Review Topical Sections

Drug delivery from engineered organisms and nanocarriers as monitored by multimodal imaging technologies

  • Received: 30 December 2016 Accepted: 08 March 2017 Published: 14 March 2017
  • In recent years, while the research budget and development times increased for different phases of drug development, the number of clinically approved new medicines declined. In fact, many promising drug candidates failed to demonstrate their full therapeutic potential in vivo. Reasons for unfavorable outcome include some intrinsic properties of drugs, like biodegradation, solubility, and systemic toxicity, as well as the ways in which they are administered or the time elapsed until therapeutic efficiency is demonstrated. Therefore, to develop the full therapeutic potential of drug candidates in vivo, there is a need for advanced drug delivery systems that would carry the drug specifically to the target and release it there at desired concentrations. In addition, there is a requirement for non-invasive biomedical imaging technologies allowing for rapid and sensitive evaluations of drug performance in vivo. This review will present recent developments in bioengineered drug delivery systems, highlighting the biomedical imaging tools needed to evaluate the success of drug delivery strategies.

    Citation: Daniel Calle, Duygu Yilmaz, Sebastian Cerdan, Armagan Kocer. Drug delivery from engineered organisms and nanocarriers as monitored by multimodal imaging technologies[J]. AIMS Bioengineering, 2017, 4(2): 198-222. doi: 10.3934/bioeng.2017.2.198

    Related Papers:

  • In recent years, while the research budget and development times increased for different phases of drug development, the number of clinically approved new medicines declined. In fact, many promising drug candidates failed to demonstrate their full therapeutic potential in vivo. Reasons for unfavorable outcome include some intrinsic properties of drugs, like biodegradation, solubility, and systemic toxicity, as well as the ways in which they are administered or the time elapsed until therapeutic efficiency is demonstrated. Therefore, to develop the full therapeutic potential of drug candidates in vivo, there is a need for advanced drug delivery systems that would carry the drug specifically to the target and release it there at desired concentrations. In addition, there is a requirement for non-invasive biomedical imaging technologies allowing for rapid and sensitive evaluations of drug performance in vivo. This review will present recent developments in bioengineered drug delivery systems, highlighting the biomedical imaging tools needed to evaluate the success of drug delivery strategies.


    加载中
    [1] Bunker A, Magarkar A, Viitala T (2016) Rational design of liposomal drug delivery systems, a review: combined experimental and computational studies of lipid membranes, liposomes and their PEGylation. BBA Biomembranes 1858: 2334–2352. doi: 10.1016/j.bbamem.2016.02.025
    [2] Mak IW, Evaniew N, Ghert M (2014) Lost in translation: animal models and clinical trials in cancer treatment. Am J Transl Res 6: 114–118.
    [3] Kopecek J (2010) Biomaterials and drug delivery: past, present, and future. Mol Pharm 7: 922–925. doi: 10.1021/mp1001813
    [4] Goldstein DB (2003) Pharmacogenetics in the laboratory and the clinic. N Engl J Med 348: 553–556. doi: 10.1056/NEJMe020173
    [5] Uetrecht J (2003) Screening for the potential of a drug candidate to cause idiosyncratic drug reactions. Drug Discov Today 8: 832–837. doi: 10.1016/S1359-6446(03)02816-2
    [6] Lindon JC, Nicholson JK, Holmes E, et al. (2003) Contemporary issues in toxicology the role of metabonomics in toxicology and its evaluation by the COMET project. Toxicol Appl Pharm 187: 137–146.
    [7] Shaw LM, Kaplan B, Kaufman D (1996) Toxic effects of immunosuppressive drugs: mechanisms and strategies for controlling them. Clin Chem 42: 1316–1321.
    [8] Serajuddin A (1999) Solid dispersion of poorly water-soluble drugs: early promises, subsequent problems, and recent breakthroughs. J Pharm Sci 88: 1058–1066. doi: 10.1021/js980403l
    [9] Brodie BB (1962) Difficulties in extrapolating data on metabolism of drugs from animal to man. Clin Pharm Th 3: 374–380. doi: 10.1002/cpt196233374
    [10] Dedrick RL, Flessner MF (1997) Pharmacokinetic problems in peritoneal drug administration: tissue penetration and surface exposure. J Natl Cancer I 89: 480–487.
