1
|
Guo Y, Guo H, Tong H, Xue W, Xie T, Wang L, Tong H. The effect Of vascular related CeRNA genes and corresponding imaging biomarkers on survival in lower grade glioma. Ir J Med Sci 2024; 193:653-663. [PMID: 37801268 DOI: 10.1007/s11845-023-03536-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND & AIMS To investigate the differential expression of vascular related ceRNA regulatory genes in LGG with different mutations of IDH1 and MGMT, and to verify imaging gene markers that can be closely associated with vascular related ceRNA regulatory genes. METHOD Five hundred fifteen patients with LGG were collected from TCGA database. CeRNA network analysis, GO analysis and Cox risk regression were used to find vascular ceRNA regulatory genes and their genetic markers related to survival. The preoperative MRI image data and postoperative tumor tissues of 14 patients with WHO grade III glioma were collected for full transcriptome analysis. The correlation between image characteristics of LGG and survival related vascular ceRNA regulatory genes was compared using nonparametric U test and Pearson correlation coefficient analysis. RESULTS Vascular related genes ranked first in the functional enrichment analysis of differentially expressed genes in LGG. EPHA2, ETS1, YAP1 and MEIS1 could significantly affect the survival of patients in each group of LGG. The volume of enhanced region was negatively correlated with IDH1 (r = -0.622, P = 0.009) mutation and TMEM100 (r = -0.535, P = 0.024), and positively correlated with MEIS1 (r = 0.551, P = 0.021), rCBFmax value was negatively correlated with TMEM100 (r = -0.492, P = 0.037). CONCLUSIONS Under different IDH1 mutations, lncRNA-dominated vascular-related ceRNA regulatory genes were the first differentially expressed subset of each group, and could be used as an effective risk factor affecting the survival of LGG. The image characteristics of LGG was an ideal image gene marker. It was a reliable imaging biological marker which can truly reflect the pathophysiological characteristics of glioma.
Collapse
Affiliation(s)
- Yu Guo
- Department of Radiology, Army Medical Center of PLA, Army Medical University, 10# Changjiangzhilu, Yuzhong District, 400024, Chongqing, China
- Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Hong Guo
- Department of Radiology, Army Medical Center of PLA, Army Medical University, 10# Changjiangzhilu, Yuzhong District, 400024, Chongqing, China
- Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Haiyan Tong
- Zhoukou Central Hospital, Zhoukou, Henan, China
| | - Wei Xue
- Department of Radiology, The 940Th Hospital of Logistics Support Force of PLA, Lanzhou, China
| | - Tian Xie
- Department of Radiology, Army Medical Center of PLA, Army Medical University, 10# Changjiangzhilu, Yuzhong District, 400024, Chongqing, China
- Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Lulu Wang
- Chongqing University Cancer Hospital, Chongqing Cancer Hospital, Chongqing, China.
| | - Haipeng Tong
- Department of Radiology, Army Medical Center of PLA, Army Medical University, 10# Changjiangzhilu, Yuzhong District, 400024, Chongqing, China.
- Department of Radiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Zhang P, Lai X, Zhu MH, Shi J, Pan H, Huang Y, Guo RJ, Lu Q, Fang C, Zhao M. Jujuboside B suppresses angiogenesis and tumor growth via blocking VEGFR2 signaling pathway. Heliyon 2023; 9:e17072. [PMID: 37484305 PMCID: PMC10361242 DOI: 10.1016/j.heliyon.2023.e17072] [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: 09/30/2022] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 07/25/2023] Open
Abstract
Jujuboside B (JuB), one of the main active triterpenoid saponins from the traditional Chinese medicine Ziziphus jujuba, possesses a wide range of pharmacological activities. However, it is unknown whether JuB can inhibit tumor angiogenesis, a crucial step in solid tumor growth. In this study, we found that JuB significantly inhibited the proliferation, migration, and tube formation of human umbilical vein endothelial cells in a dose-dependent manner. JuB also suppressed angiogenesis in chick embryo chorioallantoic membranes and Matrigel plugs. Moreover, through angiogenesis inhibition, JuB delayed the growth of human HCT-15 colorectal cancer xenograft in mice. Western blot assay demonstrated that JuB inhibited the phosphorylation of VEGFR2 and its key downstream protein kinases, such as Akt, FAK, Src, and PLCγ1. In conclusion, the antiangiogenic potency and molecular mechanism of JuB are revealed for the first time, indicating that this triterpene saponin may be further explored as a potential drug candidate or lead compound for antiangiogenic cancer therapy.
