436
Views
57
CrossRef citations to date
0
Altmetric
Review

Overcoming the stability, toxicity, and biodegradation challenges of tumor stimuli-responsive inorganic nanoparticles for delivery of cancer therapeutics

, &
Pages 1095-1112 | Received 02 Jul 2019, Accepted 29 Aug 2019, Published online: 03 Sep 2019

References

  • Dawidczyk CM, Kim C, Park JH, et al. State-of-the-art in design rules for drug delivery platforms: lessons learned from FDA-approved nanomedicines. J Control Release. 2014;187:133–144.
  • 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;65:71–79.
  • Mura S, Nicolas J, Couvreur P. Stimuli-responsive nanocarriers for drug delivery. Nat Mater. 2013;12:991–1003.
  • Lyon PC, Gray MD, Mannaris C, et al. Safety and feasibility of ultrasound-triggered targeted drug delivery of doxorubicin from thermosensitive liposomes in liver tumours (TARDOX): a single-centre, open-label, phase 1 trial. Lancet Oncol. 2018;19:1027–1039.
  • Ganta S, Devalapally H, Shahiwala A, et al. A review of stimuli-responsive nanocarriers for drug and gene delivery. J Control Release. 2008;126:187–204.
  • Paris JL, Mannaris C, Cabañas MV, et al. Ultrasound-mediated cavitation-enhanced extravasation of mesoporous silica nanoparticles for controlled-release drug delivery. Chem Eng J. 2018;340:2–8.
  • Paris JL, Manzano M, Cabañas MV, et al. Mesoporous silica nanoparticles engineered for ultrasound-induced uptake by cancer cells. Nanoscale. 2018;10:6402–6408.
  • Wang S, Huang P, Chen X. Hierarchical targeting strategy for enhanced tumor tissue accumulation/retention and cellular internalization. Adv Mater. 2016;28:7340–7364.
  • Kwan JJ, Graham S, Myers R, et al. Ultrasound-induced inertial cavitation from gas-stabilizing nanoparticles. Phys Rev E. 2015;92:023019.
  • Mahmoudi M, Sant S, Wang B, et al. Superparamagnetic iron oxide nanoparticles (SPIONs): development, surface modification and applications in chemotherapy. Adv Drug Deliv Rev. 2011;63:24–46.
  • Liu J, Detrembleur C, De Pauw-Gillet M-C, et al. Gold nanorods coated with mesoporous silica shell as drug delivery system for remote near infrared light-activated release and potential phototherapy. Small. 2015;11:2323–2332.
  • Yang K, Feng L, Liu Z. Stimuli responsive drug delivery systems based on nano-graphene for cancer therapy. Adv Drug Deliv Rev. 2016;105:228–241.
  • Argyo C, Weiss V, Bräuchle C, et al. Multifunctional mesoporous silica nanoparticles as a universal platform for drug delivery. Chem Mater. 2014;26:435–451.
  • Butler KS, Durfee PN, Theron C, et al. Protocells: modular mesoporous silica nanoparticle-supported lipid bilayers for drug delivery. Small. 2016;12:2173–2185.
  • Fadeel B, Garcia-Bennett AE. Better safe than sorry: understanding the toxicological properties of inorganic nanoparticles manufactured for biomedical applications. Adv Drug Deliv Rev. 2010;62:362–374.
  • Mukherjee S, Patra CR. Therapeutic application of anti-angiogenic nanomaterials in cancers. Nanoscale. 2016;8:12444–12470.
  • Pankhurst QA, Connolly J, Jones SK, et al. Applications of magnetic nanoparticles in biomedicine. J Phys D Appl Phys. 2003;36:R167–R181.
  • Hu Y, Mignani S, Majoral J-P, et al. Construction of iron oxide nanoparticle-based hybrid platforms for tumor imaging and therapy. Chem Soc Rev. 2018;47:1874–1900.
  • Kunzmann A, Andersson B, Vogt C, et al. Efficient internalization of silica-coated iron oxide nanoparticles of different sizes by primary human macrophages and dendritic cells. Toxicol Appl Pharmacol. 2011;253:81–93.
  • Ankamwar B, Lai TC, Huang JH, et al. Biocompatibility of Fe 3 O 4 nanoparticles evaluated by in vitro cytotoxicity assays using normal, glia and breast cancer cells. Nanotechnology. 2010;21:075102.
  • Shubayev VI, Pisanic TR, Jin S. Magnetic nanoparticles for theragnostics. Adv Drug Deliv Rev. 2009;61:467–477.
  • Eaton JW, Qian M. Molecular bases of cellular iron toxicity. Free Radic Biol Med. 2002;32:833–840.
  • Huang -C-C, Liao Z-X, Lu H-M, et al. Cellular organelle-dependent cytotoxicity of iron oxide nanoparticles and its implications for cancer diagnosis and treatment: A mechanistic investigation. Chem Mater. 2016;28:9017–9025.
  • Singh N, Jenkins GJS, Asadi R, et al. Potential toxicity of superparamagnetic iron oxide nanoparticles (SPION). Nano Rev. 2010;1:5358.
