1
|
Altai M, Nagy Á, Granit P, Zedan W, Cerezo-Magaña M, Park J, Lückerath K, Geres S, Sydoff M, Thorek DLJ, Westerlund K, Ulmert D, Karlström AE. Optimizing peptide nucleic acid-based pretargeting for enhanced targeted radionuclide therapy. J Control Release 2025; 381:113551. [PMID: 39986477 DOI: 10.1016/j.jconrel.2025.02.047] [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: 08/27/2024] [Revised: 01/17/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Radiolabeled targeting agents have emerged as valuable tools for the treatment of disseminated cancer. Monoclonal antibodies (mAbs) are widely employed as carriers for diagnostic and therapeutic radionuclides due to their exceptional specificity and affinity. However, their prolonged circulatory half-life can diminish diagnostic efficacy and increase radiation exposure to non-target tissues in therapeutic applications, resulting in dose-limiting toxicities. To overcome this limitation, pretargeting technologies emerge as promising strategies to enhance tumor-to-background ratio and reduce radiation exposure of healthy tissues. Our previous work introduced a pretargeting concept leveraging the specific interaction between two peptide nucleic acid (PNA) probes, HP1 and HP2, as the recognition mechanism. This early iteration of the PNA-based concept showed limited efficacy when used with mAb-based vectors. To improve its performance, we re-engineered the primary and secondary targeting agents by incorporating newly designed PNA-probes. As the primary targeting agent, we functionalized trastuzumab (T), a well-characterized human epidermal growth factor receptor 2 (HER2)-targeting IgG1 mAb, with a 9-mer PNA probe (HP9). Both FcIII-based covalent UV-light crosslinking and enzyme-mediated glyco-engineering click-chemistry methods were applied to generate trastuzumab-PNA conjugates T-FcIII-HP9 and T-gly-HP9, respectively. As a radionuclide-carrying secondary agent, we utilized a 9-mer complementary PNA probe, HP16, which forms a stable duplex with HP9 as well as displaying favorable in vivo kinetics. Biacore and flow cytometry assessment of the HP9-conjugated trastuzumab agents demonstrated retained HER2-binding properties. The secondary HP16 probe, labeled with either a dye or a radionuclide, showed cell surface accumulation contingent on the presence of HP9 on the primary HER2-targeting agents. In vivo, T-gly-HP9 exhibited significantly longer blood circulation half-life and superior tumor uptake compared to T-FcIII-HP9. Further, therapeutic dosing with [177Lu]-HP16 of trastuzumab-HP9 pretargeted HER2+ tumor models resulted in significantly delayed disease progression and extended survival compared to untreated subjects. Furthermore, pretargeted [177Lu]-HP16 exhibited comparable efficacy to [177Lu]-trastuzumab in both delaying disease progression and prolonging survival. In conclusion, the optimization of our PNA-based pretargeting system has resulted in exceptional in vivo targeting characteristics and therapeutic efficacy, validating the potential of this novel approach.
Collapse
Affiliation(s)
- Mohamed Altai
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
| | - Ábel Nagy
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| | - Pauline Granit
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| | - Wahed Zedan
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
| | - Myriam Cerezo-Magaña
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
| | - Julie Park
- Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
| | - Katharina Lückerath
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, DKTK, Essen, Germany
| | - Susanne Geres
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Marie Sydoff
- Lund University Bioimaging Centre (LBIC), Lund University, Lund, Sweden
| | - Daniel L J Thorek
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States of America; Department of Biomedical Engineering, Washington University, St. Louis, MO, United States of America; Oncologic Imaging Program, Siteman Cancer Center, St. Louis, MO, United States of America
| | - Kristina Westerlund
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| | - David Ulmert
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden; Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden.
| |
Collapse
|
2
|
Westerlund K, Oroujeni M, Gestin M, Clinton J, Hani Rosly A, Tano H, Vorobyeva A, Orlova A, Eriksson Karlström A, Tolmachev V. Shorter Peptide Nucleic Acid Probes Improve Affibody-Mediated Peptide Nucleic Acid-Based Pretargeting. ACS Pharmacol Transl Sci 2024; 7:1595-1611. [PMID: 38751640 PMCID: PMC11091976 DOI: 10.1021/acsptsci.4c00106] [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: 02/28/2024] [Revised: 03/28/2024] [Accepted: 04/10/2024] [Indexed: 05/18/2024]
Abstract
Affibody-mediated PNA-based pretargeting shows promise for HER2-expressing tumor radiotherapy. In our recent study, a 15-mer ZHER2:342-HP15 affibody-PNA conjugate, in combination with a shorter 9-mer [177Lu]Lu-HP16 effector probe, emerged as the most effective pretargeting strategy. It offered a superior tumor-to-kidney uptake ratio and more efficient tumor targeting compared to longer radiolabeled effector probes containing 12 or 15 complementary PNA bases. To enhance the production efficiency of our pretargeting system, we here introduce even shorter 6-, 7-, and 8-mer secondary probes, designated as HP19, HP21, and HP20, respectively. We also explore the replacement of the original 15-mer Z-HP15 primary probe with shorter 12-mer Z-HP12 and 9-mer Z-HP9 alternatives. This extended panel of shorter PNA-based probes was synthesized using automated microwave-assisted methods and biophysically screened in vitro to identify shorter probe combinations with the most effective binding properties. In a mouse xenograft model, we evaluated the biodistribution of these probes, comparing them to the Z-HP15:[177Lu]Lu-HP16 combination. Tumor-to-kidney ratios at 4 and 144 h postinjection of the secondary probe showed no significant differences among the Z-HP9:[177Lu]Lu-HP16, Z-HP9:[177Lu]Lu-HP20, and the Z-HP15:[177Lu]Lu-HP16 pairs. Importantly, tumor uptake significantly exceeded, by several hundred-fold, that of most normal tissues, with kidney uptake being the critical organ for radiation therapy. This suggests that using a shorter 9-mer primary probe, Z-HP9, in combination with 9-mer HP16 or 8-mer HP20 secondary probes effectively targets tumors while minimizing the dose-limiting kidney uptake of radionuclide. In conclusion, the Z-HP9:HP16 and Z-HP9:HP20 probe combinations offer good prospects for both cost-effective production and efficient in vivo pretargeting of HER2-expressing tumors.
Collapse
Affiliation(s)
- Kristina Westerlund
- Department
of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology
and Health, KTH Royal Institute of Technology, Stockholm 106 91, Sweden
| | - Maryam Oroujeni
- Department
of Immunology, Genetics and
Pathology, Uppsala University, Uppsala 751 23, Sweden
- Affibody
AB, Solna 171
65, Sweden
| | - Maxime Gestin
- Department
of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology
and Health, KTH Royal Institute of Technology, Stockholm 106 91, Sweden
| | - Jacob Clinton
- Department
of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology
and Health, KTH Royal Institute of Technology, Stockholm 106 91, Sweden
| | - Alia Hani Rosly
- Department
of Immunology, Genetics and
Pathology, Uppsala University, Uppsala 751 23, Sweden
| | - Hanna Tano
- Department
of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology
and Health, KTH Royal Institute of Technology, Stockholm 106 91, Sweden
| | - Anzhelika Vorobyeva
- Department
of Immunology, Genetics and
Pathology, Uppsala University, Uppsala 751 23, Sweden
| | - Anna Orlova
- Department
of Medicinal Chemistry, Uppsala University, Uppsala 751 23, Sweden
| | - Amelie Eriksson Karlström
- Department
of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology
and Health, KTH Royal Institute of Technology, Stockholm 106 91, Sweden
| | - Vladimir Tolmachev
- Department
of Immunology, Genetics and
Pathology, Uppsala University, Uppsala 751 23, Sweden
| |
Collapse
|
3
|
Gao F, Liu F, Wang J, Bi J, Zhai L, Li D. Molecular probes targeting HER2 PET/CT and their application in advanced breast cancer. J Cancer Res Clin Oncol 2024; 150:118. [PMID: 38466436 PMCID: PMC10927773 DOI: 10.1007/s00432-023-05519-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/12/2023] [Indexed: 03/13/2024]
Abstract
PURPOSE Human epidermal growth factor receptor 2 (HER2)-positive breast cancer cases are among the most aggressive breast tumor subtypes. Accurately assessing HER2 expression status is vital to determining whether patients will benefit from targeted anti-HER2 treatment. HER2-targeted positron emission tomography (PET/CT) is noninvasive, enabling the real-time evaluation of breast cancer patient HER2 status with accuracy. METHODS We summarize the research progress of PET/CT targeting HER2 in breast cancer, focusing on PET/CT molecular probes targeting HER2 and their clinical application in the management of advanced breast cancer. RESULTS At present, a variety of different HER2 targeted molecular probes for PET/CT imaging have been developed, including nucleolin-labeled antibodies, antibody fragments, nanobodies, and peptides of various affinities, among others. HER2-targeted PET/CT can relatively accurately evaluate HER2 expression status in advanced breast cancer patients. It has good performance in the early detection of small HER2-positive lesions, evaluation of HER2 status in lesions that cannot be readily biopsied, evaluation of the heterogeneity of multiple metastases, identification of lesions with altered HER2 status, and evaluation of the efficacy of anti-HER2 drugs. CONCLUSION HER2-targeted PET/CT offers a promising noninvasive approach for real-time assessment of HER2 status,which can be guide targeted treatment for HER2-positive breast cancer patients. Future prospective clinical studies will be invaluable for fully evaluating the importance of HER2-targeted molecular imaging in the management of breast cancer.
Collapse
Affiliation(s)
- Fang Gao
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Fengxu Liu
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Jun Wang
- Department of Anesthesia, Armed Police Corps Hospital in Shanxi Province, Xiaodian District, Taiyuan, Shanxi, People's Republic of China
| | - Junfang Bi
- Department of Combined Traditional Chinese Medicine and West Medicine, Traditional Chinese Medicine Hospital of Shijiazhuang City, 233 Zhongshan West Road, Qiaoxi District, Shijiazhuang, Hebei, China
| | - Luoping Zhai
- Department of Nuclear Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China.
| | - Dong Li
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China.
- Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
4
|
Lundmark F, Vorobyeva A, Liu Y, Lindbo S, Xu T, Oroujeni M, Rinne SS, Rosenström U, Garousi J. Reduction of renal activity retention of radiolabeled albumin binding domain‑derived affinity proteins using a non‑residualizing label strategy compared with a cleavable glycine‑leucine‑glycine‑lysine‑linker. Mol Med Rep 2024; 29:32. [PMID: 38186305 PMCID: PMC10784736 DOI: 10.3892/mmr.2023.13155] [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: 08/25/2023] [Accepted: 11/29/2023] [Indexed: 01/09/2024] Open
Abstract
The feasibility of targeted imaging and therapy using radiolabeled albumin‑binding domain‑derived affinity proteins (ADAPTs) has been demonstrated. However, high renal uptake of radioactivity limits the maximum tolerated dose. Successful reduction of renal retention of radiolabeled Fab fragments has been demonstrated by incorporating a cleavable linker between the targeting agent and the radiometal chelator. The present study investigated if the introduction of a glycine‑leucine‑glycine‑lysine (GLGK)‑linker would reduce the kidney uptake of radiolabeled ADAPT6 and also compared it with the non‑residualizing [125I]I‑[(4‑hydroxyphenyl)ethyl]maleimide ([125I]I‑HPEM) labeling strategy. GLGK was site‑specifically coupled to human epidermal growth factor receptor 2 (HER2)‑targeting ADAPT6. Conjugates without the cleavable linker were used as controls and all constructs were labeled with lutetium‑177 (177Lu). [125I]I‑HPEM was coupled to ADAPT6 at the C‑terminus. Biodistribution of all constructs was evaluated in NMRI mice 4 h after injection. Specific binding to HER2‑expressing cells in vitro was demonstrated for all constructs. No significant difference in kidney uptake was observed between the [177Lu]Lu‑2,2',2",2"'‑(1,4,7,10‑tetraazacyclododecane‑1,4,7,10‑tetrayl)tetraacetic acid‑GLGK‑conjugates and the controls. The renal activity of [125I]I‑HPEM‑ADAPT6 was significantly lower compared with all other constructs. In conclusion, the incorporation of the cleavable GLGK‑linker did not result in lower renal retention. Therefore, the present study emphasized that, in order to achieve a reduction of renal retention, alternative molecular design strategies may be required for different targeting agents.