Collapse
Affiliation(s)
- Pan Zhang
- Department of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Xing Lai
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Mao-Hua Zhu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Jiangpei Shi
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Hong Pan
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
- Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563003, China
| | - Yanhu Huang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Run-Jie Guo
- Department of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Qin Lu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Chao Fang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
- Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563003, China
| | - Mei Zhao
- Department of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| |
Collapse
|
3
|
Lu L, Wang L, Zhao L, Liao J, Zhao C, Xu X, Wang F, Zhang X. A Novel Blood-Brain Barrier-Penetrating and Vascular-Targeting Chimeric Peptide Inhibits Glioma Angiogenesis. Int J Mol Sci 2023; 24:ijms24108753. [PMID: 37240099 DOI: 10.3390/ijms24108753] [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: 03/31/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
The high vascularization of glioma highlights the potential value of anti-angiogenic therapeutics for glioma treatment. Previously, we designed a novel vascular-targeting and blood-brain barrier (BBB)-penetrating peptide, TAT-AT7, by attaching the cell-penetrating peptide TAT to a vascular-targeting peptide AT7, and we demonstrated that TAT-AT7 could target binding to the vascular endothelial growth factor receptor 2 (VEGFR-2) and Neuropilin-1 (NRP-1), which are both highly expressed in endothelial cells. TAT-AT7 has been proven to be a good targeting peptide which could effectively deliver the secretory endostatin gene to treat glioma via the TAT-AT7-modified polyethyleneimine (PEI) nanocomplex. In the current study, we further explored the molecular binding mechanisms of TAT-AT7 to VEGFR-2 and NRP-1 and its anti-glioma effects. Accordingly, TAT-AT7 was proven to competitively bind to VEGFR-2 and NRP-1 and prevent VEGF-A165 binding to the receptors by the surface plasmon resonance (SPR) assay. TAT-AT7 inhibited endothelial cells' proliferation, migration, invasion, and tubule formation, as well as promoted endothelial cells' apoptosis in vitro. Further research revealed that TAT-AT7 inhibited the phosphorylation of VEGFR-2 and its downstream PLC-γ, ERK1/2, SRC, AKT, and FAK kinases. Additionally, TAT-AT7 significantly inhibited angiogenesis of zebrafish embryo. Moreover, TAT-AT7 had a better penetrating ability and could penetrate the BBB into glioma tissue and target glioma neovascularization in an orthotopic U87-glioma-bearing nude mice model, and exhibited the effect of inhibiting glioma growth and angiogenesis. Taken together, the binding and function mechanisms of TAT-AT7 were firstly revealed, and TAT-AT7 was proven to be an effective and promising peptide for the development of anti-angiogenic drugs for targeted treatment of glioma.
Collapse
Affiliation(s)
- Lu Lu
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Longkun Wang
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lin Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jing Liao
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Chunqian Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiaohan Xu
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xinke Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
4
|
Cheng Y, Shi D, Ye R, Fu W, Ma P, Si Z, Xu Z, Li L, Lin Q, Cheng D. Noninvasive evaluation of PD-L1 expression in non-small cell lung cancer by immunoPET imaging using an acylating agent-modified antibody fragment. Eur J Nucl Med Mol Imaging 2023; 50:1585-1596. [PMID: 36759371 DOI: 10.1007/s00259-023-06130-6] [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: 12/23/2022] [Accepted: 01/26/2023] [Indexed: 02/11/2023]
Abstract
PURPOSE The aim of this study was to explore an effective 124I labeling strategy and improve the signal-to-noise ratio when evaluating the expression of PD-L1 using an 124I-iodinated durvalumab (durva) F(ab')2 fragment. METHODS The prepared durva F(ab')2 fragments were incubated with N-succinimidyl-3-(4-hydroxyphenyl) propionate (SHPP); after purification, the HPP-durva F(ab')2 was iodinated using Iodo-Gen method. After the radiochemical purity, stability, and specific activities were determined, the binding affinities of probes prepared using different labeling strategies were compared in vitro. The clinical application value of [124I]I-HPP-durva-F(ab')2 was confirmed by PET imaging. To more objectively evaluate the in vivo distribution and clearance of tracers, the pharmacokinetics and biodistribution assays were also performed. RESULTS After being modified with SHPP, the average conjugation number of SHPP per durva-F(ab')2 identified by LC-MS was about 8.92 ± 2.84. The prepared [124I]I-HPP-durva F(ab')2 was obtained with a satisfactory radiochemical purity of more than 98% and stability of more than 93% when incubated for 72 h. Compared with unmodified [124I]I-durva F(ab')2, the specific activity of [124I]I-HPP-durva-F(ab')2 was improved (52.91 ± 5.55 MBq/mg and 15.91 ± 0.74 MBq/mg), while the affinity did not significantly change. The biodistribution experiments and PET imaging showed that the prepared [124I]I-HPP-durva-F(ab')2 exhibited an accelerated clearance and improved tumor-to-background ratio compared with [124I]I-durva-F(ab')2. The specificity of [124I]I-HPP-durva-F(ab')2 to PD-L1 was well demonstrated both in vitro and in vivo. CONCLUSIONS A PD-L1 PET imaging probe [124I]I-HPP-durva F(ab')2 was successfully synthesized through the SHPP modification strategy. The prepared probe was able to accurately evaluate the PD-L1 expression level through high-contrast noninvasive imaging.
Collapse
Affiliation(s)
- Yuan Cheng
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Dai Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Renjie Ye
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Wenhui Fu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Pengcheng Ma
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Zhan Si
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Zhan Xu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Lixin Li
- Department of Hepatic Oncology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Qingyu Lin
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China. .,Institute of Nuclear Medicine, Fudan University, Shanghai, 200032, China. .,Shanghai Institute of Medical Imaging, Shanghai, 200032, China.
| | - Dengfeng Cheng
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China. .,Institute of Nuclear Medicine, Fudan University, Shanghai, 200032, China. .,Shanghai Institute of Medical Imaging, Shanghai, 200032, China.
| |
Collapse
|
5
|
Liu F, Peng B, Li M, Ma J, Deng G, Zhang S, Sheu WC, Zou P, Wu H, Liu J, Chen AT, Mohammed FS, Zhou J. Targeted disruption of tumor vasculature via polyphenol nanoparticles to improve brain cancer treatment. CELL REPORTS. PHYSICAL SCIENCE 2022; 3:100691. [PMID: 35199059 PMCID: PMC8863382 DOI: 10.1016/j.xcrp.2021.100691] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Despite being effective for many other solid tumors, traditional anti-angiogenic therapy has been shown to be insufficient for the treatment of malignant glioma. Here, we report the development of polyphenol nanoparticles (NPs), which not only inhibit the formation of new vessels but also enable targeted disruption of the existing tumor vasculature. The NPs are synthesized through a combinatory iron-coordination and polymer-stabilization approach, which allows for high drug loading and intrinsic tumor vessel targeting. We study a lead NP consisting of quercetin and find that the NP after intravenous administration preferentially binds to VEGFR2, which is overexpressed in tumor vasculature. We demonstrate that the binding is mediated by quercetin, and the interaction of NPs with VEGFR2 leads to disruption of the existing tumor vasculature and inhibition of new vessel development. As a result, systemic treatment with the NPs effectively inhibits tumor growth and increases drug delivery to tumors.