  • Mahmoudi M, Laurent S, Shokrgozar MA, et al. Toxicity evaluations of superparamagnetic iron oxide nanoparticles: cell “Vision” versus physicochemical properties of nanoparticles. ACS Nano. 2011;5:7263–7276.
  • Müller K, Skepper JN, Posfai M, et al. Effect of ultrasmall superparamagnetic iron oxide nanoparticles (Ferumoxtran-10) on human monocyte-macrophages in vitro. Biomaterials. 2007;28:1629–1642.
  • Malvindi MA, De Matteis V, Galeone A, et al. Toxicity assessment of silica coated iron oxide nanoparticles and biocompatibility improvement by surface engineering. Xu B, editor. PLoS One. 2014;9:e85835.
  • Iqbal MZ, Ma X, Chen T, et al. Silica-coated super-paramagnetic iron oxide nanoparticles (SPIONPs): a new type contrast agent of T 1 magnetic resonance imaging (MRI). J Mater Chem B. 2015;3:5172–5181.
  • Hanot C, Choi Y, Anani T, et al. Effects of iron-oxide nanoparticle surface chemistry on uptake kinetics and cytotoxicity in CHO-K1 cells. Int J Mol Sci. 2015;17:54.
  • Patel D, Moon JY, Chang Y, et al. Poly(d,l-lactide-co-glycolide) coated superparamagnetic iron oxide nanoparticles: synthesis, characterization and in vivo study as MRI contrast agent. Colloids Surf A Physicochem Eng Aspects. 2008;313–314:91–94.
  • Carenza E, Jordan O, Martínez-San Segundo P, et al. Encapsulation of VEGF 165 into magnetic PLGA nanocapsules for potential local delivery and bioactivity in human brain endothelial cells. J Mater Chem B. 2015;3:2538–2544.
  • Saengruengrit C, Ritprajak P, Wanichwecharungruang S, et al. The combined magnetic field and iron oxide-PLGA composite particles: effective protein antigen delivery and immune stimulation in dendritic cells. J Colloid Interface Sci. 2018;520:101–111.
  • Zhang X, Zhang H, Liang X, et al. Iron oxide nanoparticles induce autophagosome accumulation through multiple mechanisms: lysosome impairment, mitochondrial damage, and ER stress. Mol Pharm. 2016;13:2578–2587.
  • Nosrati H, Sefidi N, Sharafi A, et al. Bovine Serum Albumin (BSA) coated iron oxide magnetic nanoparticles as biocompatible carriers for curcumin-anticancer drug. Bioorg Chem. 2018;76:501–509.
  • Jedlovszky-Hajdú A, Bombelli FB, Monopoli MP, et al. Surface coatings shape the protein corona of spions with relevance to their application in vivo. Langmuir. 2012;28:14983–14991.
  • Mahmoudi M, Shokrgozar MA, Behzadi S. Slight temperature changes affect protein affinity and cellular uptake/toxicity of nanoparticles. Nanoscale. 2013;5:3240.
  • Bao Y, Wen T, Samia ACS, et al. Magnetic nanoparticles: material engineering and emerging applications in lithography and biomedicine. J Mater Sci. 2016;51:513–553.
  • Patil U, Adireddy S, Jaiswal A, et al. In vitro/in vivo toxicity evaluation and quantification of iron oxide nanoparticles. Int J Mol Sci. 2015;16:24417–24450.
  • Yang L, Kuang H, Zhang W, et al. Size dependent biodistribution and toxicokinetics of iron oxide magnetic nanoparticles in mice. Nanoscale. 2015;7:625–636.
  • Feng Q, Liu Y, Huang J, et al. Uptake, distribution, clearance, and toxicity of iron oxide nanoparticles with different sizes and coatings. Sci Rep. 2018;8:2082.
  • Yu Q, Xiong X, Zhao L, et al. Biodistribution and toxicity assessment of superparamagnetic iron oxide nanoparticles in vitro and in vivo. Curr Med Sci. 2018;38:1096–1102.
  • Mardhian DF, Storm G, Bansal R, et al. Nano-targeted relaxin impairs fibrosis and tumor growth in pancreatic cancer and improves the efficacy of gemcitabine in vivo. J Control Release. 2018;290:1–10.
  • Kim H-J, Lee J-H, Kim S-J, et al. Roles of NADPH oxidases in cisplatin-induced reactive oxygen species generation and ototoxicity. J Neurosci. 2010;30:3933–3946.
  • Ma P, Xiao H, Yu C, et al. Enhanced cisplatin chemotherapy by iron oxide nanocarrier-mediated generation of highly toxic reactive oxygen species. Nano Lett. 2017;17:928–937.
  • Yan L, Amirshaghaghi A, Huang D, et al. Protoporphyrin IX (PpIX)-coated superparamagnetic iron oxide nanoparticle (SPION) nanoclusters for magnetic resonance imaging and photodynamic therapy. Adv Funct Mater. 2018;28:1707030.
  • Boisselier E, Astruc D. Gold nanoparticles in nanomedicine: preparations, imaging, diagnostics, therapies and toxicity. Chem Soc Rev. 2009;38:1759.
