PL EN


Preferencje help
Widoczny [Schowaj] Abstrakt
Liczba wyników
Tytuł artykułu

A literature review on multimodality molecular imaging nanoprobes for cancer detection

Identyfikatory
Warianty tytułu
Języki publikacji
EN
Abstrakty
EN
Molecular imaging techniques using nanoparticles have significant potential to be widely used for the detection of various types of cancers. Nowadays, there has been an increased focus on developing novel nanoprobes as molecular imaging contrast enhancement agents in nanobiomedicine. The purpose of this review article is to summarize the use of a variety of nanoprobes and their current achievements in accurate cancer imaging and effective treatment. Nanoprobes are rapidly becoming potential tools for cancer diagnosis by using novel molecular imaging modalities such as Ultrasound (US) imaging, Computerized Tomography (CT), Single Photon Emission Tomography (SPECT) and Positron Emission Tomography (PET), Magnetic Resonance Imaging (MRI), and Optical Imaging. These imaging modalities may facilitate earlier and more accurate diagnosis and staging the most of cancers.
Słowa kluczowe
Rocznik
Strony
57--68
Opis fizyczny
Bibliogr. 107 poz., rys.
Twórcy
  • Dept. of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan
  • School of Physics, Universiti Sains Malaysia, Malaysia
  • Dept. of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan
  • Dept. of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan
  • Dept. of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan
Bibliografia
  • [1] Hussain T, Nguyen QT. Molecular imaging for cancer diagnosis and surgery. Advanced Drug Delivery Reviews. 2014;66:90-100.
  • [2] Sun X, Li Y, Liu T, et al. Peptide-based imaging agents for cancer detection. Advanced Drug Delivery Reviews. 2017;110:38-51.
  • [3] Kircher MF, Hricak H, Larson SM. Molecular imaging for personalized cancer care. Molecular Oncology. 2012;6(2):182-195.
  • [4] Khaniabadi PM, Shahbazi-Gahrouei D, Jaafar MS, et al. Magnetic iron oxide nanoparticles as T2 MR imaging contrast agent for detection of breast cancer (MCF-7) cell. Avicenna Journal of Medical Biotechnology. 2017;9(4):181-188.
  • [5] Khaniabadi PM, Majid A, Asif M. et al. Breast cancer cell targeted MR molecular imaging probe: Anti-MUC1 antibody-based magnetic nanoparticles. Journal of Physics: Conference Series. 2017;851(1):012014.
  • [6] Mirzaei M, Mohagheghi M, Shahbazi-Gahrouei D. Synthesis and development of Gd3+-ALGDG2-C595 as MR Imaging Contrast Agent. Journal of Biomaterials and Nanobiotechnology. 2013;4(01):22.
  • [7] Khemthongcharoen N, Jolivot R, Rattanavarin S, Piyawattanametha W. Advances in imaging probes and optical microendoscopic imaging techniques for early in vivo cancer assessment. Advanced Drug Delivery Reviews. 2014;74:53-74.
  • [8] Savla R, Minko T. Nanoparticle design considerations for molecular imaging of apoptosis: Diagnostic, prognostic, and therapeutic value. Advanced Drug Delivery Reviews. 2017;113:122-140.
  • [9] Muthu MS, Leong DT, Mei L, Feng S-S. Nanotheranostics˗ application and further development of nanomedicine strategies for advanced theranostics. Theranostics. 2014;4(6):660-677.
  • [10] Guo R, Lu G, Qin B, Fei B. Ultrasound Imaging Technologies for Breast Cancer Detection and Management: A Review. Ultrasound in Medicine and Biology. 2018;44(1):37-70.
  • [11] Li N, Han L, Jing H. Contrast-enhanced ultrasound with a novel nanoparticle contrast agent for clinical diagnosis in patients with non‑small cell lung cancer. Experimental and Therapeutic Medicine. 2017;14(4):3768-3773.
  • [12] Paefgen V, Doleschel D, Kiessling F. Evolution of contrast agents for ultrasound imaging and ultrasound-mediated drug delivery. Frontiers in Pharmacology. 2015;6:197.