Collapse
Affiliation(s)
- Fuyao Liu
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Bin Peng
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Miao Li
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Junning Ma
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Gang Deng
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Shenqi Zhang
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Wendy C. Sheu
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Pan Zou
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Haoan Wu
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Jun Liu
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Ann T. Chen
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Farrah S. Mohammed
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Jiangbing Zhou
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
- Lead contact
- Correspondence:
| |
Collapse
|
6
|
Wu WY, Späth F, Wibom C, Björkblom B, Dahlin AM, Melin B. Pre‐diagnostic levels of sVEGFR2, sTNFR2, sIL‐2Rα and sIL‐6R are associated with glioma risk: A nested case–control study of repeated samples. Cancer Med 2022; 11:1016-1025. [PMID: 35029050 PMCID: PMC8855896 DOI: 10.1002/cam4.4505] [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: 05/27/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/01/2022] Open
Abstract
No strong aetiological factors have been established for glioma aside from genetic mutations and variants, ionising radiation and an inverse relationship with asthmas and allergies. Our aim was to investigate the association between pre‐diagnostic immune protein levels and glioma risk. We conducted a case–control study nested in the Northern Sweden Health and Disease Study cohort. We analysed 133 glioma cases and 133 control subjects matched by age, sex and date of blood donation. ELISA or Luminex bead‐based multiplex assays were used to measure plasma levels of 19 proteins. Conditional logistic regression models were used to estimate the odds ratios and 95% CIs. To further model the protein trajectories over time, the linear mixed‐effects models were conducted. We found that the levels of sVEGFR2, sTNFR2, sIL‐2Rα and sIL‐6R were associated with glioma risk. After adjusting for the time between blood sample collection and glioma diagnosis, the odds ratios were 1.72 (95% CI = 1.01–2.93), 1.48 (95% CI = 1.01–2.16) and 1.90 (95% CI = 1.14–3.17) for sTNFR2, sIL‐2Rα and sIL‐6R, respectively. The trajectory of sVEGFR2 concentrations over time was different between cases and controls (p‐value = 0.031), increasing for cases (0.8% per year) and constant for controls. Our findings suggest these proteins play important roles in gliomagenesis.
Collapse
Affiliation(s)
- Wendy Yi‐Ying Wu
- Department of Radiation Sciences, Oncology Umeå University Umeå Sweden
| | - Florentin Späth
- Department of Radiation Sciences, Oncology Umeå University Umeå Sweden
| | - Carl Wibom
- Department of Radiation Sciences, Oncology Umeå University Umeå Sweden
| | | | - Anna M. Dahlin
- Department of Radiation Sciences, Oncology Umeå University Umeå Sweden
| | - Beatrice Melin
- Department of Radiation Sciences, Oncology Umeå University Umeå Sweden
| |
Collapse
|
7
|
Su H, Zhao L, Yu B, Zeng H, Yang J, Zhu M, Zhao J. Preparation and bioevaluation of [ 99mTc]Tc-labeled A7R and DA7R for SPECT imaging of triple-negative breast cancer. NEW J CHEM 2022. [DOI: 10.1039/d2nj04136g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[99mTc]Tc-labeled D-type A7R peptide showed better tumor-to-muscle ratios and lower renal uptake.
Collapse
Affiliation(s)
- Hongxing Su
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Buhui Yu
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Huahui Zeng
- Academy of Chinese Medicine Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, P. R. China
| | - Jiqin Yang
- Department of Nuclear Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia, P. R. China
| | - Meilin Zhu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, Ningxia, P. R. China
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| |
Collapse
|
8
|
Tolmachev VM, Chernov VI, Deyev SM. Targeted nuclear medicine. Seek and destroy. RUSSIAN CHEMICAL REVIEWS 2022. [DOI: 10.1070/rcr5034] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
9
|
Liu F, Wu H, Peng B, Zhang S, Ma J, Deng G, Zou P, Liu J, Chen AT, Li D, Bellone S, Santin AD, Moliterno J, Zhou J. Vessel-Targeting Nanoclovers Enable Noninvasive Delivery of Magnetic Hyperthermia-Chemotherapy Combination for Brain Cancer Treatment. NANO LETTERS 2021; 21:8111-8118. [PMID: 34597054 DOI: 10.1021/acs.nanolett.1c02459] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Despite being promising, the clinical application of magnetic hyperthermia for brain cancer treatment is limited by the requirement of highly invasive intracranial injections. To overcome this limitation, here we report the development of gallic acid-coated magnetic nanoclovers (GA-MNCs), which allow not only for noninvasive delivery of magnetic hyperthermia but also for targeted delivery of systemic chemotherapy to brain tumors. GA-MNCs are composed of clover-shaped MNCs in the core, which can induce magnetic heat in high efficiency, and polymerized GA on the shell, which enables tumor vessel-targeting. We demonstrate that intravenous administration of GA-MNCs following alternating magnetic field exposure effectively inhibited brain cancer development and preferentially disrupted tumor vasculature, making it possible to efficiently deliver systemic chemotherapy for further improved efficacy. Due to the noninvasive nature and high efficiency in killing tumor cells and enhancing systemic drug delivery, GA-MNCs have the potential to be translated for improved treatment of brain cancer.