  • Umair M, Javed I, Rehman M, et al. Nanotoxicity of inert materials: the case of gold, silver and iron. J Pharm Pharm Sci. 2016;19:161.
  • Han MS, Lytton-Jean AKR, Oh B-K, et al. Colorimetric Screening of DNA-Binding Molecules with Gold Nanoparticle Probes. Angew Chemie Int Ed. 2006;45:1807–1810.
  • Medley CD, Smith JE, Tang Z, et al. Gold nanoparticle-based colorimetric assay for the direct detection of cancerous cells. Anal Chem. 2008;80:1067–1072.
  • Guisasola E, Baeza A, Asín L, et al. Heating at the nanoscale through drug-delivery devices: fabrication and synergic effects in cancer treatment with nanoparticles. Small Methods. 2018;2:1800007.
  • Kim F, Song JH, Yang P. Photochemical synthesis of gold nanorods. J Am Chem Soc. 2002;124:14316–14317.
  • Yu, Chang -S-S, C-L L, et al. Gold nanorods: electrochemical synthesis and optical properties. J Phys Chem B. 1997;101:6661–6664.
  • Gole A, Murphy CJ. Seed-mediated synthesis of gold nanorods: role of the size and nature of the seed. Chem Mater. 2004;16:3633–3640.
  • Takahashi H, Niidome Y, Niidome T, et al. Modification of gold nanorods using phosphatidylcholine to reduce cytotoxicity. Langmuir. 2006;22:2–5.
  • Hauck TS, Ghazani AA, Chan WCW. Assessing the effect of surface chemistry on gold nanorod uptake, toxicity, and gene expression in mammalian cells. Small. 2008;4:153–159.
  • Rayavarapu RG, Petersen W, Hartsuiker L, et al. In vitro toxicity studies of polymer-coated gold nanorods. Nanotechnology. 2010;21:145101.
  • Pan Y, Neuss S, Leifert A, et al. Size-dependent cytotoxicity of gold nanoparticles. Small. 2007;3:1941–1949.
  • Pan Y, Leifert A, Ruau D, et al. Gold nanoparticles of diameter 1.4 nm trigger necrosis by oxidative stress and mitochondrial damage. Small. 2009;5:2067–2076.
  • Mironava T, Hadjiargyrou M, Simon M, et al. Gold nanoparticles cellular toxicity and recovery: effect of size, concentration and exposure time. Nanotoxicology. 2010;4:120–137.
  • Albanese A, Chan WCW. Effect of gold nanoparticle aggregation on cell uptake and toxicity. ACS Nano. 2011;5:5478–5489.
  • Chithrani BD, Ghazani AA, Chan WCW. Determining the size and shape dependence of gold nanoparticle uptake into mammalian cells. Nano Lett. 2006;6:662–668.
  • Singh A, Shukla R, Hassan S, et al. Cytotoxicity and cellular internalization studies of biogenic gold nanotriangles in animal cell lines. Int J Green Nanotechnol. 2011;3:251–263.
  • Favi PM, Gao M, Johana Sepúlveda Arango L, et al. Shape and surface effects on the cytotoxicity of nanoparticles: gold nanospheres versus gold nanostars. J Biomed Mater Res Part A. 2015;103:3449–3462.
  • Schaeublin NM, Braydich-Stolle LK, Schrand AM, et al. Surface charge of gold nanoparticles mediates mechanism of toxicity. Nanoscale. 2011;3:410.
  • Bertrand N, Leroux J-C. The journey of a drug-carrier in the body: an anatomo-physiological perspective. J Control Release. 2012;161:152–163.
  • Choi K, Riviere JE, Monteiro-Riviere NA. Protein corona modulation of hepatocyte uptake and molecular mechanisms of gold nanoparticle toxicity. Nanotoxicology. 2017;11:64–75.
  • Johnston HJ, Hutchison G, Christensen FM, et al. A review of the in vivo and in vitro toxicity of silver and gold particulates: particle attributes and biological mechanisms responsible for the observed toxicity. Crit Rev Toxicol. 2010;40:328–346.
  • Simpson CA, Salleng KJ, Cliffel DE, et al. In vivo toxicity, biodistribution, and clearance of glutathione-coated gold nanoparticles. Nanomedicine Nanotechnology, Biol Med. 2013;9:257–263.
  • Thakor AS, Luong R, Paulmurugan R, et al. The fate and toxicity of raman-active silica-gold nanoparticles in mice. Sci Transl Med. 2011;3: 79ra33–79ra33.
  • Lasagna-Reeves C, Gonzalez-Romero D, Barria MA, et al. Bioaccumulation and toxicity of gold nanoparticles after repeated administration in mice. Biochem Biophys Res Commun. 2010;393:649–655.
  • Villaverde G, Gómez-Graña S, Guisasola E, et al. Targeted chemo-photothermal therapy: a nanomedicine approximation to selective melanoma treatment. Part Part Syst Charact. 2018;35:1800148.
  • Kreyling WG, Abdelmonem AM, Ali Z, et al. In vivo integrity of polymer-coated gold nanoparticles. Nat Nanotechnol. 2015;10:619–623.
