1
|
Singh A, Patel A, Chaudhary H, Yadav K, Minocha N. Nanotheranostics: The Fabrication of Theranostics with Nanoparticles and their Application to Treat the Neurological Disorders. RECENT PATENTS ON NANOTECHNOLOGY 2025; 19:17-34. [PMID: 37464820 DOI: 10.2174/1872210517666230718115651] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 06/13/2023] [Accepted: 06/13/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND Theranostics is a method that focuses on providing patient-centred care and is evolving as a targeted, safe, and effective pharmacotherapy. Nanotheranostics combines diagnosis and therapeutic modalities that bridge traditional treatment and personalised medicine. Theranostics provides novel ideas for nanotechnology. This review describes the current state of nanotechnology-based therapies used to treat neurological illnesses. Some patents on theranostics are also discussed in this review. OBJECTIVE This study aims to provide a more comprehensive review of the diagnosis and therapeutic properties of nanotheranostics, the present state of nanotechnology-based treatment of neurological disorders, and the future potential of theranostics. METHODS The phrase "theranostics" refers to a treatment strategy that integrates therapeutics and diagnostics to monitor treatment response and enhance drug efficacy and safety. Theranostics is a crucial component of personalised medicine and calls for significant advancements in predictive medicine. The term "theranostics" refers to a diagnosis that screens patients for potential adverse drug reactions and targets drug delivery depending on the test results. Theranostics treats neurological disorders (like brain tumours (glioma), Parkinson's disease, Alzheimer's disease, and neurovascular diseases). Many review articles on Google Scholar, PubMed, Google Patents, and Scopus were used to gather information for this review. Data acquired from many sources was compiled in this review to provide more information on theranostics. RESULTS The role of various nanocarrier systems as theranostic agents for neurological illnesses and the fabrication of nanomaterials for theranostics are discussed in this article after evaluating a substantial number of review articles. CONCLUSION The distinctive intrinsic features of nanoparticles make them useful for functionalization and imaging. Theranostics in nuclear medicine include diagnostic imaging and therapy using the same molecule that is radiolabeled differently or the same medication at various doses. It is possible to determine if a patient will benefit from a given treatment by visualising potential targets. Targeted nuclear therapy has been shown to be beneficial in patients if chosen carefully and has a good safety profile.
Collapse
Affiliation(s)
- Astha Singh
- School of Medical and Allied Sciences, K. R. Mangalam University, Sohna Road, Gurugram, 122013, India
| | - Aakriti Patel
- School of Medical and Allied Sciences, K. R. Mangalam University, Sohna Road, Gurugram, 122013, India
| | - Hema Chaudhary
- School of Medical and Allied Sciences, K. R. Mangalam University, Sohna Road, Gurugram, 122013, India
| | - Kiran Yadav
- Department of Pharmaceutical Sciences, Chandigarh College of Pharmacy, CGC, Landran, Mohali, 140307, India
| | - Neha Minocha
- Amity Institute of Pharmacy, Amity University, Gurgaon, 122412, Haryana, India
| |
Collapse
|
2
|
Sun T, Zhao H, Hu L, Shao X, Lu Z, Wang Y, Ling P, Li Y, Zeng K, Chen Q. Enhanced optical imaging and fluorescent labeling for visualizing drug molecules within living organisms. Acta Pharm Sin B 2024; 14:2428-2446. [PMID: 38828150 PMCID: PMC11143489 DOI: 10.1016/j.apsb.2024.01.018] [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: 10/30/2023] [Revised: 01/07/2024] [Accepted: 01/25/2024] [Indexed: 06/05/2024] Open
Abstract
The visualization of drugs in living systems has become key techniques in modern therapeutics. Recent advancements in optical imaging technologies and molecular design strategies have revolutionized drug visualization. At the subcellular level, super-resolution microscopy has allowed exploration of the molecular landscape within individual cells and the cellular response to drugs. Moving beyond subcellular imaging, researchers have integrated multiple modes, like optical near-infrared II imaging, to study the complex spatiotemporal interactions between drugs and their surroundings. By combining these visualization approaches, researchers gain supplementary information on physiological parameters, metabolic activity, and tissue composition, leading to a comprehensive understanding of drug behavior. This review focuses on cutting-edge technologies in drug visualization, particularly fluorescence imaging, and the main types of fluorescent molecules used. Additionally, we discuss current challenges and prospects in targeted drug research, emphasizing the importance of multidisciplinary cooperation in advancing drug visualization. With the integration of advanced imaging technology and molecular design, drug visualization has the potential to redefine our understanding of pharmacology, enabling the analysis of drug micro-dynamics in subcellular environments from new perspectives and deepening pharmacological research to the levels of the cell and organelles.
Collapse
Affiliation(s)
- Ting Sun
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Huanxin Zhao
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Luyao Hu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xintian Shao
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- School of Life Sciences, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Zhiyuan Lu
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Yuli Wang
- Tianjin Pharmaceutical DA REN TANG Group Corporation Limited Traditional Chinese Pharmacy Research Institute, Tianjin 300457, China
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemistry Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Peixue Ling
- Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Key Laboratory of Biopharmaceuticals, Postdoctoral Scientific Research Workstation, Shandong Academy of Pharmaceutical Science, Jinan 250098, China
| | - Yubo Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Kewu Zeng
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qixin Chen
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| |
Collapse
|
3
|
Wen X, Zeng X, Liu J, Zeng X, Zhang Y, Cheng X, Huang J, Li Y, Zhuang R, Zhang X, Guo Z. In Vivo Comparative Study of Radioiodinated Folate Receptor Targeting Albumin Probes for Atherosclerosis Plaque Imaging. Bioconjug Chem 2023; 34:2387-2397. [PMID: 38055912 DOI: 10.1021/acs.bioconjchem.3c00486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
The objective of this study is to compare a series of albumin-based folate radiotracers for the potential imaging of folate receptor (FR) positive macrophages in advanced atherosclerotic plaques. Diversified radioiodinated FR-targeting albumin-binding probes ([131I]IBAbHF, [131I]IBNHF, and [131I]HF) were developed through various strategies. Among the three radiotracers, [131I]IBAbHF and [131I]IBNHF showed excellent in vitro stability (>98%) in saline and PBS 7.4 for 24 h. Also, good stability of [131I]IBNHF in mouse serum albumin was monitored using an HSA ELISA kit. The experiments in Raw264.7 macrophages activated by ox-LDL confirmed the specificity of tracers for FR-β. Biodistribution studies of radiotracers were performed to verify the prolonged blood half-life. Prolonged blood half-lives of [131I]IBAbHF, [131I]HF, and [131I]IBNHF were 17.26 ± 4.29, 6.33 ± 2.64, and 5.50 ± 1.26 h, respectively. SPECT-CT imaging of ApoE-/- mice at different stages was performed to evaluate the progression and monitor the prognosis of AS. Evident [131I]IBNHF uptake in atherosclerotic lesions could be observed along with a low background signal. In summary, we demonstrated a proof-of-concept of albumin-based radioligands for FR-targeting atherosclerosis imaging and found that different incorporation of radioiodinated groups resulted in different pharmacokinetic properties. Among these candidate compounds, [131I]IBNHF would be a satisfactory radiotracer for SPECT imaging of atherosclerosis.
Collapse
Affiliation(s)
- Xuejun Wen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xueyuan Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Jia Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xinying Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Yiren Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xingxing Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Jinxiong Huang
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, the First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Yesen Li
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, the First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Rongqiang Zhuang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Zhide Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| |
Collapse
|
4
|
Maschio R, Buonsanti F, Crivellin F, Ferretti F, Lattuada L, Maisano F, Orio L, Pizzuto L, Campanella R, Clouet A, Cavallotti C, Giovenzana GB. Improved synthesis of DA364, an NIR fluorescence RGD probe targeting α vβ 3 integrin. Org Biomol Chem 2023; 21:8584-8592. [PMID: 37855098 DOI: 10.1039/d3ob01206a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Optical imaging (OI) is gaining increasing attention in medicine as a non-invasive diagnostic imaging technology and as a useful tool for image-guided surgery. OI exploits the light emitted in the near-infrared region by fluorescent molecules able to penetrate living tissues. Cyanines are an important class of fluorescent molecules and by their conjugation to peptides it is possible to achieve optical imaging of tumours by selective targeting. We report here the improvements obtained in the synthesis of DA364, a small fluorescent probe (1.5 kDa) prepared by conjugation of pentamethine cyanine Cy5.5 to an RGD peptidomimetic, which can target tumour cells overexpressing integrin αvβ3 receptors.
Collapse
Affiliation(s)
- Rachele Maschio
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale, Largo Donegani 2/3, 28100 Novara, Italy.
| | - Federica Buonsanti
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Federico Crivellin
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Fulvio Ferretti
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Luciano Lattuada
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Federico Maisano
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Laura Orio
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Lorena Pizzuto
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Raphael Campanella
- Bracco Suisse SA, Route de la Galaise 31, 1228 Plan le Ouates, Switzerland
| | - Anthony Clouet
- Bracco Suisse SA, Route de la Galaise 31, 1228 Plan le Ouates, Switzerland
| | | | - Giovanni B Giovenzana
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale, Largo Donegani 2/3, 28100 Novara, Italy.
- CAGE Chemicals Srl, Via Bovio 6, 28100 Novara, Italy
| |
Collapse
|
5
|
Yue NN, Xu HM, Xu J, Zhu MZ, Zhang Y, Tian CM, Nie YQ, Yao J, Liang YJ, Li DF, Wang LS. Application of Nanoparticles in the Diagnosis of Gastrointestinal Diseases: A Complete Future Perspective. Int J Nanomedicine 2023; 18:4143-4170. [PMID: 37525691 PMCID: PMC10387254 DOI: 10.2147/ijn.s413141] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/02/2023] [Indexed: 08/02/2023] Open
Abstract
The diagnosis of gastrointestinal (GI) diseases currently relies primarily on invasive procedures like digestive endoscopy. However, these procedures can cause discomfort, respiratory issues, and bacterial infections in patients, both during and after the examination. In recent years, nanomedicine has emerged as a promising field, providing significant advancements in diagnostic techniques. Nanoprobes, in particular, offer distinct advantages, such as high specificity and sensitivity in detecting GI diseases. Integration of nanoprobes with advanced imaging techniques, such as nuclear magnetic resonance, optical fluorescence imaging, tomography, and optical correlation tomography, has significantly enhanced the detection capabilities for GI tumors and inflammatory bowel disease (IBD). This synergy enables early diagnosis and precise staging of GI disorders. Among the nanoparticles investigated for clinical applications, superparamagnetic iron oxide, quantum dots, single carbon nanotubes, and nanocages have emerged as extensively studied and utilized agents. This review aimed to provide insights into the potential applications of nanoparticles in modern imaging techniques, with a specific focus on their role in facilitating early and specific diagnosis of a range of GI disorders, including IBD and colorectal cancer (CRC). Additionally, we discussed the challenges associated with the implementation of nanotechnology-based GI diagnostics and explored future prospects for translation in this promising field.
