1
|
Zhang S, Sun X, Liu W, Wu J, Wu Y, Jiang S, Wang X, Gao X, Zuo Q, Zhang H, Zhang Y, Wang F, Wang R, Hu K. Determining the Multivalent Effects of d-Peptide-Based Radiotracers. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:180-190. [PMID: 40151821 PMCID: PMC11938029 DOI: 10.1021/cbmi.4c00071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/09/2024] [Accepted: 12/31/2024] [Indexed: 03/29/2025]
Abstract
Dextrorotary (d) peptides, composed of d-amino acids, are hyper-resistant to proteolytic hydrolysis, making them valuable ligands with excellent in vivo stability for radiopharmaceutical development. Multimerization is a well-established strategy for enhancing the in vivo performance of l-peptide-based radiopharmaceuticals. However, the effect of multimerization on the in vivo fate of d-peptide-based radiopharmaceuticals remains largely unexplored. Here, we synthesized the d-peptide DPA, which targets PD-L1, along with its dimer (DP2) and trimer (DP3). PET/CT imaging and ex vivo biodistribution studies were performed to delineate the pharmacokinetics and target interactions of [68Ga]DPA, [68Ga]DP2, and [68Ga]DP3 in both normal and tumor-bearing mice. Our results revealed that tumor uptake and kidney retention increased with higher valency ([68Ga]DP3 > [68Ga]DP2 > [68Ga]DPA). No significant differences were observed in the liver, heart, lung, spleen, intestine, or bone among the three radiotracers. Interestingly, a significant reduction of radioactivity in the bloodstream was detected for the [68Ga]DP3-treated group compared to the other two groups. Data analysis revealed that chiral configuration of amino acids and the linking chemistry used in multimerization are the two dominant factors in the in vivo fate of d-peptide multimers. These findings indicate that d-peptide multimerization exerts a distinct influence on in vivo profiles compared to l-peptide multimerization. This study deepens our understanding of how mirror-imaged peptides/proteins interact with the living systems, paving the way for the development of radiopharmaceuticals that harness d-peptides as targeting moieties.
Collapse
Affiliation(s)
- Siqi Zhang
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines,
Institute of Materia Medica, Chinese Academy
of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaona Sun
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines,
Institute of Materia Medica, Chinese Academy
of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wenhao Liu
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines,
Institute of Materia Medica, Chinese Academy
of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jiang Wu
- Department
of Nuclear Medicine, Nanjing First Hospital,
Nanjing Medical University, Nanjing 210008, China
| | - Yuxuan Wu
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines,
Institute of Materia Medica, Chinese Academy
of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shuo Jiang
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines,
Institute of Materia Medica, Chinese Academy
of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xingkai Wang
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines,
Institute of Materia Medica, Chinese Academy
of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xin Gao
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines,
Institute of Materia Medica, Chinese Academy
of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Quan Zuo
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines,
Institute of Materia Medica, Chinese Academy
of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hailong Zhang
- Key
Laboratory of Preclinical Study for New Drugs of Gansu Province, School
of Basic Medical Sciences & Research Unit of Peptide Science,
Chinese Academy of Medical Sciences, Lanzhou
University, 2019RU066, Lanzhou 730000, China
| | - Yingzi Zhang
- Department
of Orthopaedics, the Second Affiliated Hospital
of Soochow University, Suzhou, 215004 Jiangsu, China
| | - Feng Wang
- Department
of Nuclear Medicine, Nanjing First Hospital,
Nanjing Medical University, Nanjing 210008, China
| | - Rui Wang
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines,
Institute of Materia Medica, Chinese Academy
of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Key
Laboratory of Preclinical Study for New Drugs of Gansu Province, School
of Basic Medical Sciences & Research Unit of Peptide Science,
Chinese Academy of Medical Sciences, Lanzhou
University, 2019RU066, Lanzhou 730000, China
| | - Kuan Hu
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines,
Institute of Materia Medica, Chinese Academy
of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
2
|
Wang X, Dai D, Shen J, Zhang S, Ohkubo T, Xie L, Zhang Y, Li G, Liu C, Tian H, Zhang Y, Zhang MR, Wang R, Hu K. A Cyclic Peptide-Based Radiotheranostic Agent for Urokinase-Type Plasminogen Activator in Tumors. Chemistry 2025:e202500479. [PMID: 40100321 DOI: 10.1002/chem.202500479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 03/20/2025]
Abstract
The plasminogen activator system is critically involved in tumor progression regulation. Aberrant activation of urokinase-type plasminogen activator (uPA) induces proteolytic degradation of cellular membranes and the extracellular matrix, thereby promoting tumor invasion and metastasis. Consequently, uPA has emerged as a promising diagnostic and therapeutic target. Herein, we designed and evaluated three cyclic peptide-based radioligands ([⁶⁴Cu]CAP-1, [⁶⁴Cu]CAP-2, and [⁶⁴Cu]CAP-3) as potential PET tracers for uPA visualization in mouse tumor models, assessing their binding ability, specificity, and pharmacokinetic profiles. Among them, [⁶⁴Cu]CAP-1, featuring a native disulfide bond, emerged as the optimal candidate. This radioligand demonstrated superior tumor uptake and reduced hepatic accumulation compared to the clinically advanced uPAR-targeted tracer [⁶⁴Cu]DOTA-AE105, currently in Phase 2 trials. A single administration of [⁶⁴Cu]CAP-1 (2 mCi/mouse) significantly suppressed tumor growth and prolonged survival in mouse models. These findings position [⁶⁴Cu]CAP-1 as a potent radiotheranostic agent for uPA-overexpressing tumors, offering a novel strategy for precision targeting of the uPA/uPAR axis.
