Please wait a minute...
Frontiers of Materials Science

ISSN 2095-025X

ISSN 2095-0268(Online)

CN 11-5985/TB

Postal Subscription Code 80-974

2018 Impact Factor: 1.701

Front. Mater. Sci.    2022, Vol. 16 Issue (3) : 220604    https://doi.org/10.1007/s11706-022-0604-x
REVIEW ARTICLE
Strategies to assemble therapeutic and imaging molecules into inorganic nanocarriers
Sheikh Tanzina HAQUE1, Mark M. BANASZAK HOLL2, Ezharul Hoque CHOWDHURY1,3()
1. Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
2. Department of Chemical and Biological Engineering, Monash University, Clayton, Victoria 3800, Australia
3. Health and Wellbeing Cluster, Global Asia in the 21st Century (GA21) Platform, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
 Download: PDF(3796 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Inorganic nanocarriers are potent candidates for delivering conventional anticancer drugs, nucleic acid-based therapeutics, and imaging agents, influencing their blood half-lives, tumor targetability, and bioactivity. In addition to the high surface area-to-volume ratio, they exhibit excellent scalability in synthesis, controllable shape and size, facile surface modification, inertness, stability, and unique optical and magnetic properties. However, only a limited number of inorganic nanocarriers have been so far approved for clinical applications due to burst drug release, poor target specificity, and toxicity. To overcome these barriers, understanding the principles involved in loading therapeutic and imaging molecules into these nanoparticles (NPs) and the strategies employed in enhancing sustainability and targetability of the resultant complexes and ensuring the release of the payloads in extracellular and intracellular compartments of the target site is of paramount importance. Therefore, we will shed light on various loading mechanisms harnessed for different inorganic NPs, particularly involving physical entrapment into porous/hollow nanostructures, ionic interactions with native and surface-modified NPs, covalent bonding to surface-functionalized nanomaterials, hydrophobic binding, affinity-based interactions, and intercalation through co-precipitation or anion exchange reaction.

Keywords inorganic nanoparticle      cancer      ionic interaction      covalent bonding      affinity interaction      intercalation     
Corresponding Author(s): Ezharul Hoque CHOWDHURY   
Issue Date: 22 September 2022
 Cite this article:   
Sheikh Tanzina HAQUE,Mark M. BANASZAK HOLL,Ezharul Hoque CHOWDHURY. Strategies to assemble therapeutic and imaging molecules into inorganic nanocarriers[J]. Front. Mater. Sci., 2022, 16(3): 220604.
 URL:  
https://academic.hep.com.cn/foms/EN/10.1007/s11706-022-0604-x
https://academic.hep.com.cn/foms/EN/Y2022/V16/I3/220604
Fig.1  (a) Schematic illustration of different strategies to assemble therapeutics and imaging molecules into inorganic NPs. (b) Different surface modification approaches between inorganic NPs and drugs. (c) Change in size, morphologies, and surface properties of the inorganic nanocarriers due to their interaction (via different approaches) with drugs, highlighting their possible effect on pharmacokinetics. Note: Ag, silver; Au, gold; CA, carbonate apatite; CaCO3, calcium carbonate; CaP, calcium phosphate; CNT, carbon nanotube; CPNP, calcium phosphosilicate nanoparticle; HA, hydroxyapatite; HMnO, hollow manganese oxide; LDH, layered double hydroxide; MDR, multidrug resistance; MPS, mononuclear phagocytic system; MSN, mesoporous silica nanoparticle; QD, quantum dot; RES, reticuloendothelial system; SPION, superparamagnetic iron oxide nanoparticle; SWCNT, single-wall carbon nanotube; ZnS, zinc sulfide.
Fig.2  Schematic illustration: (a) MSN; (b) Drug-loaded MSN; (c) Gatekeepers are employed to cap the pores on the MSN in order to prevent drug release prematurely; (d) Drug-loaded MSNs which has been surface-functionalized to decrease off-target interactions and boost site-specific characteristics; (e) Ligand-conjugated MSNs which can facilitate effective cellular internalization via endocytosis for the delivery of drugs at the targeted site; (f) MSNs enter cancer cells via endocytosis and release the drugs when triggered by external or local stimuli.
Fig.3  Schematic representation of PEI-conjugated MSNs loaded with cargos and modified with and without ligands for in vitro and in vivo analyses.
Fig.4  (a) Avidin/biotin and streptavidin/biotin complex formation. For the generation of the resulting avidin/biotin and streptavidin/biotin complex, both avidin and streptavidin can bind with four conjugated biotin (biotin generally conjugated to antibody, protein, peptide, and enzyme) molecules. (b) Interaction between avidin/biotin and streptavidin/biotin. One avidin/biotin complex or streptavidin/biotin complex can be linked with another avidin/biotin complex or streptavidin/biotin complex via binding with the conjugated biotin.
Fig.5  Schematic illustration of a LDH structure with the general formula [MII1?xMIIIx(OH)2]x+(An?)x/n·mH2O (x = 0.2–0.4; n = 0.5–1), used for therapeutic purposes. MII (M2+) represents a divalent metal cation and MIII (M3+) a trivalent metal cation. Here ‘x’ is a ratio of M3+/(M2++M3+). The M2+/M3+ ratios of 2–4 are considered relatively steady. An? (in the interlamellar region) is an anion. An? represents any charge compensating organic or inorganic anions, such as oxo-anions (carbonates, nitrates, etc.), halides, oxo- and polyoxo-metallates (dichromates, (Mo7O24)6?, (V10O28)6?, etc.). ‘m’ represents the number of moles of solvent contained in the interlamellar zone where no anions are present. The LDH can be modified with inorganic ions, fluorescence (FITC), dye, polymer coating, and ligand targeting moieties.
Fig.6  Schematic illustration of the formation and cleavage of covalent bonds between NPs and drugs. The covalent bonds presented are mostly based on the functional groups (e.g., disulfide, ester, thioester, orthoester, thiol, carbonate, amide, hydrazone, maleimide-thiol, urothane, and unhydride linkage) available on nanocarriers and drugs being conjugated.
Fig.7  Incorporation of drugs or nucleic acid conjugated NPs in liposomes. The cationic liposomes can entrap inorganic NPs loaded therapeutics inside the hydrophilic core or embed them in the lipophilic bilayer outside.
Nanocarrier Drug Results with drug loaded nanocarriers vs. free drugs Refs.
PEI/PEG/MSNs Trastuzumab In trastuzumab-resistant HCC1954 xenografts, single dosage of siHER2–NPs significantly decreased (60%) HER2 protein levels. Treatment with multiple intravenous doses over three weeks resulted in a marked reduction in tumor growth. Trastuzumab (10 mg·kg?1) administered intraperitonially, trastuzumab (5 mg·kg?1) injected via tail vein, and trastuzumab (5 mg·kg?1) and paclitaxel (3.1 mg·kg?1) injected via tail vein showed a reduced efficacy in suppressing tumor progression in HCC1954 tumor-bearing mice. [54]
C60@Au-PEG DOX 1. In contrast to the DOX group, C60@Au-PEG/DOX (amide bond), and C60@Au-PEG/DOX (hydrazone bond) exhibited significant levels of DOX in the blood after injection, suggesting that C60@Au-PEG/DOX (amide bond) and C60@Au-PEG/DOX (hydrazone bond) prolonged DOX circulation time in the blood in S180 tumor-induced female BALB/c mice.2. In comparison to DOX, C60@Au-PEG/DOX (amide bond) and C60@Au-PEG/DOX (hydrazone bond) markedly lowered the accumulation of DOX in the heart and spleen, decreasing the adverse effects of DOX on heart and spleen.3. The relative tumor volume (V/V0) of DOX was 2.7±0.26 compared to 1.9±0.21 for C60@Au-PEG/DOX, indicating that more DOX could potentially enter the tumor site with C60@Au-PEG. [69]
PEI-PEG-MSNs DOX Dox and P-gp siRNA loaded PEI-PEG-MSNs complex in MCF-7/MDR tumor-bearing xenograft model demonstrated greater accumulation in the tumor as a result of prolonged blood circulation, leading to 80% of tumor growth suppression in comparison to free DOX (17%), MSN-DOX (62%) and P-gp siRNA (0%). [60]
CPNPs Cer10 MTS cytotoxicity assay demonstrating dosage responsive cytotoxic actions of Cer10-CPNPs compared to control CPNPs which exhibit modest cytotoxicity at the highest particle number concentration and Cer10 in UACC 903 melanoma cells. [67]
MSN@Gelatin DOX For DOX/MSN@Gelatin, a dose-dependent cytotoxicity was recorded (IC50 = (17.27±0.63) μg·mL?1) in Hep-G2 cells, which was higher than that of free DOX. On the contrary, minimal toxicity (IC50 > 100 μg·mL ?1) was observed for MSN@Gelatin. [58]
PEG-SWCNTs PTX In vitro PTX-PEG-SWCNTs showed higher efficacy in suppressing tumor growth compared to PTX alone in a murine 4T1 breast cancer model, owing to prolonged blood circulation and 10-fold higher tumor PTX uptake by SWNT delivery through enhanced permeability and retention (EPR). [123]
MSNs-capped MMP9 Cisplatin, Bz 1. Cisplatin and Bz-loaded MSNs capped with MMP9 were found to cause significant cell apoptosis in human tumor cells and mouse and human lung tumors. The lowest cisplatin dose (2 μmol·L?1) had the most enhanced cytotoxicity, increasing by over 35% in the presence of Bz. It represents an increase in cytotoxic potency of 5- to 10-fold for nontoxic doses of a single drug. These findings demonstrated that cisplatin and Bz delivered together via NPs were additively cytotoxic, enabling a reduction in drug dosage.2. Kras mutant mouse 3D-LTC treated with free (nonencapsulated) drug(s) induced apoptosis with no distinction between tumorous and nontumorous tissues. [87]
PEG-CA and biotinylated PEG-fibronectin-CA Gemcitabine, anastrozole 1. HPLC revealed that cellular uptake of gemcitabine and anastrozole was higher in breast cancer cells for the surface-modified NPs than CA and free drugs.2. In the cytotoxicity study, surface modified NPs showed greater toxicity than unmodified CA NPs and free gemcitabine and anastrozole.3. Tumor regression study using surface-modified gemcitabine-encapsulated NPs revealed dramatic shrinkage of tumors compared to free gemcitabine. [90]
