1
|
Meng L, Fang J, Lin X, Zhuang R, Huang L, Li Y, Zhang X, Guo Z. Development of radioligands with an albumin-binding moiety of 4-(P-Iodophenyl) butyric acid for theranostic applications. J Control Release 2025; 382:113757. [PMID: 40262707 DOI: 10.1016/j.jconrel.2025.113757] [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: 12/28/2024] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025]
Abstract
The rapid clearance of imaging probes from blood circulation is beneficial for receptor imaging, as it minimizes non-target tissue exposure and improves tumor-to-background contrast. However, this rapid clearance can hinder radioligand therapy by limiting tumor uptake of radiolabeled compounds. An optimal blood half-life is crucial, as it enhances the uptake of radiolabeled compounds in targets, improving tumor uptake and retention of small molecule drugs, and thus therapeutic outcomes. To address this, strategies to extend blood half-life have been developed, with the addition of an albumin-binding moiety (ABM) being particularly effective. Among these, 4-(p-iodophenyl)butyric acid (IPBA) has emerged as a versatile ABM for radiopharmaceutical design. IPBA conjugation has successfully enhanced tissue distribution profiles across various cancer types. This review highlights recent progress in the design, radiosynthesis, and application of IPBA-based small molecular radioligands, providing insights for future clinical development of IPBA-based radiopharmaceuticals.
Collapse
Affiliation(s)
- Lingxin Meng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Jianyang Fang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xiaoru Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Rongqiang Zhuang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Lumei Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Yesen Li
- Department of Nuclear Medicine & Minnan PET Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Xianzhong Zhang
- Theranostics and Translational Research Center, Institute of Clinical Medicine, Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Zhide Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China; Department of Nuclear Medicine & Minnan PET Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China.
| |
Collapse
|
2
|
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
|
3
|
Zhou X, Zhu H, Luo C, Xiao H, Zou X, Zou J, Zhang G. Targeting integrin α5β1 in urological tumors: opportunities and challenges. Front Oncol 2023; 13:1165073. [PMID: 37483505 PMCID: PMC10358839 DOI: 10.3389/fonc.2023.1165073] [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: 02/13/2023] [Accepted: 06/16/2023] [Indexed: 07/25/2023] Open
Abstract
Urological tumors, such as prostate cancer, renal cell carcinoma, and bladder cancer, have shown a significant rise in prevalence in recent years and account for a significant proportion of malignant tumors. It has been established that metastasis to distant organs caused by urological tumors is the main cause of death, although the mechanisms underlying metastasis have not been fully elucidated. The fibronectin receptor integrin α5β1 reportedly plays an important role in distant metastasis and is closely related to tumor development. It is widely thought to be an important cancer mediator by interacting with different ligands, mediating tumor adhesion, invasion, and migration, and leading to immune escape. In this paper, we expound on the relationship and regulatory mechanisms of integrin α5β1 in these three cancers. In addition, the clinical applications of integrin α5β1 in these cancers, especially against treatment resistance, are discussed. Last but not least, the possibility of integrin α5β1 as a potential target for treatment is examined, with new ideas for future research being proposed.
Collapse
Affiliation(s)
- Xuming Zhou
- The First Clinical College, Gannan Medical University, Ganzhou, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Hezhen Zhu
- The First Clinical College, Gannan Medical University, Ganzhou, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Cong Luo
- The First Clinical College, Gannan Medical University, Ganzhou, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Huan Xiao
- The First Clinical College, Gannan Medical University, Ganzhou, China
| | - Xiaofeng Zou
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou, China
| | - Junrong Zou
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou, China
| | - Guoxi Zhang
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou, China
| |
Collapse
|
4
|
Ebrahimi F, Noaparast Z, Abedi SM, Hosseinimehr SJ. Homodimer 99mTc-HYNIC-E(SSSLTVPWY) 2 peptide improved HER2-overexpressed tumor targeting and imaging. Med Oncol 2022; 39:204. [PMID: 36175805 DOI: 10.1007/s12032-022-01798-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/15/2022] [Indexed: 06/16/2023]
Abstract
We hypothesized that a novel design of the LTVPWY (LY) peptide might exhibit a great potential for improving binding affinity and targeting HER2-overexpressed tumors. Hence, new dimer construction of 99mTc-labeled LY [99mTc-HYNIC-E(SSSLTVPWY)2] (99mTc-DLY) was introduced. Afterward, a head-to-head comparison of in vitro and in vivo experiments was performed between 99mTc-DLY and 99mTc-HYNIC-SSSLTVPWY as the monomer analog. The blocking dosage of trastuzumab reduced the uptake of the dimer about 20% more efficiently than the monomer in the SKOV-3 cell line. A twofold increase in competitive binding affinity and biological half-life was observed for 99mTc-DLY. The ovarian-tumor-bearing mice were detected with high contrast where the tumor-to-muscle ratio of 99mTc-DLY was notably increased about 40% using a gamma camera. The biodistribution experiment revealed an approximately 10% enhancement in tumor/blood, tumor/muscle, and tumor/bone ratios for the dimer. More rapid blood clearance was another achievement of the homodimer design. Overall, 99mTc-DLY successfully affected the pharmacokinetics and consequently the visualization of HER2-overexpressing tumors.