Collapse
Affiliation(s)
- Fuyao Liu
- Department of Neurosurgery, Yale University, New Haven, Connecticut 06510, United States
| | - Haoan Wu
- Department of Neurosurgery, Yale University, New Haven, Connecticut 06510, United States
| | - Bin Peng
- Department of Neurosurgery, Yale University, New Haven, Connecticut 06510, United States
| | - Shenqi Zhang
- Department of Neurosurgery, Yale University, New Haven, Connecticut 06510, United States
| | - Junning Ma
- Department of Neurosurgery, Yale University, New Haven, Connecticut 06510, United States
| | - Gang Deng
- Department of Neurosurgery, Yale University, New Haven, Connecticut 06510, United States
| | - Pan Zou
- Department of Neurosurgery, Yale University, New Haven, Connecticut 06510, United States
| | - Jun Liu
- Department of Neurosurgery, Yale University, New Haven, Connecticut 06510, United States
| | - Ann T Chen
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06510, United States
| | - Dongfang Li
- Department of Neurosurgery, Yale University, New Haven, Connecticut 06510, United States
| | - Stefania Bellone
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, Connecticut 06510, United States
| | - Alessandro Davide Santin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, Connecticut 06510, United States
| | - Jennifer Moliterno
- Department of Neurosurgery, Yale University, New Haven, Connecticut 06510, United States
| | - Jiangbing Zhou
- Department of Neurosurgery, Yale University, New Haven, Connecticut 06510, United States
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06510, United States
| |
Collapse
|
10
|
Zaccagna F, Grist JT, Quartuccio N, Riemer F, Fraioli F, Caracò C, Halsey R, Aldalilah Y, Cunningham CH, Massoud TF, Aloj L, Gallagher FA. Imaging and treatment of brain tumors through molecular targeting: Recent clinical advances. Eur J Radiol 2021; 142:109842. [PMID: 34274843 DOI: 10.1016/j.ejrad.2021.109842] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
Molecular imaging techniques have rapidly progressed over recent decades providing unprecedented in vivo characterization of metabolic pathways and molecular biomarkers. Many of these new techniques have been successfully applied in the field of neuro-oncological imaging to probe tumor biology. Targeting specific signaling or metabolic pathways could help to address several unmet clinical needs that hamper the management of patients with brain tumors. This review aims to provide an overview of the recent advances in brain tumor imaging using molecular targeting with positron emission tomography and magnetic resonance imaging, as well as the role in patient management and possible therapeutic implications.
Collapse
Affiliation(s)
- Fulvio Zaccagna
- Division of Neuroimaging, Department of Medical Imaging, University of Toronto, Toronto, Canada.
| | - James T Grist
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom; Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom; Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom; Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Natale Quartuccio
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico Di Cristina Benfratelli, Palermo, Italy
| | - Frank Riemer
- Mohn Medical Imaging and Visualization Centre, University of Bergen, Bergen, Norway; Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Francesco Fraioli
- Institute of Nuclear Medicine, University College London, London, United Kingdom; NIHR University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Corradina Caracò
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Richard Halsey
- Institute of Nuclear Medicine, University College London, London, United Kingdom; NIHR University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Yazeed Aldalilah
- Institute of Nuclear Medicine, University College London, London, United Kingdom; NIHR University College London Hospitals Biomedical Research Centre, London, United Kingdom; Department of Radiology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Charles H Cunningham
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Tarik F Massoud
- Division of Neuroimaging and Neurointervention, Department of Radiology, Stanford University School of Medicine, Stanford, USA
| | - Luigi Aloj
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Ferdia A Gallagher
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
11
|
Bolcaen J, Kleynhans J, Nair S, Verhoeven J, Goethals I, Sathekge M, Vandevoorde C, Ebenhan T. A perspective on the radiopharmaceutical requirements for imaging and therapy of glioblastoma. Theranostics 2021; 11:7911-7947. [PMID: 34335972 PMCID: PMC8315062 DOI: 10.7150/thno.56639] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/29/2021] [Indexed: 11/26/2022] Open
Abstract
Despite numerous clinical trials and pre-clinical developments, the treatment of glioblastoma (GB) remains a challenge. The current survival rate of GB averages one year, even with an optimal standard of care. However, the future promises efficient patient-tailored treatments, including targeted radionuclide therapy (TRT). Advances in radiopharmaceutical development have unlocked the possibility to assess disease at the molecular level allowing individual diagnosis. This leads to the possibility of choosing a tailored, targeted approach for therapeutic modalities. Therapeutic modalities based on radiopharmaceuticals are an exciting development with great potential to promote a personalised approach to medicine. However, an effective targeted radionuclide therapy (TRT) for the treatment of GB entails caveats and requisites. This review provides an overview of existing nuclear imaging and TRT strategies for GB. A critical discussion of the optimal characteristics for new GB targeting therapeutic radiopharmaceuticals and clinical indications are provided. Considerations for target selection are discussed, i.e. specific presence of the target, expression level and pharmacological access to the target, with particular attention to blood-brain barrier crossing. An overview of the most promising radionuclides is given along with a validation of the relevant radiopharmaceuticals and theranostic agents (based on small molecules, peptides and monoclonal antibodies). Moreover, toxicity issues and safety pharmacology aspects will be presented, both in general and for the brain in particular.
