1
|
Olkowski CP, Basuli F, Fernandes B, Ghaemi B, Shi J, Zhang HH, Farber JM, Escorcia FE, Choyke PL, Jacobson O. Comparative Kidney Uptake of Nanobody-Based PET Tracers Labeled with Various Fluorine-18-Labeled Prosthetic Groups. Mol Pharm 2025; 22:533-543. [PMID: 39680709 DOI: 10.1021/acs.molpharmaceut.4c01101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Nanobodies, or single-domain antibody fragments, are promising candidates for molecular imaging due to their small size, rapid tissue penetration, and high target specificity. However, a significant challenge in their use is high renal uptake and retention, which can limit the therapeutic efficacy and complicate image interpretation. This study compares five different fluorine-18-labeled prosthetic groups for nanobodies, aiming to optimize pharmacokinetics and minimize kidney retention while maintaining tumor targeting. Using an epidermal growth factor receptor (EGFR) targeting nanobody as a model, two labeling approaches were evaluated; direct labeling of RESCA (with and without polyethylene glycol (PEG))-conjugated nanobody using Al[18F]F and indirect labeling using ([18F]F-fluoropyridine ([18F]F-FPy)-based prosthetic groups (site-specific and nonsite-specific). Labeled nanobodies were characterized in vitro for binding affinity and cell uptake with in vivo behavior assessed in EGFR + A431 tumor-bearing mice using PET imaging and biodistribution studies. Labeling with Al[18F]F showed high renal retention, which was partially mitigated by PEGylation. However, PEGylation also led to a decreased tumor uptake, particularly with longer PEG chains. Labeling using [18F]F-FPy prosthetic groups exhibited the most favorable pharmacokinetics, with rapid renal clearance and minimal kidney retention while maintaining high tumor uptake. These constructs showed excellent tumor-to-background contrast as early as 1 h postinjection. The study confirms that the selection of the prosthetic group significantly impacts the in vivo behavior of nanobodies, particularly regarding kidney accumulation. [18F]F-FPy-based prosthetic groups show the most promising results, with high tumor and minimal kidney uptake. Robust production of [18F]F-FPy on Sep-Pak is adaptable to clinical translation. Moreover, the potential substitution of 18F with therapeutic radioisotopes such as 131I or 211At could expand the application of these nanobodies from diagnostics to targeted radionuclide therapy while maintaining a low kidney exposure. These findings have important implications for optimizing nanobody-based radiopharmaceuticals for molecular imaging and targeted radionuclide therapy.
Collapse
Affiliation(s)
- Colleen P Olkowski
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Bruna Fernandes
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Behnaz Ghaemi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Jianfeng Shi
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Hongwei H Zhang
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Joshua M Farber
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Freddy E Escorcia
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Peter L Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Orit Jacobson
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
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
|
Mishra A, Carrascal-Miniño A, Kim J, T M de Rosales R. [ 68Ga]Ga-THP-tetrazine for bioorthogonal click radiolabelling: pretargeted PET imaging of liposomal nanomedicines. RSC Chem Biol 2024; 5:622-639. [PMID: 38966673 PMCID: PMC11221536 DOI: 10.1039/d4cb00039k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/10/2024] [Indexed: 07/06/2024] Open
Abstract
Pretargeted PET imaging using bioorthogonal chemistry is a leading strategy for the tracking of long-circulating agents such as antibodies and nanoparticle-drug delivery systems with short-lived isotopes. Here, we report the synthesis, characterisation and in vitro/vivo evaluation of a new 68Ga-based radiotracer [68Ga]Ga-THP-Tetrazine ([68Ga]Ga-THP-Tz) for bioorthogonal click radiochemistry and in vivo labelling of agents with slow pharmacokinetics. THP-tetrazine (THP-Tz) can be radiolabelled to give [68/67Ga]Ga-THP-Tz at room temperature in less than 15 minutes with excellent radiochemical stability in vitro and in vivo. [68Ga]Ga-THP-Tz was tested in vitro and in vivo for pretargeted imaging of stealth PEGylated liposomes, chosen as a leading clinically-approved platform of nanoparticle-based drug delivery, and for their known long-circulating properties. To achieve this, PEGylated liposomes were functionalised with a synthesised transcyclooctene (TCO) modified phospholipid. Radiolabelling of TCO-PEG-liposomes with [68/67Ga]Ga-THP-Tz was demonstrated in vitro in human serum, and in vivo using both healthy mice and in a syngeneic cancer murine model (WEHI-164 fibrosarcoma). Interestingly in vivo data revealed that [68Ga]Ga-THP-Tz was able to in vivo radiolabel liposomes present in the liver and spleen, and not those in the blood pool or in the tumour. Overall, these results demonstrate the potential of [68Ga]Ga-THP-Tz for pretargeted imaging/therapy but also some unexpected limitations of this system.
