期刊论文详细信息
BMC Cancer
Shortwave infrared emitting multicolored nanoprobes for biomarker-specific cancer imaging in vivo
Mark C. Pierce1  Michael Donzanti1  Vidya Ganapathy1  Shravani Barkund1  Daniel Martin1  Harini Kantamneni2  Charles M. Roth3  Prabhas V. Moghe3  Richard E. Riman4  Mei Chee Tan5  Xinyu Zhao5  Shuqing He5 
[1] Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, 08854, Piscataway, NJ, USA;Department of Chemical & Biochemical Engineering, Rutgers University, 98 Brett Road, 08854, Piscataway, NJ, USA;Department of Chemical & Biochemical Engineering, Rutgers University, 98 Brett Road, 08854, Piscataway, NJ, USA;Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, 08854, Piscataway, NJ, USA;Department of Materials Science and Engineering, Rutgers University, 607 Taylor Road, 08854, Piscataway, NJ, USA;Engineering Product Development, Singapore University of Technology and Design, 8 Somapah Rd, 487372, Singapore, Singapore;
关键词: Cancer metastasis;    Nanotechnology;    Short-wave infrared imaging;    Multiplexing;    Rare earths;   
DOI  :  10.1186/s12885-020-07604-8
来源: Springer
PDF
【 摘 要 】

BackgroundThe ability to detect tumor-specific biomarkers in real-time using optical imaging plays a critical role in preclinical studies aimed at evaluating drug safety and treatment response. In this study, we engineered an imaging platform capable of targeting different tumor biomarkers using a multi-colored library of nanoprobes. These probes contain rare-earth elements that emit light in the short-wave infrared (SWIR) wavelength region (900–1700 nm), which exhibits reduced absorption and scattering compared to visible and NIR, and are rendered biocompatible by encapsulation in human serum albumin. The spectrally distinct emissions of the holmium (Ho), erbium (Er), and thulium (Tm) cations that constitute the cores of these nanoprobes make them attractive candidates for optical molecular imaging of multiple disease biomarkers.MethodsSWIR-emitting rare-earth-doped albumin nanocomposites (ReANCs) were synthesized using controlled coacervation, with visible light-emitting fluorophores additionally incorporated during the crosslinking phase for validation purposes. Specifically, HoANCs, ErANCs, and TmANCs were co-labeled with rhodamine-B, FITC, and Alexa Fluor 647 dyes respectively. These Rh-HoANCs, FITC-ErANCs, and 647-TmANCs were further conjugated with the targeting ligands daidzein, AMD3100, and folic acid respectively. Binding specificities of each nanoprobe to distinct cellular subsets were established by in vitro uptake studies. Quantitative whole-body SWIR imaging of subcutaneous tumor bearing mice was used to validate the in vivo targeting ability of these nanoprobes.ResultsEach of the three ligand-functionalized nanoprobes showed significantly higher uptake in the targeted cell line compared to untargeted probes. Increased accumulation of tumor-specific nanoprobes was also measured relative to untargeted probes in subcutaneous tumor models of breast (4175 and MCF-7) and ovarian cancer (SKOV3). Preferential accumulation of tumor-specific nanoprobes was also observed in tumors overexpressing targeted biomarkers in mice bearing molecularly-distinct bilateral subcutaneous tumors, as evidenced by significantly higher signal intensities on SWIR imaging.ConclusionsThe results from this study show that tumors can be detected in vivo using a set of targeted multispectral SWIR-emitting nanoprobes. Significantly, these nanoprobes enabled imaging of biomarkers in mice bearing bilateral tumors with distinct molecular phenotypes. The findings from this study provide a foundation for optical molecular imaging of heterogeneous tumors and for studying the response of these complex lesions to targeted therapy.

【 授权许可】

CC BY   

【 预 览 】
附件列表
Files Size Format View
RO202104283889304ZK.pdf 3489KB PDF download
  文献评价指标  
  下载次数:3次 浏览次数:2次