    [11] Bach PB (2009) Limits on medicare's ability to control rising spending on cancer drugs. N Engl J Med 360: 626–633. doi: 10.1056/NEJMhpr0807774
    [12] Vermeire E, Hearnshaw H, Van Royen P, et al. (2001) Patient adherence to treatment: three decades of research: a comprehensive review. J Clin Pharm Ther 26: 331–342. doi: 10.1046/j.1365-2710.2001.00363.x
    [13] Cho EC, Glaus C, Chen J, et al. (2010) Inorganic nanoparticle-based contrast agents for molecular imaging. Trends Mol Med 16: 561–573. doi: 10.1016/j.molmed.2010.09.004
    [14] Dobrucki LW, Pan D, Smith AM (2015) Multiscale imaging of nanoparticle drug delivery. Curr Drug Targets 16: 560–570. doi: 10.2174/1389450116666150202163022
    [15] Garcia J, Tang T, Louie AY (2015) Nanoparticle-based multimodal PET/MRI probes. Nanomedicine 10: 1343–1359. doi: 10.2217/nnm.14.224
    [16] Janib SM, Moses AS, MacKay JA (2010) Imaging and drug delivery using theranostic nanoparticles. Adv Drug Deliver Rev 62: 1052–1063. doi: 10.1016/j.addr.2010.08.004
    [17] Llovet JM, Real MI, Montaña X, et al. (2002) Arterial embolisation or chemoembolisation versus symptomatic treatment in patients with unresectable hepatocellular carcinoma: a randomised controlled trial. Lancet 359: 1734–1739. doi: 10.1016/S0140-6736(02)08649-X
    [18] Zhou X, Tang Z, Wang J, et al. (2014) Doxorubicin-eluting beads versus conventional transarterialchemoembolization for the treatment of hepatocellular carcinoma: a meta-analysis. Int J Clin Exp Med 7: 3892–3903.
    [19] Pawelek JM, Low KB, Bermudes D (2003) Bacteria as tumour-targeting vectors. Lancet Oncol 4: 548–556. doi: 10.1016/S1470-2045(03)01194-X
    [20] King I, Bermudes D, Lin S, et al. (2004) Tumor-targeted salmonella expressing cytosine deaminase as an anticancer agent. Hum Gene Ther 13: 1225–1233.
    [21] Low KB, Ittensohn M, Le T, et al. (1999) Lipid a mutant salmonella with suppressed virulence and TNF-α induction retain tumor-targeting in vivo. Nat Biotechnol 17: 37–41.
    [22] Pawelek JM, Low KB, Bermudes D (1997) Tumor-targeted salmonella as a novel anticancer vector. Cancer Res 57: 4537–4544.
    [23] Schlechte H, Elbe B (1988) Recombinant plasmid DNA variation of clostridium oncolyticum- model experiments of cancerostatic gene transfer. Cent Sheet Bacteriol Microbiol Hyg Ser A 268: 347–356.
    [24] Sasaki T, Fujimori M, Hamaji Y, et al. (2006) Genetically engineered bifidobacterium longum for tumor-targeting enzyme-prodrug therapy of autochthonous mammary tumors in rats. Cancer Sci 97: 649–657. doi: 10.1111/j.1349-7006.2006.00221.x
    [25] Panteli JT, Forbes NS (2016) Engineered bacteria detect spatial profiles in glucose concentration within solid tumor cell masses. Biotechnol Bioeng 113: 2474–2484. doi: 10.1002/bit.26006
    [26] Hosseinidoust Z, Mostaghaci B, Yasa O, et al. (2016) Bioengineered and biohybrid bacteria-based systems for drug delivery. Adv Drug Deliver Rev 106: 27–44. doi: 10.1016/j.addr.2016.09.007
    [27] Akin D, Sturgis J, Ragheb K, et al. (2007) Bacteria-mediated delivery of nanoparticles and cargo into cells. Nat Nanotechnol 2: 441–449. doi: 10.1038/nnano.2007.149
    [28] Edwards MR, Carlsen RW, Zhuang J, et al. (2014) Swimming characterization of serratia marcescens for bio-hybrid micro-robotics. J MicroBio Robot 9: 47–60. doi: 10.1007/s12213-014-0072-1
    [29] Zhuang J, Carlsen RW, Sitti M (2015) pH-taxis of biohybrid microsystems. Sci Rep 5: 11403–11415. doi: 10.1038/srep11403
    [30] Lee JB, Hong J, Bonner DK, et al. (2012) Self-assembled RNA interference microsponges for efficient siRNA delivery. Nat Mater 11: 316–322. doi: 10.1038/nmat3253
    [31] Koudelka KJ, Pitek AS, Manchester M, et al. (2015) Virus-based nanoparticles as versatile nanomachines. Annu Rev Virol 2: 379–401. doi: 10.1146/annurev-virology-100114-055141
    [32] Douglas T, Young M (2006) Viruses: making friends with old foes. Science 312: 873–875. doi: 10.1126/science.1123223
    [33] Esfandiari N, Arzanani MK, Soleimani M, et al. (2015) A new application of plant virus nanoparticles as drug delivery in breast cancer. Tumor Biol 37: 1229–1236.
