1
|
Cong Y, Biemans R, Lieuwes NG, Suijlen D, Lambin P, Dijkgraaf I, Bauwens M, Yaromina A, Dubois LJ. Development of a novel anti-CEACAM5 VHH for SPECT imaging and potential cancer therapy applications. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07321-z. [PMID: 40358697 DOI: 10.1007/s00259-025-07321-z] [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: 01/21/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025]
Abstract
PURPOSE In this study, we investigated the utility of a novel developed anti-CEACAM5 VHH for cancer diagnosis and its potential of being a targeting-moiety of VHH-drug conjugates for cancer therapy. METHODS Anti-CEACAM5 VHH (6B11) affinity and specific cellular binding was confirmed by ELISA, FACS and immunofluorescence in cancer cell lines with varying CEACAM5 expression levels. Intracellular penetration ability within tumor spheroids was tested with Oregon Green 488 labeled 6B11 (OG488-6B11). Biodistribution and binding specificity of 99mTc-radiolabeled 6B11 was tested in A549 CEACAM5 overexpressing (A549-CEA5-OV) and knockout (A549-CEA5-KO) tumor-bearing mice upon SPECT/CT imaging, γ-counting and autoradiography. The therapeutic efficacy of 6B11 and 6F8 (anti-CEACAM5 VHH with lower binding affinity) was tested by viability, wound healing and adhesion assays. To verify the potential of VHHs as a warhead for VHH-drug conjugation, an internalization assay with OG488 labeled VHH was performed. RESULT 6B11 demonstrated high binding affinity (EC50 0.5nM) and cellular binding. OG488-6B11 penetrated tumor spheroids completely at 24 h, while a conventional antibody was only visible at the spheroid periphery. SPECT imaging indicated higher uptake (p < 0.05) in A549-CEA5-OV tumors, resulting in increased tumor-to-blood ratios especially at 4 (2.0016 ± 1.1893, p = 0.035) and 24 (2.9371 ± 2.0683, p = 0.003) hpi compared to A549-CEA5-KO tumors at 4 (0.5640 ± 0.3576) and 24 (0.8051 ± 0.4351) hpi. 99mTc-6B11 was predominantly renally cleared. Autoradiography and immunohistochemistry confirmed these uptake patterns. 6B11 nor 6F8 did exhibit significant anti-cancer therapeutic efficacy in vitro. OG488-6B11 was effectively internalized and accumulated in cells in a time-dependent manner, to end up in the lysosomes. CONCLUSION The anti-CEACAM5 VHH 6B11 is a good candidate for SPECT-based cancer diagnosis and can be potentially used as targeting moiety in the development of VHH-based drug conjugates for cancer treatments.
Collapse
Affiliation(s)
- Ying Cong
- The M-Lab, Department of Precision Medicine, GROW- Research Institute for Oncology and Reproduction, Maastricht University, UNS50/23, Maastricht, PO Box 616, 6229 ER, Maastricht, The Netherlands
| | - Rianne Biemans
- The M-Lab, Department of Precision Medicine, GROW- Research Institute for Oncology and Reproduction, Maastricht University, UNS50/23, Maastricht, PO Box 616, 6229 ER, Maastricht, The Netherlands
| | - Natasja G Lieuwes
- The M-Lab, Department of Precision Medicine, GROW- Research Institute for Oncology and Reproduction, Maastricht University, UNS50/23, Maastricht, PO Box 616, 6229 ER, Maastricht, The Netherlands
| | - Dennis Suijlen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW- Research Institute for Oncology and Reproduction, Maastricht University, UNS50/23, Maastricht, PO Box 616, 6229 ER, Maastricht, The Netherlands
| | - Ingrid Dijkgraaf
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Matthias Bauwens
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW- Research Institute for Oncology and Reproduction, Maastricht University, UNS50/23, Maastricht, PO Box 616, 6229 ER, Maastricht, The Netherlands
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW- Research Institute for Oncology and Reproduction, Maastricht University, UNS50/23, Maastricht, PO Box 616, 6229 ER, Maastricht, The Netherlands.
| |
Collapse
|
2
|
Hou J, Du K, Li J, Li Z, Cao S, Zhang S, Huang W, Liu H, Yang X, Sun S, Mo S, Qin T, Zhang X, Yin S, Nie X, Lu X. Research trends in the use of nanobodies for cancer therapy. J Control Release 2025; 381:113454. [PMID: 39922288 DOI: 10.1016/j.jconrel.2025.01.045] [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: 08/15/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 02/10/2025]
Abstract
Although there are many challenges in using nanobodies for treating various complex tumor diseases, including rapid renal clearance and the complex blood-brain barrier environment, nanobodies have shown great potential due to their high antigen affinity, excellent tumor penetration ability, and favorable safety profile. Since the discovery of the variable domain (VHH) of camelid heavy-chain antibodies in 1993, nanobodies have been progressively applied to various cancer therapy platforms, such as antagonistic drugs and targeting agents for effector domains. In recent years, several nanobody-based drugs, including Caplacizumab, KN-035, and Ozoralizumab, have been approved for clinical use. Among them, KN-035 is used for treating advanced solid tumors, and these advancements have propelled nanobody development to new heights. Currently, nanobodies are being rapidly applied to the treatment of a wide range of diseases, from viral infections to cancer, demonstrating strong advantages in areas such as targeted protein degradation, bioimaging, nanobody-drug conjugation, bispecific T-cell engagers, and vaccine applications. Bibliometric tools, including CiteSpace, HisCite Pro, and Alluvial Generator, were employed to trace the historical development of nanobodies in cancer research. The contributions of authors, countries, and institutions in this field were analyzed, and research hotspots and emerging trends were identified through keyword analysis and influential articles. Future trends were also predicted. This study provides a unique, comprehensive, and objective perspective on the use of nanobodies in tumor research, laying a foundation for future research directions and offering valuable insights for researchers in the field.
Collapse
Affiliation(s)
- Jun Hou
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Kejiang Du
- Department of Otorhinolaryngology-Head and Neck Surgery, Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou 545006, China
| | - Jinling Li
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Zhenghui Li
- Department of Neurosurgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Shaorui Cao
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Shilin Zhang
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Wenxing Huang
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Heng Liu
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Xiaomei Yang
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Shuyang Sun
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Shanzhao Mo
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Tianyu Qin
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Xilei Zhang
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Shihua Yin
- Department of Otorhinolaryngology-Head and Neck Surgery, Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China.
| | - Xinyu Nie
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230002, China.
| | - Xiaoling Lu
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
3
|
Wang J, Tolmachova KA, Bode JW. Chemoenzymatic Site-Specific Lysine Modification of Nanobodies and Subsequent Bioconjugation via Potassium Acyltrifluoroborate (KAT) Ligations. J Am Chem Soc 2025; 147:15389-15396. [PMID: 40266292 DOI: 10.1021/jacs.5c01418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Single chain camelid antigen binding domains, often called nanobodies, have emerged as powerful tools for diagnostics and therapy. Methods for their site-specific modification offer immense potential for enhancing their therapeutic applications, but established approaches, such as fusion proteins, have well-known limitations in the nanobody format. Here, we report a convenient and broadly applicable method for site-specifically functionalizing a single residue near the C-terminus of VHH nanobodies by employing lysine acylation using conjugating enzymes (LACE) to transfer short peptides bearing functional group handles for potassium acyltrifluoroborate (KAT) ligations onto a single lysine residue of the expressed nanobodies. This approach requires a LACE tag (4 residues or 11 residues) in the recombinant nanobodies and enables direct elaboration of the products via a rapid amide forming reaction. In this study, VHH nanobodies expressed in Escherichia coli could be efficiently modified through the transfer of specific chemical handles, enabling their conjugation to small molecules, nanobodies, and antibodies by chemoselective, amide-forming KAT ligations that operate at micromolar concentrations.
Collapse
Affiliation(s)
- Jinling Wang
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich 8093, Switzerland
| | - Kateryna A Tolmachova
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich 8093, Switzerland
| | - Jeffrey W Bode
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich 8093, Switzerland
| |
Collapse
|
4
|
Spahn MA, Anbuhl SM, Luyten K, Loy TV, Pronker MF, Cawthorne C, Deroose CM, Schols D, Heukers R, Bormans G, Cleeren F. Indium-111-Labeled Single-Domain Antibody for In Vivo CXCR4 Imaging Using Single-Photon Emission Computed Tomography. Bioconjug Chem 2025; 36:737-747. [PMID: 40067691 DOI: 10.1021/acs.bioconjchem.5c00024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
C-X-C chemokine receptor type 4 (CXCR4) is highly expressed in a range of pathologies, including cancers like multiple myeloma and non-Hodgkin lymphoma, inflammatory diseases such as rheumatoid arthritis, and viral infections like HIV. Currently, the most advanced radiotracer for CXCR4 imaging in clinics is [68Ga]PentixaFor. However, its structure is prone to modifications, complicating the development of a specific CXCR4 fluorine-18-labeled tracer with good pharmacokinetic properties. This study aimed to screen multiple CXCR4-targeting variable domains of heavy-chain-only antibody (VHH or single-domain antibody (sdAb)) constructs to identify the most promising sdAb as a vector molecule for the future development of a CXCR4 fluorine-18 tracer. We have generated five CXCR4-specific sdAb constructs with a cysteine-containing C-terminal tag (C-Direct tag) (VUN400-C-Direct, VUN401-C-Direct, VUN410-C-Direct, VUN411-C-Direct, and VUN415-C-Direct) and one probe (VUN400-C) without. The reduced sdAbs were coupled to maleimide-DOTAGA for 111In-labeling. Their binding affinity against human CXCR4 (hCXCR4) was assessed by using a previously described BRET-based displacement assay. The in vivo profile was assessed using naive mice. Based on the plasma stability (60 min post injection (p.i.)), we selected VUN400-C-Direct and its derivative VUN400-C for further evaluation. These compounds ([111In]In-DOTAGA-VUN400-C-Direct and [111In]In-DOTAGA-VUN400-C) were tested in mice bearing xenografts derived from U87.CD4, U87.CXCR4, and U87.CD4.CXCR4 cells through ex vivo biodistribution studies and SPECT/CT imaging. The six sdAb constructs were labeled with a high radiochemical conversion (75-97%) and purity (>95%). In radioactive binding assays using U87.CD4.CXCR4 cells, [111In]In-DOTAGA-VUN400-C-Direct and [111In]In-DOTAGA-VUN401-C-Direct displayed the highest cellular uptake, achieving 10.4 ± 1.6% and 11.5 ± 1.1%, respectively. In naive mice, [111In]In-DOTAGA-VUN400-C-Direct showed the most favorable biodistribution profile, with low uptake across all organs except the kidneys (Standardized Uptake Value (SUV) > 50, n = 3, 60 min p.i.), but average plasma stability (40.6 ± 9.4%, n = 3, 60 min p.i.). In a xenografted tumor model, [111In]In-DOTAGA-VUN400-C-Direct showed only minor uptake (SUVU87.CXCR4 0.71 ± 0.002, n = 3, 60 min p.i.). [111In]In-DOTAGA-VUN400-C demonstrated nearly identical plasma stability (41.08 ± 5.45%, n = 4) but showed high and specific uptake in the CXCR4-expressing xenografted tumor (SUVU87.CD4.CXCR4 3.75 ± 1.08 vs SUVU87.CD4 = 0.64 ± 0.19, n = 5, 60 min p.i.), which could be blocked by coinjection of AMD3100 (5 mg/kg) (SUVU87.CD4.CXCR4 0.55 ± 0.32 vs SUVU87.CD4 = 0.39 ± 0.07, n = 2, 60 min p.i.). In conclusion, all six sdAbs exhibited high in vitro affinity against hCXCR4. Among these, [111In]In-DOTAGA-VUN400-C showed high CXCR4-specific tumor uptake and favorable pharmacokinetic properties, indicating VUN400-C's potential as a promising vector for future CXCR4 PET imaging applications with fluorine-18.
Collapse
Affiliation(s)
- Muriel Aline Spahn
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven 3000, Belgium
| | - Stephanie Mareike Anbuhl
- QVQ Holding B.V., Yalelaan 1, Utrecht 3584 CL, The Netherlands
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam 1081 HV, The Netherlands
| | - Kaat Luyten
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven 3000, Belgium
| | - Tom Van Loy
- KU Leuven, Department of Microbiology, Immunology and Transplantation Rega Institute for Medical Research,Molecular Structural and Translational Virology Research Group, Leuven B-3000, Belgium
| | - Matti F Pronker
- QVQ Holding B.V., Yalelaan 1, Utrecht 3584 CL, The Netherlands
| | - Christopher Cawthorne
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven 3000, Belgium
| | - Christophe M Deroose
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven 3000, Belgium
| | - Dominique Schols
- KU Leuven, Department of Microbiology, Immunology and Transplantation Rega Institute for Medical Research,Molecular Structural and Translational Virology Research Group, Leuven B-3000, Belgium
| | - Raimond Heukers
- QVQ Holding B.V., Yalelaan 1, Utrecht 3584 CL, The Netherlands
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam 1081 HV, The Netherlands
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven 3000, Belgium
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
5
|
Wang Y, Zheng M, Zhao J, Wang C, Zhao S, Bian Y, Dai N, Zheng Y, Sang S, Guo L, Huang C, Zhang H, Jiang J, Xu C, Zhao Q, Han J, Xu T, Qin S, Miao L. Human dose-escalation study of PET imaging CD8 + T-cell infiltration in solid malignancies with [ 68Ga]Ga -NODAGA-SNA006. Eur J Nucl Med Mol Imaging 2025; 52:1332-1344. [PMID: 39653817 DOI: 10.1007/s00259-024-06999-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 11/19/2024] [Indexed: 02/20/2025]
Abstract
PURPOSE A noninvasive method for evaluating the infiltration of CD8+ T cells in tumors is urgently needed to monitor the response to immunotherapy. This study investigated the performance of a [68Ga]Ga-NODAGA-SNA006 in positron emission tomography (PET) imaging of CD8+ T cells in patients with solid malignancies. METHODS This human dose-escalation PET imaging study involved eleven patients (lung cancer, 8; gastric carcinoma, 1; esophageal carcinoma, 2). Approximately 150 MBq of [68Ga]Ga-NODAGA-SNA006 with varying nanobody masses (100 µg, 300 µg, 500 µg, 800 µg) was administered, and PET/computed tomography (CT) scans were performed at 15-30, 60-90 and 120 min postinjection (p.i.). Data regarding biodistribution, pharmacokinetics and radiation dosimetry were evaluated. CD8+ T-cell infiltration in biopsy samples was also measured via immunohistochemistry (IHC) for correlation analysis with the tumor uptake of [68Ga]Ga-NODAGA-SNA006 PET. RESULTS [68Ga]Ga-NODAGA-SNA006 was well tolerated by all eleven subjects. The highest radioactive uptake was observed in the spleen, followed by the kidneys and bladder. Liver uptake decreased with increasing nanobody mass. Rapid clearance (t1/2<30 min) of [68Ga]Ga-NODAGA-SNA006 from whole blood and serum was observed. Furthermore, 68Ga uptake in tumors (SUVmean) exhibited a linear relationship with CD8+ T-cell infiltration in biopsy samples (R2 = 0.757, p = 0.011), suggesting that the tumor uptake of [68Ga]Ga-NODAGA-SNA006 may represent the degree of CD8+ T-cell infiltration in the tumor. CONCLUSION The use of [68Ga]Ga-NODAGA-SNA006 is safe, feasible, and well tolerated. [68Ga]Ga-NONAGA-SNA006 PET imaging can accurately detect CD8 expression inside tumors with favorable pharmacokinetics, thus providing a feasible method for noninvasive quantitative assessment of CD8+ T-cell tumor infiltration and monitoring the response to immunotherapy. TRIAL REGISTRATION NCT05126927 (19 November 2021, retrospectively registered).
Collapse
Affiliation(s)
- Yan Wang
- Department of Clinical Pharmacology, First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Meng Zheng
- Department of Clinical Pharmacology, First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jun Zhao
- Department of Thoracic Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chao Wang
- Smart-Nuclide Biotech, No. 218 Xing-Hu Rd, Suzhou, Jiangsu, 215125, China
| | - Shandong Zhao
- Department of Clinical Pharmacology, First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yicong Bian
- Department of Clinical Pharmacology, First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Na Dai
- Department of Nuclear Medicine, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yushuang Zheng
- Department of Pathology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shibiao Sang
- Department of Nuclear Medicine, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Linchuan Guo
- Department of Pathology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chenrong Huang
- Department of Clinical Pharmacology, First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Hua Zhang
- Department of Clinical Pharmacology, First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jiwei Jiang
- Department of Nuclear Medicine, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Chun Xu
- Department of Thoracic Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Zhao
- Department of Radiation Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiajun Han
- Department of Thoracic Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tao Xu
- Smart-Nuclide Biotech, No. 218 Xing-Hu Rd, Suzhou, Jiangsu, 215125, China.
| | - Songbing Qin
- Department of Radiation Oncology, First Affiliated Hospital of Soochow University, Suzhou, China.
- First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd, Suzhou, Jiangsu, 215006, China.
| | - Liyan Miao
- Department of Clinical Pharmacology, First Affiliated Hospital of Soochow University, Suzhou, China.
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
- First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd, Suzhou, Jiangsu, 215006, China.
| |
Collapse
|
6
|
Lan D, Chen Z, Mu J, Chen H, Zhao Y. Approaches to Radiolabeling Nanobodies for Biomedical Applications. ACS APPLIED MATERIALS & INTERFACES 2025; 17:10265-10275. [PMID: 39904609 DOI: 10.1021/acsami.4c21014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Molecular imaging plays a vital role in diagnosing diseases, monitoring treatments, and evaluating therapeutic efficacy by enabling noninvasive visualization of biological processes. Nanobodies, single-domain antibodies derived from camelids, have emerged as promising candidates for a wide range of biomedical applications due to their unique properties, including small size, high affinity, rapid clearance, and deep-tissue penetration. While effective radiolabeling techniques remain a major challenge to fully realize their clinical potential, this review aims to present recent advances in nanobody radiolabeling, focusing on radionuclides like 64Cu, 68Ga, 89Zr, and 111In, along with their associated chelators and conjugation methods. We highlight the development of innovative chelators, including p-SCN-Bn-HOPO and desferrioxamine derivatives that enhance specificity and stability, as well as advances in conjugation techniques that influence biodistribution and pharmacokinetics. These findings highlight the essential role of nanobody-based molecular imaging for precise diagnostics and targeted therapy.
Collapse
Affiliation(s)
- Deren Lan
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
- School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Zhulan Chen
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shantou University Medical College, Shantou 515041, China
| | - Jing Mu
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Haibo Chen
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yongsheng Zhao
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| |
Collapse
|
7
|
Mehrotra S, Kaur N, Kaur S, Matharoo K, Pandey RK. From antibodies to nanobodies: The next frontier in cancer theranostics for solid tumors. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:287-329. [PMID: 39978969 DOI: 10.1016/bs.apcsb.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
The field of cancer therapeutics has witnessed significant advancements over the past decades, particularly with the emergence of immunotherapy. This chapter traces the transformative journey from traditional antibody-based therapies to the innovative use of nanobodies in the treatment and diagnosis of solid tumors. Nanobodies are the smallest fragments of antibodies derived from camelid immunoglobulins and have redefined the possibilities in cancer theranostics due to their unique structural and functional properties. We provide an overview of the biochemical characteristics of nanobodies that make them particularly suitable for theranostic applications, such as their small size, high stability, enhanced infiltration into the complex tumor microenvironment (TME) and ability to bind with high affinity to epitopes that are inaccessible to conventional antibodies. Further, their ease of modification and functionalization has enabled the development of nanobody-based drug conjugates/toxins and radiolabeled compounds for precise imaging and targeted radiotherapy. We elucidate how nanobodies are being served as valuable tools for prognostic assessment, enabling clinicians to predict disease aggressiveness, monitor treatment response, and stratify patients for personalized therapeutic interventions.
Collapse
Affiliation(s)
- Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India.
| | - Navdeep Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Sukhpreet Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Kawaljit Matharoo
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | | |
Collapse
|
8
|
Ghosh P, Davies LJ, Nitsche C. Engineered Nanobodies Bind Bismuth, Indium and Gallium for Applications in Theranostics. Angew Chem Int Ed Engl 2025; 64:e202419455. [PMID: 39481115 DOI: 10.1002/anie.202419455] [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: 10/09/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/02/2024]
Abstract
Targeted theranostics heavily rely on metal isotopes conjugated to antibodies. Single-domain antibodies, known as nanobodies, are much smaller in size without compromising specificity and affinity. The conventional way of conjugating metals to nanobodies involves non-specific modification of amino acid residues with bifunctional chelating agents. We demonstrate that mutagenesis of a single residue in a nanobody creates a triple cysteine motif that selectively binds bismuth which is, for example, used in targeted alpha therapy. Two mutations create a quadruple cysteine mutant specific for gallium and indium used in positron emission tomography and single-photon emission computed tomography, respectively. Labelling is quantitative within a few minutes. The metal nanobodies maintain structural integrity and stability over weeks, resist competition from endogenous metal binders like glutathione, and retain functionality.
Collapse
Affiliation(s)
- Pritha Ghosh
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Lani J Davies
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Christoph Nitsche
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
9
|
Hu M, Zhang C, Fan D, Yang R, Bai Y, Shi H. Advances in Preclinical Research of Theranostic Radiopharmaceuticals in Nuclear Medicine. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4337-4353. [PMID: 39800975 DOI: 10.1021/acsami.4c20602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Theranostics of nuclear medicine refers to the combination of radionuclide imaging and internal irradiation therapy, which is currently a research hotspot and an important direction for the future development of nuclear medicine. Radiopharmaceutical is a vital component of nuclear medicine and serves as one of the fundamental pillars of molecular imaging and precision medicine. At present, a variety of radiopharmaceuticals have been developed for various targets such as fibroblast activation protein (FAP), prostate-specific membrane antigen (PSMA), somatostatin receptor 2 (SSTR2), C-X-C motif chemokine receptor 4 (CXCR4), human epidermal growth factor-2 (HER2), and integrin αvβ3, and some of them have been successfully applied in clinical practice. The radiopharmaceutical with theranostic function plays an important role in the diagnosis, treatment, efficacy evaluation, and prognosis prediction of cancers and is the key to realize the personalized treatment of tumors. This Review summarizes the preclinical research progress of theranostic radiopharmaceuticals toward the above targets in the field of nuclear medicine and discusses the prospects and development directions of radiopharmaceuticals in the future.
Collapse
Affiliation(s)
- Mei Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Chenshuo Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Dandan Fan
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Ru Yang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, P. R. China
| | - Yongxiang Bai
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Haibin Shi
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
10
|
Yang Y, Ren Z, Wang D, Tang D, Wei W, Song S, Lyu Y, Ding D, Tan W. Framework Nucleic Acid-Nanobody Fusion Probe-Based Pharmacokinetics Modulation and Analysis for Efficient Positron Emission Tomography Imaging. ACS NANO 2025; 19:439-450. [PMID: 39714093 DOI: 10.1021/acsnano.4c09127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Nanobodies are promising for immunoPET imaging due to their excellent antigen recognition and tumor targeting, yet rapid clearance limits their tumor accumulation. Although multimerization and albumin binding can extend their circulation time and improve tumor targeting, a simple and universal method for creating protein multimers is still needed. Here, we leveraged the facile synthesis, controllable size, and precise assembly of DNA nanotechnology to construct CD47-targeted framework nucleic acid-nanobody fusion probes with multiple valences and sizes. Following comprehensive structural characterization, in vitro specificity assessment and in vivo PET/CT imaging analysis were conducted on a colorectal cancer LS174T mouse model. Furthermore, a pharmacokinetic model was developed and fitted with considerable in vivo data to prove its rationality, followed by testing the effects on tumor uptake prediction by changing different pharmacokinetic parameters. Indeed, by manipulating the size of the nucleic acid scaffolding and the number of attached nanobodies, we could precisely modulate the accumulation of probes at the tumor site. Overall, this study not only developed an efficient strategy for constructing nanobody multimers but also provided a pharmacokinetic model, allowing profound insight into the multidimensional data obtained experimentally and informing the design of future imaging probes with predictable delivery efficacies.
Collapse
Affiliation(s)
- Yani Yang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Zhiqiang Ren
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dan Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Decui Tang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yifan Lyu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- Furong Laboratory, Changsha, Hunan 410082, China
| | - Ding Ding
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Weihong Tan
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
11
|
He Y, Tian R, Xu D, Wu Y, Rina S, Chen T, Guan Y, Xie T, Ying T, Xie F, Han J. Preclinical evaluation and pilot clinical study of [ 68Ga]Ga-NOTA-H006 for non-invasive PET imaging of 5T4 oncofetal antigen. Eur J Nucl Med Mol Imaging 2025; 52:611-622. [PMID: 39377811 DOI: 10.1007/s00259-024-06941-1] [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: 02/28/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
PURPOSE Trophoblast glycoprotein, the so-called 5T4, is an oncofetal antigen expressed in many different cancers. However, no 5T4-specific radioligand is employed in the clinic for non-invasive diagnosis. Thus, the aim of the current study was to develop a PET radiotracer for imaging 5T4 expression in preclinical and clinical stages. METHODS A VHH library was constructed by camel immunization. The specificity of the VHHs toward 5T4 antigen was screened through phage display biopanning and periplasmic extract enzyme-linked immunosorbent assay. 1,4,7-Triazacyclononane-1,4,7-triacetate acid (NOTA) derivative was conjugated to the selected VHH. After radiolabeling, microPET/CT and ex vivo biodistribution were conducted using BxPC-3 and MDA-MB-468 tumor-bearing mice. Cold VHH was co-injected with the tracer to challenge its binding in vivo. For the pilot clinical study, PET/CT images were acquired at 1 h after injection of tracer in patients with pathologically confirmed primary and metastatic tumors. RESULTS A library with a capacity of 1.2 × 1012 colony-forming units was constructed after successful camel immunization. Nb1-40 with a median effect concentration of 0.43 nM was selected. After humanization, the resulting H006 maintained a high affinity towards 5T4. [68Ga]Ga-NOTA-H006 with the molar activities of 6.48-54.2 GBq/µmol was prepared with high radiochemical purity (> 98%). Using [68Ga]Ga-NOTA-H006, microPET/CT revealed a clear visualization of 5T4 expression in BxPC-3 tumor-bearing mice. Ex vivo biodistribution showed that the highest tumor-to-blood ratio (∼ 3-fold) and tumor-to-muscle ratio (∼ 5-fold) were achieved at 60 min post-injection. Co-injection of the cold H006 at a dose of 1.5 mg/kg significantly reduced the tumor uptake (p < 0.0001). In the pilot clinical study, [68Ga]Ga-NOTA-H006 demonstrated its capacity to map 5T4-positive lesions in humans and yielded a mean effective dose of 3.4 × 10- 2 mSv/MBq. CONCLUSIONS [68Ga]Ga-NOTA-H006, which can visualize 5T4 expression in vivo, has been successfully developed. This opens up opportunities for non-invasively studying 5T4 expression through nuclear medicine. Further clinical investigations are warranted to explore its clinical value in disease progression and companion diagnosis.
Collapse
Affiliation(s)
- Yingfang He
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Ruhua Tian
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Dong Xu
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanfei Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China
| | - Sa Rina
- Huahe Pharmaceutical Co., Ltd, Shanghai, China
| | - Tengxiang Chen
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China
| | - Tianwu Xie
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China.
| | - Junbin Han
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China.
| |
Collapse
|
12
|
De Groof TWM, Lauwers Y, De Pauw T, Saxena M, Vincke C, Van Craenenbroeck J, Chapon C, Le Grand R, Raes G, Naninck T, Van Ginderachter JA, Devoogdt N. Specific imaging of CD8 + T-Cell dynamics with a nanobody radiotracer against human CD8β. Eur J Nucl Med Mol Imaging 2024; 52:193-207. [PMID: 39218831 PMCID: PMC11599301 DOI: 10.1007/s00259-024-06896-3] [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: 04/26/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE While immunotherapy has revolutionized the oncology field, variations in therapy responsiveness limit the broad applicability of these therapies. Diagnostic imaging of immune cell, and specifically CD8+ T cell, dynamics could allow early patient stratification and result in improved therapy efficacy and safety. In this study, we report the development of a nanobody-based immunotracer for non-invasive SPECT and PET imaging of human CD8+ T-cell dynamics. METHODS Nanobodies targeting human CD8β were generated by llama immunizations and subsequent biopanning. The lead anti-human CD8β nanobody was characterized on binding, specificity, stability and toxicity. The lead nanobody was labeled with technetium-99m, gallium-68 and copper-64 for non-invasive imaging of human T-cell lymphomas and CD8+ T cells in human CD8 transgenic mice and non-human primates by SPECT/CT or PET/CT. Repeated imaging of CD8+ T cells in MC38 tumor-bearing mice allowed visualization of CD8+ T-cell dynamics. RESULTS The nanobody-based immunotracer showed high affinity and specific binding to human CD8 without unwanted immune activation. CD8+ T cells were non-invasively visualized by SPECT and PET imaging in naïve and tumor-bearing mice and in naïve non-human primates with high sensitivity. The nanobody-based immunotracer showed enhanced specificity for CD8+ T cells and/or faster in vivo pharmacokinetics compared to previous human CD8-targeting immunotracers, allowing us to follow human CD8+ T-cell dynamics already at early timepoints. CONCLUSION This study describes the development of a more specific human CD8+ T-cell-targeting immunotracer, allowing follow-up of immunotherapy responses by non-invasive imaging of human CD8+ T-cell dynamics.
Collapse
Affiliation(s)
- Timo W M De Groof
- Molecular Imaging and Therapy Laboratory, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Yoline Lauwers
- Molecular Imaging and Therapy Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Tessa De Pauw
- Molecular Imaging and Therapy Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mohit Saxena
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses & Le Kremlin-Bicêtre, Inserm, Paris, CEA, France
| | - Cécile Vincke
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
- Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jolien Van Craenenbroeck
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
- Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Catherine Chapon
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses & Le Kremlin-Bicêtre, Inserm, Paris, CEA, France
| | - Roger Le Grand
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses & Le Kremlin-Bicêtre, Inserm, Paris, CEA, France
| | - Geert Raes
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
- Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thibaut Naninck
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses & Le Kremlin-Bicêtre, Inserm, Paris, CEA, France
| | - Jo A Van Ginderachter
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
- Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- Molecular Imaging and Therapy Laboratory, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
13
|
Dierick H, Navarro L, Van den Block S, Saliën J, Lahoutte T, Caveliers V, Bridoux J. Automated radiofluorination of HER2 single domain antibody: the road towards the clinical translation of [ 18F]FB-HER2 sdAb. EJNMMI Radiopharm Chem 2024; 9:77. [PMID: 39542993 PMCID: PMC11564621 DOI: 10.1186/s41181-024-00306-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND With the next generation of Human Epidermal Growth Factor Receptor 2 (HER2) -targeting therapies, such as antibody-drug conjugates, showing benefit in "HER2 low" and even "HER2 ultralow" patients, the need for novel methods to quantify HER2 expression accurately becomes even more important for clinical decision making. A HER2 PET/CT imaging assessment could evaluate HER2 positive disease locations while improving patient care, reducing the need for invasive biopsies. A single-domain antibody (sdAb)-based PET tracer could combine the high specificity of sdAbs with short-lived radionuclides such as fluorine-18 (18F) and gallium-68 (68Ga). SdAb-based PET tracers have clinically been used via a 68Ga-chelator approach. However, the distribution of 68Ga-labelled pharmaceuticals to peripheral PET centres is more challenging to organize due to the short half-life of 68Ga, most certainly when the available activity is limited by a generator. Cyclotron produced 68Ga has removed this limitation. Distribution of 18F-labelled pharmaceuticals remains less challenging due to its slightly longer half-life, and radiofluorination of sdAbs via N-succinimidyl-4-[18F]fluorobenzoate ([18F]SFB) has shown to be a promising strategy for developing sdAb-based PET tracers. Although [18F]SFB automation has been reported, automating protein conjugation proves challenging. Herein we report the fully automated, cartridge-based production of [18F]FB-HER2 sdAb on a single synthesis module. RESULTS [18F]FB-HER2 sdAb (> 6 GBq) was obtained after a fully automated production (95 min), with a RCP > 95%, apparent molar activity > 20 GBq/µmol and decay-corrected radiochemical yield (RCY d.c.) of 14 ± 2% (n = 4). Further upscaling amounted to production batches of 16 GBq with an apparent molar activity > 40 GBq/µmol and RCY d.c. of 8 ± 1% (n = 4). Ex vivo biodistribution and PET imaging showed specific HER2-positive tumour targeting and low kidney retention. CONCLUSION The [18F]FB-HER2 sdAb tracer was produced with clinically relevant activities using a fully automated production method. The automated production method was designed to ease the translation to the clinic and has the potential to be used not only in mono-centre but also multi-centre clinical trials with one central production site. [18F]FB-HER2 sdAb showed a favourable biodistribution pattern and could be a valuable alternative to 68Ga-labelled sdAb-based PET tracers in the clinic.
Collapse
Affiliation(s)
- Herlinde Dierick
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Nuclear Medicine Department, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | | | - Sonja Van den Block
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Nuclear Medicine Department, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Jelena Saliën
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Tony Lahoutte
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Nuclear Medicine Department, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Vicky Caveliers
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Nuclear Medicine Department, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Jessica Bridoux
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| |
Collapse
|
14
|
Sun Y, Zhai L, Ma L, Zhang W. Preclinical research progress in HER2-targeted small-molecule probes for breast cancer. RADIOLOGIE (HEIDELBERG, GERMANY) 2024; 64:47-53. [PMID: 39039211 PMCID: PMC11602795 DOI: 10.1007/s00117-024-01338-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/22/2024] [Indexed: 07/24/2024]
Abstract
Breast cancer is a malignant tumor that has the highest morbidity and mortality in women worldwide. Human epidermal growth factor receptor 2 (HER2) is a key driver of breast cancer development. Therefore, accurate assessment of HER2 expression in cancer patients and timely initiation or termination of anti-HER2 treatment are crucial for the prognosis of breast cancer patients. The emergence of radiolabeled molecular probes targeting HER2 makes this assessment possible. This article describes different types of small-molecule probes that target HER2 and are used in current preclinical applications and summarizes their advantages and disadvantages.
Collapse
Affiliation(s)
- Yefan Sun
- Department of Medical Imaging, Shanxi Medical University, 030001, Taiyuan, China
| | - Luoping Zhai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, 030032, Taiyuan, China
| | - Le Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, 030032, Taiyuan, China
| | - Wanchun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, 030032, Taiyuan, China.
| |
Collapse
|
15
|
Liu X, Song Y, Cheng P, Liang B, Xing D. Targeting HER2 in solid tumors: Unveiling the structure and novel epitopes. Cancer Treat Rev 2024; 130:102826. [PMID: 39270365 DOI: 10.1016/j.ctrv.2024.102826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024]
Abstract
Human epidermal growth factor receptor-2 (HER2) is overexpressed in various solid tumor types, acting as an established therapeutic target. Over the last three decades, the fast-paced development of diverse HER2-targeted agents, notably marked by the introduction of the antibody-drug conjugate (ADC), yielding substantial improvements in survival rates. However, resistance to anti-HER2 treatments continues to pose formidable challenges. The complex structure and dynamic dimerization properties of HER2 create significant hurdles in the development of novel targeted therapeutics. In this review, we synthesize the latest insights into the structural intricacies of HER2 and present an unprecedented overview of the epitope characteristics of HER2-targeted antibodies and their derivatives. Furthermore, we delve into the correlation between anti-HER2 antibody binding epitopes and their respective functions, with a particular focus on their efficacy against resistant tumors. In addition, we highlight the potential of emerging anti-HER2 agents that target specific sites or non-overlapping epitopes, poised to transform the therapeutic landscape for HER2-positive tumors in the foreseeable future.
Collapse
Affiliation(s)
- Xinlin Liu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao 266071, China
| | - Yunlong Song
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao Institute of Preventive Medicine, Qingdao 266033, China
| | - Panpan Cheng
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao Institute of Preventive Medicine, Qingdao 266033, China
| | - Bing Liang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
16
|
Alexander E, Leong KW. Discovery of nanobodies: a comprehensive review of their applications and potential over the past five years. J Nanobiotechnology 2024; 22:661. [PMID: 39455963 PMCID: PMC11515141 DOI: 10.1186/s12951-024-02900-y] [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: 06/19/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Nanobodies (Nbs) are antibody fragments derived from heavy-chain-only IgG antibodies found in the Camelidae family as well as cartilaginous fish. Their unique structural and functional properties, such as their small size, the ability to be engineered for high antigen-binding affinity, stability under extreme conditions, and ease of production, have made them promising tools for diagnostics and therapeutics. This potential was realized in 2018 with the approval of caplacizumab, the world's first Nb-based drug. Currently, Nbs are being investigated in clinical trials for a broad range of treatments, including targeted therapies against PDL1 and Epidermal Growth Factor Receptor (EGFR), cardiovascular diseases, inflammatory conditions, and neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. They are also being studied for their potential for detecting and imaging autoimmune conditions and infectious diseases such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A variety of methods are now available to generate target-specific Nbs quickly and efficiently at low costs, increasing their accessibility. This article examines these diverse applications of Nbs and their promising roles. Only the most recent articles published in the last five years have been used to summarize the most advanced developments in the field.
Collapse
Affiliation(s)
- Elena Alexander
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| |
Collapse
|
17
|
Teodori L, Ochoa SK, Omer M, Andersen VL, Bech P, Su J, Bridoux J, Nielsen JS, Bertelsen MB, Hernot S, Gothelf KV, Kjems J. Plug-and-play nucleic acid-mediated multimerization of biparatopic nanobodies for molecular imaging. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102305. [PMID: 39281705 PMCID: PMC11402398 DOI: 10.1016/j.omtn.2024.102305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 08/12/2024] [Indexed: 09/18/2024]
Abstract
In cancer molecular imaging, selecting binders with high specificity and affinity for biomarkers is paramount for achieving high-contrast imaging within clinical time frames. Nanobodies have emerged as potent candidates, surpassing antibodies in pre-clinical imaging due to their convenient production, rapid renal clearance, and deeper tissue penetration. Multimerization of nanobodies is a popular strategy to enhance their affinity and pharmacokinetics; however, traditional methods are laborious and may yield heterogeneous products. In this study, we employ a Holliday junction (HJ)-like nucleic acid-based scaffold to create homogeneous nanostructures with precise multivalent and multiparatopic nanobody displays. The plug-and-play assembly allowed the screening of several nanobody multimer configurations for the detection of the breast cancer biomarker, human epidermal growth factor receptor 2 (HER2). In vitro studies demonstrated significant improvements in binding avidity, particularly with the biparatopic construct exhibiting high sensitivity, surpassing that of traditional antibody-based cell binding. Furthermore, our HJ platform allowed for adaptation from fluorescence-based to nuclear imaging, as demonstrated in xenografted mice, thereby allowing for future in vivo applications. This work highlights the potential of nucleic acid-mediated multimerization to markedly enhance nanobody binding, by exploring synergistic combinations and offering versatility for both in vitro diagnostics and cancer molecular imaging with prospects for future theranostic applications.
Collapse
Affiliation(s)
- Laura Teodori
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Sarah K Ochoa
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Marjan Omer
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Veronica L Andersen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Pernille Bech
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Junyi Su
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Jessica Bridoux
- Molecular Imaging and Therapy Laboratory (MITH), Vrije Universiteit Brussel (VUB), Building K, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Jesper S Nielsen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Mathias B Bertelsen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
- Department of Chemistry, Aarhus University, Langelandsgade 140, 8000 Aarhus C, Denmark
| | - Sophie Hernot
- Molecular Imaging and Therapy Laboratory (MITH), Vrije Universiteit Brussel (VUB), Building K, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Kurt V Gothelf
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
- Department of Chemistry, Aarhus University, Langelandsgade 140, 8000 Aarhus C, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
- Department of Molecular Biology and Genetics, Universitetsbyen 81, 8000 Aarhus C, Denmark
| |
Collapse
|
18
|
Höffgen KS, Dabel J, Konken CP, Depke DA, Hermann S, Dörner W, Schelhaas S, Schäfers M, Mootz HD. Combining poly-epitope MoonTags and labeled nanobodies for signal amplification in cell-specific PET imaging in vivo. Nucl Med Biol 2024; 136-137:108937. [PMID: 38964257 DOI: 10.1016/j.nucmedbio.2024.108937] [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: 03/08/2024] [Revised: 05/31/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
Immunorecognition provides an excellent basis for targeted imaging techniques covering a wide range from basic research to diagnostics and from single cells to whole organisms. Fluorescence- or radioisotope-labeled antibodies, antibody fragments or nanobodies enable a direct signal readout upon binding and allow for versatile imaging from microscopy to whole-body imaging. However, as the signal intensity directly correlates with the number of labeled antibodies bound to their epitopes (1:1 binding), sensitivity for low-expressing epitopes can be limiting for visualization. For the first time, we developed poly-epitope tags with multiple copies (1 to 7) of a short peptide epitope, specifically the MoonTag, that are recognized by a labeled nanobody and aimed at signal amplification in microscopy and cell-specific PET imaging. In transiently transfected HeLa cells or stably transduced A4573 cells we characterized complex formation and in vitro signal amplification. Indeed, using fluorescently and radioactively labeled nanobodies we found an approximately linear signal amplification with increasing numbers of epitope copies in vitro. To test the poly-epitope approach in vivo, A4573 tumor cells were injected subcutaneously into the shoulder of NSG mice, with A4573 tumor cells expressing a poly-epitope of 7 MoonTags on one side and WT cells on the other side. Using a [68Ga]-labeled NODAGA-conjugated MoonTag nanobody, we performed PET/CT imaging at day 8-9 after tumor implantation. Specific binding of a [68Ga]-labeled NODAGA-conjugated MoonTag nanobody was observed in 7xMoonTag tumors (1.7 ± 0.5%ID/mL) by PET imaging, showing significantly higher radiotracer accumulation compared to the WT tumors (1.1 ± 0.3%ID/mL; p < 0.01). Ex vivo gamma counter measurements confirmed significantly higher uptake in 7xMoonTag tumors compared to WT tumors (p < 0.001). In addition, MoonTag nanobody binding was detected by autoradiography which was spatially matched with histological analysis of the tumor tissues. In conclusion, we expect nanobody-based poly-epitope tag strategies to be widely applicable for multimodal imaging techniques given the advantageous properties of nanobodies and their amenability to genetic and chemical engineering.