Collapse
Affiliation(s)
- Fanny Lundmark
- Department of Medicinal Chemistry, Uppsala University, 75123 Uppsala, Sweden
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
| | - Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
| | - Sarah Lindbo
- Department of Protein Technology, Royal Institute of Technology, 10691 Stockholm, Sweden
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
| | - Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 75123 Uppsala, Sweden
| | - Ulrika Rosenström
- Department of Medicinal Chemistry, Uppsala University, 75123 Uppsala, Sweden
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
| |
Collapse
|
5
|
Rong J, Haider A, Jeppesen TE, Josephson L, Liang SH. Radiochemistry for positron emission tomography. Nat Commun 2023; 14:3257. [PMID: 37277339 PMCID: PMC10241151 DOI: 10.1038/s41467-023-36377-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 01/30/2023] [Indexed: 06/07/2023] Open
Abstract
Positron emission tomography (PET) constitutes a functional imaging technique that is harnessed to probe biological processes in vivo. PET imaging has been used to diagnose and monitor the progression of diseases, as well as to facilitate drug development efforts at both preclinical and clinical stages. The wide applications and rapid development of PET have ultimately led to an increasing demand for new methods in radiochemistry, with the aim to expand the scope of synthons amenable for radiolabeling. In this work, we provide an overview of commonly used chemical transformations for the syntheses of PET tracers in all aspects of radiochemistry, thereby highlighting recent breakthrough discoveries and contemporary challenges in the field. We discuss the use of biologicals for PET imaging and highlight general examples of successful probe discoveries for molecular imaging with PET - with a particular focus on translational and scalable radiochemistry concepts that have been entered to clinical use.
Collapse
Affiliation(s)
- Jian Rong
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Achi Haider
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Troels E Jeppesen
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA.
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
6
|
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
|
7
|
Oroujeni M, Tano H, Vorobyeva A, Liu Y, Vorontsova O, Xu T, Westerlund K, Orlova A, Tolmachev V, Karlström AE. Affibody-Mediated PNA-Based Pretargeted Cotreatment Improves Survival of Trastuzumab-Treated Mice Bearing HER2-Expressing Xenografts. J Nucl Med 2022; 63:1046-1051. [PMID: 34711617 PMCID: PMC9258572 DOI: 10.2967/jnumed.121.262123] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 10/15/2021] [Indexed: 01/03/2023] Open
Abstract
Treatment of patients with human epidermal growth factor receptor 2 (HER2)-expressing tumors using the monoclonal antibody trastuzumab increases survival. The Affibody-based peptide nucleic acid (PNA)-mediated pretargeted radionuclide therapy has demonstrated efficacy against HER2-expressing xenografts in mice. Structural studies suggest that Affibody molecules and trastuzumab bind to different epitopes on HER2. The aim of this study was to test the hypothesis that a combination of PNA-mediated pretargeted radionuclide therapy and trastuzumab treatment of HER2-expressing xenografts can extend survival compared with monotherapies. Methods: Mutual interference of the primary pretargeting probe ZHER2:342-SR-HP1 and trastuzumab in binding to HER2-expressing cell lines was investigated in vitro. Experimental therapy evaluated the survival of mice bearing HER2-expressing SKOV-3 xenografts after treatment with vehicle, trastuzumab only, pretargeting using Affibody-PNA chimera ZHER2:342-SR-HP1 and complementary probe 177Lu-HP2, and combination of trastuzumab and pretargeting. The ethical permit limited the study to 90 d. The animals' weights were monitored during the study. After study termination, samples of liver and kidneys were evaluated by a veterinary pathologist for toxicity signs. Results: The presence of a large molar excess of trastuzumab had no influence on the affinity of ZHER2:342-SR-HP1 binding to HER2-expressing cells in vitro. The affinity of trastuzumab was not affected by a large excess of ZHER2:342-SR-HP1 The median survival of mice treated with trastuzumab (75.5 d) was significantly longer than the survival of mice treated with a vehicle (59.5 d). Median survival of mice treated with pretargeting was not reached by day 90. Six mice of 10 in this group survived, and 2 had complete remission. All mice in the combination treatment group survived, and tumors in 7 mice had disappeared at study termination. There was no significant difference between animal weights in the different treatment groups. No significant pathologic alterations were detected in livers and kidneys of treated animals. Conclusion: Treatment of mice bearing HER2-expressing xenografts with the combination of trastuzumab and Affibody-mediated PNA-based radionuclide pretargeting significantly increased survival compared with monotherapies. Cotreatment was not toxic for normal tissues.
Collapse
Affiliation(s)
- Maryam Oroujeni
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Hanna Tano
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology, and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden;,Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia; and
| | - Yongsheng Liu
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Olga Vorontsova
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Tianqi Xu
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Kristina Westerlund
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology, and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia; and,Department of Medicinal Chemistry, Uppsala University, Uppsala University, Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden;,Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia; and
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology, and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| |
Collapse
|
8
|
Hu X, Li D, Fu Y, Zheng J, Feng Z, Cai J, Wang P. Advances in the Application of Radionuclide-Labeled HER2 Affibody for the Diagnosis and Treatment of Ovarian Cancer. Front Oncol 2022; 12:917439. [PMID: 35785201 PMCID: PMC9240272 DOI: 10.3389/fonc.2022.917439] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/20/2022] [Indexed: 12/19/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is a highly expressed tumor marker in epithelial ovarian cancer, and its overexpression is considered to be a potential factor of poor prognosis. Therefore, monitoring the expression of HER2 receptor in tumor tissue provides favorable conditions for accurate localization, diagnosis, targeted therapy, and prognosis evaluation of cancer foci. Affibody has the advantages of high affinity, small molecular weight, and stable biochemical properties. The molecular probes of radionuclide-labeled HER2 affibody have recently shown broad application prospects in the diagnosis and treatment of ovarian cancer; the aim is to introduce radionuclides into the cancer foci, display systemic lesions, and kill tumor cells through the radioactivity of the radionuclides. This process seamlessly integrates the diagnosis and treatment of ovarian cancer. Current research and development of new molecular probes of radionuclide-labeled HER2 affibody should focus on overcoming the deficiencies of non-specific uptake in the kidney, bone marrow, liver, and gastrointestinal tract, and on reducing the background of the image to improve image quality. By modifying the amino acid sequence; changing the hydrophilicity, surface charge, and lipid solubility of the affibody molecule; and using different radionuclides, chelating agents, and labeling conditions to optimize the labeling method of molecular probes, the specific uptake of molecular probes at tumor sites will be improved, while reducing radioactive retention in non-target organs and obtaining the best target/non-target value. These measures will enable the clinical use of radionuclide-labeled HER2 affibody molecular probes as soon as possible, providing a new clinical path for tumor-specific diagnosis, targeted therapy, and efficacy evaluation. The purpose of this review is to describe the application of radionuclide-labeled HER2 affibody in the imaging and treatment of ovarian cancer, including its potential clinical value and dilemmas.
Collapse
Affiliation(s)
- Xianwen Hu
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dandan Li
- Department of Obstetrics, Zunyi Hospital of Traditional Chinese Medicine, Zunyi, China
| | - Yujie Fu
- Research and Development Department, Jiangsu Yuanben Biotechnology Co., Ltd., Zunyi, China
| | - Jiashen Zheng
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zelong Feng
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiong Cai
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Jiong Cai, ; Pan Wang,
| | - Pan Wang
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Jiong Cai, ; Pan Wang,
| |
Collapse
|
9
|
Tolmachev VM, Chernov VI, Deyev SM. Targeted nuclear medicine. Seek and destroy. RUSSIAN CHEMICAL REVIEWS 2022. [DOI: 10.1070/rcr5034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
10
|
Review: Radionuclide Molecular Imaging Targeting HER2 in Breast Cancer with a Focus on Molecular Probes into Clinical Trials and Small Peptides. Molecules 2021; 26:molecules26216482. [PMID: 34770887 PMCID: PMC8588233 DOI: 10.3390/molecules26216482] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/18/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
As the most frequently occurring cancer worldwide, breast cancer (BC) is the leading cause of cancer-related death in women. The overexpression of HER2 (human epidermal growth factor receptor 2) is found in about 15% of BC patients, and it is often associated with a poor prognosis due to the effect on cell proliferation, migration, invasion, and survival. As a result of the heterogeneity of BC, molecular imaging with HER2 probes can non-invasively, in real time, and quantitatively reflect the expression status of HER2 in tumors. This will provide a new approach for patients to choose treatment options and monitor treatment response. Furthermore, radionuclide molecular imaging has the potential of repetitive measurements, and it can help solve the problem of heterogeneous expression and conversion of HER2 status during disease progression or treatment. Different imaging probes of targeting proteins, such as monoclonal antibodies, antibody fragments, nanobodies, and affibodies, are currently in preclinical and clinical development. Moreover, in recent years, HER2-specific peptides have been widely developed for molecular imaging techniques for HER2-positive cancers. This article summarized different types of molecular probes targeting HER2 used in current clinical applications and the developmental trend of some HER2-specific peptides.
Collapse
|
11
|
Jallinoja VIJ, Carney BD, Zhu M, Bhatt K, Yazaki PJ, Houghton JL. Cucurbituril-Ferrocene: Host-Guest Based Pretargeted Positron Emission Tomography in a Xenograft Model. Bioconjug Chem 2021; 32:1554-1558. [PMID: 34156824 PMCID: PMC9153067 DOI: 10.1021/acs.bioconjchem.1c00280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pretargeted positron emission tomography is a macromolecule-driven nuclear medicine technique that involves targeting a preadministered antigen target-bound macromolecule with a radioligand in vivo, aiming to minimize the overall radiation dose. This study investigates the use of antibody based host-guest chemistry methodology for pretargeted positron emission tomography. We hypothesize that the novel pretargeting approach reported here overcomes the challenges the current pretargeting platforms have with the in vivo stability and modularity of the pretargeting components. A cucurbit[7]uril host molecule modified, anti-carcinoembryonic antigen antibody (M5A; CB7-M5A) and a 68Ga-radiolabeled ferrocene guest radioligand ([68Ga]Ga-NOTA-PEG3-NMe2-Fc) were studied as potential host-guest chemistry pretargeting agents for positron emission tomography in BxPC3 xenografted nude mice. The viability of the platform was studied via in vivo biodistribution and positron emission tomography. Tumor uptake of [68Ga]Ga-NOTA-PEG3-NMe2-Fc was significantly higher in mice which received CB7-M5A prior to the radioligand injection (pretargeted) (3.3 ± 0.7%ID/g) compared to mice which only received the radioligand (nonpretargeted) (0.2 ± 0.1%ID/g).
Collapse
Affiliation(s)
- Vilma IJ Jallinoja
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
- Department of Radiology, Stony Brook University, Stony Brook, New York, 11774, USA
| | - Brandon D Carney
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
- Department of Radiology, Stony Brook University, Stony Brook, New York, 11774, USA
| | - Meiying Zhu
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
| | - Kavita Bhatt
- Department of Radiology, Stony Brook University, Stony Brook, New York, 11774, USA
| | - Paul J Yazaki
- Beckman Institute, City of Hope, Duarte, California 91010, USA
| | - Jacob L Houghton
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
- Department of Radiology, Stony Brook University, Stony Brook, New York, 11774, USA
| |
Collapse
|
12
|
Chigoho DM, Bridoux J, Hernot S. Reducing the renal retention of low- to moderate-molecular-weight radiopharmaceuticals. Curr Opin Chem Biol 2021; 63:219-228. [PMID: 34325089 DOI: 10.1016/j.cbpa.2021.06.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023]
Abstract
The field of nuclear imaging and therapy is rapidly progressing with the development of targeted radiopharmaceuticals that show rapid targeting and rapid clearance with minimal background. Unfortunately, they are often reabsorbed in the kidneys, leading to possible nephrotoxicity, limiting the therapeutic dose, and/or reducing imaging quality. The blocking of endocytic receptors has been extensively used as a strategy to reduce kidney radiation. Alternatively, the physicochemical properties of radiotracers can be modulated to either prevent their reuptake or promote the excretion of radiometabolites. Other interesting strategies focus on the insertion of a cleavable linker between the radiolabel and the targeting moiety or pretargeting approaches in which the targeting moiety and radiolabel are administered separately. In the context of this review, we will discuss the latest advances and insights on strategies used to reduce renal retention of low- to moderate-molecular-weight radiopharmaceuticals.