    [34] Cheng F, Tsvetkova IB, Khuong YL, et al. (2013) The packaging of different cargo into enveloped viral nanoparticles. Mol Pharm 10: 51–58. doi: 10.1021/mp3002667
    [35] Steinmetz NF, Mertens ME, Taurog RE, et al. (2010) Potato virus x as a novel platform for potential biomedical applications. Nano Lett 10: 305–312. doi: 10.1021/nl9035753
    [36] Aljabali AAA, Shukla S, Lomonossoff GP, et al. (2013) CPMV-DOX delivers. Mol Pharm 10: 3–10. doi: 10.1021/mp3002057
    [37] Galaway FA, Stockley PG (2013) MS2 viruslike particles: a robust, semisynthetic targeted drug delivery platform. Mol Pharm 10: 59–68. doi: 10.1021/mp3003368
    [38] Choi KM, Choi SH, Jeon H, et al. (2011) Chimeric capsid protein as a nanocarrier for siRNA delivery: stability and cellular uptake of encapsulated siRNA. Acs Nano 5: 8690–8699. doi: 10.1021/nn202597c
    [39] Choi KM, Kim K, Kwon IC, et al. (2013) Systemic delivery of siRNA by chimeric capsid protein: tumor targeting and RNAi activity in vivo. Mol Pharm 10: 18–25. doi: 10.1021/mp300211a
    [40] Kim KR, Kim DR, Lee T, et al. (2013) Drug delivery by a self-assembled DNA tetrahedron for overcoming drug resistance in breast cancer cells. Chem Commun 49: 2010–2012. doi: 10.1039/c3cc38693g
    [41] Jiang Q, Song C, Nangreave J, et al. (2012) DNA origami as a carrier for circumvention of drug resistance. J Am Chem Soc 134: 13396–13403. doi: 10.1021/ja304263n
    [42] Charoenphol P, Bermudez H (2014) Design and application of multifunctional DNA nanocarriers for therapeutic delivery. Acta Biomater 10: 1683–1691.
    [43] Kumar V, Palazzolo S, Bayda S, et al. (2016) DNA nanotechnology for cancer therapy. Theranostics 6: 710–725. doi: 10.7150/thno.14203
    [44] Angell C, Xie S, Zhang L, et al. (2016) DNA nanotechnology for precise control over drug delivery and gene therapy. Small 12: 1117–1132. doi: 10.1002/smll.201502167
    [45] Taylor AI, Beuron F, Peak CSY, et al. (2016) Nanostructures from synthetic genetic polymers. Chembiochem 17: 1107–1110. doi: 10.1002/cbic.201600136
    [46] Kim KR, Kim HY, Lee YD, et al. (2016) Self-assembled mirror DNA nanostructures for tumor-specific delivery of anticancer drugs. J Control Release 243: 121–131. doi: 10.1016/j.jconrel.2016.10.015
    [47] Kim KR, Lee T, Kim BS, et al. (2014) Utilizing the bioorthogonal base-pairing system of l-DNA to design ideal DNA nanocarriers for enhanced delivery of nucleic acid cargos. Chem Sci 5: 1533–1537. doi: 10.1039/C3SC52601A
    [48] Bangham AD, Standish MM, Watkins JC (1965) Diffusion of univalent ions across the lamellae of swollen phospholipids. J Mol Biol 13: 238–IN27. doi: 10.1016/S0022-2836(65)80093-6
    [49] Allen TM, Cullis PR (2004) Drug delivery systems: entering the mainstream. Science 303: 1818–1822. doi: 10.1126/science.1095833
    [50] Pattni BS, Chupin VV, Torchilin VP (2015) New developments in liposomal drug delivery. Chem Rev 115: 10938–10966. doi: 10.1021/acs.chemrev.5b00046
    [51] Leung SJ, Romanowski M (2012) Light-activated content release from liposomes. Theranostics 2: 1020–1036. doi: 10.7150/thno.4847
    [52] Ng LT, Yuba E, Kono K (2009) Modification of liposome surface with pH-responsive polyampholytes for the controlled-release of drugs. Res Chem Intermediat 35: 1015–1025. doi: 10.1007/s11164-009-0089-6
    [53] Obata Y, Tajima S, Takeoka S (2010) Evaluation of pH-responsive liposomes containing amino acid-based zwitterionic lipids for improving intracellular drug delivery in vitro and in vivo. J Control Release 142: 267–276. doi: 10.1016/j.jconrel.2009.10.023
    [54] Landon CD, Park JY, Needham D, et al. (2011) Nanoscale drug delivery and hyperthermia: the materials design and preclinical and clinical testing of low temperature-sensitive liposomes used in combination with mild hyperthermia in the treatment of local cancer. Open Nanomed J 3: 38–64.
    [55] Stubbs M, McSheehy PM, Griffiths JR (1999) Causes and consequences of acidic pH in tumors: a magnetic resonance study. Adv Enzyme Regul 39: 13–30. doi: 10.1016/S0065-2571(98)00018-1
    [56] Zhang J, Tao W, Chen Y, et al. (2015) Doxorubicin-loaded star-shaped copolymer PLGA-vitamin E tpgs nanoparticles for lung cancer therapy. J Mater Sci Mater M 26: 165. doi: 10.1007/s10856-015-5498-z
    [57] Kocer A (2007) A remote controlled valve in liposomes for triggered liposomal release. J Liposome Res 17: 219–225. doi: 10.1080/08982100701528203
    [58] Kocer A, Walko M, Bulten E, et al. (2006) Rationally designed chemical modulators convert a bacterial channel protein into a pH-sensory valve. Angew Chem Int Edit 45: 3126–3130. doi: 10.1002/anie.200503403
    [59] Kocer A, Walko M, Meijberg W, et al. (2005) A light-actuated nanovalve derived from a channel protein. Science 309: 755–758. doi: 10.1126/science.1114760
    [60] Pacheco TJ, Mukherjee N, Walko M, et al. (2015) Image guided drug release from pH-sensitive Ion channel-functionalized stealth liposomes into an in vivo glioblastoma model. Nanomedicine 11: 1345–1354.
    [61] Shriver LP, Koudelka KJ, Manchester M (2009) Viral nanoparticles associate with regions of inflammation and blood brain barrier disruption during CNS infection. J Neuroimmunol 211: 66–72. doi: 10.1016/j.jneuroim.2009.03.015
    [62] Rae CS, Khor IW, Wang Q, et al. (2005) Systemic trafficking of plant virus nanoparticles in mice via the oral route. Virology 343: 224–235. doi: 10.1016/j.virol.2005.08.017
    [63] Lewis JD, Destito G, Zijlstra A, et al. (2006) Viral nanoparticles as tools for intravital vascular imaging. Nat Med 12: 354–360. doi: 10.1038/nm1368
    [64] Leong HS, Steinmetz NF, Ablack A, et al. (2010) Intravital imaging of embryonic and tumor neovasculature using viral nanoparticles. Nat Protoc 5: 1406–1417. doi: 10.1038/nprot.2010.103
    [65] Manchester M, Singh P (2006) Virus-based nanoparticles (VNPs): platform technologies for diagnostic imaging. Adv Drug Deliver Rev 58: 1505–1522. doi: 10.1016/j.addr.2006.09.014
    [66] Cheng F, Mukhopadhyay S (2011) Generating enveloped virus-like particles with in vitro assembled cores. Virology 413: 153–160. doi: 10.1016/j.virol.2011.02.001
    [67] Weissleder R, Mahmood U (2001) Molecular imaging. Radiology 219: 316–333. doi: 10.1148/radiology.219.2.r01ma19316
    [68] Bao G, Mitragotri S, Tong S (2013) Multifunctional nanoparticles for drug delivery and molecular imaging. Annu Rev Biomed Eng 15: 253–282. doi: 10.1146/annurev-bioeng-071812-152409
    [69] Ding H, Wu F (2012) Image guided biodistribution of drugs and drug delivery. Theranostics 2: 1037–1039. doi: 10.7150/thno.5321
    [70] Brys AK, Gowda R, Loriaux DB, et al. (2016) Nanotechnology-based strategies for combating toxicity and resistance in melanoma therapy. Biotechnol Adv 34: 565–577. doi: 10.1016/j.biotechadv.2016.01.004
    [71] Gowda R, Jones NR, Banerjee S, et al. (2013) Use of nanotechnology to develop multi-drug inhibitors for cancer therapy. J Nanomed Nanotechnol 4: 184.