Collapse
Affiliation(s)
- Julie Bolcaen
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| | - Janke Kleynhans
- Nuclear Medicine Research Infrastructure NPC, Pretoria, South Africa
- Nuclear Medicine Department, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
| | - Shankari Nair
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| | | | - Ingeborg Goethals
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium
| | - Mike Sathekge
- Nuclear Medicine Research Infrastructure NPC, Pretoria, South Africa
- Nuclear Medicine Department, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
| | - Charlot Vandevoorde
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| | - Thomas Ebenhan
- Nuclear Medicine Research Infrastructure NPC, Pretoria, South Africa
- Nuclear Medicine Department, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
12
|
Bolcaen J, Nair S, Driver CHS, Boshomane TMG, Ebenhan T, Vandevoorde C. Novel Receptor Tyrosine Kinase Pathway Inhibitors for Targeted Radionuclide Therapy of Glioblastoma. Pharmaceuticals (Basel) 2021; 14:626. [PMID: 34209513 PMCID: PMC8308832 DOI: 10.3390/ph14070626] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GB) remains the most fatal brain tumor characterized by a high infiltration rate and treatment resistance. Overexpression and/or mutation of receptor tyrosine kinases is common in GB, which subsequently leads to the activation of many downstream pathways that have a critical impact on tumor progression and therapy resistance. Therefore, receptor tyrosine kinase inhibitors (RTKIs) have been investigated to improve the dismal prognosis of GB in an effort to evolve into a personalized targeted therapy strategy with a better treatment outcome. Numerous RTKIs have been approved in the clinic and several radiopharmaceuticals are part of (pre)clinical trials as a non-invasive method to identify patients who could benefit from RTKI. The latter opens up the scope for theranostic applications. In this review, the present status of RTKIs for the treatment, nuclear imaging and targeted radionuclide therapy of GB is presented. The focus will be on seven tyrosine kinase receptors, based on their central role in GB: EGFR, VEGFR, MET, PDGFR, FGFR, Eph receptor and IGF1R. Finally, by way of analyzing structural and physiological characteristics of the TKIs with promising clinical trial results, four small molecule RTKIs were selected based on their potential to become new therapeutic GB radiopharmaceuticals.
Collapse
Affiliation(s)
- Julie Bolcaen
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| | - Shankari Nair
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| | - Cathryn H. S. Driver
- Radiochemistry, South African Nuclear Energy Corporation, Pelindaba, Brits 0240, South Africa;
- Pre-Clinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pelindaba, Brits 0242, South Africa;
| | - Tebatso M. G. Boshomane
- Department of Nuclear Medicine, University of Pretoria Steve Biko Academic Hospital, Pretoria 0001, South Africa;
| | - Thomas Ebenhan
- Pre-Clinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pelindaba, Brits 0242, South Africa;
- Department of Nuclear Medicine, University of Pretoria Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Preclinical Drug Development Platform, Department of Science and Technology, North West University, Potchefstroom 2520, South Africa
| | - Charlot Vandevoorde
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| |
Collapse
|
13
|
The Role of VEGF Receptors as Molecular Target in Nuclear Medicine for Cancer Diagnosis and Combination Therapy. Cancers (Basel) 2021; 13:cancers13051072. [PMID: 33802353 PMCID: PMC7959315 DOI: 10.3390/cancers13051072] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/13/2021] [Accepted: 02/24/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary The rapid development of diagnostic and therapeutic methods of the cancer treatment causes that these diseases are becoming better known and the fight against them is more and more effective. Substantial contribution in this development has nuclear medicine that enables very early cancer diagnosis and early start of the so-called targeted therapy. This therapeutic concept compared to the currently used chemotherapy, causes much fewer undesirable side effects, due to targeting a specific lesion in the body. This review article discusses the possible applications of radionuclide-labelled tracers (peptides, antibodies or synthetic organic molecules) that can visualise cancer cells through pathological blood vessel system in close tumour microenvironment. Hence, at a very early step of oncological disease, targeted therapy can involve in tumour formation and growth. Abstract One approach to anticancer treatment is targeted anti-angiogenic therapy (AAT) based on prevention of blood vessel formation around the developing cancer cells. It is known that vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptors (VEGFRs) play a pivotal role in angiogenesis process; hence, application of angiogenesis inhibitors can be an effective approach in anticancer combination therapeutic strategies. Currently, several types of molecules have been utilised in targeted VEGF/VEGFR anticancer therapy, including human VEGF ligands themselves and their derivatives, anti-VEGF or anti-VEGFR monoclonal antibodies, VEGF binding peptides and small molecular inhibitors of VEGFR tyrosine kinases. These molecules labelled with diagnostic or therapeutic radionuclides can become, respectively, diagnostic or therapeutic receptor radiopharmaceuticals. In targeted anti-angiogenic therapy, diagnostic radioagents play a unique role, allowing the determination of the emerging tumour, to monitor the course of treatment, to predict the treatment outcomes and, first of all, to refer patients for AAT. This review provides an overview of design, synthesis and study of radiolabelled VEGF/VEGFR targeting and imaging agents to date. Additionally, we will briefly discuss their physicochemical properties and possible application in combination targeted radionuclide tumour therapy.