Collapse
Affiliation(s)
- Dora Mugoli Chigoho
- Laboratory for in Vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Jessica Bridoux
- Laboratory for in Vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Sophie Hernot
- Laboratory for in Vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
13
|
Tano H, Oroujeni M, Vorobyeva A, Westerlund K, Liu Y, Xu T, Vasconcelos D, Orlova A, Karlström AE, Tolmachev V. Comparative Evaluation of Novel 177Lu-Labeled PNA Probes for Affibody-Mediated PNA-Based Pretargeting. Cancers (Basel) 2021; 13:cancers13030500. [PMID: 33525578 PMCID: PMC7865858 DOI: 10.3390/cancers13030500] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Affibody molecules are small, engineered affinity proteins based on a nonimmunoglobulin scaffold. Affibody-based radionuclide imaging probes have demonstrated excellent tumor targeting. However, the renal clearance of affibody molecules is accompanied by high reabsorption and retention of activity in the kidney, which prevents their use for radionuclide therapy. We have previously shown the feasibility of overcoming the high renal uptake using a pretargeting approach for affibody-mediated therapy based on peptide nucleic acid (PNA) hybridization. In this study, we test the hypothesis that shortening the PNA pretargeting probes would further increase the difference between the accumulation of radiometals in tumor xenografts and in kidneys. A series of novel PNA probes has been designed and evaluated in vitro and in vivo. We have found that a variant containing 9 nucleobases enables a two-fold increase of the tumor-to-kidney dose ratio compared with a variant containing 15 nucleobases. This creates preconditions for more efficient therapy of cancer. Abstract Affibody-mediated PNA-based pretargeting is a promising approach to radionuclide therapy of HER2-expressing tumors. In this study, we test the hypothesis that shortening the PNA pretargeting probes would increase the tumor-to-kidney dose ratio. The primary probe ZHER2:342-SR-HP15 and the complementary secondary probes HP16, HP17, and HP18, containing 9, 12, and 15 nucleobases, respectively, and carrying a 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) chelator were designed, synthesized, characterized in vitro, and labeled with 177Lu. In vitro pretargeting was studied in HER2-expressing SKOV3 and BT474 cell lines. The biodistribution of these novel probes was evaluated in immunodeficient mice bearing SKOV3 xenografts and compared to the previously studied [177Lu]Lu-HP2. Characterization confirmed the formation of high-affinity duplexes between HP15 and the secondary probes, with the affinity correlating with the length of the complementary PNA sequences. All the PNA-based probes were bound specifically to HER2-expressing cells in vitro. In vivo studies demonstrated HER2-specific uptake of all 177Lu-labeled probes in xenografts in a pretargeting setting. The ratio of cumulated radioactivity in the tumor to the radioactivity in kidneys was dependent on the secondary probe’s size and decreased with an increased number of nucleobases. The shortest PNA probe, [177Lu]Lu-HP16, showed the highest tumor-to-kidney ratio. [177Lu]Lu-HP16 is the most promising secondary probe for affibody-mediated tumor pretargeting.
Collapse
Affiliation(s)
- Hanna Tano
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (H.T.); (K.W.); (D.V.)
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (Y.L.); (T.X.); (V.T.)
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (Y.L.); (T.X.); (V.T.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia;
| | - Kristina Westerlund
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (H.T.); (K.W.); (D.V.)
| | - Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (Y.L.); (T.X.); (V.T.)
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (Y.L.); (T.X.); (V.T.)
| | - Daniel Vasconcelos
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (H.T.); (K.W.); (D.V.)
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia;
- Department of Medicinal Chemistry, Dag Hammarskjölds väg 14C, Uppsala University, 751 23 Uppsala, Sweden
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (H.T.); (K.W.); (D.V.)
- Correspondence:
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (Y.L.); (T.X.); (V.T.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia;
| |
Collapse
|
14
|
Abstract
Affibody molecules are small engineered scaffold proteins suitable for in vivo tumor targeting. Radionuclide molecular imaging using directly radiolabelled affibody molecules provides excellent imaging. However, affibody molecules have a high renal reabsorption, which complicates their use for radionuclide therapy. The high renal reabsorption is a common problem for the use of engineered scaffold proteins for radionuclide therapy. Affibody-based PNA-mediated pretargeting reduces dramatically the absorbed dose to the kidneys and makes affibody-based radionuclide therapy possible. This methodology might, hopefully, solve the problem of high renal reabsorption for radionuclide therapy mediated by other engineered scaffold proteins.
Collapse
|
15
|
Abstract
In the recent years, progress in nanotechnology has significantly contributed to the development of novel pharmaceutical formulations to overcome the drawbacks of conventional treatments and improve the therapeutic outcome in many diseases, especially cancer. Nanoparticle vectors have demonstrated the potential to concomitantly deliver diagnostic and therapeutic payloads to diseased tissue. Due to their special physical and chemical properties, the characteristics and function of nanoparticles are tunable based on biological molecular targets and specific desired features (e.g., surface chemistry and diagnostic radioisotope labeling). Within the past decade, several theranostic nanoparticles have been developed as a multifunctional nanosystems which combine the diagnostic and therapeutic functionalities into a single drug delivery platform. Theranostic nanosystems can provide useful information on a real-time systemic distribution of the developed nanosystem and simultaneously transport the therapeutic payload. In general, the diagnostic functionality of theranostic nanoparticles can be achieved through labeling gamma-emitted radioactive isotopes on the surface of nanoparticles which facilitates noninvasive detection using nuclear molecular imaging techniques, such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT), meanwhile, the therapeutic effect arises from the potent drug released from the nanoparticle. Moreover, some radioisotopes can concurrently emit both gamma radiation and high-energy particles (e.g., alpha, beta, and Auger electrons), prompting the use either alone for radiotheranostics or synergistically with chemotherapy. This chapter provides an overview of the fundamentals of radiochemistry and relevant radiolabeling strategies for theranostic nanosystem development as well as the methods for the preclinical evaluation of radiolabeled nanoparticles. Furthermore, preclinical case studies of recently developed theranostic nanosystems will be highlighted.
Collapse
|
16
|
Mukai H, Watanabe Y. Review: PET imaging with macro- and middle-sized molecular probes. Nucl Med Biol 2021; 92:156-170. [PMID: 32660789 DOI: 10.1016/j.nucmedbio.2020.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022]
Abstract
Recent progress in radiolabeling of macro- and middle-sized molecular probes has been extending possibilities to use PET molecular imaging for dynamic application to drug development and therapeutic evaluation. Theranostics concept also accelerated the use of macro- and middle-sized molecular probes for sharpening the contrast of proper target recognition even the cellular types/subtypes and proper selection of the patients who should be treated by the same molecules recognition. Here, brief summary of the present status of immuno-PET, and then further development of advanced technologies related to immuno-PET, peptidic PET probes, and nucleic acids PET probes are described.
Collapse
Affiliation(s)
- Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| |
Collapse
|
17
|
Evaluation of an antibody-PNA conjugate as a clearing agent for antibody-based PNA-mediated radionuclide pretargeting. Sci Rep 2020; 10:20777. [PMID: 33247180 PMCID: PMC7695838 DOI: 10.1038/s41598-020-77523-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Radionuclide molecular imaging of cancer-specific targets is a promising method to identify patients for targeted antibody therapy. Radiolabeled full-length antibodies however suffer from slow clearance, resulting in high background radiation. To overcome this problem, a pretargeting system based on complementary peptide nucleic acid (PNA) probes has been investigated. The pretargeting relies on sequential injections of primary, PNA-tagged antibody and secondary, radiolabeled PNA probe, which are separated in time, to allow for clearance of non-bound primary agent. We now suggest to include a clearing agent (CA), designed for removal of primary tumor-targeting agent from the blood. The CA is based on the antibody cetuximab, which was conjugated to PNA and lactosaminated by reductive amination to improve hepatic clearance. The CA was evaluated in combination with PNA-labelled trastuzumab, T-ZHP1, for radionuclide HER2 pretargeting. Biodistribution studies in normal mice demonstrated that the CA cleared ca. 7 times more rapidly from blood than unmodified cetuximab. Injection of the CA 6 h post injection of the radiolabeled primary agent [131I]I-T-ZHP1 gave a moderate reduction of the radioactivity concentration in the blood after 1 h from 8.5 ± 1.8 to 6.0 ± 0.4%ID/g. These proof-of-principle results could guide future development of a more efficient CA.
Collapse
|
18
|
Investigation of a Pharmacological Approach for Reduction of Renal Uptake of Radiolabeled ADAPT Scaffold Protein. Molecules 2020; 25:molecules25194448. [PMID: 32998229 PMCID: PMC7583817 DOI: 10.3390/molecules25194448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022] Open
Abstract
Albumin binding domain-Derived Affinity ProTeins (ADAPTs) are small (5 kDa) engineered scaffold proteins that are promising targeting agents for radionuclide-based imaging. A recent clinical study has demonstrated that radiolabeled ADAPTs can efficiently visualize human epidermal growth factor receptor 2 (HER2) expression in breast cancer using SPECT imaging. However, the use of ADAPTs directly labeled with radiometals for targeted radionuclide therapy is limited by their high reabsorption and prolonged retention of activity in kidneys. In this study, we investigated whether a co-injection of lysine or gelofusin, commonly used for reduction of renal uptake of radiolabeled peptides in clinics, would reduce the renal uptake of [99mTc]Tc(CO)3-ADAPT6 in NMRI mice. In order to better understand the mechanism behind the reabsorption of [99mTc]Tc(CO)3-ADAPT6, we included several compounds that act on various parts of the reabsorption system in kidneys. Administration of gelofusine, lysine, probenecid, furosemide, mannitol, or colchicine did not change the uptake of [99mTc]Tc(CO)3-ADAPT6 in kidneys. Sodium maleate reduced the uptake of [99mTc]Tc(CO)3-ADAPT6 to ca. 25% of the uptake in the control, a high dose of fructose (50 mmol/kg) reduced the uptake by ca. two-fold. However, a lower dose (20 mmol/kg) had no effect. These results indicate that common clinical strategies are not effective for reduction of kidney uptake of [99mTc]Tc(CO)3-ADAPT6 and that other strategies for reduction of activity uptake or retention in kidneys should be investigated for ADAPT6.
Collapse
|
19
|
Lee JY, Park T, Hong E, Amatya R, Park KA, Park YH, Min KA, Jin M, Lee S, Hwang S, Roh GS, Shin MC. Genetic engineering of novel super long-acting Exendin-4 chimeric protein for effective treatment of metabolic and cognitive complications of obesity. Biomaterials 2020; 257:120250. [PMID: 32736262 DOI: 10.1016/j.biomaterials.2020.120250] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/21/2020] [Accepted: 07/17/2020] [Indexed: 02/08/2023]
Abstract
A common bottleneck challenge for many therapeutic proteins lies in their short plasma half-lives, which often makes the treatment far less compliant or even disables achieving sufficient therapeutic efficacy. To address this problem, we introduce a novel drug delivery strategy based on the genetic fusion of an albumin binding domain (ABD) and an anti-neonatal Fc receptor (FcRn) affibody (AFF) to therapeutic proteins. This ABD-AFF fusion strategy can provide a synergistic effect on extending the plasma residence time by, on one hand, preventing the rapid glomerular filtration via ABD-mediated albumin binding and, on the other hand, increasing the efficiency of FcRn-mediated recycling by AFF-mediated high-affinity binding to the FcRn. In this research, we explored the feasibility of applying the ABD-AFF fusion strategy to exendin-4 (EX), a clinically available anti-diabetic peptide possessing a short plasma half-life. The EX-ABD-AFF produced from the E. coli displayed a remarkably (241-fold) longer plasma half-life than the SUMO tagged-EX (SUMO-EX) (0.7 h) in mice. Furthermore, in high-fat diet (HFD)-fed obese mice model, the EX-ABD-AFF could provide significant hypoglycemic effects for over 12 days, accompanied by a reduction of body weight. In the long-term study, the EX-ABD-AFF could significantly reverse the obesity-related metabolic complications (hyperglycemia, hyperlipidemia, and hepatic steatosis) and, moreover, improve cognitive deficits. Overall, this study demonstrated that the ABD-AFF fusion could be an effective strategy to greatly increase the plasma half-lives of therapeutic proteins and thus markedly improve their druggability.