    [72] Medina FJL, Giulianotti MA, Welmaker GS, et al. (2013) Shifting from the single to the multitarget paradigm in drug discovery. Drug Discov Today 18: 495–501. doi: 10.1016/j.drudis.2013.01.008
    [73] Yuan Y, Cai T, Xia X, et al. (2016) Nanoparticle delivery of anticancer drugs overcomes multidrug resistance in breast cancer. Drug Deliv 23: 3350–3357. doi: 10.1080/10717544.2016.1178825
    [74] Zheng H, Fridkin M, Youdim M (2014) From single target to multitarget/network therapeutics in Alzheimer's therapy. Pharmaceuticals 7: 113–135. doi: 10.3390/ph7020113
    [75] Pawar S, Shevalkar G, Vavia P (2016) Glucosamine-anchored doxorubicin-loaded targeted nano-niosomes: pharmacokinetic, toxicity and pharmacodynamic evaluation. J Drug Target 24: 730–743.
    [76] Blanco E, Shen H, Ferrari M (2015) Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol 33: 941–951. doi: 10.1038/nbt.3330
    [77] Shin TH, Choi Y, Kim S, et al. (2015) Recent advances in magnetic nanoparticle-based multi-modal imaging. Chem Soc Rev 44: 4501–4516. doi: 10.1039/C4CS00345D
    [78] Johnsen KB, Moos T (2016) Revisiting nanoparticle technology for blood-brain barrier transport: unfolding at the endothelial gate improves the fate of transferrin receptor-targeted liposomes. J Control Release 222: 32–46. doi: 10.1016/j.jconrel.2015.11.032
    [79] Grabrucker AM, Ruozi B, Belletti D, et al. (2016) Nanoparticle transport across the blood brain barrier. Tissue Barriers 4: e1153568–e1153571. doi: 10.1080/21688370.2016.1153568
    [80] Ho D, Wang C-HK, Chow EKH (2015) Nanodiamonds: the intersection of nanotechnology, drug development, and personalized medicine. Sci Adv 1: e1500439–e1500439.
    [81] Webb S (1988) The physics of medical imaging, CRC Press.
    [82] Farncombe T, Iniewsky K (2013) Medical imaging: technology ad applications, CRC Press, 732.
    [83] Caravan P, Ellison JJ, McMurry TJ, et al. (1999) Gadolinium(III) chelates as MRI contrast agents: structure, dynamics, and applications. Chem Rev 99: 2293–2352. doi: 10.1021/cr980440x
    [84] Pavel DG, Zimmer M, Patterson VN (1977) In vivo labeling of red blood cells with 99mTc: a new approach to blood pool visualization. J Nucl Med 18: 305–308.
    [85] Hahn MA, Singh AK, Sharma P, et al. (2011) Nanoparticles as contrast agents for in-vivo bioimaging: current status and future perspectives. Anal Bioanal Chem 399: 3–27. doi: 10.1007/s00216-010-4207-5
    [86] Chakravarty R, Hong H, Cai W (2014) Positron emission tomography image-guided drug delivery: current status and future perspectives. Mol Pharm 11: 3777–3797. doi: 10.1021/mp500173s
    [87] de Smet M, Langereis S, van den Bosch S et al. (2013) SPECT/CT imaging of temperature-sensitive liposomes for MR-image guided drug delivery with high intensity focused ultrasound. J Control Release 169: 82–90.
    [88] Gao X, Cui Y, Levenson RM, et al. (2004) In vivo cancer targeting and imaging with semiconductor quantum dots. Nat Biotechnol 22: 969–976. doi: 10.1038/nbt994
    [89] Weissleder R, Tung CH, Mahmood U, et al. (1999) In vivo imaging of tumors with protease-activated near-infrared fluorescent probes. Nat Biotechnol 17: 375–378. doi: 10.1038/7933
    [90] Calle D, Negri V, Ballesteros P, et al. (2015) Magnetoliposomes loaded with poly-unsaturated fatty acids as novel theranostic anti-inflammatory formulations. Theranostics 5: 489–503. doi: 10.7150/thno.10069
    [91] Heinle SK, Noblin J, Goree BP, et al. (2000) Assessment of myocardial perfusion by harmonic power doppler imaging at rest and during adenosine stress: comparison with (99 m) Tc-sestamibi SPECT imaging. Circulation 102: 55–60. doi: 10.1161/01.CIR.102.1.55
    [92] Urtasun RC, Parliament MB, McEwan AJ, et al. (1996) Measurement of hypoxia in human tumours by non-invasive spect imaging of iodoazomycin arabinoside. Brit J Cancer Suppl 27: S209–S212.
    [93] Leitha T, Glaser C, Pruckmayer M, et al. (1998) Technetium-99m-MIBI in primary and recurrent head and neck tumors: contribution of bone SPECT image fusion. J Nucl Med 39: 1166–1171.
    [94] Tharp K, Israel O, Hausmann J, et al. (2004) Impact of 131I-SPECT/CT images obtained with an integrated system in the follow-up of patients with thyroid carcinoma. Eur J Nucl Med Mol I 31: 1435–1442.
    [95] Fukuyama H, Ouchi Y, Matsuzaki S, et al. (1997) Brain functional activity during gait in normal subjects: a SPECT study. Neurosci Lett 228: 183–186. doi: 10.1016/S0304-3940(97)00381-9
    [96] Rudin M, Weissleder R (2003) Molecular imaging in drug discovery and development. Nat Rev Drug Discov 2: 123–131. doi: 10.1038/nrd1007
    [97] Bushberg JT, Boone JM (2011) The essential physics of medical imaging, Lippincott Williams & Wilkins.
    [98] Finnema SJ, Scheinin M, Shahid M, et al. (2015) Application of cross-species PET imaging to assess neurotransmitter release in brain. Psychopharmacology 232: 4129–4157. doi: 10.1007/s00213-015-3938-6
    [99] Haubner R, Maschauer S, Prante O (2014) PET radiopharmaceuticals for imaging integrin expression: tracers in clinical studies and recent developments. Biomed Res Int 2014: 871609–871617.
    [100] Tateishi U, Oka T, Inoue T (2012) Radiolabeled RGD peptides as integrin alpha(v)beta3-targeted PET tracers. Curr Med Chem 19: 3301–3309. doi: 10.2174/092986712801215937
    [101] Wagner S, Breyholz HJ, Law MP, et al. (2007) Novel fluorinated derivatives of the broad-spectrum MMP inhibitors N-hydroxy-2(R)-[[(4-methoxyphenyl)sulfonyl](benzyl)- and (3-picolyl)-amino]-3-methyl-butanamide as potential tools for the molecular imaging of activated MMPs with PET. J Med Chem 50: 5752–5764. doi: 10.1021/jm0708533
    [102] Stacy MR, Maxfield MW, Sinusas AJ (2012) Targeted molecular imaging of angiogenesis in PET and SPECT: a review. Yale J Biol Med 85: 75–86.
    [103] Ripa RS, Pedersen SF, Kjaer A (2016) PET/MR imaging in vascular disease atheroscleroaia and inflammation. Positron Emission Tomo 11: 479–488.
    [104] Wu C, Li F, Niu G, et al. (2013) PET imaging of inflammation biomarkers. Theranostics 3: 448–466. doi: 10.7150/thno.6592
    [105] Pacheco TJ, Calle D, Lizarbe B, et al. (2011) Environmentally sensitive paramagnetic and diamagnetic contrast agents for nuclear magnetic resonance imaging and spectroscopy. Curr Top Med Chem 11: 115–130. doi: 10.2174/156802611793611904
    [106] Lanza GM, Moonen C, Baker JR, et al. (2013) Assessing the barriers to image-guided drug delivery. WIRE Nanomed Nanobiotechnol 6: 1–14.
    [107] Na HB, Song IC, Hyeon T (2009) Inorganic nanoparticles for MRI contrast agents. Adv Mater 21: 2133–2148.
    [108] Bonnemain B (1998) Superparamagnetic agents in magnetic resonance imaging: physicochemical characteristics and clinical applications: a review. J Drug Target 6: 167–174. doi: 10.3109/10611869808997890
    [109] Yu Y, Sun D (2010) Superparamagnetic iron oxide nanoparticle "theranostics" for multimodality tumor imaging, gene delivery, targeted drug and prodrug delivery. Expert Rev Clin Pharmaco 3: 117–130. doi: 10.1586/ecp.09.39
    [110] Liu F, Laurent S, Fattahi H, et al. (2011) Superparamagnetic nanosystems based on iron oxide nanoparticles for biomedical imaging. Nanomedicine 6: 519–528. doi: 10.2217/nnm.11.16
    [111] Al-Nahhas A, Win Z, Singh Q, et al. (2006) The role of 18F-FDG PET in oncology: clinical and resource implications. Nucl Med Rev Cent East Eur 9: 1–5.
    [112] Chopra A (2004) 18F-Labeled N-succinimidyl-4-fluorobenzoate-conjugated rat anti-mouse vascular endothelial growth factor receptor 2 monoclonal antibody linked to microbubbles, MICAD.
    [113] Gupta H, Aqil M, Khar RK, et al. (2010) Sparfloxacin-loaded PLGA nanoparticles for sustained ocular drug delivery. Nanomedicine 6: 324–333.
    [114] Luo Q, Zhao J, Zhang X, et al. (2011) Nanostructured lipid carrier (NLC) coated with chitosan oligosaccharides and its potential use in ocular drug delivery system. Int J Pharm 403: 185–191. doi: 10.1016/j.ijpharm.2010.10.013
    [115] Möller W, Schuschnig U, Celik G, et al. (2013) Topical drug delivery in chronic rhinosinusitis patients before and after sinus surgery using pulsating aerosols. Plos One 8: e74991. doi: 10.1371/journal.pone.0074991
    [116] Gallamini A, Zwarthoed C, Borra A (2014) Positron emission tomography (PET) in oncology. Cancers 6: 1821–1889. doi: 10.3390/cancers6041821
    [117] Xiao Y, Hong H, Javadi A, et al. (2012) Multifunctional unimolecular micelles for cancer-targeted drug delivery and positron emission tomography imaging. Biomaterials 33: 3071–3082. doi: 10.1016/j.biomaterials.2011.12.030
    [118] Guo J, Hong H, Chen G, et al. (2014) Theranostic unimolecular micelles based on brush-shaped amphiphilic block copolymers for tumor-targeted drug delivery and positron emission tomography imaging. Acs Appl Mater Interface 6: 21769–21779. doi: 10.1021/am5002585
    [119] Hong H, Zhang Y, Engle JW, et al. (2012) In vivo targeting and positron emission tomography imaging of tumor vasculature with (66)Ga-labeled nano-graphene. Biomaterials 33: 4147–4156. doi: 10.1016/j.biomaterials.2012.02.031
    [120] Liu Z, Cal W, He L, et al. (2007) In vivo biodistribution and highly efficient tumour targeting of carbon nanotubes in mice. Nat Nanotechnol 2: 47–52. doi: 10.1038/nnano.2006.170
    [121] Blasberg RG (2003) In vivo molecular-genetic imaging: multi-modality nuclear and optical combinations. Nucl Med Biol 30: 879–888. doi: 10.1016/S0969-8051(03)00115-X
    [122] Hoffman RM (2015) Application of GFP imaging in cancer. Lab Invest 95: 432–452. doi: 10.1038/labinvest.2014.154
    [123] Kocher B, Piwnica WD (2013) Illuminating cancer systems with genetically engineered mouse models and coupled luciferase reporters in vivo. Cancer Discov 3: 616–629.
    [124] Wang C, Cheng L, Liu Z (2011) Drug delivery with upconversion nanoparticles for multi-functional targeted cancer cell imaging and therapy. Biomaterials 32: 1110–1120. doi: 10.1016/j.biomaterials.2010.09.069
    [125] Yuan Y, Liu B (2014) Self-assembled nanoparticles based on PEGylated conjugated polyelectrolyte and drug molecules for image-guided drug delivery and photodynamic therapy. Acs Appl Mater Interface 6: 14903–14910. doi: 10.1021/am5020925
    [126] Cool SK, Geers B, Roels S, et al. (2013) Coupling of drug containing liposomes to microbubbles improves ultrasound triggered drug delivery in mice. J Control Release 172: 885–893.
    [127] Chen ML, He YJ, Chen XW, et al. (2012) Quantum dots conjugated with Fe3O4-filled carbon nanotubes for cancer-targeted imaging and magnetically guided drug delivery. Langmuir 28: 16469–16476.
    [128] Sun X, Liu Z, Welsher K, et al. (2008) Nano-graphene oxide for cellular imaging and drug delivery. Nano Res 1: 203–212. doi: 10.1007/s12274-008-8021-8
    [129] Shen Z, Wu A, Chen X (2016) Iron oxide nanoparticle based contrast agents for magnetic resonance imaging. Mol Pharm, DOI: 10.1021/acs.molpharmaceut.6b00839.
    [130] Hermann P, Kotek J, Kubíček V, et al. (2008) Gadolinium(III) complexes as MRI contrast agents : ligand design and properties of the complexes. Dalton Trans 23: 3027–3047.
    [131] Kaida S, Cabral H, Kumagai M, et al. (2010) Visible drug delivery by supramolecular nanocarriers directing to single-platformed diagnosis and therapy of pancreatic tumor model. Cancer Res 70: 7031–7041. doi: 10.1158/0008-5472.CAN-10-0303
    [132] Negussie AH, Yarmolenko PS, Partanen A, et al. (2011) Formulation and characterisation of magnetic resonance imageable thermally sensitive liposomes for use with magnetic resonance-guided high intensity focused ultrasound. Int J Hyperther 27: 140–155. doi: 10.3109/02656736.2010.528140
    [133] Tagami T, Foltz WD, Ernsting MJ, et al. (2011) MRI monitoring of intratumoral drug delivery and prediction of the therapeutic effect with a multifunctional thermosensitive liposome. Biomaterials 32: 6570–6578. doi: 10.1016/j.biomaterials.2011.05.029
    [134] Ma X, Tao H, Yang K, et al. (2012) A functionalized graphene oxide-iron oxide nanocomposite for magnetically targeted drug delivery, photothermal therapy, and magnetic resonance imaging. Nano Res 5: 199–212. doi: 10.1007/s12274-012-0200-y
    [135] Al-Jamal WT, Kostarelos K (2007) Liposome-nanoparticle hybrids for multimodal diagnostic and therapeutic applications. Nanomedicine 2: 85–98. doi: 10.2217/17435889.2.1.85
    [136] Mikhaylov G, Mikac U, Magaeva AA, et al. (2011) Ferri-liposomes as an MRI-visible drug-delivery system for targeting tumours and their microenvironment. Nat Nanotechnol 6: 594–602. doi: 10.1038/nnano.2011.112
    [137] Xu H, Cheng L, Wang C, et al. (2011) Polymer encapsulated upconversion nanoparticle/iron oxide nanocomposites for multimodal imaging and magnetic targeted drug delivery. Biomaterials 32: 9364–9373. doi: 10.1016/j.biomaterials.2011.08.053
    [138] Yang X, Hong H, Grailer JJ, et al. (2011) cRGD-functionalized, DOX-conjugated, and ⁶⁴Cu-labeled superparamagnetic iron oxide nanoparticles for targeted anticancer drug delivery and PET/MR imaging. Biomaterials 32: 4151–4160. doi: 10.1016/j.biomaterials.2011.02.006
    [139] Claesen J, Fischbach MA (2015) Synthetic microbes as drug delivery systems. Acs Synth Biol 4: 358–364. doi: 10.1021/sb500258b
    [140] Sotoudeh H, Sharma A, Fowler KJ, et al. (2016) Clinical application of PET/MRI in oncology. J Magn Reson Imaging 44: 265–276. doi: 10.1002/jmri.25161
    [141] Pan D, Caruthers SD, Chen J, et al. (2010) Nanomedicine strategies for molecular targets with MRI and optical imaging. Future Med Chem 2: 471–490. doi: 10.4155/fmc.10.5
    [142] Culver J, Akers W, Achilefu S (2008) Multimodality molecular imaging with combined optical and SPECT/PET modalities. J Nucl Med 49: 169–172. doi: 10.2967/jnumed.107.043331
  • Reader Comments
  • © 2017 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(7176) PDF downloads(1360) Cited by(4)

Article outline

Figures and Tables

Figures(4)  /  Tables(2)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog