1
|
Diao W, Lu J, Li J, Wu X, Cai H, He X, Pan L, Cheng Z, Wu H, Jia Z, Mao W. Cysteine-Specific 18F and NIR Dual Labeling of Peptides via Vinyltetrazine Bioorthogonal Conjugation for Molecular Imaging. J Med Chem 2025. [PMID: 39754584 DOI: 10.1021/acs.jmedchem.4c02165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Radiolabeled peptides are vital for positron emission tomography (PET) imaging, yet the 18F-labeling peptides remain challenging due to harsh conditions and time-consuming premodification requirements. Herein, we developed a novel vinyltetrazine-mediated bioorthogonal approach for highly efficient 18F-radiolabeling of a native peptide under mild conditions. This approach enabled radiosynthesis of various tumor-targeting PET tracers, including targeting the neurofibromin receptor (18F-P10a), the integrin αvβ3 (18F-P12a), and the platelet-derived growth factor receptor β (18F-ZPDGFRβ), with a radiochemical yield exceeding 90%. Preliminary evaluations revealed excellent hydrophilicity across these tracers, with 18F-P12a effectively visualizing integrin αvβ3 expression (tumor uptake: 1.57 ± 0.54%ID/g at 2 h). Additionally, we explored the potential for development of PET/near-infrared (NIR) dual-labeling agents using this method. The dual-modality agent 18F-Cy5-P12d enables specificity and colocalized imaging integrin αvβ3 expression (tumor uptake: 1.35 ± 0.24%ID/g at 2 h). Overall, this strategy offers a versatile platform for peptide radiolabeling and dual-modality agent development.
Collapse
Affiliation(s)
- Wei Diao
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
- Department of Nuclear Medicine, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Elctronic Science and Technology of China, Chengdu 610041, China
| | - Jing Lu
- Medical Insurance Office, West China Hospital of Sichuan University, Chengdu 610041, China
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoai Wu
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Huawei Cai
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xinyu He
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lili Pan
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Zhuzhong Cheng
- Department of Nuclear Medicine, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Elctronic Science and Technology of China, Chengdu 610041, China
| | - Haoxing Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiyun Jia
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| | - Wuyu Mao
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
2
|
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
|
3
|
van der Heide CD, Ma H, Hoorens MWH, Campeiro JD, Stuurman DC, de Ridder CMA, Seimbille Y, Dalm SU. In vitro and in vivo analyses of eFAP: a novel FAP-targeting small molecule for radionuclide theranostics and other oncological interventions. EJNMMI Radiopharm Chem 2024; 9:55. [PMID: 39073475 PMCID: PMC11286609 DOI: 10.1186/s41181-024-00283-x] [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: 06/12/2024] [Accepted: 07/09/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Fibroblast activation protein (FAP), a transmembrane serine protease overexpressed by cancer-associated fibroblasts in the tumor stroma, is an interesting biomarker for targeted radionuclide theranostics. FAP-targeting radiotracers have demonstrated to be superior to [18F]FDG PET/CT in various solid cancers. However, these radiotracers have suboptimal tumor retention for targeted radionuclide therapy (TRT). We aimed to develop a novel FAP-targeting pharmacophore with improved pharmacokinetics by introducing a substitution at the 8-position of (4-quinolinoyl)-glycyl-2-cyanopyrrolidine, which allows for conjugation of a chelator, dye, or other payloads. RESULTS Here we showed the synthesis of DOTA-conjugated eFAP-6 and sulfo-Cyanine5-conjugated eFAP-7. After chemical characterization, the uptake and specificity of both tracers were determined on FAP-expressing cells. In vitro, [111In]In-eFAP-6 demonstrated a superior affinity and a more rapid, although slightly lower, peak uptake than gold standard [111In]In-FAPI-46. Confocal microscopy demonstrated a quick FAP-mediated internalization of eFAP-7. Studies with HT1080-huFAP xenografted mice confirmed a more rapid uptake of [177Lu]Lu-eFAP-6 vs. [177Lu]Lu-FAPI-46. However, tumor retention at 24 h post injection of [177Lu]Lu-eFAP-6 was lower than that of [177Lu]Lu-FAPI-46, hereby currently limiting its use for TRT. CONCLUSION The superior affinity and faster tumor accumulation of eFAP-6 over FAPI-46 makes it a suitable compound for radionuclide imaging. After further optimization, the eFAP series has great potential for various oncological interventions, including fluorescent-guided surgery and effective targeted radionuclide theranostics.
Collapse
Affiliation(s)
- Circe D van der Heide
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Hanyue Ma
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Mark W H Hoorens
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Joana D Campeiro
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Debra C Stuurman
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Corrina M A de Ridder
- Department of Experimental Urology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
- Life Sciences Division, TRIUMF, Vancouver, BC, V6T 2A3, Canada
| | - Simone U Dalm
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands.
| |
Collapse
|
4
|
Lamba M, Singh PR, Bandyopadhyay A, Goswami A. Synthetic 18F labeled biomolecules that are selective and promising for PET imaging: major advances and applications. RSC Med Chem 2024; 15:1899-1920. [PMID: 38911154 PMCID: PMC11187557 DOI: 10.1039/d4md00033a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/14/2024] [Indexed: 06/25/2024] Open
Abstract
The concept of positron emission tomography (PET) based imaging was developed more than 40 years ago. It has been a widely adopted technique for detecting and staging numerous diseases in clinical settings, particularly cancer, neuro- and cardio-diseases. Here, we reviewed the evolution of PET and its advantages over other imaging modalities in clinical settings. Primarily, this review discusses recent advances in the synthesis of 18F radiolabeled biomolecules in light of the widely accepted performance for effective PET. The discussion particularly emphasizes the 18F-labeling chemistry of carbohydrates, lipids, amino acids, oligonucleotides, peptides, and protein molecules, which have shown promise for PET imaging in recent decades. In addition, we have deliberated on how 18F-labeled biomolecules enable the detection of metabolic changes at the cellular level and the selective imaging of gross anatomical localization via PET imaging. In the end, the review discusses the future perspective of PET imaging to control disease in clinical settings. We firmly believe that collaborative multidisciplinary research will further widen the comprehensive applications of PET approaches in the clinical management of cancer and other pathological outcomes.