  • [13] Beik J, Shiran MB, Abed Z, et al. Gold nanoparticle‐induced sonosensitization enhances the antitumor activity of ultrasound in colon tumor‐bearing mice. Medical Physics. 2018;45(9):4306-4314.
  • [14] Milgroom A, Intrator M, Madhavan K, et al. Mesoporous silica nanoparticles as a breast-cancer targeting ultrasound contrast agent. Colloids and Surfaces B, Biointerfaces. 2014;116:652-657.
  • [15] Martins SF, Garcia EA, Luz MA, et al. Clinicopathological correlation and prognostic significance of VEGF-A, VEGF-C, VEGFR-2 and VEGFR-3 expression in colorectal cancer. Cancer Genomics & Proteomics. 2013;10(2):55-67.
  • [16] Abou-Elkacem L, Wilson KE, Johnson SM, et al. Ultrasound Molecular Imaging of the Breast Cancer Neovasculature using Engineered Fibronectin Scaffold Ligands: A Novel Class of Targeted Contrast Ultrasound Agent. Theranostics. 2016;6(11):1740-1752.
  • [17] Hao L, Wang Z, Luo J, et al. Preparation of targeted high molecular polymer microbubble encapsuling quantum dots and cell experiment study in vitro. Journal of Chongqing Medical University. 2013(1):24-27.
  • [18] Lutz AM, Bachawal SV, Drescher CW, et al. Ultrasound molecular imaging in a human CD276 expression-modulated murine ovarian cancer model. Clinical Cancer Research. 2014;20(5):1313-1322.
  • [19] Yoon YI, Pang X, Jung S, et al. Smart gold nanoparticle-stabilized ultrasound microbubbles as cancer theranostics. Journal of Materials Chemistry B. 2018;6(20):3235-3239.
  • [20] Hernot S, Klibanov AL. Microbubbles in ultrasound-triggered drug and gene delivery. Adv Drug Deliv Rev. 2008;60(10):1153-1166.
  • [21] Yildirim A, Shi D, Roy S, et al. Nanoparticle-Mediated Acoustic Cavitation Enables High Intensity Focused Ultrasound Ablation Without Tissue Heating. ACS Applied Materials & Interfaces. 2018;10(43):36786-36795.
  • [22] McLaughlan J, Cowell D, Freear S. Gold nanoparticle nucleated cavitation for enhanced high intensity focused ultrasound therapy. Physics in Medicine & Biology. 2017;63(1):015004.
  • [23] Nedaei TS, Delavari HH. Preparation of Naturally Active Melanin Nano‐Platforms Chelated with Barium Ions as a Potential X‐Ray‐Computed Tomography Contrast Agent. ChemistrySelect. 2018;3(39):11098-11102.
  • [24] Wallyn J, Anton N, Serra CA, et al. A new formulation of poly (MAOTIB) nanoparticles as an efficient contrast agent for in vivo Xray imaging. Acta Biomaterialia. 2018;66:200-212.
  • [25] Hainfeld JF, Ridwan SM, Stanishevskiy Y, et al. Small, Long Blood Half-Life Iodine Nanoparticle for Vascular and Tumor Imaging. Scientific Reports. 2018;8(1):13803.
  • [26] Goldman LW. Principles of CT and CT technology. J Nucl Med Technol. 2007;35(3):115-28; quiz 29-30.
  • [27] Kim D, Yu MK, Lee TS, et al. Amphiphilic polymer-coated hybrid nanoparticles as CT/MRI dual contrast agents. Nanotechnology. 2011;22(15):155101.
  • [28] Gu X, Liu R. Application of 18F-FDG PET/CT combined with carbohydrate antigen 19-9 for differentiating pancreatic carcinoma from chronic mass-forming pancreatitis in Chinese elderly. Clin Interv Aging. 2016;11:1365-1370.
  • [29] Zheng X, Wang S, Wu L, Hou X. Microwave-assisted facile synthesis of mono-dispersed Ba/Ho co-doped nanohydroxyapatite for potential application as binary CT imaging contrast agent. Microchemical Journal. 2018;141:330-336.
