1
|
Xu Z, Sinha A, Pandya DN, Schnicker NJ, Wadas TJ. Cryo-electron microscopy reveals a single domain antibody with a unique binding epitope on fibroblast activation protein alpha. RSC Chem Biol 2025; 6:780-787. [PMID: 39975582 PMCID: PMC11834968 DOI: 10.1039/d4cb00267a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/06/2025] [Indexed: 02/21/2025] Open
Abstract
Fibroblast activation protein alpha (FAP) is a serine protease that is expressed at basal levels in benign tissues but is overexpressed in a variety of pathologies, including cancer. Despite this unique expression profile, designing functional diagnostic and therapeutic agents that effectively target this biomarker remains elusive. Here we report the structural characterization of the interaction between a novel single domain antibody (sdAb), I3, and FAP using cryo-electron microscopy. The reconstructions were determined to a resolution of 2.7 Å and contained two distinct populations; one I3 bound and two I3 molecules bound to the FAP dimer. In both cases, the sdAb bound a unique epitope that was distinct from the active site of the enzyme. Furthermore, this report describes the rational mutation of specific residues within the complementarity determining region 3 (CDR3) loop to enhance affinity and selectivity of the I3 molecule for FAP. This report represents the first sdAb-FAP structure to be described in the literature.
Collapse
Affiliation(s)
- Zhen Xu
- Protein and Crystallography Facility, University of Iowa Iowa City Iowa 52242 USA
| | - Akesh Sinha
- Department of Radiology, University of Iowa Iowa City Iowa 52242 USA
| | - Darpan N Pandya
- Department of Radiology, University of Iowa Iowa City Iowa 52242 USA
| | - Nicholas J Schnicker
- Protein and Crystallography Facility, University of Iowa Iowa City Iowa 52242 USA
- Department of Molecular Physiology and Biophysics, University of Iowa Iowa City Iowa 52242 USA
| | - Thaddeus J Wadas
- Department of Radiology, University of Iowa Iowa City Iowa 52242 USA
| |
Collapse
|
2
|
Weng D, Guo R, Gao Y, Xu S, Li Y, Zhou J, An R, Xu H. Immuno-PET Imaging of a 68Ga-Labeled Single-Domain Antibody for Detecting Tumor TIGIT Expression. Mol Pharm 2025. [PMID: 40298304 DOI: 10.1021/acs.molpharmaceut.4c00900] [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: 04/30/2025]
Abstract
Preclinical studies have shown that the expression of T cell immunoglobulin and the immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) in the tumor microenvironment is associated with the efficacy of anti-TIGIT-based immunotherapy. This study aimed to develop a TIGIT single-domain antibody (sdAb)-based positron emission tomography (PET) radiotracer, [68Ga]Ga-NOTA-NABT3A1, and evaluate its characteristics. The NABT3A1 was modified with a NOTA derivative and radiolabeled with 68Ga. In vitro stability of [68Ga]Ga-NOTA-NABT3A1 was assessed in phosphate-buffered saline (PBS) and fetal bovine serum (FBS), along with its specificity for TIGIT stably transfected A375 Human melanoma cells (A375-TIGIT) was performed. In vivo imaging was conducted on A375-TIGIT tumor-bearing nude mice at different time points after the injection of [68Ga]Ga-NOTA-NABT3A1. The synthesized [68Ga]Ga-NOTA-NABT3A1 achieved a radiochemical yield of 70.56 ± 1.14% and purity levels of 95.80 ± 0.58% in PBS and 96.79 ± 1.69% in FBS at 2 h. Immuno-PET imaging revealed specific accumulation of [68Ga]Ga-NOTA-NABT3A1 in A375-TIGIT tumor-bearing nude mice, with a maximum uptake of 3.86 ± 0.29% injected dose/g at 0.5 h. Biodistribution and immunohistochemical analyses confirmed the in vivo imaging results. In conclusion, we successfully synthesized an NABT3A1-derived PET radiotracer with the potential to noninvasively assess TIGIT expression in tumors.
Collapse
Affiliation(s)
- Dinghu Weng
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 201318 Shanghai, China
- Department of Medical Imaging, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, 430071 Wuhan, China
- Wuhan Clinical Research and Development Center of Brain Resuscitation and Functional Imaging, 430071 Wuhan, China
| | - Rong Guo
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430000, China
| | - Yu Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430000, China
| | - Shasha Xu
- Beijing Novaboody Biotechnological Ltd., Beijing 102600, China
| | - Yingying Li
- Beijing Novaboody Biotechnological Ltd., Beijing 102600, China
| | - Jun Zhou
- Interventional Diagnostic and Therapeutic Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Rui An
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430000, China
| | - Haibo Xu
- Department of Medical Imaging, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, 430071 Wuhan, China
- Wuhan Clinical Research and Development Center of Brain Resuscitation and Functional Imaging, 430071 Wuhan, China
| |
Collapse
|
3
|
Murugan D, Thirumalaiswamy HV, Murugesan V, Venkatesan J, Balachandran U, Lakshminarayanan K, Satpati D, Nikolić S, Rangasamy L. Unlocking the power of affibody conjugated radioactive metallopharmaceuticals for targeted cancer diagnosis and therapy. Pharmacol Ther 2025:108863. [PMID: 40294752 DOI: 10.1016/j.pharmthera.2025.108863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/02/2025] [Accepted: 04/20/2025] [Indexed: 04/30/2025]
Abstract
Cancer is the second-largest death-causing disease after cardiovascular diseases. Effective research on cancer diagnosis and subsequent elimination plays a vital role in reducing the cancer-related death toll. Radiotherapy is one of the best strategies that could kill masses of solid tumour tissues; however, the efficacy is limited due to the bystander effect. This issue could be solved by the emergence of targeted delivery of radiometallic complexes, enabling clinicians to monitor the tumour regions and effectively destroy the tumour. Affibody® molecules are a class of synthetic peptides known as antibody mimics having the binding sites of an antibody. The specificity of affibodies is found to be greater than that of antibodies due to their small size. This review intends to highlight the recent developments in the field of affibody-targeted radiometallopharmaceuticals. These approaches could be essential for early cancer detection, tumour staging, and monitoring the response to therapy and could produce better therapeutic outcomes. In an attempt to provide ideas and inspiration for the researchers to design affibody-conjugated radiopharmaceuticals that are clinically applicable, we have provided an in-depth exploration of the various types of affibody-conjugated radiopharmaceuticals that are currently in clinical trials and various other pre-clinically tested conjugates in this article. Only a few review reports on affibody-conjugated radiometallopharmaceuticals, typically focusing on a specific molecular target or radionuclides reported. In this review, we provide a comprehensive overview of most radiometals, such as 111In, 68Ga, 64Cu, 55Co, 57Co, 44Sc, 99mTc, 89Zr, 90Y, 211At, 188Re, and 177Lu, choice of chelators, and potential cancer-associated molecular targets such HER2, EGFR or HER1, HER3, IGF-1R, PDGFRβ, VEGFR2, PD-L1, CAIX, PD-L1, neonatal Fc receptor (FcRn) and B7-H3. This approach highlights the advancements made over the past twenty years in affibody conjugates for radio imaging and therapy in oncology.
Collapse
Affiliation(s)
- Dhanashree Murugan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; School of Biosciences & Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Harashkumar Vasanthakumari Thirumalaiswamy
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Vasanth Murugesan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Janarthanan Venkatesan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Unnikrishnan Balachandran
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Kalaiarasu Lakshminarayanan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Drishty Satpati
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, Maharashtra 400085, India; Homi Bhabha National Institute, Mumbai 400094, India
| | - Stefan Nikolić
- Innovative Centre of the Faculty of Chemistry Belgrade, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia
| | - Loganathan Rangasamy
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
4
|
Wei W, Grünwald V, Herrmann K. CD70-targeted cancer theranostics: Progress and challenges. MED 2025:100671. [PMID: 40250430 DOI: 10.1016/j.medj.2025.100671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/20/2025]
Abstract
CD70, a tumor-associated antigen, exhibits elevated expression in clear cell renal cell carcinoma (ccRCC), nasopharyngeal carcinoma, and lymphoma, among others, with minimal presence in healthy tissues. CD70 has emerged as a promising biomarker for molecular imaging, targeted therapies, and immunotherapies. ImmunoPET imaging with single-domain antibody-derived tracers, such as [18F]RCCB6 and [68Ga]Ga-NOTA-RCCB6, demonstrates exceptional diagnostic precision, identifying both common and rare metastases from ccRCC. This capability enhances staging accuracy and further enables early intervention. Beyond diagnostics, CD70-targeted imaging optimizes patient selection for emerging therapies, such as CAR-T cells and antibody-drug conjugates, by stratifying candidates based on their CD70 expression levels. It also supports real-time monitoring of therapeutic responses, enabling dynamic adjustments to treatment. The integration of these imaging tools into clinical workflows enhances personalized treatment efficacy for CD70-expressing cancers. Radiotheranostic strategies can further allow the simultaneous diagnosis and treatment of malignancies, opening new horizons for the precise management of CD70+ tumors.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, China.
| | - Viktor Grünwald
- Department of Urology, Department of Medical Oncology, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Ken Herrmann
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany; Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
5
|
Xi X, Guo S, Gu Y, Wang X, Wang Q. Challenges and opportunities in single-domain antibody-based tumor immunotherapy. Biochim Biophys Acta Rev Cancer 2025; 1880:189284. [PMID: 39947441 DOI: 10.1016/j.bbcan.2025.189284] [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: 09/17/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025]
Abstract
Single-domain antibodies (sdAbs) have emerged as a promising tool in tumor immunotherapy, garnering significant attention in recent years due to their unique structure and superior properties. Unlike traditional antibodies, sdAbs exhibit several advantages, including small molecular weight, high stability, strong affinity, and high specificity. These characteristics enable sdAbs to effectively target and eliminate tumor cells within the complex tumor microenvironment. Moreover, their structural advantages enhance tissue penetration and reduce immunogenicity, thereby increasing their potential for clinical application. The potential applications of sdAbs include novel immune checkpoint inhibitors, bispecific antibody drugs, innovative immune cell therapies, antibody-drug conjugate therapies, and tumor molecular imaging diagnostics. Despite the promising prospects, several challenges of sdAb-based tumor immunotherapy still require further investigation. This review aims to summarize the status of sdAb-based immunotherapy, identify the challenges encountered, and evaluate the clinical research and application potential of sdAbs in this field.