Collapse
Affiliation(s)
- Fatemeh Ebrahimi
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zohreh Noaparast
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mohammad Abedi
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
5
|
Hu B, Liu T, Li L, Shi L, Yao M, Li C, Ma X, Zhu H, Jia B, Wang F. IgG-Binding Nanobody Capable of Prolonging Nanobody-Based Radiotracer Plasma Half-Life and Enhancing the Efficacy of Tumor-Targeted Radionuclide Therapy. Bioconjug Chem 2022; 33:1328-1339. [PMID: 35687724 DOI: 10.1021/acs.bioconjchem.2c00209] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Nanobodies have been developed rapidly as targeted probes for molecular imaging owing to their high affinity, outstanding tissue penetration, and rapid blood clearance. However, the short retention time at the tumor site limits their application in targeted radionuclide therapy. In this study, we designed a dual-targeting nanobody referred to as MIRC213-709, which can specifically bind to the HER2 receptor in tumor cell lines with high affinity (by nanobody MIRC213) and endogenous IgG in plasma to prolong the half-life by the MIRC213 C-terminal fusion nanobody, MIRC709. The nanobodies were site-specifically radiolabeled with 99mTc and 177Lu, and radiochemical purity was >95% after purification. The long blood circulation time and tumor retention property of 99mTc/177Lu-MIRC213-709 were confirmed by a blood clearance assay, single-photon emission computed tomography (SPECT), and a biodistribution study. The blood clearance assay showed that the distribution phase half-life (T1/2α) and elimination phase half-life (T1/2β) of 99mTc-MIRC213-709 were 6.74- and 19.04-fold longer than those of 99mTc-MIRC213, respectively. The SPECT/CT and biodistribution results showed that the highest uptake of 177Lu-MIRC213 in the NCI-N87 model was 5.24 ± 0.95% ID/g at 6 h p.i., while the highest uptake of 177Lu-MIRC213-709 in the NCI-N87 model was 30.82 ± 7.29% ID/g at 48 h p.i. Compared with 177Lu-MIRC213, 177Lu-MIRC213-709 had a 16.9-fold increased tumor cumulative uptake (2606 ± 195.1 vs 153.9 ± 22.37% ID/g·h). The targeted radionuclide therapy assay was performed in the NCI-N87 tumor model, and treatment monitoring ended on day 32. The post-treatment/pretreatment tumor volumes were 12.99 ± 1.66, 3.58 ± 0.96, 1.26 ± 0.17, and 1.54 ± 0.50 in the 0, 9, and 18 MBq single-dose groups and the two 9 MBq divided dose group (14 days apart), respectively. All treatment groups showed significant therapeutic effects (P < 0.0001). Thus, fusion with the IgG-binding nanobody MIRC709 provides MIRC213 derivatives with improved metabolic properties for targeted radionuclide therapy.
Collapse
Affiliation(s)
- Biao Hu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Tianyu Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Liqiang Li
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Linqing Shi
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Meinan Yao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Chenzhen Li
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Xiaopan Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Fan Wang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
6
|
Palmitic Acid-Conjugated Radiopharmaceutical for Integrin αvβ3-Targeted Radionuclide Therapy. Pharmaceutics 2022; 14:pharmaceutics14071327. [PMID: 35890224 PMCID: PMC9321335 DOI: 10.3390/pharmaceutics14071327] [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: 04/13/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 01/25/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) is an emerging approach for patients with unresectable or metastatic tumors. Our previously optimized RGD peptide (3PRGD2) has excellent targeting specificity for a variety of integrin αvβ3/αvβ5-positive tumors and has been labeled with the therapeutic radionuclide [177Lu]LuCl3 for targeted radiotherapy of tumors. However, the rapid clearance of [177Lu]Lu-DOTA-3PRGD2 (177Lu-3PRGD2) in vivo requires two doses of 111 MBq/3 mCi to achieve effective tumor suppression, limiting its further clinical application. Albumin binders have been attached to drugs to facilitate binding to albumin in vivo to prolong the drug half-life in plasma and obtain long-term effects. In this study, we modified 3PRGD2 with albumin-binding palmitic acid (Palm-3PRGD2) and then radiolabeled Palm-3PRGD2 with 177Lu. [177Lu]Lu-DOTA-Palm-3PRGD2 (177Lu-Palm-3PRGD2) retained a specific binding affinity for integrin αvβ3/αvβ5, with an IC50 value of 5.13 ± 1.16 nM. Compared with 177Lu-3PRGD2, the 177Lu-Palm-3PRGD2 circulation time in blood was more than 6 times longer (slow half-life: 73.42 min versus 11.81 min), and the tumor uptake increased more than fivefold (21.34 ± 4.65 %IA/g and 4.11 ± 0.70 %IA/g at 12 h post-injection). Thus, the significant increase in tumor uptake and tumor retention resulted in enhanced efficacy of targeted radiotherapy, and tumor growth was completely inhibited by a single and relatively lowdose of 18.5 MBq/0.5 mCi. Thus, 177Lu-Palm-3PRGD2 shows great potential for clinical application.
Collapse
|
7
|
Guan J, Yuan C, Tian X, Cheng L, Gao H, Yao Q, Wang X, Wu H, Chen Z, Jian F. SPECT Imaging of Acute Disc Herniation by Targeting Integrin α5β1 in Rat Models. Front Neurol 2022; 13:782967. [PMID: 35614922 PMCID: PMC9124789 DOI: 10.3389/fneur.2022.782967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Traditional morphological imaging of intervertebral disc herniation (IVDH) is challenging in early disease diagnosis. Aiming at the early diagnosis of IVD by non-invasive molecular imaging targeting of integrin α5β1, we performed novel imaging in rats with acute IVDH for the first time. Methods Animal models were prepared by conducting an established needle puncture procedure through the normal intervertebral disc (IVD). The disc-injured rats underwent SPECT/CT imaging of the 99mTc-3PisoDGR2 peptide at 1 day to 2 months postinjury. The expression change of integrin α5β1 was determined by anti-integrin α5 and anti-integrin α5β1 immunohistochemistry (IHC). Magnetic resonance imaging (MRI) was performed for comparison during disease progression. The morphological changes of the disc were determined by safranin-O staining. Results Rats with acute IVDH showed gradually increased disc uptake of 99mTc-3PisoDGR2 from 1 to 7 days posttreatment, which was a significantly higher level than that of the normal disks in degenerative diseases. IHC results showed the expression of integrin α5β1 on the surface of annulus fibrosus (AF) cells and nucleus pulposus (NP) cells, which agreed with the uptake data. MRI showed a progressively decreased T2 density and MRI index throughout the investigation. Hematoxylin and eosin (HE) staining and safranin-O staining revealed a disorganized structure of the IVD as well as loss of proteoglycans after puncture. Conclusions The present study demonstrated a good correlation between integrin α5β1 expression and acute disc herniation. The SPECT/CT imaging of 99mTc-3PisoDGR2 targeting integrin α5β1 may diagnose IVDH in an acute phase for early disease management.
Collapse
Affiliation(s)
- Jian Guan
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Spine Center, China International Neuroscience Institute (CHINA-INI), Beijing, China
- Research Center of Spine and Spinal Cord, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- National Center for Neurological Disorders, Xuanwu Hospital, Beijing, China
| | - Chenghua Yuan
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Spine Center, China International Neuroscience Institute (CHINA-INI), Beijing, China
- Research Center of Spine and Spinal Cord, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- National Center for Neurological Disorders, Xuanwu Hospital, Beijing, China
| | - Xin Tian
- Center for Experimental Animals, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Lei Cheng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Spine Center, China International Neuroscience Institute (CHINA-INI), Beijing, China
- Research Center of Spine and Spinal Cord, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- National Center for Neurological Disorders, Xuanwu Hospital, Beijing, China
| | - Hannan Gao
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Qingyu Yao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Spine Center, China International Neuroscience Institute (CHINA-INI), Beijing, China
- Research Center of Spine and Spinal Cord, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- National Center for Neurological Disorders, Xuanwu Hospital, Beijing, China
| | - Xinyu Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Spine Center, China International Neuroscience Institute (CHINA-INI), Beijing, China
- Research Center of Spine and Spinal Cord, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- National Center for Neurological Disorders, Xuanwu Hospital, Beijing, China
| | - Hao Wu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Spine Center, China International Neuroscience Institute (CHINA-INI), Beijing, China
- Research Center of Spine and Spinal Cord, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- National Center for Neurological Disorders, Xuanwu Hospital, Beijing, China
| | - Zan Chen
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Spine Center, China International Neuroscience Institute (CHINA-INI), Beijing, China
- Research Center of Spine and Spinal Cord, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- National Center for Neurological Disorders, Xuanwu Hospital, Beijing, China
- *Correspondence: Zan Chen
| | - Fengzeng Jian
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Spine Center, China International Neuroscience Institute (CHINA-INI), Beijing, China
- Research Center of Spine and Spinal Cord, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- National Center for Neurological Disorders, Xuanwu Hospital, Beijing, China
- Fengzeng Jian
| |
Collapse
|
8
|
China’s radiopharmaceuticals on expressway: 2014–2021. RADIOCHIM ACTA 2022. [DOI: 10.1515/ract-2021-1137] [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
This review provides an essential overview on the progress of rapidly-developing China’s radiopharmaceuticals in recent years (2014–2021). Our discussion reflects on efforts to develop potential, preclinical, and in-clinical radiopharmaceuticals including the following areas: (1) brain imaging agents, (2) cardiovascular imaging agents, (3) infection and inflammation imaging agents, (4) tumor radiopharmaceuticals, and (5) boron delivery agents (a class of radiopharmaceutical prodrug) for neutron capture therapy. Especially, the progress in basic research, including new radiolabeling methodology, is highlighted from a standpoint of radiopharmaceutical chemistry. Meanwhile, we briefly reflect on the recent major events related to radiopharmaceuticals along with the distribution of major R&D forces (universities, institutions, facilities, and companies), clinical study status, and national regulatory supports. We conclude with a brief commentary on remaining limitations and emerging opportunities for China’s radiopharmaceuticals.