Collapse
Affiliation(s)
- Xingkai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Xian Nong Tan Street, Beijing, China
| | - Dong Dai
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin, China
- Department of Molecular Medicine, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Jieting Shen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Xian Nong Tan Street, Beijing, China
| | - Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Xian Nong Tan Street, Beijing, China
| | - Takayuki Ohkubo
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Lin Xie
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Guoqing Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Xian Nong Tan Street, Beijing, China
| | - Can Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Xian Nong Tan Street, Beijing, China
| | - Hao Tian
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Xian Nong Tan Street, Beijing, China
| | - Yingzi Zhang
- Department of Orthopaedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Xian Nong Tan Street, Beijing, China
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Xian Nong Tan Street, Beijing, China
| |
Collapse
|
3
|
Du S, Liu J, Zhang Y, Ge X, Gao S, Song J. PD-L1 peptides in cancer immunoimaging and immunotherapy. J Control Release 2025; 378:1061-1079. [PMID: 39742920 DOI: 10.1016/j.jconrel.2024.12.069] [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: 11/11/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
The interaction between programmed death protein 1 (PD-1) and programmed death ligand 1 (PD-L1) constitutes a critical immune checkpoint pathway that leads to immune tolerance in cancer cells and impacts antitumor treatment. Monoclonal antibody blockade of the PD-L1 immunoinhibitory pathway has demonstrated significant and lasting clinical antitumor responses. Furthermore, PD-L1 serves as an important biomarker for predicting the effectiveness of immune checkpoint inhibitors (ICIs). To date, numerous studies based on monoclonal antibodies have been carried out to detect the expression levels of PD-L1 and predict the antitumor effectiveness of PD-L1 ICIs. However, due to the deficiencies of monoclonal antibodies, researches of PD-L1 peptides have received increasing attention. PD-L1 peptides present promising candidates due to their advantages, including reduced manufacturing costs, enhanced stability, decreased immunogenicity, faster clearance and improved tumor or organ penetration, thereby offering broad application prospects in cancer immunoimaging and immunotherapy. In this review, we analyze the existing evidence on PD-L1 peptides in cancer immunoimaging and immunotherapy. First, the design techniques of different types of PD-L1 targeting peptides and their strengths and weaknesses are briefly introduced. Second, the recent advancements in immunoimaging and the development trends in immunotherapy are summarized. Finally, the existing challenges and future directions in this field are comprehensively deliberated.
Collapse
Affiliation(s)
- Shiye Du
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Junzhi Liu
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Youjia Zhang
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Xiaoguang Ge
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Shi Gao
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| | - Jibin Song
- College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
4
|
Zhang S, Ma X, Wu J, Shen J, Shi Y, Wang X, Xie L, Sun X, Wu Y, Tian H, Gao X, Chen X, Huang H, Chen L, Song X, Hu Q, Zhang H, Wang F, Jin ZH, Zhang MR, Wang R, Hu K. Enhanced radiotheranostic targeting of integrin α5 β1 with PEGylation-enabled peptide multidisplay platform (PEGibody): A strategy for prolonged tumor retention with fast blood clearance. Acta Pharm Sin B 2025; 15:692-706. [PMID: 40177561 PMCID: PMC11959959 DOI: 10.1016/j.apsb.2024.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/29/2024] [Accepted: 07/01/2024] [Indexed: 04/05/2025] Open
Abstract
Peptide-based radiopharmaceuticals targeting integrin α5β1 show promise for precise tumor diagnosis and treatment. However, current peptide-based radioligands that target α5β1 demonstrate inadequate in vivo performance owing to limited tumor retention. The use of PEGylation to enhance the tumor retention of radiopharmaceuticals by prolonging blood circulation time poses a risk of increased blood toxicity. Therefore, a PEGylation strategy that boosts tumor retention while minimizing blood circulation time is urgently needed. Here, we developed a PEGylation-enabled peptide multidisplay platform (PEGibody) for PR_b, an α5β1 targeting peptide. PEGibody generation involved PEGylation and self-assembly. [64Cu]QM-2303 PEGibodies displayed spherical nanoparticles ranging from 100 to 200 nm in diameter. Compared with non-PEGylated radioligands, [64Cu]QM-2303 demonstrated enhanced tumor retention time due to increased binding affinity and stability. Importantly, the biodistribution analysis confirmed rapid clearance of [64Cu]QM-2303 from the bloodstream. Administration of a single dose of [177Lu]QM-2303 led to robust antitumor efficacy. Furthermore, [64Cu]/[177Lu]QM-2303 exhibited low hematological and organ toxicity in both healthy and tumor-bearing mice. Therefore, this study presents a PEGibody-based radiotheranostic approach that enhances tumor retention time and provides long-lasting antitumor effects without prolonging blood circulation lifetime. The PEGibody-based radiopharmaceutical [64Cu]/[177Lu]QM-2303 shows great potential for positron emission tomography imaging-guided targeted radionuclide therapy for α5β1-overexpressing tumors.
Collapse
Affiliation(s)
- Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaohui Ma
- Department of Vascular and Endovascular Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Jiang Wu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Jieting Shen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yuntao Shi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xingkai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lin Xie
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan
| | - Xiaona Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yuxuan Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hao Tian
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xin Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xueyao Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hongyi Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lu Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xuekai Song
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qichen Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hailong Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Zhao-Hui Jin
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan
| |
Collapse
|
5
|
Peng T, Huang G, Zhao H, Liu J. Development of an Irreversible Peptidomimetic Radioligand for PET Imaging of ST14 Protease. Bioconjug Chem 2025; 36:116-126. [PMID: 39746783 DOI: 10.1021/acs.bioconjchem.4c00564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
To enhance the affinity of peptide ligands for their targets, covalent warheads can be engineered to facilitate irreversible binding. This study aimed at exploring the potential of a 68Ga-labeled peptidomimetic radioligand, [68Ga]Ga-DOTA-RQAR-kbt, for PET imaging through its irreversible binding to the suppression of tumorigenicity 14 (ST14). An Arg-Gln-Ala-Arg (RQAR) tetrapeptide was conjugated with 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid for gallium-68 radiolabeling. The covalent warhead ketobenzothiazole was constructed as a serine trap for ST14 protease, resulting in the formation of DOTA-RQAR-kbt. We compared both the in vitro and in vivo properties of [68Ga]Ga-DOTA-RQAR-kbt with those of its reversible-binding counterparts, [68Ga]Ga-DOTA-RQAR-OH. DOTA-RQAR-kbt exhibits high affinity for ST14 and irreversibly binds to ST14, as evidenced by the lack of ST14 activity recovery following ultrafiltration. In contrast, DOTA-RQAR-OH shows reversible binding and has low affinity for ST14. PET/CT imaging confirmed the superior tumor targeting of [68Ga]Ga-DOTA-RQAR-kbt compared to the [68Ga]Ga-DOTA-RQAR-OH, with robust signals observed at 0.5, 1, and 2 h postinjection. Blocking studies underscored the probe's specificity, as they revealed a marked reduction in tumor uptake in the presence of excess RQAR-kbt. Biodistribution studies demonstrated significantly higher tumor uptake for [68Ga]Ga-DOTA-RQAR-kbt, with 0.89 ± 0.03%ID/g at 1 h postinjection, which was reduced to 0.25 ± 0.03%ID/g (P < 0.01) in the presence of excess RQAR-kbt. In this proof-of-concept study, an irreversibly binding peptidomimetic radioligand targeting ST14 was evaluated, demonstrating improved tumor uptake in vivo compared with its reversibly binding counterparts. This approach holds promise for improving the potency of covalent radiotracers as PET agents.