  • Rengan AK, Bukhari AB, Pradhan A, et al. In vivo analysis of biodegradable liposome gold nanoparticles as efficient agents for photothermal therapy of cancer. Nano Lett. 2015;15:842–848.
  • Bunggulawa EJ, Wang W, Yin T, et al. Recent advancements in the use of exosomes as drug delivery systems. J Nanobiotechnology. 2018;16:81.
  • Betzer O, Perets N, Angel A, et al. In vivo neuroimaging of exosomes using gold nanoparticles. ACS Nano. 2017;11:10883–10893.
  • Cheng X, Sun R, Yin L, et al. Light-triggered assembly of gold nanoparticles for photothermal therapy and photoacoustic imaging of tumors in vivo. Adv Mater. 2017;29:1604894.
  • Shen H, Zhang L, Liu M, et al. Biomedical applications of graphene. Theranostics. 2012;2:283–294.
  • Seabra AB, Paula AJ, de Lima R, et al. Nanotoxicity of graphene and graphene oxide. Chem Res Toxicol. 2014;27:159–168.
  • Ou L, Song B, Liang H, et al. Toxicity of graphene-family nanoparticles: a general review of the origins and mechanisms. Part Fibre Toxicol. 2016;13:57.
  • Jastrzębska AM, Kurtycz P, Olszyna AR. Recent advances in graphene family materials toxicity investigations. J Nanopart Res. 2012;14:1320.
  • Yang K, Li Y, Tan X, et al. Behavior and toxicity of graphene and its functionalized derivatives in biological systems. Small. 2013;9:1492–1503.
  • Gurunathan S, Han JW, Eppakayala V, et al. Green synthesis of graphene and its cytotoxic effects in human breast cancercells. Int J Nanomedicine. 2013;8:1015.
  • Vallabani NVS, Mittal S, Shukla R, et al. Toxicity of graphene in normal human lung cells (BEAS-2B). J Biomed Nanotechnol. 2011;7:106–107.
  • Zhang L, Xia J, Zhao Q, et al. Functional graphene oxide as a nanocarrier for controlled loading and targeted delivery of mixed anticancer drugs. Small. 2010;6:537–544.
  • Zhang Y, Ali SF, Dervishi E, et al. Cytotoxicity effects of graphene and single-wall carbon nanotubes in neural phaeochromocytoma-derived PC12 cells. ACS Nano. 2010;4:3181–3186.
  • Chang Y, Yang S-T, Liu J-H, et al. In vitro toxicity evaluation of graphene oxide on A549 cells. Toxicol Lett. 2011;200:201–210.
  • Qiao Y, An J, Ma L. Single cell array based assay for in vitro genotoxicity study of nanomaterials. Anal Chem. 2013;85:4107–4112.
  • Robinson JT, Tabakman SM, Liang Y, et al. Ultrasmall reduced graphene oxide with high near-infrared absorbance for photothermal therapy. J Am Chem Soc. 2011;133:6825–6831.
  • Feng L, Liu Z. Graphene in biomedicine: opportunities and challenges. Nanomedicine. 2011;6:317–324.
  • Liu Z, Robinson JT, Sun X, et al. PEGylated nanographene oxide for delivery of water-insoluble cancer drugs. J Am Chem Soc. 2008;130:10876–10877.
  • Sun X, Liu Z, Welsher K, et al. Nano-graphene oxide for cellular imaging and drug delivery. Nano Res. 2008;1:203–212.
  • Sasidharan A, Panchakarla LS, Sadanandan AR, et al. Hemocompatibility and macrophage response of pristine and functionalized graphene. Small. 2012;8:1251–1263.
  • Singh SK, Singh MK, Kulkarni PP, et al. Amine-modified graphene: thrombo-protective safer alternative to graphene oxide for biomedical applications. ACS Nano. 2012;6:2731–2740.
  • Zhang X, Yin J, Peng C, et al. Distribution and biocompatibility studies of graphene oxide in mice after intravenous administration. Carbon N Y. 2011;49:986–995.
  • Liao K, Lin Y, Macosko CW, et al. Cytotoxicity of graphene oxide and graphene in human erythrocytes and skin fibroblasts. ACS Appl Mater Interfaces. 2011;3:2607–2615.
  • Wen K-P, Chen Y-C, Chuang C-H, et al. Accumulation and toxicity of intravenously-injected functionalized graphene oxide in mice. J Appl Toxicol. 2015;35:1211–1218.
  • Schinwald A, Murphy FA, Jones A, et al. Graphene-based nanoplatelets: a new risk to the respiratory system as a consequence of their unusual aerodynamic properties. ACS Nano. 2012;6:736–746.
  • Zhang S, Yang K, Feng L, et al. In vitro and in vivo behaviors of dextran functionalized graphene. Carbon N Y. 2011;49:4040–4049.
  • Wang K, Ruan J, Song H, et al. Biocompatibility of Graphene Oxide. Nanoscale Res Lett. 2011;6:8.
  • Gollavelli G, Ling Y-C. Multi-functional graphene as an in vitro and in vivo imaging probe. Biomaterials. 2012;33:2532–2545.