Collapse
|
9
|
Ebrahimi F, Hosseinimehr SJ. Homomultimer strategy for improvement of radiolabeled peptides and antibody fragments in tumor targeting. Curr Med Chem 2022; 29:4923-4957. [PMID: 35450521 DOI: 10.2174/0929867329666220420131836] [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: 11/23/2021] [Revised: 01/18/2022] [Accepted: 02/07/2022] [Indexed: 11/22/2022]
Abstract
A homomultimeric radioligand is composed of multiple identical ligands connected to the linker and radionuclide to detect a variety of overexpressed receptors on cancer cells. Multimer strategy holds great potential for introducing new radiotracers based on peptide and monoclonal antibody (mAb) derivatives in molecular imaging and therapy. It offers a reliable procedure for the preparation of biological-based targeting with diverse affinities and pharmacokinetics. In this context, we provide a useful summary and interpretation of the main results by a comprehensive look at multimeric radiopharmaceuticals in nuclear oncology. Therefore, there will be explanations for the strategy mechanisms and the main variables affecting the biodistribution results. The discussion is followed by highlights of recent work in the targeting of various types of receptors. The consequences are expressed based on comparing some parameters between monomer and multimer counterparts in each relevant section.
Collapse
Affiliation(s)
- Fatemeh Ebrahimi
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
10
|
Davis RA, Hausner SH, Harris R, Sutcliffe JL. A Comparison of Evans Blue and 4-( p-Iodophenyl)butyryl Albumin Binding Moieties on an Integrin α vβ 6 Binding Peptide. Pharmaceutics 2022; 14:pharmaceutics14040745. [PMID: 35456579 PMCID: PMC9025560 DOI: 10.3390/pharmaceutics14040745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 01/26/2023] Open
Abstract
Serum albumin binding moieties (ABMs) such as the Evans blue (EB) dye fragment and the 4-(p-iodophenyl)butyryl (IP) have been used to improve the pharmacokinetic profile of many radiopharmaceuticals. The goal of this work was to directly compare these two ABMs when conjugated to an integrin αvβ6 binding peptide (αvβ6-BP); a peptide that is currently being used for positron emission tomography (PET) imaging in patients with metastatic cancer. The ABM-modified αvβ6-BP peptides were synthesized with a 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetracetic acid (DOTA) chelator for radiolabeling with copper-64 to yield [64Cu]Cu DOTA-EB-αvβ6-BP ([64Cu]1) and [64Cu]Cu DOTA-IP-αvβ6-BP ([64Cu]2). Both peptides were evaluated in vitro for serum albumin binding, serum stability, and cell binding and internalization in the paired engineered melanoma cells DX3puroβ6 (αvβ6 +) and DX3puro (αvβ6 −), and pancreatic BxPC-3 (αvβ6 +) cells and in vivo in a BxPC-3 xenograft mouse model. Serum albumin binding for [64Cu]1 and [64Cu]2 was 53−63% and 42−44%, respectively, with good human serum stability (24 h: [64Cu]1 76%, [64Cu]2 90%). Selective αvβ6 cell binding was observed for both [64Cu]1 and [64Cu]2 (αvβ6 (+) cells: 30.3−55.8% and 48.5−60.2%, respectively, vs. αvβ6 (−) cells <3.1% for both). In vivo BxPC-3 tumor uptake for both peptides at 4 h was 5.29 ± 0.59 and 7.60 ± 0.43% ID/g ([64Cu]1 and [64Cu]2, respectively), and remained at 3.32 ± 0.46 and 4.91 ± 1.19% ID/g, respectively, at 72 h, representing a >3-fold improvement over the non-ABM parent peptide and thereby providing improved PET images. Comparing [64Cu]1 and [64Cu]2, the IP-ABM-αvβ6-BP [64Cu]2 displayed higher serum stability, higher tumor accumulation, and lower kidney and liver accumulation, resulting in better tumor-to-organ ratios for high contrast visualization of the αvβ6 (+) tumor by PET imaging.
Collapse
Affiliation(s)
- Ryan A. Davis
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA;
| | - Sven H. Hausner
- Department of Internal Medicine, Division of Hematology/Oncology, University of California, Davis, CA 95817, USA; (S.H.H.); (R.H.)
| | - Rebecca Harris
- Department of Internal Medicine, Division of Hematology/Oncology, University of California, Davis, CA 95817, USA; (S.H.H.); (R.H.)
| | - Julie L. Sutcliffe
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA;
- Department of Internal Medicine, Division of Hematology/Oncology, University of California, Davis, CA 95817, USA; (S.H.H.); (R.H.)
- Center for Molecular and Genomic Imaging, University of California, Davis, CA 95616, USA
- Correspondence: ; Tel.: +1-916-734-5536
| |
Collapse
|
11
|
Stroet MCM, de Blois E, de Jong M, Seimbille Y, Mezzanotte L, Löwik CWGM, Panth KM. Improved Multimodal Tumor Necrosis Imaging with IRDye800CW-DOTA Conjugated to an Albumin-Binding Domain. Cancers (Basel) 2022; 14:cancers14040861. [PMID: 35205609 PMCID: PMC8870237 DOI: 10.3390/cancers14040861] [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: 12/10/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Anti-tumor treatment efficacy is determined by tumor shrinkage, which takes valuable time to become apparent and poses a risk of unnecessary treatment with severe side effects. Therefore, there is an unmet need for more reliable and specific methods to monitor treatment efficacy. We explore radiolabeled cyanines for imaging tumor necrosis as a unique marker for therapy efficacy. Moreover, spontaneous tumor necrosis is a hallmark for aggressively growing tumor types with poor prognosis. We improved the binding properties of a previously reported necrosis-avid contrast agent (NACA) and successfully detected spontaneous and therapy-induced tumor necrosis in mice using radioactivity and fluorescence imaging modalities. This NACA may pave the way to in vivo detection of tumor necrosis for early-stage determination of tumor aggressiveness and therapy efficacy. Abstract Purpose: To assess our improved NACA for the detection of tumor necrosis. Methods: We increased the blood circulation time of our NACA by adding an albumin-binding domain to the molecular structure. We tested the necrosis avidity on dead or alive cultured cells and performed SPECT and fluorescence imaging of both spontaneous and treatment-induced necrosis in murine breast cancer models. We simultaneously recorded [18F]FDG-PET and bioluminescence images for complementary detection of tumor viability. Results: We generated two albumin-binding IRDye800CW derivatives which were labeled with indium-111 with high radiochemical purity. Surprisingly, both albumin-binding NACAs had >10x higher in vitro binding towards dead cells. We selected [111In]3 for in vivo experiments which showed higher dead cell binding in vitro and in vivo stability. The doxorubicin-treated tumors showed increased [111In]3-uptake (1.74 ± 0.08%ID/g after saline treatment, 2.25 ± 0.16%ID/g after doxorubicin treatment, p = 0.044) and decreased [18F]FDG-uptake (3.02 ± 0.51%ID/g after saline treatment, 1.79 ± 0.11%ID/g after doxorubicin treatment, p = 0.040), indicating therapy efficacy. Moreover, we detected increased [111In]3-uptake and tumor necrosis in more rapidly growing EMT6 tumors. Conclusions: Our albumin-binding NACA based on IRDye800CW facilitates tumor-necrosis imaging for assessment of therapy efficacy and aggressiveness in solid tumors using both fluorescence and SPECT imaging.
Collapse
Affiliation(s)
- Marcus C. M. Stroet
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Erasmus MC, Department of Molecular Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Erik de Blois
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
| | - Marion de Jong
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
| | - Yann Seimbille
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Life Sciences Division, TRIUMF, Vancouver, BC V6T 2A3, Canada
| | - Laura Mezzanotte
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Erasmus MC, Department of Molecular Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Clemens W. G. M. Löwik
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Erasmus MC, Department of Molecular Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- CHUV Department of Oncology, University of Lausanne, CH-1066 Lausanne, Switzerland
- Correspondence: (C.W.G.M.L.); (K.M.P.)
| | - Kranthi M. Panth
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Erasmus MC, Department of Molecular Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Correspondence: (C.W.G.M.L.); (K.M.P.)
| |
Collapse
|
12
|
Wang L, Zhang D, Li J, Li F, Wei R, Jiang G, Xu H, Wang X, Zhou Y, Xi L. A novel ICG-labeled cyclic TMTP1 peptide dimer for sensitive tumor imaging and enhanced photothermal therapy in vivo. Eur J Med Chem 2021; 227:113935. [PMID: 34731764 DOI: 10.1016/j.ejmech.2021.113935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/26/2022]
Abstract
TMTP1 is a polypeptide independently screened in our laboratory, which can target tumors in situ and metastases. In previous work, we have successfully developed a near-infrared (NIR) probe TMTP1-PEG4-ICG for tumor imaging. However, the limited ability to target tumor micrometastases hinders its further clinical application. Multimerization of peptides has been extensively demonstrated as an effective strategy to increase receptor binding affinity due to "multivalent effect" or "apparent cooperative affinity". In this study, a novel TMTP1 homodimer-directed NIR probe (TMTP1-PEG4)2-ICG was successfully constructed and synthesized. The cyclic TMTP1 peptides were bridged by two PEG4 linkers and then labeled with ICG-NHS for tumor imaging and photothermal therapy. In vivo biodistribution were assessed in normal BALB/c mice, and tumor targeting abilities of (TMTP1-PEG4)2-ICG and its monomer were evaluated and compared in 4T1-bearing subcutaneous tumor and lymph node metastasis model mice. Biodistribution analysis in vivo revealed that (TMTP1-PEG4)2-ICG was cleared mainly in both liver and kidney dependent way. Comparing with free ICG dye or TMTP1-PEG4-ICG probe, this improved (TMTP1-PEG4)2-ICG dimer showed more sensitive tumor imaging and could clearly identify tumors at a minimum volume of 10 mm3. Additionally, when compared to its monomer, lymph node (LN) metastases could also be apparently visualized and easily distinguished from normal LN by the novel dimer at 24 h post-injection. The blocking study revealed that the tumor accumulation of this probe was specifically medicated by receptor-ligand interaction. Furthermore, with the increase in stability and tumor targeting ability of ICG in vivo, the probe could also be an attractive photothermal agent to significantly inhibit tumor growth under 808 nm NIR laser irradiation. In conclusion, our work revealed that the novel (TMTP1-PEG4)2-ICG dimer could be a promising theranostic agent for sensitive tumor imaging and imaging-guided photothermal therapy, indicating its broad prospects for further clinical transformation.