Collapse
|
14
|
Notohamiprodjo S, Varasteh Z, Beer AJ, Niu G, Chen X(S, Weber W, Schwaiger M. Tumor Vasculature. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00090-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
15
|
Güler R, Svedmark SF, Abouzayed A, Orlova A, Löfblom J. Increasing thermal stability and improving biodistribution of VEGFR2-binding affibody molecules by a combination of in silico and directed evolution approaches. Sci Rep 2020; 10:18148. [PMID: 33097752 PMCID: PMC7585445 DOI: 10.1038/s41598-020-74560-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/23/2020] [Indexed: 11/09/2022] Open
Abstract
The family of vascular endothelial growth factor (VEGF) ligands and their interactions with VEGF receptors (VEGFRs) play important roles in both pathological and physiological angiogenesis. Hence, agonistic and antagonistic ligands targeting this signaling pathway have potential for both studies on fundamental biology and for development of therapies and diagnostics. Here, we engineer VEGFR2-binding affibody molecules for increased thermostability, refolding and improved biodistribution. We designed libraries based on the original monomeric binders with the intention of reducing hydrophobicity, while retaining high affinity for VEGFR2. Libraries were displayed on bacteria and binders were isolated by fluorescence-activated cell sorting (FACS). In parallel, we used an automated sequence- and structure-based in silico algorithm to identify potentially stabilizing mutations. Monomeric variants isolated from the screening and the in silico approach, respectively, were characterized by circular dichroism spectroscopy and biosensor assays. The most promising mutations were combined into new monomeric constructs which were finally fused into a dimeric construct, resulting in a 15 °C increase in melting temperature, complete refolding capability after heat-induced denaturation, retained low picomolar affinity and improved biodistribution profile in an in vivo mouse model. These VEGFR2-binding affibody molecules show promise as candidates for further in vivo studies to assess their suitability as molecular imaging and therapeutic agents.
Collapse
Affiliation(s)
- Rezan Güler
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Siri Flemming Svedmark
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Ayman Abouzayed
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
16
|
Ingram N, McVeigh LE, Abou-Saleh RH, Maynard J, Peyman SA, McLaughlan JR, Fairclough M, Marston G, Valleley EMA, Jimenez-Macias JL, Charalambous A, Townley W, Haddrick M, Wierzbicki A, Wright A, Volpato M, Simpson PB, Treanor DE, Thomson NH, Loadman PM, Bushby RJ, Johnson BR, Jones PF, Evans JA, Freear S, Markham AF, Evans SD, Coletta PL. Ultrasound-triggered therapeutic microbubbles enhance the efficacy of cytotoxic drugs by increasing circulation and tumor drug accumulation and limiting bioavailability and toxicity in normal tissues. Theranostics 2020; 10:10973-10992. [PMID: 33042265 PMCID: PMC7532679 DOI: 10.7150/thno.49670] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Most cancer patients receive chemotherapy at some stage of their treatment which makes improving the efficacy of cytotoxic drugs an ongoing and important goal. Despite large numbers of potent anti-cancer agents being developed, a major obstacle to clinical translation remains the inability to deliver therapeutic doses to a tumor without causing intolerable side effects. To address this problem, there has been intense interest in nanoformulations and targeted delivery to improve cancer outcomes. The aim of this work was to demonstrate how vascular endothelial growth factor receptor 2 (VEGFR2)-targeted, ultrasound-triggered delivery with therapeutic microbubbles (thMBs) could improve the therapeutic range of cytotoxic drugs. Methods: Using a microfluidic microbubble production platform, we generated thMBs comprising VEGFR2-targeted microbubbles with attached liposomal payloads for localised ultrasound-triggered delivery of irinotecan and SN38 in mouse models of colorectal cancer. Intravenous injection into tumor-bearing mice was used to examine targeting efficiency and tumor pharmacodynamics. High-frequency ultrasound and bioluminescent imaging were used to visualise microbubbles in real-time. Tandem mass spectrometry (LC-MS/MS) was used to quantitate intratumoral drug delivery and tissue biodistribution. Finally, 89Zr PET radiotracing was used to compare biodistribution and tumor accumulation of ultrasound-triggered SN38 thMBs with VEGFR2-targeted SN38 liposomes alone. Results: ThMBs specifically bound VEGFR2 in vitro and significantly improved tumor responses to low dose irinotecan and SN38 in human colorectal cancer xenografts. An ultrasound trigger was essential to achieve the selective effects of thMBs as without it, thMBs failed to extend intratumoral drug delivery or demonstrate enhanced tumor responses. Sensitive LC-MS/MS quantification of drugs and their metabolites demonstrated that thMBs extended drug exposure in tumors but limited exposure in healthy tissues, not exposed to ultrasound, by persistent encapsulation of drug prior to elimination. 89Zr PET radiotracing showed that the percentage injected dose in tumors achieved with thMBs was twice that of VEGFR2-targeted SN38 liposomes alone. Conclusions: thMBs provide a generic platform for the targeted, ultrasound-triggered delivery of cytotoxic drugs by enhancing tumor responses to low dose drug delivery via combined effects on circulation, tumor drug accumulation and exposure and altered metabolism in normal tissues.