Collapse
Affiliation(s)
| | - Jennifer Dabel
- University of Münster, European Institute for Molecular Imaging (EIMI), Münster, Germany
| | - Christian P Konken
- University of Münster, European Institute for Molecular Imaging (EIMI), Münster, Germany; University Hospital Münster, Department of Nuclear Medicine, Münster, Germany
| | - Dominic A Depke
- University of Münster, European Institute for Molecular Imaging (EIMI), Münster, Germany
| | - Sven Hermann
- University of Münster, European Institute for Molecular Imaging (EIMI), Münster, Germany
| | - Wolfgang Dörner
- University of Münster, Institute of Biochemistry, Münster, Germany
| | - Sonja Schelhaas
- University of Münster, European Institute for Molecular Imaging (EIMI), Münster, Germany
| | - Michael Schäfers
- University of Münster, European Institute for Molecular Imaging (EIMI), Münster, Germany; University Hospital Münster, Department of Nuclear Medicine, Münster, Germany.
| | - Henning D Mootz
- University of Münster, Institute of Biochemistry, Münster, Germany.
| |
Collapse
|
19
|
Zeven K, Lauwers Y, De Mey L, Debacker JM, De Pauw T, De Groof TWM, Devoogdt N. Advancements in nuclear imaging using radiolabeled nanobody tracers to support cancer immunotherapy. IMMUNOTHERAPY ADVANCES 2024; 4:ltae006. [PMID: 39281708 PMCID: PMC11402390 DOI: 10.1093/immadv/ltae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/23/2024] [Indexed: 09/18/2024] Open
Abstract
The evolving landscape of cancer immunotherapy has revolutionized cancer treatment. However, the dynamic tumor microenvironment has led to variable clinical outcomes, indicating a need for predictive biomarkers. Noninvasive nuclear imaging, using radiolabeled modalities, has aided in patient selection and monitoring of their treatment response. This approach holds promise for improving diagnostic accuracy, providing a more personalized treatment regimen, and enhancing the clinical response. Nanobodies or single-domain antibodies, derived from camelid heavy-chain antibodies, allow early timepoint detection of targets with high target-to-background ratios. To date, a plethora of nanobodies have been developed for nuclear imaging of tumor-specific antigens, immune checkpoints, and immune cells, both at a preclinical and clinical level. This review comprehensively outlines the recent advancements in nanobody-based nuclear imaging, both on preclinical and clinical levels. Additionally, the impact and expected future advancements on the use of nanobody-based radiopharmaceuticals in supporting cancer diagnosis and treatment follow-up are discussed.
Collapse
Affiliation(s)
- Katty Zeven
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Yoline Lauwers
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Lynn De Mey
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Jens M Debacker
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Tessa De Pauw
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Timo W M De Groof
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Nick Devoogdt
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
20
|
Garaulet G, Báez BB, Medrano G, Rivas-Sánchez M, Sánchez-Alonso D, Martinez-Torrecuadrada JL, Mulero F. Radioimmunotheragnosis in Cancer Research. Cancers (Basel) 2024; 16:2896. [PMID: 39199666 PMCID: PMC11352548 DOI: 10.3390/cancers16162896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
The combination of immunoPET-where an antibody (Ab) is labeled with an isotope for PET imaging-and radioimmunotherapy (RIT), using the same antibody with a therapeutic isotope, offers significant advantages in cancer management. ImmunoPET allows non-invasive imaging of antigen expression, which aids in patient selection for subsequent radioimmunotherapy. It also facilitates the assessment of tumor response to therapy, allowing for treatment adjustments if necessary. In addition, immunoPET provides critical pharmacokinetic data, including antibody biodistribution and clearance rates, which are essential for dosimetry calculations and treatment protocol optimization. There are still challenges to overcome. Identifying appropriate target antigens that are selectively expressed on cancer cells while minimally expressed on normal tissues remains a major hurdle to reduce off-target toxicity. In addition, it is critical to optimize the pharmacokinetics of radiolabeled antibodies to maximize tumor uptake and minimize normal tissue uptake, particularly in vital organs such as the liver and kidney. This approach offers the potential for targeted and personalized cancer therapy with reduced systemic toxicity by exploiting the specificity of monoclonal antibodies and the cytotoxic effects of radiation. However, further research is needed to address remaining challenges and to optimize these technologies for clinical use.
Collapse
Affiliation(s)
- Guillermo Garaulet
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - Bárbara Beatriz Báez
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - Guillermo Medrano
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - María Rivas-Sánchez
- Protein Production Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (M.R.-S.); (D.S.-A.)
| | - David Sánchez-Alonso
- Protein Production Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (M.R.-S.); (D.S.-A.)
| | | | - Francisca Mulero
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| |
Collapse
|
21
|
Ge S, Wang C, You X, He H, Zhang B, Jia T, Cai X, Sang S, Xu T, Deng S. Imaging and Monitoring HER2 Expression in Tumors during HER2 Antibody-Drug Conjugate Therapy Utilizing a Radiolabeled Site-Specific Single-Domain Antibody Probe: 68Ga-NODAGA-SNA004-GSC. J Med Chem 2024. [PMID: 39077778 DOI: 10.1021/acs.jmedchem.4c00857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
The overexpression of HER2 is pivotal in the initiation and progression of breast cancer. Developing HER2-targeted radiotracers is crucial for noninvasive assessment of HER2 expression, patient selection for HER2-targeted therapy, monitoring treatment response, and identifying resistance. Here, we reported a nonsite-specific coupled radiotracer, 68Ga-NOTA-SNA004-His6, and a site-specific coupled radiotracer, 68Ga-NODAGA-SNA004-GSC, based on a novel HER2 nanobody, SNA004. Both radiotracers exhibited high affinity, specific targeting, and rapid clearance in vitro and in vivo. Additionally, these tracers and trastuzumab showed noncompetitive binding to HER2. Compared to 68Ga-NOTA-SNA004-His6, 68Ga-NODAGA-SNA004-GSC demonstrated significantly reduced renal and liver uptake. PET/CT imaging with 68Ga-NODAGA-SNA004-GSC sensitively detected the responsiveness of various tumor models to trastuzumab and its antibody-drug conjugates (ADCs). Overall, the site-specific coupled radiotracer 68Ga-NODAGA-SNA004-GSC offered significant advantages in biodistribution and signal-to-noise ratio, making it a valuable tool for monitoring HER2 expression levels before, during, and after trastuzumab and ADC treatment.
Collapse
Affiliation(s)
- Shushan Ge
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215006, China
- Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang 621099, China
| | - Chao Wang
- Smart-Nuclide Biotech, No. 218 Xing-Hu Road, Suzhou, 215125, China
| | - Xuyang You
- Department of Nuclear Medicine, Suzhou Ninth People's Hospital, Suzhou 215006, China
| | - Huihui He
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi 214062, China
| | - Bin Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Tongtong Jia
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiaowei Cai
- Department of Nuclear Medicine, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian 223812, China
| | - Shibiao Sang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Tao Xu
- Smart-Nuclide Biotech, No. 218 Xing-Hu Road, Suzhou, 215125, China
| | - Shengming Deng
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang 621099, China
| |
Collapse
|
22
|
Medina Pérez VM, Baselga M, Schuhmacher AJ. Single-Domain Antibodies as Antibody-Drug Conjugates: From Promise to Practice-A Systematic Review. Cancers (Basel) 2024; 16:2681. [PMID: 39123409 PMCID: PMC11311928 DOI: 10.3390/cancers16152681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Antibody-drug conjugates (ADCs) represent potent cancer therapies that deliver highly toxic drugs to tumor cells precisely, thus allowing for targeted treatment and significantly reducing off-target effects. Despite their effectiveness, ADCs can face limitations due to acquired resistance and potential side effects. OBJECTIVES This study focuses on advances in various ADC components to improve both the efficacy and safety of these agents, and includes the analysis of several novel ADC formats. This work assesses whether the unique features of VHHs-such as their small size, enhanced tissue penetration, stability, and cost-effectiveness-make them a viable alternative to conventional antibodies for ADCs and reviews their current status in ADC development. METHODS Following PRISMA guidelines, this study focused on VHHs as components of ADCs, examining advancements and prospects from 1 January 2014 to 30 June 2024. Searches were conducted in PubMed, Cochrane Library, ScienceDirect and LILACS using specific terms related to ADCs and single-domain antibodies. Retrieved articles were rigorously evaluated, excluding duplicates and non-qualifying studies. The selected peer-reviewed articles were analyzed for quality and synthesized to highlight advancements, methods, payloads, and future directions in ADC research. RESULTS VHHs offer significant advantages for drug conjugation over conventional antibodies due to their smaller size and structure, which enhance tissue penetration and enable access to previously inaccessible epitopes. Their superior stability, solubility, and manufacturability facilitate cost-effective production and expand the range of targetable antigens. Additionally, some VHHs can naturally cross the blood-brain barrier or be easily modified to favor their penetration, making them promising for targeting brain tumors and metastases. Although no VHH-drug conjugates (nADC or nanoADC) are currently in the clinical arena, preclinical studies have explored various conjugation methods and linkers. CONCLUSIONS While ADCs are transforming cancer treatment, their unique mechanisms and associated toxicities challenge traditional views on bioavailability and vary with different tumor types. Severe toxicities, often linked to compound instability, off-target effects, and nonspecific blood cell interactions, highlight the need for better understanding. Conversely, the rapid distribution, tumor penetration, and clearance of VHHs could be advantageous, potentially reducing toxicity by minimizing prolonged exposure. These attributes make single-domain antibodies strong candidates for the next generation of ADCs, potentially enhancing both efficacy and safety.
Collapse
Affiliation(s)
- Víctor Manuel Medina Pérez
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Marta Baselga
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Alberto J. Schuhmacher
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Fundación Aragonesa para la Investigación y el Desarrollo (ARAID), 50018 Zaragoza, Spain
| |
Collapse
|
23
|
Abdolvahab MH, Karimi P, Mohajeri N, Abedini M, Zare H. Targeted drug delivery using nanobodies to deliver effective molecules to breast cancer cells: the most attractive application of nanobodies. Cancer Cell Int 2024; 24:67. [PMID: 38341580 DOI: 10.1186/s12935-024-03259-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Targeted drug delivery is one of the attractive ways in which cancer treatment can significantly reduce side effects. In the last two decades, the use of antibodies as a tool for accurate detection of cancer has been noted. On the other hand, the binding of drugs and carriers containing drugs to the specific antibodies of cancer cells can specifically target only these cells. However, the use of whole antibodies brings challenges, including their large size, the complexity of conjugation, the high cost of production, and the creation of immunogenic reactions in the body. The use of nanobodies, or VHHs, which are a small part of camel heavy chain antibodies, is very popular due to their small size, high craftsmanship, and low production cost. In this article, in addition to a brief overview of the structure and characteristics of nanobodies, the use of this molecule in the targeted drug delivery of breast cancer has been reviewed.
Collapse
Affiliation(s)
- Mohadeseh Haji Abdolvahab
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Pegah Karimi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Nasrin Mohajeri
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Abedini
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Hamed Zare
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
24
|
Gondry O, Caveliers V, Xavier C, Raes L, Vanhoeij M, Verfaillie G, Fontaine C, Glorieus K, De Grève J, Joris S, Luyten I, Zwaenepoel K, Vandenbroucke F, Waelput W, Thyparambil S, Vaneycken I, Cousaert J, Bourgeois S, Devoogdt N, Goethals L, Everaert H, De Geeter F, Lahoutte T, Keyaerts M. Phase II Trial Assessing the Repeatability and Tumor Uptake of [ 68Ga]Ga-HER2 Single-Domain Antibody PET/CT in Patients with Breast Carcinoma. J Nucl Med 2024; 65:178-184. [PMID: 38302159 PMCID: PMC10858381 DOI: 10.2967/jnumed.123.266254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/03/2023] [Indexed: 02/03/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) status is used for decision-making in breast carcinoma treatment. The status is obtained through immunohistochemistry or in situ hybridization. These two methods have the disadvantage of necessitating tissue sampling, which is prone to error due to tumor heterogeneity or interobserver variability. Whole-body imaging might be a solution to map HER2 expression throughout the body. Methods: Twenty patients with locally advanced or metastatic breast carcinoma (5 HER2-positive and 15 HER2-negative patients) were included in this phase II trial to assess the repeatability of uptake quantification and the extended safety of the [68Ga]Ga-NOTA-anti-HER2 single-domain antibody (sdAb). The tracer was injected, followed by a PET/CT scan at 90 min. Within 8 d, the procedure was repeated. Blood samples were taken for antidrug antibody (ADA) assessment and liquid biopsies. On available tissues, immunohistochemistry, in situ hybridization, and mass spectrometry were performed to determine the correlation of HER2 status with uptake values measured on PET. If relevant preexisting [18F]FDG PET/CT images were available (performed as standard of care), a comparison was made. Results: With a repeatability coefficient of 21.8%, this imaging technique was repeatable. No clear correlation between PET/CT uptake values and pathology could be established, as even patients with low levels of HER2 expression showed moderate to high uptake. Comparison with [18F]FDG PET/CT in 16 patients demonstrated that in 7 patients, [68Ga]Ga-NOTA-anti-HER2 shows interlesional heterogeneity within the same patient, and [18F]FDG uptake did not show the same heterogeneous uptake in all patients. In some patients, the extent of disease was clearer with the [68Ga]Ga-NOTA-anti-HER2-sdAb. Sixteen adverse events were reported but all without a clear relationship to the tracer. Three patients with preexisting ADAs did not show adverse reactions. No new ADAs developed. Conclusion: [68Ga]Ga-NOTA-anti-HER2-sdAb PET/CT imaging shows similar repeatability to [18F]FDG. It is safe for clinical use. There is tracer uptake in cancer lesions, even in patients previously determined to be HER2-low or -negative. The tracer shows potential in the assessment of interlesional heterogeneity of HER2 expression. In a subset of patients, [68Ga]Ga-NOTA-anti-HER2-sdAb uptake was seen in lesions with no or low [18F]FDG uptake. These findings support further clinical development of [68Ga]Ga-NOTA-anti-HER2-sdAb as a PET/CT tracer in breast cancer patients.
Collapse
Affiliation(s)
- Odrade Gondry
- Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium;
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Vicky Caveliers
- Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Catarina Xavier
- Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laurens Raes
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Marian Vanhoeij
- Department of Surgical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Guy Verfaillie
- Department of Surgical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Christel Fontaine
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Katrien Glorieus
- Department of Surgical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Jacques De Grève
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Sofie Joris
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ine Luyten
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Karen Zwaenepoel
- Centre for Oncological Research, University of Antwerp, Wilrijk, Belgium
| | | | - Wim Waelput
- Department of Pathology, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Experimental Pathology, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Ilse Vaneycken
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Julie Cousaert
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Sophie Bourgeois
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Nick Devoogdt
- Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lode Goethals
- Department of Radiology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Hendrik Everaert
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Frank De Geeter
- Department of Nuclear Medicine, Algemeen Ziekenhuis Sint-Jan Brugge Oostende, Bruges, Belgium
| | - Tony Lahoutte
- Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Marleen Keyaerts
- Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium;
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
25
|
Cong Y, Devoogdt N, Lambin P, Dubois LJ, Yaromina A. Promising Diagnostic and Therapeutic Approaches Based on VHHs for Cancer Management. Cancers (Basel) 2024; 16:371. [PMID: 38254860 PMCID: PMC10814765 DOI: 10.3390/cancers16020371] [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: 11/29/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
The discovery of the distinctive structure of heavy chain-only antibodies in species belonging to the Camelidae family has elicited significant interest in their variable antigen binding domain (VHH) and gained attention for various applications, such as cancer diagnosis and treatment. This article presents an overview of the characteristics, advantages, and disadvantages of VHHs as compared to conventional antibodies, and their usage in diverse applications. The singular properties of VHHs are explained, and several strategies that can augment their utility are outlined. The preclinical studies illustrating the diagnostic and therapeutic efficacy of distinct VHHs in diverse formats against solid cancers are summarized, and an overview of the clinical trials assessing VHH-based agents in oncology is provided. These investigations demonstrate the enormous potential of VHHs for medical research and healthcare.
Collapse
Affiliation(s)
- Ying Cong
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6211 LK Maastricht, The Netherlands; (Y.C.); (P.L.)
| | - Nick Devoogdt
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, 1090 Brussels, Belgium;
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6211 LK Maastricht, The Netherlands; (Y.C.); (P.L.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Ludwig J. Dubois
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6211 LK Maastricht, The Netherlands; (Y.C.); (P.L.)
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6211 LK Maastricht, The Netherlands; (Y.C.); (P.L.)
| |
Collapse
|
26
|
Wagner TR, Blaess S, Leske IB, Frecot DI, Gramlich M, Traenkle B, Kaiser PD, Seyfried D, Maier S, Rezza A, Sônego F, Thiam K, Pezzana S, Zeck A, Gouttefangeas C, Scholz AM, Nueske S, Maurer A, Kneilling M, Pichler BJ, Sonanini D, Rothbauer U. Two birds with one stone: human SIRPα nanobodies for functional modulation and in vivo imaging of myeloid cells. Front Immunol 2023; 14:1264179. [PMID: 38164132 PMCID: PMC10757926 DOI: 10.3389/fimmu.2023.1264179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Signal-regulatory protein α (SIRPα) expressed by myeloid cells is of particular interest for therapeutic strategies targeting the interaction between SIRPα and the "don't eat me" ligand CD47 and as a marker to monitor macrophage infiltration into tumor lesions. To address both approaches, we developed a set of novel human SIRPα (hSIRPα)-specific nanobodies (Nbs). We identified high-affinity Nbs targeting the hSIRPα/hCD47 interface, thereby enhancing antibody-dependent cellular phagocytosis. For non-invasive in vivo imaging, we chose S36 Nb as a non-modulating binder. By quantitative positron emission tomography in novel hSIRPα/hCD47 knock-in mice, we demonstrated the applicability of 64Cu-hSIRPα-S36 Nb to visualize tumor infiltration of myeloid cells. We envision that the hSIRPα-Nbs presented in this study have potential as versatile theranostic probes, including novel myeloid-specific checkpoint inhibitors for combinatorial treatment approaches and for in vivo stratification and monitoring of individual responses during cancer immunotherapies.
Collapse
Affiliation(s)
- Teresa R. Wagner
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Simone Blaess
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Inga B. Leske
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Desiree I. Frecot
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Marius Gramlich
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Bjoern Traenkle
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Philipp D. Kaiser
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Dominik Seyfried
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany
| | - Sandra Maier
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Amélie Rezza
- Preclinical Models & Services, genOway, Lyon, France
| | | | - Kader Thiam
- Preclinical Models & Services, genOway, Lyon, France
| | - Stefania Pezzana
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Anne Zeck
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Cécile Gouttefangeas
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany
- Department of Immunology, Institute of Cell Biology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Armin M. Scholz
- Livestock Center of the Faculty of Veterinary Medicine, Ludwig Maximilians University Munich, Oberschleissheim, Germany
| | - Stefan Nueske
- Livestock Center of the Faculty of Veterinary Medicine, Ludwig Maximilians University Munich, Oberschleissheim, Germany
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Manfred Kneilling
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Dermatology, University of Tübingen, Tübingen, Germany
| | - Bernd J. Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Dominik Sonanini
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
- Department of Medical Oncology and Pneumology, University of Tübingen, Tübingen, Germany
| | - Ulrich Rothbauer
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| |
Collapse
|
27
|
Dekempeneer Y, Massa S, Santens F, Navarro L, Berdal M, Lucero MM, Pombo Antunes AR, Lahoutte T, Van Ginderachter JA, Devoogdt N, D'Huyvetter M. Preclinical Evaluation of a Radiotheranostic Single-Domain Antibody Against Fibroblast Activation Protein α. J Nucl Med 2023; 64:1941-1948. [PMID: 38040444 DOI: 10.2967/jnumed.123.266381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/27/2023] [Indexed: 12/03/2023] Open
Abstract
Fibroblast activation protein α (FAP) is highly expressed on cancer-associated fibroblasts of epithelial-derived cancers. Breast, colon, and pancreatic tumors often show strong desmoplastic reactions, which result in a dominant presence of stromal cells. FAP has gained interest as a target for molecular imaging and targeted therapies. Single-domain antibodies (sdAbs) are the smallest antibody-derived fragments with beneficial pharmacokinetic properties for molecular imaging and targeted therapy. Methods: We describe the generation, selection, and characterization of a sdAb against FAP. In mice, we assessed its imaging and therapeutic potential after radiolabeling with tracer-dose 131I and 68Ga for SPECT and PET imaging, respectively, and with 131I and 225Ac for targeted radionuclide therapy. Results: The lead sdAb, 4AH29, exhibiting picomolar affinity for a distinct FAP epitope, recognized both purified and membrane-bound FAP protein. Radiolabeled versions, including [68Ga]Ga-DOTA-4AH29, [225Ac]Ac-DOTA-4AH29, and [131I]I-guanidinomethyl iodobenzoate (GMIB)-4AH29, displayed radiochemical purities exceeding 95% and effectively bound to recombinant human FAP protein and FAP-positive GM05389 human fibroblasts. These radiolabeled compounds exhibited rapid and specific accumulation in human FAP-positive U87-MG glioblastoma tumors, with low but specific uptake in lymph nodes, uterus, bone, and skin (∼2-3 percentage injected activity per gram of tissue [%IA/g]). Kidney clearance of unbound [131I]I-GMIB-4AH29 was fast (<1 %IA/g after 24 h), whereas [225Ac]Ac-DOTA-4AH29 exhibited slower clearance (8.07 ± 1.39 %IA/g after 24 h and 2.47 ± 0.18 %IA/g after 96 h). Mice treated with [225Ac]Ac-DOTA-4AH29 and [131I]I-GMIB-4AH29 demonstrated prolonged survival compared with those receiving vehicle solution. Conclusion: [68Ga]Ga-DOTA-4AH29 and [131I]I-GMIB-4AH29 enable precise FAP-positive tumor detection in mice. Therapeutic [225Ac]Ac-DOTA-4AH29 and [131I]I-GMIB-4AH29 exhibit strong and sustained tumor targeting, resulting in dose-dependent therapeutic effects in FAP-positive tumor-bearing mice, albeit with kidney toxicity observed later for [225Ac]Ac-DOTA-4AH29. This study confirms the potential of radiolabeled sdAb 4AH29 as a radiotheranostic agent for FAP-positive cancers, warranting clinical evaluation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Tony Lahoutte
- Precirix NV/SA, Brussels, Belgium
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Nuclear Medicine, UZ Brussel, Brussels, Belgium
| | - Jo A Van Ginderachter
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; and
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Nick Devoogdt
- Precirix NV/SA, Brussels, Belgium
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Matthias D'Huyvetter
- Precirix NV/SA, Brussels, Belgium;
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
28
|
Declerck NB, Huygen C, Mateusiak L, Stroet MCM, Hernot S. The GEM-handle as convenient labeling strategy for bimodal single-domain antibody-based tracers carrying 99mTc and a near-infrared fluorescent dye for intra-operative decision-making. Front Immunol 2023; 14:1285923. [PMID: 38035094 PMCID: PMC10684908 DOI: 10.3389/fimmu.2023.1285923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Intra-operative fluorescence imaging has demonstrated its ability to improve tumor lesion identification. However, the limited tissue penetration of the fluorescent signals hinders the detection of deep-lying or occult lesions. Integrating fluorescence imaging with SPECT and/or intra-operative gamma-probing synergistically combines the deep tissue penetration of gamma rays for tumor localization with the precision of fluorescence imaging for precise tumor resection. In this study, we detail the use of a genetically encoded multifunctional handle, henceforth referred to as a GEM-handle, for the development of fluorescent/radioactive bimodal single-domain antibody (sdAb)-based tracers. A sdAb that targets the urokinase plasminogen activator receptor (uPAR) was engineered to carry a GEM-handle containing a carboxy-terminal hexahistidine-tag and cysteine-tag. A two-step labeling strategy was optimized and applied to site-specifically label IRDye800CW and 99mTc to the sdAb. Bimodal labeling of the sdAbs proved straightforward and successful. 99mTc activity was however restricted to 18.5 MBq per nmol fluorescently-labeled sdAb to prevent radiobleaching of IRDye800CW without impeding SPECT/CT imaging. Subsequently, the in vivo biodistribution and tumor-targeting capacity of the bimodal tracer were evaluated in uPAR-positive tumor-bearing mice using SPECT/CT and fluorescence imaging. The bimodal sdAb showed expected renal background signals due to tracer clearance, along with slightly elevated non-specific liver signals. Four hours post-injection, both SPECT/CT and fluorescent images achieved satisfactory tumor uptake and contrast, with significantly higher values observed for the anti-uPAR bimodal sdAb compared to a control non-targeting sdAb. In conclusion, the GEM-handle is a convenient method for designing and producing bimodal sdAb-based tracers with adequate in vivo characteristics.
Collapse
Affiliation(s)
| | | | | | | | - Sophie Hernot
- Molecular Imaging and Therapy Laboratory (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
29
|
Zhou S, Fang X, Lv J, Yang Y, Zeng Y, Liu Y, Wei W, Huang G, Zhang B, Wu C. Site-Specific Modification of Single Domain Antibodies by Enzyme-Immobilized Magnetic Beads. Bioconjug Chem 2023; 34:1914-1922. [PMID: 37804224 DOI: 10.1021/acs.bioconjchem.3c00423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2023]
Abstract
Nanobodies as imaging agents and drug conjugates have shown great potential for cancer diagnostics and therapeutics. However, site-specific modification of a nanobody with microbial transglutaminase (mTGase) encounters problems in protein separation and purification. Here, we describe a facile yet reliable strategy of immobilizing mTGase onto magnetic beads for site-specific nanobody modification. The mTGase immobilized on magnetic beads (MB-mTGase) exhibits catalytic activity nearly equivalent to that of the free mTGase, with good reusability and universality. Magnetic separation simplifies the protein purification step and reduces the loss of nanobody bioconjugates more effectively than size exclusion chromatography. Using MB-mTGase, we demonstrate site-specific conjugation of nanobodies with fluorescent dyes and polyethylene glycol molecules, enabling targeted immunofluorescence imaging and improved circulation dynamics and tumor accumulation in vivo. The combined advantages of MB-mTGase method, including high conjugation efficiency, quick purification, less protein loss, and recycling use, are promising for site-specific nanobody functionalization and biomedical applications.
Collapse
Affiliation(s)
- Siyu Zhou
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Xiaofeng Fang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Jiahui Lv
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Yicheng Yang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Yiqi Zeng
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Ying Liu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Bo Zhang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Changfeng Wu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| |
Collapse
|
30
|
Li L, Lin X, Wang L, Ma X, Zeng Z, Liu F, Jia B, Zhu H, Wu A, Yang Z. Immuno-PET of colorectal cancer with a CEA-targeted [68 Ga]Ga-nanobody: from bench to bedside. Eur J Nucl Med Mol Imaging 2023; 50:3735-3749. [PMID: 37382662 DOI: 10.1007/s00259-023-06313-1] [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: 03/02/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023]
Abstract
PURPOSE An accurate diagnosis of colorectal carcinoma (CRC) can assist physicians in developing reasonable therapeutic regimens, thereby significantly improving the patient's prognosis. Carcinoembryonic antigen (CEA)-targeted PET imaging shows great potential for this purpose. Despite showing remarkable abilities to detect primary and metastatic CRC, previously reported CEA-specific antibody radiotracers or pretargeted imaging are not suitable for clinical use due to poor pharmacokinetics and complicated imaging procedures. In contrast, radiolabeled nanobodies exhibit ideal characteristics for PET imaging, for instance, rapid clearance rates and excellent distribution profiles, allowing same-day imaging with sufficient contrast. In this study, we developed a novel CEA-targeted nanobody radiotracer, [68 Ga]Ga-HNI01, and assessed its tumor imaging ability and biodistribution profile in preclinical xenografts and patients with primary and metastatic CRC. METHODS The novel nanobody HNI01 was acquired by immunizing the llama with CEA proteins. [68 Ga]Ga-HNI01 was synthesized by site-specifically conjugating [68 Ga]Ga with tris(hydroxypyridinone) (THP). Small-animal PET imaging and biodistribution studies were performed in CEA-overexpressed LS174T and CEA-low-expressed HT-29 tumor models. Following successful preclinical assessment, a phase I study was conducted on 9 patients with primary and metastatic CRC. Study participants received 151.21 ± 25.25 MBq of intravenous [68 Ga]Ga-HNI01 and underwent PET/CT scans at 1 h and 2 h post injection. Patients 01-03 also underwent whole-body dynamic PET imaging within 0-40 min p.i. All patients underwent [18F]F-FDG PET/CT imaging within 1 week after [68 Ga]Ga-HNI01 imaging. Tracer distribution, pharmacokinetics, and radiation dosimetry were calculated. RESULTS [68 Ga]Ga-HNI01 was successfully synthesized within 10 min under mild conditions, and the radiochemical purity was more than 98% without purification. Micro-PET imaging with [68 Ga]Ga-HNI01 revealed clear visualization of LS174T tumors, while signals from HT-29 tumors were significantly lower. Biodistribution studies indicated that uptake of [68 Ga]Ga-HNI01 in LS174T and HT-29 was 8.83 ± 3.02%ID/g and 1.81 ± 0.87%ID/g, respectively, at 2 h p.i. No adverse events occurred in all clinical participants after the injection of [68 Ga]Ga-HNI01. A fast blood clearance and low background uptake were observed, and CRC lesions could be visualized with high contrast as early as 30 min after injection. [68 Ga]Ga-HNI01 PET could clearly detect metastatic lesions in the liver, lung, and pancreas and showed superior ability in detecting small metastases. A significant accumulation of radioactivity was observed in the kidney, and normal tissues physiologically expressing CEA receptors showed slight uptakes of [68 Ga]Ga-HNI01. An interesting finding was that strong uptake of [68 Ga]Ga-HNI01 was found in non-malignant colorectal tissues adjacent to the primary tumor in some patients, suggesting abnormal CEA expression in these healthy tissues. CONCLUSION [68 Ga]Ga-HNI01 is a novel CEA-targeted PET imaging radiotracer with excellent pharmacokinetics and favorable dosimetry profiles. [68 Ga]Ga-HNI01 PET is an effective and convenient imaging tool for detecting CRC lesions, particularly for identifying small metastases. Furthermore, its high specificity for CEA in vivo makes it an ideal tool for selecting patients for anti-CEA therapy.
Collapse
Affiliation(s)
- Liqiang Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing, 100142, China
| | - Xinfeng Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing, 100142, China
| | - Lin Wang
- Department of Gastrointestinal Cancer Centre, Unit III, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing, China
| | - Xiaopan Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing, 100142, China
| | - Ziqing Zeng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing, 100142, China
| | - Futao Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing, 100142, China
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing, 100142, China.
| | - Aiwen Wu
- Department of Gastrointestinal Cancer Centre, Unit III, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing, China.
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing, 100142, China.
| |
Collapse
|
31
|
Li S, Hoefnagel SJM, Krishnadath KK. Single domain Camelid antibody fragments for molecular imaging and therapy of cancer. Front Oncol 2023; 13:1257175. [PMID: 37746282 PMCID: PMC10514897 DOI: 10.3389/fonc.2023.1257175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Despite innovations in cancer therapeutics, cancer remains associated with high mortality and is one of biggest health challenges worldwide. Therefore, developing precise cancer imaging and effective treatments is an unmet clinical need. A relatively novel type of therapeutics are heavy chain variable domain antibody fragments (VHHs) derived from llamas. Here, we explored the suitability of VHHs for cancer imaging and therapy through reviewing the existing literature. We searched the MEDLINE, EMBASE and Cochrane databases and identified 32 papers on molecular imaging and 41 papers on therapy that were suitable for comprehensive reviewing. We found that VHHs harbor a higher specificity and affinity compared to mAbs, which contributes to high-quality imaging and less side-effects on healthy cells. The employment of VHHs in cancer imaging showed remarkably shorter times between administration and imaging. Studies showed that 18F and 99mTc are two optimal radionuclides for imaging with VHHs and that site-specific labelling is the optimal conjugation modality for VHHs with radionuclide or fluorescent molecules. We found different solutions for reducing kidney retention and immunogenicity of VHHs. VHHs as anticancer therapeutics have been tested in photodynamic therapy, targeted radionuclide therapy, immunotherapy and molecular targeted therapy. These studies showed that VHHs target unique antigen epitopes, which are distinct from the ones recognized by mAbs. This advantage means that VHHs may be more effective for targeted anticancer therapy and can be combined with mAbs. We found that high cellular internalization and specificity of VHHs contributes to the effectiveness and safety of VHHs as anticancer therapeutics. Two clinical trials have confirmed that VHHs are effective and safe for cancer imaging and therapy. Together, VHHs seem to harbor several advantages compared to mAbs and show potential for application in personalized treatment for cancer patients. VHH-based imaging and therapy are promising options for improving outcomes of cancer patients.
Collapse
Affiliation(s)
- Shulin Li
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam, Netherlands
| | | | - Kausilia Krishnawatie Krishnadath
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
32
|
Ducharme M, Hall L, Eckenroad W, Cingoranelli SJ, Houson HA, Jaskowski L, Hunter C, Larimer BM, Lapi SE. Evaluation of [ 89Zr]Zr-DFO-2Rs15d Nanobody for Imaging of HER2-Positive Breast Cancer. Mol Pharm 2023; 20:4629-4639. [PMID: 37552575 PMCID: PMC11606513 DOI: 10.1021/acs.molpharmaceut.3c00360] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
One of the most aggressive forms of breast cancer involves the overexpression of human epidermal growth factor receptor 2 (HER2). HER2 is overexpressed in ∼25% of all breast cancers and is associated with increased proliferation, increased rates of metastasis, and poor prognosis. Treatment for HER2-positive breast cancer has vastly improved since the development of the monoclonal antibody trastuzumab (Herceptin) as well as other biological constructs. However, patients still commonly develop resistance, illustrating the need for newer therapies. Nanobodies have become an important focus for potential development as HER2-targeting imaging agents and therapeutics. Nanobodies have many favorable characteristics, including high stability in heat and nonphysiological pH, while maintaining their low-nanomolar affinity for their designed targets. Specifically, the 2Rs15d nanobody has been developed for targeting HER2 and has been evaluated as a diagnostic imaging agent for single-photon emission computed tomography (SPECT) and positron emission tomography (PET). While a construct of 2Rs15d with the positron emitter 68Ga is currently in phase I clinical trials, the only PET images acquired in preclinical or clinical research have been within 3 h postinjection. We evaluated our in-house produced 2Rs15d nanobody, conjugated with the chelator deferoxamine (DFO), and radiolabeled with 89Zr for PET imaging up to 72 h postinjection. [89Zr]Zr-DFO-2Rs15d demonstrated high stability in both phosphate-buffered saline (PBS) and human serum. Cell binding studies showed high binding and specificity for HER2, as well as prominent internalization. Our in vivo PET imaging confirmed high-quality visualization of HER2-positive tumors up to 72 h postinjection, whereas HER2-negative tumors were not visualized. Subsequent biodistribution studies quantitatively supported the significant HER2-positive tumor uptake compared to the negative control. Our studies fill an important gap in understanding the imaging and binding properties of the 2Rs15d nanobody at extended time points. As many therapeutic radioisotopes have single or multiday half-lives, this information will directly benefit the potential of the radiotherapy development of 2Rs15d for HER2-positive breast cancer patients.
Collapse
Affiliation(s)
- Maxwell Ducharme
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Lucinda Hall
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Whitney Eckenroad
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Shelbie J Cingoranelli
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Hailey A Houson
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Luke Jaskowski
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Chanelle Hunter
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Benjamin M Larimer
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Suzanne E Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| |
Collapse
|
33
|
Tang H, Gao Y, Han J. Application Progress of the Single Domain Antibody in Medicine. Int J Mol Sci 2023; 24:ijms24044176. [PMID: 36835588 PMCID: PMC9967291 DOI: 10.3390/ijms24044176] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/07/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
The camelid-derived single chain antibody (sdAb), also termed VHH or nanobody, is a unique, functional heavy (H)-chain antibody (HCAb). In contrast to conventional antibodies, sdAb is a unique antibody fragment consisting of a heavy-chain variable domain. It lacks light chains and a first constant domain (CH1). With a small molecular weight of only 12~15 kDa, sdAb has a similar antigen-binding affinity to conventional Abs but a higher solubility, which exerts unique advantages for the recognition and binding of functional, versatile, target-specific antigen fragments. In recent decades, with their unique structural and functional features, nanobodies have been considered promising agents and alternatives to traditional monoclonal antibodies. As a new generation of nano-biological tools, natural and synthetic nanobodies have been used in many fields of biomedicine, including biomolecular materials, biological research, medical diagnosis and immune therapies. This article briefly overviews the biomolecular structure, biochemical properties, immune acquisition and phage library construction of nanobodies and comprehensively reviews their applications in medical research. It is expected that this review will provide a reference for the further exploration and unveiling of nanobody properties and function, as well as a bright future for the development of drugs and therapeutic methods based on nanobodies.
Collapse
Affiliation(s)
- Huaping Tang
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Yuan Gao
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
- Correspondence:
| | - Jiangyuan Han
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
34
|
Qin X, Guo X, Liu T, Li L, Zhou N, Ma X, Meng X, Liu J, Zhu H, Jia B, Yang Z. High in-vivo stability in preclinical and first-in-human experiments with [ 18F]AlF-RESCA-MIRC213: a 18F-labeled nanobody as PET radiotracer for diagnosis of HER2-positive cancers. Eur J Nucl Med Mol Imaging 2023; 50:302-313. [PMID: 36129493 DOI: 10.1007/s00259-022-05967-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 09/11/2022] [Indexed: 01/10/2023]
Abstract
PURPOSE [18F]AlF-RESCA was introduced as a core particularly useful for 18F-labeling of heat-sensitive biomolecules. However, no translational studies have been reported up to now. Herein, we reported the first-in-human evaluation of an 18F-labeled anti-HER2 nanobody MIRC213 as a PET radiotracer for imaging HER2-positive cancers. METHODS MIRC213 was produced by E. coli and conjugated with ( ±)-H3RESCA-Mal. [18F]AlF-RESCA-MIRC213 was prepared at room temperature. Its radiochemical purity and stability of were determined by radio-HPLC with the size-exclusion chromatographic column. Cell uptake was performed in NCI-N87 (HER2 +) and MCF-7 (HER2-) cells and the cell-binding affinity was verified in SK-OV-3 (HER2 +) cells. Small-animal PET/CT was performed using SK-OV-3, NCI-N87, and MCF-7 tumor-bearing mice at 30 min, 1 h, and 2 h post-injection. For blocking experiment, excess MIRC213 was co-injected with radiotracer. Biodistribution were performed on SKOV-3 and MCF-7 tumor-bearing mice at 2 h post-injection. For clinical study, PET/CT images were acquired at 2 h and 4 h after injection of [18F]AlF-RESCA-MIRC213 (1.85-3.7 MBq/kg) in six breast cancer patients (3 HER2-positive and 3 HER2-negative). All patients underwent [18F]-FDG PET/CT within a week for tissue selection purpose. Distribution and dosimetry were calculated. Standardized uptake values (SUV) were measured in tumors and normal organs. RESULTS MIRC213 was produced with > 95% purity and modified with RESCA to obtain RESCA-MIRC213. [18F]AlF-RESCA-MIRC213 was prepared within 20 min at room temperature with the radiochemical yield of 50.48 ± 7.6% and radiochemical purity of > 98% (n > 10), and remained stable in both PBS (88%) and 5% HSA (92%) after 6 h. The 2 h cellular uptake of [18F]AlF-RESCA-MIRC213 in NCI-N87 cells was 11.22 ± 0.60 AD%/105 cells. Its binding affinity Kd value was determined to be 1.23 ± 0.58 nM. Small-animal PET/CT with [18F]AlF-RESCA-MIRC213 can clearly differentiate SK-OV-3 and NCI-N87 tumors from MCF-7 tumors and background with a high uptake of 4.73 ± 1.18 ID%/g and substantially reduced to 1.70 ± 0.13 ID%/g for the blocking group (p < 0.05) in SK-OV-3 tumors at 2 h post-injection. No significant bone radioactivity was seen in the tumor-bearing animals. In all six breast cancer patients, there was no adverse reaction during study. The uptake of [18F]AlF-RESCA-MIRC213 was mainly in lacrimal gland, parotid gland, submandibular gland, thyroid gland, gallbladder, kidneys, liver, and intestines. There was no significant bone radioactivity accumulation in cancer patients. [18F]AlF-RESCA-MIRC213 had significantly higher tumor uptake in lesions from HER2-positive patients than that lesions from HER2-negative patients (SUVmax of 3.62 ± 1.56 vs. 1.41 ± 0.41, p = 0.0012) at 2 h post-injection. The kidneys received the highest radiation dose of 2.42 × 10-1 mGy/MBq, and the effective dose was 1.56 × 10-2 mSv/MBq. CONCLUSIONS [18F]AlF-RESCA-MIRC213 could be prepared with high radiolabeling yield under mild conditions. [18F]AlF-RESCA-MIRC213 has relatively high stability both in vitro and in vivo. The results from clinical transformation suggest that [18F]AlF-RESCA-MIRC213 PET/CT is a safe procedure with favorable pharmacokinetics and dosimetry profile, and it is a promising new PET radiotracer for noninvasive diagnosis of HER2-positive cancers.
Collapse
Affiliation(s)
- Xue Qin
- Guizhou University School of Medicine, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaoyi Guo
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Tianyu Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Liqiang Li
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Nina Zhou
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaopan Ma
- Guizhou University School of Medicine, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jiayue Liu
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Hua Zhu
- Guizhou University School of Medicine, Guizhou University, Guiyang, 550025, China.
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Zhi Yang
- Guizhou University School of Medicine, Guizhou University, Guiyang, 550025, China.
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
35
|
Guilbaud A, Pecorari F. Construction of Synthetic VHH Libraries in Ribosome Display Format. Methods Mol Biol 2023; 2681:19-31. [PMID: 37405640 DOI: 10.1007/978-1-0716-3279-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
Single-domain antibodies, or VHH, represent an attractive molecular basis to design affinity proteins with favorable properties. Beyond high affinity and specificity for their cognate target, they usually show high stability and high production yields in bacteria, yeast, or mammalian cells. In addition to these favorable properties, their ease of engineering makes them useful for many applications. Until the past few years, the generation of VHH involved the immunization of a Camelidae with the target antigen, followed by a phage display selection using phage libraries encoding the VHH repertoire of the animal blood sample. However, this approach is constrained by the accessibility to the animals, and the output relies on the animal's immune system.Recently, synthetic VHH libraries have been designed to avoid the use of animals. Here, we describe the construction of VHH combinatorial libraries and their use for the selection of binders by ribosome display, a fully in vitro selection technique.
Collapse
Affiliation(s)
- Audrey Guilbaud
- Nantes Université, Univ Angers, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302/EMR6001, Nantes, France
| | - Frédéric Pecorari
- Nantes Université, Univ Angers, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302/EMR6001, Nantes, France.
| |
Collapse
|
36
|
Zhao L, Gong J, Qi Q, Liu C, Su H, Xing Y, Zhao J. 131I-Labeled Anti-HER2 Nanobody for Targeted Radionuclide Therapy of HER2-Positive Breast Cancer. Int J Nanomedicine 2023; 18:1915-1925. [PMID: 37064291 PMCID: PMC10094415 DOI: 10.2147/ijn.s399322] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 04/04/2023] [Indexed: 04/18/2023] Open
Abstract
Purpose The unique structure of nanobodies is advantageous for the development of radiopharmaceuticals for nuclear medicine. Nanobodies targeted to human epidermal growth factor receptor 2 (HER2) can be used as tools for the imaging and therapy of HER2-overexpressing tumors. In this study, we aimed to describe the generation of a 131I-labeled anti-HER2 nanobody as a targeted radionuclide therapy (TRNT) agent for HER2-positive breast cancer. Methods The anti-HER2 nanobody NM-02 was labeled with 131I using the iodogen method, and its radiochemical purity and stability in vitro were assessed. The pharmacokinetic profile of 131I-NM-02 was investigated in normal mice. Tumor accumulation, biodistribution, and therapeutic potential of 131I-NM-02 were evaluated in HER2-positive SKBR3 xenografts; HER2-negative MB-MDA-231 xenografts were used as the control group. Results 131I-NM-02 could be readily prepared with satisfactory radiochemical purity and stability in vitro. Apparent tumor uptake was observed in HER2-positive tumor-bearing mice with rapid blood clearance and favorable biodistribution. 131I-NM-02 could significantly inhibit tumor growth and extend the life of these mice with good organ compatibility. Negligible tumor accumulation and inhibitory effects of 131I-NM-02 were observed in the negative control group. Conclusion 131I-NM-02 has the potential to be explored as a novel tool for TRNT of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jiali Gong
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Qinli Qi
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Changcun Liu
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Hongxing Su
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yan Xing
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Correspondence: Jinhua Zhao; Yan Xing, Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Road, Shanghai, 200080, People’s Republic of China, Tel/Fax +86 21 3779 8352, Email ;
| |
Collapse
|
37
|
Rodak M, Dekempeneer Y, Wojewódzka M, Caveliers V, Covens P, Miller BW, Sevenois MB, Bruchertseifer F, Morgenstern A, Lahoutte T, D'Huyvetter M, Pruszyński M. Preclinical Evaluation of 225Ac-Labeled Single-Domain Antibody for the Treatment of HER2pos Cancer. Mol Cancer Ther 2022; 21:1835-1845. [PMID: 36129807 PMCID: PMC9716241 DOI: 10.1158/1535-7163.mct-21-1021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/25/2022] [Accepted: 09/16/2022] [Indexed: 01/12/2023]
Abstract
Human epidermal growth factor receptor type 2 (HER2) is overexpressed in various cancers; thus, HER2-targeting single-domain antibodies (sdAb) could offer a useful platform for radioimmunotherapy. In this study, we optimized the labeling of an anti-HER2-sdAb with the α-particle-emitter 225Ac through a DOTA-derivative. The formed radioconjugate was tested for binding affinity, specificity and internalization properties, whereas cytotoxicity was evaluated by clonogenic and DNA double-strand-breaks assays. Biodistribution studies were performed in mice bearing subcutaneous HER2pos tumors to estimate absorbed doses delivered to organs and tissues. Therapeutic efficacy and potential toxicity were assessed in HER2pos intraperitoneal ovarian cancer model and in healthy C57Bl/6 mice. [225Ac]Ac-DOTA-2Rs15d exhibited specific cell uptake and cell-killing capacity in HER2pos cells (EC50 = 3.9 ± 1.1 kBq/mL). Uptake in HER2pos lesions peaked at 3 hours (9.64 ± 1.69% IA/g), with very low accumulation in other organs (<1% IA/g) except for kidneys (11.69 ± 1.10% IA/g). α-camera imaging presented homogeneous uptake of radioactivity in tumors, although heterogeneous in kidneys, with a higher signal density in cortex versus medulla. In mice with HER2pos disseminated tumors, repeated administration of [225Ac]Ac-DOTA-2Rs15d significantly prolonged survival (143 days) compared to control groups (56 and 61 days) and to the group treated with HER2-targeting mAb trastuzumab (100 days). Histopathologic evaluation revealed signs of kidney toxicity after repeated administration of [225Ac]Ac-DOTA-2Rs15d. [225Ac]Ac-DOTA-2Rs15d efficiently targeted HER2pos cells and was effective in treatment of intraperitoneal disseminated tumors, both alone and as an add-on combination with trastuzumab, albeit with substantial signs of inflammation in kidneys. This study warrants further development of [225Ac]Ac-DOTA-2Rs15d.
Collapse
Affiliation(s)
- Magdalena Rodak
- Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - Yana Dekempeneer
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Vicky Caveliers
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Peter Covens
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Brian W. Miller
- Department of Medical Imaging, University of Arizona, Tucson, Arizona
| | - Matthijs B. Sevenois
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | - Tony Lahoutte
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Matthias D'Huyvetter
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marek Pruszyński
- Institute of Nuclear Chemistry and Technology, Warsaw, Poland
- NOMATEN Centre of Excellence, National Centre for Nuclear Research, Otwock, Poland
| |
Collapse
|
38
|
Evaluation of 68Ga-Radiolabeled Peptides for HER2 PET Imaging. Diagnostics (Basel) 2022; 12:diagnostics12112710. [PMID: 36359554 PMCID: PMC9689602 DOI: 10.3390/diagnostics12112710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/25/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
One in eight women will be diagnosed with breast cancer in their lifetime and approximately 25% of those cases will be HER2-positive. Current methods for diagnosing HER2-positive breast cancer involve using IHC and FISH from suspected cancer biopsies to quantify HER2 expression. HER2 PET imaging could potentially increase accuracy and improve the diagnosis of lesions that are not available for biopsies. Using two previously discovered HER2-targeting peptides, we modified each peptide with the chelator DOTA and a PEG2 linker resulting in DOTA-PEG2-GSGKCCYSL (P5) and DOTA-PEG2-DTFPYLGWWNPNEYRY (P6). Each peptide was labeled with 68Ga and was evaluated for HER2 binding using in vitro cell studies and in vivo tumor xenograft models. Both [68Ga]P5 and [68Ga]P6 showed significant binding to HER2-positive BT474 cells versus HER2-negative MDA-MB-231 cells ([68Ga]P5; 0.68 ± 0.20 versus 0.47 ± 0.05 p < 0.05 and [68Ga]P6; 0.55 ± 0.21 versus 0.34 ± 0.12 p < 0.01). [68Ga]P5 showed a higher percent injected dose per gram (%ID/g) binding to HER2-positive tumors two hours post-injection compared to HER2-negative tumors (0.24 ± 0.04 versus 0.12 ± 0.06; p < 0.05), while the [68Ga]P6 peptide showed significant binding (0.98 ± 0.22 versus 0.51 ± 0.08; p < 0.05) one hour post-injection. These results lay the groundwork for the use of peptides to image HER2-positive breast cancer.
Collapse
|
39
|
Lugat A, Bailly C, Chérel M, Rousseau C, Kraeber-Bodéré F, Bodet-Milin C, Bourgeois M. Immuno-PET: Design options and clinical proof-of-concept. Front Med (Lausanne) 2022; 9:1026083. [PMID: 36314010 PMCID: PMC9613928 DOI: 10.3389/fmed.2022.1026083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/29/2022] [Indexed: 11/23/2022] Open
Abstract
Radioimmunoconjugates have been used for over 30 years in nuclear medicine applications. In the last few years, advances in cancer biology knowledge have led to the identification of new molecular targets specific to certain patient subgroups. The use of these targets in targeted therapies approaches has allowed the developments of specifically tailored therapeutics for patients. As consequence of the PET-imaging progresses, nuclear medicine has developed powerful imaging tools, based on monoclonal antibodies, to in vivo characterization of these tumor biomarkers. This imaging modality known as immuno-positron emission tomography (immuno-PET) is currently in fastest-growing and its medical value lies in its ability to give a non-invasive method to assess the in vivo target expression and distribution and provide key-information on the tumor targeting. Currently, immuno-PET presents promising probes for different nuclear medicine topics as staging/stratification tool, theranostic approaches or predictive/prognostic biomarkers. To develop a radiopharmaceutical drug that can be used in immuno-PET approach, it is necessary to find the best compromise between the isotope choice and the immunologic structure (full monoclonal antibody or derivatives). Through some clinical applications, this paper review aims to discuss the most important aspects of the isotope choice and the usable proteic structure that can be used to meet the clinical needs.
Collapse
Affiliation(s)
- Alexandre Lugat
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France
| | - Clément Bailly
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Michel Chérel
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Department of Nuclear Medicine, Institut de Cancérologie de l'Ouest (ICO) – Site Gauducheau, Saint-Herblain, France
| | - Caroline Rousseau
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Department of Nuclear Medicine, Institut de Cancérologie de l'Ouest (ICO) – Site Gauducheau, Saint-Herblain, France
| | - Françoise Kraeber-Bodéré
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Caroline Bodet-Milin
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Mickaël Bourgeois
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France,ARRONAX Cyclotron, Saint-Herblain, France,*Correspondence: Mickaël Bourgeois
| |
Collapse
|
40
|
Ebrahimi F, Noaparast Z, Abedi SM, Hosseinimehr SJ. Homodimer 99mTc-HYNIC-E(SSSLTVPWY) 2 peptide improved HER2-overexpressed tumor targeting and imaging. Med Oncol 2022; 39:204. [PMID: 36175805 DOI: 10.1007/s12032-022-01798-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/15/2022] [Indexed: 06/16/2023]
Abstract
We hypothesized that a novel design of the LTVPWY (LY) peptide might exhibit a great potential for improving binding affinity and targeting HER2-overexpressed tumors. Hence, new dimer construction of 99mTc-labeled LY [99mTc-HYNIC-E(SSSLTVPWY)2] (99mTc-DLY) was introduced. Afterward, a head-to-head comparison of in vitro and in vivo experiments was performed between 99mTc-DLY and 99mTc-HYNIC-SSSLTVPWY as the monomer analog. The blocking dosage of trastuzumab reduced the uptake of the dimer about 20% more efficiently than the monomer in the SKOV-3 cell line. A twofold increase in competitive binding affinity and biological half-life was observed for 99mTc-DLY. The ovarian-tumor-bearing mice were detected with high contrast where the tumor-to-muscle ratio of 99mTc-DLY was notably increased about 40% using a gamma camera. The biodistribution experiment revealed an approximately 10% enhancement in tumor/blood, tumor/muscle, and tumor/bone ratios for the dimer. More rapid blood clearance was another achievement of the homodimer design. Overall, 99mTc-DLY successfully affected the pharmacokinetics and consequently the visualization of HER2-overexpressing tumors.
Collapse
Affiliation(s)
- Fatemeh Ebrahimi
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zohreh Noaparast
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mohammad Abedi
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
41
|
Uccelli L, Martini P, Urso L, Ghirardi T, Marvelli L, Cittanti C, Carnevale A, Giganti M, Bartolomei M, Boschi A. Rhenium Radioisotopes for Medicine, a Focus on Production and Applications. Molecules 2022; 27:5283. [PMID: 36014521 PMCID: PMC9412410 DOI: 10.3390/molecules27165283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/08/2022] [Accepted: 08/17/2022] [Indexed: 11/28/2022] Open
Abstract
In recent decades, the use of alpha; pure beta; or beta/gamma emitters in oncology, endocrinology, and interventional cardiology rheumatology, has proved to be an important alternative to the most common therapeutic regimens. Among radionuclides used for therapy in nuclear medicine, two rhenium radioisotopes are of particular relevance: rhenium-186 and rhenium-188. The first is routinely produced in nuclear reactors by direct neutron activation of rhenium-186 via 185Re(n,γ)186Re nuclear reaction. Rhenium-188 is produced by the decay of the parent tungsten-188. Separation of rhenium-188 is mainly performed using a chromatographic 188W/188Re generator in which tungsten-188 is adsorbed on the alumina column, similar to the 99Mo/99mTc generator system, and the radionuclide eluted in saline solution. The application of rhenium-186 and rhenium-188 depends on their specific activity. Rhenium-186 is produced in low specific activity and is mainly used for labeling particles or diphosphonates for bone pain palliation. Whereas, rhenium-188 of high specific activity can be used for labeling peptides or bioactive molecules. One of the advantages of rhenium is its chemical similarity with technetium. So, diagnostic technetium analogs labeled with radiorhenium can be developed for therapeutic applications. Clinical trials promoting the use of 186/188Re-radiopharmaceuticals is, in particular, are discussed.
Collapse
Affiliation(s)
- Licia Uccelli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy
| | - Petra Martini
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Luca Urso
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy
| | - Teresa Ghirardi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Lorenza Marvelli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Corrado Cittanti
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy
| | - Aldo Carnevale
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Radiology Unit, University Hospital, 44124 Ferrara, Italy
| | - Melchiore Giganti
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Radiology Unit, University Hospital, 44124 Ferrara, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy
| | - Alessandra Boschi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
42
|
Wang Y, Wang C, Huang M, Qin S, Zhao J, Sang S, Zheng M, Bian Y, Huang C, Zhang H, Guo L, Jiang J, Xu C, Dai N, Zheng Y, Han J, Yang M, Xu T, Miao L. Pilot study of a novel nanobody 68 Ga-NODAGA-SNA006 for instant PET imaging of CD8 + T cells. Eur J Nucl Med Mol Imaging 2022; 49:4394-4405. [PMID: 35829748 DOI: 10.1007/s00259-022-05903-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/30/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Positron emission tomography (PET) with specific diagnostic probes for quantifying CD8+ T cells has emerged as a powerful technique for monitoring the immune response. However, most CD8+ T cell radiotracers are based on antibodies or antibody fragments, which are slowly cleared from circulation. Herein, we aimed to develop and assess 68 Ga-NODAGA-SNA006 for instant PET (iPET) imaging of CD8+ T cells. METHODS A novel nanobody without a hexahistidine (His6) tag, SNA006-GSC, was designed, site-specifically conjugated with NODAGA-maleimide and radiolabelled with 68 Ga. The PET imaging profiles of 68 Ga-NODAGA-SNA006 were evaluated in BALB/c MC38-CD8+/CD8- tumour models and cynomolgus monkeys. Three volunteers with lung cancer underwent whole-body PET/CT imaging after 68 Ga-NODAGA-SNA006 administration. The biodistribution, pharmacokinetics and dosimetry of patients were also investigated. In addition, combined with immunohistochemistry (IHC), the quantitative performance of the tracer for monitoring CD8 expression was evaluated in BALB/c MC38-CD8+/CD8- and human subjects. RESULTS 68 Ga-NODAGA-SNA006 was prepared with RCP > 98% and SA > 100 GBq/μmol. 68 Ga-NODAGA-SNA006 exhibited specific uptake in MC38-CD8+ xenografts tumours, CD8-rich tissues (such as the spleen) in monkeys and CD8+ tumour lesions in patients within 1 h. Fast washout from circulation was observed in three volunteers (t1/2 < 20 min). A preliminary quantitative linear relationship (R2 = 0.9668, p < 0.0001 for xenografts and R2 = 0.7924, p = 0.0013 for lung patients) appeared between 68 Ga-NODAGA-SNA006 uptake and CD8 expression. 68 Ga-NODAGA-SNA006 was well tolerated by all patients. CONCLUSION 68 Ga-NODAGA-SNA006 PET imaging can instantly quantify CD8 expression with an ideal safety profile and is expected to be important for dynamically tracking CD8+ T cells and monitoring immune responses for individualised cancer immunotherapy. TRIAL REGISTRATION NCT05126927 (19 November 2021, retrospectively registered).
Collapse
Affiliation(s)
- Yan Wang
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Chao Wang
- Smart-Nuclide Biotech, No. 218 Xing-Hu Rd., Suzhou, 215125, Jiangsu, China
| | - Minzhou Huang
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Songbing Qin
- Department of Radiotherapy, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shibiao Sang
- Department of Nuclear Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Meng Zheng
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yicong Bian
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Chenrong Huang
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Hua Zhang
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Lingchuan Guo
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiwei Jiang
- Department of Nuclear Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Chun Xu
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Na Dai
- Department of Nuclear Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yushuang Zheng
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiajun Han
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Yang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, No. 20 Qian-Rong Rd., Wuxi, 214063, Jiangsu, China.
| | - Tao Xu
- Smart-Nuclide Biotech, No. 218 Xing-Hu Rd., Suzhou, 215125, Jiangsu, China.
| | - Liyan Miao
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China. .,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
43
|
Shoari A, Tahmasebi M, Khodabakhsh F, Cohan RA, Oghalaie A, Behdani M. Angiogenic biomolecules specific nanobodies application in cancer imaging and therapy; review and updates. Int Immunopharmacol 2022; 105:108585. [DOI: 10.1016/j.intimp.2022.108585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 11/05/2022]
|
44
|
Wang S, Gai Y, Sun L, Lan X, Zeng D, Xiang G, Ma X. Synthesis and evaluation of novel 1,4,7-triazacyclononane derivatives as Cu2+ and Ga3+ chelators. J Inorg Biochem 2022; 229:111719. [DOI: 10.1016/j.jinorgbio.2022.111719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/31/2021] [Accepted: 01/02/2022] [Indexed: 12/25/2022]
|
45
|
Yang E, Liu Q, Huang G, Liu J, Wei W. Engineering nanobodies for next-generation molecular imaging. Drug Discov Today 2022; 27:1622-1638. [PMID: 35331925 DOI: 10.1016/j.drudis.2022.03.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 02/04/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022]
Abstract
In recent years, nanobodies have emerged as ideal imaging agents for molecular imaging. Molecular nanobody imaging combines the specificity of nanobodies with the sensitivity of state-of-the-art molecular imaging modalities, such as positron emission tomography (PET). Given that modifications of nanobodies alter their pharmacokinetics (PK), the engineering strategies that combine nanobodies with radionuclides determine the effectiveness, reliability, and safety of the molecular imaging probes. In this review, we introduce conjugation strategies that have been applied to nanobodies, including random conjugation, 99mTc tricarbonyl chemistry, sortase A-mediated site-specific conjugation, maleimide-cysteine chemistry, and click chemistries. We also summarize the latest advances in nanobody tracers, emphasizing their preclinical and clinical use. In addition, we elaborate on nanobody-based near-infrared fluorescence (NIRF) imaging and image-guided surgery.
Collapse
Affiliation(s)
- Erpeng Yang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Qiufang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| |
Collapse
|
46
|
Barakat S, Berksöz M, Zahedimaram P, Piepoli S, Erman B. Nanobodies as molecular imaging probes. Free Radic Biol Med 2022; 182:260-275. [PMID: 35240292 DOI: 10.1016/j.freeradbiomed.2022.02.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 12/12/2022]
Abstract
Camelidae derived single-domain antibodies (sdAbs), commonly known as nanobodies (Nbs), are the smallest antibody fragments with full antigen-binding capacity. Owing to their desirable properties such as small size, high specificity, strong affinity, excellent stability, and modularity, nanobodies are on their way to overtake conventional antibodies in terms of popularity. To date, a broad range of nanobodies have been generated against different molecular targets with applications spanning basic research, diagnostics, and therapeutics. In the field of molecular imaging, nanobody-based probes have emerged as a powerful tool. Radioactive or fluorescently labeled nanobodies are now used to detect and track many targets in different biological systems using imaging techniques. In this review, we provide an overview of the use of nanobodies as molecular probes. Additionally, we discuss current techniques for the generation, conjugation, and intracellular delivery of nanobodies.
Collapse
Affiliation(s)
- Sarah Barakat
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Tuzla, Istanbul, Turkey.
| | - Melike Berksöz
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Tuzla, Istanbul, Turkey.
| | - Pegah Zahedimaram
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Tuzla, Istanbul, Turkey.
| | - Sofia Piepoli
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bogazici University, 34342, Bebek, Istanbul, Turkey.
| | - Batu Erman
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bogazici University, 34342, Bebek, Istanbul, Turkey.
| |
Collapse
|
47
|
Feng Y, Meshaw R, McDougald D, Zhou Z, Zhao XG, Jannetti SA, Reiman RE, Pippen E, Marjoram R, Schaal JL, Vaidyanathan G, Zalutsky MR. Evaluation of an 131I-labeled HER2-specific single domain antibody fragment for the radiopharmaceutical therapy of HER2-expressing cancers. Sci Rep 2022; 12:3020. [PMID: 35194100 PMCID: PMC8864007 DOI: 10.1038/s41598-022-07006-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
Radiopharmaceutical therapy (RPT) is an attractive strategy for treatment of disseminated cancers including those overexpressing the HER2 receptor including breast, ovarian and gastroesophageal carcinomas. Single-domain antibody fragments (sdAbs) exemplified by the HER2-targeted VHH_1028 evaluated herein are attractive for RPT because they rapidly accumulate in tumor and clear faster from normal tissues than intact antibodies. In this study, VHH_1028 was labeled using the residualizing prosthetic agent N-succinimidyl 3-guanidinomethyl 5-[131I]iodobenzoate (iso-[131I]SGMIB) and its tissue distribution evaluated in the HER2-expressing SKOV-3 ovarian and BT474 breast carcinoma xenograft models. In head-to-head comparisons to [131I]SGMIB-2Rs15d, a HER2-targeted radiopharmaceutical currently under clinical investigation, iso-[131I]SGMIB-VHH_1028 exhibited significantly higher tumor uptake and significantly lower kidney accumulation. The results demonstrated 2.9 and 6.3 times more favorable tumor-to-kidney radiation dose ratios in the SKOV-3 and BT474 xenograft models, respectively. Iso-[131I]SGMIB-VHH_1028 was prepared using a solid-phase extraction method for purification of the prosthetic agent intermediate Boc2-iso-[131I]SGMIB that reproducibly scaled to therapeutic-level doses and obviated the need for its HPLC purification. Single-dose (SKOV-3) and multiple-dose (BT474) treatment regimens demonstrated that iso-[131I]SGMIB-VHH_1028 was well tolerated and provided significant tumor growth delay and survival prolongation. This study suggests that iso-[131I]SGMIB-VHH_1028 is a promising candidate for RPT of HER2-expressing cancers and further development is warranted.
Collapse
Affiliation(s)
- Yutian Feng
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Rebecca Meshaw
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Darryl McDougald
- Department of Radiology, Duke University Medical Center, Durham, NC, USA.,Cereius Inc, Durham, NC, USA
| | - Zhengyuan Zhou
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Xiao-Guang Zhao
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Stephen A Jannetti
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Robert E Reiman
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
48
|
Oliveira MC, Correia JDG. Clinical application of radioiodinated antibodies: where are we? Clin Transl Imaging 2022. [DOI: 10.1007/s40336-021-00477-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
49
|
Rafidi H, Rajan S, Urban K, Shatz-Binder W, Hui K, Ferl GZ, Kamath AV, Boswell CA. Effect of molecular size on interstitial pharmacokinetics and tissue catabolism of antibodies. MAbs 2022; 14:2085535. [PMID: 35867780 PMCID: PMC9311319 DOI: 10.1080/19420862.2022.2085535] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Advances in antibody engineering have enabled the construction of novel molecular formats in diverse shapes and sizes, providing new opportunities for biologic therapies and expanding the need to understand how various structural aspects affect their distribution properties. To assess the effect of antibody size on systemic pharmacokinetics (PK) and tissue distribution with or without neonatal Fc receptor (FcRn) binding, we evaluated a series of non-mouse-binding anti-glycoprotein D monoclonal antibody formats, including IgG [~150 kDa], one-armed IgG [~100 kDa], IgG-HAHQ (attenuated FcRn binding) [~150 kDa], F(ab')2 [~100 kDa], and F(ab) [~50 kDa]. Tissue-specific concentration-time profiles were corrected for blood content based on vascular volumes and normalized based on interstitial volumes to allow estimation of interstitial concentrations and interstitial:serum concentration ratios. Blood correction demonstrated that the contribution of circulating antibody on total uptake was greatest at early time points and for highly vascularized tissues. Tissue interstitial PK largely mirrored serum exposure profiles. Similar interstitial:serum ratios were obtained for the two FcRn-binding molecules, IgG and one-armed IgG, which reached pseudo-steady-state kinetics in most tissues. For non-FcRn-binding molecules, interstitial:serum ratios changed over time, suggesting that these molecules did not reach steady-state kinetics during the study. Furthermore, concentration-time profiles of both intact and catabolized molecule were measured by a dual tracer approach, enabling quantification of tissue catabolism and demonstrating that catabolism levels were highest for IgG-HAHQ. Overall, these data sets provide insight into factors affecting preclinical distribution and may be useful in estimating interstitial concentrations and/or catabolism in human tissues.
Collapse
Affiliation(s)
- Hanine Rafidi
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Sharmila Rajan
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Konnie Urban
- Safety Assessment, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Whitney Shatz-Binder
- Protein Chemistry, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Keliana Hui
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Gregory Z Ferl
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA.,Biomedical Imaging, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Amrita V Kamath
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - C Andrew Boswell
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA.,Biomedical Imaging, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| |
Collapse
|
50
|
Declerck NB, Mateusiak L, Hernot S. Design and Validation of Site-Specifically Labeled Single-Domain Antibody-Based Tracers for in Vivo Fluorescence Imaging and Image-Guided Surgery. Methods Mol Biol 2022; 2446:395-407. [PMID: 35157285 DOI: 10.1007/978-1-0716-2075-5_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Near-infrared fluorescence molecular imaging has become an established preclinical technique to investigate molecular processes in vivo and to study novel therapies. Furthermore, fluorescence molecular imaging is gaining significant interest from clinicians as an intra-operative guidance tool. This technique makes use of targeted fluorescent tracers as contrast agents that recognize specific biomarkers expressed at the site of disease. Single-domain antibodies have shown to possess excellent properties for in vivo imaging in comparison to conventional antibodies. In this chapter, we describe a method for site-specific conjugation of a near-infrared fluorophore to single-domain antibodies by exploiting cysteine-maleimide chemistry. As opposed to random conjugation, site-specific conjugation results in a homogenously labeled fluorescent tracer and avoids inference with antigen binding.
Collapse
Affiliation(s)
- Noemi B Declerck
- Laboratory for In Vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lukasz Mateusiak
- Laboratory for In Vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sophie Hernot
- Laboratory for In Vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|