  • [30] Naha PC, Lau KC, Hsu JC, et al. Gold silver alloy nanoparticles (GSAN): an imaging probe for breast cancer screening with dualenergy mammography or computed tomography. Nanoscale. 2016;8(28):13740-13754.
  • [31] Shi X, Wang SH, Van Antwerp ME, et al. Targeting and detecting cancer cells using spontaneously formed multifunctional dendrimer-stabilized gold nanoparticles. The Analyst. 2009;134(7):1373-1379.
  • [32] Eck W, Nicholson AI, Zentgraf H, et al. Anti-CD4-targeted gold nanoparticles induce specific contrast enhancement of peripheral lymph nodes in X-ray computed tomography of live mice. Nano Letters. 2010;10(7):2318-2322.
  • [33] Zhang Z, Ross RD, Roeder RK. Preparation of functionalized gold nanoparticles as a targeted X-ray contrast agent for damaged bone tissue. Nanoscale. 2010;2(4):582-586.
  • [34] Aydogan B, Li J, Rajh T, et al. AuNP-DG: Deoxyglucose-labeled gold nanoparticles as X-ray computed tomography contrast agents for cancer imaging. Molecular Imaging and Biology. 2010;12(5):463-467.
  • [35] Zhou B, Yang J, Peng C, et al. PEGylated polyethylenimine-entrapped gold nanoparticles modified with folic acid for targeted tumor CT imaging. Colloids and surfaces B, Biointerfaces. 2016;140:489-496.
  • [36] Khademi S, Sarkar S, Shakeri-Zadeh A, et al. Folic acid-cysteamine modified gold nanoparticle as a nanoprobe for targeted computed tomography imaging of cancer cells. Materials Science and Engineering: C. 2018;89:182-193.
  • [37] Zhang Y, Wen S, Zhao L, et al. Ultrastable polyethyleneimine-stabilized gold nanoparticles modified with polyethylene glycol for blood pool, lymph node and tumor CT imaging. Nanoscale. 2016;8(10):5567-5577.
  • [38] Nakagawa T, Gonda K, Kamei T, et al. X-ray computed tomography imaging of a tumor with high sensitivity using gold nanoparticles conjugated to a cancer-specific antibody via polyethylene glycol chains on their surface. Science and Technology of Advanced Materials. 2016;17(1):387-297.
  • [39] Kobayashi Y, Shibuya K, Tokunaga M, et al. Preparation of high-concentration colloidal solution of silica-coated gold nanoparticles and their application to X-ray imaging. Journal of Sol-Gel Science and Technology. 2016;78(1):82-90.
  • [40] Cole LE, Ross RD, Tilley JM, et al. Gold nanoparticles as contrast agents in X-ray imaging and computed tomography. Nanomedicine. 2015;10(2):321-341.
  • [41] Jing L, Liang X, Deng Z, et al. Prussian blue coated gold nanoparticles for simultaneous photoacoustic/CT bimodal imaging and photothermal ablation of cancer. Biomaterials. 2014;35(22):5814-5821.
  • [42] Zhang J, Li C, Zhang X, et al. In vivo tumor-targeted dual-modal fluorescence/CT imaging using a nanoprobe co-loaded with an aggregation-induced emission dye and gold nanoparticles. Biomaterials. 2015;42:103-111.
  • [43] Bi H, He F, Dong Y, et al. Bismuth Nanoparticles with “Light” Property Served as a Multifunctional Probe for X-ray Computed Tomography and Fluorescence Imaging. Chemistry of Materials. 2018;30(10):3301-3307.
  • [44] Fazel-Ghaziyani M, Shahbazi-Gahrouei D, Pourhassan-Moghaddam M, et al. Targeted detection of the cancer cells using the anti-CD24 bio modified PEGylated gold nanoparticles: the application of CD24 as a vital cancer biomarker. Nanomedicine J. 2018;5(3):172-179.
  • [45] Tang D, Gao W, Yuan Y, et al. Novel Biocompatible Au Nanostars@PEG Nanoparticles for In Vivo CT Imaging and Renal Clearance Properties. Nanoscale Research Letters. 2017;12(1):565.