Collapse
Affiliation(s)
- Nicola Ingram
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Laura E. McVeigh
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Radwa H. Abou-Saleh
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
- Department of Physics, Faculty of Science, Mansoura University, Egypt
| | - Juliana Maynard
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Sally A. Peyman
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
| | - James R. McLaughlan
- Faculty of Electronic and Electrical Engineering, University of Leeds, LS2 9JT, United Kingdom
| | - Michael Fairclough
- Wolfson Molecular Imaging Centre, University of Manchester, Palatine Road, Manchester, M20 3LI, United Kingdom
| | - Gemma Marston
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Elizabeth M. A. Valleley
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Jorge L. Jimenez-Macias
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Antonia Charalambous
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - William Townley
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Malcolm Haddrick
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Antonia Wierzbicki
- Institute of Cancer Therapeutics, University of Bradford, BD7 1DP, United Kingdom
| | - Alexander Wright
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Milène Volpato
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Peter B. Simpson
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Darren E. Treanor
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Neil H. Thomson
- School of Dentistry, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Paul M. Loadman
- Institute of Cancer Therapeutics, University of Bradford, BD7 1DP, United Kingdom
| | - Richard J. Bushby
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Benjamin R.G. Johnson
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
| | - Pamela F. Jones
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - J. Anthony Evans
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Steven Freear
- Faculty of Electronic and Electrical Engineering, University of Leeds, LS2 9JT, United Kingdom
| | - Alexander F. Markham
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Stephen D. Evans
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
| | - P. Louise Coletta
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| |
Collapse
|
17
|
Imaging using radiolabelled targeted proteins: radioimmunodetection and beyond. EJNMMI Radiopharm Chem 2020; 5:16. [PMID: 32577943 PMCID: PMC7311618 DOI: 10.1186/s41181-020-00094-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 04/14/2020] [Indexed: 12/18/2022] Open
Abstract
The use of radiolabelled antibodies was proposed in 1970s for staging of malignant tumours. Intensive research established chemistry for radiolabelling of proteins and understanding of factors determining biodistribution and targeting properties. The use of radioimmunodetection for staging of cancer was not established as common practice due to approval and widespread use of [18F]-FDG, which provided a more general diagnostic use than antibodies or their fragments. Expanded application of antibody-based therapeutics renewed the interest in radiolabelled antibodies. RadioimmunoPET emerged as a powerful tool for evaluation of pharmacokinetics of and target engagement by biotherapeutics. In addition to monoclonal antibodies, new radiolabelled engineered proteins have recently appeared, offering high-contrast imaging of expression of therapeutic molecular targets in tumours shortly after injection. This creates preconditions for noninvasive determination of a target expression level and stratification of patients for targeted therapies. Radiolabelled proteins hold great promise to play an important role in development and implementation of personalised targeted treatment of malignant tumours. This article provides an overview of biodistribution and tumour-seeking features of major classes of targeting proteins currently utilized for molecular imaging. Such information might be useful for researchers entering the field of the protein-based radionuclide molecular imaging.
Collapse
|
18
|
Hu K, Shang J, Xie L, Hanyu M, Zhang Y, Yang Z, Xu H, Wang L, Zhang MR. PET Imaging of VEGFR with a Novel 64Cu-Labeled Peptide. ACS OMEGA 2020; 5:8508-8514. [PMID: 32337411 PMCID: PMC7178340 DOI: 10.1021/acsomega.9b03953] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/12/2020] [Indexed: 05/11/2023]
Abstract
Vascular endothelial growth factor receptors (VEGFRs) are well recognized as significant biomarkers of tumor angiogenesis. Herein, we have developed a first-of-its-kind peptide-based VEGFR positron emission tomography (PET) tracer. The novel [64Cu]VEGF125-136 peptide possessed satisfactory radio-characteristics and showed good specificity for the visualization of VEGFR in various mouse models, in which the tumor-specific radioactivity uptake was highly correlated to the VEGFR expression level. Moreover, the tracer showed high tumor uptake (ca. 5.89 %ID/g at 20 min postinjection in B16F10 mice) and excellent pharmacokinetics, achieving the maximum imaging quality within 1 h after injection. These features convey [64Cu]VEGF125-136 as a promising, clinically translatable PET tracer for the imaging of tumor angiogenesis.
Collapse
Affiliation(s)
- Kuan Hu
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Jingjie Shang
- Center
of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine
and PET/CT-MRI Center, The First Affiliated
Hospital of Jinan University, Guangzhou 510630, China
| | - Lin Xie
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Masayuki Hanyu
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Yiding Zhang
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Zhimin Yang
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
and Radiological Science and Technology, Chiba 263-8555, Japan
- Center
of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine
and PET/CT-MRI Center, The First Affiliated
Hospital of Jinan University, Guangzhou 510630, China
| | - Hao Xu
- Center
of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine
and PET/CT-MRI Center, The First Affiliated
Hospital of Jinan University, Guangzhou 510630, China
| | - Lu Wang
- Center
of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine
and PET/CT-MRI Center, The First Affiliated
Hospital of Jinan University, Guangzhou 510630, China
| | - Ming-Rong Zhang
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
and Radiological Science and Technology, Chiba 263-8555, Japan
| |
Collapse
|
19
|
Affibody Molecules as Targeting Vectors for PET Imaging. Cancers (Basel) 2020; 12:cancers12030651. [PMID: 32168760 PMCID: PMC7139392 DOI: 10.3390/cancers12030651] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022] Open
Abstract
Affibody molecules are small (58 amino acids) engineered scaffold proteins that can be selected to bind to a large variety of proteins with a high affinity. Their small size and high affinity make them attractive as targeting vectors for molecular imaging. High-affinity affibody binders have been selected for several cancer-associated molecular targets. Preclinical studies have shown that radiolabeled affibody molecules can provide highly specific and sensitive imaging on the day of injection; however, for a few targets, imaging on the next day further increased the imaging sensitivity. A phase I/II clinical trial showed that 68Ga-labeled affibody molecules permit an accurate and specific measurement of HER2 expression in breast cancer metastases. This paper provides an overview of the factors influencing the biodistribution and targeting properties of affibody molecules and the chemistry of their labeling using positron emitters.