Collapse
Affiliation(s)
- Aishwarya Mishra
- School of Biomedical Engineering & Imaging Sciences, King's College London St Thomas' Hospital London SE1 7EH UK
| | - Amaia Carrascal-Miniño
- School of Biomedical Engineering & Imaging Sciences, King's College London St Thomas' Hospital London SE1 7EH UK
| | - Jana Kim
- School of Biomedical Engineering & Imaging Sciences, King's College London St Thomas' Hospital London SE1 7EH UK
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London St Thomas' Hospital London SE1 7EH UK
| |
Collapse
|
4
|
Tian R, Kong J, He Y, Xu G, Chen T, Han J. Radiosynthesis and preclinical evaluations of [ 18F]AlF-RESCA-5F7 as a novel molecular probe for HER2 tumor imaging. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2024; 14:175-181. [PMID: 39027646 PMCID: PMC11253080 DOI: 10.62347/bvpk1360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/15/2024] [Indexed: 07/20/2024]
Abstract
HER2 overexpression is associated with various tumor types and prompted the development of targeted therapies. Previously, iso-[211At]SGMAB-5F7 was developed as a HER2-targeted alpha therapy agent, demonstrating promising therapeutic efficacy in the preclinical stage. Aiming for an 18F-labeled tracer for companion diagnostics in clinical translation, we employed the Al18F-RESCA strategy in our current work and investigated whether [18F]AlF-RESCA-5F7 could visualize HER2 expression in vivo. [18F]AlF-RESCA-5F7 was attained with high radiochemical purity (> 99%) and molar activity in the range of 16.5 ± 8.8 GBq/μmol (n = 8). Compared to previously reported radiotracers that contained 5F7 as the HER2-targeting carrier and fluorine-18 as the positron-emitting isotope, the radiosynthesis was simplified to one single step within 30 min. The dissociation constant of [18F]AlF-RESCA-5F7 was determined as 3.3 nM via saturation binding assay using SKOV3 ovarian carcinoma cells. Tumor uptake of the novel tracer in Balb/c nude mice bearing SKOV3 xenografts was 4.69 ± 1.51, 3.34 ± 0.82 and 3.77 ± 0.99 %ID/g at 1, 2, and 4 h post-injection. Even though high retention of radioactivity was seen in the kidneys, micro-PET/CT imaging of [18F]AlF-RESCA-5F7 delineated the tumor up to 4 h post-injection with minimal activity in the gallbladder, intestines, and bone. This study suggests that [18F]AlF-RESCA-5F7 is a promising HER2 PET radiotracer with an eased radiolabeling method. Whether [18F]AlF-RESCA-5F7 could work as a companion diagnostic agent to assist in patient stratification and treatment monitoring of iso-[211At]SGMAB-5F7 warrants further investigation.
Collapse
Affiliation(s)
- Ruhua Tian
- Department of Physiology, School of Basic Medicine, Guizhou Medical UniversityGuiyang 550009, Guizhou, China
| | - Jinping Kong
- Department of Physiology, School of Basic Medicine, Guizhou Medical UniversityGuiyang 550009, Guizhou, China
| | - Yingfang He
- Institute of Radiation Medicine, Fudan UniversityNo. 2094 Xietu Road, Shanghai 200032, China
| | - Guoqiang Xu
- Department of Physiology, School of Basic Medicine, Guizhou Medical UniversityGuiyang 550009, Guizhou, China
| | - Tengxiang Chen
- Department of Physiology, School of Basic Medicine, Guizhou Medical UniversityGuiyang 550009, Guizhou, China
| | - Junbin Han
- Department of Physiology, School of Basic Medicine, Guizhou Medical UniversityGuiyang 550009, Guizhou, China
- Institute of Radiation Medicine, Fudan UniversityNo. 2094 Xietu Road, Shanghai 200032, China
| |
Collapse
|
5
|
Gao F, Liu F, Wang J, Bi J, Zhai L, Li D. Molecular probes targeting HER2 PET/CT and their application in advanced breast cancer. J Cancer Res Clin Oncol 2024; 150:118. [PMID: 38466436 PMCID: PMC10927773 DOI: 10.1007/s00432-023-05519-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/12/2023] [Indexed: 03/13/2024]
Abstract
PURPOSE Human epidermal growth factor receptor 2 (HER2)-positive breast cancer cases are among the most aggressive breast tumor subtypes. Accurately assessing HER2 expression status is vital to determining whether patients will benefit from targeted anti-HER2 treatment. HER2-targeted positron emission tomography (PET/CT) is noninvasive, enabling the real-time evaluation of breast cancer patient HER2 status with accuracy. METHODS We summarize the research progress of PET/CT targeting HER2 in breast cancer, focusing on PET/CT molecular probes targeting HER2 and their clinical application in the management of advanced breast cancer. RESULTS At present, a variety of different HER2 targeted molecular probes for PET/CT imaging have been developed, including nucleolin-labeled antibodies, antibody fragments, nanobodies, and peptides of various affinities, among others. HER2-targeted PET/CT can relatively accurately evaluate HER2 expression status in advanced breast cancer patients. It has good performance in the early detection of small HER2-positive lesions, evaluation of HER2 status in lesions that cannot be readily biopsied, evaluation of the heterogeneity of multiple metastases, identification of lesions with altered HER2 status, and evaluation of the efficacy of anti-HER2 drugs. CONCLUSION HER2-targeted PET/CT offers a promising noninvasive approach for real-time assessment of HER2 status,which can be guide targeted treatment for HER2-positive breast cancer patients. Future prospective clinical studies will be invaluable for fully evaluating the importance of HER2-targeted molecular imaging in the management of breast cancer.
Collapse
Affiliation(s)
- Fang Gao
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Fengxu Liu
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Jun Wang
- Department of Anesthesia, Armed Police Corps Hospital in Shanxi Province, Xiaodian District, Taiyuan, Shanxi, People's Republic of China
| | - Junfang Bi
- Department of Combined Traditional Chinese Medicine and West Medicine, Traditional Chinese Medicine Hospital of Shijiazhuang City, 233 Zhongshan West Road, Qiaoxi District, Shijiazhuang, Hebei, China
| | - Luoping Zhai
- Department of Nuclear Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China.
| | - Dong Li
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China.
- Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
6
|
Steffann M, Bluet G, Roy S, Aubert C, Fouquet E, Hermange P. 18 F-Fluorination of a supported 2-(aryl-di-tert-butylsilyl)-N-methyl-imidazole for indirect 18 F-labeling of a V H H single-variable domain. J Labelled Comp Radiopharm 2024; 67:104-110. [PMID: 38224624 DOI: 10.1002/jlcr.4082] [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: 08/22/2023] [Revised: 11/15/2023] [Accepted: 12/18/2023] [Indexed: 01/17/2024]
Abstract
Anchoring an imidazole-di-tert-butyl-arylsilane possessing an azido group to a polystyrene resin provided a heterogeneous precursor that was radiolabeled easily using aqueous [18 F]fluoride. After optimizing the conditions (i.e., using DMSO as solvent and heating at 160°C for 15 min), the desired [18 F]fluorosilane was obtained in 24% radiochemical yield (RCY) and 78% radiochemical purity (RCP) using solid-phase extraction as sole purification. Then, this compound was conjugated by strain-promoted alkyne-azide cycloaddition to a model single-variable domain possessing a cyclooctyne tag, yielding to the desired 18 F-labeled bioconjugate in 2% RCY and >95% RCP after purification by a size exclusion chromatography.
Collapse
Affiliation(s)
- Marine Steffann
- Institut des Sciences Moléculaires (ISM), UMR 5255, Univ. Bordeaux, CNRS, Bordeaux INP, Talence Cedex, France
- Sanofi, Integrated Drug Discovery (IDD) Isotope Chemistry (IC), Vitry-sur-Seine, France
| | - Guillaume Bluet
- Sanofi, Integrated Drug Discovery (IDD) Isotope Chemistry (IC), Vitry-sur-Seine, France
| | - Sébastien Roy
- Sanofi, Integrated Drug Discovery (IDD) Isotope Chemistry (IC), Vitry-sur-Seine, France
| | - Catherine Aubert
- Sanofi, Integrated Drug Discovery (IDD) Isotope Chemistry (IC), Vitry-sur-Seine, France
| | - Eric Fouquet
- Institut des Sciences Moléculaires (ISM), UMR 5255, Univ. Bordeaux, CNRS, Bordeaux INP, Talence Cedex, France
| | - Philippe Hermange
- Institut des Sciences Moléculaires (ISM), UMR 5255, Univ. Bordeaux, CNRS, Bordeaux INP, Talence Cedex, France
| |
Collapse
|
7
|
Huang W, Liang C, Zhang Y, Zhang D, An S, Wu Q, Li J, Zhao H, Wang C, Cui J, Bao Z, Huang G, Wei W, Liu J. ImmunoPET imaging of Trop2 expression in solid tumors with nanobody tracers. Eur J Nucl Med Mol Imaging 2024; 51:380-394. [PMID: 37792026 DOI: 10.1007/s00259-023-06454-3] [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: 08/08/2023] [Accepted: 09/21/2023] [Indexed: 10/05/2023]
Abstract
PURPOSE The high expression of the transmembrane glycoprotein trophoblast cell-surface antigen 2 (Trop2) was strongly associated with the progression of solid tumors, including pancreatic and gastric cancers. Our study aimed to construct Trop2-specific immuno-positron emission tomography (immunoPET) probes and assess the diagnostic abilities in preclinical pancreatic and gastric cancer models. METHODS The expression of Trop2 in pancreatic cancer was determined by single-cell sequencing and immunohistochemistry on tissue microarray (TMA). Flow cytometry was used to screen the expression of Trop2 in pancreatic cancer cell lines. Two nanobodies (i.e., RTD98 and RTD01) targeting Trop2 were developed and labeled with gallium-68 (68Ga, T1/2 = 1.1 h) to construct immunoPET imaging probes. The agents were researched in cell-derived pancreatic and patient-derived gastric cancer models expressing varying Trop2. RESULTS Single-cell sequencing results showed high expression of Trop2 in pancreatic ductal cells as well as acinar cells and immunohistochemical staining of TMA from pancreatic