  • [46] Yu N, Wang Z, Zhang J, et al. Thiol-capped Bi nanoparticles as stable and all-in-one type theranostic nanoagents for tumor imaging and thermoradiotherapy. Biomaterials. 2018;161:279-291.
  • [47] Wu B, Lu S-T, Yu H, et al. Gadolinium-chelate functionalized bismuth nanotheranostic agent for in vivo MRI/CT/PAI imagingguided photothermal cancer therapy. Biomaterials. 2018;159:37-47.
  • [48] Wang Y, Cai D, Wu H, et al. Functionalized Cu 3 BiS 3 nanoparticles for dual-modal imaging and targeted photothermal/photodynamic therapy. Nanoscale. 2018;10(9):4452-1162.
  • [49] Kaufmann PA, Di Carli MF, editors. Hybrid SPECT/CT and PET/CT imaging: the next step in noninvasive cardiac imaging. Seminars in nuclear medicine; 2009: Elsevier.
  • [50] Antoch G, Bockisch A. Combined PET/MRI: a new dimension in whole-body oncology imaging? European Journal of Nuclear Medicine and Molecular Imaging. 2009;36(Suppl1):113-120.
  • [51] Giovacchini G, Picchio M, Garcia-Parra R, et al. 11C-choline PET/CT predicts prostate cancer–specific survival in patients with biochemical failure during androgen-deprivation therapy. Journal of Nuclear Medicine. 2014;55(2):233-241.
  • [52] Takenaka D, Ohno Y, Koyama H, et al. Integrated FDG-PET/CT vs. standard radiological examinations: comparison of capability for assessment of postoperative recurrence in non-small cell lung cancer patients. European Journal of Radiology. 2010;74(3):458-464.
  • [53] Buck AK, Herrmann K, Stargardt T, et al. Economic evaluation of PET and PET/CT in oncology: evidence and methodologic approaches. Journal of Nuclear Medicine Technology. 2010;38(1):6-17.
  • [54] Miyazaki T, Sohda M, Higuchi T, et al. Effectiveness of FDG-PET in screening of synchronous cancer of other organs in patients with esophageal cancer. Anticancer Research. 2014;34(1):283-287.
  • [55] Khan N, Oriuchi N, Higuchi T, et al. PET in the follow-up of differentiated thyroid cancer. British Journal of Radiology. 2003;76(910):690-695.
  • [56] Skovgaard D, Persson M, Brandt-Larsen M, et al. Safety, dosimetry, and tumor detection ability of 68Ga-NOTA-AE105: First-inhuman study of a novel radioligand for uPAR PET imaging. Journal of Nuclear Medicine. 2017;58(3):379-386.
  • [57] McDonagh PR, Sundaresan G, Yang L, et al. Biodistribution and PET imaging of 89-zirconium labeled cerium oxide nanoparticles synthesized with several surface coatings. Nanomedicine: Nanotechnology, Biology and Medicine. 2018;14(4):1429-1440.
  • [58] Berke S, Kampmann A-L, Wuest M, et al. 18F-Radiolabeling and In Vivo Analysis of SiFA-Derivatized Polymeric Core–Shell Nanoparticles. Bioconjugate Chemistry. 2017;29(1):89-95.
  • [59] Rowe SP, Gage KL, Faraj SF, et al. 18F-DCFBC PET/CT for PSMA-based detection and characterization of primary prostate cancer. Journal of Nuclear Medicine. 2015;56(7):1003-1010.
  • [60] Rainone P, Riva B, Belloli S, et al. Development of 99mTc-radiolabeled nanosilica for targeted detection of her2-positive breast cancer. International Journal of Nanomedicine. 2017;12:3447-3461.
  • [61] Polyak A, Nagy LN, Mihaly J, et al. Preparation and 68Ga-radiolabeling of porous zirconia nanoparticle platform for PET/CTimaging guided drug delivery. Journal of Pharmaceutical and Biomedical Analysis. 2017;137:146-150.