Collapse
Affiliation(s)
- Xiaozhi Xi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan 250022, People's Republic of China.; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, People's Republic of China.; Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003 Qingdao, People's Republic of China
| | - Shasha Guo
- Shandong Women's University, 250355 Jinan, People's Republic of China
| | - Yuchao Gu
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xuekai Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan 250022, People's Republic of China.; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, People's Republic of China
| | - Qiang Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan 250022, People's Republic of China.; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, People's Republic of China.; Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003 Qingdao, People's Republic of China.
| |
Collapse
|
6
|
Li W, Xu Z, He Q, Pan J, Zhang Y, El-Sheikh ESA, Hammock BD, Li D. Nanobody-Based Immunoassays for the Detection of Food Hazards-A Review. BIOSENSORS 2025; 15:183. [PMID: 40136980 PMCID: PMC11939871 DOI: 10.3390/bios15030183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025]
Abstract
Food safety remains a significant global challenge that affects human health. Various hazards, including microbiological and chemical threats, can compromise food safety throughout the supply chain. To address food safety issues and ensure public health, it is necessary to adopt rapid, accurate, and highly specific detection methods. Immunoassays are considered to be an effective method for the detection of highly sensitive biochemical indicators and provide an efficient platform for the identification of food hazards. In immunoassays, antibodies function as the primary recognition elements. Nanobodies have significant potential as valuable biomolecules in diagnostic applications. Their distinctive physicochemical and structural characteristics make them excellent candidates for the development of reliable diagnostic assays, and as promising alternatives to monoclonal and polyclonal antibodies. Herein, we summarize a comprehensive overview of the status and prospects of nanobody-based immunoassays in ensuring food safety. First, we begin with a historical perspective on the development of nanobodies and their unique characteristics. Subsequently, we explore the definitions and boundaries of immunoassays and immunosensors, before discussing the potential applications of nanobody-based immunoassays in food safety testing that have emerged over the past five years, and follow the different immunoassays, highlighting their advantages over traditional detection methods. Finally, the directions and challenges of nanobody-based immunoassays in food safety are discussed. Due to their remarkable sensitivity, specificity and versatility, nanobody-based immunoassays hold great promise in revolutionizing food safety testing and ensuring public health and well-being.
Collapse
Affiliation(s)
- Wenkai Li
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (W.L.); (Z.X.); (Q.H.); (J.P.); (Y.Z.)
- Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | - Zhihao Xu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (W.L.); (Z.X.); (Q.H.); (J.P.); (Y.Z.)
- Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | - Qiyi He
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (W.L.); (Z.X.); (Q.H.); (J.P.); (Y.Z.)
- Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | - Junkang Pan
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (W.L.); (Z.X.); (Q.H.); (J.P.); (Y.Z.)
- Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | - Yijia Zhang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (W.L.); (Z.X.); (Q.H.); (J.P.); (Y.Z.)
- Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | | | - Bruce D. Hammock
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, USA
| | - Dongyang Li
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (W.L.); (Z.X.); (Q.H.); (J.P.); (Y.Z.)
- Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| |
Collapse
|
7
|
Ma X, Liu T, Guo R, Zhou W, Yao Y, Wen D, Tao J, Zhu J, Wang F, Zhu H, Yang Z. Radioiodinated Nanobody immunoPET probe for in vivo detection of CD147 in pan-cancer. Eur J Nucl Med Mol Imaging 2025; 52:1406-1421. [PMID: 39549046 DOI: 10.1007/s00259-024-06985-3] [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: 06/30/2024] [Accepted: 11/09/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND To develop the extracellular matrix metalloproteinase inducer (CD147)-targeting therapeutic strategies, accurate detection of CD147 expression in tumors is crucial. Owing to their relatively low molecular weights and high affinities, nanobodies (Nbs) may be powerful candidates for cancer diagnosis and therapy. In this study, we developed a novel CD147-targeted nanobody radiotracer, [124I]I-NB147, which provides guidance for the noninvasive detection of CD147-overexpressing cancers. METHODS CD147 expression in human cancers was detected via immunohistochemistry (IHC) on tissue microarrays (TMAs). Western blot (WB) and flow cytometry were used to screen CD147-positive malignant melanoma (MM), triple-negative breast cancer (TNBC), and pancreatic cancer (PCA) cell lines. The CD147 nanobody (NB147) was labeled with [124I]INa using Iodogen as the oxidizing agent and was purified by the PD-10 column. The physicochemical properties, affinity, metabolic characteristics, biodistribution, and immunoPET imaging of [124I]I-NB147 were evaluated Moreover, [18F]F-FDG was used as a control. Finally, CD147 expression analysis was performed via multiplex immunofluorescence (MxIF) and autoradiography on human cancer specimens and tumor-bearing mice tissues. RESULTS TMAs results revealed that CD147 is highly expressed in MM, TNBC, and PCA. A CD147-specific nanobody, NB147, was successfully generated with excellent in vitro binding characteristics. [124I]I-NB147 was obtained with high radiochemical yield and purity, and was stable for at least 4 h in vitro. WB and FCM revealed that CD147 was positive in A375, MDA-MB-435 and ASPC1 cells, whereas SK-MEL-28, 4T1 and BXPC3 cells presented low expression levels. The radio-ELISA results indicated that [124I]I-NB147 had a high binding affinity to CD147. The uptake of [124I]I-NB147 was significantly different between CD147 high-expression cells and CD147 low-expression cells (P < 0.001). The biological half-life of the distribution and clearance phases were 0.05 h and 1.58 h, respectively. In CD147-positive tumor models, the [124I]I-NB147 accumulated in A375, MDA-MB-435, and ASPC1 tumors, and the uptake value was significantly higher than that of [18F]F-FDG. Uptake in SK-MEL-28, BXPC3, and 4T1 tumors was not clearly observed. Finally, through autoradiography and histological studies, the correlation analysis between tumor uptake and CD147 expression level was determined. CONCLUSIONS The high expression of CD147 in MM, TNBC, and PCA tissuesand in tumor cells was verified. The CD147 nanobody, NB147 was produced and radiolabeled to obtain the immunoPET probe, [124I]I-NB147, which showed high affinity to CD147 and precise visualization for accurate diagnosis of CD147-expressing lesions in different cancers. These results provide insight into the imaging and binding properties of nanobody NB147 over extended periods of time, reinforcing its potential in developing radionuclide therapies for CD147-positive cancer patients.
Collapse
Affiliation(s)
- Xiaokun Ma
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Teli Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Rui Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Wenyuan Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yuan Yao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Dan Wen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, 100142, China
- Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637002, Sichuan, China
| | - Jinping Tao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jinyu Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Feng Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Hua Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Zhi Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
8
|
Martinez-Orengo N, Shah S, Lai J, Basuli F, Lyndaker A, Turner ML, Peiravi M, Sourabh S, Sampson K, Zhang P, Swenson RE, Lusso P, Maldarelli F, Nath A, Lau CY, Hammoud DA. PET imaging of HIV-1 envelope protein gp120 using 18F-labeled nanobodies. iScience 2025; 28:111795. [PMID: 39917021 PMCID: PMC11800091 DOI: 10.1016/j.isci.2025.111795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/02/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025] Open
Abstract
Radiolabeled antibodies against the HIV-1 envelope protein, gp120, have been previously tested in animal models and in people with HIV (PWH). Nanobodies offer advantages over antibodies, including smaller size and faster clearance, which allow labeling with fluorine-18. In this study, three nanobodies (J3, 3E3, B9) chosen based on their binding properties to the conserved CD4-binding site of gp120 were labeled with fluorine-18 and used for PET imaging in mice bearing wild-type (WT) and/or gp120-expressing (Env+) tumors. [18F]J3 and [18F]3E3 selectively targeted Env+ tumors and not WT tumors, with minimal background signal. Switching from non-site-specific radiolabeling method to sortase A-mediated site-specific conjugation at the C-terminus improved binding to Env+ tumors for all nanobodies. Site-specifically 18F-labeled J3 nanobody is the most promising candidate with the highest level of binding. These results establish an Env+ imaging method that will enable next stage testing in an HIV-1 preclinical infection model and potentially in PWH.