Collapse
Affiliation(s)
- Ning-ning Yue
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Hao-ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
| | - Min-zheng Zhu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong, People’s Republic of China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Yu-qiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Yu-jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - De-feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Li-sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
6
|
Patras L, Shaashua L, Matei I, Lyden D. Immune determinants of the pre-metastatic niche. Cancer Cell 2023; 41:546-572. [PMID: 36917952 PMCID: PMC10170403 DOI: 10.1016/j.ccell.2023.02.018] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 03/16/2023]
Abstract
Primary tumors actively and specifically prime pre-metastatic niches (PMNs), the future sites of organotropic metastasis, preparing these distant microenvironments for disseminated tumor cell arrival. While initial studies of the PMN focused on extracellular matrix alterations and stromal reprogramming, it is increasingly clear that the far-reaching effects of tumors are in great part achieved through systemic and local PMN immunosuppression. Here, we discuss recent advances in our understanding of the tumor immune microenvironment and provide a comprehensive overview of the immune determinants of the PMN's spatiotemporal evolution. Moreover, we depict the PMN immune landscape, based on functional pre-clinical studies as well as mounting clinical evidence, and the dynamic, reciprocal crosstalk with systemic changes imposed by cancer progression. Finally, we outline emerging therapeutic approaches that alter the dynamics of the interactions driving PMN formation and reverse immunosuppression programs in the PMN ensuring early anti-tumor immune responses.
Collapse
Affiliation(s)
- Laura Patras
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Lee Shaashua
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
| | - David Lyden
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Zhang Q, Liu N, Wang J, Liu Y, Wang K, Zhang J, Pan X. The Recent Advance of Cell-Penetrating and Tumor-Targeting Peptides as Drug Delivery Systems Based on Tumor Microenvironment. Mol Pharm 2023; 20:789-809. [PMID: 36598861 DOI: 10.1021/acs.molpharmaceut.2c00629] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cancer has become the primary reason for industrial countries death. Although first-line treatments have achieved remarkable results in inhibiting tumors, they could have serious side effects because of insufficient selectivity. Therefore, specific localization of tumor cells is currently the main desire for cancer treatment. In recent years, cell-penetrating peptides (CPPs), as a kind of promising delivery vehicle, have attracted much attention because they mediate the high-efficiency import of large quantities of cargos in vivo and vitro. Unfortunately, the poor targeting of CPPs is still a barrier to their clinical application. In order to solve this problem, researchers use the various characteristics of tumor microenvironment and multiple receptors to improve the specificity toward tumors. This review focuses on the characteristics of the tumor microenvironment, and introduces the development of strategies and peptides based on these characteristics as drug delivery system in the tumor-targeted therapy.
Collapse
Affiliation(s)
- Qingqing Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Nanxin Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yuying Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Kai Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
8
|
Xu Y, Nie Z, Ni N, Zhang X, Yuan J, Gao Y, Gong Y, Liu S, Wu M, Sun X. Shield-activated two-way imaging nanomaterials for enhanced cancer theranostics. Biomater Sci 2022; 10:6893-6910. [PMID: 36317535 DOI: 10.1039/d2bm01317g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Smart nanomaterials with stimuli-responsive imaging enhancement have been widely developed to meet the requirements of accurate cancer diagnosis. However, these imaging nanoenhancers tend to be always on during circulation, which significantly increases the background signal when assessing the imaging performance. To improve unfavorable signal-to-noise ratios, an effective way is to shield the noise signal of these nanoprobes in non-targeted areas. Fortunately, there is a natural mutual shielding effect between some imaging nanomaterials, which provides the possibility of designing engineered nanomaterials with imaging quenching between two different components at the beginning. Once in the tumor microenvironment, the two components will present activated dual-mode imaging ability because of their separation, designated as two-way imaging tuning. This review highlights the design and mechanism of a series of engineered nanomaterials with two-way imaging tuning and their latest applications in the fields of cancer magnetic resonance imaging, fluorescence imaging, and their combination. The challenges and future directions for the improvement of these engineered nanomaterials towards clinical transformation are also discussed. This review aims to introduce the special constraint relationships of imaging components and provide scientists with simpler and more efficient nanoplatform construction ideas, promoting the development of engineered nanomaterials with two-way imaging tuning in cancer theranostics.
Collapse
Affiliation(s)
- Yang Xu
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Zhaokun Nie
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Nengyi Ni
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore 117585, Singapore
| | - Xinyu Zhang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Jia Yuan
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Yuan Gao
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Yufang Gong
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Shuangqing Liu
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Min Wu
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China.
| | - Xiao Sun
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| |
Collapse
|
9
|
Metal Peptide Conjugates in Cell and Tissue Imaging and Biosensing. Top Curr Chem (Cham) 2022; 380:30. [PMID: 35701677 PMCID: PMC9197911 DOI: 10.1007/s41061-022-00384-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 05/10/2022] [Indexed: 11/05/2022]
Abstract
Metal complex luminophores have seen dramatic expansion in application as imaging probes over the past decade. This has been enabled by growing understanding of methods to promote their cell permeation and intracellular targeting. Amongst the successful approaches that have been applied in this regard is peptide-facilitated delivery. Cell-permeating or signal peptides can be readily conjugated to metal complex luminophores and have shown excellent response in carrying such cargo through the cell membrane. In this article, we describe the rationale behind applying metal complexes as probes and sensors in cell imaging and outline the advantages to be gained by applying peptides as the carrier for complex luminophores. We describe some of the progress that has been made in applying peptides in metal complex peptide-driven conjugates as a strategy for cell permeation and targeting of transition metal luminophores. Finally, we provide key examples of their application and outline areas for future progress.
Collapse
|
10
|
Li Y, Xie M, Jones JB, Zhang Z, Wang Z, Dang T, Wang X, Lipowska M, Mao H. Targeted Delivery of DNA Topoisomerase Inhibitor SN38 to Intracranial Tumors of Glioblastoma Using Sub-5 Ultrafine Iron Oxide Nanoparticles. Adv Healthc Mater 2022; 11:e2102816. [PMID: 35481625 DOI: 10.1002/adhm.202102816] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 04/07/2022] [Indexed: 11/09/2022]
Abstract
Effectively delivering therapeutics for treating brain tumors is hindered by the physical and biological barriers in the brain. Even with the compromised blood-brain barrier and highly angiogenic blood-tumor barrier seen in glioblastoma (GBM), most drugs, including nanomaterial-based formulations, hardly reach intracranial tumors. This work investigates sub-5 nm ultrafine iron oxide nanoparticles (uIONP) with 3.5 nm core diameter as a carrier for delivering DNA topoisomerase inhibitor 7-ethyl-10-hydroxyl camptothecin (SN38) to treat GBM. Given a higher surface-to-volume ratio, uIONP shows one- or three-folds higher SN38 loading efficiency (48.3 ± 6.1%, mg/mg Fe) than those with core sizes of 10 or 20 nm. SN38 encapsulated in the coating polymer exhibits pH sensitive release with <10% over 48 h at pH 7.4, but 86% at pH 5, thus being protected from converting to inactive glucuronide by UDP-glucuronosyltransferase 1A1. Conjugating αv β3 -integrin-targeted cyclo(Arg-Gly-Asp-D-Phe-Cys) (RGD) as ligands, RGD-uIONP/SN38 demonstrates targeted cytotoxicity to αv β3 -integrin-overexpressed U87MG GBM cells with a half-maximal inhibitory concentration (IC50 ) of 30.9 ± 2.2 nm. The efficacy study using an orthotopic mouse model of GBM reveals tumor-specific delivery of 11.5% injected RGD-uIONP/SN38 (10 mg Fe kg-1 ), significantly prolonging the survival in mice by 41%, comparing to those treated with SN38 alone (p < 0.001).
Collapse
Affiliation(s)
- Yuancheng Li
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
- 5M Biomed, LLC Atlanta GA 30303 USA
| | - Manman Xie
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| | - Joshua B. Jones
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| | - Zhaobin Zhang
- Department of Neurosurgery Emory University Atlanta GA 30329 USA
| | - Zi Wang
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| | - Tu Dang
- Division of Research Philadelphia College of Osteopathic Medicine – Georgia Campus Suwanee GA 30024 USA
| | - Xinyu Wang
- Department of Pharmaceutical Sciences Philadelphia College of Osteopathic Medicine – Georgia Campus Suwanee GA 30024 USA
| | - Malgorzata Lipowska
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| |
Collapse
|
11
|
Alhalhooly L, Confeld MI, Woo SO, Mamnoon B, Jacobson R, Ghosh S, Kim J, Mallik S, Choi Y. Single-Molecule Force Probing of RGD-Binding Integrins on Pancreatic Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2022; 14:7671-7679. [PMID: 35113515 PMCID: PMC8890904 DOI: 10.1021/acsami.1c23361] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Integrin-targeting arginine-glycine-aspartic acid (RGD)-based nanocarriers have been widely used for tumor imaging, monitoring of tumor development, and delivery of anticancer drugs. However, the thermodynamics of an RGD-integrin formation and dissociation associated with binding dynamics, affinity, and stability remains unclear. Here, we probed the binding strength of the binary complex to live pancreatic cancer cells using single-molecule binding force spectroscopy methods, in which RGD peptides were functionalized on a force probe tip through poly(ethylene glycol) (PEG)-based bifunctional linker molecules. While the density of integrin αV receptors on the cell surface varies more than twofold from cell line to cell line, the individual RGD-integrin complexes exhibited a cell type-independent, monovalent bond strength. The load-dependent bond strength of multivalent RGD-integrin interactions scaled sublinearly with increasing bond number, consistent with the noncooperative, parallel bond model. Furthermore, the multivalent bonds ruptured sequentially either by one or in multiples, and the force strength was comparable to the synchronous rupture force. Comparison of energy landscapes of the bond number revealed a substantial decrease of kinetic off-rates for multivalent bonds, along with the increased width of the potential well and the increased potential barrier height between bound and unbound states, enhancing the stability of the multivalent bonds between them.
Collapse
Affiliation(s)
- Lina Alhalhooly
- Department of Physics, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Matthew I. Confeld
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Sung Oh Woo
- Department of Physics, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Babak Mamnoon
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Reed Jacobson
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Shrinwanti Ghosh
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Jiha Kim
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
- Molecular and Cellular Biology Program, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Yongki Choi
- Department of Physics, North Dakota State University, Fargo, North Dakota 58108, United State
- Molecular and Cellular Biology Program, North Dakota State University, Fargo, North Dakota 58108, United State
- Materials and Nanotechnology Program, North Dakota State University, Fargo, North Dakota 58108, United State
| |
Collapse
|
12
|
Zhao Z, Swartchick CB, Chan J. Targeted contrast agents and activatable probes for photoacoustic imaging of cancer. Chem Soc Rev 2022; 51:829-868. [PMID: 35094040 PMCID: PMC9549347 DOI: 10.1039/d0cs00771d] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Photoacoustic (PA) imaging has emerged as a powerful technique for the high resolution visualization of biological processes within deep tissue. Through the development and application of exogenous targeted contrast agents and activatable probes that can respond to a given cancer biomarker, researchers can image molecular events in vivo during cancer progression. This information can provide valuable details that can facilitate cancer diagnosis and therapy monitoring. In this tutorial review, we provide a step-by-step guide to select a cancer biomarker and subsequent approaches to design imaging agents for in vivo use. We envision this information will be a useful summary to those in the field, new members to the community, and graduate students taking advanced imaging coursework. We also highlight notable examples from the recent literature, with emphasis on the molecular designs and their in vivo PA imaging performance. To conclude, we provide our outlook and future perspective in this exciting field.
Collapse
Affiliation(s)
- Zhenxiang Zhao
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois, USA.
| | - Chelsea B Swartchick
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois, USA.
| | - Jefferson Chan
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois, USA.
| |
Collapse
|
13
|
Li H, Kim Y, Jung H, Hyun JY, Shin I. Near-infrared (NIR) fluorescence-emitting small organic molecules for cancer imaging and therapy. Chem Soc Rev 2022; 51:8957-9008. [DOI: 10.1039/d2cs00722c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We discuss recent advances made in the development of NIR fluorescence-emitting small organic molecules for tumor imaging and therapy.
Collapse
Affiliation(s)
- Hui Li
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea
| | - Yujun Kim
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea
| | - Hyoje Jung
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea
| | - Ji Young Hyun
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Injae Shin
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea
| |
Collapse
|
14
|
Gambino G, Gambino T, Connah L, La Cava F, Evrard H, Angelovski G. RGD-Peptide Functionalization Affects the In Vivo Diffusion of a Responsive Trimeric MRI Contrast Agent through Interactions with Integrins. J Med Chem 2021; 64:7565-7574. [PMID: 33961422 PMCID: PMC8279402 DOI: 10.1021/acs.jmedchem.1c00264] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The relevance of
MRI as a diagnostic methodology has been expanding
significantly with the development of molecular imaging. Partially,
the credit for this advancement is due to the increasing potential
and performance of targeted MRI contrast agents, which are able to
specifically bind distinct receptors or biomarkers. Consequently,
these allow for the identification of tissues undergoing a disease,
resulting in the over- or underexpression of the particular molecular
targets. Here we report a multimeric molecular probe, which combines
the established targeting properties of the Arg-Gly-Asp (RGD) peptide
sequence toward the integrins with three calcium-responsive, Gd-based
paramagnetic moieties. The bifunctional probe showed excellent 1H MRI contrast enhancement upon Ca2+ coordination
and demonstrated a longer retention time in the tissue due to the
presence of the RGD moiety. The obtained results testify to the potential
of combining bioresponsive contrast agents with targeting vectors
to develop novel functional molecular imaging methods.
Collapse
Affiliation(s)
- Giuseppe Gambino
- Max Planck Institute for Biological Cybernetics, Department for Physiology of Cognitive Processes, Max-Planck-Ring 11, 72072 Tübingen, Germany
| | - Tanja Gambino
- Max Planck Institute for Biological Cybernetics, Department for Physiology of Cognitive Processes, Max-Planck-Ring 11, 72072 Tübingen, Germany
| | - Liam Connah
- Max Planck Institute for Biological Cybernetics, Department for Physiology of Cognitive Processes, Max-Planck-Ring 11, 72072 Tübingen, Germany
| | - Francesca La Cava
- Max Planck Institute for Biological Cybernetics, Department for Physiology of Cognitive Processes, Max-Planck-Ring 11, 72072 Tübingen, Germany
| | - Henry Evrard
- Max Planck Institute for Biological Cybernetics, Department for Physiology of Cognitive Processes, Max-Planck-Ring 11, 72072 Tübingen, Germany.,Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, New York 10962, United States.,Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Strasse 25, 72076 Tübingen, Germany
| | - Goran Angelovski
- Max Planck Institute for Biological Cybernetics, Department for Physiology of Cognitive Processes, Max-Planck-Ring 11, 72072 Tübingen, Germany.,Laboratory of Molecular and Cellular Neuroimaging, International Center for Primate Brain Research (ICPBR), Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Sciences (CAS), Shanghai 200031, PR China
| |
Collapse
|
15
|
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: 131] [Impact Index Per Article: 32.8] [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
|
16
|
Brennecke B, Wang Q, Haap W, Grether U, Hu HY, Nazaré M. DOTAM-Based, Targeted, Activatable Fluorescent Probes for the Highly Sensitive and Selective Detection of Cancer Cells. Bioconjug Chem 2021; 32:702-712. [PMID: 33691062 DOI: 10.1021/acs.bioconjchem.0c00699] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The utilization of an activatable, substrate-based probe design in combination with a cellular targeting approach has been rarely explored for cancer imaging on a small-molecule basis, although such probes could benefit from advantages of both concepts. Cysteine proteases like cathepsin S are known to be involved in fundamental processes associated with tumor development and progression and thus are valuable cancer markers. We report the development of a combined dual functional DOTAM-based, RGD-targeted internally quenched fluorescent probe that is activated by cathepsin S. The probe exhibits excellent in vitro activation kinetics which can be fully translated to human cancer cell lines. We demonstrate that the targeted, activatable probe is superior to its nontargeted analog, exhibiting improved uptake into ανβ3-integrin expressing human sarcoma cells (HT1080) and significantly higher resultant fluorescence staining. However, profound activation was also found in cancer cells with a lower integrin expression level, whereas in healthy cells almost no probe activation could be observed, highlighting the high selectivity of our probe toward cancer cells. These auspicious results show the outstanding potential of the dual functionality concept combining a substrate-based probe design with a targeting approach, which could form the basis for highly sensitive and selective in vivo imaging probes.
Collapse
Affiliation(s)
- Benjamin Brennecke
- Medicinal Chemistry, Leibniz-Forschungsinstitut für Molekulare Pharmakologie Berlin, 13125 Berlin, Germany
| | - Qinghua Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Wolfgang Haap
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Uwe Grether
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Hai-Yu Hu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Marc Nazaré
- Medicinal Chemistry, Leibniz-Forschungsinstitut für Molekulare Pharmakologie Berlin, 13125 Berlin, Germany
| |
Collapse
|
17
|
Cai C, Sun H, Hu L, Fan Z. Visualization of integrin molecules by fluorescence imaging and techniques. ACTA ACUST UNITED AC 2021; 45:229-257. [PMID: 34219865 PMCID: PMC8249084 DOI: 10.32604/biocell.2021.014338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Integrin molecules are transmembrane αβ heterodimers involved in cell adhesion, trafficking, and signaling. Upon activation, integrins undergo dynamic conformational changes that regulate their affinity to ligands. The physiological functions and activation mechanisms of integrins have been heavily discussed in previous studies and reviews, but the fluorescence imaging techniques -which are powerful tools for biological studies- have not. Here we review the fluorescence labeling methods, imaging techniques, as well as Förster resonance energy transfer assays used to study integrin expression, localization, activation, and functions.
Collapse
Affiliation(s)
- Chen Cai
- Department of Immunology, School of Medicine, UConn Health, Farmington, 06030, USA
| | - Hao Sun
- Department of Medicine, University of California, San Diego, La Jolla, 92093, USA
| | - Liang Hu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450051, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, 06030, USA
| |
Collapse
|
18
|
Sebak AA, Gomaa IEO, ElMeshad AN, Farag MH, Breitinger U, Breitinger HG, AbdelKader MH. Distinct Proteins in Protein Corona of Nanoparticles Represent a Promising Venue for Endogenous Targeting - Part I: In vitro Release and Intracellular Uptake Perspective. Int J Nanomedicine 2020; 15:8845-8862. [PMID: 33204091 PMCID: PMC7667594 DOI: 10.2147/ijn.s273713] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022] Open
Abstract
Introduction Protein corona (PC) deposition on nanoparticles (NPs) in biological systems contributes to a great extent to NPs' fates; their targeting potential, the interaction with different biological systems and the subsequent functions. PC - when properly tuned - can serve as a potential avenue for optimization of NPs' use in cancer therapy. Methods Poly-lactic co-glycolic acid (PLGA)-based NPs exhibiting different physicochemical properties were fabricated and characterized. The PC makeup of these NPs were qualitatively and quantitatively analyzed by Western blot and Bradford assay, respectively. The effect of PC on the release of NPs' cargos and the intracellular uptake into B16F10 melanoma cells has been studied. Results The composition of NPs (polymeric PLGA NPs vs lipid-polymer hybrid NPs) and the conjugation of an active targeting ligand (cRGDyk peptide) represented the major determinants of the PC makeup of NPs. The in vitro release of the loaded cargos from the NPs depended on the PC and the presence of serum proteins in the release medium. Higher cumulative release has been recorded in the presence of proteins in the case of peptide conjugated NPs, cNPs, while the unconjugated formulations, uNPs, showed an opposite pattern. NPs intracellular uptake studies revealed important roles of distinct serum and cellular proteins on the extent of NPs' accumulation in melanoma cells. For example, the abundance of vitronectin (VN) protein from serum has been positively related to the intracellular accumulation of the NPs. Conclusion Careful engineering of nanocarriers can modulate the recruitment of some proteins suggesting a potential use for achieving endogenous targeting to overcome the current limitations of targeted delivery of chemotherapeutic agents.
Collapse
Affiliation(s)
- Aya Ahmed Sebak
- Pharmaceutical Technology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Iman Emam Omar Gomaa
- Biochemistry Department, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Aliaa Nabil ElMeshad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Mahmoud Hussien Farag
- Pharmaceutical Technology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Ulrike Breitinger
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Hans-Georg Breitinger
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Mahmoud Hashem AbdelKader
- National Institute of Laser Enhanced Sciences (NILES), Cairo University (CU), Giza, Egypt.,European University in Egypt (EUE), New Administrative Capital, Cairo, Egypt
| |
Collapse
|
19
|
Zhang M, Zhai X, Sun M, Ma T, Huang Y, Huang B, Du Y, Yan C. When rare earth meets carbon nanodots: mechanisms, applications and outlook. Chem Soc Rev 2020; 49:9220-9248. [PMID: 33165456 DOI: 10.1039/d0cs00462f] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Rare earth (RE) elements are widely used in the luminescence and magnetic fields by virtue of their abundant 4f electron configurations. However, the overall performance and aqueous stability of single-component RE materials need to be urgently improved to satisfy the requirements for multifunctional applications. Carbon nanodots (CNDs) are excellent nanocarriers with abundant functional surface groups, excellent hydrophilicity, unique photoluminescence (PL) and tunable features. Accordingly, RE-CND hybrids combine the merits of both RE and CNDs, which dramatically enhance their overall properties such as luminescent and magnetic-optical imaging performances, leading to highly promising practical applications in the future. Nevertheless, a comprehensive review focusing on the introduction and in-depth understanding of RE-CND hybrid materials has not been reported to date. This review endeavors to summarize the recent advances of RE-CNDs, including their interaction mechanisms, general synthetic strategies and applications in fluorescence, biosensing and multi-modal biomedical imaging. Finally, we present the current challenges and the possible application perspectives of newly developed RE-CND materials. We hope this review will inspire new design ideas and valuable references in this promising field in the future.