  • [62] Pascual L, Cerqueira-Coutinho C, García-Fernández A, et al. MUC1 aptamer-capped mesoporous silica nanoparticles for controlled drug delivery and radio-imaging applications. Nanomedicine: Nanotechnology, Biology and Medicine. 2017;13(8):2495-2505.
  • [63] Norregaard K, Jørgensen JT, Simón M, et al. 18F-FDG PET/CT-based early treatment response evaluation of nanoparticle-assisted photothermal cancer therapy. PloS one. 2017;12(5):e0177997.
  • [64] Cassidy PJ, Radda GK. Molecular imaging perspectives. Journal of the Royal Society Interface. 2005;2(3):133-144.
  • [65] Szigeti K, Hegedűs N, Rácz K, et al. Thallium Labeled Citrate-Coated Prussian Blue Nanoparticles as Potential Imaging Agent. Contrast Media and Molecular Imaging. 2018;2018:2023604.
  • [66] de Souza Albernaz M, Toma SH, Clanton J, et al. Decorated Superparamagnetic Iron Oxide Nanoparticles with Monoclonal Antibody and Diethylene-Triamine-Pentaacetic Acid Labeled with Thechnetium-99m and Galium-68 for Breast Cancer Imaging. Pharmaceutical Research. 2018;35(1):24.
  • [67] Thomas R, Park I-K, Jeong Y. Magnetic iron oxide nanoparticles for multimodal imaging and therapy of cancer. International Journal of Molecular Sciences. 2013;14(8):15910-15930.
  • [68] Misri R, Meier D, Yung AC, et al. Development and evaluation of a dual-modality (MRI/SPECT) molecular imaging bioprobe. Nanomedicine: Nanotechnology, Biology and Medicine. 2012;8(6):1007-1016.
  • [69] Pham TN, Lengkeek NA, Greguric I, et al. Tunable and noncytotoxic PET/SPECT-MRI multimodality imaging probes using colloidally stable ligand-free superparamagnetic iron oxide nanoparticles. International Journal of Nanomedicine. 2017;12:899-909.
  • [70] Bennett KM, Jo J, Cabral H, et al. MR imaging techniques for nano-pathophysiology and theranostics. Advanced Drug Delivery Reviews. 2014;74:75-94.
  • [71] Shahbazi-Gahrouei D. Novel MR imaging contrast agents for cancer detection. Journal of Research in Medical Sciences. 2009;14(3):141-147.
  • [72] Lauterbur P. Image-formation by induced local interactions-examples employing nuclear magnetic-resonance. Clinical Orthopaedics and Related Research. 1989;244:3-6.
  • [73] Shahbazi‐Gahrouei D, Williams M, Rizvi S, Allen B. In vivo studies of Gd‐DTPA‐monoclonal antibody and gd‐porphyrins: Potential magnetic resonance imaging contrast agents for melanoma. Journal of Magnetic Resonance Imaging. 2001;14(2):169-174.
  • [74] Shahbazi-Gahrouei D, Rizvi S, Williams M, Allen B. In vitro studies of gadolinium-DTPA conjugated with monoclonal antibodies as cancer-specific magnetic resonance imaging contrast agents. Australasian Physics and Engineering Sciences in Medicine. 2002;25(1):31-38.
  • [75] Mirzaei M, Mohagheghi M, Shahbazi-Gahrouei D, Khatami A. Gd3+-Anionic Linear Globular Dendrimer-G2-C595 A Dual Novel Nanoprobe for MR Imaging and Therapeutic Agent. J Nanomed Nanotechnol. 2012;3(7):1000147.
  • [76] Zhang M, Cao Y, Chong Y, et al. Graphene oxide based theranostic platform for T 1-weighted magnetic resonance imaging and drug delivery. ACS Applied Materials and Interfaces. 2013;5(24):13325-13332.
  • [77] Shahbazi-Gahrouei D, Abdolahi M. A novel method for quantitative analysis of anti-MUC1 expressing ovarian cancer cell surface based on magnetic cell separation. Journal of Medical Sciences. 2012;12(8):256-266.
