General Review Article

Ultrasound-Targeted Delivery Technology: A Novel Strategy for Tumor- Targeted Therapy

Author(s): Meng Du, Zhiyi Chen*, Yuhao Chen and Yue Li

Volume 20, Issue 2, 2019

Page: [220 - 231] Pages: 12

DOI: 10.2174/1389450119666180731095441

Price: $65

Abstract

Background: Ultrasound has been widely used in clinical diagnosis because it is noninvasive, inexpensive, simple, and reproducible. With the development of molecular imaging, material science, and ultrasound contrast agents, ultrasound-targeted delivery technology has emerged. The interaction of ultrasound and molecular probes can be exploited to change the structures of cells and tissues in order to promote the targeted release of therapeutic substances to local tumors. The targeted delivery of drugs, genes, and gases would not only improve the efficacy of tumor treatment but also avoid the systemic toxicity and side effects caused by antitumor treatments. This technology was recently applied in clinical trials and showed enormous potential for clinical application.

Objective: This article briefly introduces the characteristics of the tumor microenvironment and the principle of ultrasound-targeted delivery technology. To present recent progress in this field, this review focuses on the application of ultrasound-targeted delivery technology in tumor-targeted therapy, including drug delivery, gene transfection, and gas treatment.

Results: The results of this study show that ultrasound-targeted delivery technology is a promising therapeutic strategy for tumor treatment.

Conclusion: Ultrasound-targeted delivery technology shows promise with regard to cancer treatment.

Keywords: Drug delivery, targeted therapy, ultrasound, tumor, microbubble, gene therapy, gas treatment.