Collapse
Affiliation(s)
- Jong Youl Lee
- Department of Anatomy and Convergence Medical Science, Bio Anti-Aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, 52727, Republic of Korea
| | - Taehoon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju, Gyeongnam, 52828, Republic of Korea
| | - Eunmi Hong
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, Republic of Korea
| | - Reeju Amatya
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju, Gyeongnam, 52828, Republic of Korea
| | - Kyung-Ah Park
- Department of Anatomy and Convergence Medical Science, Bio Anti-Aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, 52727, Republic of Korea
| | - Young-Hoon Park
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, Republic of Korea
| | - Kyoung Ah Min
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam, 50834, Republic of Korea
| | - Minki Jin
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam, 50834, Republic of Korea
| | - Sumi Lee
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam, 50834, Republic of Korea
| | - Seungmi Hwang
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam, 50834, Republic of Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, Bio Anti-Aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, 52727, Republic of Korea.
| | - Meong Cheol Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju, Gyeongnam, 52828, Republic of Korea.
| |
Collapse
|
20
|
Hapuarachchige S, Artemov D. Theranostic Pretargeting Drug Delivery and Imaging Platforms in Cancer Precision Medicine. Front Oncol 2020; 10:1131. [PMID: 32793481 PMCID: PMC7387661 DOI: 10.3389/fonc.2020.01131] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 06/05/2020] [Indexed: 12/29/2022] Open
Abstract
Theranostics are nano-size or molecular-level agents serving for both diagnosis and therapy. Structurally, they are drug delivery systems integrated with molecular or targeted imaging agents. Theranostics are becoming popular because they are targeted therapeutics and can be used with no or minimal changes for diagnostic imaging to aid in precision medicine. Thus, there is a close relation between theranostics and image-guided therapy (IGT), and theranostics are actually a subclass of IGT in which both therapeutic and imaging functionalities are attributed to a single platform. An important theranostics strategy is biological pretargeting. In pretargeted IGT, first, the target is identified by a target-specific natural or synthetic bioligand followed by a nano-scale or molecular drug delivery component, which form therapeutic clusters by in situ conjugation reactions. If pretargeted drug delivery platforms are labeled with multimodal imaging probes, they can be used as theranostics for both diagnostic imaging and therapy. Optical and nuclear imaging techniques have mostly been used in proof-of-concept studies with pretargeted theranostics. The concept of pretargeting in theranostics is comparatively novel and generally requires a confirmed overexpression of surface receptors on targeted cells/tissue. In addition, the receptors should have natural or synthetic bioligands to be used as pretargeting components. Therefore, applications of pretargeting theranostics are still limited to several cancer types, which overexpress cell-surface markers on the target cancer cells. In this review, recent discoveries of pretargeting theranostics in breast, ovarian, prostate, and colorectal cancers are discussed to highlight main strengths and potential limitations the strategy.
Collapse
Affiliation(s)
- Sudath Hapuarachchige
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Dmitri Artemov
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
21
|
Garousi J, Vorobyeva A, Altai M. Influence of Several Compounds and Drugs on the Renal Uptake of Radiolabeled Affibody Molecules. Molecules 2020; 25:molecules25112673. [PMID: 32526905 PMCID: PMC7321166 DOI: 10.3390/molecules25112673] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 12/19/2022] Open
Abstract
Affibody molecules are the most studied class of engineered scaffold proteins (ESPs) in radionuclide molecular imaging. Attempts to use affibody molecules directly labelled with radiometals for targeted radionuclide therapy were hampered by the high uptake and retention of radioactivity in kidneys. Several promising strategies have been implemented to circumvent this problem. Here, we investigated whether a pharmacological approach targeting different components of the reabsorption system could be used to lower the uptake of [99mTc]Tc-ZHER:2395 affibody molecule in kidneys. Pre-injection of probenecid, furosemide, mannitol or colchicine had no influence on activity uptake in kidneys compared to the control group. Mice pre-injected with maleate and fructose had 33% and 51% reduction in the kidney-associated activity, respectively, compared to the control group. Autoradiography images showed that the accumulation of activity after [99mTc]Tc-ZHER2:2395 injection was in the renal cortex and that both maleate and fructose could significantly reduce it. Results from this study demonstrate that pharmacological intervention with maleate and fructose was effective in reducing the kidney uptake of affibody molecules. A presumable mechanism is the disruption of ATP-mediated cellular uptake and endocytosis processes of affibody molecules by tubular cells.
Collapse
Affiliation(s)
- Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (J.G.); (A.V.)
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (J.G.); (A.V.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia
| | - Mohamed Altai
- Division of Oncology and Pathology, Kamprad Lab, Department of Clinical Sciences, Lund University, 222 43 Lund, Sweden
- Correspondence: ; Tel.: +46-704128699
| |
Collapse
|
22
|
Qi S, Hoppmann S, Xu Y, Cheng Z. PET Imaging of HER2-Positive Tumors with Cu-64-Labeled Affibody Molecules. Mol Imaging Biol 2020; 21:907-916. [PMID: 30617730 DOI: 10.1007/s11307-018-01310-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE Previous studies has demonstrated the utility of human epidermal growth factor receptor type 2 (HER2) as an attractive target for cancer molecular imaging and therapy. An affibody protein with strong binding affinity for HER2, ZHER2:342, has been reported. Various methods of chelator conjugation for radiolabeling HER2 affibody molecules have been described in the literature including N-terminal conjugation, C-terminal conjugation, and other methods. Cu-64 has recently been extensively evaluated due to its half-life, decay properties, and availability. Our goal was to optimize the radiolabeling method of this affibody molecule with Cu-64, and translate a positron emission tomography (PET) probe with the best in vivo performance to clinical PET imaging of HER2-positive cancers. PROCEDURES In our study, three anti-HER2 affibody proteins-based PET probes were prepared, and their in vivo performance was evaluated in mice bearing HER2-positive subcutaneous SKOV3 tumors. The affibody analogues, Ac-Cys-ZHER2:342, Ac-ZHER2:342(Cys39), and Ac-ZHER2:342-Cys, were synthesized using the solid phase peptide synthesis method. The purified small proteins were site-specifically conjugated with the maleimide-functionalized chelator, 1,4,7,10-tetraazacyclododecane-1,4,7-tris- aceticacid-10-maleimidethylacetamide (maleimido-mono-amide-DOTA). The resulting DOTA-affibody conjugates were then radiolabeled with Cu-64. Cell uptake assay of the resulting PET probes, [64Cu]DOTA-Cys-ZHER2:342, [64Cu]DOTA-ZHER2:342(Cys39), and [64Cu]DOTA-ZHER2:342-Cys, was performed in HER2-positive human ovarian SKOV3 carcinoma cells at 4 and 37 °C. The binding affinities of the radiolabeled peptides were tested by cell saturation assay using SKOV3 cells. PET imaging, biodistribution, and metabolic stability studies were performed in mice bearing SKOV3 tumors. RESULTS Cell uptake assays showed high and specific uptake by incubation of Cu-64-labeled affibodies with SKOV3 cells. The affinities (KD) of the PET radio probes as tested by cell saturation analysis were in the low nanomolar range with the ranking of [64Cu]DOTA-Cys-ZHER2:342 (25.2 ± 9.2 nM) ≈ [64Cu]DOTA-ZHER2:342-Cys (32.6 ± 14.7 nM) > [64Cu]DOTA-ZHER2:342(Cys39) (77.6 ± 22.2 nM). In vitro stability and in vivo metabolite analysis study revealed that all three probes were stable enough for in vivo imaging applications, while [64Cu]DOTA-Cys-ZHER2:342 showed the highest stability. In vivo small-animal PET further demonstrated fast tumor targeting, good tumor accumulation, and good tumor to normal tissue contrast of all three probes. For [64Cu]DOTA-Cys-ZHER2:342, [64Cu]DOTA-ZHER2:342(Cys39), and [64Cu]DOTA-ZHER2:342-Cys, tumor uptake at 24 h are 4.0 ± 1.0 % ID/g, 4.0 ± 0.8 %ID/g, and 4.3 ± 0.7 %ID/g, respectively (mean ± SD, n = 4). Co-injection of the probes with non-labeled anti-HER2 affibody proteins confirmed in vivo specificities of the compounds by tumor uptake reduction. CONCLUSIONS The three Cu-64-labeled ZHER2:342 analogues all display excellent HER2 targeting ability and tumor PET imaging quality. Although varied in the position of the radiometal labeling of these three Cu-64-labeled ZHER2:342 analogues, there is no significant difference in tumor and normal tissue uptakes among the three probes. [64Cu]DOTA-Cys-ZHER2:342 stands out as the most superior PET probe because of its highest affinities and in vivo stability.
Collapse
Affiliation(s)
- Shibo Qi
- School of Environmental and Chemical Engineering, Tianjin Polytechnic University, Tianjin, 300387, China.,Molecular Imaging Program at Stanford (MIPS), Department of Radiology, and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, 94305-5344, USA
| | - Susan Hoppmann
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, 94305-5344, USA
| | - Yingding Xu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, 94305-5344, USA
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, 94305-5344, USA.
| |
Collapse
|
23
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
24
|
Perrin J, Capitao M, Mougin-Degraef M, Guérard F, Faivre-Chauvet A, Rbah-Vidal L, Gaschet J, Guilloux Y, Kraeber-Bodéré F, Chérel M, Barbet J. Cell Tracking in Cancer Immunotherapy. Front Med (Lausanne) 2020; 7:34. [PMID: 32118018 PMCID: PMC7033605 DOI: 10.3389/fmed.2020.00034] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
The impressive development of cancer immunotherapy in the last few years originates from a more precise understanding of control mechanisms in the immune system leading to the discovery of new targets and new therapeutic tools. Since different stages of disease progression elicit different local and systemic inflammatory responses, the ability to longitudinally interrogate the migration and expansion of immune cells throughout the whole body will greatly facilitate disease characterization and guide selection of appropriate treatment regiments. While using radiolabeled white blood cells to detect inflammatory lesions has been a classical nuclear medicine technique for years, new non-invasive methods for monitoring the distribution and migration of biologically active cells in living organisms have emerged. They are designed to improve detection sensitivity and allow for a better preservation of cell activity and integrity. These methods include the monitoring of therapeutic cells but also of all cells related to a specific disease or therapeutic approach. Labeling of therapeutic cells for imaging may be performed in vitro, with some limitations on sensitivity and duration of observation. Alternatively, in vivo cell tracking may be performed by genetically engineering cells or mice so that may be revealed through imaging. In addition, SPECT or PET imaging based on monoclonal antibodies has been used to detect tumors in the human body for years. They may be used to detect and quantify the presence of specific cells within cancer lesions. These methods have been the object of several recent reviews that have concentrated on technical aspects, stressing the differences between direct and indirect labeling. They are briefly described here by distinguishing ex vivo (labeling cells with paramagnetic, radioactive, or fluorescent tracers) and in vivo (in vivo capture of injected radioactive, fluorescent or luminescent tracers, or by using labeled antibodies, ligands, or pre-targeted clickable substrates) imaging methods. This review focuses on cell tracking in specific therapeutic applications, namely cell therapy, and particularly CAR (Chimeric Antigen Receptor) T-cell therapy, which is a fast-growing research field with various therapeutic indications. The potential impact of imaging on the progress of these new therapeutic modalities is discussed.
Collapse
Affiliation(s)
- Justine Perrin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marisa Capitao
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marie Mougin-Degraef
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - François Guérard
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Alain Faivre-Chauvet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - Latifa Rbah-Vidal
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Joëlle Gaschet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Yannick Guilloux
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Françoise Kraeber-Bodéré
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | - Michel Chérel
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | | |
Collapse
|
25
|
Garousi J, Huizing FJ, Vorobyeva A, Mitran B, Andersson KG, Leitao CD, Frejd FY, Löfblom J, Bussink J, Orlova A, Heskamp S, Tolmachev V. Comparative evaluation of affibody- and antibody fragments-based CAIX imaging probes in mice bearing renal cell carcinoma xenografts. Sci Rep 2019; 9:14907. [PMID: 31624303 PMCID: PMC6797765 DOI: 10.1038/s41598-019-51445-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/25/2019] [Indexed: 12/11/2022] Open
Abstract
Carbonic anhydrase IX (CAIX) is a cancer-associated molecular target for several classes of therapeutics. CAIX is overexpressed in a large fraction of renal cell carcinomas (RCC). Radionuclide molecular imaging of CAIX-expression might offer a non-invasive methodology for stratification of patients with disseminated RCC for CAIX-targeting therapeutics. Radiolabeled monoclonal antibodies and their fragments are actively investigated for imaging of CAIX expression. Promising alternatives are small non-immunoglobulin scaffold proteins, such as affibody molecules. A CAIX-targeting affibody ZCAIX:2 was re-designed with the aim to decrease off-target interactions and increase imaging contrast. The new tracer, DOTA-HE3-ZCAIX:2, was labeled with 111In and characterized in vitro. Tumor-targeting properties of [111In]In-DOTA-HE3-ZCAIX:2 were compared head-to-head with properties of the parental variant, [99mTc]Tc(CO)3-HE3-ZCAIX:2, and the most promising antibody fragment-based tracer, [111In]In-DTPA-G250(Fab’)2, in the same batch of nude mice bearing CAIX-expressing RCC xenografts. Compared to the 99mTc-labeled parental variant, [111In]In-DOTA-HE3-ZCAIX:2 provides significantly higher tumor-to-lung, tumor-to-bone and tumor-to-liver ratios, which is essential for imaging of CAIX expression in the major metastatic sites of RCC. [111In]In-DOTA-HE3-ZCAIX:2 offers significantly higher tumor-to-organ ratios compared with [111In]In-G250(Fab’)2. In conclusion, [111In]In-DOTA-HE3-ZCAIX:2 can be considered as a highly promising tracer for imaging of CAIX expression in RCC metastases based on our results and literature data.
Collapse
Affiliation(s)
- Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Fokko J Huizing
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bogdan Mitran
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Ken G Andersson
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Charles Dahlsson Leitao
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Y Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Sandra Heskamp
- Department of Radiology and Nuclear medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
26
|
Yonekura Y, Mattsson S, Flux G, Bolch WE, Dauer LT, Fisher DR, Lassmann M, Palm S, Hosono M, Doruff M, Divgi C, Zanzonico P. ICRP Publication 140: Radiological Protection in Therapy with Radiopharmaceuticals. Ann ICRP 2019; 48:5-95. [PMID: 31565950 DOI: 10.1177/0146645319838665] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Radiopharmaceuticals are increasingly used for the treatment of various cancers with novel radionuclides, compounds, tracer molecules, and administration techniques. The goal of radiation therapy, including therapy with radiopharmaceuticals, is to optimise the relationship between tumour control probability and potential complications in normal organs and tissues. Essential to this optimisation is the ability to quantify the radiation doses delivered to both tumours and normal tissues. This publication provides an overview of therapeutic procedures and a framework for calculating radiation doses for various treatment approaches. In radiopharmaceutical therapy, the absorbed dose to an organ or tissue is governed by radiopharmaceutical uptake, retention in and clearance from the various organs and tissues of the body, together with radionuclide physical half-life. Biokinetic parameters are determined by direct measurements made using techniques that vary in complexity. For treatment planning, absorbed dose calculations are usually performed prior to therapy using a trace-labelled diagnostic administration, or retrospective dosimetry may be performed on the basis of the activity already administered following each therapeutic administration. Uncertainty analyses provide additional information about sources of bias and random variation and their magnitudes; these analyses show the reliability and quality of absorbed dose calculations. Effective dose can provide an approximate measure of lifetime risk of detriment attributable to the stochastic effects of radiation exposure, principally cancer, but effective dose does not predict future cancer incidence for an individual and does not apply to short-term deterministic effects associated with radiopharmaceutical therapy. Accident prevention in radiation therapy should be an integral part of the design of facilities, equipment, and administration procedures. Minimisation of staff exposures includes consideration of equipment design, proper shielding and handling of sources, and personal protective equipment and tools, as well as education and training to promote awareness and engagement in radiological protection. The decision to hold or release a patient after radiopharmaceutical therapy should account for potential radiation dose to members of the public and carers that may result from residual radioactivity in the patient. In these situations, specific radiological protection guidance should be provided to patients and carers.
Collapse
|
27
|
Bodet-Milin C, Bailly C, Touchefeu Y, Frampas E, Bourgeois M, Rauscher A, Lacoeuille F, Drui D, Arlicot N, Goldenberg DM, Faivre-Chauvet A, Barbet J, Rousseau C, Kraeber-Bodéré F. Clinical Results in Medullary Thyroid Carcinoma Suggest High Potential of Pretargeted Immuno-PET for Tumor Imaging and Theranostic Approaches. Front Med (Lausanne) 2019; 6:124. [PMID: 31214593 PMCID: PMC6558173 DOI: 10.3389/fmed.2019.00124] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/17/2019] [Indexed: 12/03/2022] Open
Abstract
Monoclonal antibody (mAb)-based therapies have experienced considerable growth in cancer management. When labeled with radionuclides, mAbs also represent promising probes for imaging or theranostic approaches. Initially, mAbs have been radiolabeled with single-photon emitters, such as 131I, 99mTc, or 111In, for diagnostic purposes or to improve radioimmunotherapy (RIT) using dosimetry estimations. Today, more accurate imaging is achieved using positron- emission tomography (PET). Thanks to the important technical advances in the production of PET emitters and their related radiolabeling methods, the last decade has witnessed the development of a broad range of new probes for specific PET imaging. Immuno-PET, which combines the high sensitivity and resolution of a PET camera with the specificity of a monoclonal antibody, is fully in line with this approach. As RIT, immuno-PET can be performed using directly radiolabeled mAbs or using pretargeting to improve imaging contrast. Pretargeted immuno-PET has been developed against different antigens, and promising results have been reported in tumor expressing carcinoembryonic antigen (CEA; CEACAM5) using a bispecific mAb (BsmAb) and a radiolabeled peptide. Medullary thyroid carcinoma (MTC) is an uncommon thyroid cancer subtype which accounts for <10% of all thyroid neoplasms. Characterized by an intense expression of CEA, MTC represents a relevant tumor model for immuno-PET. High sensitivity of pretargeted immunoscintigraphy using murine or chimeric anti-CEA BsMAb and pretargeted haptens-peptides labeled with 111In or 131I were reported in metastatic MTC patients 20 years ago. Recently, an innovative clinical study reported high tumor uptake and contrast using pretargeted anti-CEA immuno-PET in relapsed MTC patients. This review focuses on MTC as an example, but the same pretargeting technique has been applied with success for clinical PET imaging of other CEA-expressing tumors and other pretargeting systems. In particular, those exploiting bioorthogonal chemistry also appear interesting in preclinical animal models, suggesting the high potential of pretargeting for diagnostic and theranostic applications.
Collapse
Affiliation(s)
- Caroline Bodet-Milin
- Nuclear Medicine, University Hospital, Nantes, France
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Clément Bailly
- Nuclear Medicine, University Hospital, Nantes, France
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Yann Touchefeu
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
- Hepato-Gastro-enterology, University Hospital, Nantes, France
| | - Eric Frampas
- Nuclear Medicine, University Hospital, Nantes, France
- Radiology, University Hospital, Nantes, France
| | - Mickael Bourgeois
- Nuclear Medicine, University Hospital, Nantes, France
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Aurore Rauscher
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
- Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | | | - Delphine Drui
- Endocrinology Department, University Hospital, Nantes, France
| | | | - David M. Goldenberg
- IBC Pharmaceuticals, Inc., Morris Plains, NJ, United States
- Immunomedics, Inc., Morris Plains, NJ, United States
| | - Alain Faivre-Chauvet
- Nuclear Medicine, University Hospital, Nantes, France
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | | | - Caroline Rousseau
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
- Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | - Françoise Kraeber-Bodéré
- Nuclear Medicine, University Hospital, Nantes, France
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
- Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| |
Collapse
|
28
|
Site-specific conjugation of recognition tags to trastuzumab for peptide nucleic acid-mediated radionuclide HER2 pretargeting. Biomaterials 2019; 203:73-85. [PMID: 30877838 DOI: 10.1016/j.biomaterials.2019.02.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/01/2019] [Accepted: 02/12/2019] [Indexed: 01/11/2023]
Abstract
Pretargeting is a promising strategy to reach high imaging contrast in a shorter time than by targeting with directly radiolabeled monoclonal antibodies (mAbs). One of problems in pretargeting is a site-specific, reproducible and uniform conjugation of recognition tags to mAbs. To solve this issue we propose a photoconjugation to covalently couple a recognition tag to a mAb via a photoactivatable Z domain. The Z-domain, a 58-amino acid protein derived from the IgG-binding B-domain of Staphylococcus aureus protein A, has a well-characterized binding site in the Fc portion of IgG. We tested the feasibility of this approach using pretargeting based on hybridization between peptide nucleic acids (PNAs). We have used photoconjugation to couple trastuzumab with the PNA-based hybridization probe, HP1. A complementary [57Co]Co-labeled PNA hybridization probe ([57Co]Co-HP2) was used as the secondary targeting probe. In vitro studies demonstrated that trastuzumab-ZHP1 bound specifically to human epidermal growth factor receptor 2 (HER2)-expressing cells with nanomolar affinity. The binding of the secondary [57Co]Co-HP2 probe to trastuzumab-PNA-pretreated cells was in the picomolar affinity range. A two-fold increase in SKOV-3 tumor targeting was achieved when [57Co]Co-HP2 (0.7 nmol) was injected 48 h after injection of trastuzumab-ZHP1 (0.5 nmol) compared with trastuzumab-ZHP1 alone (0.8 ± 0.2 vs. 0.33 ± 0.06 %ID/g). Tumor accumulation of [57Co]Co-HP2 was significantly reduced by pre-saturation with trastuzumab or when no trastuzumab-ZHP1 was preinjected. A tumor-to-blood uptake ratio of 1.5 ± 0.3 was achieved resulting in a clear visualization of HER2-expressing xenografts as confirmed by SPECT imaging. In conclusion, the feasibility of stable site-specific coupling of a PNA-based recognition tag to trastuzumab and successful pretargeting has been demonstrated. This approach can hopefully be used for a broad range of mAbs and recognition tags.
Collapse
|
29
|
Nabil G, Bhise K, Sau S, Atef M, El-Banna HA, Iyer AK. Nano-engineered delivery systems for cancer imaging and therapy: Recent advances, future direction and patent evaluation. Drug Discov Today 2019; 24:462-491. [PMID: 30121330 PMCID: PMC6839688 DOI: 10.1016/j.drudis.2018.08.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/20/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022]
Abstract
Cancer is the second highest cause of death worldwide. Several therapeutic approaches, such as conventional chemotherapy, antibodies and small molecule inhibitors and nanotherapeutics have been employed in battling cancer. Amongst them, nanotheranostics is an example of successful personalized medicine bearing dual role of early diagnosis and therapy to cancer patients. In this review, we have focused on various types of theranostic polymer and metal nanoparticles for their role in cancer therapy and imaging concerning their limitation, future application such as dendritic cell cancer vaccination, gene delivery, T-cell activation and immune modulation. Also, some of the recorded patent applications and clinical trials have been illustrated. The impact of the biological microenvironment on the biodistribution and accumulation of nanoparticles have been discussed.
Collapse
Affiliation(s)
- Ghazal Nabil
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA; Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Ketki Bhise
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Samaresh Sau
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Mohamed Atef
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Hossny A El-Banna
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA; Molecular Imaging Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
30
|
Liu G. A Revisit to the Pretargeting Concept-A Target Conversion. Front Pharmacol 2018; 9:1476. [PMID: 30618765 PMCID: PMC6304396 DOI: 10.3389/fphar.2018.01476] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/03/2018] [Indexed: 01/22/2023] Open
Abstract
Pretargeting is often used as a tumor targeting strategy that provides much higher tumor to non-tumor ratios than direct-targeting using radiolabeled antibody. Due to the multiple injections, pretargeting is investigated less than direct targeting, but the high T/NT ratios have rendered it more useful for therapy. While the progress in using this strategy for tumor therapy has been regularly reviewed in the literature, this review focuses on the nature and quantitative understanding of the pretargeting concept. By doing so, it is the goal of this review to accelerate pretargeting development and translation to the clinic and to prepare the researchers who are not familiar with the pretargeting concept but are interested in applying it. The quantitative understanding is presented in a way understandable to the average researchers in the areas of drug development and clinical translation who have the basic concept of calculus and general chemistry.
Collapse
Affiliation(s)
- Guozheng Liu
- Department of Radiology, University of Massachusetts Medical School Worcester, MA, United States
| |
Collapse
|
31
|
Vorobyeva A, Westerlund K, Mitran B, Altai M, Rinne S, Sörensen J, Orlova A, Tolmachev V, Karlström AE. Development of an optimal imaging strategy for selection of patients for affibody-based PNA-mediated radionuclide therapy. Sci Rep 2018; 8:9643. [PMID: 29942011 PMCID: PMC6018533 DOI: 10.1038/s41598-018-27886-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/08/2018] [Indexed: 01/03/2023] Open
Abstract
Affibody molecules are engineered scaffold proteins, which demonstrated excellent binding to selected tumor-associated molecular abnormalities in vivo and highly sensitive and specific radionuclide imaging of Her2-expressing tumors in clinics. Recently, we have shown that peptide nucleic acid (PNA)-mediated affibody-based pretargeted radionuclide therapy using beta-emitting radionuclide 177Lu extended significantly survival of mice bearing human Her2-expressing tumor xenografts. In this study, we evaluated two approaches to use positron emission tomography (PET) for stratification of patients for affibody-based pretargeting therapy. The primary targeting probe ZHER2:342-SR-HP1 and the secondary probe HP2 (both conjugated with DOTA chelator) were labeled with the positron-emitting radionuclide 68Ga. Biodistribution of both probes was measured in BALB/C nu/nu mice bearing either SKOV-3 xenografts with high Her2 expression or DU-145 xenografts with low Her2 expression. 68Ga-HP2 was evaluated in the pretargeting setting. Tumor uptake of both probes was compared with the uptake of pretargeted 177Lu-HP2. The uptake of both 68Ga-ZHER2:342-SR-HP1 and 68Ga-HP2 depended on Her2-expression level providing clear discrimination of between tumors with high and low Her2 expression. Tumor uptake of 68Ga-HP2 correlated better with the uptake of 177Lu-HP2 than the uptake of 68Ga-ZHER2:342-SR-HP1. The use of 68Ga-HP2 as a theranostics counterpart would be preferable approach for clinical translation.