  • [78] Abdolahi M, Shahbazi‐Gahrouei D, Laurent S, et al. Synthesis and in vitro evaluation of MR molecular imaging probes using J591 mAb‐conjugated SPIONs for specific detection of prostate cancer. Contrast Media and Molecular Imaging. 2013;8(2):175-184.
  • [79] Shahbazi-Gahrouei D, Abdolahi M. Detection of MUC1-expressing ovarian cancer by C595 monoclonal antibody-conjugated SPIONs using MR imaging. The Scientific World Journal. 2013;2013:609151.
  • [80] Shahbazi-Gahrouei D, Abdolahi M. Superparamagnetic iron oxide-C595: Potential MR imaging contrast agents for ovarian cancer detection. Journal of Medical Physics. 2013;38(4):198-204.
  • [81] Shahbazi-Gahrouei D, Khodamoradi E. Porphyrin-based agents: potential MR imaging contrast agents for colorectal (HT29/219) detection in mice. Journal of Medical Sciences. 2007;7(6):1015-1020.
  • [82] Fang H, Liu C, Liu C, et al. Gd-DTPA-dialkylamine derivatives: Synthesis and self-assembled behaviors for T1-enhanced magnetic resonance imaging and drug carriers. Journal of Molecular Liquids. 2018;268:77-86.
  • [83] Keshtkar M, Shahbazi-Gahrouei D, Mehrgardi M, Aghaei M. Synthesis and Cytotoxicity Assessment of Gold-coated Magnetic Iron Oxide Nanoparticles. Journal of Biomedical Physics and Engineering. 2018;8(4):357-364.
  • [84] Keshtkar M, Shahbazi-Gahrouei D, Khoshfetrat SM, et al. Aptamer-conjugated magnetic nanoparticles as targeted magnetic resonance imaging contrast agent for breast cancer. Journal of Medical Signals and Sensors. 2016;6(4):243-247.
  • [85] Ghahremani F, Shahbazi-Gahrouei D, Kefayat A, et al. AS1411 aptamer conjugated gold nanoclusters as a targeted radiosensitizer for megavoltage radiation therapy of 4T1 breast cancer cells. RSC Advances. 2018;8(8):4249-4258.
  • [86] Ghahremani F, Kefayat A, Shahbazi-Gahrouei D, Motaghi H, Mehrgardi MA, Javanmard SH. AS1411 Aptamer targeted gold nanoclusters effect on enhancement of radiation therapy efficacy in 4T1 breast tumor-bearing mice. Nanomedicine. 2018; 13(20): 2563-2578.
  • [87] Li J, Wu C, Hou P, et al. One-pot preparation of hydrophilic manganese oxide nanoparticles as T1 nano-contrast agent for molecular magnetic resonance imaging of renal carcinoma in vitro and in vivo. Biosensors and Bioelectronics. 2018;102:1-8.
  • [88] Liu K, Yan X, Xu Y-J, et al. Sequential growth of CaF 2: Yb, Er@ CaF 2: Gd nanoparticles for efficient magnetic resonance angiography and tumor diagnosis. Biomaterials Science. 2017;5(12):2403-2415.
  • [89] Ma L, Liu Y, Liu Liu L, et al. Simultaneous activation of short‐wave infrared (SWIR) light and paramagnetism by a functionalized shell for high penetration and spatial resolution theranostics. Advanced Functional Materials. 2018;28(6):1705057.
  • [90] You Q, Sun Q, Yu M, et al. BSA–Bioinspired Gadolinium Hybrid-Functionalized Hollow Gold Nanoshells for NIRF/PA/CT/MR Quadmodal Diagnostic Imaging-Guided Photothermal/Photodynamic Cancer Therapy. ACS Applied Materials & Interfaces. 2017;9(46):40017-40030.
  • [91] Payne WM, Hill TK, Svechkarev D, et al. Multimodal imaging nanoparticles derived from hyaluronic acid for integrated preoperative and intraoperative cancer imaging. Contrast Media and Molecular Imaging. 2017;2017:9616791.
  • [92] Chan WC, Nie S. Quantum dot bioconjugates for ultrasensitive nonisotopic detection. Science. 1998;281(5385):2016-2018.