Collapse
Affiliation(s)
- Mengzhen Zhang
- Tianjin Key Lab for Rare Earth Materials and Applications, Center for Rare Earth and Inorganic Functional Materials, School of Materials Science and Engineering & National Institute for Advanced Materials, Nankai University, Tianjin, 300350, China.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Ruman U, Fakurazi S, Masarudin MJ, Hussein MZ. Nanocarrier-Based Therapeutics and Theranostics Drug Delivery Systems for Next Generation of Liver Cancer Nanodrug Modalities. Int J Nanomedicine 2020; 15:1437-1456. [PMID: 32184597 PMCID: PMC7060777 DOI: 10.2147/ijn.s236927] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/18/2020] [Indexed: 12/13/2022] Open
Abstract
The development of therapeutics and theranostic nanodrug delivery systems have posed a challenging task for the current researchers due to the requirement of having various nanocarriers and active agents for better therapy, imaging, and controlled release of drugs efficiently in one platform. The conventional liver cancer chemotherapy has many negative effects such as multiple drug resistance (MDR), high clearance rate, severe side effects, unwanted drug distribution to the specific site of liver cancer and low concentration of drug that finally reaches liver cancer cells. Therefore, it is necessary to develop novel strategies and novel nanocarriers that will carry the drug molecules specific to the affected cancerous hepatocytes in an adequate amount and duration within the therapeutic window. Therapeutics and theranostic systems have advantages over conventional chemotherapy due to the high efficacy of drug loading or drug encapsulation efficiency, high cellular uptake, high drug release, and minimum side effects. These nanocarriers possess high drug accumulation in the tumor area while minimizing toxic effects on healthy tissues. This review focuses on the current research on nanocarrier-based therapeutics and theranostic drug delivery systems excluding the negative consequences of nanotechnology in the field of drug delivery systems. However, clinical developments of theranostics nanocarriers for liver cancer are considered outside of the scope of this article. This review discusses only the recent developments of nanocarrier-based drug delivery systems for liver cancer therapy and diagnosis. The negative consequences of individual nanocarrier in the drug delivery system will also not be covered in this review.
Collapse
Affiliation(s)
- Umme Ruman
- Materials Synthesis and Characterization Laboratory, Institute of Advanced Technology (ITMA), Universiti Putra Malaysia, Serdang, Selangor43400, Malaysia
| | - Sharida Fakurazi
- Laboratory of Vaccine and Immunotherapeutics, Institute of Bioscience Universiti, Putra43400, Malaysia
- Department of Human Anatomy, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Serdang, Selangor43400, Malaysia
| | - Mas Jaffri Masarudin
- Materials Synthesis and Characterization Laboratory, Institute of Advanced Technology (ITMA), Universiti Putra Malaysia, Serdang, Selangor43400, Malaysia
- Laboratory of Vaccine and Immunotherapeutics, Institute of Bioscience Universiti, Putra43400, Malaysia
- Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Selangor43400, Malaysia
| | - Mohd Zobir Hussein
- Materials Synthesis and Characterization Laboratory, Institute of Advanced Technology (ITMA), Universiti Putra Malaysia, Serdang, Selangor43400, Malaysia
| |
Collapse
|
21
|
Qi B, Crawford AJ, Wojtynek NE, Talmon GA, Hollingsworth MA, Ly QP, Mohs AM. Tuned near infrared fluorescent hyaluronic acid conjugates for delivery to pancreatic cancer for intraoperative imaging. Theranostics 2020; 10:3413-3429. [PMID: 32206099 PMCID: PMC7069077 DOI: 10.7150/thno.40688] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
The prognosis of pancreatic cancer remains poor. Intraoperative fluorescence imaging of tumors could improve staging and surgical resection, thereby improving prognosis. However, imaging pancreatic cancer with macromolecular delivery systems, is often hampered by nonspecific organ accumulation. Methods: We describe the rational development of hyaluronic acid (HA) conjugates that vary in molecular weight and are conjugated to near infrared fluorescent (NIRF) dyes that have differences in hydrophilicity, serum protein binding affinity, and clearance mechanism. We systematically investigated the roles of each of these properties on tumor accumulation, relative biodistribution, and the impact of intraoperative imaging of orthotopic, syngeneic pancreatic cancer. Results: Each HA-NIRF conjugate displayed intrapancreatic tumor enhancement. Regardless of HA molecular weight, Cy7.5 conjugation directed biodistribution to the liver, spleen, and bowels. Conjugation of IRDye800 to 5 and 20 kDa HA resulted in low liver and spleen signal while enhancing the tumor up to 14-fold compared to healthy pancreas, while 100 kDa HA conjugated to IRDye800 resulting in liver and spleen accumulation. Conclusion: These studies demonstrate that by tuning HA molecular weight and the physicochemical properties of the conjugated moiety, in this case a NIRF probe, peritoneal biodistribution can be substantially altered to achieve optimized delivery to tumors intraoperative abdominal imaging.
Collapse
|
22
|
Kim H, Choi HS, Eom JB, Choi Y. Mini-Platform for Off-On Near-Infrared Fluorescence Imaging Using Peptide-Targeting Ligands. Bioconjug Chem 2020; 31:721-728. [PMID: 31895549 DOI: 10.1021/acs.bioconjchem.9b00844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Here, we propose a zwitterionic near-infrared (NIR) fluorophore-tryptophan (Trp) conjugate with a cleavable linker as a minimal-sized versatile platform (MP) for the preparation of peptide ligand-based off-on type molecular probes. The zwitterionic NIR fluorophore in MP undergoes fluorescence quenching via a photoinduced electron transfer mechanism when in close proximity to tryptophan, and nonspecific binding with serum proteins is minimized by the zwitterionicity of the fluorophore. The linker can be cleaved inside cancer cells in response to tumor-associated stimuli. As a proof-of-concept experiment, ATTO655 was covalently linked with Trp via a diarginine linker to form an MP. A cyclic peptide consisting of Arg-Gly-Asp-d-Phe-Lys (cRGD) was used as a cancer-targeting ligand and was conjugated to the MP to form cRGD-MP. The NIR fluorescence of cRGD-MP could be selectively turned on inside the target cancer cells, thereby enabling specific fluorescence imaging of integrin αvβ3-overexpressing cancer cells in vitro and in vivo.
Collapse
Affiliation(s)
- Hyunjin Kim
- National Cancer Center, 323 Ilsan-ro, Goyang, Gyeonggi-do 10408, Republic of Korea
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Joo Beom Eom
- Medical Photonics Research Center, Korea Photonics Technology Institute (KOPTI), Gwangju-si 61007, Republic of Korea
| | - Yongdoo Choi
- National Cancer Center, 323 Ilsan-ro, Goyang, Gyeonggi-do 10408, Republic of Korea
| |
Collapse
|
23
|
Alipour M, Baneshi M, Hosseinkhani S, Mahmoudi R, Jabari Arabzadeh A, Akrami M, Mehrzad J, Bardania H. Recent progress in biomedical applications of RGD-based ligand: From precise cancer theranostics to biomaterial engineering: A systematic review. J Biomed Mater Res A 2019; 108:839-850. [PMID: 31854488 DOI: 10.1002/jbm.a.36862] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/03/2019] [Accepted: 12/06/2019] [Indexed: 12/17/2022]
Abstract
Arginine-glycine-aspartic acid (RGD) peptide family is known as the most prominent ligand for extracellular domain of integrin receptors. Specific expression of these receptors in various tissue of human body and tight association of their expression profile with various pathophysiological conditions made these receptors a suitable targeting candidate for several disease diagnosis and treatment as well as regeneration of various organs. For these reasons, various forms of RGD-based integrins ligands have been greatly used in biomedical studies. Here, we summarized the last decade application progress of RGD for cancer theranostics, control of inflammation, thrombosis inhibition and critically discussed the effect of RGD peptides structure and sequence on the efficacy of gene/drug delivery systems in preclinical studies. Furthermore, we will show recent advances in application of RGD functionalized biomaterials for various tissue regenerations including cornea repair, artificial neovascularization and bone tissue regeneration. Finally, we analyzed clinically translatability of RGD peptides, considering examples of integrin ligands in clinical trials. In conclusion, prospects on using RGD peptide for precise drug delivery and biomaterial engineering are well discussed.
Collapse
Affiliation(s)
- Mohsen Alipour
- Department of Advanced Medical Sciences and Technologies, School of Medicine, Jahrom University of Medical Sciences (JUMS), Jahrom, Iran
- Department of Nano Biotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Marzieh Baneshi
- Department of Chemistry, Yazd University, Yazd, Iran
- Department of Chemistry, Cape Breton University, Sydney, Nova Scotia, Canada
| | - Saman Hosseinkhani
- Department of Nano Biotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza Mahmoudi
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Ali Jabari Arabzadeh
- Department of Radiopharmaceutical Sciences, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Akrami
- Department of Pharmaceutical Biomaterials, and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Jalil Mehrzad
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hassan Bardania
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| |
Collapse
|
24
|
Choi N, Jeong HS. Precision surgery for cancer: a new surgical concept in individual tumor biology-based image-guided surgery. PRECISION AND FUTURE MEDICINE 2019. [DOI: 10.23838/pfm.2019.00072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
25
|
Chen P, Kuang W, Zheng Z, Yang S, Liu Y, Su L, Zhao K, Liang G. Carboxylesterase-Cleavable Biotinylated Nanoparticle for Tumor-Dual Targeted Imaging. Theranostics 2019; 9:7359-7369. [PMID: 31695773 PMCID: PMC6831296 DOI: 10.7150/thno.37625] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 08/29/2019] [Indexed: 12/30/2022] Open
Abstract
Near-infrared (NIR) nanoprobes with fluorescence "Turn-On" property are advantageous in cancer diagnosis but, to the best of our knowledge, "smart" nanoprobe that simultaneously targets both biotin receptor and carboxylesterase (CES) for HepG2 tumor-dual targeted imaging has not been reported. Methods: Using CBT-Cys click condensation reaction, we rationally designed a "smart" NIR fluorescence probe H2N-Cys(StBu)-Lys(Biotin)-Ser(Cy5.5)-CBT (NIR-CBT) and used it to facilely prepare the fluorescence-quenched nanoparticle NIR-CBT-NP. Results: In vitro results indicated that, after NIR-CBT-NP was incubated with CES for 6 h, its fluorescence was turned "On" by 69 folds. Cell experiments verified that NIR-CBT-NP was uptaken by HepG2 cells via biotin receptor-assisted endocytosis and its fluorescence was turned "On" by intracellular CES hydrolysis. Moreover, NIR-CBT-NP was successfully applied to image both biotin receptor- and CES-overexpressing HepG2 tumors. Conclusion: Fluorescence-quenched nanoparticle NIR-CBT-NP was facilely prepared to actively target biotin receptor-overexpressing HepG2 cancer cells and turn the fluorescence "On" by intracellular CES hydrolysis for tumor-dual targeted imaging. We anticipate that our fluorescence "Turn-On" nanoparticle could be applied for liver cancer diagnosis in clinic in the near future.
Collapse
Affiliation(s)
- Peiyao Chen
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Wen Kuang
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Zhen Zheng
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Shuye Yang
- Department of PET Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, China
| | - Yaling Liu
- Jiangsu Institute of Nuclear Medicine, 20 Qianrong Road, Wuxi, Jiangsu 214063, China
| | - Lanhong Su
- School of Life Sciences, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui 230027, China
| | - Kui Zhao
- Department of PET Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, China
| | - Gaolin Liang
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| |
Collapse
|
26
|
Cheng J, Zhang R, Li C, Tao H, Dou Y, Wang Y, Hu H, Zhang J. A Targeting Nanotherapy for Abdominal Aortic Aneurysms. J Am Coll Cardiol 2019; 72:2591-2605. [PMID: 30466517 DOI: 10.1016/j.jacc.2018.08.2188] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 08/14/2018] [Accepted: 08/20/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a leading cause of mortality and morbidity in the elderly. Currently, there remain no effective drugs that can prevent the growth of aneurysms and delay aneurysm rupture in the clinical setting. OBJECTIVES The aim of this study was to develop a nanotherapy that can target aneurysms and release drug molecules in response to the inflammatory microenvironment. METHODS Using a reactive oxygen species (ROS)-responsive nanoparticle and a candidate drug rapamycin, in combination with a peptide ligand for integrin and biomimetic cloaking with macrophage cell membrane, a nanotherapy was developed. Its effectiveness was demonstrated by in vitro and in vivo studies. RESULTS Based on a facile and translational method, a rapamycin-loaded responsive nanotherapy was successfully prepared, which could release drug molecules upon triggering by the high level of ROS. In cells associated with the development of AAAs, the nanotherapy significantly inhibited calcification and attenuated ROS-mediated oxidative stress and apoptosis. By passively targeting aneurysms and releasing drug molecules in response to the inflammatory microenvironment, the intravenously injected ROS-responsive nanotherapy more effectively prevented aneurysm expansion in AAA rats than a nonresponsive control nanotherapy. After decoration with a peptide ligand cRGDfK and macrophage cell membrane, the aneurysmal targeting capability and therapeutic effects of a ROS-responsive nanotherapy with a mean diameter of 190 nm were further enhanced. Moreover, the nanotherapy showed a good safety profile in a preliminary safety test. CONCLUSIONS The multifunctional nanotherapy can be further studied as a promising targeted drug for treatment of aneurysms. The underlying design principles enable the development of a broad range of nanomedicines for targeted therapy of other vascular diseases.
Collapse
Affiliation(s)
- Juan Cheng
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Runjun Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China; Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hui Tao
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yin Dou
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yuquan Wang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China; Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Houyuan Hu
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, China.
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China.
| |
Collapse
|
27
|
Huang D, He B, Mi P. Calcium phosphate nanocarriers for drug delivery to tumors: imaging, therapy and theranostics. Biomater Sci 2019; 7:3942-3960. [PMID: 31414096 DOI: 10.1039/c9bm00831d] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Calcium phosphate (CaP) was engineered as a drug delivery nanocarrier nearly 50 years ago due to its biocompatibility and biodegradability. In recent years, several approaches have been developed for the preparation of size-controllable, stable and multifunctional CaP nanocarriers, and several targeting moieties have also been decorated on the surface of these nanocarriers for active targeting. The CaP nanocarriers have been utilized for loading probes, nucleic acids, anticancer drugs and photosensitizers for cancer imaging, therapy and theranostics. Herein, we reviewed the recent advances in the preparation strategies of CaP nanocarriers and the applications of these nanocarriers in tumor diagnosis, gene delivery, drug delivery and theranostics and finally provided perspectives.
Collapse
Affiliation(s)
- Dan Huang
- Department of Radiology, Center for Medical Imaging, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Number 17, 3rd Section, Renmin South Road, Chengdu, Sichuan 610041, P.R. China.
| | - Bin He
- Department of Radiology, Center for Medical Imaging, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Number 17, 3rd Section, Renmin South Road, Chengdu, Sichuan 610041, P.R. China.
| | - Peng Mi
- Department of Radiology, Center for Medical Imaging, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Number 17, 3rd Section, Renmin South Road, Chengdu, Sichuan 610041, P.R. China.
| |
Collapse
|
28
|
An integrin-targeting glutathione-activated zinc(II) phthalocyanine for dual targeted photodynamic therapy. Eur J Med Chem 2019; 174:56-65. [DOI: 10.1016/j.ejmech.2019.04.049] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/15/2022]
|
29
|
|
30
|
Thoreau F, Vanwonterghem L, Henry M, Coll JL, Boturyn D. Design of RGD-ATWLPPR peptide conjugates for the dual targeting of α Vβ 3 integrin and neuropilin-1. Org Biomol Chem 2019; 16:4101-4107. [PMID: 29774910 DOI: 10.1039/c8ob00669e] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Targeting the tumour microenvironment is a promising strategy to detect and/or treat cancer. The design of selective compounds that co-target several receptors frequently overexpressed in solid tumours may allow a reliable and selective detection of tumours. Here we report the modular synthesis of compounds encompassing ligands of αVβ3 integrin and neuropilin-1 that are overexpressed in the tumour microenvironment. These compounds were then evaluated through cellular experiments and imaging of tumours in mice. We observed that the peptide that displays both ligands is more specifically accumulating in the tumours than in controls. Simultaneous interaction with αVβ3 integrin and NRP1 induces NRP1 stabilization at the cell membrane surface which is not observed with the co-injection of the controls.
Collapse
Affiliation(s)
- Fabien Thoreau
- Univ. Grenoble Alpes, CNRS, Department of Molecular Chemistry, UMR 5250, F-38000 Grenoble, France.
| | | | | | | | | |
Collapse
|
31
|
Sharma M, Dube T, Chibh S, Kour A, Mishra J, Panda JJ. Nanotheranostics, a future remedy of neurological disorders. Expert Opin Drug Deliv 2019; 16:113-128. [PMID: 30572726 DOI: 10.1080/17425247.2019.1562443] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Effective therapy of various neurological disorders is hindered on account of the failure of various therapeutics crossing blood-brain-barrier (BBB). Nanotheranostics has emerged as a cutting-edge unconventional theranostic nanomedicine, capable of realizing accurate diagnosis together with effective and targeted delivery of therapeutics across BBB to the unhealthy regions of the brain for potential clinical success. AREAS COVERED We have tried to review the current status of nanotheranostic based approaches followed to manage neurological disorders. The focus has been majorly laid on to explore various theranostic nanoparticles and their application potential towards image-guided neurotherapies. Additionally, the usefulness of exceptional diagnostic, imaging techniques including magnetic resonance imaging and fluorescence imaging are being discussed by highlighting their promising opportunities in the detection, diagnosis, and treatment of the neurological disorders. EXPERT OPINION Inimitable diagnostic and therapeutic potential of nanotheranostics have accomplished the aim of personalized therapies by governing the therapeutic efficacy of the system along with facilitating patient pre-selection grounded on non-invasive imaging, thereby predicting the responses of patients to nanomedicine treatments. While these accomplishments are encouraging, they are still the minority and demands for a continuous effort to improve sensitivity and precision in screening/diagnosis along with improving therapeutic efficacy in various neural disorders.
Collapse
Affiliation(s)
- Manju Sharma
- a Institute of Nano Science and Technology , Mohali , India
| | - Taru Dube
- a Institute of Nano Science and Technology , Mohali , India
| | - Sonika Chibh
- a Institute of Nano Science and Technology , Mohali , India
| | - Avneet Kour
- a Institute of Nano Science and Technology , Mohali , India
| | - Jibanananda Mishra
- b School of Bioengineering and Biosciences , Lovely Professional University , Phagwara , India
| | | |
Collapse
|
32
|
Facciolà A, Visalli G, La Maestra S, Ceccarelli M, D'Aleo F, Nunnari G, Pellicanò GF, Di Pietro A. Carbon nanotubes and central nervous system: Environmental risks, toxicological aspects and future perspectives. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 65:23-30. [PMID: 30500734 DOI: 10.1016/j.etap.2018.11.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/05/2018] [Accepted: 11/22/2018] [Indexed: 06/09/2023]
Abstract
Due to their morphological and physicochemical properties, carbon nanotubes (CNTs) enhance the structural properties of several materials and are produced in great volumes. The production and the manufacturing of CNTs-incorporated products can lead to the potential environmental release of CNTs. For these reasons, CNTs can represent a serious concern for human health. Humans are exposed to nanoparticles through inhalation, ingestion and skin uptake. After their entrance, the particles can reach the Central Nervous System (CNS) through three different pathways: the systemic, olfactory and trigeminal pathways. In the first, through systemic blood circulation, nanoparticles cross both the blood-brain and blood-spinal cord barriers, which are highly selective semipermeable barriers that protect the CNS compartments. The second is the step from the nose to brain route and occurs along axons and via nerve bundles that cross the cribriform plate to the olfactory bulb. In the third, the compounds diffuse through the nasal cavity mucosa to reach the branches of the trigeminal nerve in the olfactory and respiratory regions, and they reach brain stem via axonal transport. After their entrance, CNTs reach the CNS where they may cause cytotoxicity of selected neurons in several CNS regions, impairing molecular pathways and contributing to the onset and progression of chronic brain inflammation, microglia activation and white matter abnormalities with an increased risk for autism spectrum disorders, lower IQ in children, neurodegenerative diseases and stroke. The large surface area to mass ratio of CNTs greatly increases surface reactivity. Despite this property considerable contributes to their toxicological profile in biological systems, also makes CNTs very attractive in the medical field, where they can be used as carriers of bioactive molecules, contrast agents, biological platforms and for many other applications in medicine.