Collapse
|
20
|
Wang X, Liu G, Chen N, Wu J, Zhang J, Qian Y, Zhang L, Zhou D, Yu Y. Angiopep2-Conjugated Star-Shaped Polyprodrug Amphiphiles for Simultaneous Glioma-Targeting Therapy and MR Imaging. ACS APPLIED MATERIALS & INTERFACES 2020; 12:12143-12154. [PMID: 32078286 DOI: 10.1021/acsami.0c00509] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The development of valuable theranostic agents for overcoming the blood-brain barrier (BBB) to achieve efficient imaging-guided glioma-targeting delivery of therapeutics remains a great challenge for personalized glioma therapy. We herein developed a novel functional star-shaped polyprodrug amphiphile (denoted as CPP-2) via a combination of successive reversible addition-fragmentation chain transfer (RAFT) polymerization and click functionalization. In a diluted solution, the star amphiphile existed as structurally stable unimolecular micelles, containing hydrophobic cores conjugated with reduction-responsive camptothecin prodrugs Camptothecin (CPT) prodrug monomer (CPTM) and a tertiary amine monomer (2-(diethylamine) ethyl methacrylate, DEA) and hydrophilic oligo-(ethylene glycol) monomethyl ether methacrylat (OEGMA) outer coronas covalently decorated with dual-targeting moieties Angiopep2 (ANG) and small magnetic resonance imaging (MRI) contrast agents DOTA-Gd. In vitro and in vivo data in this study demonstrated that the ANG-modified micelles were capable of efficiently penetrating the BBB and delivering loaded cargoes such as CPT and Gd3+ contrast agents to glioma cells, leading to a considerably enhanced t1 relaxivity as well as antiglioma efficacy. Simultaneously, the targeted antiglioma efficacy and noninvasive MR imaging for a visualized therapy were realized. These collective findings augured well for the star polyprodrug amphiphiles to be utilized as a novel theranostic platform for clinical application in glioma therapy.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Guhuan Liu
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Ni Chen
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Jing Wu
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Jingjing Zhang
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Yinfeng Qian
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Lei Zhang
- Department of Pharmaceutics, School of Pharmacy, Anhui Medical University, Hefei 230032, P. R. China
| | - Dandan Zhou
- Department of Pharmaceutics, School of Pharmacy, Anhui Medical University, Hefei 230032, P. R. China
| | - Yongqiang Yu
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| |
Collapse
|
21
|
Güler R, Thatikonda N, Ghani HA, Hedhammar M, Löfblom J. VEGFR2-Specific Ligands Based on Affibody Molecules Demonstrate Agonistic Effects when Tetrameric in the Soluble Form or Immobilized via Spider Silk. ACS Biomater Sci Eng 2019; 5:6474-6484. [PMID: 33417800 DOI: 10.1021/acsbiomaterials.9b00994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Strategies to promote vascularization are being developed in order to improve long-term survival of artificial tissue constructs. Vascular endothelial growth factor A (VEGFA) has an important role in both pathological and physiological angiogenesis, mediated by binding to VEGF receptors (VEGFRs). In nature, signaling can be modulated by presentation of growth factors in either soluble form or bound to the extracellular matrix. Herein, a previously reported VEGFR2-binding antagonistic affibody heterodimer (di-ZVEGFR2) was formatted into a tetrameric construct (tetra-ZVEGFR2) with the intention of generating artificial agonistic ligands for VEGFR2 signaling. In vitro cell assays demonstrated that tetra-ZVEGFR2 induced VEGFR2 phosphorylation and increased cell proliferation, in contrast to di-ZVEGFR2. In order to simulate matrix-bound factors, both constructs were fused at the genetic level to a partial spider silk protein, 4RepCT. Assembly of the silk fusion proteins onto a solid surface was verified by quartz crystal microbalance with dissipation analysis. Moreover, surface plasmon resonance studies demonstrated retained VEGFR2 binding ability of di-ZVEGFR2-silk and tetra-ZVEGFR2-silk after silk-mediated immobilization. Cell culture studies demonstrated that VEGFR2-overexpressing cells adhered to di-ZVEGFR2-silk and tetra-ZVEGFR2-silk and had activated VEGFR2 signaling. Altogether, we demonstrate the potential of especially tetra-ZVEGFR2-silk to promote angiogenesis in tissue-engineering applications. The results from the study also show that molecules can obtain completely new functions when presented on materials, and verifying the biological effects after functionalizing materials is thus always recommended.
Collapse
Affiliation(s)
- Rezan Güler
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, SE-10691 Stockholm, Sweden
| | - Naresh Thatikonda
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, SE-10691 Stockholm, Sweden
| | - Hawraa Ali Ghani
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, SE-10691 Stockholm, Sweden
| | - My Hedhammar
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, SE-10691 Stockholm, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, SE-10691 Stockholm, Sweden
| |
Collapse
|
22
|
Puttemans J, Lahoutte T, D'Huyvetter M, Devoogdt N. Beyond the Barrier: Targeted Radionuclide Therapy in Brain Tumors and Metastases. Pharmaceutics 2019; 11:pharmaceutics11080376. [PMID: 31374991 PMCID: PMC6723032 DOI: 10.3390/pharmaceutics11080376] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/12/2019] [Accepted: 07/17/2019] [Indexed: 01/10/2023] Open
Abstract
Brain tumors are notoriously difficult to treat. The blood-brain barrier provides a sanctuary site where residual and metastatic cancer cells can evade most therapeutic modalities. The delicate nature of the brain further complicates the decision of eliminating as much tumorous tissue as possible while protecting healthy tissue. Despite recent advances in immunotherapy, radiotherapy and systemic treatments, prognosis of newly diagnosed patients remains dismal, and recurrence is still a universal problem. Several strategies are now under preclinical and clinical investigation to optimize delivery and maximize the cytotoxic potential of pharmaceuticals with regards to brain tumors. This review provides an overview of targeted radionuclide therapy approaches for the treatment of primary brain tumors and brain metastases, with an emphasis on biological targeting moieties that specifically target key biomarkers involved in cancer development.
Collapse
Affiliation(s)
- Janik Puttemans
- In Vivo Cellular and Molecular Imaging Lab, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Tony Lahoutte
- In Vivo Cellular and Molecular Imaging Lab, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Matthias D'Huyvetter
- In Vivo Cellular and Molecular Imaging Lab, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Lab, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| |
Collapse
|