Collapse
Affiliation(s)
- Tukang Peng
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 210000, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 210000, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 210000, China
| | - Haitao Zhao
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 210000, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 210000, China
| |
Collapse
|
6
|
Zhang S, Wang X, Gao X, Chen X, Li L, Li G, Liu C, Miao Y, Wang R, Hu K. Radiopharmaceuticals and their applications in medicine. Signal Transduct Target Ther 2025; 10:1. [PMID: 39747850 PMCID: PMC11697352 DOI: 10.1038/s41392-024-02041-6] [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: 05/15/2024] [Revised: 08/30/2024] [Accepted: 10/28/2024] [Indexed: 01/04/2025] Open
Abstract
Radiopharmaceuticals involve the local delivery of radionuclides to targeted lesions for the diagnosis and treatment of multiple diseases. Radiopharmaceutical therapy, which directly causes systematic and irreparable damage to targeted cells, has attracted increasing attention in the treatment of refractory diseases that are not sensitive to current therapies. As the Food and Drug Administration (FDA) approvals of [177Lu]Lu-DOTA-TATE, [177Lu]Lu-PSMA-617 and their complementary diagnostic agents, namely, [68Ga]Ga-DOTA-TATE and [68Ga]Ga-PSMA-11, targeted radiopharmaceutical-based theranostics (radiotheranostics) are being increasingly implemented in clinical practice in oncology, which lead to a new era of radiopharmaceuticals. The new generation of radiopharmaceuticals utilizes a targeting vector to achieve the accurate delivery of radionuclides to lesions and avoid off-target deposition, making it possible to improve the efficiency and biosafety of tumour diagnosis and therapy. Numerous studies have focused on developing novel radiopharmaceuticals targeting a broader range of disease targets, demonstrating remarkable in vivo performance. These include high tumor uptake, prolonged retention time, and favorable pharmacokinetic properties that align with clinical standards. While radiotheranostics have been widely applied in tumor diagnosis and therapy, their applications are now expanding to neurodegenerative diseases, cardiovascular diseases, and inflammation. Furthermore, radiotheranostic-empowered precision medicine is revolutionizing the cancer treatment paradigm. Diagnostic radiopharmaceuticals play a pivotal role in patient stratification and treatment planning, leading to improved therapeutic outcomes in targeted radionuclide therapy. This review offers a comprehensive overview of the evolution of radiopharmaceuticals, including both FDA-approved and clinically investigated agents, and explores the mechanisms of cell death induced by radiopharmaceuticals. It emphasizes the significance and future prospects of theranostic-based radiopharmaceuticals in advancing precision medicine.
Collapse
Grants
- 82372002 National Natural Science Foundation of China (National Science Foundation of China)
- 0104002 Beijing Nova Program
- L248087; L234044 Natural Science Foundation of Beijing Municipality (Beijing Natural Science Foundation)
- Nonprofit Central Research Institute Fund of the Chinese Academy of Medical Sciences (No. 2022-RC350-04), the CAMS Innovation Fund for Medical Sciences (Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001), the National Key Research and Development Program of China (No. 2022YFE0111700),the Fundamental Research Funds for the Central Universities (Nos. 3332023044 and 3332023151), the CIRP Open Fund of Radiation Protection Laboratories (No. ZHYLYB2021005), and the China National Nuclear Corporation Young Talent Program.
- Fundamental Research Funds for the Central Universities,Nos. 3332023044
- Fundamental Research Funds for the Central Universities,Nos. 3332023151
- he Nonprofit Central Research Institute Fund of Chinese Academy of Medical Sciences,No. 2022-RC350-04;the CAMS Innovation Fund for Medical Sciences,Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001;the National Key Research and Development Program of China,No. 2022YFE0111700
Collapse
Affiliation(s)
- Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xingkai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xin Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xueyao Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Linger Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Guoqing Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Can Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Yuan Miao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 2019RU066, 730000, Lanzhou, China.
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
| |
Collapse
|
7
|
Gong J, Zhu M, Zhao L, Wang T, Qiao W, Huang Q, Xing Y, Zhao J. 99mTc-Labeled D-Type PTP as a Plectin-Targeting Single-Photon Emission Computed Tomography Probe for Hepatocellular Carcinoma Imaging. Bioconjug Chem 2024; 35:1997-2005. [PMID: 39571181 DOI: 10.1021/acs.bioconjchem.4c00492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Plectin, a scaffolding protein overexpressed in tumor cells, plays a significant role in hepatocellular carcinoma (HCC) proliferation, invasion, and migration. However, the use of L-type peptides for targeting plectin is hindered by their limited stability and retention. We designed a D-type plectin-targeting peptide (DPTP) and developed a novel single-photon emission computed tomography (SPECT) probe for HCC imaging. The DPTP targeting ability was evaluated in vitro using flow cytometry and ex vivo fluorescence imaging. 99mTc radiolabeling was performed using tricine and ethylenediamine-N,N'-diacetic acid (EDDA) as coligands after modification with 6-hydrazino nicotinamide (HYNIC) at the N termini of DPTP. The radiochemical purity (RCP), in vitro stability, and binding affinity of the prepared 99mTc-HYNIC-DPTP were analyzed. Tumor uptake, metabolic stability, biodistribution, and pharmacokinetics of 99mTc-HYNIC-DPTP were investigated and compared with those of 99mTc-labeled L-type PTP (99mTc-HYNIC-PTP) in HCC tumor-bearing mice. DPTP could be efficiently radiolabeled with 99mTc using the HYNIC/tricine/EDDA system with a high RCP and good in vitro stability. Compared with the L-type PTP, DPTP exhibited improved targeting ability, and 99mTc-HYNIC-DPTP displayed higher tumor uptake, better metabolic stability, longer blood circulation time, and lower kidney retention, resulting in superior imaging performance and biodistribution in vivo. 99mTc-HYNIC-DPTP has great potential as a novel SPECT probe for diagnosing HCC.
Collapse
Affiliation(s)
- JiaLi Gong
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Meilin Zhu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, Ningxia,China
| | - Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Taisong Wang
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Wenli Qiao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Qingqing Huang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Yan Xing
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
8
|
Bojarska J, Wolf WM. Short Peptides as Powerful Arsenal for Smart Fighting Cancer. Cancers (Basel) 2024; 16:3254. [PMID: 39409876 PMCID: PMC11476321 DOI: 10.3390/cancers16193254] [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: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Short peptides have been coming around as a strong weapon in the fight against cancer on all fronts-in immuno-, chemo-, and radiotherapy, and also in combinatorial approaches. Moreover, short peptides have relevance in cancer imaging or 3D culture. Thanks to the natural 'smart' nature of short peptides, their unique structural features, as well as recent progress in biotechnological and bioinformatics development, short peptides are playing an enormous role in evolving cutting-edge strategies. Self-assembling short peptides may create excellent structures to stimulate cytotoxic immune responses, which is essential for cancer immunotherapy. Short peptides can help establish versatile strategies with high biosafety and effectiveness. Supramolecular short peptide-based cancer vaccines entered clinical trials. Peptide assemblies can be platforms for the delivery of antigens, adjuvants, immune cells, and/or drugs. Short peptides have been unappreciated, especially in the vaccine aspect. Meanwhile, they still hide the undiscovered unlimited potential. Here, we provide a timely update on this highly active and fast-evolving field.
Collapse
Affiliation(s)
- Joanna Bojarska
- Chemistry Department, Institute of Inorganic and Ecological Chemistry, Łódź University of Technology, S. Żeromskiego Str. 116, 90-924 Łódź, Poland;
| | | |
Collapse
|
9
|
Li H, Gong Q, Luo K. Biomarker-driven molecular imaging probes in radiotherapy. Theranostics 2024; 14:4127-4146. [PMID: 38994026 PMCID: PMC11234278 DOI: 10.7150/thno.97768] [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: 04/25/2024] [Accepted: 06/23/2024] [Indexed: 07/13/2024] Open
Abstract
Background: Biomarker-driven molecular imaging has emerged as an integral part of cancer precision radiotherapy. The use of molecular imaging probes, including nanoprobes, have been explored in radiotherapy imaging to precisely and noninvasively monitor spatiotemporal distribution of biomarkers, potentially revealing tumor-killing mechanisms and therapy-induced adverse effects during radiation treatment. Methods: We summarized literature reports from preclinical studies and clinical trials, which cover two main parts: 1) Clinically-investigated and emerging imaging biomarkers associated with radiotherapy, and 2) instrumental roles, functions, and activatable mechanisms of molecular imaging probes in the radiotherapy workflow. In addition, reflection and future perspectives are proposed. Results: Numerous imaging biomarkers have been continuously explored in decades, while few of them have been successfully validated for their correlation with radiotherapeutic outcomes and/or radiation-induced toxicities. Meanwhile, activatable molecular imaging probes towards the emerging biomarkers have exhibited to be promising in animal or small-scale human studies for precision radiotherapy. Conclusion: Biomarker-driven molecular imaging probes are essential for precision radiotherapy. Despite very inspiring preliminary results, validation of imaging biomarkers and rational design strategies of probes await robust and extensive investigations. Especially, the correlation between imaging biomarkers and radiotherapeutic outcomes/toxicities should be established through multi-center collaboration involving a large cohort of patients.
Collapse
Affiliation(s)
- Haonan Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, 699 Jinyuan Xi Road, Jimei District, 361021 Xiamen, Fujian, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|
10
|
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
|
11
|
Xu Y, Chen J, Zhang Y, Zhang P. Recent Progress in Peptide-Based Molecular Probes for Disease Bioimaging. Biomacromolecules 2024; 25:2222-2242. [PMID: 38437161 DOI: 10.1021/acs.biomac.3c01413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Recent strides in molecular pathology have unveiled distinctive alterations at the molecular level throughout the onset and progression of diseases. Enhancing the in vivo visualization of these biomarkers is crucial for advancing disease classification, staging, and treatment strategies. Peptide-based molecular probes (PMPs) have emerged as versatile tools due to their exceptional ability to discern these molecular changes with unparalleled specificity and precision. In this Perspective, we first summarize the methodologies for crafting innovative functional peptides, emphasizing recent advancements in both peptide library technologies and computer-assisted peptide design approaches. Furthermore, we offer an overview of the latest advances in PMPs within the realm of biological imaging, showcasing their varied applications in diagnostic and therapeutic modalities. We also briefly address current challenges and potential future directions in this dynamic field.
Collapse
Affiliation(s)
- Ying Xu
- School of Biomedical Engineering and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Junfan Chen
- School of Biomedical Engineering and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Yuan Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Pengcheng Zhang
- School of Biomedical Engineering and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
12
|
Zhang Y, Cao M, Wu Y, Malih S, Xu D, Yang E, Younis MH, Lin W, Zhao H, Wang C, Liu Q, Engle JW, Rasaee MJ, Guan Y, Huang G, Liu J, Cai W, Xie F, Wei W. Preclinical development of novel PD-L1 tracers and first-in-human study of [ 68Ga]Ga-NOTA-RW102 in patients with lung cancers. J Immunother Cancer 2024; 12:e008794. [PMID: 38580333 PMCID: PMC11002357 DOI: 10.1136/jitc-2024-008794] [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] [Accepted: 03/20/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND The programmed cell death protein-1 (PD-1)/programmed death receptor ligand 1 (PD-L1) axis critically facilitates cancer cells' immune evasion. Antibody therapeutics targeting the PD-1/PD-L1 axis have shown remarkable efficacy in various tumors. Immuno-positron emission tomography (ImmunoPET) imaging of PD-L1 expression may help reshape solid tumors' immunotherapy landscape. METHODS By immunizing an alpaca with recombinant human PD-L1, three clones of the variable domain of the heavy chain of heavy-chain only antibody (VHH) were screened, and RW102 with high binding affinity was selected for further studies. ABDRW102, a VHH derivative, was further engineered by fusing RW102 with the albumin binder ABD035. Based on the two targeting vectors, four PD-L1-specific tracers ([68Ga]Ga-NOTA-RW102, [68Ga]Ga-NOTA-ABDRW102, [64Cu]Cu-NOTA-ABDRW102, and [89Zr]Zr-DFO-ABDRW102) with different circulation times were developed. The diagnostic efficacies were thoroughly evaluated in preclinical solid tumor models, followed by a first-in-human translational investigation of [68Ga]Ga-NOTA-RW102 in patients with non-small cell lung cancer (NSCLC). RESULTS While RW102 has a high binding affinity to PD-L1 with an excellent KD value of 15.29 pM, ABDRW102 simultaneously binds to human PD-L1 and human serum albumin with an excellent KD value of 3.71 pM and 3.38 pM, respectively. Radiotracers derived from RW102 and ABDRW102 have different in vivo circulation times. In preclinical studies, [68Ga]Ga-NOTA-RW102 immunoPET imaging allowed same-day annotation of differential PD-L1 expression with specificity, while [64Cu]Cu-NOTA-ABDRW102 and [89Zr]Zr-DFO-ABDRW102 enabled longitudinal visualization of PD-L1. More importantly, a pilot clinical trial shows the safety and diagnostic value of [68Ga]Ga-NOTA-RW102 immunoPET imaging in patients with NSCLCs and its potential to predict immune-related adverse effects following PD-L1-targeted immunotherapies. CONCLUSIONS We developed and validated a series of PD-L1-targeted tracers. Initial preclinical and clinical evidence indicates that immunoPET imaging with [68Ga]Ga-NOTA-RW102 holds promise in visualizing differential PD-L1 expression, selecting patients for PD-L1-targeted immunotherapies, and monitoring immune-related adverse effects in patients receiving PD-L1-targeted treatments. TRIAL REGISTRATION NUMBER NCT06165874.
Collapse
Affiliation(s)
- You Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Min Cao
- Department of Thoracic Surgery,Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanfei Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Sara Malih
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Dong Xu
- Department of Thoracic Surgery, Huashan Hospital Fudan University, Shanghai, China
| | - Erpeng Yang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Muhsin H Younis
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Wilson Lin
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Haitao Zhao
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng Wang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiufang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jonathan W Engle
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mohammad J Rasaee
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
13
|
Sun X, Wu Y, Wang X, Gao X, Zhang S, Sun Z, Liu R, Hu K. Beyond Small Molecules: Antibodies and Peptides for Fibroblast Activation Protein Targeting Radiopharmaceuticals. Pharmaceutics 2024; 16:345. [PMID: 38543239 PMCID: PMC10974899 DOI: 10.3390/pharmaceutics16030345] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 04/05/2025] Open
Abstract
Fibroblast activation protein (FAP) is a serine protease characterized by its high expression in cancer-associated fibroblasts (CAFs) and near absence in adult normal tissues and benign lesions. This unique expression pattern positions FAP as a prospective biomarker for targeted tumor radiodiagnosis and therapy. The advent of FAP-based radiotheranostics is anticipated to revolutionize cancer management. Among various types of FAP ligands, peptides and antibodies have shown advantages over small molecules, exemplifying prolonged tumor retention in human volunteers. Within its scope, this review summarizes the recent research progress of the FAP radiopharmaceuticals based on antibodies and peptides in tumor imaging and therapy. Additionally, it incorporates insights from recent studies, providing valuable perspectives on the clinical utility of FAP-targeted radiopharmaceuticals.
Collapse
Affiliation(s)
- Xiaona Sun
- School of Printing and Packaging Engineer, Beijing Institute of Graphic Communication, Beijing 102600, China; (X.S.); (Y.W.); (Z.S.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (X.W.); (X.G.); (S.Z.)
| | - Yuxuan Wu
- School of Printing and Packaging Engineer, Beijing Institute of Graphic Communication, Beijing 102600, China; (X.S.); (Y.W.); (Z.S.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (X.W.); (X.G.); (S.Z.)
| | - Xingkai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (X.W.); (X.G.); (S.Z.)
| | - Xin Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (X.W.); (X.G.); (S.Z.)
| | - Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (X.W.); (X.G.); (S.Z.)
| | - Zhicheng Sun
- School of Printing and Packaging Engineer, Beijing Institute of Graphic Communication, Beijing 102600, China; (X.S.); (Y.W.); (Z.S.)
| | - Ruping Liu
- School of Printing and Packaging Engineer, Beijing Institute of Graphic Communication, Beijing 102600, China; (X.S.); (Y.W.); (Z.S.)
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (X.W.); (X.G.); (S.Z.)
| |
Collapse
|
14
|
Sui X, Niu X, Zhou X, Gao Y. Peptide drugs: a new direction in cancer immunotherapy. Cancer Biol Med 2023; 21:j.issn.2095-3941.2023.0297. [PMID: 38062861 PMCID: PMC10976324 DOI: 10.20892/j.issn.2095-3941.2023.0297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/28/2023] [Indexed: 09/19/2024] Open
Affiliation(s)
- Xinghua Sui
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Xiaoshuang Niu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Xiuman Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
15
|
Zhang S, Shen J, Wang X, Sun X, Wu Y, Zhang M, Wang R, Hu K. Integration of organoids in peptide drug discovery: Rise of the high‐throughput screening. VIEW 2023; 4. [DOI: 10.1002/viw.20230010] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/13/2023] [Indexed: 04/05/2025] Open
Abstract
AbstractOrganoids are three‐dimensional cell aggregates with near‐physiologic cell behaviors and can undergo long‐term expansion in vitro. They are amenable to high‐throughput drug screening processes, which renders them a viable preclinical model for drug development. The procedure of organoid‐based high‐throughput screening has been extensively employed to discover small‐molecule drugs, encompassing the steps of generating organoids, examining efficient drugs in organoid cultures, and data assessment. Compared to small molecules, peptides are more straightforward to synthesize, can be modified chemically, and demonstrate high target specificity and low cytotoxicity. Therefore, they have emerged as promising carriers to deliver drugs to disease‐associated targets and could be efficient therapeutic drugs for various diseases. To date, organoids have been used to evaluate the efficacy of certain peptide agents; however, no organoid‐based high‐throughput screening of peptide drugs has been reported. Given the advantages of peptide drugs, there is an urgent need to establish organoid‐based peptide high‐throughput screening platforms. In this review, we discuss the typical approach of screening small‐molecular drugs with the use of organoid cultures, as well as provide an overview of the studies that have incorporated organoids in peptide research. Drawing on the knowledge from small molecular screens, we explore the difficulties and potential avenues for creating new platforms to identify peptide agents using organoid models.
Collapse
Affiliation(s)
- Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Jieting Shen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Xingkai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Xiaona Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Yuxuan Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Ming‐Rong Zhang
- Department of Advanced Nuclear Medicine Sciences Institute of Quantum Medical Science National Institutes for Quantum Science and Technology Chiba Japan
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| |
Collapse
|
16
|
Zhang L, Zhang S, Wu J, Wang Y, Wu Y, Sun X, Wang X, Shen J, Xie L, Zhang Y, Zhang H, Hu K, Wang F, Wang R, Zhang MR. Linear Peptide-Based PET Tracers for Imaging PD-L1 in Tumors. Mol Pharm 2023; 20:4256-4267. [PMID: 37368947 DOI: 10.1021/acs.molpharmaceut.3c00382] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Programmed cell death receptor 1 (PD-1) and its ligand PD-L1 are particularly interesting immune checkpoint proteins for human cancer treatment. Positron emission tomography (PET) imaging allows for the dynamic monitoring of PD-L1 status during tumor progression, thus informing patients' response index. Herein, we report the synthesis of two linear peptide-based radiotracers, [64Cu]/[68Ga]HKP2201 and [64Cu]/[68Ga]HKP2202, and validate their utility for PD-L1 visualization in preclinical models. The precursor peptide HKP2201 was derived from a linear peptide ligand, CLP002, which was previously identified by phage display and showed nanomolar affinity toward PD-L1. Appropriate modification of CLP002 via PEGylation and DOTA conjugation yielded HKP2201. The dimerization of HKP2201 generated HKP2202. The 64Cu and 68Ga radiolabeling of both precursors was studied and optimized. PD-L1 expression in mouse melanoma cell line B16F10, mouse colon cancer cell line MC38, and their allografts were assayed by immunofluorescence and immunohistochemistry staining. Cellular uptake and binding assays were conducted in both cell lines. PET imaging and ex vivo biodistribution studies were employed in tumor mouse models bearing B16F10 and MC38 allografts. [64Cu]/[68Ga]HKP2201 and [64Cu]/[68Ga]HKP2202 were obtained with satisfactory radiocharacteristics. They all showed lower liver accumulation compared to [64Cu]/[68Ga]WL12. B16F10 and MC38 cells and their tumor allografts sections were verified to express PD-L1. These tracers demonstrated a concentration-dependent cell affinity and a comparable half-maximal effect concentration (EC50) with radiolabeled WL12. Competitive binding and blocking studies demonstrated the specific target of these tracers to PD-L1. PET imaging and ex vivo biodistribution studies revealed notable tumor uptake in tumor-bearing mice and rapid clearance from blood and major organs. Importantly, [64Cu]/[68Ga]HKP2202 showed higher tumor uptake compared to [64Cu]/[68Ga]HKP2201. Of note, [64Cu] labeled tracers showed longer retention in tumors than [68Ga] labeled traces, indicating advantages in the long-term tracking of PD-L1 dynamics. In comparison, [68Ga]HKP2201 and [68Ga]HKP2202 showed lower liver accumulation, enabling its great potential in the fast detection of both primary and metastatic tumors, including hepatic carcinoma. [64Cu]/[68Ga]HKP2201 and [64Cu]/[68Ga]HKP2202 are promising PET tracers for visualizing PD-L1 status. Notably, their combination would cooperate in rapid diagnosis and subsequent treatment guidance. Future assessment of the radiotracers in patients is needed to fully evaluate their clinical value.
Collapse
Affiliation(s)
- Lulu Zhang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210008, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jiang Wu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Yanrong Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Yuxuan Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaona Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xingkai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jieting Shen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lin Xie
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Hailong Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| |
Collapse
|
17
|
Chen Y, Guo Y, Liu Z, Hu X, Hu M. An overview of current advances of PD-L1 targeting immuno-imaging in cancers. J Cancer Res Ther 2023; 19:866-875. [PMID: 37675710 DOI: 10.4103/jcrt.jcrt_88_23] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
The programmed death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) pathway plays a significant role in immune evasion. PD-1 or PD-L1 immune checkpoint inhibitors (ICIs) have become a standard treatment for multiple types of cancer. To date, PD-L1 has served as a biomarker for predicting the efficacy of ICIs in several cancers. The need to establish an effective detection method that could visualize PD-L1 expression and predict the efficacy of PD-1/PD-L1 ICIs has promoted a search for new imaging strategies. PD-L1-targeting immuno-imaging could provide a noninvasive, real-time, repeatable, dynamic, and quantitative assessment of the characteristics of all tumor lesions in individual patients. This study analyzed the existing evidence in the literature on PD-L1-based immuno-imaging (2015-2022). Original English-language articles were searched using PubMed and Google Scholar. Keywords, such as "PD-L1," "PET," "SPECT," "PET/CT," and "SPECT/CT," were used in various combinations. A total of nearly 50 preclinical and clinical studies of PD-L1-targeting immuno-imaging were selected, reviewed, and included in this study. Therefore, in this review, we conducted a study of the advances in PD-L1-targeting immuno-imaging for detecting the expression of PD-L1 and the efficacy of ICIs. We focused on the different types of PD-L1-targeting agents, including antibodies and small PD-L1-binding agents, and illustrated the strength and weakness of these probes. Furthermore, we summarized the trends in the development of PD-L1-targeting immuno-imaging, as well as the current challenges and future directions for clinical workflow.