Collapse
Affiliation(s)
- Neysha Martinez-Orengo
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Swati Shah
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Jianhao Lai
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Rockville, MD, USA
| | - Anna Lyndaker
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Mitchell L. Turner
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Morteza Peiravi
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Suman Sourabh
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Kevon Sampson
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA
| | - Peng Zhang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Rolf E. Swenson
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Rockville, MD, USA
| | - Paolo Lusso
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Frank Maldarelli
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, USA
| | - Avindra Nath
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA
| | - Chuen-Yen Lau
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, USA
| | - Dima A. Hammoud
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
9
|
Li Y, Li D, Lin H, Wang D, Zhao J, Wang Z, Hong H, Wu Z. Reconstituting the immune killing functions and improving the pharmacokinetics of nanobodies by rhamnolipid conjugation. J Control Release 2025; 378:18-26. [PMID: 39637990 DOI: 10.1016/j.jconrel.2024.11.080] [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: 06/25/2024] [Revised: 10/20/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
Nanobodies (Nbs) hold great promise as next-generation cancer immunotherapies, but their efficacy is hindered by their poor pharmacokinetics and the inability to trigger Fc-mediated immune killing functions. To address these limitations, we designed and synthesized rhamnolipid-modified Nbs as a type of antibody-recruiting molecule by site-specifically conjugating EGFR-targeting Nb 7D12 to a series of rhamnolipid derivatives, and their biological profiles were evaluated in vitro and in vivo. Investigation of the structure-activity relationship revealed that the number of rhamnose (Rha) units and the length of the PEG linker in the conjugates affected anti-tumor activities. Conjugate R5, which contained two Rha units and a PEG2 linker, exhibited the most potent antibody-dependent cell-mediated phagocytosis (ADCP) and complement-dependent cytotoxicity (CDC) activities. In vivo, R5 had a significantly longer half-life because of its ability to bind to serum albumin and endogenous anti-Rha antibodies, and it demonstrated potent in vivo antitumor activity in a xenograft mouse model of A431 tumor. Our findings highlight the potential of rhamnolipidation as a strategy to enhance the efficacy of Nbs in cancer immunotherapy and provide a cost-effective platform for improving the therapeutic efficiency of Nbs.
Collapse
Affiliation(s)
- Yanchun Li
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Dan Li
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Han Lin
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Di Wang
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Jie Zhao
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Zheng Wang
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Haofei Hong
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Zhimeng Wu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
10
|
Tran HH, Yamaguchi A, Manning HC. Radiotheranostic landscape: A review of clinical and preclinical development. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07103-7. [PMID: 39891713 DOI: 10.1007/s00259-025-07103-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Radiotheranostics combines diagnostic imaging with targeted radionuclide therapy, representing a transformative approach in precision oncology. Landmark approvals of Lutathera® and Pluvicto® have catalyzed significant advancements in this field, driving research into novel radionuclides, targeting strategies, and clinical applications. This review evaluates the evolving clinical and preclinical landscape of radiotheranostics, highlighting advancements, emerging trends, and persistent challenges in radionuclide therapy. METHODS A comprehensive analysis was performed, encompassing active clinical trials as of December 2024, sourced from ClinicalTrials.gov and TheranosticTrials.org. Preclinical developments were evaluated through a review of recent literature, focusing on innovations in radionuclide production, targeting molecules, and radiochemistry. RESULTS In reviewing the clinical landscape, agents targeting somatostatin receptors (SSTR) and prostate-specific membrane antigen (PSMA) still dominate the field, but new targets such as fibroblast activation protein (FAP), integrins, and gastrin-releasing peptide receptors (GRPR) are gaining traction in both clinical and preclinical development. While small molecules and peptides remain the most common radionuclide carriers, antibody-based carriers including bispecific antibodies, immunoglobin-derived antigen-binding fragments, and antibody-mimetic proteins are on the rise due to their specificity and adaptability. Innovations in radioligand design are driving a shift from agonists to antagonists, accompanied by the development of modified peptides with enhanced pharmacokinetics and tumor-targeting properties. Next-generation therapeutic radionuclides, such as the beta-emitter terbium-161 and alpha-emitters actinium-225 and lead-212, are under investigation to complement or replace lutetium-177, addressing the need for improved efficacy and reduced toxicity. Paired isotopic radionuclides are gaining popularity for their ability to optimize imaging and therapeutic dosimetry as they offer near-identical specificity, biodistribution, and metabolism. Additionally, radiohybrid systems represent an innovative approach to chelating chemically distinct radionuclide pairs within a single molecule, further enhancing flexibility in radiotheranostic design. CONCLUSION Radiotheranostics has transformed cancer care through its precision and adaptability, but challenges in radionuclide production, regulatory frameworks, and workforce training hinder broader adoption. Advances in isotopic pairing, next-generation radionuclides, and radiohybrid systems in preclinical and clinical settings hold promise to overcome these barriers. Collaborative efforts among academia, industry, and regulatory bodies are critical to accelerating innovation and optimizing clinical outcomes.
Collapse
Affiliation(s)
- Ha H Tran
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aiko Yamaguchi
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - H Charles Manning
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
11
|
Yang Y, Ren Z, Wang D, Tang D, Wei W, Song S, Lyu Y, Ding D, Tan W. Framework Nucleic Acid-Nanobody Fusion Probe-Based Pharmacokinetics Modulation and Analysis for Efficient Positron Emission Tomography Imaging. ACS NANO 2025; 19:439-450. [PMID: 39714093 DOI: 10.1021/acsnano.4c09127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Nanobodies are promising for immunoPET imaging due to their excellent antigen recognition and tumor targeting, yet rapid clearance limits their tumor accumulation. Although multimerization and albumin binding can extend their circulation time and improve tumor targeting, a simple and universal method for creating protein multimers is still needed. Here, we leveraged the facile synthesis, controllable size, and precise assembly of DNA nanotechnology to construct CD47-targeted framework nucleic acid-nanobody fusion probes with multiple valences and sizes. Following comprehensive structural characterization, in vitro specificity assessment and in vivo PET/CT imaging analysis were conducted on a colorectal cancer LS174T mouse model. Furthermore, a pharmacokinetic model was developed and fitted with considerable in vivo data to prove its rationality, followed by testing the effects on tumor uptake prediction by changing different pharmacokinetic parameters. Indeed, by manipulating the size of the nucleic acid scaffolding and the number of attached nanobodies, we could precisely modulate the accumulation of probes at the tumor site. Overall, this study not only developed an efficient strategy for constructing nanobody multimers but also provided a pharmacokinetic model, allowing profound insight into the multidimensional data obtained experimentally and informing the design of future imaging probes with predictable delivery efficacies.
Collapse
Affiliation(s)
- Yani Yang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Zhiqiang Ren
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dan Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Decui Tang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yifan Lyu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- Furong Laboratory, Changsha, Hunan 410082, China
| | - Ding Ding
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Weihong Tan
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
12
|
He Y, Tian R, Xu D, Wu Y, Rina S, Chen T, Guan Y, Xie T, Ying T, Xie F, Han J. Preclinical evaluation and pilot clinical study of [ 68Ga]Ga-NOTA-H006 for non-invasive PET imaging of 5T4 oncofetal antigen. Eur J Nucl Med Mol Imaging 2025; 52:611-622. [PMID: 39377811 DOI: 10.1007/s00259-024-06941-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
PURPOSE Trophoblast glycoprotein, the so-called 5T4, is an oncofetal antigen expressed in many different cancers. However, no 5T4-specific radioligand is employed in the clinic for non-invasive diagnosis. Thus, the aim of the current study was to develop a PET radiotracer for imaging 5T4 expression in preclinical and clinical stages. METHODS A VHH library was constructed by camel immunization. The specificity of the VHHs toward 5T4 antigen was screened through phage display biopanning and periplasmic extract enzyme-linked immunosorbent assay. 1,4,7-Triazacyclononane-1,4,7-triacetate acid (NOTA) derivative was conjugated to the selected VHH. After radiolabeling, microPET/CT and ex vivo biodistribution were conducted using BxPC-3 and MDA-MB-468 tumor-bearing mice. Cold VHH was co-injected with the tracer to challenge its binding in vivo. For the pilot clinical study, PET/CT images were acquired at 1 h after injection of tracer in patients with pathologically confirmed primary and metastatic tumors. RESULTS A library with a capacity of 1.2 × 1012 colony-forming units was constructed after successful camel immunization. Nb1-40 with a median effect concentration of 0.43 nM was selected. After humanization, the resulting H006 maintained a high affinity towards 5T4. [68Ga]Ga-NOTA-H006 with the molar activities of 6.48-54.2 GBq/µmol was prepared with high radiochemical purity (> 98%). Using [68Ga]Ga-NOTA-H006, microPET/CT revealed a clear visualization of 5T4 expression in BxPC-3 tumor-bearing mice. Ex vivo biodistribution showed that the highest tumor-to-blood ratio (∼ 3-fold) and tumor-to-muscle ratio (∼ 5-fold) were achieved at 60 min post-injection. Co-injection of the cold H006 at a dose of 1.5 mg/kg significantly reduced the tumor uptake (p < 0.0001). In the pilot clinical study, [68Ga]Ga-NOTA-H006 demonstrated its capacity to map 5T4-positive lesions in humans and yielded a mean effective dose of 3.4 × 10- 2 mSv/MBq. CONCLUSIONS [68Ga]Ga-NOTA-H006, which can visualize 5T4 expression in vivo, has been successfully developed. This opens up opportunities for non-invasively studying 5T4 expression through nuclear medicine. Further clinical investigations are warranted to explore its clinical value in disease progression and companion diagnosis.
Collapse
Affiliation(s)
- Yingfang He
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Ruhua Tian
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Dong Xu
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanfei Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China
| | - Sa Rina
- Huahe Pharmaceutical Co., Ltd, Shanghai, China
| | - Tengxiang Chen
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China
| | - Tianwu Xie
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China.
| | - Junbin Han
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China.
| |
Collapse
|
13
|
Wang Y, Zhang G, Zhou Z, Zhang N, Jiang H, Liu Y, Fu T, Zhu Y, Li J. Antitumor Activity of a Bispecific Chimera Targeting EGFR and Met in Gefitinib-Resistant Non-Small Cell Lung Cancer. Adv Healthc Mater 2025; 14:e2402884. [PMID: 39586988 DOI: 10.1002/adhm.202402884] [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: 08/03/2024] [Revised: 09/28/2024] [Indexed: 11/27/2024]
Abstract
Non-small cell lung cancers (NSCLC) frequently acquire resistance to tyrosine kinase inhibitors (TKI) due to epidermal growth factor receptor (EGFR) mutation or activation of the bypass pathway involving mesenchymal-epithelial transition factor (Met). To address this challenge, a bispecific nanobody-aptamer chimera is designed to target mutated EGFR and Met simultaneously to block their cross-talk in NSCLC. The EGFR-Met chimera is cost-effectively engineered using microbial transglutaminase and click chemistry strategies. With enhanced binding affinity toward the target proteins, the as-developed chimera inhibits efficiently the cross-talk between signaling pathways associated with EGFR and Met. This inhibition leads to the suppression of downstream pathways, such as Erk and Akt, and induces upregulation of cell cycle arrest-related proteins, including Rb, p21, and p27. Additionally, the chimera activates the caspase-dependent apoptotic signaling pathway. Consequently, it inhibits cell migration, induces cell death, and causes cell cycle arrest in vitro. Moreover, the chimera exhibits significant antitumor efficacy in drug-resistant xenograft mouse models, showcasing improved tissue penetration and low toxicity. This study accentuates the potential of the bispecific EGFR-Met chimera as a promising therapeutic option for NSCLC resistant to EGFR TKIs.
Collapse
Affiliation(s)
- Ya Wang
- School of Chemistry and Materials, University of Science and Technology of China, Hefei, 230026, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Guixi Zhang
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Zhilan Zhou
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Ning Zhang
- School of Chemistry and Materials, University of Science and Technology of China, Hefei, 230026, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Hang Jiang
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yichang Liu
- School of Pharmacy, Nantong University, Nantong, 226019, China
| | - Ting Fu
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yingdi Zhu
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Juan Li
- School of Chemistry and Materials, University of Science and Technology of China, Hefei, 230026, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| |
Collapse
|
14
|
Xu Z, Sinha A, Pandya DN, Schnicker NJ, Wadas TJ. Cryo-electron microscopy reveals a single domain antibody with a unique binding epitope on fibroblast activation protein alpha. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.619146. [PMID: 39463996 PMCID: PMC11507940 DOI: 10.1101/2024.10.18.619146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Fibroblast activation protein alpha (FAP) is a serine protease that is expressed at basal levels in benign tissues but is overexpressed in a variety of pathologies, including cancer. Despite this unique expression profile, designing effective diagnostic and therapeutic agents that effectively target this biomarker remain elusive. Here we report the structural characterization of the interaction between a novel single domain antibody (sdAbs), I3, and FAP using cryo-electron microscopy. The reconstructions were determined to a resolution of 2.7 Å and contained two distinct populations; one I3 bound and two I3 molecules bound to the FAP dimer. In both cases, the sdAbs bound a unique epitope that was distinct from the active site of the enzyme. Furthermore, this report describes the rational mutation of specific residues within the complementarity determining region 3 (CDR3) loop to enhance affinity and selectivity of the I3 molecule for FAP. This report represents the first sdAb-FAP structure to be described in the literature.
Collapse
Affiliation(s)
- Zhen Xu
- Protein and Crystallography Facility, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Akesh Sinha
- Department of Radiology, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Darpan N. Pandya
- Department of Radiology, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Nicholas J. Schnicker
- Protein and Crystallography Facility, University of Iowa, Iowa City, Iowa, 52242, USA
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Thaddeus J. Wadas
- Department of Radiology, University of Iowa, Iowa City, Iowa, 52242, USA
| |
Collapse
|
15
|
Hao Z, Dong X, Zhang Z, Qin Z. A Nanobody of PEDV S1 Protein: Screening and Expression in Escherichia coli. Biomolecules 2024; 14:1116. [PMID: 39334881 PMCID: PMC11430113 DOI: 10.3390/biom14091116] [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: 08/02/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) has caused significant economic losses to the pig farming industry in various countries for a long time. Currently, there are no highly effective preventive or control measures available. Research into the pathogenic mechanism of PEDV has shown that it primarily causes infection by binding the S protein to the CD13 (APN) receptor on the membrane of porcine intestinal epithelial cells. The S1 region contains three neutralization epitopes and multiple receptor-binding domains, which are closely related to viral antigenicity and ad-sorption invasion. Nanobodies are a type of single-domain antibody that have been discovered in recent years. They can be expressed on a large scale through prokaryotic expression systems, which makes them cost-effective, stable, and less immunogenic. This study used a phage display library of nanobodies against the PEDV S1 protein. After three rounds of selection and enrichment, the DNA sequence of the highly specific nanobody S1Nb1 was successfully obtained. To obtain soluble nanobody S1Nb1, its DNA sequence was inserted into the vector Pcold and a solubility-enhancing SUMO tag was added. The resulting recombinant vector, Pcold-SUMO-S1Nb1, was then transformed into E. coli BL21(DE3) to determine the optimal expression conditions for the nanobody. Following purification using Ni-column affinity chromatography, Western blot analysis confirmed the successful purification of S1Nb1 carrying the solubility-enhancing tag. ELISA results demonstrated a strong affinity between the S1Nb1 nanobody and PEDV S1 protein.
Collapse
Affiliation(s)
| | | | | | - Zhihua Qin
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China; (Z.H.); (X.D.); (Z.Z.)
| |
Collapse
|
16
|
Zhang J, Du B, Wang Y, Cui Y, Wang S, Zhao Y, Li Y, Li X. The role of CD8 PET imaging in guiding cancer immunotherapy. Front Immunol 2024; 15:1428541. [PMID: 39072335 PMCID: PMC11272484 DOI: 10.3389/fimmu.2024.1428541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Currently, immunotherapy is being widely used for treating cancers. However, the significant heterogeneity in patient responses is a major challenge for its successful application. CD8-positive T cells (CD8+ T cells) play a critical role in immunotherapy. Both their infiltration and functional status in tumors contribute to treatment outcomes. Therefore, accurate monitoring of CD8+ T cells, a potential biomarker, may improve therapeutic strategy. Positron emission tomography (PET) is an optimal option which can provide molecular imaging with enhanced specificity. This review summarizes the mechanism of action of CD8+ T cells in immunotherapy, and highlights the recent advancements in PET-based tracers that can visualize CD8+ T cells and discusses their clinical applications to elucidate their potential role in cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yaming Li
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuena Li
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
17
|
Wu Q, Wu Y, Zhang Y, Guan Y, Huang G, Xie F, Liu J, Zhai W, Wei W. ImmunoPET/CT imaging of clear cell renal cell carcinoma with [ 18F]RCCB6: a first-in-human study. Eur J Nucl Med Mol Imaging 2024; 51:2444-2457. [PMID: 38480552 DOI: 10.1007/s00259-024-06672-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/05/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE The cluster of differentiation (CD70) is a potential biomarker of clear cell renal cell carcinoma (ccRCC). This study aims to develop CD70-targeted immuno-positron emission tomography/computed tomography (immunoPET/CT) imaging tracers and explore the diagnostic value in preclinical studies and the potential value in detecting metastases in ccRCC patients. METHODS Four novel CD70-specific single-domain antibodies (sdAbs) were produced and labelled with 18F by the aluminium fluoride restrained complexing agent (AlF-RESCA) method to develop radiotracers. The visualisation properties of the tracers were evaluated in a subcutaneous ccRCC patient-derived xenograft (PDX) model. In a registered prospective clinical trial (NCT06148220), six patients with pathologically confirmed RCC were included and underwent immunoPET/CT examination exploiting one of the developed tracers (i.e., [18F]RCCB6). RESULTS We engineered four sdAbs (His-tagged RCCB3 and RCCB6, His-tag-free RB3 and RB6) specifically targeting recombinant human CD70 without cross-reactivity to murine CD70. ImmunoPET/CT imaging with [18F]RCCB3 and [18F]RCCB6 demonstrated a high tumour-to-background ratio in a subcutaneous ccRCC PDX model, with the latter showing better diagnostic potential supported by higher tumour uptake and lower bone accumulation. In comparison, [18F]RB6, developed by sequence optimisation, has significantly lower kidney accumulation than that of [18F]RCCB6. In a pilot translational study, [18F]RCCB6 immunoPET/CT displayed ccRCC metastases in multiple patients and demonstrated improved imaging contrast and diagnostic value than 18F-FDG PET/CT in a patient with ccRCC. CONCLUSION The work successfully developed a series of CD70-targeted immunoPET/CT imaging tracers. Of them, [18F]RCCB6 clearly and specifically identified inoculated ccRCCs in preclinical studies. Clinical translation of [18F]RCCB6 suggests potential for identifying recurrence and/or metastasis in ccRCC patients.
Collapse
Affiliation(s)
- 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
| | - Yanfei Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, 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
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, 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
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, 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.
| | - Wei Zhai
- Department of Urology, 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.
| |
Collapse
|
18
|
Badenhorst M, Windhorst AD, Beaino W. Navigating the landscape of PD-1/PD-L1 imaging tracers: from challenges to opportunities. Front Med (Lausanne) 2024; 11:1401515. [PMID: 38915766 PMCID: PMC11195831 DOI: 10.3389/fmed.2024.1401515] [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: 03/15/2024] [Accepted: 05/20/2024] [Indexed: 06/26/2024] Open
Abstract
Immunotherapy targeted to immune checkpoint inhibitors, such as the program cell death receptor (PD-1) and its ligand (PD-L1), has revolutionized cancer treatment. However, it is now well-known that PD-1/PD-L1 immunotherapy response is inconsistent among patients. The current challenge is to customize treatment regimens per patient, which could be possible if the PD-1/PD-L1 expression and dynamic landscape are known. With positron emission tomography (PET) imaging, it is possible to image these immune targets non-invasively and system-wide during therapy. A successful PET imaging tracer should meet specific criteria concerning target affinity, specificity, clearance rate and target-specific uptake, to name a few. The structural profile of such a tracer will define its properties and can be used to optimize tracers in development and design new ones. Currently, a range of PD-1/PD-L1-targeting PET tracers are available from different molecular categories that have shown impressive preclinical and clinical results, each with its own advantages and disadvantages. This review will provide an overview of current PET tracers targeting the PD-1/PD-L1 axis. Antibody, peptide, and antibody fragment tracers will be discussed with respect to their molecular characteristics and binding properties and ways to optimize them.
Collapse
Affiliation(s)
- Melinda Badenhorst
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| | - Albert D. Windhorst
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| | - Wissam Beaino
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| |
Collapse
|
19
|
Rizk SS, Moustafa DM, ElBanna SA, Nour El-Din HT, Attia AS. Nanobodies in the fight against infectious diseases: repurposing nature's tiny weapons. World J Microbiol Biotechnol 2024; 40:209. [PMID: 38771414 PMCID: PMC11108896 DOI: 10.1007/s11274-024-03990-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/15/2024] [Indexed: 05/22/2024]
Abstract
Nanobodies are the smallest known antigen-binding molecules to date. Their small size, good tissue penetration, high stability and solubility, ease of expression, refolding ability, and negligible immunogenicity in the human body have granted them excellence over conventional antibodies. Those exceptional attributes of nanobodies make them promising candidates for various applications in biotechnology, medicine, protein engineering, structural biology, food, and agriculture. This review presents an overview of their structure, development methods, advantages, possible challenges, and applications with special emphasis on infectious diseases-related ones. A showcase of how nanobodies can be harnessed for applications including neutralization of viruses and combating antibiotic-resistant bacteria is detailed. Overall, the impact of nanobodies in vaccine design, rapid diagnostics, and targeted therapies, besides exploring their role in deciphering microbial structures and virulence mechanisms are highlighted. Indeed, nanobodies are reshaping the future of infectious disease prevention and treatment.
Collapse
Affiliation(s)
- Soha S Rizk
- Microbiology and Immunology Postgraduate Program, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Dina M Moustafa
- Department of Medical Sciences, Faculty of Dentistry, The British University in Egypt, El Sherouk City, Cairo, 11837, Egypt
| | - Shahira A ElBanna
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Hanzada T Nour El-Din
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ahmed S Attia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
20
|
Huang W, Zhang Y, Cao M, Wu Y, Jiao F, Chu Z, Zhou X, Li L, Xu D, Pan X, Guan Y, Huang G, Liu J, Xie F, Wei W. ImmunoPET imaging of Trop2 in patients with solid tumours. EMBO Mol Med 2024; 16:1143-1161. [PMID: 38565806 PMCID: PMC11099157 DOI: 10.1038/s44321-024-00059-5] [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: 12/31/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024] Open
Abstract
Accurately predicting and selecting patients who can benefit from targeted or immunotherapy is crucial for precision therapy. Trophoblast cell surface antigen 2 (Trop2) has been extensively investigated as a pan-cancer biomarker expressed in various tumours and plays a crucial role in tumorigenesis through multiple signalling pathways. Our laboratory successfully developed two 68Ga-labelled nanobody tracers that can rapidly and specifically target Trop2. Of the two tracers, [68Ga]Ga-NOTA-T4, demonstrated excellent pharmacokinetics in preclinical mouse models and a beagle dog. Moreover, [68Ga]Ga-NOTA-T4 immuno-positron emission tomography (immunoPET) allowed noninvasive visualisation of Trop2 heterogeneous and differential expression in preclinical solid tumour models and ten patients with solid tumours. [68Ga]Ga-NOTA-T4 immunoPET could facilitate clinical decision-making through patient stratification and response monitoring during Trop2-targeted therapies.
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
| | - 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
| | - Min Cao
- Department of Thoracic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200217, China
| | - Yanfei Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Feng Jiao
- Department of Oncology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhaohui Chu
- Department of Oncology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xinyuan Zhou
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Lianghua 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
| | - Dongsheng Xu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Xinbing Pan
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, 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
| | - 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.
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, 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.
| |
Collapse
|
21
|
Li Q, Kong Y, Zhong Y, Huang A, Ying T, Wu Y. Half-life extension of single-domain antibody-drug conjugates by albumin binding moiety enhances antitumor efficacy. MedComm (Beijing) 2024; 5:e557. [PMID: 38737471 PMCID: PMC11082534 DOI: 10.1002/mco2.557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 05/14/2024] Open
Abstract
Single-domain antibody-drug conjugates (sdADCs) have been proven to have deeper solid tumor penetration and intratumor accumulation capabilities due to their smaller size compared with traditional IgG format ADCs. However, one of the key challenges for improving clinical outcomes of sdADCs is their abbreviated in vivo half-life. In this study, we innovatively fused an antihuman serum albumin (αHSA) nanobody to a sdADCs targeting oncofetal antigen 5T4, conferring serum albumin binding to enhance the pharmacokinetic profiles of sdADCs. The fusion protein was conjugated with monomethyl auristatin E (MMAE) at s224c site mutation. The conjugate exhibited potent cytotoxicity against various tumor cells. Compared with the nonalbumin-binding counterparts, the conjugate exhibited a 10-fold extended half-life in wild-type mice and fivefold prolonged serum half-life in BxPC-3 xenograft tumor models as well as enhanced tumor accumulation and retention in mice. Consequently, n501-αHSA-MMAE showed potent antitumor effects, which were comparable to n501-MMAE in pancreatic cancer BxPC-3 xenograft tumor models; however, in human ovarian teratoma PA-1 xenograft tumor models, n501-αHSA-MMAE significantly improved antitumor efficacy. Moreover, the conjugate showed mitigated hepatotoxicity. In summary, our results suggested that fusion to albumin-binding moiety as a viable strategy can enhance the therapeutic potential of sdADCs through optimized pharmacokinetics.