Graphical Abstract
[1]
Li WP, Su CH, Chang YC, Lin YJ, Yeh CS. Ultrasound-induced reactive oxygen species mediated therapy and imaging using a fenton reaction activable polymersome. ACS Nano 2016; 10(2): 2017-27.
[2]
Kobayashi H, Watanabe R, Choyke PL. Improving conventional Enhanced Permeability and Retention (EPR) effects; what is the appropriate target? Theranostics 2013; 4(1): 81-9.
[3]
Bae YH, Park K. Targeted drug delivery to tumors: Myths, reality and possibility. J Control Release 2011; 153(3): 198-205.
[4]
Maeda H, Nakamura H, Fang J. The EPR effect for macromolecular drug delivery to solid tumors: Improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo. Adv Drug Deliv Rev 2013; 65(1): 71-9.
[5]
Barua S, Mitragotri S. Challenges associated with penetration of nanoparticles across cell and tissue barriers: A review of current status and future prospects. Nano Today 2014; 9(2): 223-43.
[6]
Eales KL, Hollinshead KER, Tennant DA. Hypoxia and metabolic adaptation of cancer cells. Oncogenesis 2016; 5(1): e190.
[7]
Klemm Florian, Joyce Johanna A. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol 2015; 25(4): 198-213.
[8]
Gasser S, Lim L, Cheung F. The role of the tumour microenvironment in immunotherapy. Endocr Relat Cancer 2017; 24(12): T283-95.
[9]
Tzu-Yin W, Wilson KE, Machtaler S, Willmann JK. Ultrasound and microbubble guided drug delivery: mechanistic understanding and clinical implications. Curr Pharm Biotechnol 2013; 14(8): 743-52.
[10]
Lentacker I, De Cock I, Deckers R, De Smedt SC, Moonen CT. Understanding ultrasound induced sonoporation: definitions and underlying mechanisms. Adv Drug Deliv Rev 2014; 72: 49-64.
[11]
Dasgupta A, Liu M, Ojha T, Storm G, Kiessling F, Lammers T. Ultrasound-mediated drug delivery to the brain: principles, progress and prospects. Drug Discov Today Technol 2016; 20: 41-8.
[12]
Abou-Elkacem L, Bachawal SV, Willmann JK. Ultrasound molecular imaging: Moving toward clinical translation. Eur J Radiol 2015; 84(9): 1685-93.
[13]
Kilroy JP, Klibanov AL, Wamhoff BR, Bowles DK, Hossack JA. Localized in vivo model drug delivery with intravascular ultrasound and microbubbles. Ultrasound Med Biol 2014; 40(10): 2458-67.
[14]
Zhao S, Borden M, Bloch SH, Kruse D, Ferrara KW, Dayton PA. Radiation-force assisted targeting facilitates ultrasonic molecular imaging. Mol Imaging 2004; 3(3): 135-48.
[15]
Miller DL, Smith NB, Bailey MR, et al. Overview of therapeutic ultrasound applications and safety considerations. J Ultrasound Med 2012; 31(4): 623-34.
[16]
Luo MH, Yeh CK, Situ B, et al. Microbubbles: A novel strategy for chemotherapy. Curr Pharm Des 2017; 23(23): 3383-90.
[17]
Rapoport N, Gao Z, Kennedy A. Multifunctional nanoparticles for combining ultrasonic tumor imaging and targeted chemotherapy. J Natl Cancer Inst 2007; 99(14): 1095-106.
[18]
Min HS, You DG, Son S, et al. echogenic glycol chitosan nanoparticles for ultrasound-triggered cancer theranostics. Theranostics 2015; 5(12): 1402-18.
[19]
Minchinton AI, Tannock IF. Drug penetration in solid tumours. Nat Rev Cancer 2006; 6(8): 583-92.
[20]
Ho YJ, Chang YC, Yeh CK. Improving nanoparticle penetration in tumors by vascular disruption with acoustic droplet vaporization. Theranostics 2016; 6(3): 392-403.
[21]
Zhao H, Wu M, Zhu L, et al. Cell-penetrating peptide-modified targeted drug-loaded phase-transformation lipid nanoparticles combined with low-intensity focused ultrasound for precision theranostics against hepatocellular carcinoma. Theranostics 2018; 8(7): 1892-910.
[22]
Zhu L, Zhao H, Zhou Z, et al. Peptide-functionalized phase-transformation nanoparticles for low intensity focused ultrasound-assisted tumor imaging and therapy. Nano Lett 2018; 18(3): 1831-41.
[23]
Cao Y, Chen Y, Yu T, et al. Drug release from phase-changeable nanodroplets triggered by low-intensity focused ultrasound. Theranostics 2018; 8(5): 1327-39.
[24]
Huynh E, Leung BY, Helfield BL, et al. In situ conversion of porphyrin microbubbles to nanoparticles for multimodality imaging. Nat Nanotechnol 2015; 10(4): 325-32.
[25]
Paefgen V, Doleschel D, Kiessling F. Evolution of contrast agents for ultrasound imaging and ultrasound-mediated drug delivery. Front Pharmacol 2015; 6: 197.
[26]
Ma YY, Jin KT, Wang SB, et al. Molecular imaging of cancer with nanoparticle-based theranostic probes. Contrast Media Mol Imaging 2017; 2017: 1026270.
[27]
Ma J, Xu CS, Gao F, et al. Diagnostic and therapeutic research on ultrasound microbubble/nanobubble contrast agents.(Review). Mol Med Rep 2015; 12(3): 4022-8.
[28]
Xing L, Shi Q, Zheng K, et al. Ultrasound-mediated microbubble destruction (ummd) facilitates the delivery of ca19-9 targeted and paclitaxel loaded mpeg-plga-pll nanoparticles in pancreatic cancer. Theranostics 2016; 6(10): 1573-87.
[29]
Cerroni B, Chiessi E, Margheritelli S, Oddo L, Paradossi G. Polymer shelled microparticles for a targeted doxorubicin delivery in cancer therapy. Biomacromolecules 2011; 12(3): 593-601.
[30]
Liu H, Chang S, Sun J, et al. Ultrasound-mediated destruction of LHRHa-targeted and paclitaxel-loaded lipid microbubbles induces proliferation inhibition and apoptosis in ovarian cancer cells. Mol Pharm 2014; 11(1): 40-8.
[31]
Wu M, Wang Y, Wang Y, et al. Paclitaxel-loaded and A10-3.2 aptamer-targeted poly(lactide-co-glycolic acid) nanobubbles for ultrasound imaging and therapy of prostate cancer. Int J Nanomedicine 2017; 12: 5313-30.
[32]
An L, Hu H, Du J, et al. Paramagnetic hollow silica nanospheres for in vivo targeted ultrasound and magnetic resonance imaging. Biomaterials 2014; 35(20): 5381-92.
[33]
Gaertner FC, Kessler H, Wester HJ, Schwaiger M, Beer AJ. Radiolabelled RGD peptides for imaging and therapy. Eur J Nucl Med Mol Imaging 2012; 39(Suppl. 1): S126-38.
[34]
Deng Z, Xiao Y, Pan M, et al. Hyperthermia-triggered drug delivery from iRGD-modified temperature-sensitive liposomes enhances the anti-tumor efficacy using high intensity focused ultrasound. J Control Release 2016; 243: 333-41.
[35]
Luo W, Wen G, Yang L, et al. Dual-targeted and ph-sensitive doxorubicin prodrug-microbubble complex with ultrasound for tumor treatment. Theranostics 2017; 7(2): 452-65.
[36]
Lin W, Xie X, Deng J, et al. Cell-penetrating peptide-doxorubicin conjugate loaded NGR-modified nanobubbles for ultrasound triggered drug delivery. J Drug Target 2016; 24(2): 134-46.
[37]
Trendowski M. The promise of sonodynamic therapy. Cancer Metastasis Rev 2014; 33(1): 143-60.
[38]
Li Y, An H, Wang X, et al. Ultrasound-triggered release of sinoporphyrin sodium from liposome-microbubble complexes and its enhanced sonodynamic toxicity in breast cancer. Nano Res 2018; 11(2): 1038-56.
[39]
Wang X, Liu P, Yang W, et al. Microbubbles coupled to methotrexate-loaded liposomes for ultrasound-mediated delivery of methotrexate across the blood-brain barrier. Int J Nanomedicine 2014; 9((issue 1)): 4899.
[40]
Zhao Y, Lin Q, Wong HL, et al. Glioma-targeted therapy using Cilengitide nanoparticles combined with UTMD enhanced delivery. J Control Release 2016; 224: 112-25.
[41]
Fan CH, Cheng YH, Ting CY, et al. Ultrasound/magnetic targeting with spio-dox-microbubble complex for image-guided drug delivery in brain tumors. Theranostics 2016; 6(10): 1542-56.
[42]
Huang P, You X, Pan M, et al. A novel therapeutic strategy using ultrasound mediated microbubbles destruction to treat colon cancer in a mouse model. Cancer Lett 2013; 335(1): 183-90.
[43]
Zhang C, Huang P, Zhang Y, et al. Anti-tumor efficacy of ultrasonic cavitation is potentiated by concurrent delivery of anti-angiogenic drug in colon cancer. Cancer Lett 2014; 347(1): 105-13.
[44]
Moen I, Stuhr LEB. Hyperbaric oxygen therapy and cancer—a review. Target Oncol 2012; 7(4): 233-42.
[45]
Yang C, Xiao H, Sun Y, et al. Lipid microbubbles as ultrasound-stimulated oxygen carriers for controllable oxygen release for tumor reoxygenation. Ultrasound Med Biol 2018; 44(2): 416-25.
[46]
Liu L, Chang S, Sun J, et al. Ultrasound-mediated destruction of paclitaxel and oxygen loaded lipid microbubbles for combination therapy in ovarian cancer xenografts. Cancer Lett 2015; 361(1): 147-54.
[47]
Zhang K, Xu H, Chen H, et al. CO2 bubbling-based ‘Nanobomb’ System for Targetedly Suppressing Panc-1 Pancreatic Tumor via Low Intensity Ultrasound-activated Inertial Cavitation. Theranostics 2015; 5(11): 1291-302.
[48]
Min KH, Min HS, Lee HJ, et al. pH-controlled gas-generating mineralized nanoparticles: a theranostic agent for ultrasound imaging and therapy of cancers. ACS Nano 2015; 9(1): 134-45.
[49]
Carpenter AW, Schoenfisch MH. Nitric oxide release: part II. Therapeutic applications. Chem Soc Rev 2012; 41(10): 3742-52.
[50]
Zhang K, Xu H, Jia X, et al. Ultrasound-triggered nitric oxide release platform based on energy transformation for targeted inhibition of pancreatic tumor. ACS Nano 2016; 10(12): 10816-28.
[51]
Wang B, Zhai Y, Shi J, et al. Simultaneously overcome tumor vascular endothelium and extracellular matrix barriers via a non-destructive size-controlled nanomedicine. J Control Release 2017; 268: 225-36.
[52]
Dunbar CE, High KA, Joung JK, et al. Gene therapy comes of age. Science 2018; 359(6372): n4672.
[53]
Huang C, Zhang H, Bai R. Advances in ultrasound-targeted microbubble-mediated gene therapy for liver fibrosis. Acta Pharm Sin B 2017; 7(4): 447-52.
[54]
Bykov V, Eriksson SE, Bianchi J, Wiman KG. Targeting mutant p53 for efficient cancer therapy. NAT REV CANCER 2018; 18(2): 89-102.
[55]
Chang S, Guo J, Sun J, et al. Targeted microbubbles for ultrasound mediated gene transfection and apoptosis induction in ovarian cancer cells. Ultrason Sonochem 2013; 20(1): 171-9.
[56]
Gao R, Zhou X, Yang Y, Wang Z. Transfection of wtp53 and Rb94 genes into retinoblastomas of nude mice by ultrasound-targeted microbubble destruction. Ultrasound Med Biol 2014; 40(11): 2662-70.
[57]
Fan CH, Chang EL, Ting CY, et al. Folate-conjugated gene-carrying microbubbles with focused ultrasound for concurrent blood-brain barrier opening and local gene delivery. Biomaterials 2016; 106: 46-57.
[58]
Chang EL, Ting CY, Hsu PH, et al. Angiogenesis-targeting microbubbles combined with ultrasound-mediated gene therapy in brain tumors. J Control Release 2017; 255: 164-75.
[59]
Nana-Sinkam SP, Croce CM. MicroRNA regulation of tumorigenesis, cancer progression and interpatient heterogeneity: towards clinical use. Genome Biol 2014; 15(9): 445.
[60]
Li T, Sun X, Xu K. The suppressing role of miR-622 in renal cell carcinoma progression by down-regulation of CCL18/MAPK signal pathway. Cell Biosci 2018; 8: 17.
[61]
Dowdy SF. Overcoming cellular barriers for RNA therapeutics. Nat Biotechnol 2017; 35(3): 222-9.
[62]
Devulapally R, Sekar NM, Sekar TV, et al. Polymer nanoparticles mediated codelivery of antimiR-10b and antimiR-21 for achieving triple negative breast cancer therapy. ACS Nano 2015; 9(3): 2290-302.
[63]
Wang TY, Choe JW, Pu K, et al. Ultrasound-guided delivery of microRNA loaded nanoparticles into cancer. J Control Release 2015; 203: 99-108.
[64]
Chen W, Du J, Li X, et al. miR-509-3p promotes cisplatin-induced apoptosis in ovarian cancer cells through the regulation of anti-apoptotic genes. Pharmacogenomics 2017; 18(18): 1671-82.
[65]
Mullick CS, Wang TY, Bachawal S, et al. Ultrasound-guided therapeutic modulation of hepatocellular carcinoma using complementary microRNAs. J Control Release 2016; 238: 272-80.
[66]
Lin L, Fan Y, Gao F, et al. UTMD-promoted co-delivery of gemcitabine and mir-21 inhibitor by dendrimer-entrapped gold nanoparticles for pancreatic cancer therapy. Theranostics 2018; 8(7): 1923-39.
[67]
Slaby O, Laga R, Sedlacek O. Therapeutic targeting of non-coding RNAs in cancer. Biochem J 2017; 474(24): 4219-51.