Collapse
Affiliation(s)
- Yunhao Chen
- Department of Radiation Oncology, Shandong University Cancer Center; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yujiao Guo
- Department of Oncology, The Affiliated Hospital of Jining Medical University, Jining, China
| | - Zhiguo Liu
- Department of PET/CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaokun Hu
- Department of the Interventional Medical Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Man Hu
- Department of Radiation Oncology, Shandong University Cancer Center; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
18
|
Su L, Dalby KS, Luehmann H, Elkassih SA, Cho S, He X, Detering L, Lin YN, Kang N, Moore DA, Laforest R, Sun G, Liu Y, Wooley KL. Ultrasmall, elementary and highly translational nanoparticle X-ray contrast media from amphiphilic iodinated statistical copolymers. Acta Pharm Sin B 2023; 13:1660-1670. [PMID: 37139426 PMCID: PMC10149980 DOI: 10.1016/j.apsb.2022.09.009] [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: 07/21/2022] [Revised: 08/18/2022] [Accepted: 09/05/2022] [Indexed: 11/01/2022] Open
Abstract
To expand the single-dose duration over which noninvasive clinical and preclinical cancer imaging can be conducted with high sensitivity, and well-defined spatial and temporal resolutions, a facile strategy to prepare ultrasmall nanoparticulate X-ray contrast media (nano-XRCM) as dual-modality imaging agents for positron emission tomography (PET) and computed tomography (CT) has been established. Synthesized from controlled copolymerization of triiodobenzoyl ethyl acrylate and oligo(ethylene oxide) acrylate monomers, the amphiphilic statistical iodocopolymers (ICPs) could directly dissolve in water to afford thermodynamically stable solutions with high aqueous iodine concentrations (>140 mg iodine/mL water) and comparable viscosities to conventional small molecule XRCM. The formation of ultrasmall iodinated nanoparticles with hydrodynamic diameters of ca. 10 nm in water was confirmed by dynamic and static light scattering techniques. In a breast cancer mouse model, in vivo biodistribution studies revealed that the 64Cu-chelator-functionalized iodinated nano-XRCM exhibited extended blood residency and higher tumor accumulation compared to typical small molecule imaging agents. PET/CT imaging of tumor over 3 days showed good correlation between PET and CT signals, while CT imaging allowed continuous observation of tumor retention even after 10 days post-injection, enabling longitudinal monitoring of tumor retention for imaging or potentially therapeutic effect after a single administration of nano-XRCM.
Collapse
Affiliation(s)
- Lu Su
- Department of Chemistry, Department of Materials Science and Engineering, and Department of Chemical Engineering, Texas A&M University, College Station, TX 77842, USA
- Laboratory of Macromolecular and Organic Chemistry and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, MB 5600, The Netherlands
| | - Kellie S. Dalby
- Department of Chemistry, Department of Materials Science and Engineering, and Department of Chemical Engineering, Texas A&M University, College Station, TX 77842, USA
| | - Hannah Luehmann
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Sussana A. Elkassih
- Department of Chemistry, Department of Materials Science and Engineering, and Department of Chemical Engineering, Texas A&M University, College Station, TX 77842, USA
| | - Sangho Cho
- Department of Chemistry, Department of Materials Science and Engineering, and Department of Chemical Engineering, Texas A&M University, College Station, TX 77842, USA
| | - Xun He
- Department of Chemistry, Department of Materials Science and Engineering, and Department of Chemical Engineering, Texas A&M University, College Station, TX 77842, USA
| | - Lisa Detering
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Yen-Nan Lin
- Department of Chemistry, Department of Materials Science and Engineering, and Department of Chemical Engineering, Texas A&M University, College Station, TX 77842, USA
| | - Nari Kang
- Department of Chemistry, Department of Materials Science and Engineering, and Department of Chemical Engineering, Texas A&M University, College Station, TX 77842, USA
| | | | - Richard Laforest
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Guorong Sun
- Department of Chemistry, Department of Materials Science and Engineering, and Department of Chemical Engineering, Texas A&M University, College Station, TX 77842, USA
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Karen L. Wooley
- Department of Chemistry, Department of Materials Science and Engineering, and Department of Chemical Engineering, Texas A&M University, College Station, TX 77842, USA
| |
Collapse
|
19
|
Xu L, Zhang L, Liang B, Zhu S, Lv G, Qiu L, Lin J. Design, Synthesis, and Biological Evaluation of a Small-Molecule PET Agent for Imaging PD-L1 Expression. Pharmaceuticals (Basel) 2023; 16:213. [PMID: 37259361 PMCID: PMC9968138 DOI: 10.3390/ph16020213] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/10/2023] [Accepted: 01/23/2023] [Indexed: 10/29/2023] Open
Abstract
Immunotherapy blocking programmed cell death protein 1/programmed death ligand 1 (PD-1/PD-L1) pathway has achieved great therapeutic effect in the clinic, but the overall response rate is not satisfactory. Early studies showed that response to treatment and overall survival could be positively related to PD-L1 expression in tumors. Therefore, accurate measurement of PD-L1 expression will help to screen cancer patients and improve the overall response rate. A small molecular positron emission tomography (PET) probe [18F]LP-F containing a biphenyl moiety was designed and synthesized for measurement of PD-L1 expression in tumors. The PET probe [18F]LP-F was obtained with a radiochemical yield of 12.72 ± 1.98%, a radiochemical purity of above 98% and molar activity of 18.8 GBq/μmol. [18F]LP-F had good stability in phosphate buffer saline (PBS) and mouse serum. In vitro assay indicated that [18F]LP-F showed moderate affinity to PD-L1. Micro-PET results showed that the tumor accumulation of [18F]LP-F in A375 tumor was inferior to that in A375-hPD-L1 tumor. All the results demonstrated that [18F]LP-F could specifically bind to PD-L1 and had a potential application in non-invasive evaluation of PD-L1 expression in tumors.