  • [93] Chen H, Wang Y, Wang T, et al. Application prospective of nanoprobes with MRI and FI dual-modality imaging on breast cancer stem cells in tumor. Journal of Nanobiotechnology. 2016;14(1):52.
  • [94] Srinivas M, Heerschap A, Ahrens ET, et al. 19F MRI for quantitative in vivo cell tracking. Trends in Biotechnology. 2010;28(7):363-370.
  • [95] Xu M, Guo C, Hu G, et al. Organic Nanoprobes for Fluorescence and 19F Magnetic Resonance Dual‐Modality Imaging. Chinese Journal of Chemistry. 2018;36(1):25-30.
  • [96] Depalo N, Corricelli M, De Paola I, et al. NIR Emitting Nanoprobes Based on Cyclic RGD Motif Conjugated PbS Quantum Dots for Integrin-Targeted Optical Bioimaging. ACS Appl Mater Interfaces. 2017;9(49):43113-43126.
  • [97] Xie W, Wang L, Zhang Y, et al. Nuclear targeted nanoprobe for single living cell detection by surface-enhanced Raman scattering. Bioconjug Chem. 2009;20(4):768-773.
  • [98] Repenko T, Rix A, Nedilko A, et al. Strong photoacoustic signal enhancement by coating gold nanoparticles with melanin for biomedical imaging. Advanced Functional Materials. 2018;28(7):1705607.
  • [99] Liu Y, Lv X, Liu H, et al. Porous gold nanocluster-decorated manganese monoxide nanocomposites for microenvironmentactivatable MR/photoacoustic/CT tumor imaging. Nanoscale. 2018;10(8):3631-3638.
  • [100] Zhang L, Wang D, Yang K, et al. Mitochondria‐Targeted Artificial “Nano‐RBCs” for Amplified Synergistic Cancer Phototherapy by a Single NIR Irradiation. Advanced Science. 2018;5(5):1800049.
  • [101] Pansare V, Hejazi S, Faenza W, Prud'homme RK. Review of Long-Wavelength Optical and NIR Imaging Materials: Contrast Agents, Fluorophores and Multifunctional Nano Carriers. Chemistry of Materials. 2012;24(5):812-827.
  • [102] Ren F, Ding L, Liu H, et al. Ultra-small nanocluster mediated synthesis of Nd3+-doped core-shell nanocrystals with emission in the second near-infrared window for multimodal imaging of tumor vasculature. Biomaterials. 2018;175:30-43.
  • [103] Pisanic TR 2nd, Zhang Y, Wang TH. Quantum dots in diagnostics and detection: principles and paradigms. The Analyst. 2014;139(12):2968-2981.
  • [104] Liu H, Li Z, Sun Y, et al. Synthesis of Luminescent Carbon Dots with Ultrahigh Quantum Yield and Inherent Folate Receptor-Positive Cancer Cell Targetability. Scientific Reports. 2018;8(1):1086.
  • [105] Pellerin M, Glais E, Lecuyer T, et al. LaAlO3: Cr3+, Sm3+: Nano-perovskite with persistent luminescence for in vivo optical imaging. Journal of Luminescence. 2018;202:83-88.
  • [106] Schroeder KL, Goreham RV, Nann T. Graphene Quantum Dots for Theranostics and Bioimaging. Pharm Res. 2016;33(10):2337-2357.
  • [107] Hai X, Guo Z, Lin X, et al. Fluorescent TPA@GQDs Probe for Sensitive Assay and Quantitative Imaging of Hydroxyl Radicals in Living Cells. ACS Appl Mater Interfaces. 2018;10(6):5853-5861.
Uwagi
Opracowanie rekordu ze środków MNiSW, umowa Nr 461252 w ramach programu "Społeczna odpowiedzialność nauki" - moduł: Popularyzacja nauki i promocja sportu (2020).
Typ dokumentu
Bibliografia
Identyfikator YADDA
bwmeta1.element.baztech-e2a1a607-f022-4b5e-8431-adb69b937371
JavaScript jest wyłączony w Twojej przeglądarce internetowej. Włącz go, a następnie odśwież stronę, aby móc w pełni z niej korzystać.