Collapse
Affiliation(s)
- Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kristina Westerlund
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Bogdan Mitran
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Mohamed Altai
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sara Rinne
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Jens Sörensen
- Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Medical Imaging Centre, Uppsala University Hospital, Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
32
|
Krasniqi A, D'Huyvetter M, Devoogdt N, Frejd FY, Sörensen J, Orlova A, Keyaerts M, Tolmachev V. Same-Day Imaging Using Small Proteins: Clinical Experience and Translational Prospects in Oncology. J Nucl Med 2018; 59:885-891. [PMID: 29545374 DOI: 10.2967/jnumed.117.199901] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/06/2018] [Indexed: 12/12/2022] Open
Abstract
Imaging of expression of therapeutic targets may enable stratification of patients for targeted treatments. The use of small radiolabeled probes based on the heavy-chain variable region of heavy-chain-only immunoglobulins or nonimmunoglobulin scaffolds permits rapid localization of radiotracers in tumors and rapid clearance from normal tissues. This makes high-contrast imaging possible on the day of injection. This mini review focuses on small proteins for radionuclide-based imaging that would allow same-day imaging, with the emphasis on clinical applications and promising preclinical developments within the field of oncology.
Collapse
Affiliation(s)
- Ahmet Krasniqi
- In Vivo Cellular and Molecular Imaging Laboratory (ICMI), VUB, Brussels, Belgium
| | - Matthias D'Huyvetter
- In Vivo Cellular and Molecular Imaging Laboratory (ICMI), VUB, Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory (ICMI), VUB, Brussels, Belgium
| | - Fredrik Y Frejd
- Affibody AB, Solna, Sweden.,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Jens Sörensen
- Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.,Medical Imaging Centre, Uppsala University Hospital, Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden; and
| | - Marleen Keyaerts
- In Vivo Cellular and Molecular Imaging Laboratory (ICMI), VUB, Brussels, Belgium .,Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
33
|
Zhou Z, Chitneni SK, Devoogdt N, Zalutsky MR, Vaidyanathan G. Fluorine-18 labeling of an anti-HER2 VHH using a residualizing prosthetic group via a strain-promoted click reaction: Chemistry and preliminary evaluation. Bioorg Med Chem 2018. [PMID: 29534937 DOI: 10.1016/j.bmc.2018.02.040] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In a previous study, we evaluated a HER2-specific single domain antibody fragment (sdAb) 2Rs15d labeled with 18F via conjugation of a residualizing prosthetic agent that was synthesized by copper-catalyzed azide-alkyne cycloaddition (CuAAC). In order to potentially increase overall efficiency and decrease the time required for labeling, we now investigate the use of a strain-promoted azide-alkyne cycloaddition (SPAAC) between the 2Rs15d sdAb, which had been pre-derivatized with an azide-containing residualizing moiety, and an 18F-labeled aza-dibenzocyclooctyne derivative. The HER2-targeted sdAb 2Rs15d and a nonspecific sdAb R3B23 were pre-conjugated with a moiety containing both azide- and guanidine functionalities. The thus derivatized sdAbs were radiolabeled with 18F using an 18F-labeled aza-dibenzocyclooctyne derivative ([18F]F-ADIBO) via SPAAC, generating the desired conjugate ([18F]RL-II-sdAb). For comparison, unmodified 2Rs15d was labeled with N-succinimidyl 4-guanidinomethyl-3-[125I]iodobenzoate ([125I]SGMIB), the prototypical residualizing agent for radioiodination. Radiochemical purity (RCP), immunoreactive fraction (IRF), HER2-binding affinity and cellular uptake of [18F]RL-II-2Rs15d were assessed in vitro. Paired label biodistribution of [18F]RL-II-2Rs15d and [125I]SGMIB-2Rs15d, and microPET/CT imaging of [18F]RL-II-2Rs15d and the [18F]RL-II-R3B23 control sdAb were performed in nude mice bearing HER2-expressing SKOV-3 xenografts. A radiochemical yield of 23.9 ± 6.9% (n = 8) was achieved for the SPAAC reaction between [18F]F-ADIBO and azide-modified 2Rs15d and the RCP of the labeled sdAb was >95%. The affinity (Kd) and IRF for the binding of [18F]RL-II-2Rs15d to HER2 were 5.6 ± 1.3 nM and 73.1 ± 22.5% (n = 3), respectively. The specific uptake of [18F]RL-II-2Rs15d by HER2-expressing BT474M1 breast carcinoma cells in vitro was 14-17% of the input dose at 1, 2, and 4 h, slightly higher than seen for co-incubated [125I]SGMIB-2Rs15d. The uptake of [18F]RL-II-2Rs15d in SKOV-3 xenografts at 1 h and 2 h p.i. were 5.54 ± 0.77% ID/g and 6.42 ± 1.70% ID/g, respectively, slightly higher than those for co-administered [125I]SGMIB-2Rs15d (4.80 ± 0.78% ID/g and 4.78 ± 1.39% ID/g). MicroPET/CT imaging with [18F]RL-II-2Rs15d at 1-3 h p.i. clearly delineated SKOV-3 tumors while no significant accumulation of activity in tumor was seen for [18F]RL-II-R3B23. With the exception of kidneys, normal tissue levels for [18F]RL-II-2Rs15d were low and cleared rapidly. To our knowledge, this is the first time SPAAC method has been used to label an sdAb with 18F, especially with residualizing functionality.
Collapse
Affiliation(s)
- Zhengyuan Zhou
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Satish K Chitneni
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, (VUB), 1090 Brussels, Belgium
| | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
34
|
Pretargeting in the context of theranostics and companion diagnostics in nuclear oncology. Clin Transl Imaging 2018. [DOI: 10.1007/s40336-018-0271-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
35
|
Westerlund K, Altai M, Mitran B, Konijnenberg M, Oroujeni M, Atterby C, de Jong M, Orlova A, Mattsson J, Micke P, Karlström AE, Tolmachev V. Radionuclide Therapy of HER2-Expressing Human Xenografts Using Affibody-Based Peptide Nucleic Acid-Mediated Pretargeting: In Vivo Proof of Principle. J Nucl Med 2018; 59:1092-1098. [PMID: 29439013 DOI: 10.2967/jnumed.118.208348] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 01/23/2018] [Indexed: 12/25/2022] Open
Abstract
Affibody molecules are small proteins engineered using a nonantibody scaffold. Radiolabeled Affibody molecules are excellent imaging probes, but their application to radionuclide therapy has been prevented by high renal reabsorption. The aim of this study was to test the hypothesis that Affibody-based peptide nucleic acid (PNA)-mediated pretargeted therapy of human epidermal growth factor receptor 2 (HER2)-expressing cancer extends survival without accompanying renal toxicity. Methods: A HER2-targeting Affibody molecule ligated with an AGTCGTGATGTAGTC PNA hybridization probe (ZHER2:342-SR-HP1) was used as the primary pretargeting agent. A complementary AGTCGTGATGTAGTC PNA conjugated to the chelator DOTA and labeled with the radionuclide 177Lu (177Lu-HP2) was used as the secondary agent. The influence of different factors on pretargeting was investigated. Experimental radionuclide therapy in mice bearing SKOV-3 xenografts was performed in 6 cycles separated by 7 d. Results: Optimal tumor targeting was achieved when 16 MBq/3.5 μg (0.65 nmol) of 177Lu-HP2 was injected 16 h after injection of 100 μg (7.7 nmol) of ZHER2:342-SR-HP1. The calculated absorbed dose to tumors was 1,075 mGy/MBq, whereas the absorbed dose to kidneys was 206 mGy/MBq and the absorbed dose to blood (surrogate of bone marrow) was 4 mGy/MBq. Survival of mice was significantly longer (P < 0.05) in the treatment group (66 d) than in the control groups treated with the same amount of ZHER2:342-SR-HP1 only (37 d), the same amount and activity of 177Lu-HP2 only (32 d), or phosphate-buffered saline (37 d). Conclusion: The studied pretargeting system can deliver an absorbed dose to tumors appreciably exceeding absorbed doses to critical organs, making Affibody-based PNA-mediated pretargeted radionuclide therapy highly attractive.
Collapse
Affiliation(s)
- Kristina Westerlund
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mohamed Altai
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Bogdan Mitran
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden; and
| | - Mark Konijnenberg
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Maryam Oroujeni
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Christina Atterby
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Anna Orlova
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden; and
| | - Johanna Mattsson
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | | | - Vladimir Tolmachev
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
36
|
Ji A, Zhang Y, Lv G, Lin J, Qi N, Ji F, Du M. 131
I radiolabeled immune albumin nanospheres loaded with doxorubicin for in vivo combinatorial therapy. J Labelled Comp Radiopharm 2018; 61:362-369. [PMID: 29247459 DOI: 10.1002/jlcr.3593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 01/10/2023]
Affiliation(s)
- Anping Ji
- Nuclear Medicine Department; the Affiliated Hospital of Nanjing University of Traditional Chinese Medicine; Nanjing China
| | - Yuanchao Zhang
- Nuclear Medicine Department; the Affiliated Hospital of Nanjing University of Traditional Chinese Medicine; Nanjing China
| | - Gaochao Lv
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine; Jiangsu Institute of Nuclear Medicine; Wuxi China
| | - Jianguo Lin
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine; Jiangsu Institute of Nuclear Medicine; Wuxi China
| | - Ning Qi
- College of Clinical Medicine; Southeast University; Nanjing China
| | - Faquan Ji
- Nuclear Medicine Department; the Affiliated Hospital of Nanjing University of Traditional Chinese Medicine; Nanjing China
| | - Minghua Du
- Nuclear Medicine Department; the Affiliated Hospital of Nanjing University of Traditional Chinese Medicine; Nanjing China
| |
Collapse
|
37
|
Cai C, Guo Z, Cao Y, Zhang W, Chen Y. A dual biomarker detection platform for quantitating circulating tumor DNA (ctDNA). Nanotheranostics 2018; 2:12-20. [PMID: 29291160 PMCID: PMC5743835 DOI: 10.7150/ntno.22419] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 10/12/2017] [Indexed: 01/11/2023] Open
Abstract
Circulating tumor DNA (ctDNA), which includes DNA mutations, epigenetic alterations and other forms of tumor-specific abnormalities, is a promising “real-time” biomarker for noninvasive cancer assessment. Tumor DNA is of great value in the process of cancer treatment, including diagnostic and prognostic information before, during treatment and at progression. Here we introduce a peptide nucleic acids probe-gold nanoparticles (PNA-AuNPs) and lead phosphate apoferritin (LPA)-based dual biomarker detection platform, which could be used in a DNA biosensor to quantify ctDNA by detection of tumor-specific mutations and methylation of PIK3CA gene. On the one hand, PNA probe and anti-5-Methylcytosine monoclonal antibody (anti-5-mC) were used to recognize the different parts of ctDNA, forming a sandwich-structure on a screen-printed electrode (SPE) surface. On the other hand, AuNPs and LPA were introduced to construct the biosensor for double signal amplification. Square-wave voltammetry (SWV) was used to monitor the electrochemical signal of lead ions released from apoferritin. The proposed DNA biosensor yielded a linear current response to ctDNA concentrations over a broad range of 50-10000 fM with a detection limit of 10 fM. It also successfully detected ctDNA collected from cancer patient serum. Therefore, we anticipate this new platform opens up an approach to detect and monitor diverse malignancies, facilitating personalized cancer therapy.
Collapse
Affiliation(s)
- Chunyan Cai
- Institute for Interdisciplinary Research, Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education, Jianghan University, Wuhan 430056, PR China
| | - Zhenzhong Guo
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan 430065, P.R.China
| | - Yiping Cao
- Institute for Interdisciplinary Research, Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education, Jianghan University, Wuhan 430056, PR China
| | - Weiying Zhang
- Institute for Interdisciplinary Research, Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education, Jianghan University, Wuhan 430056, PR China
| | - Yong Chen
- Institute for Interdisciplinary Research, Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education, Jianghan University, Wuhan 430056, PR China.,Ecole Normale Supérieure, CNRS-ENS-UPMC UMR 8640, 24 Rue Lhomond, Paris 75005, France
| |
Collapse
|
38
|
Bedford R, Tiede C, Hughes R, Curd A, McPherson MJ, Peckham M, Tomlinson DC. Alternative reagents to antibodies in imaging applications. Biophys Rev 2017; 9:299-308. [PMID: 28752365 PMCID: PMC5578921 DOI: 10.1007/s12551-017-0278-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/06/2017] [Indexed: 12/21/2022] Open
Abstract
Antibodies have been indispensable tools in molecular biology, biochemistry and medical research. However, a number of issues surrounding validation, specificity and batch variation of commercially available antibodies have prompted research groups to develop novel non-antibody binding reagents. The ability to select highly specific monoclonal non-antibody binding proteins without the need for animals, the ease of production and the ability to site-directly label has enabled a wide variety of applications to be tested, including imaging. In this review, we discuss the success of a number of non-antibody reagents in imaging applications, including the recently reported Affimer.
Collapse
Affiliation(s)
- R Bedford
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - C Tiede
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - R Hughes
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - A Curd
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - M J McPherson
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - Michelle Peckham
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK.
| | - Darren C Tomlinson
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK.
| |
Collapse
|
39
|
Altai M, Westerlund K, Velletta J, Mitran B, Honarvar H, Karlström AE. Evaluation of affibody molecule-based PNA-mediated radionuclide pretargeting: Development of an optimized conjugation protocol and 177Lu labeling. Nucl Med Biol 2017; 54:1-9. [PMID: 28810153 DOI: 10.1016/j.nucmedbio.2017.07.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/30/2017] [Accepted: 07/04/2017] [Indexed: 12/20/2022]
Abstract
INTRODUCTION We have previously developed a pretargeting approach for affibody-mediated cancer therapy based on PNA-PNA hybridization. In this article we have further developed this approach by optimizing the production of the primary agent, ZHER2:342-SR-HP1, and labeling the secondary agent, HP2, with the therapeutic radionuclide 177Lu. We also studied the biodistribution profile of 177Lu-HP2 in mice, and evaluated pretargeting with 177Lu-HP2 in vitro and in vivo. METHODS The biodistribution profile of 177Lu-HP2 was evaluated in NMRI mice and compared to the previously studied 111In-HP2. Pretargeting using 177Lu-HP2 was studied in vitro using the HER2-expressing cell lines BT-474 and SKOV-3, and in vivo in mice bearing SKOV-3 xenografts. RESULTS AND CONCLUSION Using an optimized production protocol for ZHER2:342-SR-HP1 the ligation time was reduced from 15h to 30min, and the yield increased from 45% to 70%. 177Lu-labeled HP2 binds specifically in vitro to BT474 and SKOV-3 cells pre-treated with ZHER2:342-SR-HP1. 177Lu-HP2 was shown to have a more rapid blood clearance compared to 111In-HP2 in NMRI mice, and the measured radioactivity in blood was 0.22±0.1 and 0.68±0.07%ID/g for 177Lu- and 111In-HP2, respectively, at 1h p.i. In contrast, no significant difference in kidney uptake was observed (4.47±1.17 and 3.94±0.58%ID/g for 177Lu- and 111In-HP2, respectively, at 1h p.i.). Co-injection with either Gelofusine or lysine significantly reduced the kidney uptake for 177Lu-HP2 (1.0±0.1 and 1.6±0.2, respectively, vs. 2.97±0.87%ID/g in controls at 4h p.i.). 177Lu-HP2 accumulated in SKOV-3 xenografts in BALB/C nu/nu mice when administered after injection of ZHER2:342-SR-HP1. Without pre-injection of ZHER2:342-SR-HP1, the uptake of 177Lu-HP2 was about 90-fold lower in tumor (0.23±0.08 vs. 20.7±3.5%ID/g). The tumor-to-kidney radioactivity accumulation ratio was almost 5-fold higher in the group of mice pre-injected with ZHER2:342-SR-HP1. In conclusion, 177Lu-HP2 was shown to be a promising secondary agent for affibody-mediated tumor pretargeting in vivo.
Collapse
Affiliation(s)
- Mohamed Altai
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kristina Westerlund
- Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Justin Velletta
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bogdan Mitran
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Hadis Honarvar
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Amelie Eriksson Karlström
- Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
40
|
Altai M, Membreno R, Cook B, Tolmachev V, Zeglis BM. Pretargeted Imaging and Therapy. J Nucl Med 2017; 58:1553-1559. [PMID: 28687600 DOI: 10.2967/jnumed.117.189944] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 06/27/2017] [Indexed: 12/22/2022] Open
Abstract
In vivo pretargeting stands as a promising approach to harnessing the exquisite tumor-targeting properties of antibodies for nuclear imaging and therapy while simultaneously skirting their pharmacokinetic limitations. The core premise of pretargeting lies in administering the targeting vector and radioisotope separately and having the 2 components combine within the body. In this manner, pretargeting strategies decrease the circulation time of the radioactivity, reduce the uptake of the radionuclide in healthy nontarget tissues, and facilitate the use of short-lived radionuclides that would otherwise be incompatible with antibody-based vectors. In this short review, we seek to provide a brief yet informative survey of the 4 preeminent mechanistic approaches to pretargeting, strategies predicated on streptavidin and biotin, bispecific antibodies, complementary oligonucleotides, and bioorthogonal click chemistry.
Collapse
Affiliation(s)
- Mohamed Altai
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Rosemery Membreno
- Department of Chemistry, Hunter College of the City University of New York, New York, New York.,PhD Program in Chemistry, Graduate Center of the City University of New York, New York, New York; and.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brendon Cook
- Department of Chemistry, Hunter College of the City University of New York, New York, New York.,PhD Program in Chemistry, Graduate Center of the City University of New York, New York, New York; and.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vladimir Tolmachev
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Brian M Zeglis
- Department of Chemistry, Hunter College of the City University of New York, New York, New York .,PhD Program in Chemistry, Graduate Center of the City University of New York, New York, New York; and.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
41
|
Bailly C, Bodet-Milin C, Rousseau C, Faivre-Chauvet A, Kraeber-Bodéré F, Barbet J. Pretargeting for imaging and therapy in oncological nuclear medicine. EJNMMI Radiopharm Chem 2017; 2:6. [PMID: 29503847 PMCID: PMC5824696 DOI: 10.1186/s41181-017-0026-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/24/2017] [Indexed: 12/27/2022] Open
Abstract
Background Oncological pretargeting has been implemented and tested in several different ways in preclinical models and clinical trials over more than 30 years. Despite highly promising results, pretargeting has not achieved market approval even though it could be considered the ultimate theranostic, combining PET imaging with short-lived positron emitters and therapy with radionuclides emitting beta or alpha particles. Results We have reviewed the pretargeting approaches proposed over the years, discussing their suitability for imaging, particularly PET imaging, and therapy, as well as their limitations. The reviewed pretargeting modalities are the avidin-biotin system, bispecific anti-tumour x anti-hapten antibodies and bivalent haptens, antibody-oligonucleotide conjugates and radiolabelled complementary oligonucleotides, and approaches using click chemistry. Finally, we discuss recent developments, such as the use of small binding proteins for pretargeting that may offer new perspectives to cancer pretargeting. Conclusions While pretargeting has shown promise and demonstrated preclinical and clinical proof of principle, full-scale clinical development programs are needed to translate pretargeting into a clinical reality that could ideally fit into current theranostic and precision medicine perspectives.
Collapse
Affiliation(s)
- Clément Bailly
- 1Service de Médecine Nucléaire, CHU de Nantes, Nantes, France.,3Centre de Recherche en Cancérologie Nantes/Angers (CRCNA), Nantes, France.,6299 CNRS, Nantes, France.,UMR892 Inserm, Nantes, France.,6Université de Nantes, Nantes, France
| | - Caroline Bodet-Milin
- 1Service de Médecine Nucléaire, CHU de Nantes, Nantes, France.,3Centre de Recherche en Cancérologie Nantes/Angers (CRCNA), Nantes, France.,6299 CNRS, Nantes, France.,UMR892 Inserm, Nantes, France.,6Université de Nantes, Nantes, France
| | - Caroline Rousseau
- 2Service de Médecine Nucléaire, Institut de Cancérologie de l'Ouest, Saint-Herblain, France.,3Centre de Recherche en Cancérologie Nantes/Angers (CRCNA), Nantes, France.,6299 CNRS, Nantes, France.,UMR892 Inserm, Nantes, France.,6Université de Nantes, Nantes, France
| | - Alain Faivre-Chauvet
- 1Service de Médecine Nucléaire, CHU de Nantes, Nantes, France.,3Centre de Recherche en Cancérologie Nantes/Angers (CRCNA), Nantes, France.,6299 CNRS, Nantes, France.,UMR892 Inserm, Nantes, France.,6Université de Nantes, Nantes, France
| | - Françoise Kraeber-Bodéré
- 1Service de Médecine Nucléaire, CHU de Nantes, Nantes, France.,2Service de Médecine Nucléaire, Institut de Cancérologie de l'Ouest, Saint-Herblain, France.,3Centre de Recherche en Cancérologie Nantes/Angers (CRCNA), Nantes, France.,6299 CNRS, Nantes, France.,UMR892 Inserm, Nantes, France.,6Université de Nantes, Nantes, France
| | - Jacques Barbet
- 6Université de Nantes, Nantes, France.,GIP Arronax, 1, rue Arronax, 44187 Saint-Herblain cedex, France
| |
Collapse
|
42
|
Ståhl S, Gräslund T, Eriksson Karlström A, Frejd FY, Nygren PÅ, Löfblom J. Affibody Molecules in Biotechnological and Medical Applications. Trends Biotechnol 2017; 35:691-712. [PMID: 28514998 DOI: 10.1016/j.tibtech.2017.04.007] [Citation(s) in RCA: 243] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 01/08/2023]
Abstract
Affibody molecules are small (6.5-kDa) affinity proteins based on a three-helix bundle domain framework. Since their introduction 20 years ago as an alternative to antibodies for biotechnological applications, the first therapeutic affibody molecules have now entered clinical development and more than 400 studies have been published in which affibody molecules have been developed and used in a variety of contexts. In this review, we focus primarily on efforts over the past 5 years to explore the potential of affibody molecules for medical applications in oncology, neurodegenerative, and inflammation disorders, including molecular imaging, receptor signal blocking, and delivery of toxic payloads. In addition, we describe recent examples of biotechnological applications, in which affibody molecules have been exploited as modular affinity fusion partners.
Collapse
Affiliation(s)
- Stefan Ståhl
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden.
| | - Torbjörn Gräslund
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | | | - Fredrik Y Frejd
- Unit of Biomedical Radiation Sciences, Uppsala University, SE-751 85 Uppsala, Sweden; Affibody AB, Gunnar Asplunds Allé 24, SE-171 69 Solna, Sweden
| | - Per-Åke Nygren
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | - John Löfblom
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| |
Collapse
|
43
|
Xu Y, Bai Z, Huang Q, Pan Y, Pan D, Wang L, Yan J, Wang X, Yang R, Yang M. PET of HER2 Expression with a Novel 18FAl Labeled Affibody. J Cancer 2017; 8:1170-1178. [PMID: 28607591 PMCID: PMC5463431 DOI: 10.7150/jca.18070] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/24/2017] [Indexed: 12/27/2022] Open
Abstract
Background: Human epidermal growth factor receptor type 2 (HER2) is abundant in a wide variety of tumors and associated with the poor prognosis. Radiolabeled affibodies are potential candidates for detecting HER2-positive lesions. However, laborious multiple-step synthetic procedure and high abdomen background may hinder the widespread use. Herein, cysteinylated ZHER2:342 modified with a new hydrophilic linker (denoted as MZHER2:342) was designed and labeled using 18FAl-NOTA strategies. The biologic efficacy of the novel tracer and its feasibilities for in vivo monitoring HER2 levels were also investigated in xenograft models with different HER2 expressions. Method: MZHER2:342 was conjugated with MAL-NOTA under standard reaction conditions. The affibody molecule was then radiolabeled with 18FAl complex. The binding specificity of the tracer, 18FAl-NOTA-MAL-MZHER2:342, with HER2 was primarily characterized via in vitro studies. MicroPET imaging were performed in nude mice bearing tumors (SKOV-3, JIMT-1 and MCF-7) after injection. The HER2 levels of xenografts were determined using Western blotting analysis. Results:18FAl-NOTA-MAL-MZHER2:342 can be efficiently produced within 30 min with a non-decaycorrected yield of about 10% and a radiochemical purity of more than 95%. In vitro experiments revealed that the modified affibody retained the specific affinity to HER2. PET imaging showed that SKOV-3 and JIMT-1 xenografts were clearly visualized with excellent contrast and low abdomen backgrounds. On the contrary, the signals of MCF-7 tumor were difficult to visualize. The ROI values ranged from16.54±2.69% ID/g for SKOV-3 to 8.42±1.20 %ID/g for JIMT-1 tumors at 1h postinjection respectively. Poor uptake was observed from MCF-7 tumors with 1.71±0.34% ID/g at the same time point. Besides, a significant linear correlation between % ID/g values and relative HER2 expression levels was also found. Conclusions:18FAl-NOTA-MAL-MZHER2:342 is a promising tracer for in vivo detecting HER2 status with the advantages of facile synthesis and favorable pharmacokinetics. It may be useful in differential diagnosis, molecularly targeted therapy and prognosis of the cancers.