Collapse
Affiliation(s)
- Liang Xu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Lixia Zhang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Beibei Liang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Shiyu Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Gaochao Lv
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Ling Qiu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Jianguo Lin
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| |
Collapse
|
20
|
Sun J, Huangfu Z, Yang J, Wang G, Hu K, Gao M, Zhong Z. Imaging-guided targeted radionuclide tumor therapy: From concept to clinical translation. Adv Drug Deliv Rev 2022; 190:114538. [PMID: 36162696 DOI: 10.1016/j.addr.2022.114538] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 09/03/2022] [Accepted: 09/11/2022] [Indexed: 01/24/2023]
Abstract
Since the first introduction of sodium iodide I-131 for use with thyroid patients almost 80 years ago, more than 50 radiopharmaceuticals have reached the markets for a wide range of diseases, especially cancers. The nuclear medicine paradigm also shifts from solely molecular imaging or radionuclide therapy to imaging-guided radionuclide therapy, which is deemed a vital component of precision cancer therapy and an emerging medical modality for personalized medicine. The imaging-guided radionuclide therapy highlights the systematic integration of targeted nuclear diagnostics and radionuclide therapeutics. Regarding this, nuclear imaging serves to "visualize" the lesions and guide the therapeutic strategy, followed by administration of a precise patient specific dose of radiotherapeutics for treatment according to the absorbed dose to different organs and tumors calculated by dosimetry tools, and finally repeated imaging to predict the prognosis. This strategy leads to significantly enhanced therapeutic efficacy, improved patient outcomes, and manageable adverse events. In this review, we provide an overview of imaging-guided targeted radionuclide therapy for different tumors such as advanced prostate cancer and neuroendocrine tumors, with a focus on development of new radioligands and their preclinical and clinical results, and further discuss about challenges and future perspectives.
Collapse
Affiliation(s)
- Juan Sun
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhenyuan Huangfu
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Jiangtao Yang
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China.
| | - Kuan Hu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan.
| | - Mingyuan Gao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|
21
|
Huang C, Xu Y, Wang D, Chen Z, Fang W, Shi C, Xiao Z, Luo L. Interference With Redox Homeostasis Through a G6PD-Targeting Self-Assembled Hydrogel for the Enhancement of Sonodynamic Therapy in Breast Cancer. Front Chem 2022; 10:908892. [PMID: 35601559 PMCID: PMC9114499 DOI: 10.3389/fchem.2022.908892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/19/2022] [Indexed: 01/10/2023] Open
Abstract
Sonodynamics has emerged as a new potential therapy for breast cancer in recent years. However, GSH-mediated redox systems in cancer cells make them tolerable to oxidative stress-related therapy. Herein, in this study, with G6PD, the gatekeeper enzyme of the pentose phosphate pathway, as the regulative target, a self-assembled thermosensitive chitosan-pluronic hydrogel coloaded with ICG (sono-sensitive agent) and RRx-001 (IR@CPGel) was successfully prepared to enhance SDT through interference with redox homeostasis. Both in vitro and in vivo antitumor investigations verified that when integrated with sonodynamic therapy applied in breast cancer treatment, local administration of IR@CPgel could enhance ROS generation under LIFU irradiation and trigger the intrinsic apoptotic pathway of cancer cells, thus effectively inhibiting tumor growth in a safe manner. Moreover, RRx-001 may interfere with redox homeostasis in cancer cells by downregulating G6PD expression. Due to this redox imbalance, proapoptotic signals, such as P21 and P53, were enhanced, and metastasis-related signals, including MMP-2, ZEB1 and HIF-1α, were effectively reduced. Taken together, this work aimed to enhance the efficacy of sonodynamic therapy through local administration of self-assembled IR@CPGel to interfere with redox homeostasis and thus amplify the oxidative stress microenvironment in tumor tissues. In a word, this work provides a new strategy for the SDT enhancement in breast cancer therapy.
Collapse
Affiliation(s)
- Cuiqing Huang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Ultrasound, Guangdong Women and Children Hospital, Guangzhou, China
| | - Yuan Xu
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Duo Wang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zerong Chen
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Weimin Fang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Changzheng Shi
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Liangping Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
22
|
Special radionuclide production activities – recent developments at QST and throughout Japan. RADIOCHIM ACTA 2022. [DOI: 10.1515/ract-2021-1124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
National Institutes for Quantum Science and Technology (QST), formerly known as the National Institute of Radiological Sciences (NIRS), has been engaged in work on radiopharmaceutical science using cyclotrons since 1974. Eight pioneering researchers founded the basis of this field of research at NIRS, and to the present, many researchers and technicians have accumulated both scientific and technical achievements, as well as inherited the spirit of research. Besides, in recent years, we have developed production systems with AVF-930 cyclotron for various ‘non-standard’ radioisotopes applied in both diagnosis and therapy. Here, we review the past 50 years of our activities on radioisotope and radiopharmaceutical development, as well as more recent activities.
Collapse
|
23
|
Hu K, Ma X, Xie L, Zhang Y, Hanyu M, Obata H, Zhang L, Nagatsu K, Suzuki H, Shi R, Wang W, Zhang MR. Development of a Stable Peptide-Based PET Tracer for Detecting CD133-Expressing Cancer Cells. ACS OMEGA 2022; 7:334-341. [PMID: 35036703 PMCID: PMC8756568 DOI: 10.1021/acsomega.1c04711] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 12/09/2021] [Indexed: 05/08/2023]
Abstract
CD133 has been recognized as a prominent biomarker for cancer stem cells (CSCs), which promote tumor relapse and metastasis. Here, we developed a clinically relevant, stable, and peptide-based positron emission tomography (PET) tracer, [64Cu]CM-2, for mapping CD133 protein in several kinds of cancers. Through the incorporation of a 6-aminohexanoic acid (Ahx) into the N terminus of a CM peptide, we constructed a stable peptide tracer [64Cu]CM-2, which exhibited specific binding to CD133-positive CSCs in multiple preclinical tumor models. Both PET imaging and ex vivo biodistribution verified the superb performance of [64Cu]CM-2. Furthermore, the matched physical and biological half-life of [64Cu]CM-2 makes it a state-of-the-art PET tracer for CD133. Therefore, [64Cu]CM-2 PET may not only enable the longitudinal tracking of CD133 dynamics in the cancer stem cell niche but also provide a powerful and noninvasive imaging tool to track down CSCs in refractory cancers.
Collapse
Affiliation(s)
- Kuan Hu
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Xiaohui Ma
- Department
of Vascular Surgery, General Hospital of
People’s Liberation Army, Beijing 100853, P. R.
China
| | - Lin Xie
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Yiding Zhang
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Masayuki Hanyu
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Honoka Obata
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Lulu Zhang
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Kotaro Nagatsu
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Hisashi Suzuki
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Rui Shi
- Institute
of Traumatology and Orthopaedics Beijing
Jishuitan Hospital Beijing Laboratory of Biomedical Materials, Beijing 100035, P. R. China
| | - Weizhi Wang
- School
of Chemistry and Chemical Engineering, Beijing
Institute of Technology, Beijing 100081, P. R. China
| | - Ming-Rong Zhang
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| |
Collapse
|