[68]
Babu A, Muralidharan R, Amreddy N, et al. Nanoparticles for siRNA-based gene silencing in tumor therapy. IEEE Trans Nanobioscience 2016; 15(8): 849-63.
[69]
Zajac M, Muszalska I, Jelinska A. New molecular targets of anticancer therapy-current status and perspectives. Curr Med Chem 2016; 23(37): 4176-220.
[70]
Liang J, Huang W, Cai W, et al. Inhibition of microRNA-495 enhances therapeutic angiogenesis of human induced pluripotent stem cells. Stem Cells 2017; 35(2): 337-50.
[71]
Frinking PJ, Tardy I, Theraulaz M, et al. Effects of acoustic radiation force on the binding efficiency of BR55, a VEGFR2-specific ultrasound contrast agent. Ultrasound Med Biol 2012; 38(8): 1460-9.
[72]
Florinas S, Kim J, Nam K, Janat-Amsbury MM, Kim SW. Ultrasound-assisted siRNA delivery via arginine-grafted bioreducible polymer and microbubbles targeting VEGF for ovarian cancer treatment. J Control Release 2014; 183: 1-8.
[73]
Lyu H, Huang J, He Z, Liu B. Epigenetic mechanism of survivin dysregulation in human cancer. Sci China Life Sci 2018.
[74]
Li J, Cheng D, Yin T, et al. Copolymer of poly(ethylene glycol) and poly(L-lysine) grafting polyethylenimine through a reducible disulfide linkage for siRNA delivery. Nanoscale 2014; 6(3): 1732-40.
[75]
Zhang Y, Chang S, Sun J, et al. Targeted microbubbles for ultrasound mediated short hairpin rna plasmid transfection to inhibit survivin gene expression and induce apoptosis of ovarian cancer A2780/DDP Cells. Mol Pharm 2015; 12(9): 3137-45.
[76]
Chen ZY, Liang K, Lin Y, Yang F. Study of the UTMD-based delivery system to induce cervical cancer cell apoptosis and inhibit proliferation with shRNA targeting Survivin. Int J Mol Sci 2013; 14(1): 1763-77.
[77]
Wang P, Yin T, Li J, et al. Ultrasound-responsive microbubbles for sonography-guided siRNA delivery. Nanomedicine-UK 2016; 12(4): 1139-49.
[78]
Wu B, Qiao Q, Han X, et al. Targeted nanobubbles in low-frequency ultrasound-mediated gene transfection and growth inhibition of hepatocellular carcinoma cells. Tumour Biol 2016; 37(9): 12113-21.
[79]
Bae YJ, Yoon YI, Yoon TJ, Lee HJ. Ultrasound-guided delivery of sirna and a chemotherapeutic drug by using microbubble complexes: In Vitro and in vivo evaluations in a prostate cancer model. Korean J Radiol 2016; 17(4): 497-508.
[80]
Youle RJ, Strasser A. The BCL-2 protein family: Opposing activities that mediate cell death. Nat Rev Mol Cell Biol 2008; 9(1): 47-59.
[81]
Yin T, Wang P, Li J, et al. Tumor-penetrating codelivery of siRNA and paclitaxel with ultrasound-responsive nanobubbles hetero-assembled from polymeric micelles and liposomes. Biomaterials 2014; 35(22): 5932-43.
[82]
Scherman D, Rousseau A, Bigey P, Escriou V. Genetic pharmacology: progresses in siRNA delivery and therapeutic applications. Gene Ther 2017; 24(3): 151-6.
[83]
Zhao G, Huang Q, Wang F, et al. Targeted shRNA-loaded liposome complex combined with focused ultrasound for blood brain barrier disruption and suppressing glioma growth. Cancer Lett 2018; 418: 147-58.
[84]
Matkar PN, Singh KK, Prud Homme GJ, Hedley DW, Leong-Poi H. Abstract 2919: Ultrasound-mediated neuropilin-1 shRNA minicircle delivery inhibits tumour growth in an orthotopic human pancreatic adenocarcinoma model. Cancer Res 2016; 76(14)(Suppl.): 2919.
[85]
Zhou R, Curry JM, Roy LD, et al. A novel association of neuropilin-1 and MUC1 in pancreatic ductal adenocarcinoma: role in induction of VEGF signaling and angiogenesis. Oncogene 2016; 35(43): 5608-18.
[86]
Carpentier A, Canney M, Vignot A, et al. Clinical trial of blood-brain barrier disruption by pulsed ultrasound. Sci Transl Med 2016; 8(343): 342r-3r.
[87]
Dimcevski G, Kotopoulis S, Bjanes T, et al. A human clinical trial using ultrasound and microbubbles to enhance gemcitabine treatment of inoperable pancreatic cancer. J Control Release 2016; 243: 172-81.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy