1,611
Views
0
CrossRef citations to date
0
Altmetric
Review

Controlled Actuation of Therapeutic nanoparticles: an Update on Recent Progress

, , , , &
Pages 335-352 | Received 14 Jan 2016, Accepted 23 Mar 2016, Published online: 14 Apr 2016

References

  • Delehanty JB Mattoussi H Medintz IL . Delivering quantum dots into cells: strategies, progress and remaining issues. Anal. Bioanal. Chem.393 (4), 1091–1105 (2009).
  • Delehanty JB Susumu K Manthe RL Algar WR Medintz IL . Active cellular sensing with quantum dots: transitioning from research tool to reality; a review. Anal. Chim. Acta750, 63–81 (2012).
  • Ruedas-Rama MJ Walters JD Orte A Hall EA . Fluorescent nanoparticles for intracellular sensing: a review. Anal. Chim. Acta751, 1–23 (2012).
  • Roldo M . Carbon nanotubes in drug delivery: just a carrier?Ther. Deliv.7 (2), 55–57 (2016).
  • Nazarenus M Zhang Q Soliman MG et al. In vitro interaction of colloidal nanoparticles with mammalian cells: what have we learned thus far? Beilstein. J. Nanotechnol. 5, 1477–1490 (2014).
  • Paranjpe M Müller-Goymann C . Nanoparticle-mediated pulmonary drug delivery: a review. Int. J. Mol. Sci.15 (4), 5852 (2014).
  • Ryvolova M Chomoucka J Drbohlavova J et al. Modern micro and nanoparticle-based imaging techniques. Sensors12 (11), 14792 (2012).
  • Patel SC Lee S Lalwani G Suhrland C Chowdhury SM Sitharaman B . Graphene-based platforms for cancer therapeutics. Ther. Deliv.7 (2), 101–116 (2016).
  • Delehanty JB Breger JC Gemmill KB Stewart MH Medintz IL . Controlling the actuation of therapeutic nanomaterials: enabling nanoparticle-mediated drug delivery. Ther. Deliv.4 (11), 1411–1429 (2013).
  • Delehanty JB Medintz IL . Controlled actuation of therapeutic nanoparticles: moving beyond passive delivery modalities. Ther. Deliv.4 (2), 127–129 (2013).
  • Hu X Tian J Liu T Zhang G Liu S . Photo-triggered release of caged camptothecin prodrugs from dually responsive shell cross-linked micelles. Macromolecules46 (15), 6243–6256 (2013).
  • Huu VAN Luo J Zhu J et al. Light-responsive nanoparticle depot to control release of a small molecule angiogenesis inhibitor in the posterior segment of the eye. J. Control. Rel.200 (0), 71–77 (2015).
  • Lee J-H Chen K-J Noh S-H et al. On-demand drug release system for in vivo cancer rreatment through self-assembled magnetic nanoparticles. Angew Chem. Int. Ed. Engl.52 (16), 4384–4388 (2013).
  • Huang P Song H Wang W et al. Integrin-rargeted zwitterionic polymeric nanoparticles with acid-induced disassembly property for enhanced drug accumulation and release in tumor. Biomacromolecules15 (8), 3128–3138 (2014).
  • Wang Y Han N Zhao Q et al. Redox-responsive mesoporous silica as carriers for controlled drug delivery: a comparative study based on silica and PEG gatekeepers. Eur. J. Pharm. Sci.72, 12–20 (2015).
  • Matsumura Y Yoshikata K Kunisaki S Tsuchido T . Mode of bactericidal action of silver zeolite and its comparison with that of silver nitrate. Appl. Environ. Microb.69 (7), 4278–4281 (2003).
  • Chen X Schluesener HJ . Nanosilver: a nanoproduct in medical application. Toxicol. Lett.176 (1), 1–12 (2008).
  • Prabhu S Poulose E . Silver nanoparticles: mechanism of antimicrobial action, synthesis, medical applications, and toxicity effects. Int. Nano Lett.2 (1), 1–10 (2012).
  • Kasivelu Govindaraju KK Alsagaby SA Ganesan SMP . Green synthesis of silver nanoparticles for selective toxicity towards cancer cells. IET Nanobiotechnol.9 (6), 325–330 (2015).
  • Suliman YA Ali D Alarifi S Harrath AH Mansour L Alwasel SH . Evaluation of cytotoxic, oxidative stress, proinflammatory and genotoxic effect of silver nanoparticles in human lung epithelial cells. Environ. Toxicol.30 (2), 149–160 (2015).
  • Zhang H Wang X Wang M et al. Mammalian cells exhibit a range of sensitivities to silver nanoparticles that are partially explicable by variations in antioxidant defense and metallothionein expression. Small11 (31), 3797–3805 (2015).
  • Oh E Delehanty JB Sapsford KE et al. Cellular uptake and fate of PEGylated gold nanoparticles is dependent on both cell-penetration peptides and particle size. ACS Nano.5 (8), 6434–6448 (2011).
  • Alkilany AM Murphy CJ . Toxicity and cellular uptake of gold nanoparticles: what we have learned so far?J. Nanopart. Res.12 (7), 2313–2333 (2010).
  • Seo JM Kim EB Hyun MS Kim BB Park TJ . Self-assembly of biogenic gold nanoparticles and their use to enhance drug delivery into cells. Colloids Surf. B Biointerfaces135, 27–34 (2015).
  • Elbialy NS Fathy MM Khalil WM . Doxorubicin loaded magnetic gold nanoparticles for in vivo targeted drug delivery. Int. J. Pharm.490 (1–2), 190–199 (2015).
  • Afifi MM Austin LA Mackey MA El-Sayed MA . XAV939: from a small inhibitor to a potent drug bioconjugate when delivered by gold nanoparticles. Bioconjug. Chem.25 (2), 207–215 (2014).
  • Mu Q Kievit FM Kant RJ Lin G Jeon M Zhang M . Anti-HER2/neu peptide-conjugated iron oxide nanoparticles for targeted delivery of paclitaxel to breast cancer cells. Nanoscale7 (43), 18010–18014 (2015).
  • Hsiao M-H Mu Q Stephen ZR Fang C Zhang M . Hexanoyl-Chitosan-PEG Copolymer Coated Iron Oxide Nanoparticles for Hydrophobic Drug Delivery. ACS Macro. Lett.4 (4), 403–407 (2015).
  • Kossatz S Grandke J Couleaud P et al. Efficient treatment of breast cancer xenografts with multifunctionalized iron oxide nanoparticles combining magnetic hyperthermia and anti-cancer drug delivery. Breast Cancer Res.17, 66 (2015).
  • Chen X Tang Y Cai B Fan H . ‘One-pot’ synthesis of multifunctional GSH-CdTe quantum dots for targeted drug delivery. Nanotechnology25 (23), 235101 (2014).
  • Wang X Sun X Lao J et al. Multifunctional graphene quantum dots for simultaneous targeted cellular imaging and drug delivery. Colloids Surf. B Biointerfaces122, 638–644 (2014).
  • Murata M Narahara S Kawano T et al. Design and function of engineered protein nanocages as a drug delivery system for targeting pancreatic cancer cells via neuropilin-1. Mol. Pharm.12 (5), 1422–1430 (2015).
  • Bellini M Mazzucchelli S Galbiati E et al. Protein nanocages for self-triggered nuclear delivery of DNA-targeted chemotherapeutics in Cancer Cells. J. Control. Rel.196, 184–196 (2014).
  • Kambhampati SP Mishra MK Mastorakos P Oh Y Lutty GA Kannan RM . Intracellular delivery of dendrimer triamcinolone acetonide conjugates into microglial and human retinal pigment epithelial cells. Eur. J. Pharm. Biopharm.95 (Pt B), 239–249 (2015).
  • Scarano W de Souza P Stenzel MH . Dual-drug delivery of curcumin and platinum drugs in polymeric micelles enhances the synergistic effects: a double act for the treatment of multidrug-resistant cancer. Biomater. Sci.3 (1), 163–174 (2015).
  • Kroon J Buijs JT van der Horst G et al. Liposomal delivery of dexamethasone attenuates prostate cancer bone metastatic tumor growth in vivo. Prostate75 (8), 815–824 (2015).
  • Rodrigues FP Carneiro ZA Mascharak P Curti C da Silva RS . Incorporation of a ruthenium nitrosyl complex into liposomes, the nitric oxide released from these liposomes and HepG2 cell death mechanism. Coord. Chem. Rev.306, 701–707 (2016).
  • Ding Q Si X Liu D et al. Targeting and liposomal drug delivery to CD40L expressing T cells for treatment of autoimmune diseases. J. Control. Rel.207, 86–92 (2015).
  • Wei X Gao J Zhan C et al. Liposome-based glioma targeted drug delivery enabled by stable peptide ligands. J. Control. Rel.218, 13–21 (2015).
  • Dasa SS Suzuki R Gutknecht M et al. Development of target-specific liposomes for delivering small molecule drugs after reperfused myocardial infarction. J. Control. Rel.220 (Pt A), 556–567 (2015).
  • Moles E Urban P Jimenez-Diaz MB et al. Immunoliposome-mediated drug delivery to Plasmodium-infected and non-infected red blood cells as a dual therapeutic/prophylactic antimalarial strategy. J. Control. Rel.210, 217–229 (2015).
  • Spillmann CM Naciri J Algar WR Medintz IL Delehanty JB . Multifunctional liquid crystal nanoparticles for intracellular fluorescent imaging and drug delivery. ACS Nano.8 (7), 6986–6997 (2014).
  • Wang C-F Mäkilä EM Kaasalainen MH et al. Dual-drug delivery by porous silicon nanoparticles for improved cellular uptake, sustained release, and combination therapy. Acta Biomater.16, 206–214 (2015).
  • Khandelia R Bhandari S Pan UN Ghosh SS Chattopadhyay A . Gold nanocluster embedded albumin nanoparticles for two-photon imaging of cancer cells accompanying drug delivery. Small11 (33), 4075–4081 (2015).
  • Nigam P Waghmode S Louis M Wangnoo S Chavan P Sarkar D . Graphene quantum dots conjugated albumin nanoparticles for targeted drug delivery and imaging of pancreatic cancer. J. Mater. Chem. B2 (21), 3190–3195 (2014).
  • Park E-J Umh H Choi D-H et al. Magnetite- and maghemite-induced different toxicity in murine alveolar macrophage cells. Arch. Toxicol.88 (8), 1607–1618 (2014).
  • Soenen SJ De Cuyper M De Smedt SC Braeckmans K . Chapter ten–investigating the toxic effects of iron oxide nanoparticles. In : Methods Enzymol. NejatD ( Ed.). Academic Press195–224 (2012).
  • Hayashi K Nakamura M Miki H et al. Magnetically responsive smart nanoparticles for cancer treatment with a combination of magnetic hyperthermia and remote-control drug release. Theranostics4 (8), 834–844 (2014).
  • Tietze R Lyer S Dürr S et al. Efficient drug-delivery using magnetic nanoparticles–biodistribution and therapeutic effects in tumour bearing rabbits. Nanomedicine9 (7), 961–971 (2013).
  • Di Corato R Béalle G Kolosnjaj-Tabi J et al. Combining magnetic hyperthermia and photodynamic therapy for tumor ablation with photoresponsive magnetic liposomes. ACS Nano.9 (3), 2904–2916 (2015).
  • Qin Y Chen J Bi Y et al. Near-infrared light remote-controlled intracellular anti-cancer drug delivery using thermo/pH sensitive nanovehicle. Acta Biomater.17 (0), 201–209 (2015).
  • Timko BP Arruebo M Shankarappa SA et al. Near-infrared-actuated devices for remotely controlled drug delivery. Proc. Natl Acad. Sci. USA111 (4), 1349–1354 (2014).
  • Noh MS Lee S Kang H et al. Target-specific near-IR induced drug release and photothermal therapy with accumulated Au/Ag hollow nanoshells on pulmonary cancer cell membranes. Biomaterials45 (0), 81–92 (2015).
  • Yin W Yan L Yu J et al. High-throughput synthesis of single-layer MoS2 nanosheets as a near-infrared photothermal-triggered drug delivery for effective eancer therapy. ACS Nano.8 (7), 6922–6933 (2014).
  • Lugo K Miao X Rieke F Lin LY . Remote switching of cellular activity and cell signaling using light in conjunction with quantum dots. Biomed. Opt. Express3 (3), 447–454 (2012).
  • Carvalho-de-Souza João L Treger Jeremy S Dang B Kent Stephen BH Pepperberg David R Bezanilla F . Photosensitivity of neurons enabled by cell-targeted gold nanoparticles. Neuron86 (1), 207–217 (2015).
  • Packer AM Roska B Hausser M . Targeting neurons and photons for optogenetics. Nat. Neurosci.16 (7), 805–815 (2013).
  • Husseini GA Pitt WG Martins AM . Ultrasonically triggered drug delivery: breaking the barrier. Colloids Surf. B Biointerface123, 364–386 (2014).
  • Sirsi SR Borden MA . State-of-the-art materials for ultrasound-triggered drug delivery. Adv. Drug Deliv. Rev.72, 3–14 (2014).
  • Di J Price J Gu X Jiang X Jing Y Gu Z . Ultrasound-triggered regulation of blood glucose levels using injectable nano-network. Adv. Healthc. Mater.3 (6), 811–816 (2014).
  • Yang P Li D Jin S et al. Stimuli-responsive biodegradable poly(methacrylic acid) based nanocapsules for ultrasound traced and triggered drug delivery system. Biomaterials35 (6), 2079–2088 (2014).
  • Huebsch N Kearney CJ Zhao X et al. Ultrasound-triggered disruption and self-healing of reversibly cross-linked hydrogels for drug delivery and enhanced chemotherapy. Proc. Natl Acad Sci. USA111 (27), 9762–9767 (2014).
  • Ninomiya K Yamashita T Kawabata S Shimizu N . Targeted and ultrasound-triggered drug delivery using liposomes co-modified with cancer cell-targeting aptamers and a thermosensitive polymer. Ultrason. Sonochem.21 (4), 1482–1488 (2014).
  • Liang X Gao J Jiang L et al. Nanohybrid liposomal cerasomes with good physiological stability and rapid temperature responsiveness for high intensity focused ultrasound triggered local chemotherapy of cancer. ACS Nano.9 (2), 1280–1293 (2015).
  • Gao D Xu M Cao Z et al. Ultrasound-triggered phase-transition cationic nanodroplets for enhanced gene delivery. ACS Appl. Mater. Interfaces7 (24), 13524–13537 (2015).
  • Wang T-Y Choe JW Pu K et al. Ultrasound-guided delivery of microRNA loaded nanoparticles into cancer. J. Control. Rel.203, 99–108 (2015).
  • Wong PT Choi SK . Mechanisms of drug release in nanotherapeutic delivery systems. Chem. Rev.115 (9), 3388–3432 (2015).
  • Kato Y Ozawa S Miyamoto C et al. Acidic extracellular microenvironment and cancer. Cancer Cell. Int.13 (1), 1–8 (2013).
  • Liang K Such GK Johnston AP et al. Endocytic pH-triggered degradation of nanoengineered multilayer capsules. Adv. Mater.26 (12), 1901–1905 (2014).
  • Samanta D Meiser JL Zare RN . Polypyrrole nanoparticles for tunable, pH-sensitive and sustained drug release. Nanoscale7 (21), 9497–9504 (2015).
  • Xu X Li Y Li H et al. Smart nanovehicles based on pH-triggered disassembly of supramolecular peptide-amphiphiles for efficient intracellular drug delivery. Small10 (6), 1133–1140 (2014).
  • Bhang SH Han J Jang HK et al. pH-triggered release of manganese from MnAu nanoparticles that enables cellular neuronal differentiation without cellular toxicity. Biomaterials55, 33–43 (2015).
  • Zolata H Abbasi Davani F Afarideh H . Synthesis characterization and theranostic evaluation of Indium-111 labeled multifunctional superparamagnetic iron oxide nanoparticles. Nucl. Med. Biol.42 (2), 164–170 (2015).
  • Liu J Zhang B Luo Z et al. Enzyme responsive mesoporous silica nanoparticles for targeted tumor therapy in vitro and in vivo. Nanoscale7 (8), 3614–3626 (2015).
  • Zhao Y Burkert SC Tang Y et al. Nano-gold corking and enzymatic uncorking of carbon nanotube cups. J. Am. Chem. Soc.137 (2), 675–684 (2015).
  • Hou XF Chen Y Liu Y . Enzyme-responsive protein/polysaccharide supramolecular nanoparticles. Soft Matter.11 (12), 2488–2493 (2015).
  • Medina SH Chevliakov MV Tiruchinapally G Durmaz YY Kuruvilla SP Elsayed ME . Enzyme-activated nanoconjugates for tunable release of doxorubicin in hepatic cancer cells. Biomaterials34 (19), 4655–4666 (2013).
  • Guarnieri D Biondi M Yu H et al. Tumor-activated prodrug (TAP)-conjugated nanoparticles with cleavable domains for safe doxorubicin delivery. Biotechnol. Bioeng.112 (3), 601–611 (2015).
  • Hwang C Sinskey A Lodish H . Oxidized redox state of glutathione in the endoplasmic reticulum. Science257 (5076), 1496–1502 (1992).
  • Zhao Q Wang C Liu Y et al. PEGylated mesoporous silica as a redox-responsive drug delivery system for loading thiol-containing drugs. Int. J. Pharm.477 (1–2), 613–622 (2014).
  • Zhao Q Geng H Wang Y et al. Hyaluronic acid oligosaccharide modified redox-responsive mesoporous silica nanoparticles for targeted drug delivery. ACS Appl. Mater. Interfaces6 (22), 20290–20299 (2014).
  • Xu J Singh A Amiji MM . Redox-responsive targeted gelatin nanoparticles for delivery of combination wt-p53 expressing plasmid DNA and gemcitabine in the treatment of pancreatic cancer. BMC Cancer14, 75 (2014).
  • Muhammad F Wang A Qi W Zhang S Zhu G . Intracellular antioxidants dissolve man-made antioxidant nanoparticles: using redox vulnerability of nanoceria to develop a responsive drug delivery system. ACS Appl. Mater. Interfaces6 (21), 19424–19433 (2014).
  • Naeem M Kim W Cao J Jung Y Yoo JW . Enzyme/pH dual sensitive polymeric nanoparticles for targeted drug delivery to the inflamed colon. Colloids Surf. B Biointerfaces123, 271–278 (2014).
  • Ruan S He Q Gao H . Matrix metalloproteinase triggered size-shrinkable gelatin-gold fabricated nanoparticles for tumor microenvironment sensitive penetration and diagnosis of glioma. Nanoscale7 (21), 9487–9496 (2015).
  • Moore RG Brown AK Miller MC et al. The use of multiple novel tumor biomarkers for the detection of ovarian carcinoma in patients with a pelvic mass. Gynecol Oncol.108 (2), 402–408 (2008).
  • NBTXR3 crystalline nanoparticles and radiation therapy in treating patients with soft tissue sarcoma of the extremity. clinicaltrials.gov/show/NCT01433068.
  • Pilot trial of crlx101 in treatment of patients with advanced or metastatic stomach, gastroesophageal, or esophageal cancer that cannot be removed by surgery. clinicaltrials.gov/show/NCT01612546.
  • Gao S Simon MJ Hue CD Morrison B 3rd Banta S . An unusual cell penetrating peptide identified using a plasmid display-based functional selection platform. ACS Chem. Biol.6 (5), 484–491 (2011).
  • Sajid M Ilyas M Basheer C et al. Impact of nanoparticles on human and environment: review of toxicity factors, exposures, control strategies, and future prospects. Environ. Sci. Pollut. Res. Int.22 (6), 4122–4143 (2014).
  • Liu B Campo EM Bossing T . Drosophila embryos as model to assess cellular and developmental toxicity of multi-walled carbon nanotubes (MWCNT) in living organisms. PLoS One9 (2), e88681 (2014).
  • Arami H Khandhar A Liggitt D Krishnan KM . In vivo delivery, pharmacokinetics, biodistribution and toxicity of iron oxide nanoparticles. Chem. Soc. Rev.44 (23), 8576–8607 (2015).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.