  • Zanni E, De Bellis G, Bracciale MP, et al. Graphite nanoplatelets and Caenorhabditis elegans : insights from an in vivo model. Nano Lett. 2012;12:2740–2744.
  • Zhang W, Wang C, Li Z, et al. Unraveling stress-induced toxicity properties of graphene oxide and the underlying mechanism. Adv Mater. 2012;24:5391–5397.
  • Syama S, Mohanan PV. Safety and biocompatibility of graphene: A new generation nanomaterial for biomedical application. Int J Biol Macromol. 2016;86:546–555.
  • Zhu C, Guo S, Fang Y, et al. Reducing sugar: new functional molecules for the green synthesis of graphene nanosheets. ACS Nano. 2010;4:2429–2437.
  • Gao J, Liu F, Liu Y, et al. Environment-friendly method to produce graphene that employs vitamin C and amino acid. Chem Mater. 2010;22:2213–2218.
  • Urbas K, Aleksandrzak M, Jedrzejczak M, et al. Chemical and magnetic functionalization of graphene oxide as a route to enhance its biocompatibility. Nanoscale Res Lett. 2014;9:656.
  • Chong Y, Ma Y, Shen H, et al. The in vitro and in vivo toxicity of graphene quantum dots. Biomaterials. 2014;35:5041–5048.
  • Jasim DA, Boutin H, Fairclough M, et al. Thickness of functionalized graphene oxide sheets plays critical role in tissue accumulation and urinary excretion: A pilot PET/CT study. Appl Mater Today. 2016;4:24–30.
  • Yang K, Zhang S, Zhang G, et al. Graphene in mice: ultrahigh in vivo tumor uptake and efficient photothermal therapy. Nano Lett. 2010;10:3318–3323.
  • Miao W, Shim G, Lee S, et al. Safety and tumor tissue accumulation of pegylated graphene oxide nanosheets for co-delivery of anticancer drug and photosensitizer. Biomaterials. 2013;34:3402–3410.
  • Yang K, Gong H, Shi X, et al. In vivo biodistribution and toxicology of functionalized nano-graphene oxide in mice after oral and intraperitoneal administration. Biomaterials. 2013;34:2787–2795.
  • Yan L, Wang Y, Xu X, et al. Can graphene oxide cause damage to eyesight?. Chem Res Toxicol. 2012;25:1265–1270.
  • Kiew SF, Kiew LV, Lee HB, et al. Assessing biocompatibility of graphene oxide-based nanocarriers: A review. J Control Release. 2016;226:217–228.
  • Kim Y-K, Kim M-H, Min D-H. Biocompatible reduced graphene oxide prepared by using dextran as a multifunctional reducing agent. Chem Commun. 2011;47:3195–3197.
  • Wate PS, Banerjee SS, Jalota-Badhwar A, et al. Cellular imaging using biocompatible dendrimer-functionalized graphene oxide-based fluorescent probe anchored with magnetic nanoparticles. Nanotechnology. 2012;23:415101.
  • Hong BJ, Compton OC, An Z, et al. successful stabilization of graphene oxide in electrolyte solutions: enhancement of biofunctionalization and cellular uptake. ACS Nano. 2012;6:63–73.
  • Cheng C, Nie S, Li S, et al. Biopolymer functionalized reduced graphene oxide with enhanced biocompatibility via mussel inspired coatings/anchors. J Mater Chem B. 2013;1:265–275.
  • Xu M, Zhu J, Wang F, et al. Improved in vitro and in vivo biocompatibility of graphene oxide through surface modification: poly(Acrylic Acid)-functionalization is superior to PEGylation. ACS Nano. 2016;10:3267–3281.
  • Croissant JG, Fatieiev Y, Khashab NM. Degradability and clearance of silicon, organosilica, silsesquioxane, silica mixed oxide, and mesoporous silica nanoparticles. Adv Mater. 2017;29:1604634.
  • Santos HA, Mäkilä E, Airaksinen AJ, et al. Porous silicon nanoparticles for nanomedicine: preparation and biomedical applications. Nanomedicine. 2014;9:535–554.
  • Xu W, Rytkönen J, Rönkkö S, et al. A nanostopper approach to selectively engineer the surfaces of mesoporous silicon. Chem Mater. 2014;26:6734–6742.
  • Kafshgari MH, Delalat B, Tong WY, et al. Oligonucleotide delivery by chitosan-functionalized porous silicon nanoparticles. Nano Res. 2015;8:2033–2046.
  • Godin B, Gu J, Serda RE, et al. Tailoring the degradation kinetics of mesoporous silicon structures through PEGylation. J Biomed Mater Res A. 2010;94:1236–1243.
  • Hou H, Nieto A, Ma F, et al. Tunable sustained intravitreal drug delivery system for daunorubicin using oxidized porous silicon. J Control Release. 2014;178:46–54.
  • Martinez JO, Chiappini C, Ziemys A, et al. Engineering multi-stage nanovectors for controlled degradation and tunable release kinetics. Biomaterials. 2013;34:8469–8477.