Collapse
Affiliation(s)
- Quanxiao Li
- MOE/NHC/CAMS Key Laboratory of Medical Molecular VirologyShanghai Institute of Infectious Disease and BiosecurityShanghai Frontiers Science Center of Pathogenic Microorganisms and InfectionShanghai Engineering Research Center for Synthetic ImmunologyDepartment of medical microbiology and parasitology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Yu Kong
- MOE/NHC/CAMS Key Laboratory of Medical Molecular VirologyShanghai Institute of Infectious Disease and BiosecurityShanghai Frontiers Science Center of Pathogenic Microorganisms and InfectionShanghai Engineering Research Center for Synthetic ImmunologyDepartment of medical microbiology and parasitology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Yuxuan Zhong
- MOE/NHC/CAMS Key Laboratory of Medical Molecular VirologyShanghai Institute of Infectious Disease and BiosecurityShanghai Frontiers Science Center of Pathogenic Microorganisms and InfectionShanghai Engineering Research Center for Synthetic ImmunologyDepartment of medical microbiology and parasitology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Ailing Huang
- College of Life SciencesHebei Agricultural UniversityBaodingChina
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular VirologyShanghai Institute of Infectious Disease and BiosecurityShanghai Frontiers Science Center of Pathogenic Microorganisms and InfectionShanghai Engineering Research Center for Synthetic ImmunologyDepartment of medical microbiology and parasitology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Yanling Wu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular VirologyShanghai Institute of Infectious Disease and BiosecurityShanghai Frontiers Science Center of Pathogenic Microorganisms and InfectionShanghai Engineering Research Center for Synthetic ImmunologyDepartment of medical microbiology and parasitology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| |
Collapse
|
22
|
Perez KA, Deppe DW, Filas A, Singh SA, Aikawa E. Multimodal Analytical Tools to Enhance Mechanistic Understanding of Aortic Valve Calcification. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:539-550. [PMID: 37517686 PMCID: PMC10988764 DOI: 10.1016/j.ajpath.2023.06.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/14/2023] [Accepted: 06/29/2023] [Indexed: 08/01/2023]
Abstract
This review focuses on technologies at the core of calcific aortic valve disease (CAVD) and drug target research advancement, including transcriptomics, proteomics, and molecular imaging. We examine how bulk RNA sequencing and single-cell RNA sequencing have engendered organismal genomes and transcriptomes, promoting the analysis of tissue gene expression profiles and cell subpopulations, respectively. We bring into focus how the field is also largely influenced by increasingly accessible proteome profiling techniques. In unison, global transcriptional and protein expression analyses allow for increased understanding of cellular behavior and pathogenic pathways under pathologic stimuli including stress, inflammation, low-density lipoprotein accumulation, increased calcium and phosphate levels, and vascular injury. We also look at how direct investigation of protein signatures paves the way for identification of targetable pathways for pharmacologic intervention. Here, we note that imaging techniques, once a clinical diagnostic tool for late-stage CAVD, have since been refined to address a clinical need to identify microcalcifications using positron emission tomography/computed tomography and even detect in vivo cellular events indicative of early stage CAVD and map the expression of identified proteins in animal models. Together, these techniques generate a holistic approach to CAVD investigation, with the potential to identify additional novel regulatory pathways.
Collapse
Affiliation(s)
- Katelyn A Perez
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel W Deppe
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aidan Filas
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
23
|
Zhang Q, Zhang N, Xiao H, Wang C, He L. Small Antibodies with Big Applications: Nanobody-Based Cancer Diagnostics and Therapeutics. Cancers (Basel) 2023; 15:5639. [PMID: 38067344 PMCID: PMC10705070 DOI: 10.3390/cancers15235639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/16/2023] [Accepted: 11/27/2023] [Indexed: 02/13/2025] Open
Abstract
Monoclonal antibodies (mAbs) have exhibited substantial potential as targeted therapeutics in cancer treatment due to their precise antigen-binding specificity. Despite their success in tumor-targeted therapies, their effectiveness is hindered by their large size and limited tissue permeability. Camelid-derived single-domain antibodies, also known as nanobodies, represent the smallest naturally occurring antibody fragments. Nanobodies offer distinct advantages over traditional mAbs, including their smaller size, high stability, lower manufacturing costs, and deeper tissue penetration capabilities. They have demonstrated significant roles as both diagnostic and therapeutic tools in cancer research and are also considered as the next generation of antibody drugs. In this review, our objective is to provide readers with insights into the development and various applications of nanobodies in the field of cancer treatment, along with an exploration of the challenges and strategies for their prospective clinical trials.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Q.Z.); (C.W.)
| | - Nan Zhang
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China;
| | - Han Xiao
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730030, China;
| | - Chen Wang
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Q.Z.); (C.W.)
| | - Lian He
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Q.Z.); (C.W.)
| |
Collapse
|
24
|
Affiliation(s)
- Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
25
|
Jumapili NA, Zivalj M, Barthelmess RM, Raes G, De Groof TWM, Devoogdt N, Stijlemans B, Vincke C, Van Ginderachter JA. A few good reasons to use nanobodies for cancer treatment. Eur J Immunol 2023; 53:e2250024. [PMID: 37366246 DOI: 10.1002/eji.202250024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/29/2023] [Accepted: 05/17/2023] [Indexed: 06/28/2023]
Abstract
mAbs have been instrumental for targeted cancer therapies. However, their relatively large size and physicochemical properties result in a heterogenous distribution in the tumor microenvironment, usually restricted to the first cell layers surrounding blood vessels, and a limited ability to penetrate the brain. Nanobodies are tenfold smaller, resulting in a deeper tumor penetration and the ability to reach cells in poorly perfused tumor areas. Nanobodies are rapidly cleared from the circulation, which generates a fast target-to-background contrast that is ideally suited for molecular imaging purposes but may be less optimal for therapy. To circumvent this problem, nanobodies have been formatted to noncovalently bind albumin, increasing their serum half-life without majorly increasing their size. Finally, nanobodies have shown superior qualities to infiltrate brain tumors as compared to mAbs. In this review, we discuss why these features make nanobodies prime candidates for targeted therapy of cancer.
Collapse
Affiliation(s)
- Neema Ahishakiye Jumapili
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Maida Zivalj
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Romina Mora Barthelmess
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Geert Raes
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Timo W M De Groof
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Benoît Stijlemans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Cécile Vincke
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Jo A Van Ginderachter
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| |
Collapse
|
26
|
Zhu M, Zhang J, Yang M, Zhang H, Xu T, Kan F, Zhang X, Zhang S, Yin Y, Yu F. In vitro and in vivo study on the treatment of non-small cell lung cancer with radionuclide labeled PD-L1 nanobody. J Cancer Res Clin Oncol 2023; 149:8429-8442. [PMID: 37085729 DOI: 10.1007/s00432-023-04793-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/15/2023] [Indexed: 04/23/2023]
Abstract
PURPOSE Nanobodies have become promising carriers due to excellent in vivo properties. Radiopharmaceutical therapy targeting programmed cell death ligand 1 (PD-L1) is an effective therapeutic strategy. Our study aimed to explore therapeutic efficacy of 131I labeled PD-L1 nanobody (Nb109) in non-small cell lung cancers (NSCLCs) in vitro and in vivo. METHODS 131I-Nb109 was synthesized by chloramine-T method. We implemented stability analysis, SDS-PAGE and lipid-water partition coefficient test to assess its quality. Cell uptake assay and SPECT/CT scan were applied to evaluate its ability to target NSCLCs (H460 and A549). CCK8 assay and in vivo efficacy assay were conducted to estimate its therapeutic effect in H460 tumors. Damage-associated molecular patterns (DAMPs) release in H460 cells incubated with 131I-Nb109 was investigated by western blot and ATP test kit. RESULTS 131I-Nb109 was hydrophilic with high labeling rate (69.51-98.06%), radiochemical purity (99.17% ± 0.76%) and stability. Cell uptake experiments showed that H460 cells (PD-L1 positive) compared with A549 cells (PD-L1 negative) had higher 131I-Nb109 uptake. SPECT/CT imaging revealed the accumulation of 131I-Nb109 in H460 tumor within 48 h. 131I-Nb109 inhibited H460 tumor growth without toxic side effects in contrast with control group. It also induced H460 cells to release DAMPs (adenosine triphosphate, high mobility group box 1, and heat shock protein 70). CONCLUSION 131I-Nb109 had high stability, excellent ability to target and treatment PD-L1 positive tumors, and can improve tumor immunogenicity. The results of our study were expected to inspire the development of more novel radiopharmaceuticals to treat NSCLCs.
Collapse
Affiliation(s)
- Mengqin Zhu
- Shanghai Clinical College, Anhui Medical University, Shanghai, 200040, China
- The Fifth Clinical Medical College, Anhui Medical University, Hefei, 230032, China
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200040, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200040, China
| | - Jiajia Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200040, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200040, China
| | - Mengdie Yang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200040, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200040, China
| | - Han Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200040, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200040, China
| | - Tao Xu
- Smart-Nuclide Biopharma Co. Ltd, No. 218 Xing-Hu Rd., Suzhou, 215125, China
| | - Fei Kan
- Smart-Nuclide Biopharma Co. Ltd, No. 218 Xing-Hu Rd., Suzhou, 215125, China
| | - Xiaoyi Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200040, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200040, China
| | - Shenghong Zhang
- Shanghai Clinical College, Anhui Medical University, Shanghai, 200040, China
- The Fifth Clinical Medical College, Anhui Medical University, Hefei, 230032, China
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200040, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200040, China
| | - Yuzhen Yin
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200040, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200040, China
| | - Fei Yu
- Shanghai Clinical College, Anhui Medical University, Shanghai, 200040, China.
- The Fifth Clinical Medical College, Anhui Medical University, Hefei, 230032, China.
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200040, China.
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200040, China.
| |
Collapse
|
27
|
Song W, Wei W, Lan X, Cai W. Albumin binding improves nanobody pharmacokinetics for dual-modality PET/NIRF imaging of CEACAM5 in colorectal cancer models. Eur J Nucl Med Mol Imaging 2023; 50:2591-2594. [PMID: 37191678 PMCID: PMC10330897 DOI: 10.1007/s00259-023-06266-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Affiliation(s)
- Wenyu Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Madison, WI, USA
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China.
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Madison, WI, USA.
| |
Collapse
|
28
|
Santos A, Macedo de Souza Brandão AP, Hryniewicz BM, Abreu H, Bach-Toledo L, Schuster da Silva S, Deller AE, Rogerio VZ, Baêta Rodrigues DS, Hiraiwa PM, Guimarães BG, Marchesi LF, Carvalho de Oliveira J, Gradia DF, Soares FLF, Zanchin NIT, Camargo de Oliveira C, Vidotti M. COVID-19 impedimetric biosensor based on polypyrrole nanotubes, nickel hydroxide and VHH antibody fragment: specific, sensitive, and rapid viral detection in saliva samples. MATERIALS TODAY. CHEMISTRY 2023; 30:101597. [PMID: 37284350 PMCID: PMC10236006 DOI: 10.1016/j.mtchem.2023.101597] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/30/2023] [Accepted: 05/09/2023] [Indexed: 06/08/2023]
Abstract
SARS-CoV-2 rapid spread required urgent, accurate, and prompt diagnosis to control the virus dissemination and pandemic management. Several sensors were developed using different biorecognition elements to obtain high specificity and sensitivity. However, the task to achieve these parameters in combination with fast detection, simplicity, and portability to identify the biorecognition element even in low concentration remains a challenge. Therefore, we developed an electrochemical biosensor based on polypyrrole nanotubes coupled via Ni(OH)2 ligation to an engineered antigen-binding fragment of heavy chain-only antibodies (VHH) termed Sb#15. Herein we report Sb#15-His6 expression, purification, and characterization of its interaction with the receptor-binding domain (RBD) of SARS-CoV-2 in addition to the construction and validation of a biosensor. The recombinant Sb#15 is correctly folded and interacts with the RBD with a dissociation constant (KD) of 27.1 ± 6.4 nmol/L. The biosensing platform was developed using polypyrrole nanotubes and Ni(OH)2, which can properly orientate the immobilization of Sb#15-His6 at the electrode surface through His-tag interaction for the sensitive SARS-CoV-2 antigen detection. The quantification limit was determined as 0.01 pg/mL using recombinant RBD, which was expressively lower than commercial monoclonal antibodies. In pre-characterized saliva, both Omicron and Delta SARS-CoV-2 were accurately detected only in positive samples, meeting all the requirements recommended by the World Health Organization for in vitro diagnostics. A low sample volume of saliva is needed to perform the detection, providing results within 15 min without further sample preparations. In summary, a new perspective allying recombinant VHHs with biosensor development and real sample detection was explored, addressing the need for accurate, rapid, and sensitive biosensors.
Collapse
Affiliation(s)
- A Santos
- Laboratory of Inflammatory and Neoplastic Cells, Cell Biology Department, Section of Biological Sciences - Universidade Federal do Paraná (UFPR), 81531-980, Curitiba, PR, Brazil
| | - A P Macedo de Souza Brandão
- Laboratory of Structural Biology and Protein Engineering, Carlos Chagas Institute, FIOCRUZ Paraná, 81350-010, Curitiba, PR, Brazil
| | - B M Hryniewicz
- Grupo de Pesquisa em Macromoléculas e Interfaces, Universidade Federal do Paraná (UFPR), 81531-980, Curitiba, PR, Brazil
| | - H Abreu
- Laboratory of Human Cytogenetics and Oncogenetics, Postgraduate Program in Genetics, Department of Genetics, Universidade Federal do Paraná (UFPR), 81531-980, Curitiba, Brazil
| | - L Bach-Toledo
- Grupo de Pesquisa em Macromoléculas e Interfaces, Universidade Federal do Paraná (UFPR), 81531-980, Curitiba, PR, Brazil
- Centro de Tecnologia da Informação Renato Archer (CTI), Rod. D. Pedro I, KM143.6, 13069-901, Campinas, SP, Brazil
| | - S Schuster da Silva
- Laboratory of Inflammatory and Neoplastic Cells, Cell Biology Department, Section of Biological Sciences - Universidade Federal do Paraná (UFPR), 81531-980, Curitiba, PR, Brazil
| | - A E Deller
- Grupo de Pesquisa em Macromoléculas e Interfaces, Universidade Federal do Paraná (UFPR), 81531-980, Curitiba, PR, Brazil
| | - V Z Rogerio
- Laboratory of Structural Biology and Protein Engineering, Carlos Chagas Institute, FIOCRUZ Paraná, 81350-010, Curitiba, PR, Brazil
| | - D S Baêta Rodrigues
- Laboratory of Structural Biology and Protein Engineering, Carlos Chagas Institute, FIOCRUZ Paraná, 81350-010, Curitiba, PR, Brazil
| | - P M Hiraiwa
- Laboratory of Structural Biology and Protein Engineering, Carlos Chagas Institute, FIOCRUZ Paraná, 81350-010, Curitiba, PR, Brazil
| | - B G Guimarães
- Laboratory of Structural Biology and Protein Engineering, Carlos Chagas Institute, FIOCRUZ Paraná, 81350-010, Curitiba, PR, Brazil
| | - L F Marchesi
- Grupo de Estudos em Espectroscopia de Impedância Eletroquímica (GEIS), Universidade Tecnológica Federal Do Paraná, Rua Dr. Washington Subtil Chueire, 330 - Jd. Carvalho, CEP 84017-220, Ponta Grossa, PR, Brazil
| | - J Carvalho de Oliveira
- Laboratory of Human Cytogenetics and Oncogenetics, Postgraduate Program in Genetics, Department of Genetics, Universidade Federal do Paraná (UFPR), 81531-980, Curitiba, Brazil
| | - D F Gradia
- Laboratory of Human Cytogenetics and Oncogenetics, Postgraduate Program in Genetics, Department of Genetics, Universidade Federal do Paraná (UFPR), 81531-980, Curitiba, Brazil
| | - F L F Soares
- Grupo de Pesquisa em Macromoléculas e Interfaces, Universidade Federal do Paraná (UFPR), 81531-980, Curitiba, PR, Brazil
| | - N I T Zanchin
- Laboratory of Structural Biology and Protein Engineering, Carlos Chagas Institute, FIOCRUZ Paraná, 81350-010, Curitiba, PR, Brazil
| | - C Camargo de Oliveira
- Laboratory of Inflammatory and Neoplastic Cells, Cell Biology Department, Section of Biological Sciences - Universidade Federal do Paraná (UFPR), 81531-980, Curitiba, PR, Brazil
| | - M Vidotti
- Grupo de Pesquisa em Macromoléculas e Interfaces, Universidade Federal do Paraná (UFPR), 81531-980, Curitiba, PR, Brazil
| |
Collapse
|
29
|
Wen G, Lei H, Qi B, Duan S, Xiao Z, Han C, Xia Y, Jing C, Liu J, Li C. Noninvasive Imaging OX40 + Activated T Cells Provides Early Warning of Rheumatoid Arthritis. Mol Imaging Biol 2023:10.1007/s11307-023-01819-4. [PMID: 37012517 DOI: 10.1007/s11307-023-01819-4] [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: 12/02/2022] [Revised: 03/09/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023]
Abstract
PURPOSE The goal of this study was to develop an imaging probe-IRDye-680RD-OX40 mAb-that can be used for noninvasive imaging and optical imaging of rheumatoid arthritis (RA). OX40/OX40 ligand (OX40L) interactions have been shown to exert potent costimulatory effects on T cell activation. Detectable change in T cell activation profiles was observed in early RA. METHODS OX40 expression pattern was analyzed by flow cytometry. N-hydroxysuccinimide (NHS) esters are used to label proteins selectively on free amino groups of OX40 monoclonal antibody (mAb). Characterization of IRDye-680RD-OX40 mAb was measured and a fluorescence spectrum gathered. Cell binding assay was also performed between activated and naïve murine T cells. Longitudinal near-infrared fluorescence (NIRF) imaging of the probe was performed on day 8, day 9, day 10, and day 11 of adjuvant-induced arthritis (AIA) mouse model. Paw thickness and body weight were compared between the OX40 mAb and IgG injection groups. RESULTS NIRF imaging with IRDye-680RD-OX40 mAb revealed strong OX40-positive responses with high specificity. Flow analysis showed that OX40 was specifically expressed on the surface of T cells in RP and spleen of AIA model. The AIA group was significantly differentiated from the control group at all time points with imaging monitoring. The region of interest (ROI) was in line with ex vivo imaging and biodistribution study. This study highlights the potential utility of the OX40 NIRF imaging as a new strategy for RA prediction and T cell monitoring. CONCLUSION The results provide evidence that IRDye-680RD-OX40 mAb detects organized T cells activation in early RA. The optical probe was capable of detection of RA pathogenesis. It identified transcriptional responses to RA that mediate its immune functions. Thus, it may be an ideal probe for RA imaging.