Collapse
Affiliation(s)
- Manisha Lamba
- Department of Chemistry, Indian Institute of Technology Birla Farms Ropar Punjab-140001 India
| | - Prasoon Raj Singh
- Department of Chemistry, Indian Institute of Technology Birla Farms Ropar Punjab-140001 India
| | - Anupam Bandyopadhyay
- Department of Chemistry, Indian Institute of Technology Birla Farms Ropar Punjab-140001 India
| | - Avijit Goswami
- Department of Chemistry, Indian Institute of Technology Birla Farms Ropar Punjab-140001 India
| |
Collapse
|
5
|
Bauer D, Cornejo MA, Hoang TT, Lewis JS, Zeglis BM. Click Chemistry and Radiochemistry: An Update. Bioconjug Chem 2023; 34:1925-1950. [PMID: 37737084 PMCID: PMC10655046 DOI: 10.1021/acs.bioconjchem.3c00286] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/16/2023] [Indexed: 09/23/2023]
Abstract
The term "click chemistry" describes a class of organic transformations that were developed to make chemical synthesis simpler and easier, in essence allowing chemists to combine molecular subunits as if they were puzzle pieces. Over the last 25 years, the click chemistry toolbox has swelled from the canonical copper-catalyzed azide-alkyne cycloaddition to encompass an array of ligations, including bioorthogonal variants, such as the strain-promoted azide-alkyne cycloaddition and the inverse electron-demand Diels-Alder reaction. Without question, the rise of click chemistry has impacted all areas of chemical and biological science. Yet the unique traits of radiopharmaceutical chemistry have made it particularly fertile ground for this technology. In this update, we seek to provide a comprehensive guide to recent developments at the intersection of click chemistry and radiopharmaceutical chemistry and to illuminate several exciting trends in the field, including the use of emergent click transformations in radiosynthesis, the clinical translation of novel probes synthesized using click chemistry, and the advent of click-based in vivo pretargeting.
Collapse
Affiliation(s)
- David Bauer
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10021, United States
| | - Mike A. Cornejo
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10021, United States
- Department
of Chemistry, Hunter College, City University
of New York, New York, New York 10065, United States
- Ph.D.
Program in Chemistry, Graduate Center of
the City University of New York, New York, New York 10016, United States
| | - Tran T. Hoang
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10021, United States
- Department
of Pharmacology, Weill Cornell Medical College, New York, New York 10065, United States
| | - Jason S. Lewis
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10021, United States
- Department
of Radiology, Weill Cornell Medical College, New York 10021, New York United States
| | - Brian M. Zeglis
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10021, United States
- Department
of Chemistry, Hunter College, City University
of New York, New York, New York 10065, United States
- Ph.D.
Program in Chemistry, Graduate Center of
the City University of New York, New York, New York 10016, United States
- Department
of Pharmacology, Weill Cornell Medical College, New York, New York 10065, United States
- Department
of Radiology, Weill Cornell Medical College, New York 10021, New York United States
- Ph.D.
Program
in Biochemistry, Graduate Center of the
City University of New York, New
York, New York 10016, United States
| |
Collapse
|
6
|
Novak A, Kersaudy F, Berger S, Morisset-Lopez S, Lefoulon F, Pifferi C, Aucagne V. An efficient site-selective, dual bioconjugation approach exploiting N-terminal cysteines as minimalistic handles to engineer tailored anti-HER2 affibody conjugates. Eur J Med Chem 2023; 260:115747. [PMID: 37657270 DOI: 10.1016/j.ejmech.2023.115747] [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: 06/26/2023] [Revised: 08/07/2023] [Accepted: 08/17/2023] [Indexed: 09/03/2023]
Abstract
Site-selective, dual-conjugation approaches for the incorporation of distinct payloads are key for the development of molecularly targeted biomolecules, such as antibody conjugates, endowed with better properties. Combinations of cytotoxic drugs, imaging probes, or pharmacokinetics modulators enabled for improved outcomes in both molecular imaging, and therapeutic settings. We have developed an efficacious dual-bioconjugation strategy to target the N-terminal cysteine of a chemically-synthesized, third-generation anti-HER2 affibody. Such two-step, one-purification approach can be carried out under mild conditions (without chaotropic agents, neutral pH) by means of a slight excess of commercially available N-hydroxysuccinimidyl esters and maleimido-functionalized payloads, to generate dual conjugates displaying drugs (DM1/MMAE) or probes (sulfo-Cy5/biotin) in high yields and purity. Remarkably, the double drug conjugate exhibited an exacerbated cytoxicity against HER2-expressing cell lines as compared to a combination of two monoconjugates, demonstrating a potent synergistic effect. Consistently, affibody-drug conjugates did not decrease the viability of HER2-negative cells, confirming their specificity for the target.