  • Hon NK, Shaposhnik Z, Diebold ED, et al. Tailoring the biodegradability of porous silicon nanoparticles. J Biomed Mater Res Part A. 2012;100A:3416–3421.
  • Park J-H, Gu L, von Maltzahn G, et al. Biodegradable luminescent porous silicon nanoparticles for in vivo applications. Nat Mater. 2009;8:331–336.
  • Martinez JO, Evangelopoulos M, Chiappini C, et al. Degradation and biocompatibility of multistage nanovectors in physiological systems. J Biomed Mater Res Part A. 2014;102:3540–3549.
  • Tolstik E, Osminkina LA, Matthäus C, et al. Studies of silicon nanoparticles uptake and biodegradation in cancer cells by Raman spectroscopy. Nanomedicine Nanotechnology, Biol Med. 2016;12:1931–1940.
  • Tzur-Balter A, Shatsberg Z, Beckerman M, et al. Mechanism of erosion of nanostructured porous silicon drug carriers in neoplastic tissues. Nat Commun. 2015;6:6208.
  • Wang C-F, Mäkilä EM, Kaasalainen MH, et al. Copper-free azide–alkyne cycloaddition of targeting peptides to porous silicon nanoparticles for intracellular drug uptake. Biomaterials. 2014;35:1257–1266.
  • Shahbazi MA, Hamidi M, Mäkilä EM, et al. The mechanisms of surface chemistry effects of mesoporous silicon nanoparticles on immunotoxicity and biocompatibility. Biomaterials. 2013;34:7776–7789.
  • Ivanov S, Zhuravsky S, Yukina G, et al. In vivo toxicity of intravenously administered silica and silicon nanoparticles. Materials. 2012;5:1873–1889.
  • Sarparanta M, Bimbo LM, Rytkoänen J, et al. Intravenous delivery of hydrophobin-functionalized porous silicon nanoparticles: stability, plasma protein adsorption and biodistribution. Mol Pharm. 2012;9:654–663.
  • Sarparanta MP, Bimbo LM, Mäkilä EM, et al. The mucoadhesive and gastroretentive properties of hydrophobin-coated porous silicon nanoparticle oral drug delivery systems. Biomaterials. 2012;33:3353–3362.
  • Shahbazi M, Almeida PV, Mäkilä E, et al. Functionalized porous silicon nanoparticles for enhanced stability and cellular internalization. Macromol Rapid Commun. 2014;35:624–629.
  • Xia B, Zhang W, Shi J, et al. Engineered stealth porous silicon nanoparticles via surface encapsulation of bovine serum albumin for prolonging blood circulation in vivo. ACS Appl Mater Interfaces. 2013;5:11718–11724.
  • Paris JL, Colilla M, Izquierdo-Barba I, et al. Tuning mesoporous silica dissolution in physiological environments: a review. J Mater Sci. 2017;52:8761–8771.
  • Vallet-Regí M, Rámila A, Del Real RP, et al. A new property of MCM-41: drug delivery system. Chem Mater. 2001;13:308–311.
  • Vallet-Regí M, Ruiz-Hernández E. Bioceramics: from bone regeneration to cancer nanomedicine. Adv Mater. 2011;23:5177–5218.
  • Villaverde G, Alfranca A, Gonzalez‐Murillo Á, et al. Molecular scaffolds as double‐targeting agents for the diagnosis and treatment of neuroblastoma. Angew Chemie. 2019;131:3099–3104.
  • Mora-Raimundo P, Lozano D, Manzano M, et al. Nanoparticles to Knockdown osteoporosis-related gene and promote osteogenic marker expression for osteoporosis treatment. ACS Nano. 2019;13:5451–5464.
  • Baeza A, Colilla M, Vallet-Regí M. Advances in mesoporous silica nanoparticles for targeted stimuli-responsive drug delivery. Expert Opin Drug Deliv. 2015;12:319–337.
  • Baeza A, Ruiz-Molina D, Vallet-Regí M. Recent advances in porous nanoparticles for drug delivery in antitumoral applications: inorganic nanoparticles and nanoscale metal-organic frameworks. Expert Opin Drug Deliv. 2017;14:783–796.
  • Martínez-Carmona M, Lozano D, Baeza A, et al. A novel visible light responsive nanosystem for cancer treatment. Nanoscale. 2017;9:15967–15973.
  • Martínez-Carmona M, Baeza A, Rodriguez-Milla MA, et al. Mesoporous silica nanoparticles grafted with a light-responsive protein shell for highly cytotoxic antitumoral therapy. J Mater Chem B. 2015;3:5746–5752.
  • Guisasola E, Asín L, Beola L, et al. Beyond traditional hyperthermia: in vivo cancer treatment with magnetic-responsive mesoporous silica nanocarriers. ACS Appl Mater Interfaces. 2018;10:12518–12525.
  • Baeza A, Guisasola E, Ruiz-Hernández E, et al. Magnetically triggered multidrug release by hybrid mesoporous silica nanoparticles. Chem Mater. 2012;24:517–524.
  • Guisasola E, Baeza A, Talelli M, et al. Magnetic-responsive release controlled by hot spot effect. Langmuir. 2015;31:12777–12782.
  • Paris JL, Cabañas MV, Manzano M, et al. Polymer-grafted mesoporous silica nanoparticles as ultrasound-responsive drug carriers. ACS Nano. 2015;9:11023–11033.
  • Martínez-Carmona M, Lozano D, Colilla M, et al. Selective topotecan delivery to cancer cells by targeted pH-sensitive mesoporous silica nanoparticles. RSC Adv. 2016;6:50923–50932.
  • Hu C, Huang P, Zheng Z, et al. A facile strategy to prepare an enzyme-responsive mussel mimetic coating for drug delivery based on mesoporous silica nanoparticles. Langmuir. 2017;33:5511–5518.
  • Wu M, Meng Q, Chen Y, et al. Large pore-sized hollow mesoporous organosilica for redox-responsive gene delivery and synergistic cancer chemotherapy. Adv Mater. 2016;28:1963–1969.
  • Lin YS, Abadeer N, Haynes CL. Stability of small mesoporous silica nanoparticles in biological media. Chem Commun. 2011;47:532–534.
  • Murugadoss S, Lison D, Godderis L, et al. Toxicology of silica nanoparticles: an update. Arch Toxicol. 2017;91:2967–3010.
  • Kim I-Y, Joachim E, Choi H, et al. Toxicity of silica nanoparticles depends on size, dose, and cell type. Nanomedicine Nanotechnology, Biol Med. 2015;11:1407–1416.
  • Kim D, Lin YS, Haynes CL. On-chip evaluation of shear stress effect on cytotoxicity of mesoporous silica nanoparticles. Anal Chem. 2011;83:8377–8382.
  • Heidegger S, Gößl D, Schmidt A, et al. Immune response to functionalized mesoporous silica nanoparticles for targeted drug delivery. Nanoscale. 2016;8:938–948.
  • Lin Y-S, Haynes CL. Impacts of mesoporous silica nanoparticle size, pore ordering, and pore integrity on hemolytic activity. J Am Chem Soc. 2010;132:4834–4842.
  • Slowing II, Wu CW, Vivero-Escoto JL, et al. Mesoporous silica nanoparticles for reducing hemolytic activity towards mammalian red blood cells. Small. 2009;5:57–62.
  • Urata C, Yamada H, Wakabayashi R, et al. Aqueous colloidal mesoporous nanoparticles with ethenylene-bridged silsesquioxane frameworks. J Am Chem Soc. 2011;133:8102–8105.
  • Liu T, Li L, Teng X, et al. Single and repeated dose toxicity of mesoporous hollow silica nanoparticles in intravenously exposed mice. Biomaterials. 2011;32:1657–1668.
  • Fu C, Liu T, Li L, et al. The absorption, distribution, excretion and toxicity of mesoporous silica nanoparticles in mice following different exposure routes. Biomaterials. 2013;34:2565–2575.
  • Hudson SP, Padera RF, Langer R, et al. The biocompatibility of mesoporous silicates. Biomaterials. 2008;29:4045–4055.
  • Villaverde G, Baeza A, Melen GJ, et al. A new targeting agent for the selective drug delivery of nanocarriers for treating neuroblastoma. J Mater Chem B. 2015;3:4831–4842.
  • Dogra P, Adolphi NL, Wang Z, et al. Establishing the effects of mesoporous silica nanoparticle properties on in vivo disposition using imaging-based pharmacokinetics. Nat Commun. 2018;9:4551.
  • Yu T, Greish K, McGill LD, et al. Influence of geometry, porosity, and surface characteristics of silica nanoparticles on acute toxicity: their vasculature effect and tolerance threshold. ACS Nano. 2012;6:2289–2301.
  • Lee S, Kim MS, Lee D, et al. The comparative immunotoxicity of mesoporous silica nanoparticles and colloidal silica nanoparticles in mice. Int J Nanomedicine. 2013;8:147–158.
  • Li L, Tang F, Liu H, et al. In vivo delivery of silica nanorattle encapsulated docetaxel for liver cancer therapy with low toxicity and high efficacy. ACS Nano. 2011;5:679.
  • He Q, Shi J, Zhu M, et al. The three-stage in vitro degradation behavior of mesoporous silica in simulated body fluid. Microporous Mesoporous Mater. 2010;131:314–320.
  • Braun K, Pochert A, Beck M, et al. Dissolution kinetics of mesoporous silica nanoparticles in different simulated body fluids. J Sol-Gel Sci Technol. 2016;79:319–327.
  • Yamada H, Urata C, Aoyama Y, et al. Preparation of colloidal mesoporous silica nanoparticles with different diameters and their unique degradation behavior in static aqueous systems. Chem Mater. 2012;24:1462–1471.
  • Hao N, Liu H, Li L, et al. In vitro degradation behavior of silica nanoparticles under physiological conditions. J Nanosci Nanotechnol. 2012;12:6346–6354.
  • Huang X, Young NP, Townley HE. Characterization and comparison of mesoporous silica particles for optimized drug delivery. Nanomater Nanotechnol. 2014;4:2.
  • Cauda V, Schlossbauer A, Bein T. Bio-degradation study of colloidal mesoporous silica nanoparticles: effect of surface functionalization with organo-silanes and poly(ethylene glycol). Microporous Mesoporous Mater. 2010;132:60–71.
  • Li X, Zhang L, Dong X, et al. Preparation of mesoporous calcium doped silica spheres with narrow size dispersion and their drug loading and degradation behavior. Microporous Mesoporous Mater. 2007;102:151–158.
  • Yu L, Chen Y, Wu M, et al. “Manganese extraction” strategy enables tumor-sensitive biodegradability and theranostics of nanoparticles. J Am Chem Soc. 2016;138:9881–9894.
  • Fontecave T, Sanchez C, Azaïs T, et al. Chemical modification as a versatile tool for tuning stability of silica based mesoporous carriers in biologically relevant conditions. Chem Mater. 2012;24:4326–4336.
  • Maggini L, Cabrera I, Ruiz-Carretero A, et al. Breakable mesoporous silica nanoparticles for targeted drug delivery. Nanoscale. 2016;8:7240–7247.
  • Chen Y, Meng Q, Wu M, et al. Hollow mesoporous organosilica nanoparticles: a generic intelligent framework-hybridization approach for biomedicine. J Am Chem Soc. 2014;136:16326–16334.
  • Cauda V, Argyo C, Bein T. Impact of different PEGylation patterns on the long-term bio-stability of colloidal mesoporous silica nanoparticles. J Mater Chem. 2010;20:8693–8699.
  • Shen D, Yang J, Li X, et al. Biphase stratification approach to three-dimensional dendritic biodegradable mesoporous silica nanospheres. Nano Lett. 2014;14:923–932.
  • Möller K, Bein T. Degradable drug carriers: vanishing mesoporous silica nanoparticles. Chem Mater. 2019;31:4364–4378.
  • Zhang Q, Wang X, Li PZ, et al. Biocompatible, uniform, and redispersible mesoporous silica nanoparticles for cancer-targeted drug delivery in vivo. Adv Funct Mater. 2014;24:2450–2461.
  • He Q, Zhang Z, Gao F, et al. In vivo biodistribution and urinary excretion of mesoporous silica nanoparticles: effects of particle size and PEGylation. Small. 2011;7:271–280.
  • Huang X, Li L, Liu T, et al. The shape effect of mesoporous silica nanoparticles on biodistribution, clearance, and biocompatibility in vivo. ACS Nano. 2011;5:5390–5399.
  • Li L, Liu T, Fu C, et al. Biodistribution, excretion, and toxicity of mesoporous silica nanoparticles after oral administration depend on their shape. Nanomedicine Nanotechnology, Biol Med. 2015;11:1915–1924.
  • Tarn D, Ashley CE, Xue M, et al. Mesoporous silica nanoparticle nanocarriers: biofunctionality and biocompatibility. Acc Chem Res. 2013;46:792–801.
  • Desai D, Zhang J, Sandholm J, et al. Lipid bilayer-gated mesoporous silica nanocarriers for tumor-targeted delivery of zoledronic acid in vivo. Mol Pharm. 2017;14:3218–3227.
  • Gonzalez Porras MA, Durfee PN, Gregory AM, et al. A novel approach for targeted delivery to motoneurons using cholera toxin-B modified protocells. J Neurosci Methods. 2016;273:160–174.
  • Ashley CE, Carnes EC, Epler KE, et al. Delivery of small interfering RNA by peptide-targeted mesoporous silica nanoparticle-supported lipid bilayers. ACS Nano. 2012;6:2174–2188.
  • Epler K, Padilla D, Phillips G, et al. Delivery of ricin toxin A-chain by peptide-targeted mesoporous silica nanoparticle-supported lipid bilayers. Adv Healthc Mater. 2012;1:348–353.
  • Sharifabad ME, Mercer T, Sen T. Drug-loaded liposome-capped mesoporous core–shell magnetic nanoparticles for cellular toxicity study. Nanomedicine. 2016;11:2753–2755.
  • Han N, Zhao Q, Wan L, et al. Hybrid lipid-capped mesoporous silica for stimuli-responsive drug release and overcoming multidrug resistance. ACS Appl Mater Interfaces. 2015;7:3342–3351.
  • Villegas MR, Baeza A, Noureddine A, et al. Multifunctional protocells for enhanced penetration in 3D extracellular tumoral matrices. Chem Mater. 2018;30:112–120.
  • Villegas MR, Baeza A, Vallet-Regí M. Hybrid collagenase nanocapsules for enhanced nanocarrier penetration in tumoral tissues. ACS Appl Mater Interfaces. 2015;7:24075–24081.
  • Akin D, Sturgis J, Ragheb K, et al. Bacteria-mediated delivery of nanoparticles and cargo into cells. Nat Nanotechnol. 2007;2:441–449.
  • Bonvalot S, Le Pechoux C, De Baere T, et al. First-in-human study testing a new radioenhancer using nanoparticles (NBTXR3) activated by radiation therapy in patients with locally advanced soft tissue sarcomas. Clin Cancer Res. 2017;23:908–917.

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.