Collapse
Affiliation(s)
- Ling Wang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Danya Zhang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jie Li
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Fei Li
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Rui Wei
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Guiying Jiang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Hanjie Xu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xueqian Wang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Ying Zhou
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Ling Xi
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
13
|
Abstract
A growing theme in chemistry is the joining of multiple organic molecular building blocks to create functional molecules. Diverse derivatizable structures—here termed “scaffolds” comprised of “hubs”—provide the foundation for systematic covalent organization of a rich variety of building blocks. This review encompasses 30 tri- or tetra-armed molecular hubs (e.g., triazine, lysine, arenes, dyes) that are used directly or in combination to give linear, cyclic, or branched scaffolds. Each scaffold is categorized by graph theory into one of 31 trees to express the molecular connectivity and overall architecture. Rational chemistry with exacting numbers of derivatizable sites is emphasized. The incorporation of water-solubilization motifs, robust or self-immolative linkers, enzymatically cleavable groups and functional appendages affords immense (and often late-stage) diversification of the scaffolds. Altogether, 107 target molecules are reviewed along with 19 syntheses to illustrate the distinctive chemistries for creating and derivatizing scaffolds. The review covers the history of the field up through 2020, briefly touching on statistically derivatized carriers employed in immunology as counterpoints to the rationally assembled and derivatized scaffolds here, although most citations are from the past two decades. The scaffolds are used widely in fields ranging from pure chemistry to artificial photosynthesis and biomedical sciences.
Collapse
|
14
|
Zhao H, Gao H, Luo C, Yang G, Zhao X, Gao S, Ma Q, Jia B, Shi J, Wang F. An Integrin-α vβ 6/α 5β 1-Bitargeted Probe for the SPECT Imaging of Pancreatic Adenocarcinoma in Preclinical and Primary Clinical Studies. Bioconjug Chem 2021; 32:1298-1305. [PMID: 34137602 DOI: 10.1021/acs.bioconjchem.1c00296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Pancreatic adenocarcinoma (PA) is one of the deadliest human malignancies. However, early detection, prediction of surgical resectability, and prognosis of PA are challenging with current conventional imaging technologies in the clinic. Molecular imaging technologies combined with novel imaging probes could be useful for early detection and accurate staging of PA. Integrin αvβ6 and α5β1 are found to be overexpressed in PA. In this study, integrin αvβ6/α5β1-bitargeted probes 99mTc-HYNIC-isoDGR (99mTc-isoDGR) and 99mTc-HYNIC-PEG4-PisoDGR2 (99mTc-3PisoDGR2) were prepared and evaluated in the BxPC-3 human pancreatic tumor model. Both subcutaneous and in situ BxPC-3 tumors could be clearly visualized by 99mTc-isoDGR nanoScan SPECT/CT imaging with a high ratio of tumor to background. The blocking study with excess nonradioactive peptide showed a significantly reduced tumor uptake, which confirmed the specificity of 99mTc-isoDGR. Biodistribution results confirmed the imaging results. The dimer tracer 99mTc-3PisoDGR2 significantly enhanced tumor uptake compared with 99mTc-isoDGR, and the spontaneous PA lesion in the mouse model could be clearly visualized by 99mTc-3PisoDGR2. The primary clinical study also verified the ability of 99mTc-3PisoDGR2 for detection of PA. Therefore, SPECT/CT imaging using the integrin αvβ6/α5β1-bitargeted 99mTc-3PisoDGR2 provided a potential approach for the noninvasive detection of PA.