Collapse
Affiliation(s)
- Ana Novak
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Rue Charles Sadron, 45071, Orléans, France
| | - Florian Kersaudy
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Rue Charles Sadron, 45071, Orléans, France
| | - Sylvie Berger
- Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - Séverine Morisset-Lopez
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Rue Charles Sadron, 45071, Orléans, France.
| | | | - Carlo Pifferi
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Rue Charles Sadron, 45071, Orléans, France.
| | - Vincent Aucagne
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Rue Charles Sadron, 45071, Orléans, France.
| |
Collapse
|
7
|
Jin R, Zhou W, Xu Y, Jiang D, Fang D. Electrochemical Visualization of Membrane Proteins in Single Cells at a Nanoscale Using Scanning Electrochemical Cell Microscopy. Anal Chem 2023. [PMID: 37358933 DOI: 10.1021/acs.analchem.3c00114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
The electrochemical visualization of proteins in the plasma membrane of single fixed cells was achieved with a spatial resolution of 160 nm using scanning electrochemical cell microscopy. The model protein, the carcinoembryonic antigen (CEA), is linked with a ruthenium complex (Ru(bpy)32+)-tagged antibody, which exhibits redox peaks in its cyclic voltammetry curves after a nanopipette tip contacts the cellular membrane. Based on the potential-resolved oxidation or reduction currents, an uneven distribution of membrane CEAs on the cells is electrochemically visualized, which could only be achieved previously using super-resolution optical microscopy. Compared with current electrochemical microscopy, the single-cell scanning electrochemical cell microscopy (SECCM) strategy not only improves the spatial resolution but also utilizes the potential-resolved current from the antibody-antigen complex to increase electrochemical imaging accuracy. Eventually, the electrochemical visualization of cellular proteins at the nanoscale enables the super-resolution study of cells to provide more biological information.
Collapse
Affiliation(s)
- Rong Jin
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211126, China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Wenting Zhou
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211126, China
| | - Yanyan Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211126, China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Dechen Jiang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Danjun Fang
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211126, China
| |
Collapse
|
8
|
Muilenburg KM, Isder CC, Radhakrishnan P, Batra SK, Ly QP, Carlson MA, Bouvet M, Hollingsworth MA, Mohs AM. Mucins as contrast agent targets for fluorescence-guided surgery of pancreatic cancer. Cancer Lett 2023; 561:216150. [PMID: 36997106 PMCID: PMC10150776 DOI: 10.1016/j.canlet.2023.216150] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/16/2023] [Accepted: 03/26/2023] [Indexed: 03/31/2023]
Abstract
Pancreatic cancer is difficult to resect due to its unique challenges, often leading to incomplete tumor resections. Fluorescence-guided surgery (FGS), also known as intraoperative molecular imaging and optical surgical navigation, is an intraoperative tool that can aid surgeons in complete tumor resection through an increased ability to detect the tumor. To target the tumor, FGS contrast agents rely on biomarkers aberrantly expressed in malignant tissue compared to normal tissue. These biomarkers allow clinicians to identify the tumor and its stage before surgical resection and provide a contrast agent target for intraoperative imaging. Mucins, a family of glycoproteins, are upregulated in malignant tissue compared to normal tissue. Therefore, these proteins may serve as biomarkers for surgical resection. Intraoperative imaging of mucin expression in pancreatic cancer can potentially increase the number of complete resections. While some mucins have been studied for FGS, the potential ability to function as a biomarker target extends to the entire mucin family. Therefore, mucins are attractive proteins to investigate more broadly as FGS biomarkers. This review summarizes the biomarker traits of mucins and their potential use in FGS for pancreatic cancer.
Collapse
Affiliation(s)
- Kathryn M Muilenburg
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA.
| | - Carly C Isder
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA.
| | - Prakash Radhakrishnan
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, S 45th St, Omaha, NE, 68198, USA.
| | - Quan P Ly
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Department of Surgery, University of Nebraska Medical Center, 983280 Nebraska Medical Center, Omaha, NE, 68198-3280, USA.
| | - Mark A Carlson
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Department of Surgery, University of Nebraska Medical Center, 983280 Nebraska Medical Center, Omaha, NE, 68198-3280, USA.
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA; VA San Diego Healthcare System, 3350 La Jolla Village Dr, San Diego, CA, 92161, USA.
| | - Michael A Hollingsworth
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA.
| | - Aaron M Mohs
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, S 45th St, Omaha, NE, 68198, USA.
| |
Collapse
|
9
|
Development of a radiolabeled site-specific single-domain antibody positron emission tomography probe for monitoring PD-L1 expression in cancer. J Pharm Anal 2022; 12:869-878. [PMID: 36605578 PMCID: PMC9805943 DOI: 10.1016/j.jpha.2022.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 09/06/2022] [Accepted: 09/13/2022] [Indexed: 01/07/2023] Open
Abstract
Despite advances in immunotherapy for the treatment of cancers, not all patients can benefit from programmed cell death ligand 1 (PD-L1) immune checkpoint blockade therapy. Anti-PD-L1 therapeutic effects reportedly correlate with the PD-L1 expression level; hence, accurate detection of PD-L1 expression can guide immunotherapy to achieve better therapeutic effects. Therefore, based on the high affinity antibody Nb109, a new site-specifically radiolabeled tracer, 68Ga-NODA-cysteine, aspartic acid, and valine (CDV)-Nb109, was designed and synthesized to accurately monitor PD-L1 expression. The tracer 68Ga-NODA-CDV-Nb109 was obtained using a site-specific conjugation strategy with a radiochemical yield of about 95% and radiochemical purity of 97%. It showed high affinity for PD-L1 with a dissociation constant of 12.34 ± 1.65 nM. Both the cell uptake assay and positron emission tomography (PET) imaging revealed higher tracer uptake in PD-L1-positive A375-hPD-L1 and U87 tumor cells than in PD-L1-negative A375 tumor cells. Meanwhile, dynamic PET imaging of a NCI-H1299 xenograft indicated that doxorubicin could upregulate PD-L1 expression, allowing timely interventional immunotherapy. In conclusion, this tracer could sensitively and dynamically monitor changes in PD-L1 expression levels in different cancers and help screen patients who can benefit from anti-PD-L1 immunotherapy.
