Minireviews Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Radiol. Dec 28, 2013; 5(12): 468-471
Published online Dec 28, 2013. doi: 10.4329/wjr.v5.i12.468
Nano/microparticles and ultrasound contrast agents
Shu-Guang Zheng, Hui-Xiong Xu, Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, Shanghai 200072, China
Hang-Rong Chen, State Key Laboratory of High Performance Ceramic and Superfine Microstructures, Shanghai Institute of Ceramic Chinese Academy of Science, Shanghai 200050, China
Author contributions: Zheng SG, Xu HX and Chen HR conducted the literature research; Zheng SG wrote the paper; Xu HX edited and revised the paper.
Supported by National Natural Science Foundation of China, No. 81371570; Key Project from Shanghai Health Bureau, No. 20114003; Shanghai Talent Development Project from Shanghai Human Resource and Social Security Bureau, No. 2012045
Correspondence to: Hui-Xiong Xu, MD, PhD, Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, No. 301 Yanchangzhong Road, Shanghai 200072, China. xuhuixiong@hotmail.com
Telephone: +86-21-66301031 Fax: +86-21-66301031
Received: July 17, 2013
Revised: October 31, 2013
Accepted: November 15, 2013
Published online: December 28, 2013
Processing time: 172 Days and 13.6 Hours

Abstract

Microbubbles have been used for many years now in clinical practice as contrast agents in ultrasound imaging. Recently, their therapeutic applications have also attracted more attention. However, the short circulation time (minutes) and relatively large size (two to ten micrometers) of currently used commercial microbubbles do not allow effective extravasation into tumor tissue, preventing efficient tumor targeting. Fortunately, more multifunctional and theranostic nanoparticles with some special advantages over the traditional microbubbles have been widely investigated and explored for biomedical applications. The way to synthesize an ideal ultrasound contrast agent based on nanoparticles in order to achieve an expected effect on contrast imaging is a key technique. Currently a number of nanomaterials, including liposomes, polymers, micelles, dendrimers, emulsions, quantum dots, solid nanoparticles etc., have already been applied to pre or clinical trials. Multifunctional and theranostic nanoparticles with some special advantages, such as the tumor-targeted (passive or active), multi-mode contrast agents (magnetic resonance imaging, ultrasonography or fluorescence), carrier or enhancer of drug delivery, and combined chemo or thermal therapy etc., are rapidly gaining popularity and have shown a promising application in the field of cancer treatment. In this mini review, the trends and the advances of multifunctional and theranostic nanoparticles are briefly discussed.

Key Words: Ultrasound contrast agent, Microbubble, Nanoparticle, Imaging, Nanomaterial

Core tip: The theranostic nanoparticles are defined as nanoparticles with double functions (for both therapeutic and diagnostic purposes) and are commonly applied to simultaneous drug delivery and molecular imaging.



MICROBUBBLES

Conventionally, the ultrasound contrast agents (UCAs) commercially used are microbubbles with sizes in the micrometer range, such as SonoVue®, Definity®, Luminity®, Sonazoid®etc., which are mainly composed of insoluble gas (perfluorocarbons or sulfur hexafluoride) and an encapsulating shell (lipids, proteins or polymers)[1-5]. During the past decades, these UCAs were mainly used for imaging in clinical practice[2-11]. Recently, microbubbles and their associated cavitation are playing an increasingly significant role in both diagnostic and therapeutic applications of ultrasound[12]. Besides that, microbubbles have also attracted more attention as carriers and enhancers of drug and gene delivery and have been widely investigated for these applications (especially in the area of anticancer research)[13]. However, short circulation time (minutes) and relatively large size (two to ten micrometers) of currently used commercial microbubbles do not allow effective extravasation into tumor tissue (pore size of tumor endothelium is typically in the range 380-780 nm), preventing efficient tumor targeting[14,15].

NANOMATERIALS

Fortunately, with the development of nanotechnology, nanomaterials have also evolved and now are widely recognized as a novel type of biomaterial, with very promising applications in the field of drug delivery[13,16-21]. Currently, a number of nanomaterials, including liposomes, polymers, micelles, dendrimers, emulsions, quantum dots, solid nanoparticles etc., have already been applied to pre or clinical trials[22-30]. In theory, nanomaterials can overcome the aforementioned shortcomings of microbubbles due to their smaller sizes (in the nanometer range)[29,31,32]. The National Cancer Institute has defined the nanoparticle as any particle with at least one dimension under 100 nm, while in many articles, some sub-micrometer particles have also been regarded as nanoparticles[1].

The way to synthesize an ideal ultrasound contrast agent based on nanoparticles in order to achieve an expected effect on contrast imaging is a key technique. According to some recently published articles, most researchers were able to enhance the acoustic backscatter by using nanomaterial equipped with gas (perfluorocarbon), although the nanoparticle-based contrast agents for the imaging modalities discussed were in various stages of development[25-27,33-35].

The use of nanomaterials as carriers for drug delivery represents the mainstream in the field of biomedical research[29,31,36-39]. However, recently, multifunctional and theranostic nanoparticles with some special advantages, such as the tumor-targeted (passive or active), multi-mode contrast agents [magnetic resonance imaging (MRI), ultrasonography (US) or fluorescence], carrier or enhancer of drug delivery, and combined chemo or thermal therapy, etc., are rapidly gaining popularity and have shown a promising application in the field of cancer treatment[13,18-20,29,38-42].

For instance, Lammers et al[31] discussed the principles, pitfalls and (pre) clinical progress of drugs (including those with nanoparticles) used for tumor-targeting. Most nanoparticles can be easily functionalized with a wide variety of biomolecules or antibodies and could be used for the targeted recognition or imaging of specific tissue/organs[19,43-47]. Besides, regarding its multifunction and multi-mode imaging, Malvindi et al[48] reported a magnetic/silica nanocomposite as a dual-mode contrast agent for combined MRI and US which enables non-invasive detection of the molecular components of pathological processes through multiple-mode imaging techniques[14,48]. Wang et al[35] proposed that Au nanoparticle coated mesoporous silica nanocapsule-based enhancement agents can be used as an inorganic theranostic platform for contrast-intensified US imaging, combined chemotherapy and efficient high intensity focused ultrasound tumor ablation[42,45,49].

Nowadays, these kinds of nanoparticles with more novel functions are still the research focus of biomaterials and are being explored for further biomedical applications[1,14,18,20,35,43,48,50].

We hope that the information presented in this mini review will stimulate the readers’ interest regarding the field of nano/microparticles and UCAs.

Footnotes

P- Reviewers: Martins WP, Tsalafoutas I S- Editor: Wen LL L- Editor: Roemmele A E- Editor: Liu SQ

References
1.  Mullin LB, Phillips LC, Dayton PA. Nanoparticle delivery enhancement with acoustically activated microbubbles. IEEE Trans Ultrason Ferroelectr Freq Control. 2013;60:65-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
2.  Claudon M, Cosgrove D, Albrecht T, Bolondi L, Bosio M, Calliada F, Correas JM, Darge K, Dietrich C, D’Onofrio M. Guidelines and good clinical practice recommendations for contrast enhanced ultrasound (CEUS) - update 2008. Ultraschall Med. 2008;29:28-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 526]  [Cited by in F6Publishing: 485]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
3.  Claudon M, Dietrich CF, Choi BI, Cosgrove DO, Kudo M, Nolsøe CP, Piscaglia F, Wilson SR, Barr RG, Chammas MC. Guidelines and good clinical practice recommendations for Contrast Enhanced Ultrasound (CEUS) in the liver - update 2012: A WFUMB-EFSUMB initiative in cooperation with representatives of AFSUMB, AIUM, ASUM, FLAUS and ICUS. Ultrasound Med Biol. 2013;39:187-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 473]  [Cited by in F6Publishing: 479]  [Article Influence: 43.5]  [Reference Citation Analysis (0)]
4.  Xu HX. Contrast-enhanced ultrasound: The evolving applications. World J Radiol. 2009;1:15-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 53]  [Cited by in F6Publishing: 44]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
5.  Quaia E. Microbubble ultrasound contrast agents: an update. Eur Radiol. 2007;17:1995-2008.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 230]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
6.  Xu HX. Contrast-enhanced ultrasound in the biliary system: Potential uses and indications. World J Radiol. 2009;1:37-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 34]  [Cited by in F6Publishing: 38]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
7.  Xu HX. Era of diagnostic and interventional ultrasound. World J Radiol. 2011;3:141-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
8.  Xu HX, Lu MD. The current status of contrast-enhanced ultrasound in China. J Med Ultrason. 2010;37:97-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 14]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
9.  Liu GJ, Lu MD. Diagnosis of liver cirrhosis with contrast-enhanced ultrasound. World J Radiol. 2010;2:32-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
10.  Ignee A, Straub B, Schuessler G, Dietrich CF. Contrast enhanced ultrasound of renal masses. World J Radiol. 2010;2:15-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 42]  [Cited by in F6Publishing: 27]  [Article Influence: 1.9]  [Reference Citation Analysis (1)]
11.  Molins IG, Font JM, Alvaro JC, Navarro JL, Gil MF, Rodríguez CM. Contrast-enhanced ultrasound in diagnosis and characterization of focal hepatic lesions. World J Radiol. 2010;2:455-462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 16]  [Cited by in F6Publishing: 19]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
12.  Stride EP, Coussios CC. Cavitation and contrast: the use of bubbles in ultrasound imaging and therapy. Proc Inst Mech Eng H. 2010;224:171-191.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Mohan P, Rapoport N. Doxorubicin as a molecular nanotheranostic agent: effect of doxorubicin encapsulation in micelles or nanoemulsions on the ultrasound-mediated intracellular delivery and nuclear trafficking. Mol Pharm. 2010;7:1959-1973.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 256]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
14.  Casciaro S, Soloperto G, Greco A, Casciaro E, Franchini R, Conversano F. Effectiveness of functionalized nanosystems for multimodal molecular sensing and imaging in medicine. IEEE Sens J. 2013;6:2305-2312.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J Control Release. 2000;65:271-284.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Suzuki R, Oda Y, Utoguchi N, Namai E, Taira Y, Okada N, Kadowaki N, Kodama T, Tachibana K, Maruyama K. A novel strategy utilizing ultrasound for antigen delivery in dendritic cell-based cancer immunotherapy. J Control Release. 2009;133:198-205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 71]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
17.  Petros RA, DeSimone JM. Strategies in the design of nanoparticles for therapeutic applications. Nat Rev Drug Discov. 2010;9:615-627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2702]  [Cited by in F6Publishing: 2508]  [Article Influence: 179.1]  [Reference Citation Analysis (0)]
18.  Janib SM, Moses AS, MacKay JA. Imaging and drug delivery using theranostic nanoparticles. Adv Drug Deliv Rev. 2010;62:1052-1063.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 916]  [Cited by in F6Publishing: 762]  [Article Influence: 54.4]  [Reference Citation Analysis (0)]
19.  Byrne JD, Betancourt T, Brannon-Peppas L. Active targeting schemes for nanoparticle systems in cancer therapeutics. Adv Drug Deliv Rev. 2008;60:1615-1626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1200]  [Cited by in F6Publishing: 1078]  [Article Influence: 67.4]  [Reference Citation Analysis (0)]
20.  Chen Y, Chen H, Zeng D, Tian Y, Chen F, Feng J, Shi J. Core/shell structured hollow mesoporous nanocapsules: a potential platform for simultaneous cell imaging and anticancer drug delivery. ACS Nano. 2010;4:6001-6013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 559]  [Cited by in F6Publishing: 335]  [Article Influence: 23.9]  [Reference Citation Analysis (0)]
21.  De Jong WH, Borm PJ. Drug delivery and nanoparticles: applications and hazards. Int J Nanomedicine. 2008;3:133-149.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Nie S, Xing Y, Kim GJ, Simons JW. Nanotechnology applications in cancer. Annu Rev Biomed Eng. 2007;9:257-288.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 783]  [Cited by in F6Publishing: 611]  [Article Influence: 35.9]  [Reference Citation Analysis (0)]
23.  Kratz F. Albumin as a drug carrier: design of prodrugs, drug conjugates and nanoparticles. J Control Release. 2008;132:171-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1512]  [Cited by in F6Publishing: 1511]  [Article Influence: 94.4]  [Reference Citation Analysis (0)]
24.  Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov. 2005;4:145-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3592]  [Cited by in F6Publishing: 3305]  [Article Influence: 173.9]  [Reference Citation Analysis (0)]
25.  Chung MF, Chen KJ, Liang HF, Liao ZX, Chia WT, Xia Y, Sung HW. A liposomal system capable of generating CO2 bubbles to induce transient cavitation, lysosomal rupturing, and cell necrosis. Angew Chem Int Ed Engl. 2012;51:10089-10093.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 101]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
26.  Wang X, Chen H, Chen Y, Ma M, Zhang K, Li F, Zheng Y, Zeng D, Wang Q, Shi J. Perfluorohexane-encapsulated mesoporous silica nanocapsules as enhancement agents for highly efficient high intensity focused ultrasound (HIFU). Adv Mater. 2012;24:785-791.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 171]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
27.  Kang E, Min HS, Lee J, Han MH, Ahn HJ, Yoon IC, Choi K, Kim K, Park K, Kwon IC. Nanobubbles from gas-generating polymeric nanoparticles: ultrasound imaging of living subjects. Angew Chem Int Ed Engl. 2010;49:524-528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 71]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
28.  Wang L, Shi J, Zhang H, Li H, Gao Y, Wang Z, Wang H, Li L, Zhang C, Chen C. Synergistic anticancer effect of RNAi and photothermal therapy mediated by functionalized single-walled carbon nanotubes. Biomaterials. 2013;34:262-274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 162]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
29.  Eifler AC, Thaxton CS. Nanoparticle therapeutics: FDA approval, clinical trials, regulatory pathways, and case study. Methods Mol Biol. 2011;726:325-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 86]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
30.  Chen H, Li B, Ren X, Li S, Ma Y, Cui S, Gu Y. Multifunctional near-infrared-emitting nano-conjugates based on gold clusters for tumor imaging and therapy. Biomaterials. 2012;33:8461-8476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 79]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
31.  Lammers T, Kiessling F, Hennink WE, Storm G. Drug targeting to tumors: principles, pitfalls and (pre-) clinical progress. J Control Release. 2012;161:175-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 994]  [Cited by in F6Publishing: 918]  [Article Influence: 76.5]  [Reference Citation Analysis (0)]
32.  Allen TM, Cullis PR. Drug delivery systems: entering the mainstream. Science. 2004;303:1818-1822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3286]  [Cited by in F6Publishing: 2903]  [Article Influence: 145.2]  [Reference Citation Analysis (0)]
33.  Liberman A, Martinez HP, Ta CN, Barback CV, Mattrey RF, Kono Y, Blair SL, Trogler WC, Kummel AC, Wu Z. Hollow silica and silica-boron nano/microparticles for contrast-enhanced ultrasound to detect small tumors. Biomaterials. 2012;33:5124-5129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 59]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
34.  Yang F, Chen P, He W, Gu N, Zhang X, Fang K, Zhang Y, Sun J, Tong J. Bubble microreactors triggered by an alternating magnetic field as diagnostic and therapeutic delivery devices. Small. 2010;6:1300-1305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 37]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
35.  Wang X, Chen H, Zheng Y, Ma M, Chen Y, Zhang K, Zeng D, Shi J. Au-nanoparticle coated mesoporous silica nanocapsule-based multifunctional platform for ultrasound mediated imaging, cytoclasis and tumor ablation. Biomaterials. 2013;34:2057-2068.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 109]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
36.  Wang AZ, Langer R, Farokhzad OC. Nanoparticle delivery of cancer drugs. Annu Rev Med. 2012;63:185-198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1110]  [Cited by in F6Publishing: 1052]  [Article Influence: 80.9]  [Reference Citation Analysis (0)]
37.  Egusquiaguirre SP, Igartua M, Hernández RM, Pedraz JL. Nanoparticle delivery systems for cancer therapy: advances in clinical and preclinical research. Clin Transl Oncol. 2012;14:83-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 160]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
38.  Chen Y, Chen H, Guo L, He Q, Chen F, Zhou J, Feng J, Shi J. Hollow/rattle-type mesoporous nanostructures by a structural difference-based selective etching strategy. ACS Nano. 2010;4:529-539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 583]  [Cited by in F6Publishing: 419]  [Article Influence: 29.9]  [Reference Citation Analysis (0)]
39.  Chen Y, Chen H, Zhang S, Chen F, Zhang L, Zhang J, Zhu M, Wu H, Guo L, Feng J. Multifunctional Mesoporous Nanoellipsoids for Biological Bimodal Imaging and Magnetically Targeted Delivery of Anticancer Drugs. Adv Funct Mater. 2011;21:270-278.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Rapoport N, Gao Z, Kennedy A. Multifunctional nanoparticles for combining ultrasonic tumor imaging and targeted chemotherapy. J Natl Cancer Inst. 2007;99:1095-1106.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Popović Z, Liu W, Chauhan VP, Lee J, Wong C, Greytak AB, Insin N, Nocera DG, Fukumura D, Jain RK. A nanoparticle size series for in vivo fluorescence imaging. Angew Chem Int Ed Engl. 2010;49:8649-8652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 248]  [Cited by in F6Publishing: 251]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
42.  Wang ZY, Song J, Zhang DS. Nanosized As2O3/Fe2O3 complexes combined with magnetic fluid hyperthermia selectively target liver cancer cells. World J Gastroenterol. 2009;15:2995-3002.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Behnke T, Mathejczyk JE, Brehm R, Würth C, Gomes FR, Dullin C, Napp J, Alves F, Resch-Genger U. Target-specific nanoparticles containing a broad band emissive NIR dye for the sensitive detection and characterization of tumor development. Biomaterials. 2013;34:160-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
44.  Liu J, Li J, Rosol TJ, Pan X, Voorhees JL. Biodegradable nanoparticles for targeted ultrasound imaging of breast cancer cells in vitro. Phys Med Biol. 2007;52:4739-4747.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Sheeran PS, Luois SH, Mullin LB, Matsunaga TO, Dayton PA. Design of ultrasonically-activatable nanoparticles using low boiling point perfluorocarbons. Biomaterials. 2012;33:3262-3269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 163]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
46.  Brannon-Peppas L, Blanchette JO. Nanoparticle and targeted systems for cancer therapy. Adv Drug Deliv Rev. 2004;56:1649-1659.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Veiseh O, Gunn JW, Zhang M. Design and fabrication of magnetic nanoparticles for targeted drug delivery and imaging. Adv Drug Deliv Rev. 2010;62:284-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1378]  [Cited by in F6Publishing: 1052]  [Article Influence: 75.1]  [Reference Citation Analysis (0)]
48.  Malvindi MA, Greco A, Conversano F, Figuerola A, Corti M, Bonora M, Lascialfari A, Doumari HA, Moscardini M, Cingolani R. Magnetic/Silica Nanocomposites as Dual-Mode Contrast Agents for Combined Magnetic Resonance Imaging and Ultrasonography. Adv Funct Mater. 2011;21:2548-2555.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Wei H, Huang J, Yang J, Zhang X, Lin L, Xue E, Chen Z. Ultrasound exposure improves the targeted therapy effects of galactosylated docetaxel nanoparticles on hepatocellular carcinoma xenografts. PLoS One. 2013;8:e58133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
50.  Dai LC, Yao X, Wang X, Niu SQ, Zhou LF, Fu FF, Yang SX, Ping JL. In vitro and in vivo suppression of hepatocellular carcinoma growth by midkine-antisense oligonucleotide-loaded nanoparticles. World J Gastroenterol. 2009;15:1966-1972.  [PubMed]  [DOI]  [Cited in This Article: ]