Collapse
Affiliation(s)
- Yuping Xu
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.,Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Zhicheng Bai
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Qianhuan Huang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Yunyun Pan
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Donghui Pan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Lizhen Wang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Junjie Yan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Xinyu Wang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Runlin Yang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Min Yang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.,Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| |
Collapse
|
44
|
Abstract
Affibody molecules can be used as tools for molecular recognition in diagnostic and therapeutic applications. There are several preclinical studies reported on diagnostic and therapeutic use of this molecular class of alternative scaffolds, and early clinical evidence is now beginning to accumulate that suggests the Affibody molecules to be efficacious and safe in man. The small size and ease of engineering make Affibody molecules suitable for use in multispecific constructs where AffiMabs is one such that offers the option to potentiate antibodies for use in complex disease.
Collapse
|
45
|
A Conjugate Based on Anti-HER2 Diaffibody and Auristatin E Targets HER2-Positive Cancer Cells. Int J Mol Sci 2017; 18:ijms18020401. [PMID: 28216573 PMCID: PMC5343935 DOI: 10.3390/ijms18020401] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/20/2017] [Accepted: 01/31/2017] [Indexed: 01/29/2023] Open
Abstract
Antibody-drug conjugates (ADCs) have recently emerged as efficient and selective cancer treatment therapeutics. Currently, alternative forms of drug carriers that can replace monoclonal antibodies are under intensive investigation. Here, a cytotoxic conjugate of an anti-HER2 (Human Epidermal Growth Factor Receptor 2) diaffibody with monomethyl-auristatin E (MMAE) is proposed as a potential anticancer therapeutic. The anti-HER2 diaffibody was based on the ZHER2:4 affibody amino acid sequence. The anti-HER2 diaffibody has been expressed as a His-tagged protein in E. coli and purified by Ni-nitrilotriacetyl (Ni-NTA) agarose chromatography. The molecule was properly folded, and the high affinity and specificity of its interaction with HER2 was confirmed by surface plasmon resonance (SPR) and flow cytometry, respectively. The (ZHER2:4)2DCS-MMAE conjugate was obtained by coupling the maleimide group linked with MMAE to cysteines, which were introduced in a drug conjugation sequence (DCS). Cytotoxicity of the conjugate was evaluated using the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide MTT assay and the xCELLigence Real-Time Cell Analyzer. Our experiments demonstrated that the conjugate delivered auristatin E specifically to HER2-positive tumor cells, which finally led to their death. These results indicate that the cytotoxic diaffibody conjugate is a highly potent molecule for the treatment of various types of cancer overexpressing HER2 receptors.
Collapse
|
46
|
Abstract
Differing from the conventional direct-targeting strategy in which a probe or payload is directly loaded onto a targeting molecule that binds to the native target, pretargeting is an improved targeting strategy. It converts the native target to an artificial target specific for a secondary targeting molecule loaded with the probe or payload (effector). The effector is small and does not accumulate in normal tissues, which accelerates the targeting process and generates high target to nontarget ratios. DNA/cDNA analogs can serve as the recognition pair, i.e., the artificial target and the secondary targeting effector. Morpholino oligomers are so far the most investigated and the most successful DNA/cDNA analog recognition pairs for pretargeting. Herein, we describe the pretargeting principles, the pretargeting strategy using Morpholino oligomers, and the preclinical success so far achieved.
Collapse
Affiliation(s)
- Guozheng Liu
- Department of Radiology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA.
| |
Collapse
|
47
|
Meyer JP, Adumeau P, Lewis JS, Zeglis BM. Click Chemistry and Radiochemistry: The First 10 Years. Bioconjug Chem 2016; 27:2791-2807. [PMID: 27787983 DOI: 10.1021/acs.bioconjchem.6b00561] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The advent of click chemistry has had a profound influence on almost all branches of chemical science. This is particularly true of radiochemistry and the synthesis of agents for positron emission tomography (PET), single photon emission computed tomography (SPECT), and targeted radiotherapy. The selectivity, ease, rapidity, and modularity of click ligations make them nearly ideally suited for the construction of radiotracers, a process that often involves working with biomolecules in aqueous conditions with inexorably decaying radioisotopes. In the following pages, our goal is to provide a broad overview of the first 10 years of research at the intersection of click chemistry and radiochemistry. The discussion will focus on four areas that we believe underscore the critical advantages provided by click chemistry: (i) the use of prosthetic groups for radiolabeling reactions, (ii) the creation of coordination scaffolds for radiometals, (iii) the site-specific radiolabeling of proteins and peptides, and (iv) the development of strategies for in vivo pretargeting. Particular emphasis will be placed on the four most prevalent click reactions-the Cu-catalyzed azide-alkyne cycloaddition (CuAAC), the strain-promoted azide-alkyne cycloaddition (SPAAC), the inverse electron demand Diels-Alder reaction (IEDDA), and the Staudinger ligation-although less well-known click ligations will be discussed as well. Ultimately, it is our hope that this review will not only serve to educate readers but will also act as a springboard, inspiring synthetic chemists and radiochemists alike to harness click chemistry in even more innovative and ambitious ways as we embark upon the second decade of this fruitful collaboration.
Collapse
Affiliation(s)
| | - Pierre Adumeau
- Department of Chemistry, Hunter College of the City University of New York , 413 East 69th Street, New York, New York 10028, United States
| | - Jason S Lewis
- Department of Radiology, Weill Cornell Medical College , 520 East 70th Street, New York, New York 10065, United States
| | - Brian M Zeglis
- Department of Chemistry, Hunter College of the City University of New York , 413 East 69th Street, New York, New York 10028, United States.,Department of Radiology, Weill Cornell Medical College , 520 East 70th Street, New York, New York 10065, United States.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York , 365 5th Avenue, New York, New York 10016, United States
| |
Collapse
|
48
|
Leenheer D, ten Dijke P, Hipolito CJ. A current perspective on applications of macrocyclic-peptide-based high-affinity ligands. Biopolymers 2016; 106:889-900. [PMID: 27352774 PMCID: PMC5132055 DOI: 10.1002/bip.22900] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 05/24/2016] [Accepted: 05/31/2016] [Indexed: 01/05/2023]
Abstract
Monoclonal antibodies can bind with high affinity and high selectivity to their targets. As a tool in therapeutics or diagnostics, however, their large size (∼150 kDa) reduces penetration into tissue and prevents passive cellular uptake. To overcome these and other problems, minimized protein scaffolds have been chosen or engineered, with care taken to not compromise binding affinity or specificity. An alternate approach is to begin with a minimal non-antibody scaffold and select functional ligands from a de novo library. We will discuss the structure, production, applications, strengths, and weaknesses of several classes of antibody-derived ligands, that is, antibodies, intrabodies, and nanobodies, and nonantibody-derived ligands, that is, monobodies, affibodies, and macrocyclic peptides. In particular, this review is focussed on macrocyclic peptides produced by the Random non-standard Peptides Integrated Discovery (RaPID) system that are small in size (typically ∼2 kDa), but are able to perform tasks typically handled by larger proteinaceous ligands.
Collapse
Affiliation(s)
- Daniël Leenheer
- Ph.D. Program in Human Biology, School of Integrative and Global MajorsUniversity of TsukubaTsukubaIbarakiJapan
| | - Peter ten Dijke
- Leiden University Medical Center, Department of Molecular Cell BiologyLeidenSouth HollandThe Netherlands
- Cancer Signaling, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of TsukubaTsukubaIbarakiJapan
| | - Christopher John Hipolito
- Cancer Signaling, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of TsukubaTsukubaIbarakiJapan
| |
Collapse
|
49
|
Dong D, Xia G, Li Z, Li Z. Human Serum Albumin and HER2-Binding Affibody Fusion Proteins for Targeted Delivery of Fatty Acid-Modified Molecules and Therapy. Mol Pharm 2016; 13:3370-3380. [DOI: 10.1021/acs.molpharmaceut.6b00265] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
| | - Guanjun Xia
- Department
of Pathology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | | | | |
Collapse
|
50
|
Westerlund K, Honarvar H, Tolmachev V, Eriksson Karlström A. Design, Preparation, and Characterization of PNA-Based Hybridization Probes for Affibody-Molecule-Mediated Pretargeting. Bioconjug Chem 2015; 26:1724-36. [PMID: 26086597 DOI: 10.1021/acs.bioconjchem.5b00292] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In radioimmunotherapy, the contrast between tumor and normal tissue can be improved by using a pretargeting strategy with a primary targeting agent, which is conjugated to a recognition tag, and a secondary radiolabeled molecule binding specifically to the recognition tag. The secondary molecule is injected after the targeting agent has accumulated in the tumor and is designed to have a favorable biodistribution profile, with fast clearance from blood and low uptake in normal tissues. In this study, we have designed and evaluated two complementary peptide nucleic acid (PNA)-based probes for specific and high-affinity association in vivo. An anti-HER2 Affibody-PNA chimera, Z(HER2:342)-SR-HP1, was produced by a semisynthetic approach using sortase A catalyzed ligation of a recombinantly produced Affibody molecule to a PNA-based HP1-probe assembled using solid-phase chemistry. A complementary HP2 probe carrying a DOTA chelator and a tyrosine for dual radiolabeling was prepared by solid-phase synthesis. Circular dichroism (CD) spectroscopy and UV thermal melts showed that the probes can hybridize to form a structured duplex with a very high melting temperature (T(m)), both in HP1:HP2 and in Z(HER2:342)-SR-HP1:HP2 (T(m) = 86-88 °C), and the high binding affinity between Z(HER2:342)-SR-HP1 and HP2 was confirmed in a surface plasmon resonance (SPR)-based binding study. Following a moderately fast association (1.7 × 10(5) M(-1) s(-1)), the dissociation of the probes was extremely slow and <5% dissociation was observed after 17 h. The equilibrium dissociation constant (K(D)) for Z(HER2:342)-SR-HP1:HP2 binding to HER2 was estimated by SPR to be 212 pM, suggesting that the conjugation to PNA does not impair Affibody binding to HER2. The biodistribution profiles of (111)In- and (125)I-labeled HP2 were measured in NMRI mice, showing very fast blood clearance rates and low accumulation of radioactivity in kidneys and other organs. The measured radioactivity in blood was 0.63 ± 0.15 and 0.41 ± 0.15%ID/g for (125)I- and (111)In-HP2, respectively, at 1 h p.i., and at 4 h p.i., the kidney accumulation of radioactivity was 0.17 ± 0.04%ID/g for (125)I-HP2 and 3.83 ± 0.39%ID/g for (111)In-HP2. Taken together, the results suggest that a PNA-based system has suitable biophysical and in vivo properties and is a promising approach for pretargeting of Affibody molecules.
Collapse
Affiliation(s)
- Kristina Westerlund
- †School of Biotechnology, Division of Protein Technology, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Hadis Honarvar
- ‡Department of Immunology, Genetics and Pathology, Uppsala University, 751 05 Uppsala, Sweden
| | - Vladimir Tolmachev
- ‡Department of Immunology, Genetics and Pathology, Uppsala University, 751 05 Uppsala, Sweden
| | - Amelie Eriksson Karlström
- †School of Biotechnology, Division of Protein Technology, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| |
Collapse
|