Collapse
|
10
|
Clickable C-Glycosyl Scaffold for the Development of a Dual Fluorescent and [ 18F]fluorinated Cyanine-Containing Probe and Preliminary In Vitro/Vivo Evaluation by Fluorescence Imaging. Pharmaceuticals (Basel) 2022; 15:ph15121490. [PMID: 36558941 PMCID: PMC9782470 DOI: 10.3390/ph15121490] [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: 10/27/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
Considering the individual characteristics of positron emission tomography (PET) and optical imaging (OI) in terms of sensitivity, spatial resolution, and tissue penetration, the development of dual imaging agents for bimodal PET/OI imaging is a growing field. A current major breakthrough in this field is the design of monomolecular agent displaying both a radioisotope for PET and a fluorescent dye for OI. We took advantage of the multifunctionalities allowed by a clickable C-glycosyl scaffold to gather the different elements. We describe, for the first time, the synthesis of a cyanine-based dual PET/OI imaging probe based on a versatile synthetic strategy and its direct radiofluorination via [18F]F-C bond formation. The non-radioactive dual imaging probe coupled with two c(RGDfK) peptides was evaluated in vitro and in vivo in fluorescence imaging. The binding on αvβ3 integrin (IC50 = 16 nM) demonstrated the efficiency of the dimeric structure and PEG linkers in maintaining the affinity. In vivo fluorescence imaging of U-87 MG engrafted nude mice showed a high tumor uptake (40- and 100-fold increase for orthotopic and ectopic brain tumors, respectively, compared to healthy brain). In vitro and in vivo evaluations and resection of the ectopic tumor demonstrated the potential of the conjugate in glioblastoma cancer diagnosis and image-guided surgery.
Collapse
|
11
|
Ariztia J, Solmont K, Moïse NP, Specklin S, Heck MP, Lamandé-Langle S, Kuhnast B. PET/Fluorescence Imaging: An Overview of the Chemical Strategies to Build Dual Imaging Tools. Bioconjug Chem 2022; 33:24-52. [PMID: 34994545 DOI: 10.1021/acs.bioconjchem.1c00503] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Molecular imaging is a biomedical research discipline that has quickly emerged to afford the observation, characterization, monitoring, and quantification of biomarkers and biological processes in living organism. It covers a large array of imaging techniques, each of which provides anatomical, functional, or metabolic information. Multimodality, as the combination of two or more of these techniques, has proven to be one of the best options to boost their individual properties, hence offering unprecedented tools for human health. In this review, we will focus on the combination of positron emission tomography and fluorescence imaging from the specific perspective of the chemical synthesis of dual imaging agents. Based on a detailed analysis of the literature, this review aims at giving a comprehensive overview of the chemical strategies implemented to build adequate imaging tools considering radiohalogens and radiometals as positron emitters, fluorescent dyes mostly emitting in the NIR window and all types of targeting vectors.
Collapse
Affiliation(s)
- Julen Ariztia
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| | - Kathleen Solmont
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| | | | - Simon Specklin
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| | - Marie Pierre Heck
- Université Paris-Saclay, INRAE, Département Médicaments et Technologies pour la santé (DMTS), SCBM, 91191, Gif-sur-Yvette cedex, France
| | | | - Bertrand Kuhnast
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| |
Collapse
|
12
|
Handula M, Verhoeven M, Chen KT, Haeck J, de Jong M, Dalm SU, Seimbille Y. Towards Complete Tumor Resection: Novel Dual-Modality Probes for Improved Image-Guided Surgery of GRPR-Expressing Prostate Cancer. Pharmaceutics 2022; 14:pharmaceutics14010195. [PMID: 35057090 PMCID: PMC8778164 DOI: 10.3390/pharmaceutics14010195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 02/01/2023] Open
Abstract
Nuclear and optical dual-modality probes can be of great assistance in prostate cancer localization, providing the means for both preoperative nuclear imaging and intraoperative surgical guidance. We developed a series of probes based on the backbone of the established GRPR-targeting radiotracer NeoB. The inverse electron demand of the Diels–Alder reaction was used to integrate the sulfo-cyanine 5 dye. Indium-111 radiolabeling, stability studies and a competition binding assay were carried out. Pilot biodistribution and imaging studies were performed in PC-3 tumor-bearing mice, using the best two dual-labeled probes. The dual-modality probes were radiolabeled with a high yield (>92%), were proven to be hydrophilic and demonstrated high stability in mouse serum (>94% intact labeled ligand at 4 h). The binding affinity for the GRPR was in the nanomolar range (21.9–118.7 nM). SPECT/CT images at 2 h p.i. clearly visualized the tumor xenograft and biodistribution studies, after scanning confirmed the high tumor uptake (8.47 ± 0.46%ID/g and 6.90 ± 0.81%ID/g for probe [111In]In-12 and [111In]In-15, respectively). Receptor specificity was illustrated with blocking studies, and co-localization of the radioactive and fluorescent signal was verified by ex vivo fluorescent imaging. Although optimal tumor-to-blood and tumor-to-kidney ratios might not yet have been reached due to the prolonged blood circulation, our probes are promising candidates for the preoperative and intraoperative visualization of GRPR-positive prostate cancer.
Collapse
Affiliation(s)
- Maryana Handula
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.H.); (M.V.); (S.U.D.)
| | - Marjolein Verhoeven
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.H.); (M.V.); (S.U.D.)
| | - Kuo-Ting Chen
- Department of Chemistry, National Dong Hwa University, Shoufeng, Hualien 974301, Taiwan;
| | - Joost Haeck
- AMIE Core Facility, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands;
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.H.); (M.V.); (S.U.D.)
| | - Simone U. Dalm
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.H.); (M.V.); (S.U.D.)
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.H.); (M.V.); (S.U.D.)
- Life Sciences Division, TRIUMF, Vancouver, BC V6T 2A3, Canada
- Correspondence: ; Tel.: +31-10-703-8961
| |
Collapse
|
13
|
Madden PJ, Arif MS, Becker ME, McRaven MD, Carias AM, Lorenzo-Redondo R, Xiao S, Midkiff CC, Blair RV, Potter EL, Martin-Sancho L, Dodson A, Martinelli E, Todd JPM, Villinger FJ, Chanda SK, Aye PP, Roy CJ, Roederer M, Lewis MG, Veazey RS, Hope TJ. Development of an In Vivo Probe to Track SARS-CoV-2 Infection in Rhesus Macaques. Front Immunol 2021; 12:810047. [PMID: 35003140 PMCID: PMC8739270 DOI: 10.3389/fimmu.2021.810047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/10/2021] [Indexed: 01/02/2023] Open
Abstract
Infection with the novel coronavirus, SARS-CoV-2, results in pneumonia and other respiratory symptoms as well as pathologies at diverse anatomical sites. An outstanding question is whether these diverse pathologies are due to replication of the virus in these anatomical compartments and how and when the virus reaches those sites. To answer these outstanding questions and study the spatiotemporal dynamics of SARS-CoV-2 infection a method for tracking viral spread in vivo is needed. We developed a novel, fluorescently labeled, antibody-based in vivo probe system using the anti-spike monoclonal antibody CR3022 and demonstrated that it could successfully identify sites of SARS-CoV-2 infection in a rhesus macaque model of COVID-19. Our results showed that the fluorescent signal from our antibody-based probe could differentiate whole lungs of macaques infected for 9 days from those infected for 2 or 3 days. Additionally, the probe signal corroborated the frequency and density of infected cells in individual tissue blocks from infected macaques. These results provide proof of concept for the use of in vivo antibody-based probes to study SARS-CoV-2 infection dynamics in rhesus macaques.
Collapse
Affiliation(s)
- Patrick J. Madden
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Muhammad S. Arif
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Mark E. Becker
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Michael D. McRaven
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Ann M. Carias
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Ramon Lorenzo-Redondo
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Institute for Global Health, Chicago, IL, United States
| | - Sixia Xiao
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Cecily C. Midkiff
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Robert V. Blair
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Elizabeth Lake Potter
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Laura Martin-Sancho
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | | | - Elena Martinelli
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - John-Paul M. Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Francois J. Villinger
- New Iberia Research Center, University of Louisiana-Lafayette, New Iberia, LA, United States
| | - Sumit K. Chanda
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Pyone Pyone Aye
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Chad J. Roy
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | | | - Ronald S. Veazey
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Thomas J. Hope
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
14
|
Zhou Z, Meshaw R, Zalutsky MR, Vaidyanathan G. Site-Specific and Residualizing Linker for 18F Labeling with Enhanced Renal Clearance: Application to an Anti-HER2 Single-Domain Antibody Fragment. J Nucl Med 2021; 62:1624-1630. [PMID: 33637584 PMCID: PMC8612331 DOI: 10.2967/jnumed.120.261446] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/01/2021] [Indexed: 11/16/2022] Open
Abstract
Single-domain antibody fragments (sdAbs) are promising vectors for immuno-PET; however, better methods for labeling sdAbs with 18F are needed. Herein, we evaluate a site-specific strategy using an 18F residualizing motif and the anti-epidermal growth factor receptor 2 (HER2) sdAb 5F7 bearing an engineered C-terminal GGC tail (5F7GGC). Methods: 5F7GGC was site-specifically attached with a tetrazine-bearing agent via thiol-maleimide reaction. The resultant conjugate was labeled with 18F by inverse electron demand Diels-Alder cycloaddition with a trans-cyclooctene attached to 6-18F-fluoronicotinoyl moiety via a renal brush border enzyme-cleavable linker and a PEG4 chain (18F-5F7GGC). For comparisons, 5F7 sdAb was labeled using the prototypical residualizing agent, N-succinimidyl 3-(guanidinomethyl)-5-125I-iodobenzoate (iso-125I-SGMIB). The 2 labeled sdAbs were compared in paired-label studies performed in the HER2-expressing BT474M1 breast carcinoma cell line and athymic mice bearing BT474M1 subcutaneous xenografts. Small-animal PET/CT imaging after administration of 18F-5F7GGC in the above mouse model was also performed. Results:18F-5F7GGC was synthesized in an overall radiochemical yield of 8.9% ± 3.2% with retention of HER2 binding affinity and immunoreactivity. The total cell-associated and intracellular activity for 18F-5F7GGC was similar to that for coincubated iso-125I-SGMIB-5F7. Likewise, the uptake of 18F-5F7GGC in BT474M1 xenografts in mice was similar to that for iso-125I-SGMIB-5F7; however, 18F-5F7GGC exhibited significantly more rapid clearance from the kidney. Small-animal PET/CT imaging confirmed high uptake and retention in the tumor with very little background activity at 3 h except in the bladder. Conclusion: This site-specific and residualizing 18F-labeling strategy could facilitate clinical translation of 5F7 anti-HER2 sdAb as well as other sdAbs for immuno-PET.
Collapse
Affiliation(s)
- Zhengyuan Zhou
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Rebecca Meshaw
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | | |
Collapse
|
15
|
Xu L, Kuan SL, Weil T. Contemporary Approaches for Site-Selective Dual Functionalization of Proteins. Angew Chem Int Ed Engl 2021; 60:13757-13777. [PMID: 33258535 PMCID: PMC8248073 DOI: 10.1002/anie.202012034] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Indexed: 12/16/2022]
Abstract
Site-selective protein functionalization serves as an invaluable tool for investigating protein structures and functions in complicated cellular environments and accomplishing semi-synthetic protein conjugates such as traceable therapeutics with improved features. Dual functionalization of proteins allows the incorporation of two different types of functionalities at distinct location(s), which greatly expands the features of native proteins. The attachment and crosstalk of a fluorescence donor and an acceptor dye provides fundamental insights into the folding and structural changes of proteins upon ligand binding in their native cellular environments. Moreover, the combination of drug molecules with different modes of action, imaging agents or stabilizing polymers provides new avenues to design precision protein therapeutics in a reproducible and well-characterizable fashion. This review aims to give a timely overview of the recent advancements and a future perspective of this relatively new research area. First, the chemical toolbox for dual functionalization of proteins is discussed and compared. The strengths and limitations of each strategy are summarized in order to enable readers to select the most appropriate method for their envisaged applications. Thereafter, representative applications of these dual-modified protein bioconjugates benefiting from the synergistic/additive properties of the two synthetic moieties are highlighted.
Collapse
Affiliation(s)
- Lujuan Xu
- Max Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Institute of Inorganic Chemistry IUlm UniversityAlbert-Einstein-Allee 1189081UlmGermany
| | - Seah Ling Kuan
- Max Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Institute of Inorganic Chemistry IUlm UniversityAlbert-Einstein-Allee 1189081UlmGermany
| | - Tanja Weil
- Max Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Institute of Inorganic Chemistry IUlm UniversityAlbert-Einstein-Allee 1189081UlmGermany
| |
Collapse
|
16
|
Sharma SK, Adumeau P, Keinänen O, Sisodiya V, Sarvaiya H, Tchelepi R, Korsen JA, Pourat J, Edwards KJ, Ragupathi A, Hamdy O, Saunders LR, Rudin CM, Poirier JT, Lewis JS, Zeglis BM. Synthesis and Comparative In Vivo Evaluation of Site-Specifically Labeled Radioimmunoconjugates for DLL3-Targeted ImmunoPET. Bioconjug Chem 2021; 32:1255-1262. [PMID: 33835770 PMCID: PMC8295218 DOI: 10.1021/acs.bioconjchem.1c00121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Delta-like ligand 3 (DLL3) is a therapeutic target for the treatment of small cell lung cancer, neuroendocrine prostate cancer, and isocitrate dehydrogenase mutant glioma. In the clinic, DLL3-targeted 89Zr-immunoPET has the potential to aid in the assessment of disease burden and facilitate the selection of patients suitable for therapies that target the antigen. The overwhelming majority of 89Zr-labeled radioimmunoconjugates are synthesized via the random conjugation of desferrioxamine (DFO) to lysine residues within the immunoglobulin. While this approach is admittedly facile, it can produce heterogeneous constructs with suboptimal in vitro and in vivo behavior. In an effort to circumvent these issues, we report the development and preclinical evaluation of site-specifically labeled radioimmunoconjugates for DLL3-targeted immunoPET. To this end, we modified a cysteine-engineered variant of the DLL3-targeting antibody SC16-MB1 with two thiol-reactive variants of DFO: one bearing a maleimide moiety (Mal-DFO) and the other containing a phenyloxadiazolyl methyl sulfone group (PODS-DFO). In an effort to obtain immunoconjugates with a DFO-to-antibody ratio (DAR) of 2, we explored both the reduction of the antibody with tris(2-carboxyethyl) phosphine (TCEP) as well as the use of a combination of glutathione and arginine as reducing and stabilizing agents, respectively. While exerting control over the DAR of the immunoconjugate proved cumbersome using TCEP, the use of glutathione and arginine enabled the selective reduction of the engineered cysteines and thus the formation of homogeneous immunoconjugates. A head-to-head comparison of the resulting 89Zr-radioimmunoconjugates in mice bearing DLL3-expressing H82 xenografts revealed no significant differences in tumoral uptake and showed comparable radioactivity concentrations in most healthy nontarget organs. However, 89Zr-DFOPODS-DAR2SC16-MB1 produced 30% lower uptake (3.3 ± 0.5 %ID/g) in the kidneys compared to 89Zr-DFOMal-DAR2SC16-MB1 (4.7 ± 0.5 %ID/g). In addition, H82-bearing mice injected with a 89Zr-labeled isotype-control radioimmunoconjugate synthesized using PODS exhibited ∼40% lower radioactivity in the kidneys compared to mice administered its maleimide-based counterpart. Taken together, these results demonstrate the improved in vivo performance of the PODS-based radioimmunoconjugate and suggest that a stable, well-defined DAR2 radiopharmaceutical may be suitable for the clinical immunoPET of DLL3-expressing cancers.
Collapse
Affiliation(s)
- Sai Kiran Sharma
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Department of Chemistry, Hunter College, City University of New York, New York, New York 10021, United States
| | - Pierre Adumeau
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10021, United States
| | - Outi Keinänen
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Department of Chemistry, Hunter College, City University of New York, New York, New York 10021, United States
| | - Vikram Sisodiya
- Abbvie Stemcentrx, South San Francisco, California 94080, United States
| | - Hetal Sarvaiya
- Abbvie Stemcentrx, South San Francisco, California 94080, United States
| | - Robert Tchelepi
- Abbvie Stemcentrx, South San Francisco, California 94080, United States
| | - Joshua A Korsen
- Department of Pharmacology, Weill Cornell Medical College, New York, New York 10021, United States
| | - Jacob Pourat
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Kimberly J Edwards
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Ashwin Ragupathi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Omar Hamdy
- Abbvie Stemcentrx, South San Francisco, California 94080, United States
| | - Laura R Saunders
- Abbvie Stemcentrx, South San Francisco, California 94080, United States
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - John T Poirier
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Department of Pharmacology, Weill Cornell Medical College, New York, New York 10021, United States.,Department of Radiology, Weill Cornell Medical College, New York, New York 10021, United States.,Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Brian M Zeglis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Department of Chemistry, Hunter College, City University of New York, New York, New York 10021, United States.,Department of Radiology, Weill Cornell Medical College, New York, New York 10021, United States
| |
Collapse
|
17
|
Zhao L, Chen H, Lu L, Zhao C, Malichewe CV, Wang L, Guo X, Zhang X. Design and screening of a novel neuropilin-1 targeted penetrating peptide for anti-angiogenic therapy in glioma. Life Sci 2021; 270:119113. [PMID: 33508290 DOI: 10.1016/j.lfs.2021.119113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 12/20/2022]
Abstract
AIMS This study aimed to design and screen a dual functional fusion peptide that could penetrate the blood-brain barrier and target neuropilin 1 (NRP1) overexpressed in vascular endothelial cells for the anti-angiogenesis of glioma treatment. MAIN METHODS At the cellular level, the in vitro anti-angiogenic activity of six NRP1 targeting peptides was screened by testing the ability to inhibit the proliferation and tube formation of HUVECs. Then, the in vitro anti-angiogenic activity of two fusion peptides containing different linkers was screened by testing the ability to inhibit HUVECs proliferation, tube formation and migration. The effect of fusion peptide on VEGFR2 related signal pathway was confirmed by Western-blotting. Surface plasmon resonance technology was used to detect the affinity of the fusion peptide to NRP1. The ability of FITC-labeled peptides to penetrate cells was confirmed by cell uptake assay. By establishing an orthotopic glioma model, we evaluated the ability of FITC-labeled peptides to penetrate the blood-brain barrier and their anti-glioma growth activity in vivo. KEY FINDINGS We found that NRP1 targeting peptide RP7 and linker cysteine were the most suitable key components in the fusion peptide. We also found that the fusion peptide Tat-C-RP7 we constructed had the strongest ability to penetrate the blood-brain barrier and anti-angiogenic activity in vitro and in vivo. SIGNIFICANCE At present, NRP1 targeting peptide as a drug delivery tool and molecular probe seems to have received more attention. We constructed a fusion peptide Tat-C-RP7 with strong anti-angiogenic activity for the treatment of glioma.
Collapse
Affiliation(s)
- Lin Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Hongyuan Chen
- Department of General Surgery, Shandong University Affiliated Shandong Provincial Hospital, Jinan 250021, China
| | - Lu Lu
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Chunqian Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Christina V Malichewe
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Lei Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xiuli Guo
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
| | - Xinke Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
| |
Collapse
|
18
|
Xu L, Kuan SL, Weil T. Contemporary Approaches for Site‐Selective Dual Functionalization of Proteins. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202012034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Lujuan Xu
- Max Planck Institute for Polymer Research Ackermannweg 10 55128 Mainz Germany
- Institute of Inorganic Chemistry I Ulm University Albert-Einstein-Allee 11 89081 Ulm Germany
| | - Seah Ling Kuan
- Max Planck Institute for Polymer Research Ackermannweg 10 55128 Mainz Germany
- Institute of Inorganic Chemistry I Ulm University Albert-Einstein-Allee 11 89081 Ulm Germany
| | - Tanja Weil
- Max Planck Institute for Polymer Research Ackermannweg 10 55128 Mainz Germany
- Institute of Inorganic Chemistry I Ulm University Albert-Einstein-Allee 11 89081 Ulm Germany
| |
Collapse
|
19
|
Liu M, Li L, Jin D, Liu Y. Nanobody-A versatile tool for cancer diagnosis and therapeutics. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1697. [PMID: 33470555 DOI: 10.1002/wnan.1697] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/19/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022]
Abstract
In spite of the successful use of monoclonal antibodies (mAbs) in clinic for tumor treatment, their applications are still hampered in therapeutic development due to limitations, such as tumor penetration and high cost of manufacture. Nanobody, a single domain antibody that holds the strong antigen targeting and binding capacity, has demonstrated various advantages relative to antibody. Nanobody is considered as a next-generation of antibody-derived tool in the antigen related recognition and modulation. A number of nanobodies have been developed and evaluated in different stages of clinical trials for cancer treatment. Here we summarized the current progress of nanobody in tumor diagnosis and therapeutics, particularly on the conjugation of nanobody with functional moieties. The nanobody conjugation of diagnostic agents, such as radionuclide and optical tracers, can achieve specific tumor imaging. The nanobody-drug conjugates can enhance the therapeutic efficacy of anti-tumor drugs and reduce the adverse effects. The decoration of nanobody on nanodrug delivery systems can further improve the drug targeting to specific tumors. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Manman Liu
- CAS Key Laboratory of Soft Matter Chemistry, Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Li Li
- CAS Key Laboratory of Soft Matter Chemistry, Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Duo Jin
- CAS Key Laboratory of Soft Matter Chemistry, Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Yangzhong Liu
- CAS Key Laboratory of Soft Matter Chemistry, Department of Chemistry, University of Science and Technology of China, Hefei, China
| |
Collapse
|
20
|
Wu AM. Protein Engineering for Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00045-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
21
|
Ji Y, Jones C, Baek Y, Park GK, Kashiwagi S, Choi HS. Near-infrared fluorescence imaging in immunotherapy. Adv Drug Deliv Rev 2020; 167:121-134. [PMID: 32579891 DOI: 10.1016/j.addr.2020.06.012] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022]
Abstract
Near-infrared (NIR) light possesses many suitable optophysical properties for medical imaging including low autofluorescence, deep tissue penetration, and minimal light scattering, which together allow for high-resolution imaging of biological tissue. NIR imaging has proven to be a noninvasive and effective real-time imaging methodology that provides a high signal-to-background ratio compared to other potential optical imaging modalities. In response to this, the use of NIR imaging has been extensively explored in the field of immunotherapy. To date, NIR fluorescence imaging has successfully offered reliable monitoring of the localization, dynamics, and function of immune responses, which are vital in assessing not only the efficacy but also the safety of treatments to design immunotherapies optimally. This review aims to provide an overview of the current research on NIR imaging of the immune response. We expect that the use of NIR imaging will expand further in response to the recent success in cancer immunotherapy. We will also offer our insights on how this technology will meet rapidly growing expectations in the future.
Collapse
Affiliation(s)
- Yuanyuan Ji
- Scientific Research Centre, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China; Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Catherine Jones
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yoonji Baek
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - G Kate Park
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Satoshi Kashiwagi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
22
|
Li Y, Zhou Y, Yue X, Dai Z. Cyanine Conjugate-Based Biomedical Imaging Probes. Adv Healthc Mater 2020; 9:e2001327. [PMID: 33000915 DOI: 10.1002/adhm.202001327] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/11/2020] [Indexed: 12/12/2022]
Abstract
Cyanine is a class of fluorescent dye with meritorious fluorescence properties and has motivated numerous researchers to explore its imaging capabilities by miscellaneous structural modification and functionalization strategies. The covalent conjugation with other functional molecules represents a distinctive design strategy and has shown immense potential in both basic and clinical research. This review article summarizes recent achievements in cyanine conjugate-based probes for biomedical imaging. Particular attention is paid to the conjugation with targeting warheads and other contrast agents for targeted fluorescence imaging and multimodal imaging, respectively. Additionally, their clinical potential in cancer diagnostics is highlighted and some concurrent impediments for clinical translation are discussed.
Collapse
Affiliation(s)
- Yang Li
- Department of Biomedical Engineering College of Engineering Peking University Beijing 100871 China
| | - Yiming Zhou
- Department of Biomedical Engineering College of Engineering Peking University Beijing 100871 China
| | - Xiuli Yue
- School of Environment Harbin Institute of Technology Harbin 150090 China
| | - Zhifei Dai
- Department of Biomedical Engineering College of Engineering Peking University Beijing 100871 China
| |
Collapse
|
23
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
24
|
Zhao X, Ning Q, Mo Z, Tang S. A promising cancer diagnosis and treatment strategy: targeted cancer therapy and imaging based on antibody fragment. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:3621-3630. [PMID: 31468992 DOI: 10.1080/21691401.2019.1657875] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With the arrival of the precision medicine and personalized treatment era, targeted therapy that improves efficacy and reduces side effects has become the mainstream approach of cancer treatment. Antibody fragments that further enhance penetration and retain the most critical antigen-specific binding functions are considered the focus of research targeting cancer imaging and therapy. Thanks to the superior penetration and rapid blood clearance of antibody fragments, antibody fragment-based imaging agents enable efficient and sensitive imaging of tumour sites. In tumour-targeted therapy, antibody fragments can directly inhibit tumour proliferation and growth, serve as an ideal carrier for delivery of anti-tumour drugs, or manipulate the immune system to eliminate tumour cells. In this review, the excellent physicochemical properties and the basic structure of antibody fragments are expressly depicted depicted, the progress of antibody fragments in cancer therapy and imaging are thoroughly summarized, and the future development of antibody fragments is predicted.
Collapse
Affiliation(s)
- Xuhong Zhao
- Learning Key Laboratory for Pharmacoproteomics of Hunan Province, Institute of Pharmacy and Pharmacology, University of South China , Hengyang , China.,Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine , Huaihua , China
| | - Qian Ning
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine , Huaihua , China
| | - Zhongcheng Mo
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China , Hengyang , China
| | - Shengsong Tang
- Learning Key Laboratory for Pharmacoproteomics of Hunan Province, Institute of Pharmacy and Pharmacology, University of South China , Hengyang , China.,Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine , Huaihua , China
| |
Collapse
|
25
|
Gamache RF, Zettlitz KA, Tsai WTK, Collins J, Wu AM, Murphy JM. Tri-functional platform for construction of modular antibody fragments for in vivo 18F-PET or NIRF molecular imaging. Chem Sci 2020; 11:1832-1838. [PMID: 34123276 PMCID: PMC8148382 DOI: 10.1039/c9sc05007h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Positron emission tomography (PET) molecular imaging is a powerful tool for interrogating physiological and biochemical processes to understand the biology of disease and advance therapeutic developments. Near-infrared fluorescence (NIRF) optical imaging has become increasingly popular for intraoperative staging to enable cellular resolution imaging of tumor margins during surgical resection. In addition, engineered antibody fragments have emerged as promising molecular imaging agents given their exquisite target selectivity, rapid systemic clearance and site-selective chemical modification. We report a tri-functional platform for construction of a modular antibody fragment that can rapidly be labeled with radionuclides or fluorophores for PET or NIRF molecular imaging of prostate stem cell antigen (PSCA). To provide a universal approach towards the targeted delivery of PET and optical imaging agents, we have developed a tri-functional platform (TFP) for the facile construction of modular, target-specific tracers.![]()
Collapse
Affiliation(s)
- Raymond F Gamache
- Department of Chemistry and Biochemistry, University of California Los Angeles CA 90095 USA
| | - Kirstin A Zettlitz
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| | - Wen-Ting K Tsai
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| | - Jeffrey Collins
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| | - Anna M Wu
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| | - Jennifer M Murphy
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| |
Collapse
|
26
|
Debie P, Hernot S. Emerging Fluorescent Molecular Tracers to Guide Intra-Operative Surgical Decision-Making. Front Pharmacol 2019; 10:510. [PMID: 31139085 PMCID: PMC6527780 DOI: 10.3389/fphar.2019.00510] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/24/2019] [Indexed: 12/26/2022] Open
Abstract
Fluorescence imaging is an emerging technology that can provide real-time information about the operating field during cancer surgery. Non-specific fluorescent agents, used for the assessment of blood flow and sentinel lymph node detection, have so far dominated this field. However, over the last decade, several clinical studies have demonstrated the great potential of targeted fluorescent tracers to visualize tumor lesions in a more specific way. This has led to an exponential growth in the development of novel molecular fluorescent contrast agents. In this review, the design of fluorescent molecular tracers will be discussed, with particular attention for agents and approaches that are of interest for clinical translation.
Collapse
Affiliation(s)
| | - Sophie Hernot
- Laboratory for in vivo Cellular and Molecular Imaging (ICMI-BEFY/MIMA), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|