Collapse
Affiliation(s)
- Haitao Zhao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China.,Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hannan Gao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Chuangwei Luo
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Guangjie Yang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Xiaoyu Zhao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Shi Gao
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, China.,NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, Jilin 130021, China
| | - Qingjie Ma
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, China.,NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, Jilin 130021, China
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Jiyun Shi
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fan Wang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China.,Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
15
|
Zhao W, Yu X, Peng S, Luo Y, Li J, Lu L. Construction of nanomaterials as contrast agents or probes for glioma imaging. J Nanobiotechnology 2021; 19:125. [PMID: 33941206 PMCID: PMC8091158 DOI: 10.1186/s12951-021-00866-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023] Open
Abstract
Malignant glioma remains incurable largely due to the aggressive and infiltrative nature, as well as the existence of blood-brain-barrier (BBB). Precise diagnosis of glioma, which aims to accurately delineate the tumor boundary for guiding surgical resection and provide reliable feedback of the therapeutic outcomes, is the critical step for successful treatment. Numerous imaging modalities have been developed for the efficient diagnosis of tumors from structural or functional aspects. However, the presence of BBB largely hampers the entrance of contrast agents (Cas) or probes into the brain, rendering the imaging performance highly compromised. The development of nanomaterials provides promising strategies for constructing nano-sized Cas or probes for accurate imaging of glioma owing to the BBB crossing ability and other unique advantages of nanomaterials, such as high loading capacity and stimuli-responsive properties. In this review, the recent progress of nanomaterials applied in single modal imaging modality and multimodal imaging for a comprehensive diagnosis is thoroughly summarized. Finally, the prospects and challenges are offered with the hope for its better development.
Collapse
Affiliation(s)
- Wei Zhao
- Zhuhai Precision Medical Center, Zhuhai Interventional Medical Center, Zhuhai People's Hospital (Affiliated With Jinan University), Zhuhai, 519000, Guangdong, China
| | - Xiangrong Yu
- Zhuhai Precision Medical Center, Zhuhai Interventional Medical Center, Zhuhai People's Hospital (Affiliated With Jinan University), Zhuhai, 519000, Guangdong, China
| | - Shaojun Peng
- Zhuhai Precision Medical Center, Zhuhai Interventional Medical Center, Zhuhai People's Hospital (Affiliated With Jinan University), Zhuhai, 519000, Guangdong, China
| | - Yu Luo
- School of Chemical Science and Engineering, Tongji University, 1239 Siping Road, Shanghai, China.
| | - Jingchao Li
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China.
| | - Ligong Lu
- Zhuhai Precision Medical Center, Zhuhai Interventional Medical Center, Zhuhai People's Hospital (Affiliated With Jinan University), Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
16
|
Ludwig BS, Kessler H, Kossatz S, Reuning U. RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field. Cancers (Basel) 2021; 13:1711. [PMID: 33916607 PMCID: PMC8038522 DOI: 10.3390/cancers13071711] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
Integrins have been extensively investigated as therapeutic targets over the last decades, which has been inspired by their multiple functions in cancer progression, metastasis, and angiogenesis as well as a continuously expanding number of other diseases, e.g., sepsis, fibrosis, and viral infections, possibly also Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). Although integrin-targeted (cancer) therapy trials did not meet the high expectations yet, integrins are still valid and promising targets due to their elevated expression and surface accessibility on diseased cells. Thus, for the future successful clinical translation of integrin-targeted compounds, revisited and innovative treatment strategies have to be explored based on accumulated knowledge of integrin biology. For this, refined approaches are demanded aiming at alternative and improved preclinical models, optimized selectivity and pharmacological properties of integrin ligands, as well as more sophisticated treatment protocols considering dose fine-tuning of compounds. Moreover, integrin ligands exert high accuracy in disease monitoring as diagnostic molecular imaging tools, enabling patient selection for individualized integrin-targeted therapy. The present review comprehensively analyzes the state-of-the-art knowledge on the roles of RGD-binding integrin subtypes in cancer and non-cancerous diseases and outlines the latest achievements in the design and development of synthetic ligands and their application in biomedical, translational, and molecular imaging approaches. Indeed, substantial progress has already been made, including advanced ligand designs, numerous elaborated pre-clinical and first-in-human studies, while the discovery of novel applications for integrin ligands remains to be explored.
Collapse
Affiliation(s)
- Beatrice S. Ludwig
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
| | - Horst Kessler
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics and Gynecology, University Hospital Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
| |
Collapse
|
17
|
Haque S, Norbert CC, Patra CR. Nanomedicine: future therapy for brain cancers. NANO DRUG DELIVERY STRATEGIES FOR THE TREATMENT OF CANCERS 2021:37-74. [DOI: 10.1016/b978-0-12-819793-6.00003-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
18
|
Hou J, Yan D, Liu Y, Huang P, Cui H. The Roles of Integrin α5β1 in Human Cancer. Onco Targets Ther 2020; 13:13329-13344. [PMID: 33408483 PMCID: PMC7781020 DOI: 10.2147/ott.s273803] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/21/2020] [Indexed: 12/19/2022] Open
Abstract
Cell adhesion to the extracellular matrix has important roles in tissue integrity and human health. Integrins are heterodimeric cell surface receptors that are composed by two non-covalently linked alpha and beta subunits that mainly participate in the interaction of cell-cell adhesion and cell-extracellular matrix and regulate cell motility, adhesion, differentiation, migration, proliferation, etc. In mammals, there have been eighteen α subunits and 8 β subunits and so far 24 distinct types of αβ integrin heterodimers have been identified in humans. Integrin α5β1, also known as the fibronectin receptor, is a heterodimer with α5 and β1 subunits and has emerged as an essential mediator in many human carcinomas. Integrin α5β1 alteration is closely linked to the progression of several types of human cancers, including cell proliferation, angiogenesis, tumor metastasis, and cancerogenesis. In this review, we will introduce the functions of integrin α5β1 in cancer progression and also explore its regulatory mechanisms. Additionally, the potential clinical applications as a target for cancer imaging and therapy are discussed. Collectively, the information reviewed here may increase the understanding of integrin α5β1 as a potential therapeutic target for cancer.