Collapse
Affiliation(s)
- Alessio Facciolà
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Messina, Italy
| | - Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Italy
| | | | - Manuela Ceccarelli
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Messina, Italy
| | - Francesco D'Aleo
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Messina, Italy
| | - Giuseppe Nunnari
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Messina, Italy
| | | | - Angela Di Pietro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Italy.
| |
Collapse
|
33
|
Targeted photodynamic-induced singlet oxygen production by peptide-conjugated biodegradable nanoparticles for treatment of skin melanoma. Photodiagnosis Photodyn Ther 2018; 23:181-189. [DOI: 10.1016/j.pdpdt.2018.05.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/15/2018] [Accepted: 05/31/2018] [Indexed: 01/30/2023]
|
34
|
Xiao Y, Zhang Q, Wang Y, Wang B, Sun F, Han Z, Feng Y, Yang H, Meng S, Wang Z. Dual-functional protein for one-step production of a soluble and targeted fluorescent dye. Theranostics 2018; 8:3111-3125. [PMID: 29896306 PMCID: PMC5996361 DOI: 10.7150/thno.24613] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 03/17/2018] [Indexed: 01/17/2023] Open
Abstract
Low water solubility and poor selectivity are two fundamental limitations that compromise applications of near-infrared (NIR) fluorescent probes. Methods: Here, a simple strategy that can resolve these problems simultaneously was developed by using a novel hybrid protein named RGD-HFBI that is produced by fusion of hydrophobin HFBI and arginine-glycine-aspartic acid (RGD) peptide. This unique hybrid protein inherits self-assembly and targeting functions from HFBI and RGD peptide respectively. Results: Boron-dipyrromethene (BODIPY) used as a model NIR dye can be efficiently dispersed in the RGD-HFBI solution by simple mixing and sonication for 30 min. The data shows that self-assembled RGD-HFBI forms a protein nanocage by using the BODIPY as the assembly template. Cell uptake assay proves that RGD-HFBI/BODIPY can efficiently stain αvβ3 integrin-positive cancer cells. Finally, in vivo affinity tests fully demonstrate that the soluble RGD-HFBI/BODIPY complex selectively targets and labels tumor sites of tumor-bearing mice due to the high selectivity of the RGD peptide. Conclusion: Our one-step strategy using dual-functional RGD-HFBI opens a novel route to generate soluble and targeted NIR fluorescent dyes in a very simple and efficient way and may be developed as a general strategy to broaden their applications.
Collapse
Affiliation(s)
- Yunjie Xiao
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Qian Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Yanyan Wang
- College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Bin Wang
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Fengnan Sun
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Ziyu Han
- College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Yaqing Feng
- Collaborative Innovation Center of Chemical Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Haitao Yang
- School of Life Sciences, Tianjin University, Tianjin 300072, China
- Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin 300457, China
| | - Shuxian Meng
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Zefang Wang
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| |
Collapse
|
35
|
Tang L, Sun X, Liu N, Zhou Z, Yu F, Zhang X, Sun X, Chen X. Radiolabeled Angiogenesis-Targeting Croconaine Nanoparticles for Trimodality Imaging Guided Photothermal Therapy of Glioma. ACS APPLIED NANO MATERIALS 2018; 1:1741-1749. [PMID: 30506043 PMCID: PMC6261507 DOI: 10.1021/acsanm.8b00195] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
To meet the criteria of effective theranostics, biocompatible nanomedicine endowing intrinsic therapeutic and imaging properties have gained extraordinary momentum. In this study, an ultra-stable near-infrared (NIR) dye croconaine (CR780) was engineered with arginine-glycine-aspartic acid (RGD) peptide and polyethylene glycol (PEG), which was then self-assembled into uniform nanoparticles (NPs). These RGD-CR780-PEG5K assemblies were radiolabeled with 125I through a facile standard Iodo-Gen method. The resulting [125I]RGD-CR780-PEG5K NPs showed effective accumulation in αvβ3 integrin expressing glioblastoma, as evidenced by single photon emission computed tomography (SPECT)/CT and NIR fluorescence imaging. More importantly, high-resolution photoacoustic imaging revealed that these NPs selectively targeted to angiogenic tumor vessels. With the favorable tumor selective accumulation and high photothermal conversion efficiency, the [125I]RGD-CR780-PEG5K NPs allowed thorough tumor ablation and inhibition of tumor relapse at a relatively low laser energy (0.5 W/cm2). Overall, this work offers a proper methodology to fabricate tumor-targeted multi-modal nanotheranostic agents, providing great opportunity for precision imaging and cancer therapy.
Collapse
Affiliation(s)
- Longguang Tang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361005, China
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Xiaoli Sun
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Nian Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361005, China
| | - Zijian Zhou
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Fei Yu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361005, China
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361005, China
- Corresponding Authors: , ,
| | - Xiaolian Sun
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
- Corresponding Authors: , ,
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland 20892, United States
- Corresponding Authors: , ,
| |
Collapse
|
36
|
Kong X, Dong B, Song X, Wang C, Zhang N, Lin W. Dual turn-on fluorescence signal-based controlled release system for real-time monitoring of drug release dynamics in living cells and tumor tissues. Theranostics 2018; 8:800-811. [PMID: 29344307 PMCID: PMC5771094 DOI: 10.7150/thno.21577] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 11/18/2017] [Indexed: 01/24/2023] Open
Abstract
Controlled release systems with capabilities for direct and real-time monitoring of the release and dynamics of drugs in living systems are of great value for cancer chemotherapy. Herein, we describe a novel dual turn-on fluorescence signal-based controlled release system (CDox), in which the chemotherapy drug doxorubicin (Dox) and the fluorescent dye (CH) are conjugated by a hydrazone moiety, a pH-responsive cleavable linker. CDox itself shows nearly no fluorescence as the fluorescence of CH and Dox is essentially quenched by the C=N isomerization and N-N free rotation. However, when activated under acidic conditions, CDox could be hydrolyzed to afford Dox and CH, resulting in dual turn-on signals with emission peaks at 595 nm and 488 nm, respectively. Notably, CDox exhibits a desirable controlled release feature as the hydrolysis rate is limited by the steric hindrance effect from both the Dox and CH moieties. Cytotoxicity assays indicate that CDox shows much lower cytotoxicity relative to Dox, and displays higher cell inhibition rate to cancer than normal cells. With the aid of the dual turn-on fluorescence at different wavelengths, the drug release dynamics of CDox in living HepG2 and 4T-1 cells was monitored in double channels in a real-time fashion. Importantly, two-photon fluorescence imaging of CDox in living tumor tissues was also successfully performed by high-definition 3D imaging. We expect that the unique controlled release system illustrated herein could provide a powerful means to investigate modes of action of drugs, which is critical for development of much more robust and effective chemotherapy drugs.
Collapse
|
37
|
Comegna D, Zannetti A, Del Gatto A, de Paola I, Russo L, Di Gaetano S, Liguoro A, Capasso D, Saviano M, Zaccaro L. Chemical Modification for Proteolytic Stabilization of the Selective α vβ 3 Integrin RGDechi Peptide: in Vitro and in Vivo Activities on Malignant Melanoma Cells. J Med Chem 2017; 60:9874-9884. [PMID: 29144748 DOI: 10.1021/acs.jmedchem.7b01590] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Herein, we report the synthesis and biological characterization of the new peptide ψRGDechi as the first step toward novel-targeted theranostics in melanoma. This pseudopeptide is designed from our previously reported RGDechi peptide, known to bind selectively αvβ3 integrin, and differs for a modified amide bond at the main protease cleavage site. This chemical modification drastically reduces the enzymatic degradation in serum, compared to its parental peptide, resulting in an overall magnification of the biological activity on a highly expressing αvβ3 human metastatic melanoma cell line. Selective inhibition of cell adhesion, wound healing, and invasion are demonstrated; near-infrared fluorescent ψRGDechi derivative is able to detect αvβ3 integrin in human melanoma xenografts in a selective fashion. More, molecular docking studies confirm that ψRGDechi recognizes the receptor similarly to RGDechi. All these findings pave the way for the future employment of this novel peptide as promising targeting probe and therapeutic agent in melanoma disease.
Collapse
Affiliation(s)
- Daniela Comegna
- Institute of Biostructures and Bioimaging-CNR , Via Mezzocannone 16, 80134 Naples, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging-CNR , Via De Amicis 95, 80145 Naples, Italy
| | - Annarita Del Gatto
- Institute of Biostructures and Bioimaging-CNR , Via Mezzocannone 16, 80134 Naples, Italy.,Interdepartmental Center of Bioactive Peptide, University of Naples Federico II , Via Mezzocannone 16, 80134 Naples, Italy
| | - Ivan de Paola
- Institute of Biostructures and Bioimaging-CNR , Via Mezzocannone 16, 80134 Naples, Italy
| | - Luigi Russo
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania Luigi Vanvitelli , via Vivaldi 43, 81100 Caserta, Italy
| | - Sonia Di Gaetano
- Institute of Biostructures and Bioimaging-CNR , Via Mezzocannone 16, 80134 Naples, Italy.,Interdepartmental Center of Bioactive Peptide, University of Naples Federico II , Via Mezzocannone 16, 80134 Naples, Italy
| | - Annamaria Liguoro
- Institute of Biostructures and Bioimaging-CNR , Via Mezzocannone 16, 80134 Naples, Italy
| | - Domenica Capasso
- Department of Pharmacy, University of Naples Federico II , Via Mezzocannone 16, 80134 Naples, Italy
| | - Michele Saviano
- Interdepartmental Center of Bioactive Peptide, University of Naples Federico II , Via Mezzocannone 16, 80134 Naples, Italy.,Institute of Crystallography-CNR , Via Amendola 122/O, 70126 Bari, Italy
| | - Laura Zaccaro
- Institute of Biostructures and Bioimaging-CNR , Via Mezzocannone 16, 80134 Naples, Italy.,Interdepartmental Center of Bioactive Peptide, University of Naples Federico II , Via Mezzocannone 16, 80134 Naples, Italy
| |
Collapse
|
38
|
Molecular Ultrasound Imaging of αvβ3-Integrin Expression in Carotid Arteries of Pigs After Vessel Injury. Invest Radiol 2017; 51:767-775. [PMID: 27119438 DOI: 10.1097/rli.0000000000000282] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Interventions such as balloon angioplasty can cause vascular injury leading to platelet activation, thrombus formation, and inflammatory response. This induces vascular smooth muscle cell activation and subsequent re-endothelialization with expression of αvβ3-integrin by endothelial cells and vascular smooth muscle cell. Thus, poly-N-butylcyanoacrylate microbubbles (MBs) targeted to αvβ3-integrin were evaluated for monitoring vascular healing after vessel injury in pigs using molecular ultrasound imaging. MATERIALS AND METHODS Approval for animal experiments was obtained. The binding specificity of αvβ3-integrin-targeted MB to human umbilical vein endothelial cells was tested with fluorescence microscopy. In vivo imaging was performed using a clinical ultrasound system and an 8-MHz probe. Six mini pigs were examined after vessel injury in the left carotid artery. The right carotid served as control. Uncoated MB, cDRG-coated MB, and αvβ3-integrin-specific cRGD-coated MB were injected sequentially. Bound MBs were assessed 8 minutes after injection using ultrasound replenishment analysis. Measurements were performed 2 hours, 1 and 5 weeks, and 3 and 6 months after injury. In vivo data were validated by immunohistochemistry. RESULTS Significantly stronger binding of cRGD-MB than MB and cDRG-MB to human umbilical vein endothelial cells was found (P < 0.01). As vessel injury leads to upregulation of αvβ3-integrin, cRGD-MBs bound significantly stronger (P < 0.05) in injured carotid arteries than at the counter side 1 week after vessel injury and significant differences could also be observed after 5 weeks. After 3 months, αvβ3-integrin expression decreased to baseline and binding of cRGD-MB was comparable in both vessels. Values remained at baseline also after 6 months. CONCLUSIONS Ultrasound imaging with RGD-MB is promising for monitoring vascular healing after vessel injury. This may open new perspectives to assess vascular damage after radiological interventions.
Collapse
|
39
|
Zheng F, Zhang P, Xi Y, Chen X, He Z, Meng T, Chen J, Li L, Zhu JJ. Hierarchical Nanocarriers for Precisely Regulating the Therapeutic Process via Dual-Mode Controlled Drug Release in Target Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:36655-36664. [PMID: 28975792 DOI: 10.1021/acsami.7b12251] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A precisely controlled drug release is a great challenge in exploring methodologies of drug administration and fighting drug resistance for successful cancer chemotherapy. Herein, we developed a dual-mode nanocarrier to specifically deliver doxorubicin (Dox) and precisely control the drug release in target tumor cells. This hierarchical nanocarrier consisted of a gold nanorod as the heating core, biodegradable mesoporous silica as the storage chamber, and graphene quantum dot (GQD) as a drug carrier. The Arg-Gly-Asp peptides on the nanocarrier surface facilitated the specific interaction with integrin-overexpressed tumor cells and subsequent uptake via receptor-mediated endocytosis. Once exposed under the near-infrared (NIR) laser, the internalized nanocarrier rapidly heated the surrounding environment, which led to an instantaneous drug release by collapsing the π-π interaction between Dox and GQDs at high temperature and thereby intensified therapeutic efficacy. On the other hand, the silica shells underwent gradual degradation in the cellular matrix environment, along with stepwise liberation of the embedded GQD-Dox composites from the confined porous structure for the Dox release, exerting a long-term lethality to the tumor cells. By virtue of the physicochemical properties and synergistic behavior of the multiple components in this hierarchical nanocarrier, the NIR-triggered prompt release mode and the biodegradation-mediated slow release mode functioned in a precise and collaborative fashion, providing a promising way to manipulate the pharmacokinetics for precise cancer treatment.
Collapse
Affiliation(s)
- Fenfen Zheng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210093, People's Republic of China
| | - Penghui Zhang
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an 710049, People's Republic of China
| | - Yu Xi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210093, People's Republic of China
| | - Xueqin Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210093, People's Republic of China
| | - Zhimei He
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210093, People's Republic of China
| | - Tiantian Meng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210093, People's Republic of China
| | - Jingjia Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210093, People's Republic of China
| | - Lingling Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210093, People's Republic of China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210093, People's Republic of China
| |
Collapse
|
40
|
Staderini M, Megia-Fernandez A, Dhaliwal K, Bradley M. Peptides for optical medical imaging and steps towards therapy. Bioorg Med Chem 2017; 26:2816-2826. [PMID: 29042225 DOI: 10.1016/j.bmc.2017.09.039] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/22/2017] [Accepted: 09/29/2017] [Indexed: 12/20/2022]
Abstract
Optical medical imaging is a rapidly growing area of research and development that offers a multitude of healthcare solutions both diagnostically and therapeutically. In this review, some of the most recently described peptide-based optical probes are reviewed with a special emphasis on their in vivo use and potential application in a clinical setting.
Collapse
Affiliation(s)
- Matteo Staderini
- School of Chemistry, EaStChem, University of Edinburgh, Joseph Black Building, David Brewster Road, Edinburgh EH9 3FJ, UK
| | - Alicia Megia-Fernandez
- School of Chemistry, EaStChem, University of Edinburgh, Joseph Black Building, David Brewster Road, Edinburgh EH9 3FJ, UK
| | - Kevin Dhaliwal
- EPSRC IRC Proteus Hub, MRC Centre of Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Mark Bradley
- School of Chemistry, EaStChem, University of Edinburgh, Joseph Black Building, David Brewster Road, Edinburgh EH9 3FJ, UK; EPSRC IRC Proteus Hub, MRC Centre of Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
41
|
Zhang X, Wang B, Zhao N, Tian Z, Dai Y, Nie Y, Tian J, Wang Z, Chen X. Improved Tumor Targeting and Longer Retention Time of NIR Fluorescent Probes Using Bioorthogonal Chemistry. Am J Cancer Res 2017; 7:3794-3802. [PMID: 29109777 PMCID: PMC5667349 DOI: 10.7150/thno.20912] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 07/15/2017] [Indexed: 01/27/2023] Open
Abstract
The traditional labeling method for targeted NIR fluorescence probes requires directly covalent-bonded conjugation of targeting domains and fluorophores in vitro. Although this strategy works well, it is not sufficient for detecting or treating cancers in vivo, due to steric hindrance effects that relatively large fluorophore molecules exert on the configurations and physiological functions of specific targeting domains. The copper-free, “click-chemistry”-assisted assembly of small molecules in living systems may enhance tumor accumulation of fluorescence probes by improving the binding affinities of the targeting factors. Here, we employed a vascular homing peptide, GEBP11, as a targeting factor for gastric tumors, and we demonstrate its effectiveness for in vivo imaging via click-chemistry-mediated conjugation with fluorescence molecules in tumor xenograft mouse models. This strategy showed higher binding affinities than those of the traditional conjugation method, and our results showed that the tumor accumulation of click-chemistry-mediated probes are 11-fold higher than that of directly labeled probes. The tracking life was prolonged by 12-fold, and uptake of the probes into the kidney was reduced by 6.5-fold. For lesion tumors of different sizes, click-chemistry-mediated probes can achieve sufficient signal-to-background ratios (3.5-5) for in vivo detection, and with diagnostic sensitivity approximately 3.5 times that of traditional labeling probes. The click-chemistry-assisted detection strategy utilizes the advantages of “small molecule” probes while not perturbing their physiological functions; this enables tumor detection with high sensitivity and specific selectivity.
Collapse
|
42
|
Haedicke K, Brand C, Omar M, Ntziachristos V, Reiner T, Grimm J. Sonophore labeled RGD: a targeted contrast agent for optoacoustic imaging. PHOTOACOUSTICS 2017; 6:1-8. [PMID: 28393018 PMCID: PMC5376267 DOI: 10.1016/j.pacs.2017.03.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/20/2017] [Accepted: 03/01/2017] [Indexed: 05/10/2023]
Abstract
Optoacoustic imaging is a rapidly expanding field for the diagnosis, characterization, and treatment evaluation of cancer. However, the availability of tumor specific exogenous contrast agents is still limited. Here, we report on a small targeted contrast agent for optoacoustic imaging using a black hole quencher® (BHQ) dye. The sonophore BHQ-1 exhibited strong, concentration-dependent, optoacoustic signals in phantoms, demonstrating its ideal suitability for optoacoustic imaging. After labeling BHQ-1 with cyclic RGD-peptide, BHQ-1-cRGD specifically bound to αvβ3-integrin expressing glioblastoma cell spheroids in vitro. The excellent optoacoustic properties of BHQ-1-cRGD could furthermore be proven in vivo. Together with this emerging imaging modality, our sonophore labeled small peptide probe offers new possibilities for non-invasive detection of molecular structures with high resolution in vivo and furthers the specificity of optoacoustic imaging. Ultimately, the discovery of tailor-made sonophores might offer new avenues for various molecular optoacoustic imaging applications, similar to what we see with fluorescence imaging.
Collapse
Affiliation(s)
- Katja Haedicke
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christian Brand
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Murad Omar
- Chair of Biological Imaging, Technische Universitaet Muenchen, Munich, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Vasilis Ntziachristos
- Chair of Biological Imaging, Technische Universitaet Muenchen, Munich, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jan Grimm
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, Pharmacology Program and Department of Radiology
- Corresponding author. 1275 York Ave, Box 248, New York, NY 10065.
| |
Collapse
|
43
|
Abstract
Conjugates of cytotoxic agents with RGD peptides (Arg-Gly-Asp) addressed to ανβ3, α5β1 and ανβ6 integrin receptors overexpressed by cancer cells, have recently gained attention as potential selective anticancer chemotherapeutics. In this review, the design and the development of RGD conjugates coupled to different small molecules including known cytotoxic drugs and natural products will be discussed.
Collapse
|
44
|
Vahabi S, Eatemadi A. Nanoliposome encapsulated anesthetics for local anesthesia application. Biomed Pharmacother 2017; 86:1-7. [DOI: 10.1016/j.biopha.2016.11.137] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/28/2016] [Accepted: 11/28/2016] [Indexed: 12/14/2022] Open
|
45
|
Optical imaging of MMP-12 active form in inflammation and aneurysm. Sci Rep 2016; 6:38345. [PMID: 27917892 PMCID: PMC5137160 DOI: 10.1038/srep38345] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 11/08/2016] [Indexed: 01/08/2023] Open
Abstract
Matrix metalloproteinase (MMP)-12 plays a key role in the development of aneurysm. Like other members of MMP family, MMP-12 is produced as a proenzyme, mainly by macrophages, and undergoes proteolytic activation to generate an active form. Accordingly, molecular imaging of the MMP-12 active form can inform of the pathogenic process in aneurysm. Here, we developed a novel family of fluorescent probes based on a selective MMP-12 inhibitor, RXP470.1 to target the active form of MMP-12. These probes were stable in complex media and retained the high affinity and selectivity of RXP470.1 for MMP-12. Amongst these, probe 3 containing a zwitterionic fluorophore, ZW800-1, combined a favorable affinity profile toward MMP-12 and faster blood clearance. In vivo binding of probe 3 was observed in murine models of sterile inflammation and carotid aneurysm. Binding specificity was demonstrated using a non-binding homolog. Co-immunostaining localized MMP-12 probe binding to MMP-12 positive areas and F4/80 positive macrophages in aneurysm. In conclusion, the active form of MMP-12 can be detected by optical imaging using RXP470.1-based probes. This is a valuable adjunct for pathophysiology research, drug development, and potentially clinical applications.
Collapse
|
46
|
Xu F, Liu J, Tian J, Gao L, Cheng X, Pan Y, Sun Z, Li X. Supramolecular Self-Assemblies with Nanoscale RGD Clusters Promote Cell Growth and Intracellular Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2016; 8:29906-29914. [PMID: 27759366 DOI: 10.1021/acsami.6b08624] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
In this work, we reported the generation of a novel supramolecular hydrogelator from a peptide derivative which consisted of a structural motif (e.g., Fc-FF) for supramolecular self-assembly and a functional moiety (e.g., RGD) for integrin binding. Following self-assembly in water at neutral pH, this molecule first tended to form metastable spherical aggregates, which subsequently underwent a morphological transformation to form high-aspect-ratio nanostructures over 2 h when aged at room temperature. More importantly, because of the presence of nanoscale RGD clusters on the surface of nanostructures, the self-assembled nanomaterials (e.g., nanoparticles and nanofibers) can be potentially used as a biomimetic matrix for cell culture and as a vector for cell-targeting drug delivery via multivalent RGD-integrin interactions.
Collapse
Affiliation(s)
- Fengyang Xu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, China
| | - Jie Liu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, China
| | - Jian Tian
- School of Biology and Basic Medical Science, Soochow University , Suzhou 215123, China
| | - Linfeng Gao
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, China
| | - Xiaju Cheng
- School of Biology and Basic Medical Science, Soochow University , Suzhou 215123, China
| | - Yue Pan
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, China
| | - Ziling Sun
- School of Biology and Basic Medical Science, Soochow University , Suzhou 215123, China
| | - Xinming Li
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University , Shanghai 200433, China
| |
Collapse
|
47
|
Progress of Multimodal Molecular Imaging Technology in Diagnosis of Tumor. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2016. [DOI: 10.1016/s1872-2040(16)60966-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
48
|
Bordenave T, Helle M, Beau F, Georgiadis D, Tepshi L, Bernes M, Ye Y, Levenez L, Poquet E, Nozach H, Razavian M, Toczek J, Stura EA, Dive V, Sadeghi MM, Devel L. Synthesis and in Vitro and in Vivo Evaluation of MMP-12 Selective Optical Probes. Bioconjug Chem 2016; 27:2407-2417. [PMID: 27564088 DOI: 10.1021/acs.bioconjchem.6b00377] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In designing new tracers consisting of a small peptide conjugated to a reporter of comparable size, particular attention needs to be paid to the selection of the reporter group, which can dictate both the in vitro and the in vivo performances of the whole conjugate. In the case of fluorescent tracers, this is particularly true given the large numbers of available dye moieties differing in their structures and properties. Here, we have investigated the in vitro and in vivo properties of a novel series of MMP-12 selective probes composed of cyanine dyes varying in their structure, net charge, and hydrophilic character, tethered through a linker to a potent and specific MMP-12 phosphinic pseudopeptide inhibitor. The impact of linker length has been also explored. The crystallographic structure of one tracer in complex with MMP-12 has been obtained, providing the first crystal structure of a Cy5.5-derived probe and confirming that the binding of the targeting moiety is unaffected. MMP-12 remains the tracers' privileged target, as attested by their affinity selectivity profile evaluated in solution toward a panel of 12 metalloproteases. In vivo assessment of four selected probes has highlighted not only the impact of the dye structure but also that of the linker length on the probes' blood clearance rates and their biodistributions. These experiments have also provided valuable data on the stability of the dye moieties in vivo. This has permitted the identification of one probe, which combines favorable binding to MMP-12 in solution and on cells with optimized in vivo performance including blood clearance rate suitable for short-time imaging. Through this series of tracers, we have identified various critical factors modulating the tracers' in vivo behavior, which is both useful for the development and optimization of MMP-12 selective radiolabeled tracers and informative for the design of fluorescent probes in general.
Collapse
Affiliation(s)
- Thomas Bordenave
- Service d'ingénierie moléculaire des protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay , Gif-sur-Yvette F-91191, France
| | - Marion Helle
- Service d'ingénierie moléculaire des protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay , Gif-sur-Yvette F-91191, France
| | - Fabrice Beau
- Service d'ingénierie moléculaire des protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay , Gif-sur-Yvette F-91191, France
| | - Dimitris Georgiadis
- Department of Chemistry, Laboratory of Organic Chemistry, University of Athens , Panepistimiopolis, Zografou, Athens 15771, Greece
| | - Livia Tepshi
- Service d'ingénierie moléculaire des protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay , Gif-sur-Yvette F-91191, France
| | - Mylène Bernes
- Service d'ingénierie moléculaire des protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay , Gif-sur-Yvette F-91191, France
| | - Yunpeng Ye
- Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine and Yale Cardiovascular Research Center, Yale University School of Medicine , New Haven, Connecticut 06511, United States.,Veterans Affairs Connecticut Healthcare System , West Haven, Connecticut 06516, United States
| | - Laure Levenez
- Service d'ingénierie moléculaire des protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay , Gif-sur-Yvette F-91191, France
| | - Enora Poquet
- Service d'ingénierie moléculaire des protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay , Gif-sur-Yvette F-91191, France
| | - Hervé Nozach
- Service d'ingénierie moléculaire des protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay , Gif-sur-Yvette F-91191, France
| | - Mahmoud Razavian
- Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine and Yale Cardiovascular Research Center, Yale University School of Medicine , New Haven, Connecticut 06511, United States.,Veterans Affairs Connecticut Healthcare System , West Haven, Connecticut 06516, United States
| | - Jakub Toczek
- Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine and Yale Cardiovascular Research Center, Yale University School of Medicine , New Haven, Connecticut 06511, United States.,Veterans Affairs Connecticut Healthcare System , West Haven, Connecticut 06516, United States
| | - Enrico A Stura
- Service d'ingénierie moléculaire des protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay , Gif-sur-Yvette F-91191, France
| | - Vincent Dive
- Service d'ingénierie moléculaire des protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay , Gif-sur-Yvette F-91191, France
| | - Mehran M Sadeghi
- Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine and Yale Cardiovascular Research Center, Yale University School of Medicine , New Haven, Connecticut 06511, United States.,Veterans Affairs Connecticut Healthcare System , West Haven, Connecticut 06516, United States
| | - Laurent Devel
- Service d'ingénierie moléculaire des protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay , Gif-sur-Yvette F-91191, France
| |
Collapse
|
49
|
Li Y, Li Y, Zhang X, Xu X, Zhang Z, Hu C, He Y, Gu Z. Supramolecular PEGylated Dendritic Systems as pH/Redox Dual-Responsive Theranostic Nanoplatforms for Platinum Drug Delivery and NIR Imaging. Theranostics 2016; 6:1293-305. [PMID: 27375780 PMCID: PMC4924500 DOI: 10.7150/thno.15081] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 04/27/2016] [Indexed: 01/10/2023] Open
Abstract
Recently, self-assembling small dendrimers into supramolecular dendritic systems offers an alternative strategy to develop multifunctional nanoplatforms for biomedical applications. We herein report a dual-responsive supramolecular PEGylated dendritic system for efficient platinum-based drug delivery and near-infrared (NIR) tracking. With a refined molecular/supramolecular engineering, supramolecular dendritic systems were stabilized by bioreducible disulfide bonds and endowed with NIR fluorescence probes, and PEGylated platinum derivatives coordinated onto the abundant peripheral groups of supramolecular dendritic templates to generate pH/redox dual-responsive theranostic supramolecular PEGylated dendritic systems (TSPDSs). TSPDSs markedly improved the pharmacokinetics and biodistribution of platinum-based drugs, owing to their stable nanostructures and PEGylated shells during the blood circulation. Tumor intracellular environment (low pH value and high glutathione concentration) could trigger the rapid disintegration of TSPDSs due to acid-labile coordination bonds and redox-cleavable disulfide linkages, and then platinum-based drugs were delivered into the nuclei to exert antitumor activity. In vivo antitumor treatments indicated TSPDSs not only provided high antitumor efficiency which was comparable to clinical cisplatin, but also reduced renal toxicity of platinum-based drugs. Moreover, NIR fluorescence of TSPDSs successfully visualized in vitro and in vivo fate of nanoplatforms and disclosed the intracellular platinum delivery and pharmacokinetics. These results confirm tailor-made supramolecular dendritic system with sophisticated nanostructure and excellent performance is a promising candidate as smart theranostic nanoplatforms.
Collapse
Affiliation(s)
| | | | | | - Xianghui Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, Sichuan, China
| | | | | | | | - Zhongwei Gu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, Sichuan, China
| |
Collapse
|
50
|
Bunschoten A, van Willigen DM, Buckle T, van den Berg NS, Welling MM, Spa SJ, Wester HJ, van Leeuwen FWB. Tailoring Fluorescent Dyes To Optimize a Hybrid RGD-Tracer. Bioconjug Chem 2016; 27:1253-1258. [PMID: 27074375 DOI: 10.1021/acs.bioconjchem.6b00093] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Quantitative assessment of affinity and kinetics is a critical component in the development of (receptor-targeted) radiotracers. For fluorescent tracers, such an assessment is currently not yet applied, while (small) changes in chemical composition of the fluorescent component might have substantial influence on the overall properties of a fluorescent tracer. Hybrid imaging labels that contain both a radiolabel and a fluorescent dye can be used to evaluate both the affinity (fluorescent label) and the in vivo distribution (radiolabel) of a targeted tracer. We present a hybrid label oriented and matrix-based scoring approach that enabled quantitative assessment of the influence of (overall) charge and lipophilicity of the fluorescent label on the (in vivo) characteristics of αvβ3-integrin targeted tracers. Systematic chemical alterations in the fluorescent dye were shown to result in a clear difference in the in vivo distribution of the different hybrid tracers. The applied evaluation technique resulted in an optimized targeted tracer for αvβ3-integrin, which combined the highest T/M ratio with the lowest uptake in other organs. Obviously this selection concept would also be applicable during the development of other (receptor-targeted) imaging tracers.
Collapse
Affiliation(s)
- Anton Bunschoten
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center , Albinusdreef 2, PO BOX 9600, 2300 RC, Leiden, The Netherlands
| | - Danny M van Willigen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center , Albinusdreef 2, PO BOX 9600, 2300 RC, Leiden, The Netherlands
| | - Tessa Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center , Albinusdreef 2, PO BOX 9600, 2300 RC, Leiden, The Netherlands
| | - Nynke S van den Berg
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center , Albinusdreef 2, PO BOX 9600, 2300 RC, Leiden, The Netherlands
- Departments of Urology and Head and Neck Surgery & Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital , Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Mick M Welling
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center , Albinusdreef 2, PO BOX 9600, 2300 RC, Leiden, The Netherlands
| | - Silvia J Spa
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center , Albinusdreef 2, PO BOX 9600, 2300 RC, Leiden, The Netherlands
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Faculties of Chemistry and Medicine, Technische Universität München , 85748 Garching, Germany
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center , Albinusdreef 2, PO BOX 9600, 2300 RC, Leiden, The Netherlands
- Departments of Urology and Head and Neck Surgery & Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital , Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|