Collapse
Affiliation(s)
- Gang Wen
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Hongwei Lei
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Baochang Qi
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Shao Duan
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Zunyu Xiao
- Molecular Imaging Research Center of Harbin Medical University, Harbin, 150001, China
| | - Chaozhe Han
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Yifei Xia
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Chengwei Jing
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jianyu Liu
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| | - Chao Li
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
30
|
Zhang Y, Zhang D, An S, Liu Q, Liang C, Li J, Liu P, Wu C, Huang G, Wei W, Liu J. Development and Characterization of Nanobody-Derived CD47 Theranostic Pairs in Solid Tumors. RESEARCH (WASHINGTON, D.C.) 2023; 6:0077. [PMID: 36939440 PMCID: PMC10017100 DOI: 10.34133/research.0077] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/23/2023] [Indexed: 02/04/2023]
Abstract
Overexpression of CD47 is frequently observed in various types of human malignancies, inhibiting myeloid-mediated elimination of tumor cells and affecting the prognosis of cancer patients. By mapping biomarker expression, immuno-positron emission tomography has been increasingly used for patient screening and response monitoring. By immunization alpacas with recombinant human CD47, we prepared a CD47-targeting nanobody C2 and developed [68Ga]Ga-NOTA-C2, followed by an exploration of the diagnostic value in CD47-expressing tumor models including gastric-cancer patient-derived xenograft models. By fusing C2 to an albumin binding domain (ABD), we synthesized ABDC2, which had increased in vivo half-life and improved targeting properties. We further labeled ABDC2 with 68Ga/89Zr/177Lu to develop radionuclide theranostic pairs and evaluated the pharmacokinetics and theranostic efficacies of the agents in cell- and patient-derived models. Both C2 and ABDC2 specifically reacted with human CD47 with a high K D value of 23.50 and 84.57 pM, respectively. [68Ga]Ga-NOTA-C2 was developed with high radiochemical purity (99 >%, n = 4) and visualized CD47 expression in the tumors. In comparison to the rapid renal clearance and short half-life of [68Ga]Ga-NOTA-C2, both [68Ga]Ga-NOTA-ABDC2 and [89Zr]Zr-DFO-ABDC2 showed prolonged circulation and increased tumor uptake, with the highest uptake of [89Zr]Zr-DFO-ABDC2 occurring at 72 h post-injection. Moreover, [177Lu]Lu-DOTA-ABDC2 radioimmunotherapy suppressed the tumor growth but was associated with toxicity, warranting further optimization of the treatment schedules. Taken together, we reported a series of nanobody-derived CD47-targeted agents, of which [68Ga]Ga-NOTA-C2 and [89Zr]Zr-DFO-ABDC2 are readily translatable. Optimization and translation of CD47-targeted theranostic pair may provide new prospects for CD47-targeted management of solid tumors.
Collapse
Affiliation(s)
- You Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
| | - Di Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shuxian An
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qiufang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center,
Fudan University, Shanghai 200030, China
| | - Chenyi Liang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
| | - Juan Li
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences,
The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Ping Liu
- School of Biomedical Engineering and Med-X Research Institute,
Shanghai Jiao Tong University, Shanghai 200030, China
| | - Changfeng Wu
- Department of Biomedical Engineering,
Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
- Address correspondence to: (W.W.); (G.H.); (J.L.)
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
- Address correspondence to: (W.W.); (G.H.); (J.L.)
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
- Address correspondence to: (W.W.); (G.H.); (J.L.)
| |
Collapse
|
31
|
Ma J, Xu X, Fu C, Xia P, Tian M, Zheng L, Chen K, Liu X, Li Y, Yu L, Zhu Q, Yu Y, Fan R, Jiang H, Li Z, Yang C, Xu C, Long Y, Wang J, Li Z. CDH17 nanobodies facilitate rapid imaging of gastric cancer and efficient delivery of immunotoxin. Biomater Res 2022; 26:64. [PMID: 36435809 PMCID: PMC9701387 DOI: 10.1186/s40824-022-00312-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/27/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND It is highly desirable to develop new therapeutic strategies for gastric cancer given the low survival rate despite improvement in the past decades. Cadherin 17 (CDH17) is a membrane protein highly expressed in cancers of digestive system. Nanobody represents a novel antibody format for cancer targeted imaging and drug delivery. Nanobody targeting CHD17 as an imaging probe and a delivery vehicle of toxin remains to be explored for its theragnostic potential in gastric cancer. METHODS Naïve nanobody phage library was screened against CDH17 Domain 1-3 and identified nanobodies were extensively characterized with various assays. Nanobodies labeled with imaging probe were tested in vitro and in vivo for gastric cancer detection. A CDH17 Nanobody fused with toxin PE38 was evaluated for gastric cancer inhibition in vitro and in vivo. RESULTS Two nanobodies (A1 and E8) against human CDH17 with high affinity and high specificity were successfully obtained. These nanobodies could specifically bind to CDH17 protein and CDH17-positive gastric cancer cells. E8 nanobody as a lead was extensively determined for tumor imaging and drug delivery. It could efficiently co-localize with CDH17-positive gastric cancer cells in zebrafish embryos and rapidly visualize the tumor mass in mice within 3 h when conjugated with imaging dyes. E8 nanobody fused with toxin PE38 showed excellent anti-tumor effect and remarkably improved the mice survival in cell-derived (CDX) and patient-derived xenograft (PDX) models. The immunotoxin also enhanced the anti-tumor effect of clinical drug 5-Fluorouracil. CONCLUSIONS The study presents a novel imaging and drug delivery strategy by targeting CDH17. CDH17 nanobody-based immunotoxin is potentially a promising therapeutic modality for clinical translation against gastric cancer.
Collapse
Affiliation(s)
- Jingbo Ma
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China.,College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, P. R. China
| | - Xiaolong Xu
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China
| | - Chunjin Fu
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China
| | - Peng Xia
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China.,Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430072, Hubei, P. R. China
| | - Ming Tian
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China.,Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430072, Hubei, P. R. China
| | - Liuhai Zheng
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China
| | - Kun Chen
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China
| | - Xiaolian Liu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, P. R. China
| | - Yilei Li
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, P. R. China
| | - Le Yu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, P. R. China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, P.R. China
| | - Qinchang Zhu
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, P.R. China
| | - Yangyang Yu
- Health Science Center, Shenzhen University, Shenzhen, 518055, Guangdong, P. R. China
| | - Rongrong Fan
- Deapartment of Biosciences and Nutrition, Karolinska Institute, 14157, Stockholm, Sweden
| | - Haibo Jiang
- Department of Chemistry, The University of Hong Kong, Pok Fu Lam, Hong Kong, P. R. China
| | - Zhifen Li
- School of Chemistry and Chemical Engineering, Shanxi Datong University, Xing Yun Street, Pingcheng District, Datong, 037009, Shanxi, P. R. China
| | - Chuanbin Yang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China
| | - Chengchao Xu
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China
| | - Ying Long
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China.
| | - Jigang Wang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China. .,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, P.R. China. .,Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China.
| | - Zhijie Li
- Department of Hyperbaric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China. .,Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, P. R. China.
| |
Collapse
|
32
|
Wang Q, Zhang X, Wei W, Cao M. PET Imaging of Lung Cancers in Precision Medicine: Current Landscape and Future Perspective. Mol Pharm 2022; 19:3471-3483. [PMID: 35771950 DOI: 10.1021/acs.molpharmaceut.2c00353] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Despite the recent advances in cancer treatment, lung cancer remains the leading cause of cancer mortality worldwide. Immunotherapies using immune checkpoint inhibitors (ICIs) achieved substantial efficacy in nonsmall cell lung cancer (NSCLC). Currently, most ICIs are still a monoclonal antibody (mAb). Using mAbs or antibody derivatives labeled with radionuclide as the tracers, immunopositron emission tomography (immunoPET) possesses multiple advantages over traditional 18F-FDG PET in imaging lung cancers. ImmunoPET presents excellent potential in detecting, diagnosing, staging, risk stratification, treatment guidance, and recurrence monitoring of lung cancers. By using radiolabeled mAbs, immunoPET can visualize the biodistribution and uptake of ICIs, providing a noninvasive modality for patient stratification and response evaluation. Some novel targets and associated tracers for immunoPET have been discovered and investigated. This Review introduces the value of immunoPET in imaging lung cancers by summarizing both preclinical and clinical evidence. We also emphasize the value of immunoPET in optimizing immunotherapy in NSCLC. Lastly, immunoPET probes developed for imaging small cell lung cancer (SCLC) will also be discussed. Although the major focus is to summarize the immunoPET tracers for lung cancers, we also highlighted several small-molecule PET tracers to give readers a balanced view of the development status.
Collapse
Affiliation(s)
- Qing Wang
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200217, China
| | - Xindi Zhang
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 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
| | - Min Cao
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200217, China
| |
Collapse
|
33
|
Ayala I, Colanzi A. Structural Organization and Function of the Golgi Ribbon During Cell Division. Front Cell Dev Biol 2022; 10:925228. [PMID: 35813197 PMCID: PMC9263219 DOI: 10.3389/fcell.2022.925228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/06/2022] [Indexed: 11/24/2022] Open
Abstract
The Golgi complex has a central role in the secretory traffic. In vertebrate cells it is generally organized in polarized stacks of cisternae that are laterally connected by membranous tubules, forming a structure known as Golgi ribbon. The steady state ribbon arrangement results from a dynamic equilibrium between formation and cleavage of the membrane tubules connecting the stacks. This balance is of great physiological relevance as the unlinking of the ribbon during G2 is required for mitotic entry. A block of this process induces a potent G2 arrest of the cell cycle, indicating that a mitotic “Golgi checkpoint” controls the correct pre-mitotic segregation of the Golgi ribbon. Then, after mitosis onset, the Golgi stacks undergo an extensive disassembly, which is necessary for proper spindle formation. Notably, several Golgi-associated proteins acquire new roles in spindle formation and mitotic progression during mitosis. Here we summarize the current knowledge about the basic principle of the Golgi architecture and its functional relationship with cell division to highlight crucial aspects that need to be addressed to help us understand the physiological significance of the ribbon and the pathological implications of alterations of this organization.
Collapse
|