Collapse
Affiliation(s)
- Jianbing Hou
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400716, People's Republic of China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, People's Republic of China.,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, People's Republic of China
| | - Du Yan
- Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400716, People's Republic of China
| | - Yudong Liu
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400716, People's Republic of China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, People's Republic of China.,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, People's Republic of China
| | - Pan Huang
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400716, People's Republic of China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, People's Republic of China.,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, People's Republic of China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400716, People's Republic of China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, People's Republic of China.,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, People's Republic of China
| |
Collapse
|
19
|
Luo Q, Yang G, Gao H, Wang Y, Luo C, Ma X, Gao Y, Li X, Zhao H, Jia B, Shi J, Wang F. An Integrin Alpha 6-Targeted Radiotracer with Improved Receptor Binding Affinity and Tumor Uptake. Bioconjug Chem 2020; 31:1510-1521. [PMID: 32347718 DOI: 10.1021/acs.bioconjchem.0c00170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this study, we reported a 99mTc-labeled integrin α6-targeted peptide as the molecular imaging probe for tumor imaging by single-photon emission computed tomography (SPECT). We found that replacing Cys-Cys cyclized RWY peptide (sequence: cCRWYDENAC) with lactam-bridged cyclic cKiE peptide (sequence: cKRWYDENAisoE) did not sacrifice the integrin α6-binding affinity and specificity of cKiE radiotracer. To further improve the radiotracer's tumor targeting capability, the dimerized cKiE peptide (termed cKiE2) was designed, and the corresponding radiotracer 99mTc-cKiE2 was evaluated for tumor uptake and in vivo pharmacokinetics properties in tumor models. We found that cKiE2 showed higher binding affinity to integrin α6 than did monomeric RWY or cKiE peptide. The biodistribution results showed that the tumor uptake of 99mTc-cKiE2 was twice higher than that of 99mTc-RWY (3.20 ± 0.12 vs 1.26 ± 0.06 %ID/g, P < 0.001) at 0.5 h postinjection. The tumor to nontargeting tissue ratios were also enhanced in most normal organs. Specificity of 99mTc-cKiE2 for integrin α6 was demonstrated by competitive blocking of tumor uptake with excess cold peptide (3.20 ± 0.24 to 1.38 ± 0.23 %ID/g, P < 0.001). The integrin α6-positive tumors were clearly visualized by 99mTc-cKiE2/SPECT with low background except with a relatively high kidney uptake. The tumor uptake of 99mTc-cKiE2 correlates well with the tumor integrin α6 expression levels in a linear fashion (R2 = 0.9623). We also compared 99mTc-cKiE2 with an integrin αvβ3-targeted radiotracer 99mTc-3PRGD2 in the orthotopic hepatocellular carcinoma tumor models. We found that the orthotopic tumor was clearly visualized with 99mTc-cKiE2. 99mTc-3PRGD2 imaging did not show tumor contours in situ as clearly as 99mTc-cKiE2. The tumor-to-liver ratios of 99mTc-cKiE2 and 99mTc-3PRGD2 were 2.20 ± 0.17 and 0.85 ± 0.20. In conclusion, 99mTc-cKiE2 is an improved SPECT radiotracer for imaging integrin α6-positive tumors and has great potential for further clinical application.
Collapse
Affiliation(s)
- Qi Luo
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China.,University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Guangjie Yang
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Hannan Gao
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Yanpu Wang
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Chuangwei Luo
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Xiaotu Ma
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China.,University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yu Gao
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China.,University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Xiaoda Li
- Medical and Healthy Analytical Center, Peking University, Beijing 100191, People's Republic of China
| | - Huiyun Zhao
- Medical and Healthy Analytical Center, Peking University, Beijing 100191, People's Republic of China
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Jiyun Shi
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Fan Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China.,Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
| |
Collapse
|
20
|
Um W, Park J, Youn A, Cho H, Lim S, Lee JW, Yoon HY, Lim DK, Park JH, Kim K. A Comparative Study on Albumin-Binding Molecules for Targeted Tumor Delivery through Covalent and Noncovalent Approach. Bioconjug Chem 2019; 30:3107-3118. [DOI: 10.1021/acs.bioconjchem.9b00760] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Wooram Um
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Jooho Park
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Ahye Youn
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Hanhee Cho
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Seungho Lim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Jong Won Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Hong Yeol Yoon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Jae Hyung Park
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|