AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (3.2 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review | Open Access

Phase-transition Perfluorocarbon Nanoparticles for Ultrasound Molecular Imaging and Therapy

Jinshun Xu1,#( )Yang Cao1,2,#Chunyan Xu1Xuan Zhou3Jianxin Liu1Yuanzhi Yao1Pan Li1,2( )Li Zhou1Yufeng You1Lan Hao1,2Yang Sun1,2Weixiang Song1Yajing Zhao1Zhigang Wang1,2( )
Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400016, China
The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
Department of Emergency of Chinese PLA General Hospital, China

#Both authors contributed equally to this study.

Show Author Information

Abstract

Recently, the advances in perfluorocarbon nanoparticles have been introduced to expand the diagnostic and therapeutic capability of ultrasound molecular imaging, a non-invasive diagnostic imaging, which passed from robust anatomical presentation to detection of physiological processes and recognition of specific tissue epitopes at the cellular level. The good properties of perfluorocarbon nanoparticles, such as nontoxicity, small size, phase-shift capability, etc., have been resulted in tremendous potential prospects in clinical application. Herein, this review focuses on the mechanisms of perfluorocarbon nanoparticles in terms of phase transition and ultrasonic theranostic. And the potential applications of perfluorocarbon nanoparticles in ultrasound medicine are also discussed.

References

[1]

J.M. Yang, C. Favazza, R. Chen, et al., Simultaneous functional photoacoustic and ultrasonic endoscopy of internal organs in vivo. Nature medicine. 2012; 18: 1297-1302.

[2]

J. Bohannon, Ultrasound uses in medicine heat up. Science (New York, NY). 2008; 321: 338-339.

[3]

L.V. Wang, S. Hu, Photoacoustic tomography: in vivo imaging from organelles to organs. Science. 2012; 335: 1458-1462.

[4]

E. Terreno, D.D. Castelli, A. Viale, et al., Challenges for molecular magnetic resonance imaging. Chemical reviews. 2010; 110: 3019-3042.

[5]

W. Cai, X. Chen, Nanoplatforms for targeted molecular imaging in living subjects. Small. 2007; 3: 1840-1854.

[6]

F. Yang, S. Hu, Y. Zhang,, et al., A hydrogen peroxide- responsive O(2) nanogenerator for ultrasound and magnetic-resonance dual modality imaging. Advanced materials (Deerfield Beach, Fla). 2012; 24: 5205-5211.

[7]

M.A. Nakatsuka, M.J. Hsu, S.C. Esener, et al., DNA- coated microbubbles with biochemically tunable ultrasound contrast activity. Advanced materials (Deerfield Beach, Fla). 2011; 23: 4908-4912.

[8]

O.D. Kripfgans, J.B. Fowlkes, D.L. Miller, et al., Acoustic droplet vaporization for therapeutic and diagnostic applications. Ultrasound in medicine & biology. 2000; 26: 1177-1189.

[9]

E. Huynh, J.F. Lovell, B.L. Helfield, et al., Porphyrin shell microbubbles with intrinsic ultrasound and photoacoustic properties. Journal of the American Chemical Society. 2012; 134: 16464-16467.

[10]

J. Zhang, R.J. Coulston, S.T. Jones, et al., One-step fabrication of supramolecular microcapsules from microfluidic droplets. Science (New York, NY). 2012; 335: 690-694.

[11]

Y. Zhou, Z. Wang, Y. Chen, et al., Microbubbles from gas- generating perfluorohexane nanoemulsions for targeted temperature-sensitive ultrasonography and synergistic HIFU ablation of tumors. Advanced materials. 2013; 25: 4123-4130.

[12]

N. Rapoport, Phase-shift, stimuli-responsive perfluoro- carbon nanodroplets for drug delivery to cancer. Wiley interdisciplinary reviews Nanomedicine and nanobiotechnology. 2012; 4: 492-510.

[13]

Y. Chen, Q. Yin, X. Ji, et al., Manganese oxide-based multifunctionalized mesoporous silica nanoparticles for pH-responsive MRI, ultrasonography and circumvention of MDR in cancer cells. Biomaterials. 2012; 33: 7126-7137.

[14]

H. Ke, J. Wang, Z. Dai, et al., Gold-nanoshelled microcapsules: a theranostic agent for ultrasound contrast imaging and photothermal therapy. Angewandte Chemie (International ed in English). 2011; 50: 3017-3021.

[15]

M.S. Hughes, J.N. Marsh, H. Zhang, et al., Characteriza- tion of digital waveforms using thermodynamic analogs: detection of contrast-targeted tissue in vivo. Ultrasonics, Ferroelectrics and Frequency Control, IEEE Transactions on. 2006; 53: 1609-1616.

[16]

P. Grayburn, Perflenapent emulsion (echogen®): A new long-acting phase-shift agent for contrast echocardiography. Clinical cardiology. 1997; 20: 12-18.

[17]

G.M. Lanza, S.A. Wickline, Targeted ultrasonic contrast agents for molecular imaging and therapy. Progress in cardiovascular diseases. 2001; 44: 13-31.

[18]

R.B. Campbell, Tumor physiology and delivery of nanopharmaceuticals. Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry-Anti- Cancer Agents). 2006; 6: 503-512.

[19]

N. Rapoport, A.M. Kennedy, J.E. Shea, et al., Ultrasonic nanotherapy of pancreatic cancer: lessons from ultrasound imaging. Molecular pharmaceutics. 2009; 7: 22-31.

[20]

A. Kheirolomoom, L.M. Mahakian, C.Y. Lai, et al., Copper-Doxorubicin as a Nanoparticle Cargo Retains Efficacy with Minimal Toxicity. Molecular pharmaceutics. 2010; 7: 1948-1958.

[21]

M.A. Hahn, A.K. Singh, P. Sharma, et al., Nanoparticles as contrast agents for in-vivo bioimaging: current status and future perspectives. Analytical and bioanalytical chemistry. 2011; 399: 3-27.

[22]

M. Javadi, W.G. Pitt, C.M. Tracy, et al., Ultrasonic gene and drug delivery using eLiposomes. Journal of Controlled Release. 2013; 167: 92-100.

[23]

M. Beck-Broichsitter, A.C. Dalla-Bona, T. Kissel, et al., Polymer Nanoparticle-Based Controlled Pulmonary Drug Delivery. Drug Delivery System: Springer; 2014, 133- 145.

[24]

P.E. Keipert, S. Otto, S.F. Flaim, et al., Influence of perflubron emulsion particle size on blood half-life and febrile response in rats. Artificial cells, blood substitutes, and immobilization biotechnology. 1994; 22: 1169-1174.

[25]

D.R. Spahn, Blood substitutes. Artificial oxygen carriers: perfluorocarbon emulsions. Critical care (London, England). 1999; 3: R93-97.

[26]

R.J. Noveck, E.J. Shannon, P.T. Leese, et al., Randomized safety studies of intravenous perflubron emulsion. Ⅱ. Effects on immune function in healthy volunteers. Anesthesia and analgesia. 2000; 91: 812-822.

[27]
C.S. Cohn, M.M. Cushing, Oxygen therapeutics: perfluorocarbons and blood substitute safety. Critical care clinics. 2009;25: 399-414, Table of Contents.
[28]

N. Rapoport, D.A. Christensen, A.M. Kennedy, et al., Cavitation properties of block copolymer stabilized phase- shift nanoemulsions used as drug carriers. Ultrasound in medicine & biology. 2010;36: 419-429.

[29]

N. Rapoport, K.H. Nam, R. Gupta, et al., Ultrasound- mediated tumor imaging and nanotherapy using drug loaded, block copolymer stabilized perfluorocarbon nanoemulsions. Journal of Controlled Release. 2011; 153: 4-15.

[30]

G. Lanza, P. Winter, S. Caruthers, et al., Theragnostics for tumor and plaque angiogenesis with perfluorocarbon nanoemulsions. Angiogenesis. 2010; 13: 189-202.

[31]

S. M. Janib, A.S. Moses, J.A. MacKay, Imaging and drug delivery using theranostic nanoparticles. Advanced drug delivery reviews. 2010; 62: 1052-1063.

[32]
R.E. Apfel, Activatable infusable dispersions containing drops of a superheated liquid for methods of therapy and diagnosis. Google Patents; 1998.
[33]

O.D. Kripfgans, J.B. Fowlkes, M. Woydt, et al., In vivo droplet vaporization for occlusion therapy and phase aberration correction. IEEE transactions on ultrasonics, ferroelectrics, and frequency control. 2002; 49: 726-738.

[34]

O.D. Kripfgans, M.L. Fabiilli, P.L. Carson, et al., On the acoustic vaporization of micrometer-sized droplets. The Journal of the Acoustical Society of America. 2004; 116: 272-281.

[35]

A.H. Lo, O.D. Kripfgans, P.L. Carson, et al., Acoustic droplet vaporization threshold: effects of pulse duration and contrast agent. IEEE transactions on ultrasonics, ferroelectrics, and frequency control. 2007; 54: 933-946.

[36]

M.L. Fabiilli, K.J. Haworth, N.H. Fakhri, et al., The role of inertial cavitation in acoustic droplet vaporization. IEEE transactions on ultrasonics, ferroelectrics, and frequency control. 2009; 56: 1006-1017.

[37]

M.L. Fabiilli, J.A. Lee, O.D. Kripfgans, et al., Delivery of water-soluble drugs using acoustically triggered perfluorocarbon double emulsions. Pharmaceutical research. 2010; 27: 2753-2765.

[38]

M. Zhang, M.L. Fabiilli, K.J. Haworth, et al., Initial investigation of acoustic droplet vaporization for occlusion in canine kidney. Ultrasound Med Biol. 2010; 36: 1691-1703.

[39]

C.M. Carneal, O.D. Kripfgans, J. Krucker, et al., A tissue-mimicking ultrasound test object using droplet vaporization to create point targets. IEEE transactions on ultrasonics, ferroelectrics, and frequency control. 2011; 58: 2013-2025.

[40]

M. Zhang, M.L. Fabiilli, K.J. Haworth, et al., Acoustic droplet vaporization for enhancement of thermal ablation by high intensity focused ultrasound. Academic radiology. 2011; 18: 1123-1132.

[41]

O.D. Kripfgans, M. Zhang, M.L. Fabiilli, et al., Acceleration of ultrasound thermal therapy by patterned acoustic droplet vaporization. The Journal of the Acoustical Society of America. 2014; 135: 537-544.

[42]

D.S. Li, O.D. Kripfgans, M.L. Fabiilli, et al., Formation of toroidal bubbles from acoustic droplet vaporization. Applied physics letters. 2014; 104: 063706.

[43]

D.S. Li, O.D. Kripfgans, M.L. Fabiilli, et al., Initial nucleation site formation due to acoustic droplet vaporization. Applied physics letters. 2014; 104: 063703.

[44]
P.C. Hiemenz, R. Rajagopalan, Principles of Colloid and Surface Chemistry, revised and expanded: CRC Press; 1997.
[45]

P.S. Sheeran, T.O. Matsunaga, P.A. Dayton, Phase- transition thresholds and vaporization phenomena for ultrasound phase-change nanoemulsions assessed via high-speed optical microscopy. Physics in medicine and biology. 2013; 58: 4513.

[46]

C.Y. Lin, W.G. Pitt, Acoustic droplet vaporization in biology and medicine. BioMed research international. 2013;2013.

[47]

L.Y. Clasohm, I.U. Vakarelski, R.R. Dagastine, et al., Anomalous pH dependent stability behavior of surfactant- free nonpolar oil drops in aqueous electrolyte solutions. Langmuir. 2007; 23: 9335-9340.

[48]

P.S. Sheeran, S. Luois, P.A. Dayton, et al., Formulation and acoustic studies of a new phase-shift agent for diagnostic and therapeutic ultrasound. Langmuir. 2011; 27: 10412-10420.

[49]

P.S. Sheeran, S.H. Luois, L.B. Mullin, et al., Design of ultrasonically-activatable nanoparticles using low boiling point perfluorocarbons. Biomaterials. 2012; 33: 3262-3269.

[50]

Z.Z. Wong, O.D. Kripfgans, A. Qamar, et al., Bubble evolution in acoustic droplet vaporization at physiological temperature via ultra-high speed imaging. Soft Matter. 2011; 7: 4009-4016.

[51]

K.J. Haworth, J.B. Fowlkes, P.L. Carson, et al., Towards aberration correction of transcranial ultrasound using acoustic droplet vaporization. Ultrasound in medicine & biology. 2008; 34: 435-445.

[52]

S.T. Kang, C.K. Yeh, Intracellular Acoustic Droplet Vaporization in a Single Peritoneal Macrophage for Drug Delivery Applications. Langmuir. 2011; 27 : 13183-13188.

[53]

W.G. Pitt, R.N. Singh, K.X. Perez, et al., Phase transitions of perfluorocarbon nanoemulsion induced with ultrasound: a mathematical model. Ultrasonics sonochemistry. 2014; 21: 879-891.

[54]

A. Kabalnov, J. Weers, R. Arlauskas, et al., Phospholipids as emulsion stabilizers. 1. Interfacial tensions. Langmuir. 1995; 11: 2966-2974.

[55]
R. Rowley, W. Wilding, J. Oscarson, et al., DIPPR Data Compilation of Pure Chemical Properties. http://dipprbyuedu/. 2012.
[56]

O. Shpak, L. Stricker, M. Versluis, et al., The role of gas in ultrasonically driven vapor bubble growth. Physics in medicine and biology. 2013; 58: 2523.

[57]

N. Reznik, O. Shpak, E.C. Gelderblom, et al., The efficiency and stability of bubble formation by acoustic vaporization of submicron perfluorocarbon droplets. Ultrasonics. 2013; 53: 1368-1376.

[58]

J. Lattin, D. Belnap, W. Pitt, Formation of eLiposomes as a drug delivery vehicle. Colloids and Surfaces B: Biointerfaces. 2012; 89: 93-100.

[59]

J.W. Wijlemans, M. de Greef, G. Schubert, et al., A Clinically Feasible Treatment Protocol for Magnetic Resonance-Guided High-Intensity Focused Ultrasound Ablation in the Liver. Investigative radiology. 2015; 50: 24-31.

[60]

S. Xu, Y. Zong, Y. Feng, et al., Dependence of pulsed focused ultrasound induced thrombolysis on duty cycle and cavitation bubble size distribution. Ultrasonics sonochemistry. 2015; 22: 160-166.

[61]

B.J.J. Abdullah, C.H. Yeong, K.L. Goh, et al., Robotic- assisted thermal ablation of liver tumours. European radiology. 2015; 25: 246-257.

[62]

M.A. Chinnaratha, D. Sathananthan, P. Pateria, et al., High local recurrence of early-stage hepatocellular carcinoma after percutaneous thermal ablation in routine clinical practice. European Journal of Gastroenterology & Hepatology. 2015.

[63]

U. Leung, D. Kuk, M.D. Angelica, et al., Long-term outcomes following microwave ablation for liver malignancies. British Journal of Surgery. 2015; 102: 85-91.

[64]

K.C. Huang, W.T. Hsiao, C.H. Hwang, et al., The laser ablation model development of glass substrate cutting assisted with the thermal fracture and ultrasonic mechanisms. Optics and Lasers in Engineering. 2015; 67: 31-35.

[65]

K.F. Chu, D.E. Dupuy, Thermal ablation of tumours: biological mechanisms and advances in therapy. Nature Reviews Cancer. 2014; 14: 199-208.

[66]

T.M. Krupka, D. Dremann, A.A. Exner, Time and dose dependence of pluronic bioactivity in hyperthermia- induced tumor cell death. Experimental Biology and Medicine. 2009; 234: 95-104.

[67]

M.R. Dreher, W. Liu, C.R. Michelich, et al., Tumor vascular permeability, accumulation, and penetration of macromolecular drug carriers. Journal of the National Cancer Institute. 2006; 98: 335-344.

[68]

P.S. Yarmolenko, Y. Zhao, C. Landon, et al., Comparative effects of thermosensitive doxorubicin-containing liposomes and hyperthermia in human and murine tumours. International Journal of Hyperthermia. 2010; 26: 485-498.

[69]

Z. Vujaskovic, D.W. Kim, E. Jones, et al., A phase I/Ⅱ study of neoadjuvant liposomal doxorubicin, paclitaxel, and hyperthermia in locally advanced breast cancer. International Journal of Hyperthermia. 2010; 26: 514-521.

[70]

A.H. Negussie, P.S. Yarmolenko, A. Partanen, et al., Formulation and characterisation of magnetic resonance imageable thermally sensitive liposomes for use with magnetic resonance-guided high intensity focused ultrasound. International Journal of Hyperthermia. 2011; 27: 140-155.

[71]

R. Deckers, C.T. Moonen, Ultrasound triggered, image guided, local drug delivery. Journal of Controlled Release. 2010; 148: 25-33.

[72]

R.T. Poon, N. Borys, Lyso-thermosensitive liposomal doxorubicin: a novel approach to enhance efficacy of thermal ablation of liver cancer. Expert Opin Pharmacother. 2009; 10: 333-343.

[73]

S. Song, H. Guo, Z. Jiang, et al., Self-Assembled Fe3O4/ Polymer Hybrid Microbubble with MRI/Ultrasound Dual- Imaging Enhancement. Langmuir. 2014; 30: 10557-10561.

[74]

C. Tremblay-Darveau, R. Williams, L. Milot, et al., Combined perfusion and doppler imaging using plane- wave nonlinear detection and microbubble contrast agents. Ultrasonics, Ferroelectrics, and Frequency Control, IEEE Transactions on. 2014; 61: 1988-2000.

[75]

Y. Yang, J. Wang, X. Li, et al., A near infrared fluorescent/ ultrasonic bimodal contrast agent for imaging guided pDNA delivery via ultrasound targeted microbubble destruction. RSC Advances. 2015; 5: 8404-8414.

[76]
L.J. M, Juffermans, Ultrasound and microbubble-targeted delivery of drugs and genes: Cellular bioeffects and mechanisms. 2009.
[77]

I. De Cock, E. Zagato, K. Braeckmans, et al., Ultrasound and microbubble mediated drug delivery: Acoustic pressure as determinant for uptake via membrane pores or endocytosis. Journal of Controlled Release. 2014.

[78]

Z. Fan, R.E. Kumon, C.X. Deng, Mechanisms of microbubble-facilitated sonoporation for drug and gene delivery. Therapeutic delivery. 2014; 5: 467-486.

[79]

O.F. Eker, B. Quesson, C. Rome, et al., Combination of cell delivery and thermoinducible transcription for in vivo spatiotemporal control of gene expression: a feasibility study. Radiology. 2011; 258: 496-504.

[80]

G.A. Husseini, L. Kherbeck, W.G. Pitt, et al., Kinetics of ultrasonic drug delivery from targeted micelles. Journal of Nanoscience and Nanotechnology. 2015; 15: 2099-2104.

[81]

I. Lentacker, I. De Cock, R. Deckers, et al., Understanding ultrasound induced sonoporation: Definitions and underlying mechanisms. Advanced drug delivery reviews. 2014; 72: 49-64.

[82]

H. Yu, S. Chen, A model to calculate microstreaming- shear stress generated by oscillating microbubbles on the cell membrane in sonoporation. Bio-medical materials and engineering. 2014; 24: 861-868.

[83]

J.J. Rychak, A.L. Klibanov, Nucleic acid delivery with microbubbles and ultrasound. Advanced drug delivery reviews. 2014.

[84]

E. Unger, T. Porter, J. Lindner, et al., Cardiovascular drug delivery with ultrasound and microbubbles. Advanced drug delivery reviews. 2014; 72: 110-126.

[85]

S.R. Sirsi, M.A. Borden, State-of-the-art materials for ultrasound-triggered drug delivery. Advanced drug delivery reviews. 2014; 72: 3-14.

[86]

E. Paoli, D. Kruse, J. Seo, et al., An optical and microPET assessment of thermally-sensitive liposome biodistribution in the Met-1 tumor model: Importance of formulation. Journal of Controlled Release. 2010; 143: 13-22.

[87]

A. Kheirolomoom, D.E. Kruse, S. Qin, et al., Enhanced in vivo bioluminescence imaging using liposomal luciferin delivery system. Journal of Controlled Release. 2010; 141: 128-136.

[88]

A.L. Klibanov, T.I. Shevchenko, B.I. Raju, et al., Ultrasound-triggered release of materials entrapped in microbubble–liposome constructs: a tool for targeted drug delivery. Journal of Controlled Release. 2010; 148: 13-17.

[89]

S. Hernot, A.L. Klibanov, Microbubbles in ultrasound- triggered drug and gene delivery. Advanced drug delivery reviews. 2008; 60: 1153-1166.

[90]

P. Mohan, N. Rapoport, Doxorubicin as a molecular nanotheranostic agent: effect of doxorubicin encapsulation in micelles or nanoemulsions on the ultrasound-mediated intracellular delivery and nuclear trafficking. Molecular pharmaceutics. 2010; 7: 1959-1973.

[91]

M. BBöhmer, C. Chlon, B. Raju, et al., Focused ultrasound and microbubbles for enhanced extravasation. Journal of Controlled Release. 2010; 148: 18-24.

[92]

F. Guenther, C. von zur Muhlen, E.A. Ferrante, et al., An ultrasound contrast agent targeted to P-selectin detects activated platelets at supra-arterial shear flow conditions. Investigative radiology. 2010; 45: 586.

[93]

L.C. Phillips, A.L. Klibanov, D.K. Bowles, et al., Focused in vivo delivery of plasmid DNA to the porcine vascular wall via intravascular ultrasound destruction of microbubbles. Journal of vascular research. 2009; 47: 270-274.

[94]

P. Dayton, A. Klibanov, G. Brandenburger, et al., Acoustic radiation force in vivo: a mechanism to assist targeting of microbubbles. Ultrasound in medicine & biology. 1999; 25: 1195-1201.

[95]

P.A. Dayton, S. Zhao, S.H. Bloch, et al., Application of ultrasound to selectively localize nanodroplets for targeted imaging and therapy. Molecular imaging. 2006; 5: 160.

[96]

M.J. Shortencarier, P.A. Dayton, S.H. Bloch, et al., A method for radiation-force localized drug delivery using gas-filled lipospheres. Ultrasonics, Ferroelectrics and Frequency Control, IEEE Transactions on. 2004; 51: 822-831.

[97]

N. R. Soman, J.N. Marsh, M. S. Hughes, et al., Acoustic activation of targeted liquid perfluorocarbon nanoparticles does not compromise endothelial integrity. NanoBioscience, IEEE Transactions on. 2006; 5: 69-75.

[98]
C.K. Holland, D. D. McPherson, Echogenic lipsomes for targeted drug delivery. Biomedical Imaging: From Nano to Macro, 2009 ISBI'09 IEEE International Symposium on: IEEE; 2009; 755-758.
[99]

S.M. Stieger, C.F. Caskey, R.H. Adamson, et al., Enhancement of Vascular Permeability with Low- Frequency Contrast-enhanced Ultrasound in the Chorioallantoic Membrane Model. Radiology. 2007; 243: 112-121.

[100]

K.W. Ferrara, Driving delivery vehicles with ultrasound. Advanced drug delivery reviews. 2008; 60: 1097- 1102.

[101]

A.F. Lum, M.A. Borden, P.A. Dayton, et al., Ultrasound radiation force enables targeted deposition of model drug carriers loaded on microbubbles. Journal of Controlled Release. 2006; 111: 128-134.

[102]

N. McDannold, N. Vykhodtseva, K. Hynynen, Effects of acoustic parameters and ultrasound contrast agent dose on focused-ultrasound induced blood-brain barrier disruption. Ultrasound in medicine & biology. 2008; 34: 930-937.

[103]

N.Y. Rapoport, A.M. Kennedy, J.E. Shea, et al., Controlled and targeted tumor chemotherapy by ultrasound-activated nanoemulsions/microbubbles. Journal of Controlled Release. 2009; 138: 268-276.

[104]

N.J. McDannold, N.I. Vykhodtseva, K. Hynynen, Microbubble Contrast Agent with Focused Ultrasound to Create Brain Lesions at Low Power Levels: MR Imaging and Histologic Study in Rabbits. Radiology. 2006; 241: 95-106.

[105]

L.H. Treat, N. McDannold, N. Vykhodtseva, et al., Targeted delivery of doxorubicin to the rat brain at therapeutic levels using MRI‐guided focused ultrasound. International journal of cancer. 2007; 121: 901-907.

[106]

A.K. Iyer, G. Khaled, J. Fang, et al., Exploiting the enhanced permeability and retention effect for tumor targeting. Drug discovery today. 2006; 11: 812-818.

[107]

S.K. Hobbs, W.L. Monsky, F. Yuan, et al., Regulation of transport pathways in tumor vessels: role of tumor type and microenvironment. Proceedings of the National Academy of Sciences. 1998; 95: 4607-4612.

[108]

P.M. Winter, A.H. Schmieder, S.D. Caruthers, et al., Minute dosages of ανβ3-targeted fumagillin nanoparticles impair Vx-2 tumor angiogenesis and development in rabbits. The FASEB Journal. 2008; 22: 2758-2767.

[109]

P.M. Winter, S.D. Caruthers, H. Zhang, et al., Antiangiogenic synergism of integrin-targeted fumagillin nanoparticles and atorvastatin in atherosclerosis. JACC: Cardiovascular Imaging. 2008; 1: 624-634.

[110]

H.F. Zhou, H.W. Chan, S.A. Wickline, et al., Alphavbeta3- targeted nanotherapy suppresses inflammatory arthritis in mice. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2009; 23: 2978-2985.

[111]

N.R. Soman, S.L. Baldwin, G. Hu, et al., Molecularly targeted nanocarriers deliver the cytolytic peptide melittin specifically to tumor cells in mice, reducing tumor growth. The Journal of clinical investigation. 2009; 119: 2830-2842.

[112]

N.R. Soman, G.M. Lanza, J.M. Heuser, et al., Synthesis and characterization of stable fluorocarbon nanostructures as drug delivery vehicles for cytolytic peptides. Nano letters. 2008; 8: 1131-1136.

[113]

G. Hu, M. Lijowski, H. Zhang, et al., Imaging of Vx-2 rabbit tumors with alpha(nu)beta3-integrin-targeted 111In nanoparticles. International journal of cancer Journal international du cancer. 2007; 120: 1951-1957.

[114]

C.M. Sehgal, P.H. Arger, C.R. Pugh, Sonographic enhancement of renal cortex by contrast media. Journal of ultrasound in medicine. 1995; 14: 741-748.

[115]

J. Correas, S. Quay, EchoGen emulsion: a new ultrasound contrast agent based on phase shift colloids. Clinical radiology. 1996; 51: 11-14.

[116]

J.D. Kasprzak, F.J. Ten Cate, New ultrasound contrast agents for left ventricular and myocardial opacification. Herz. 1998; 23: 474-482.

[117]

P.A. Grayburn, J.L. Weiss, T.C. Hack, et al., Phase Ⅲ multicenter trial comparing the efficacy of 2% dodecafluoropentane emulsion (EchoGen) and sonicated 5% human albumin (Albunex) as ultrasound contrast agents in patients with suboptimal echocardiograms. Journal of the American College of Cardiology. 1998; 32: 230-236.

[118]

M.L. Main, P.A. Grayburn, Clinical applications of transpulmonary contrast echocardiography. American heart journal. 1999; 137: 144-153.

[119]

W.G. Hundley, A.M. Kizilbash, I. Afridi, et al., Administration of an intravenous perfluorocarbon contrast agent improves echocardiographic determination of left ventricular volumes and ejection fraction: comparison with cine magnetic resonance imaging. Journal of the American College of Cardiology. 1998; 32: 1426-1432.

[120]

H. Ragde, G. M. Kenny, G.P. Murphy, K. Landin, Transrectal ultrasound microbubble contrast angiography of the prostate. The Prostate. 1997; 32: 279-283.

[121]

J.M. Correas, A.R. Meuter, E. Singlas, et al., Human pharmacokinetics of a perfluorocarbon ultrasound contrast agent evaluated with gas chromatography. Ultrasound in medicine & biology. 2001; 27: 565-570.

[122]

R. Totaro, M. Del Sette, C. Marini, Echocontrast agents in neurosonology. Functional neurology. 1998; 14: 235-239.

[123]

K. Tiemann, S. Loheier, S. Kuntz, et al., Real-Time Contrast Echo Assessment of Myocardial Perfusion at Low Emission Power. Echocardiography. 1999; 16: 799-809.

[124]

N. Rapoport, Z. Gao, A. Kennedy, Multifunctional nanoparticles for combining ultrasonic tumor imaging and targeted chemotherapy. Journal of the National Cancer Institute. 2007; 99: 1095-1106.

[125]

J.A. Kopechek, E. Park, C.S. Mei, et al., Accumulation of phase-shift nanoemulsions to enhance MR-guided ultrasound-mediated tumor ablation in vivo. Journal of healthcare engineering. 2013; 4: 109-126.

[126]

J. Jian, C. Liu, Y. Gong, et al., India ink incorporated multifunctional phase-transition nanodroplets for photoacoustic/ultrasound dual-modality imaging and photoacoustic effect based tumor therapy. Theranostics. 2014; 4: 1026-1038.

[127]

M. Ma, H. Xu, H. Chen, et al., A drug-perfluorocarbon nanoemulsion with an ultrathin silica coating for the synergistic effect of chemotherapy and ablation by high- intensity focused ultrasound. Advanced materials. 2014; 26: 7378-7385.

[128]
G.M. Lanza, C.H. Lorenz, S.E. Fischer, et al., Enhanced detection of thrombi with a novel fibrin-targeted magnetic resonance imaging agent. Academic radiology. 1998; 5 Suppl 1: S173-176; discussion S183-174.
[129]

G.M. Lanza, K.D. Wallace, M.J. Scott, et al., A novel site- targeted ultrasonic contrast agent with broad biomedical application. Circulation. 1996; 94: 3334-3340.

[130]

G.M. Lanza, D.R. Abendschein, C.S. Hall, et al., In vivo molecular imaging of stretch-induced tissue factor in carotid arteries with ligand-targeted nanoparticles. Journal of the American Society of Echocardiography: official publication of the American Society of Echocardiography. 2000; 13: 608-614.

[131]

G.M. Lanza, D.R. Abendschein, C.S. Hall, et al. Molecular imaging of stretch-induced tissue factor expression in carotid arteries with intravascular ultrasound. Invest Radiol. 2000; 35: 227-234.

[132]
A. Morawski, P. Winter, S. Caruthers, et al., "Magnetic resonance immunocytochemistry": Characterization of "Tissue factor" expression by smooth muscle cells with targeted paramagnetic nanoparticles. Circulation: LIPPINCOTT WILLIAMS & WILKINS 530 WALNUT ST, PHILADELPHIA, PA 19106-3621 USA; 2003, 139-139.
[133]
G. Lanza, D. Abendschein, C. Hall, et al., In vivo molecular imaging of tissue factor in carotid arteries with a one-step ligand conjugated acoustic nanoparticle. Circulation: LIPPINCOTT WILLIAMS & WILKINS 530 WALNUT ST, PHILADELPHIA, PA 19106-3621 USA; 1999, 72-72.
[134]
N. Matsuura, R. Williams, I. Gorelikov, et al., Nanoparticle-loaded perfluorocarbon droplets for imaging and therapy. Ultrasonics Symposium (IUS), 2009 IEEE International: IEEE; 2009, 5-8.
[135]

S. Samuel, A. Duprey, M.L. Fabiilli, et al., In vivo microscopy of targeted vessel occlusion employing acoustic droplet vaporization. Microcirculation. 2012; 19: 501-509.

Nano Biomedicine and Engineering
Pages 8-19
Cite this article:
Xu J, Cao Y, Xu C, et al. Phase-transition Perfluorocarbon Nanoparticles for Ultrasound Molecular Imaging and Therapy. Nano Biomedicine and Engineering, 2015, 7(1): 8-19. https://doi.org/10.5101/nbe.v7i1.p8-19

364

Views

10

Downloads

2

Crossref

3

Scopus

Altmetrics

Received: 10 February 2015
Accepted: 15 March 2015
Published: 24 March 2015
© 2015 Jinshun Xu, Yang Cao, Chunyan Xu, Xuan Zhou, Jianxin Liu, Yuanzhi Yao, Pan Li, Li Zhou, Yufeng You, Lan Hao, Yang Sun, Weixiang Song, Yajing Zhao and Zhigang Wang.

This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Return