cancers showed significantly higher expression of Trop2 in cancerous than in paracancerous tissues. ImmunoPET utilizing [68Ga]Ga-NOTA-RTD98 could clearly delineate subcutaneous tumors, both in cell-derived pancreatic cancer models and patient-derived gastric cancer models, superior to imaging using [18F]-FDG or a non-specific probe [68Ga]Ga-NOTA-RTD161. Another probe with improved pharmacokinetics targeting Trop2, [68Ga]Ga-NOTA-RTD01, was further prepared and showed advantageous diagnostic capabilities in preclinical pancreatic cancer models. CONCLUSION In the work, we reported two nanobody tracers targeting human Trop2 which may facilitate better use of Trop2-targeted therapeutics by noninvasively displaying expression dynamics of the target.
Collapse
Affiliation(s)
- Wei Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Chenyi Liang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - You Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Di Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Shuxian An
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Qianyun Wu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Jiajin Li
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Haitao Zhao
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Cheng Wang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Jiujie Cui
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Zhouzhou Bao
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
- Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China.
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China.
| |
Collapse
|
8
|
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
|
9
|
Feng Y, Meshaw R, Zhao XG, Jannetti S, Vaidyanathan G, Zalutsky MR. Effective Treatment of Human Breast Carcinoma Xenografts with Single-Dose 211At-Labeled Anti-HER2 Single-Domain Antibody Fragment. J Nucl Med 2023; 64:124-130. [PMID: 35618478 PMCID: PMC9841253 DOI: 10.2967/jnumed.122.264071] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 01/06/2023] Open
Abstract
Single-domain antibody fragments (sdAbs) are attractive for targeted α-particle therapy, particularly with 211At, because of their rapid accumulation in tumor and clearance from normal tissues. Here, we evaluate the therapeutic potential of this strategy with 5F7 and VHH_1028-2 sdAbs that bind with high affinity to domain IV of human epidermal growth factor receptor type 2 (HER2). Methods: The HER2-specific sdAbs and HER2-irrelevant VHH_2001 were labeled using N-succinimidyl-3-211At-astato-5-guanidinomethyl benzoate (iso-211At-SAGMB). The cytotoxicity of iso- 211At-SAGMB-5F7 and iso- 211At-SAGMB-VHH_2001 were compared on HER2-expressing BT474 breast carcinoma cells. Three experiments in mice with subcutaneous BT474 xenografts were performed to evaluate the therapeutic effectiveness of single doses of iso- 211At-SAGMB-5F7 (0.7-3.0 MBq), iso- 211At-SAGMB-VHH_1028 (1.0-3.0 MBq), and iso- 211At-SAGMB-VHH_1028 and iso- 211At-SAGMB-VHH_2001 (∼1.0 MBq). Results: Clonogenic survival of BT474 cells was reduced after exposure to iso- 211At-SAGMB-5F7 (D0 = 1.313 kBq/mL) whereas iso- 211At-SAGMB-VHH_2001 was ineffective. Dose-dependent tumor growth inhibition was observed with 211At-labeled HER2-specific 5F7 and VHH_1028 but not with HER2-irrelevant VHH_2001. At the 3.0-MBq dose, complete tumor regression was seen in 3 of 4 mice treated with iso- 211At-SAGMB-5F7 and 8 of 11 mice treated with iso- 211At-SAGMB-VHH_1028; prolongation in median survival was 495% and 414%, respectively. Conclusion: Combining rapidly internalizing, high-affinity HER2-targeted sdAbs with the iso- 211At-SAGMB residualizing prosthetic agent is a promising strategy for targeted α-particle therapy of HER2-expressing cancers.
Collapse
Affiliation(s)
- Yutian Feng
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Rebecca Meshaw
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Xiao-Guang Zhao
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Stephen Jannetti
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | | | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
10
|
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: 2] [Impact Index Per Article: 0.7] [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
|