LDH MTX The antitumor effects of intact MTX (30 mg·kg?1) and MTX-LDH (75 mg·kg?1 was equivalent to 30 mg·kg?1 MTX) using HOS-bearing xenograft mice models demonstrated that MTX-LDH treated tumors were considerably smaller than intact MTX. [105]
LDH 5-Fu The 5-fluorouracil-LDH (5-Fu–LDH) revealed sustained release, prolonged half-life, and increased distribution of 5-Fu in tumors in comparison to free 5-Fu. [96]
LDH Mercaptoundecahydro-closo-dodecaborate (BSH) anionic molecules In biodistribution experiments with xenografted mice, the tumor-to-blood ratio of BSH in the BSH-LDH (boron delivery system) treated group was shown to be 4.4 times greater than in the intact BSH treated group 2 h after drug administration. [113]
Ferrous ions doped MgAl-LDH (Fe-LDH) DOX Tumor growth in 4T1 bearing mice was suppressed following treatment of PTT and chemotherapy with Fe-LDH/DOX compared to free DOX. [114]
Pt-PEG-GNRs Cisplatin In comparison to free cisplatin, cytotoxicity analyses (MTT assay) revealed increased toxicity for cervical cancer HeLa cells, human lung carcinoma A549 cells and human breast adenocarcinoma MCF-7 cells, with IC50 values around 9 to 65 fold lower. [135]
TCL-SPION DOX In vivo studies showed more significant tumor growth suppression with DOX@TCL-SPIONs than mice treated with 5% glucose, TCL-SPION, DOX (0.64 mg·kg?1), and DOX (5 mg·kg?1). [138]
CaCO3 DOX The in vitro chemosensitivity test utilizing MTT, modified neutral red/trypan blue assay, and LDH were used to demonstrate that CaCO3/DOX nanocrystals could mitigate tumor cell growth more than free DOX. [140]
Mesoporous CaCO3 Etoposide The MTT assay showed that etoposide-loaded mesoporous CaCO3 inhibited SGC-7901 cells more effectively and lessened the toxic effects of etoposide in HEK 293 T cells compared with free etoposide. [142]
α-Ketoglutaric acid-modified CA AZ628 (Raf-kinase inhibitor) The MTT assay on MCF-7 and 4T1 cells showed that the %cytotoxicity of AZ628-loaded α-KAMCA NPs was substantially greater than that of AZ628-loaded CA NPs, and only AZ628 at similar doses. In MCF-7 and 4T1 cells, AZ628-loaded α-KAMCA NPs displayed around 9% and 12% higher cytotoxicity, respectively, than AZ628-loaded CA NPs, and a 30% greater cytotoxic efficacy compared to free drugs. [150]
Citrate-modified CA and α-ketoglutaric acid-modified CA CYP, DOX 1. Compared with CYP alone, the antitumor efficacy of the group treated with CYP-loaded α-ketoglutaric acid-modified CA NPs showed a five-fold reduction in tumor growth in murine breast cancer models. In addition, the CYP-loaded NPs accumulated more in the tumor than free CYP.2. Biodistribution studies showed less accumulation of DOX-loaded NPs in the heart than free DOX, suggesting their effectiveness in attenuating cardiotoxicity in mice. These findings indicated that both citrate-modified CA and α-ketoglutaric acid-modified CA carriers could prolong the circulation time, increase the antitumor effect, and abate the toxicity of the chemotherapeutic drugs in healthy tissues. [148]
Citrate-modified CA DOX In MCF-7 cells, citrate-modified CA enhanced the cellular uptake and had a half-maximal inhibitory concentration 1000 times lower than free DOX. [151]
Fe-PEI NPs Cisplatin Fe-PEI NPs loaded cisplatin could more effectively suppress cancer cell growth (in vitro and in vivo) than free cisplatin. [163]
Anti-EGFR-TCS-GNSs PTX In MDA-MB-231 cells, the anti-EGFR-PTX-TCS-GNSs + laser irradiation group induced 81.9% apoptosis. In comparison, the photothermal therapeutic potencies of free PTX, PTX-TCSGNSs, and anti-EGFR-PTX-TCS-GNSs were 25.45%, 17.15%, and 13.27%, respectively. The anti-EGFR-PTX-TCS-GNSs + laser irradiation group demonstrated a 100% survival rate over a period of 34 d, compared to all other groups. In addition, antibody-decorated conjugates uptake was four times than that of non-targeted samples after 5 h of treatment, demonstrating the receptor-mediated internalization of anti-EGFR-PTX-TCS-GNS. [181]
Lysine/SWCNTs DOX The inhibition of tumor growth on SMMC-7721 cells and the sarcoma 180-bearing mice was significantly enhanced after treatment with DOX-lysine/SWCNTs in near-infrared laser light at 808 nm. A dramatic rise in body weight, food and water intake, and mental state was noticed in sarcoma 180 tumor-induced mice on DOX-lysine/SWCNTs treatment compared to free DOX. [208]
Mn@CaCO3-loaded PD-L1-targeting siRNA ICG In Lewis lung cancer cells, Mn@CaCO3/ICG-loaded PD-L1-targeting siRNA increased the PDT effect in vitro. Furthermore, in vivo tests revealed that this nanoplatform was capable of transporting the cargos to the tumor and inhibit tumor growth more efficiently than free ICG. [218]
PEI/PEG/MSNs DOX Using near-infrared fluorescence imaging and elemental Si analysis, the human squamous carcinoma xenografts in nude mice after intravenous injection revealed passive accumulation of 12% of the tail vein-injected PEI/PEG/MSNs at the tumour site, indicating successful cellular internalization and DOX delivery to KB-31 cells. The resultant NPs showed apoptosis and tumor shrinkage that was greater than that of free DOX. [219]
F-FMSNs CPT In tumor regression study using nude mice induced with MCF-7 cells, administration of CPT suppressed tumor growth by 14% at the end of the study (the 68th day) compared to control, indicating that it is an efficient tumor-suppressing agent when dissolved in DMSO. In contrast, from day 48 on, the average tumor volumes in groups treated with CPT-loaded FMSN or CPT-loaded F-FMSN continued to decrease at a faster rate. [220]
ZnO-Au-PLA-GPPS-FA-1 and ZnO-Au-PLA-GPPS-FA-2 CPT The CPT-loaded nanocarriers were more cytotoxic to Hela cells at the same concentration of up to 75 μg·mL?1 for ZnO-Au-PLA-GPPS-FA-1 and ZnO-Au-PLA-GPPS-FA-2 nanocarriers, containing about 4.7 and 6.5 μg·mL?1 free CPT in both systems, respectively. Despite the same levels of CPT, the inhibition rates of the CPT-loaded nanocarriers were greater than those of the free CPT. [221]
ZnO DOX The cytotoxicity study showed that DOX-loaded ZnO NPs had a greater anticancer effect than either blank ZnO NPs or DOX alone. [222]
LDH RH The MTT assay reveals that RH-LDHs delivered drugs more effectively to cancer cells cancer cells compared to pure RH due to pure RH’s poor bioavailability. [223]
LDH MTX The half maximal inhibitory concentration of MTX-LDH (48 h) suggested that MTX-LDH had improved efficacy and sensitivity in human colon cancer cells HCT-116. In comparison, naked MTX took 72 h to achieve similar outcomes. [224]
Carboxylate-modified MgAl LDH Cisplatin The anti-proliferative effect of cisplatin-conjugated carboxylate-modified MgAl LDH on colon cancer cell lines was stronger than that of the free drug, according to in vivo data. [225]
NaCa-LDH DAC On malignant (A-375) melanoma and breast cancer (MCF-7) cell lines, the anticancer activity of DAC-NaCa-LDH is higher than that of free DAC. [226]
PLGA-coated MgAl LDH (PLGA-LDH) MTX Greater therapeutic efficacy was observed with PLGA-LDH-MTX than that of the bare MTX, according to in vitro and in vivo experiments. [227228]
FA-modified nanocarrier based on the self-assembly of delaminated CoAl LDH and MnO2 DOX, PTX Based on both in vitro cytotoxicity studies and a xenograft tumor model of hepatoma, this nanocarrier was more effective in fighting cancer than either the free drugs alone or the corresponding cocktail solutions. [229]
GE11-Se NPs Ori GE11-Ori-Se NPs caused a larger level of ROS in KYSE-150 cells than oridonin or Chi-Se NPs, implying that the combination of Chi-Se NPs and oridonin synergistically increased intracellular ROS levels in KYSE-150 cells, increasing the anticancer activity. [230]
FA-QD DOX 1. Fluorescence microscopy and the MTT assay revealed that the folate-targeted DOX-QD NPs exhibit greater toxic effects than non-targeted NPs and the free DOX, demonstrating their preferential accumulation in 4T1 and MCF-7 cells in vitro.2. The in vivo tumor inhibitory impact of FA-QD-DOX NPs revealed that the targeted formulation outperformed the non-targeted formulation and free DOX in terms of therapeutic efficacy. [231]
Herceptin coupled Au-Fe3O4 NPs (Au-Fe3O4-Herceptin) Cisplatin The platin-Au-Fe3O4-Herceptin NPs serve as nanocarriers for the targeted delivery of platin into Her2-positive breast cancer cells (Sk-Br3). The half-maximal inhibitory concentration (IC50) of platin-Au-Fe3O4-Herceptin NPs against Sk-Br3 cells is 1.76 μg of Pt/mL, which is significantly less than the 3.5 μg·mL?1 required for cisplatin. [232]
Au NPs with a PEG spacer linked via an acid-labile linkage (DOX-Hyd@AuNPs) DOX When compared to free DOX, DOX-Hyd@AuNPs boosted drug accumulation and retention in multidrug-resistant MCF-7/ADR cancer cells, resulting in higher toxicity and greater apoptosis in MCF-7/ADR cancer cells. [233]
MSA-capped gold nanoconstructs loaded with SMI#9 (Rad6 protein inhibitor) Cisplatin In vitro, the effective dose of cisplatin required to suppress the development of 50% of cancer cells (4.9 μmol·L?1) was about five times lower than free cisplatin (> 25 μmol·L?1). [234]
Calcium phosphate-polymer hybrid with inhibitors for microRNA-221 and microRNA-222 pac MTS assay was used to evaluate the cytotoxic efficacy of pac-encapsulated NPs (NP(pac)), co-delivery NPs (NP(pac/miRi) or NP(pac/miRi NC)), and free pac in MDA-MB-231 cells after 72 h. The difference between the encapsulated form (NP(pac)) and the free form of pac in terms of cell viability was small. At a high dosage of 0.67 μg·mL?1, pac reduced cell viability by 40%. At the same dosage of 0.67 μg·mL?1, the cell viability of NP(pac/miRi) decreased by 80%. The cytotoxicity evaluation at a broader range of concentrations demonstrated that the NP(pac/miRi) only needed 1% of the pac in the free form to produce the same cytotoxicity (around 80%). [235]
PEGylated Au NP Pc 4 The in vivo drug delivery period for PDT has been lowered to less than 2 h with the Au NP-Pc 4 conjugates, compared to 2 d with the free Pc 4 conjugates. [236]
Tab.1  Comparison of efficacy between drugs and drugs-loaded inorganic nanocarriers
1 G, Wang Y, Chen P, Wang , et al.. Preferential tumor accumulation and desirable interstitial penetration of poly(lactic-co-glycolic acid) nanoparticles with dual coating of chitosan oligosaccharide and polyethylene glycol-poly(D,L-lactic acid). Acta Biomaterialia, 2016, 29: 248– 260
https://doi.org/10.1016/j.actbio.2015.10.017 pmid: 26476340
2 D, Laha A, Pramanik S, Chattopadhyay , et al.. Folic acid modified copper oxide nanoparticles for targeted delivery in in vitro and in vivo systems. RSC Advances, 2015, 5( 83): 68169– 68178
https://doi.org/10.1039/C5RA08110F
3 J, Williams R, Lansdown R, Sweitzer , et al.. Nanoparticle drug delivery system for intravenous delivery of topoisomerase inhibitors. Journal of Controlled Release, 2003, 91( 1–2): 167– 172
https://doi.org/10.1016/S0168-3659(03)00241-4 pmid: 12932648
4 J C, Leroux E, Allémann Jaeghere F, De , et al.. Biodegradable nanoparticles — from sustained release formulations to improved site specific drug delivery. Journal of Controlled Release, 1996, 39( 2–3): 339– 350
https://doi.org/10.1016/0168-3659(95)00164-6
5 S, Gupta M K Gupta . Possible role of nanocarriers in drug delivery against cervical cancer. Nano Reviews & Experiments, 2017, 8( 1): 1335567
https://doi.org/10.1080/20022727.2017.1335567 pmid: 30410707
6 K T Nguyen . Targeted nanoparticles for cancer therapy: promises and challenge. Journal of Nanomedicine & Nanotechnology, 2011, 2( 5): 103e
https://doi.org/10.4172/2157-7439.1000103e
7 K Ky . Nanotechnology platforms and physiological challenges for cancer therapeutic. Nanomedicine, 2007, 3: 103– 110
8 P, Foroozandeh A A Aziz . Insight into cellular uptake and intracellular trafficking of nanoparticles. Nanoscale Research Letters, 2018, 13( 1): 339
https://doi.org/10.1186/s11671-018-2728-6 pmid: 30361809
9 S T, Haque R A, Islam S H, Gan , et al.. Characterization and evaluation of bone-derived nanoparticles as a novel pH-responsive carrier for delivery of doxorubicin into breast cancer cells. International Journal of Molecular Sciences, 2020, 21( 18): 6721
https://doi.org/10.3390/ijms21186721 pmid: 32937817
10 S M, Moghimi A C, Hunter J C Murray . Long-circulating and target-specific nanoparticles: theory to practice. Pharmacological Reviews, 2001, 53( 2): 283– 318
pmid: 11356986
11 H C, Huang S, Barua G, Sharma , et al.. Inorganic nanoparticles for cancer imaging and therapy. Journal of Controlled Release, 2011, 155( 3): 344– 357
https://doi.org/10.1016/j.jconrel.2011.06.004 pmid: 21723891
12 S T, Haque E H Chowdhury . Recent progress in delivery of therapeutic and imaging agents utilizing organic-inorganic hybrid nanoparticles. Current Drug Delivery, 2018, 15( 4): 485– 496
https://doi.org/10.2174/1567201814666171120114034 pmid: 29165073
13 S T, Haque M E, Karim I, Othman , et al.. Mitigating off-target distribution and enhancing cytotoxicity in breast cancer cells with alpha-ketoglutaric acid-modified Fe/Mg-CA nanoparticles. Journal of Pharmaceutical Investigation, 2022, 52( 3): 367– 386
https://doi.org/10.1007/s40005-022-00571-1
14 A C, Anselmo S Mitragotri . Nanoparticles in the clinic: an update. Bioengineering & Translational Medicine, 2019, 4( 3): e10143
https://doi.org/10.1002/btm2.10143 pmid: 31572799
15 M J, Mitchell M M, Billingsley R M, Haley , et al.. Engineering precision nanoparticles for drug delivery. Nature Reviews Drug Discovery, 2021, 20( 2): 101– 124
https://doi.org/10.1038/s41573-020-0090-8 pmid: 33277608
16 Y, Chen Z, Xue D, Zheng , et al.. Sodium chloride modified silica nanoparticles as a non-viral vector with a high efficiency of DNA transfer into cells. Current Gene Therapy, 2003, 3( 3): 273– 279
https://doi.org/10.2174/1566523034578339 pmid: 12762484
17 Z P, Xu Q H, Zeng G Q, Lu , et al.. Inorganic nanoparticles as carriers for efficient cellular delivery. Chemical Engineering Science, 2006, 61( 3): 1027– 1040
https://doi.org/10.1016/j.ces.2005.06.019
18 M C Garnett . Gene-delivery systems using cationic polymers. Critical Reviews™ in Therapeutic Drug Carrier Systems, 1999, 16( 2): 147– 207
19 F, Wang C, Li J, Cheng , et al.. Recent advances on inorganic nanoparticle-based cancer therapeutic agents. International Journal of Environmental Research and Public Health, 2016, 13( 12): 1182
https://doi.org/10.3390/ijerph13121182 pmid: 27898016
20 J V, Jokerst T, Lobovkina R N, Zare , et al.. Nanoparticle PEGylation for imaging and therapy. Nanomedicine, 2011, 6( 4): 715– 728
https://doi.org/10.2217/nnm.11.19 pmid: 21718180
21 J D, Byrne T, Betancourt L Brannon-Peppas . Active targeting schemes for nanoparticle systems in cancer therapeutics. Advanced Drug Delivery Reviews, 2008, 60( 15): 1615– 1626
https://doi.org/10.1016/j.addr.2008.08.005 pmid: 18840489
22 G, Yang X, Sun J, Liu , et al.. Light-responsive, singlet-oxygen-triggered on-demand drug release from photosensitizer-doped mesoporous silica nanorods for cancer combination therapy. Advanced Functional Materials, 2016, 26( 26): 4722– 4732
https://doi.org/10.1002/adfm.201600722
23 Z, Zhang J, Wang X, Nie , et al.. Near infrared laser-induced targeted cancer therapy using thermoresponsive polymer encapsulated gold nanorods. Journal of the American Chemical Society, 2014, 136( 20): 7317– 7326
https://doi.org/10.1021/ja412735p pmid: 24773323
24 Y Q, Ye F L, Yang F Q, Hu , et al.. Core-modified chitosan-based polymeric micelles for controlled release of doxorubicin. International Journal of Pharmaceutics, 2008, 352( 1–2): 294– 301
https://doi.org/10.1016/j.ijpharm.2007.10.035 pmid: 18096336
25 Y Q, Ye F L, Yang F Q, Hu , et al.. Core-modified chitosan-based polymeric micelles for controlled release of doxorubicin. International Journal of Pharmaceutics, 2008, 352( 1–2): 294– 301
https://doi.org/10.1016/j.ijpharm.2007.10.035 pmid: 18096336
26 K, Maier-Hauff F, Ulrich D, Nestler , et al.. Efficacy and safety of intratumoral thermotherapy using magnetic iron-oxide nanoparticles combined with external beam radiotherapy on patients with recurrent glioblastoma multiforme. Journal of Neuro-Oncology, 2011, 103( 2): 317– 324
https://doi.org/10.1007/s11060-010-0389-0 pmid: 20845061
27 C C, Berry S, Wells S, Charles , et al.. Dextran and albumin derivatised iron oxide nanoparticles: influence on fibroblasts in vitro. Biomaterials, 2003, 24( 25): 4551– 4557
https://doi.org/10.1016/S0142-9612(03)00237-0 pmid: 12950997
28 A K, Gupta A S Curtis . Lactoferrin and ceruloplasmin derivatized superparamagnetic iron oxide nanoparticles for targeting cell surface receptors. Biomaterials, 2004, 25( 15): 3029– 3040
https://doi.org/10.1016/j.biomaterials.2003.09.095 pmid: 14967536
29 A K, Gupta M Gupta . Cytotoxicity suppression and cellular uptake enhancement of surface modified magnetic nanoparticles. Biomaterials, 2005, 26( 13): 1565– 1573
https://doi.org/10.1016/j.biomaterials.2004.05.022 pmid: 15522758
30 F Masood . Polymeric nanoparticles for targeted drug delivery system for cancer therapy. Materials Science and Engineering C, 2016, 60: 569– 578
https://doi.org/10.1016/j.msec.2015.11.067 pmid: 26706565
31 D C, Drummond O, Meyer K, Hong , et al.. Optimizing liposomes for delivery of chemotherapeutic agents to solid tumors. Pharmacological Reviews, 1999, 51( 4): 691– 743
pmid: 10581328
32 H, Shmeeda Y, Amitay D, Tzemach , et al.. Liposome encapsulation of zoledronic acid results in major changes in tissue distribution and increase in toxicity. Journal of Controlled Release, 2013, 167( 3): 265– 275
https://doi.org/10.1016/j.jconrel.2013.02.003 pmid: 23419948
33 C G, Hadjipanayis R, Machaidze M, Kaluzova , et al.. EGFRvIII antibody-conjugated iron oxide nanoparticles for magnetic resonance imaging-guided convection-enhanced delivery and targeted therapy of glioblastoma. Cancer Research, 2010, 70( 15): 6303– 6312
https://doi.org/10.1158/0008-5472.CAN-10-1022 pmid: 20647323
34 I H, El-Sayed X, Huang M A El-Sayed . Surface plasmon resonance scattering and absorption of anti-EGFR antibody conjugated gold nanoparticles in cancer diagnostics: applications in oral cancer. Nano Letters, 2005, 5( 5): 829– 834
https://doi.org/10.1021/nl050074e pmid: 15884879
35 M A, Khan D, Singh A, Ahmad , et al.. Revisiting inorganic nanoparticles as promising therapeutic agents: a paradigm shift in oncological theranostics. European Journal of Pharmaceutical Sciences, 2021, 164: 105892
https://doi.org/10.1016/j.ejps.2021.105892 pmid: 34052295
36 M C, Scicluna L Vella-Zarb . Evolution of nanocarrier drug-delivery systems and recent advancements in covalent organic framework-drug systems. ACS Applied Nano Materials, 2020, 3( 4): 3097– 3115
https://doi.org/10.1021/acsanm.9b02603
37 C, Bharti U, Nagaich A K, Pal , et al.. Mesoporous silica nanoparticles in target drug delivery system: a review. International Journal of Pharmaceutical Investigation, 2015, 5( 3): 124– 133
https://doi.org/10.4103/2230-973X.160844 pmid: 26258053
38 S W, Song K, Hidajat S Kawi . Functionalized SBA-15 materials as carriers for controlled drug delivery: influence of surface properties on matrix-drug interactions. Langmuir, 2005, 21( 21): 9568– 9575
https://doi.org/10.1021/la051167e pmid: 16207037
39 N, Varga M, Benkő D, Sebők , et al.. Mesoporous silica core–shell composite functionalized with polyelectrolytes for drug delivery. Microporous and Mesoporous Materials, 2015, 213: 134– 141
https://doi.org/10.1016/j.micromeso.2015.02.008
40 Y, Wang Q, Zhao N, Han , et al.. Mesoporous silica nanoparticles in drug delivery and biomedical applications. Nanomedicine: Nanotechnology, Biology, and Medicine, 2015, 11( 2): 313– 327
https://doi.org/10.1016/j.nano.2014.09.014 pmid: 25461284
41 L, Xiong X, Du B, Shi , et al.. Tunable stellate mesoporous silica nanoparticles for intracellular drug delivery. Journal of Materials Chemistry B: Materials for Biology and Medicine, 2015, 3( 8): 1712– 1721
https://doi.org/10.1039/C4TB01601G pmid: 32262444
42 M, Karimi P S, Zangabad A, Ghasemi, et al.. Chapter 7: Nanotoxicology and future scope for smart nanoparticles. In: M, Karimi P S, Zangabad A, Ghasemi, et al.., eds. Smart External Stimulus-Responsive Nanocarriers for Drug and Gene Delivery. Morgan & Claypool Publishers, 2015
43 T, Asefa Z Tao . Biocompatibility of mesoporous silica nanoparticles. Chemical Research in Toxicology, 2012, 25( 11): 2265– 2284
https://doi.org/10.1021/tx300166u pmid: 22823891
44 N, Wang X, Cheng N, Li , et al.. Nanocarriers and their loading strategies. Advanced Healthcare Materials, 2019, 8( 6): 1801002
https://doi.org/10.1002/adhm.201801002 pmid: 30450761
45 J, Lu M, Liong J I, Zink , et al.. Mesoporous silica nanoparticles as a delivery system for hydrophobic anticancer drugs. Small, 2007, 3( 8): 1341– 1346
https://doi.org/10.1002/smll.200700005 pmid: 17566138
46 L, Tang J Cheng . Nonporous silica nanoparticles for nanomedicine application. Nano Today, 2013, 8( 3): 290– 312
https://doi.org/10.1016/j.nantod.2013.04.007 pmid: 23997809
47 J J, Corbalan C, Medina A, Jacoby , et al.. Amorphous silica nanoparticles aggregate human platelets: potential implications for vascular homeostasis. International Journal of Nanomedicine, 2012, 7: 631– 639
pmid: 22334785
48 F, Chen H, Hong Y, Zhang , et al.. In vivo tumor targeting and image-guided drug delivery with antibody-conjugated, radiolabeled mesoporous silica nanoparticles. ACS Nano, 2013, 7( 10): 9027– 9039
https://doi.org/10.1021/nn403617j pmid: 24083623
49 M, Karimi M, Eslami P, Sahandi-Zangabad , et al.. pH-Sensitive stimulus-responsive nanocarriers for targeted delivery of therapeutic agents. Wiley Interdisciplinary Reviews: Nanomedicine and Nanobiotechnology, 2016, 8( 5): 696– 716
https://doi.org/10.1002/wnan.1389 pmid: 26762467
50 M, Karimi P, Sahandi-Zangabad A, Ghasemi , et al.. Temperature-responsive smart nanocarriers for delivery of therapeutic agents: applications and recent advances. ACS Applied Materials & Interfaces, 2016, 8( 33): 21107– 21133
https://doi.org/10.1021/acsami.6b00371 pmid: 27349465
51 J M, Rosenholm A, Meinander E, Peuhu , et al.. Targeting of porous hybrid silica nanoparticles to cancer cells. ACS Nano, 2009, 3( 1): 197– 206
https://doi.org/10.1021/nn800781r pmid: 19206267
52 D S, Karaman D, Desai R, Senthilkumar , et al.. Shape engineering vs organic modification of inorganic nanoparticles as a tool for enhancing cellular internalization. Nanoscale Research Letters, 2012, 7( 1): 358
https://doi.org/10.1186/1556-276X-7-358 pmid: 22747910
53 T, Xia M, Kovochich M, Liong , et al.. Polyethyleneimine coating enhances the cellular uptake of mesoporous silica nanoparticles and allows safe delivery of siRNA and DNA constructs. ACS Nano, 2009, 3( 10): 3273– 3286
https://doi.org/10.1021/nn900918w pmid: 19739605
54 W, Ngamcherdtrakul J, Morry S, Gu , et al.. Cationic polymer modified mesoporous silica nanoparticles for targeted siRNA delivery to HER2+ breast cancer. Advanced Functional Materials, 2015, 25( 18): 2646– 2659
https://doi.org/10.1002/adfm.201404629 pmid: 26097445
55 Y, Wang Y, Cui J, Huang , et al.. Redox and pH dual-responsive mesoporous silica nanoparticles for site-specific drug delivery. Applied Surface Science, 2015, 356: 1282– 1288
https://doi.org/10.1016/j.apsusc.2015.07.151
56 D R, Radu C Y, Lai K, Jeftinija , et al.. A polyamidoamine dendrimer-capped mesoporous silica nanosphere-based gene transfection reagent. Journal of the American Chemical Society, 2004, 126( 41): 13216– 13217
https://doi.org/10.1021/ja046275m pmid: 15479063
57 M, Kar N, Tiwari M, Tiwari , et al.. Poly-L-arginine grafted silica mesoporous nanoparticles for enhanced cellular uptake and their application in DNA delivery and controlled drug release. Particle & Particle Systems Characterization, 2013, 30( 2): 166– 179
https://doi.org/10.1002/ppsc.201200089
58 Z, Zou D, He X, He , et al.. Natural gelatin capped mesoporous silica nanoparticles for intracellular acid-triggered drug delivery. Langmuir, 2013, 29( 41): 12804– 12810
https://doi.org/10.1021/la4022646 pmid: 24073830
59 I Y, Park I Y, Kim M K, Yoo , et al.. Mannosylated polyethylenimine coupled mesoporous silica nanoparticles for receptor-mediated gene delivery. International Journal of Pharmaceutics, 2008, 359( 1–2): 280– 287
https://doi.org/10.1016/j.ijpharm.2008.04.010 pmid: 18490119
60 H, Meng W X, Mai H, Zhang , et al.. Codelivery of an optimal drug/siRNA combination using mesoporous silica nanoparticles to overcome drug resistance in breast cancer in vitro and in vivo. ACS Nano, 2013, 7( 2): 994– 1005
https://doi.org/10.1021/nn3044066 pmid: 23289892
61 M, Jang Y I, Yoon Y S, Kwon , et al.. Trastuzumab-conjugated liposome-coated fluorescent magnetic nanoparticles to target breast cancer. Korean Journal of Radiology, 2014, 15( 4): 411– 422
https://doi.org/10.3348/kjr.2014.15.4.411 pmid: 25053899
62 Q, Sun Q, You J, Wang , et al.. Theranostic nanoplatform: triple-modal imaging-guided synergistic cancer therapy based on liposome-conjugated mesoporous silica nanoparticles. ACS Applied Materials & Interfaces, 2018, 10( 2): 1963– 1975
https://doi.org/10.1021/acsami.7b13651 pmid: 29276824
63 W, Wei G H, Ma G, Hu , et al.. Preparation of hierarchical hollow CaCO3 particles and the application as anticancer drug carrier. Journal of the American Chemical Society, 2008, 130( 47): 15808– 15810
https://doi.org/10.1021/ja8039585 pmid: 18980322
64 Y, Ueno H, Futagawa Y, Takagi , et al.. Drug-incorporating calcium carbonate nanoparticles for a new delivery system. Journal of Controlled Release, 2005, 103( 1): 93– 98
https://doi.org/10.1016/j.jconrel.2004.11.015 pmid: 15710503
65 S, Chen D, Zhao F, Li , et al.. Co-delivery of genes and drugs with nanostructured calcium carbonate for cancer therapy. RSC Advances, 2012, 2( 5): 1820– 1826
https://doi.org/10.1039/c1ra00527h
66 J, Wang J S, Chen J Y, Zong , et al.. Calcium carbonate/carboxymethyl chitosan hybrid microspheres and nanospheres for drug delivery. The Journal of Physical Chemistry C, 2010, 114( 44): 18940– 18945
https://doi.org/10.1021/jp105906p
67 M, Kester Y, Heakal T, Fox , et al.. Calcium phosphate nanocomposite particles for in vitro imaging and encapsulated chemotherapeutic drug delivery to cancer cells. Nano Letters, 2008, 8( 12): 4116– 4121
https://doi.org/10.1021/nl802098g pmid: 19367878
68 K H, Bae K, Lee C, Kim , et al.. Surface functionalized hollow manganese oxide nanoparticles for cancer targeted siRNA delivery and magnetic resonance imaging. Biomaterials, 2011, 32( 1): 176– 184
https://doi.org/10.1016/j.biomaterials.2010.09.039 pmid: 20934746
69 J, Shi Z, Chen L, Wang , et al.. A tumor-specific cleavable nanosystem of PEG-modified C60@Au hybrid aggregates for radio frequency-controlled release, hyperthermia, photodynamic therapy and X-ray imaging. Acta Biomaterialia, 2016, 29: 282– 297
https://doi.org/10.1016/j.actbio.2015.10.027 pmid: 26485168
70 S, Chen X, Zhao J, Chen , et al.. Mechanism-based tumor-targeting drug delivery system. Validation of efficient vitamin receptor-mediated endocytosis and drug release. Bioconjugate Chemistry, 2010, 21( 5): 979– 987
https://doi.org/10.1021/bc9005656 pmid: 20429547
71 W M, Pardridge R J Boado . Enhanced cellular uptake of biotinylated antisense oligonucleotide or peptide mediated by avidin, a cationic protein. FEBS Letters, 1991, 288( 1–2): 30– 32
https://doi.org/10.1016/0014-5793(91)80996-G pmid: 1879560
72 X, Zeng Y X, Sun X Z, Zhang , et al.. Biotinylated disulfide containing PEI/avidin bioconjugate shows specific enhanced transfection efficiency in HepG2 cells. Organic & Biomolecular Chemistry, 2009, 7( 20): 4201– 4210
https://doi.org/10.1039/b910831a pmid: 19795058
73 U, Wojda P, Goldsmith J L Miller . Surface membrane biotinylation efficiently mediates the endocytosis of avidin bioconjugates into nucleated cells. Bioconjugate Chemistry, 1999, 10( 6): 1044– 1050
https://doi.org/10.1021/bc990059z pmid: 10563774
74 S F Rosebrough . Pharmacokinetics and biodistribution of radiolabeled avidin, streptavidin and biotin. Nuclear Medicine and Biology, 1993, 20( 5): 663– 668
https://doi.org/10.1016/0969-8051(93)90037-U pmid: 8358353
75 B, Schechter R, Silberman R, Arnon , et al.. Tissue distribution of avidin and streptavidin injected to mice ― effect of avidin carbohydrate, streptavidin truncation and exogenous biotin. European Journal of Biochemistry, 1990, 189( 2): 327– 331
https://doi.org/10.1111/j.1432-1033.1990.tb15493.x pmid: 2186907
76 Z, Yao M, Zhang H, Sakahara , et al.. Avidin targeting of intraperitoneal tumor xenografts. Journal of the National Cancer Institute, 1998, 90( 1): 25– 29
https://doi.org/10.1093/jnci/90.1.25 pmid: 9428779
77 M, González C E, Argaraña G D Fidelio . Extremely high thermal stability of streptavidin and avidin upon biotin binding. Biomolecular Engineering, 1999, 16( 1–4): 67– 72
https://doi.org/10.1016/S1050-3862(99)00041-8 pmid: 10796986
78 G Elia . Biotinylation reagents for the study of cell surface proteins. Proteomics, 2008, 8( 19): 4012– 4024
https://doi.org/10.1002/pmic.200800097 pmid: 18763706
79 A, Jain K Cheng . The principles and applications of avidin-based nanoparticles in drug delivery and diagnosis. Journal of Controlled Release, 2017, 245: 27– 40
https://doi.org/10.1016/j.jconrel.2016.11.016 pmid: 27865853
80 K, Hoya L R, Guterman L, Miskolczi , et al.. A novel intravascular drug delivery method using endothelial biotinylation and avidin-biotin binding. Drug Delivery, 2001, 8( 4): 215– 222
https://doi.org/10.1080/107175401317245895 pmid: 11757779
81 N P, Singh E S, Yolcu N, Askenasy , et al.. ProtEx: a novel technology to display exogenous proteins on the cell surface for immunomodulation. Annals of the New York Academy of Sciences, 2005, 1056( 1): 344– 358
https://doi.org/10.1196/annals.1352.036 pmid: 16387700
82 T T, Nguyen K L, Sly J C Conboy . Comparison of the energetics of avidin, streptavidin, neutrAvidin, and anti-biotin antibody binding to biotinylated lipid bilayer examined by second-harmonic generation. Analytical Chemistry, 2012, 84( 1): 201– 208
https://doi.org/10.1021/ac202375n pmid: 22122646
83 D, Artemov N, Mori B, Okollie , et al.. MR molecular imaging of the Her-2/neu receptor in breast cancer cells using targeted iron oxide nanoparticles. Magnetic Resonance in Medicine, 2003, 49( 3): 403– 408
https://doi.org/10.1002/mrm.10406 pmid: 12594741
84 C, Yan Y, Wu J, Feng , et al.. Anti-αvβ3 antibody guided three-step pretargeting approach using magnetoliposomes for molecular magnetic resonance imaging of breast cancer angiogenesis. International Journal of Nanomedicine, 2013, 8: 245– 255
pmid: 23345972
85 A, Barve A, Jain H, Liu , et al.. An enzyme-responsive conjugate improves the delivery of a PI3K inhibitor to prostate cancer. Nanomedicine: Nanotechnology, Biology, and Medicine, 2016, 12( 8): 2373– 2381
https://doi.org/10.1016/j.nano.2016.07.007 pmid: 27478108
86 J M, Steinbach Y E, Seo W M Saltzman . Cell penetrating peptide-modified poly(lactic-co-glycolic acid) nanoparticles with enhanced cell internalization. Acta Biomaterialia, 2016, 30: 49– 61
https://doi.org/10.1016/j.actbio.2015.11.029 pmid: 26602822
87 Rijt S H, van D A, Bölükbas C, Argyo , et al.. Protease-mediated release of chemotherapeutics from mesoporous silica nanoparticles to ex vivo human and mouse lung tumors. ACS Nano, 2015, 9( 3): 2377– 2389
https://doi.org/10.1021/nn5070343 pmid: 25703655
88 V, Oliveri R, D’Agata V, Giglio , et al.. Cyclodextrin-functionalised gold nanoparticles via streptavidin: a supramolecular approach. Supramolecular Chemistry, 2013, 25( 8): 465– 473
https://doi.org/10.1080/10610278.2013.794278
89 B M, Barth R, Sharma E I, Altinoğlu , et al.. Bioconjugation of calcium phosphosilicate composite nanoparticles for selective targeting of human breast and pancreatic cancers in vivo. ACS Nano, 2010, 4( 3): 1279– 1287
https://doi.org/10.1021/nn901297q pmid: 20180585
90 F S, Mozar E H Chowdhury . Surface-modification of carbonate apatite nanoparticles enhances delivery and cytotoxicity of gemcitabine and anastrozole in breast cancer cells. Pharmaceutics, 2017, 9( 2): 21
https://doi.org/10.3390/pharmaceutics9020021 pmid: 28590445
91 P, Bajaj C, Mikoryak R, Wang , et al.. A carbon nanotube-based Raman-imaging immunoassay for evaluating tumor targeting ligands. Analyst, 2014, 139( 12): 3069– 3076
https://doi.org/10.1039/C4AN00258J pmid: 24776815
92 G, Lai J, Wu H, Ju , et al.. Streptavidin-functionalized silver-nanoparticle-enriched carbon nanotube tag for ultrasensitive multiplexed detection of tumor markers. Advanced Functional Materials, 2011, 21( 15): 2938– 2943
https://doi.org/10.1002/adfm.201100396
93 K K, Cotí M E, Belowich M, Liong , et al.. Mechanised nanoparticles for drug delivery. Nanoscale, 2009, 1( 1): 16– 39
https://doi.org/10.1039/b9nr00162j pmid: 20644858
94 K, Ladewig Z P, Xu G Q Lu . Layered double hydroxide nanoparticles in gene and drug delivery. Expert Opinion on Drug Delivery, 2009, 6( 9): 907– 922
https://doi.org/10.1517/17425240903130585 pmid: 19686052
95 S J, Choi J H Choy . Layered double hydroxide nanoparticles as target-specific delivery carriers: uptake mechanism and toxicity. Nanomedicine, 2011, 6( 5): 803– 814
https://doi.org/10.2217/nnm.11.86 pmid: 21793673
96 S J, Choi J M, Oh J H Choy . Biocompatible nanoparticles intercalated with anticancer drug for target delivery: pharmacokinetic and biodistribution study. Journal of Nanoscience and Nanotechnology, 2010, 10( 4): 2913– 2916
https://doi.org/10.1166/jnn.2010.1415 pmid: 20355523
97 J H, Choy J S, Jung J M, Oh , et al.. Layered double hydroxide as an efficient drug reservoir for folate derivatives. Biomaterials, 2004, 25( 15): 3059– 3064
https://doi.org/10.1016/j.biomaterials.2003.09.083 pmid: 14967539
98 J M, Oh S J, Choi G E, Lee , et al.. Inorganic metal hydroxide nanoparticles for targeted cellular uptake through clathrin-mediated endocytosis. Chemistry: An Asian Journal, 2009, 4( 1): 67– 73
https://doi.org/10.1002/asia.200800290 pmid: 18988236
99 A, Nel T, Xia L, Mädler , et al.. Toxic potential of materials at the nanolevel. Science, 2006, 311( 5761): 622– 627
https://doi.org/10.1126/science.1114397 pmid: 16456071
100 S J, Choi J M, Oh J H Choy . Safety aspect of inorganic layered nanoparticles: size-dependency in vitro and in vivo. Journal of Nanoscience and Nanotechnology, 2008, 8( 10): 5297– 5301
https://doi.org/10.1166/jnn.2008.1143 pmid: 19198442
101 J M, Oh T T, Biswick J H Choy . Layered nanomaterials for green materials. Journal of Materials Chemistry, 2009, 19( 17): 2553– 2563
https://doi.org/10.1039/b819094a
102 J, Panyam V Labhasetwar . Biodegradable nanoparticles for drug and gene delivery to cells and tissue. Advanced Drug Delivery Reviews, 2003, 55( 3): 329– 347
https://doi.org/10.1016/S0169-409X(02)00228-4 pmid: 12628320
103 J M, Oh S J, Choi S T, Kim , et al.. Cellular uptake mechanism of an inorganic nanovehicle and its drug conjugates: enhanced efficacy due to clathrin-mediated endocytosis. Bioconjugate Chemistry, 2006, 17( 6): 1411– 1417
https://doi.org/10.1021/bc0601323 pmid: 17105218
104 J M, Oh M, Park S T, Kim , et al.. Efficient delivery of anticancer drug MTX through MTX-LDH nanohybrid system. Journal of Physics and Chemistry of Solids, 2006, 67( 5–6): 1024– 1027
https://doi.org/10.1016/j.jpcs.2006.01.033
105 S J, Choi J M, Oh H E, Chung , et al.. In vivo anticancer activity of methotrexate-loaded layered double hydroxide nanoparticles. Current Pharmaceutical Design, 2013, 19( 41): 7196– 7202
https://doi.org/10.2174/138161281941131219123718 pmid: 23489199
106 A, Javaid M, Bone C Stanley . Effect of fenbufen on the quality of life of patients with pain from squamous-cell carcinoma of the bronchus. In: Proceedings of the Thorax, 1988, 244
107 B, Li J, He D G, Evans , et al.. Inorganic layered double hydroxides as a drug delivery system — intercalation and in vitro release of fenbufen. Applied Clay Science, 2004, 27( 3–4): 199– 207
https://doi.org/10.1016/j.clay.2004.07.002
108 V, Ambrogi G, Fardella G, Grandolini , et al.. Intercalation compounds of hydrotalcite-like anionic clays with antiinflammatory agents ― I. Intercalation and in vitro release of ibuprofen. International Journal of Pharmaceutics, 2001, ( 1–2): 23– 32
https://doi.org/10.1016/S0378-5173(01)00629-9 pmid: 11376964
109 N T, Whilton P J, Vickers S Mann . Bioinorganic clays: synthesis and characterization of amino-andpolyamino acid intercalated layered double hydroxides. Journal of Materials Chemistry, 1997, 7( 8): 1623– 1629
https://doi.org/10.1039/a701237c
110 Y H, Xue R, Zhang X Y, Sun , et al.. The construction and characterization of layered double hydroxides as delivery vehicles for podophyllotoxins. Journal of Materials Science: Materials in Medicine, 2008, 19( 3): 1197– 1202
https://doi.org/10.1007/s10856-007-3221-4 pmid: 17701296
111 D H, Park J, Cho O J, Kwon , et al.. Biodegradable inorganic nanovector: passive versus active tumor targeting in siRNA transportation. Angewandte Chemie International Edition in English, 2016, 55( 14): 4582– 4586
https://doi.org/10.1002/anie.201510844 pmid: 26879376
112 L, Li Y, Qian L, Sun , et al.. Albumin-stabilized layered double hydroxide nanoparticles synergized combination chemotherapy for colorectal cancer treatment. Nanomedicine: Nanotechnology, Biology, and Medicine, 2021, 34: 102369
https://doi.org/10.1016/j.nano.2021.102369 pmid: 33636347
113 G, Choi I R, Jeon H, Piao , et al.. Highly condensed boron cage cluster anions in 2d carrier and its enhanced antitumor efficiency for boron neutron capture therapy. Advanced Functional Materials, 2018, 28( 27): 1704470
https://doi.org/10.1002/adfm.201704470
114 Z, Guo W, Xie J, Lu , et al.. Ferrous ions doped layered double hydroxide: smart 2D nanotheranostic platform with imaging-guided synergistic chemo/photothermal therapy for breast cancer. Biomaterials Science, 2021, 9( 17): 5928– 5938
https://doi.org/10.1039/D1BM00765C pmid: 34308465
115 T, Xu J, Liu L, Sun , et al.. Enhancing tumor accumulation and cellular uptake of layered double hydroxide nanoparticles by coating/detaching pH-triggered charge-convertible polymers. ACS Omega, 2021, 6( 5): 3822– 3830
https://doi.org/10.1021/acsomega.0c05520 pmid: 33585761
116 M, Baek I S, Kim J, Yu , et al.. Effect of different forms of anionic nanoclays on cytotoxicity. Journal of Nanoscience and Nanotechnology, 2011, 11( 2): 1803– 1806
https://doi.org/10.1166/jnn.2011.3408 pmid: 21456296
117 Z P, Xu M, Niebert K, Porazik , et al.. Subcellular compartment targeting of layered double hydroxide nanoparticles. Journal of Controlled Release, 2008, 130( 1): 86– 94
https://doi.org/10.1016/j.jconrel.2008.05.021 pmid: 18614254
118 R, Hong G, Han J M, Fernández , et al.. Glutathione-mediated delivery and release using monolayer protected nanoparticle carriers. Journal of the American Chemical Society, 2006, 128( 4): 1078– 1079
https://doi.org/10.1021/ja056726i pmid: 16433515
119 T R, Fadel T M Fahmy . Immunotherapy applications of carbon nanotubes: from design to safe applications. Trends in Biotechnology, 2014, 32( 4): 198– 209
https://doi.org/10.1016/j.tibtech.2014.02.005 pmid: 24630474
120 C H, Villa T, Dao I, Ahearn , et al.. Single-walled carbon nanotubes deliver peptide antigen into dendritic cells and enhance IgG responses to tumor-associated antigens. ACS Nano, 2011, 5( 7): 5300– 5311
https://doi.org/10.1021/nn200182x pmid: 21682329
121 C A, Dyke M P, Stewart J M Tour . Separation of single-walled carbon nanotubes on silica gel. Materials morphology and Raman excitation wavelength affect data interpretation. Journal of the American Chemical Society, 2005, 127( 12): 4497– 4509
https://doi.org/10.1021/ja042828h pmid: 15783233
122 K M, Lee L, Li L Dai . Asymmetric end-functionalization of multi-walled carbon nanotubes. Journal of the American Chemical Society, 2005, 127( 12): 4122– 4123
https://doi.org/10.1021/ja0423670 pmid: 15783165
123 Z, Liu K, Chen C, Davis , et al.. Drug delivery with carbon nanotubes for in vivo cancer treatment. Cancer Research, 2008, 68( 16): 6652– 6660
https://doi.org/10.1158/0008-5472.CAN-08-1468 pmid: 18701489
124 Z, Sobhani M A, Behnam F, Emami , et al.. Photothermal therapy of melanoma tumor using multiwalled carbon nanotubes. International Journal of Nanomedicine, 2017, 12: 4509– 4517
https://doi.org/10.2147/IJN.S134661 pmid: 28684911
125 C, Sacchetti N, Rapini A, Magrini , et al.. In vivo targeting of intratumor regulatory T cells using PEG-modified single-walled carbon nanotubes. Bioconjugate Chemistry, 2013, 24( 6): 852– 858
https://doi.org/10.1021/bc400070q pmid: 23682992
126 J S, Lee J J, Green K T, Love , et al.. Gold, poly(β-amino ester) nanoparticles for small interfering RNA delivery. Nano Letters, 2009, 9( 6): 2402– 2406
https://doi.org/10.1021/nl9009793 pmid: 19422265
127 L, Li M, Nurunnabi M, Nafiujjaman , et al.. A photosensitizer-conjugated magnetic iron oxide/gold hybrid nanoparticle as an activatable platform for photodynamic cancer therapy. Journal of Materials Chemistry B: Materials for Biology and Medicine, 2014, 2( 19): 2929– 2937
https://doi.org/10.1039/c4tb00181h pmid: 32261487
128 Y, Cheng T L, Doane C H, Chuang , et al.. Near infrared light-triggered drug generation and release from gold nanoparticle carriers for photodynamic therapy. Small, 2014, 10( 9): 1799– 1804
https://doi.org/10.1002/smll.201303329 pmid: 24515950
129 M D, Massich D A, Giljohann A L, Schmucker , et al.. Cellular response of polyvalent oligonucleotide-gold nanoparticle conjugates. ACS Nano, 2010, 4( 10): 5641– 5646
https://doi.org/10.1021/nn102228s pmid: 20860397
130 R, Huschka J, Zuloaga M W, Knight , et al.. Light-induced release of DNA from gold nanoparticles: nanoshells and nanorods. Journal of the American Chemical Society, 2011, 133( 31): 12247– 12255
https://doi.org/10.1021/ja204578e pmid: 21736347
131 C C, Chen Y P, Lin C W, Wang , et al.. DNA-gold nanorod conjugates for remote control of localized gene expression by near infrared irradiation. Journal of the American Chemical Society, 2006, 128( 11): 3709– 3715
https://doi.org/10.1021/ja0570180 pmid: 16536544
132 S, Dhar F X, Gu R, Langer , et al.. Targeted delivery of cisplatin to prostate cancer cells by aptamer functionalized Pt(IV) prodrug-PLGA-PEG nanoparticles. Proceedings of the National Academy of Sciences of the United States of America, 2008, 105( 45): 17356– 17361
https://doi.org/10.1073/pnas.0809154105 pmid: 18978032
133 S, Dhar Z, Liu J, Thomale , et al.. Targeted single-wall carbon nanotube-mediated Pt(IV) prodrug delivery using folate as a homing device. Journal of the American Chemical Society, 2008, 130( 34): 11467– 11476
https://doi.org/10.1021/ja803036e pmid: 18661990
134 S, Dhar W L, Daniel D A, Giljohann , et al.. Polyvalent oligonucleotide gold nanoparticle conjugates as delivery vehicles for platinum(IV) warheads. Journal of the American Chemical Society, 2009, 131( 41): 14652– 14653
https://doi.org/10.1021/ja9071282 pmid: 19778015
135 Y, Min C, Mao D, Xu , et al.. Gold nanorods for platinum based prodrug delivery. Chemical Communications, 2010, 46( 44): 8424– 8426
https://doi.org/10.1039/c0cc03108a pmid: 20936244
136 W, Ding L Guo . Immobilized transferrin Fe3O4@SiO2 nanoparticle with high doxorubicin loading for dual-targeted tumor drug delivery. International Journal of Nanomedicine, 2013, 8: 4631– 4639
pmid: 24348038
137 M, Kresse S, Wagner D, Pfefferer , et al.. Targeting of ultrasmall superparamagnetic iron oxide (USPIO) particles to tumor cells in vivo by using transferrin receptor pathways. Magnetic Resonance in Medicine, 1998, 40( 2): 236– 242
https://doi.org/10.1002/mrm.1910400209 pmid: 9702705
138 M K, Yu Y Y, Jeong J, Park , et al.. Drug-loaded superparamagnetic iron oxide nanoparticles for combined cancer imaging and therapy in vivo. Angewandte Chemie International Edition in English, 2008, 47( 29): 5362– 5365
https://doi.org/10.1002/anie.200800857 pmid: 18551493
139 A, Som R, Raliya L, Tian , et al.. Monodispersed calcium carbonate nanoparticles modulate local pH and inhibit tumor growth in vivo. Nanoscale, 2016, 8( 25): 12639– 12647
https://doi.org/10.1039/C5NR06162H pmid: 26745389
140 A S, Kamba M, Ismail T A T, Ibrahim , et al.. A pH-sensitive, biobased calcium carbonate aragonite nanocrystal as a novel anticancer delivery system. BioMed Research International, 2013, 2013: 587451
https://doi.org/10.1155/2013/587451
141 N I, Hammadi Y, Abba M N M, Hezmee , et al.. Formulation of a sustained release docetaxel loaded cockle shell-derived calcium carbonate nanoparticles against breast cancer. Pharmaceutical Research, 2017, 34( 6): 1193– 1203
https://doi.org/10.1007/s11095-017-2135-1 pmid: 28382563
142 H, Peng K, Li T, Wang , et al.. Preparation of hierarchical mesoporous CaCO3 by a facile binary solvent approach as anticancer drug carrier for etoposide. Nanoscale Research Letters, 2013, 8( 1): 321
https://doi.org/10.1186/1556-276X-8-321 pmid: 23849350
143 J, Li Y, Yang L Huang . Calcium phosphate nanoparticles with an asymmetric lipid bilayer coating for siRNA delivery to the tumor. Journal of Controlled Release, 2012, 158( 1): 108– 114
https://doi.org/10.1016/j.jconrel.2011.10.020 pmid: 22056915
144 Z, Wu J, Chen Y, Sun , et al.. Tumor microenvironment-response calcium phosphate hybrid nanoparticles enhanced siRNAs targeting tumors in vivo. Journal of Biomedical Nanotechnology, 2018, 14( 10): 1816– 1825
https://doi.org/10.1166/jbn.2018.2606 pmid: 30041727
145 Y, Dong H, Liao H, Fu , et al.. pH-sensitive shell–core platform block DNA repair pathway to amplify irreversible DNA damage of triple negative breast cancer. ACS Applied Materials & Interfaces, 2019, 11( 42): 38417– 38428
https://doi.org/10.1021/acsami.9b12140 pmid: 31556584
146 C, Qiu W, Wei J, Sun , et al.. Systemic delivery of siRNA by hyaluronan-functionalized calcium phosphate nanoparticles for tumor-targeted therapy. Nanoscale, 2016, 8( 26): 13033– 13044
https://doi.org/10.1039/C6NR04034A pmid: 27314204
147 S T, Haque M E, Karim S A Z, Abidin , et al.. Fe/Mg-modified carbonate apatite with uniform particle size and unique transport protein-related protein corona efficiently delivers doxorubicin into breast cancer cells. Nanomaterials, 2020, 10( 5): 834
https://doi.org/10.3390/nano10050834 pmid: 32349272
148 S M, Hossain Abidin S A, Zainal E H Chowdhury . Krebs cycle intermediate-modified carbonate apatite nanoparticles drastically reduce mouse tumor burden and toxicity by restricting broad tissue distribution of anticancer drugs. Cancers, 2020, 12( 1): 161
https://doi.org/10.3390/cancers12010161 pmid: 31936503
149 F S, Mozar E H Chowdhury . PEGylation of carbonate apatite nanoparticles prevents opsonin binding and enhances tumor accumulation of gemcitabine. Journal of Pharmaceutical Sciences, 2018, 107( 9): 2497– 2508
https://doi.org/10.1016/j.xphs.2018.05.020 pmid: 29883662
150 S M, Hossain J, Shetty K K, Tha , et al.. α-Ketoglutaric acid-modified carbonate apatite enhances cellular uptake and cytotoxicity of a Raf-kinase inhibitor in breast cancer cells through inhibition of MAPK and PI-3 kinase pathways. Biomedicines, 2019, 7( 1): 4
https://doi.org/10.3390/biomedicines7010004 pmid: 30609867
151 S M, Hossain E H Chowdhury . Citrate- and succinate-modified carbonate apatite nanoparticles with loaded doxorubicin exhibit potent anticancer activity against breast cancer cells. Pharmaceutics, 2018, 10( 1): 32
https://doi.org/10.3390/pharmaceutics10010032 pmid: 29534497
152 G, Verma K, Barick N G, Shetake , et al.. Citrate-functionalized hydroxyapatite nanoparticles for pH-responsive drug delivery. RSC Advances, 2016, 6( 81): 77968– 77976
https://doi.org/10.1039/C6RA10659E
153 I, Rodríguez-Ruiz J M, Delgado-López M A, Durán-Olivencia , et al.. pH-responsive delivery of doxorubicin from citrate-apatite nanocrystals with tailored carbonate content. Langmuir, 2013, 29( 26): 8213– 8221
https://doi.org/10.1021/la4008334 pmid: 23735159
154 E Bilensoy . Cationic nanoparticles for cancer therapy. Expert Opinion on Drug Delivery, 2010, 7( 7): 795– 809
https://doi.org/10.1517/17425247.2010.485983 pmid: 20446858
155 A, Slita A, Egorova E, Casals , et al.. Characterization of modified mesoporous silica nanoparticles as vectors for siRNA delivery. Asian Journal of Pharmaceutical Sciences, 2018, 13( 6): 592– 599
https://doi.org/10.1016/j.ajps.2018.01.006
156 A, Zakeri M A J, Kouhbanani N, Beheshtkhoo , et al.. Polyethylenimine-based nanocarriers in co-delivery of drug and gene: a developing horizon. Nano Reviews & Experiments, 2018, 9( 1): 1488497
https://doi.org/10.1080/20022727.2018.1488497 pmid: 30410712
157 S, Vaidyanathan J, Chen B G, Orr , et al.. Cationic polymer intercalation into the lipid membrane enables intact polyplex DNA escape from endosomes for gene delivery. Molecular Pharmaceutics, 2016, 13( 6): 1967– 1978
https://doi.org/10.1021/acs.molpharmaceut.6b00139 pmid: 27111496
158 R V, Benjaminsen M A, Mattebjerg J R, Henriksen , et al.. The possible “proton sponge” effect of polyethylenimine (PEI) does not include change in lysosomal pH. Molecular Therapy, 2013, 21( 1): 149– 157
https://doi.org/10.1038/mt.2012.185 pmid: 23032976
159 X, Wang D, Niu C, Hu , et al.. Polyethyleneimine-based nanocarriers for gene delivery. Current Pharmaceutical Design, 2015, 21( 42): 6140– 6156
https://doi.org/10.2174/1381612821666151027152907 pmid: 26503146
160 T, Zhang X, Xue D, He , et al.. A prostate cancer-targeted polyarginine-disulfide linked PEI nanocarrier for delivery of microRNA. Cancer Letters, 2015, 365( 2): 156– 165
https://doi.org/10.1016/j.canlet.2015.05.003 pmid: 26054847
161 X, Li Y, Chen M, Wang , et al.. A mesoporous silica nanoparticle–PEI–fusogenic peptide system for siRNA delivery in cancer therapy. Biomaterials, 2013, 34( 4): 1391– 1401
https://doi.org/10.1016/j.biomaterials.2012.10.072 pmid: 23164421
162 J, Shen H C, Kim H, Su , et al.. Cyclodextrin and polyethylenimine functionalized mesoporous silica nanoparticles for delivery of siRNA cancer therapeutics. Theranostics, 2014, 4( 5): 487– 497
https://doi.org/10.7150/thno.8263 pmid: 24672582
163 R, Tutuianu L M, Popescu M B, Preda , et al.. Evaluation of the ability of nanostructured PEI-coated iron oxide nanoparticles to incorporate cisplatin during synthesis. Nanomaterials, 2017, 7( 10): 314
https://doi.org/10.3390/nano7100314 pmid: 29023373
164 G, Liu J, Xie F, Zhang , et al.. N-Alkyl-PEI-functionalized iron oxide nanoclusters for efficient siRNA delivery. Small, 2011, 7( 19): 2742– 2749
https://doi.org/10.1002/smll.201100825 pmid: 21861295
165 L, Zhang T, Wang L, Li , et al.. Multifunctional fluorescent-magnetic polyethyleneimine functionalized Fe3O4–mesoporous silica yolk–shell nanocapsules for siRNA delivery. Chemical Communications, 2012, 48( 69): 8706– 8708
https://doi.org/10.1039/c2cc33472k pmid: 22824833
166 K S, Siu D, Chen X, Zheng , et al.. Non-covalently functionalized single-walled carbon nanotube for topical siRNA delivery into melanoma. Biomaterials, 2014, 35( 10): 3435– 3442
https://doi.org/10.1016/j.biomaterials.2013.12.079 pmid: 24424208
167 H, Wu H, Shi H, Zhang , et al.. Prostate stem cell antigen antibody-conjugated multiwalled carbon nanotubes for targeted ultrasound imaging and drug delivery. Biomaterials, 2014, 35( 20): 5369– 5380
https://doi.org/10.1016/j.biomaterials.2014.03.038 pmid: 24709520
168 Y, Lee S H, Lee J S, Kim , et al.. Controlled synthesis of PEI-coated gold nanoparticles using reductive catechol chemistry for siRNA delivery. Journal of Controlled Release, 2011, 155( 1): 3– 10
https://doi.org/10.1016/j.jconrel.2010.09.009 pmid: 20869409
169 V, Cebrián F, Martín-Saavedra C, Yagüe , et al.. Size-dependent transfection efficiency of PEI-coated gold nanoparticles. Acta Biomaterialia, 2011, 7( 10): 3645– 3655
https://doi.org/10.1016/j.actbio.2011.06.018 pmid: 21704738
170 L, Zhang Z, Lu Q, Zhao , et al.. Enhanced chemotherapy efficacy by sequential delivery of siRNA and anticancer drugs using PEI-grafted graphene oxide. Small, 2011, 7( 4): 460– 464
https://doi.org/10.1002/smll.201001522 pmid: 21360803
171 J, Sheng L, Han J, Qin , et al.. N-trimethyl chitosan chloride-coated PLGA nanoparticles overcoming multiple barriers to oral insulin absorption. ACS Applied Materials & Interfaces, 2015, 7( 28): 15430– 15441
https://doi.org/10.1021/acsami.5b03555 pmid: 26111015
172 S, Şenel S J McClure . Potential applications of chitosan in veterinary medicine. Advanced Drug Delivery Reviews, 2004, 56( 10): 1467– 1480
https://doi.org/10.1016/j.addr.2004.02.007 pmid: 15191793
173 T, Kean M Thanou . Biodegradation, biodistribution and toxicity of chitosan. Advanced Drug Delivery Reviews, 2010, 62( 1): 3– 11
https://doi.org/10.1016/j.addr.2009.09.004 pmid: 19800377
174 J, Lin Y, Li Y, Li , et al.. Drug/dye-loaded, multifunctional PEG–chitosan–iron oxide nanocomposites for methotraxate synergistically self-targeted cancer therapy and dual model imaging. ACS Applied Materials & Interfaces, 2015, 7( 22): 11908– 11920
https://doi.org/10.1021/acsami.5b01685 pmid: 25978458
175 S, Mao W, Sun T Kissel . Chitosan-based formulations for delivery of DNA and siRNA. Advanced Drug Delivery Reviews, 2010, 62( 1): 12– 27
https://doi.org/10.1016/j.addr.2009.08.004 pmid: 19796660
176 M K, Gurka D, Pender P, Chuong , et al.. Identification of pancreatic tumors in vivo with ligand-targeted, pH responsive mesoporous silica nanoparticles by multispectral optoacoustic tomography. Journal of Controlled Release, 2016, 231: 60– 67
https://doi.org/10.1016/j.jconrel.2015.12.055 pmid: 26763377
177 C, Murugan K, Rayappan R, Thangam , et al.. Combinatorial nanocarrier based drug delivery approach for amalgamation of anti-tumor agents in breast cancer cells: an improved nanomedicine strategy. Scientific Reports, 2016, 6: 34053
https://doi.org/10.1038/srep34053
178 T, Liao C, Liu J, Ren , et al.. A chitosan/mesoporous silica nanoparticle-based anticancer drug delivery system with a “tumor-triggered targeting” property. International Journal of Biological Macromolecules, 2021, 183: 2017– 2029
https://doi.org/10.1016/j.ijbiomac.2021.06.004 pmid: 34097958
179 Q, Yan X, Chen H, Gong , et al.. Delivery of a TNF-α-derived peptide by nanoparticles enhances its antitumor activity by inducing cell-cycle arrest and caspase-dependent apoptosis. FASEB Journal, 2018, 32( 12): 6948– 6964
https://doi.org/10.1096/fj.201800377R pmid: 30161002
180 A, Jayasree S, Sasidharan M, Koyakutty , et al.. Mannosylated chitosan-zinc sulphide nanocrystals as fluorescent bioprobes for targeted cancer imaging. Carbohydrate Polymers, 2011, 85( 1): 37– 43
https://doi.org/10.1016/j.carbpol.2011.01.034
181 P, Manivasagan V T, Nguyen S W, Jun , et al.. Anti-EGFR antibody conjugated thiol chitosan-layered gold nanoshells for dual-modal imaging-guided cancer combination therapy. Journal of Controlled Release, 2019, 311–312: 26– 42
https://doi.org/10.1016/j.jconrel.2019.08.007 pmid: 31401198
182 P, Li Y, Yan H, Zhang , et al.. Treatment of cervical cancer by siRNA-loaded chitosan-coated calcium phosphate nanoparticles. Journal of Chinese Pharmaceutical Sciences, 2018, 27( 8): 517– 529
https://doi.org/10.5246/jcps.2018.08.053
183 K, Roy R K, Kanwar J R Kanwar . LNA aptamer based multi-modal, Fe3O4-saturated lactoferrin (Fe3O4-bLf) nanocarriers for triple positive (EpCAM, CD133, CD44) colon tumor targeting and NIR, MRI and CT imaging. Biomaterials, 2015, 71: 84– 99
https://doi.org/10.1016/j.biomaterials.2015.07.055 pmid: 26318819
184 C, Lonez M, Vandenbranden J M Ruysschaert . Cationic liposomal lipids: from gene carriers to cell signaling. Progress in Lipid Research, 2008, 47( 5): 340– 347
https://doi.org/10.1016/j.plipres.2008.03.002 pmid: 18424270
185 N J Caplen . Nucleic acid transfer using cationic lipids. Methods in Molecular Biology, 2000, 133: 1– 19
https://doi.org/10.1385/1-59259-215-5:1
186 N, Zhu D, Liggitt Y, Liu , et al.. Systemic gene expression after intravenous DNA delivery into adult mice. Science, 1993, 261( 5118): 209– 211
https://doi.org/10.1126/science.7687073 pmid: 7687073
187 L H, Lindner R, Brock D, Arndt-Jovin , et al.. Structural variation of cationic lipids: minimum requirement for improved oligonucleotide delivery into cells. Journal of Controlled Release, 2006, 110( 2): 444– 456
https://doi.org/10.1016/j.jconrel.2005.10.009 pmid: 16297484
188 G, Pillai A, Cox L Yuen . The science and technology of cancer theranostic nanomedicines: a primer for clinicians and pharmacists. SOJ Pharmacy and Pharmaceutical Sciences, 2018, 5( 2): 1– 7
https://doi.org/10.15226/2374-6866/5/2/00178
189 S R, Mudshinge A B, Deore S, Patil , et al.. Nanoparticles: emerging carriers for drug delivery. Saudi Pharmaceutical Journal, 2011, 19( 3): 129– 141
https://doi.org/10.1016/j.jsps.2011.04.001 pmid: 23960751
190 W T, Al-Jamal K T, Al-Jamal B, Tian , et al.. Lipid-quantum dot bilayer vesicles enhance tumor cell uptake and retention in vitro and in vivo. ACS Nano, 2008, 2( 3): 408– 418
https://doi.org/10.1021/nn700176a pmid: 19206564
191 S J, Leung M Romanowski . Light-activated content release from liposomes. Theranostics, 2012, 2( 10): 1020– 1036
https://doi.org/10.7150/thno.4847 pmid: 23139729
192 V P Torchilin . Recent advances with liposomes as pharmaceutical carriers. Nature Reviews. Drug Discovery, 2005, 4( 2): 145– 160
https://doi.org/10.1038/nrd1632 pmid: 15688077
193 Y, Nie L, Ji H, Ding , et al.. Cholesterol derivatives based charged liposomes for doxorubicin delivery: preparation, in vitro and in vivo characterization. Theranostics, 2012, 2( 11): 1092– 1103
https://doi.org/10.7150/thno.4949 pmid: 23227125
194 D R, Sørensen M, Leirdal M Sioud . Gene silencing by systemic delivery of synthetic siRNAs in adult mice. Journal of Molecular Biology, 2003, 327( 4): 761– 766
https://doi.org/10.1016/S0022-2836(03)00181-5 pmid: 12654261
195 S, Zhang B, Zhao H, Jiang , et al.. Cationic lipids and polymers mediated vectors for delivery of siRNA. Journal of Controlled Release, 2007, 123( 1): 1– 10
https://doi.org/10.1016/j.jconrel.2007.07.016 pmid: 17716771
196 W, Tao X, Mao J P, Davide , et al.. Mechanistically probing lipid-siRNA nanoparticle-associated toxicities identifies Jak inhibitors effective in mitigating multifaceted toxic responses. Molecular Therapy, 2011, 19( 3): 567– 575
https://doi.org/10.1038/mt.2010.282 pmid: 21179008
197 Y, Yang J, Li F, Liu , et al.. Systemic delivery of siRNA via LCP nanoparticle efficiently inhibits lung metastasis. Molecular Therapy, 2012, 20( 3): 609– 615
https://doi.org/10.1038/mt.2011.270 pmid: 22186791
198 N, Reinhardt L, Adumeau O, Lambert , et al.. Quaternary ammonium groups exposed at the surface of silica nanoparticles suitable for DNA complexation in the presence of cationic lipids. The Journal of Physical Chemistry B, 2015, 119( 21): 6401– 6411
https://doi.org/10.1021/acs.jpcb.5b01834 pmid: 25950202
199 W T, Al-Jamal K T, Al-Jamal A, Cakebread , et al.. Blood circulation and tissue biodistribution of lipid-quantum dot (L-QD) hybrid vesicles intravenously administered in mice. Bioconjugate Chemistry, 2009, 20( 9): 1696– 1702
https://doi.org/10.1021/bc900047n pmid: 19655725
200 W T, Al-Jamal K T, Al-Jamal B, Tian , et al.. Lipid-quantum dot bilayer vesicles enhance tumor cell uptake and retention in vitro and in vivo. ACS Nano, 2008, 2( 3): 408– 418
https://doi.org/10.1021/nn700176a pmid: 19206564
201 W T, Al-Jamal K T, Al-Jamal P H, Bomans , et al.. Functionalized-quantum-dot-liposome hybrids as multimodal nanoparticles for cancer. Small, 2008, 4( 9): 1406– 1415
https://doi.org/10.1002/smll.200701043 pmid: 18711753
202 F, Wang Z, Chen L Zhu . cRGD-conjugated magnetic-fluorescent liposomes for targeted dual-modality imaging of bone metastasis from prostate cancer. Journal of Liposome Research, 2015, 25( 2): 89– 100
https://doi.org/10.3109/08982104.2014.928890 pmid: 24960451
203 S J, Mattingly M G, O’Toole K T, James , et al.. Magnetic nanoparticle-supported lipid bilayers for drug delivery. Langmuir, 2015, 31( 11): 3326– 3332
https://doi.org/10.1021/la504830z pmid: 25714501
204 W H, Kong K H, Bae S D, Jo , et al.. Cationic lipid-coated gold nanoparticles as efficient and non-cytotoxic intracellular siRNA delivery vehicles. Pharmaceutical Research, 2012, 29( 2): 362– 374
https://doi.org/10.1007/s11095-011-0554-y pmid: 21842305
205 A, Chakraborty J C, Boer C, Selomulya , et al.. Amino acid functionalized inorganic nanoparticles as cutting-edge therapeutic and diagnostic agents. Bioconjugate Chemistry, 2018, 29( 3): 657– 671
https://doi.org/10.1021/acs.bioconjchem.7b00455 pmid: 28876902
206 S, Biswas S H, Medina J J Jr Barchi . Synthesis and cell-selective antitumor properties of amino acid conjugated tumor-associated carbohydrate antigen-coated gold nanoparticles. Carbohydrate Research, 2015, 405: 93– 101
https://doi.org/10.1016/j.carres.2014.11.002 pmid: 25556664
207 J, Shi X, Sun X, Zou , et al.. Amino acid-dependent transformations of citrate-coated silver nanoparticles: impact on morphology, stability and toxicity. Toxicology Letters, 2014, 229( 1): 17– 24
https://doi.org/10.1016/j.toxlet.2014.06.014 pmid: 24910988
208 X, Zhu Y, Xie Y, Zhang , et al.. Thermo-sensitive liposomes loaded with doxorubicin and lysine modified single-walled carbon nanotubes as tumor-targeting drug delivery system. Journal of Biomaterials Applications, 2014, 29( 5): 769– 779
https://doi.org/10.1177/0885328214543211 pmid: 25033825
209 Y, Feng J, Su Z, Zhao , et al.. Differential effects of amino acid surface decoration on the anticancer efficacy of selenium nanoparticles. Dalton Transactions, 2014, 43( 4): 1854– 1861
https://doi.org/10.1039/C3DT52468J pmid: 24257441
210 H M, Yang H J, Lee C W, Park , et al.. Endosome-escapable magnetic poly(amino acid) nanoparticles for cancer diagnosis and therapy. Chemical Communications, 2011, 47( 18): 5322– 5324
https://doi.org/10.1039/c1cc10371g pmid: 21451847
211 L, Agemy D, Friedmann-Morvinski V R, Kotamraju , et al.. Targeted nanoparticle enhanced proapoptotic peptide as potential therapy for glioblastoma. Proceedings of the National Academy of Sciences of the United States of America, 2011, 108( 42): 17450– 17455
https://doi.org/10.1073/pnas.1114518108 pmid: 21969599
212 Z, Shen T, Liu Z, Yang , et al.. Small-sized gadolinium oxide based nanoparticles for high-efficiency theranostics of orthotopic glioblastoma. Biomaterials, 2020, 235: 119783
https://doi.org/10.1016/j.biomaterials.2020.119783 pmid: 31981762
213 O, Taratula O B, Garbuzenko A M, Chen , et al.. Innovative strategy for treatment of lung cancer: targeted nanotechnology-based inhalation co-delivery of anticancer drugs and siRNA. Journal of Drug Targeting, 2011, 19( 10): 900– 914
https://doi.org/10.3109/1061186X.2011.622404 pmid: 21981718
214 W, Fei Y, Zhang S, Han , et al.. RGD conjugated liposome-hollow silica hybrid nanovehicles for targeted and controlled delivery of arsenic trioxide against hepatic carcinoma. International Journal of Pharmaceutics, 2017, 519( 1–2): 250– 262
https://doi.org/10.1016/j.ijpharm.2017.01.031 pmid: 28109899
215 G F, Luo W H, Chen Y, Liu , et al.. Multifunctional enveloped mesoporous silica nanoparticles for subcellular co-delivery of drug and therapeutic peptide. Scientific Reports, 2014, 4: 6064
https://doi.org/10.1038/srep06064 pmid: 25317538
216 X Z, Yang J Z, Du S, Dou , et al.. Sheddable ternary nanoparticles for tumor acidity-targeted siRNA delivery. ACS Nano, 2012, 6( 1): 771– 781
https://doi.org/10.1021/nn204240b pmid: 22136582
217 K T, Jin Z B, Lu J Y, Chen , et al.. Recent trends in nanocarrier-based targeted chemotherapy: selective delivery of anticancer drugs for effective lung, colon, cervical, and breast cancer treatment. Journal of Nanomaterials, 2020, 2020: 9184284
https://doi.org/10.1155/2020/9184284
218 Y, Liu Y, Pan W, Cao , et al.. A tumor microenvironment responsive biodegradable CaCO3/MnO2-based nanoplatform for the enhanced photodynamic therapy and improved PD-L1 immunotherapy. Theranostics, 2019, 9( 23): 6867– 6884
https://doi.org/10.7150/thno.37586 pmid: 31660074
219 H, Meng M, Xue T, Xia , et al.. Use of size and a copolymer design feature to improve the biodistribution and the enhanced permeability and retention effect of doxorubicin-loaded mesoporous silica nanoparticles in a murine xenograft tumor model. ACS Nano, 2011, 5( 5): 4131– 4144
https://doi.org/10.1021/nn200809t pmid: 21524062
220 J, Lu M, Liong Z, Li , et al.. Biocompatibility, biodistribution, and drug-delivery efficiency of mesoporous silica nanoparticles for cancer therapy in animals. Small, 2010, 6( 16): 1794– 1805
https://doi.org/10.1002/smll.201000538 pmid: 20623530
221 T, Chen T, Zhao D, Wei , et al.. Core–shell nanocarriers with ZnO quantum dots-conjugated Au nanoparticle for tumor-targeted drug delivery. Carbohydrate Polymers, 2013, 92( 2): 1124– 1132
https://doi.org/10.1016/j.carbpol.2012.10.022 pmid: 23399137
222 H, Sharma K, Kumar C, Choudhary , et al.. Development and characterization of metal oxide nanoparticles for the delivery of anticancer drug. Artificial Cells, Nanomedicine, and Biotechnology, 2016, 44( 2): 672– 679
https://doi.org/10.3109/21691401.2014.978980 pmid: 25406734
223 S, Senapati R, Thakur S P, Verma , et al.. Layered double hydroxides as effective carrier for anticancer drugs and tailoring of release rate through interlayer anions. Journal of Controlled Release, 2016, 224: 186– 198
https://doi.org/10.1016/j.jconrel.2016.01.016 pmid: 26774219
224 J, Chakraborty S, Roychowdhury S, Sengupta , et al.. Mg–Al layered double hydroxide-methotrexate nanohybrid drug delivery system: evaluation of efficacy. Materials Science and Engineering C, 2013, 33( 4): 2168– 2174
https://doi.org/10.1016/j.msec.2013.01.047 pmid: 23498245
225 Y M, Kuo Y, Kuthati R K, Kankala , et al.. Layered double hydroxide nanoparticles to enhance organ-specific targeting and the anti-proliferative effect of cisplatin. Journal of Materials Chemistry B: Materials for Biology and Medicine, 2015, 3( 17): 3447– 3458
https://doi.org/10.1039/C4TB01989J pmid: 32262227
226 H, Asiabi Y, Yamini M, Alipour , et al.. Synthesis and characterization of a novel biocompatible pseudo-hexagonal NaCa-layered double metal hydroxides for smart pH-responsive drug release of dacarbazine and enhanced anticancer activity in malignant melanoma. Materials Science and Engineering C, 2019, 97: 96– 102
https://doi.org/10.1016/j.msec.2018.12.017 pmid: 30678984
227 S, Ray M, Joy B, Sa , et al.. pH dependent chemical stability and release of methotrexate from a novel nanoceramic carrier. RSC Advances, 2015, 5( 49): 39482– 39494
https://doi.org/10.1039/C5RA03546E
228 S, Ray A, Mishra T K, Mandal , et al.. Optimization of the process parameters for the fabrication of a polymer coated layered double hydroxide-methotrexate nanohybrid for the possible treatment of osteosarcoma. RSC Advances, 2015, 5( 124): 102574– 102592
https://doi.org/10.1039/C5RA15859A
229 J, Wen Y, Lv Y, Xu , et al.. Construction of a biodegradable, versatile nanocarrier for optional combination cancer therapy. Acta Biomaterialia, 2019, 83: 359– 371
https://doi.org/10.1016/j.actbio.2018.11.009 pmid: 30414486
230 J, Pi J, Jiang H, Cai , et al.. GE11 peptide conjugated selenium nanoparticles for EGFR targeted oridonin delivery to achieve enhanced anticancer efficacy by inhibiting EGFR-mediated PI3K/AKT and Ras/Raf/MEK/ERK pathways. Drug Delivery, 2017, 24( 1): 1549– 1564
https://doi.org/10.1080/10717544.2017.1386729 pmid: 29019267
231 M, Alibolandi K, Abnous F, Sadeghi , et al.. Folate receptor-targeted multimodal polymersomes for delivery of quantum dots and doxorubicin to breast adenocarcinoma: in vitro and in vivo evaluation. International Journal of Pharmaceutics, 2016, 500( 1–2): 162– 178
https://doi.org/10.1016/j.ijpharm.2016.01.040 pmid: 26802496
232 C, Xu B, Wang S Sun . Dumbbell-like Au-Fe3O4 nanoparticles for target-specific platin delivery. Journal of the American Chemical Society, 2009, 131( 12): 4216– 4217
https://doi.org/10.1021/ja900790v pmid: 19275156
233 F, Wang Y C, Wang S, Dou , et al.. Doxorubicin-tethered responsive gold nanoparticles facilitate intracellular drug delivery for overcoming multidrug resistance in cancer cells. ACS Nano, 2011, 5( 5): 3679– 3692
https://doi.org/10.1021/nn200007z pmid: 21462992
234 B, Haynes Y, Zhang F, Liu , et al.. Gold nanoparticle conjugated Rad6 inhibitor induces cell death in triple negative breast cancer cells by inducing mitochondrial dysfunction and PARP-1 hyperactivation: synthesis and characterization. Nanomedicine: Nanotechnology, Biology, and Medicine, 2016, 12( 3): 745– 757
https://doi.org/10.1016/j.nano.2015.10.010 pmid: 26563438
235 Z, Zhou C, Kennell J Y, Lee , et al.. Calcium phosphate-polymer hybrid nanoparticles for enhanced triple negative breast cancer treatment via co-delivery of paclitaxel and miR-221/222 inhibitors. Nanomedicine: Nanotechnology, Biology, and Medicine, 2017, 13( 2): 403– 410
https://doi.org/10.1016/j.nano.2016.07.016 pmid: 27520723
236 Y, Cheng A C, Samia J D, Meyers , et al.. Highly efficient drug delivery with gold nanoparticle vectors for in vivo photodynamic therapy of cancer. Journal of the American Chemical Society, 2008, 130( 32): 10643– 10647
https://doi.org/10.1021/ja801631c pmid: 18642918
237 O, Zelphati L S, Uyechi L G, Barron , et al.. Effect of serum components on the physico-chemical properties of cationic lipid/oligonucleotide complexes and on their interactions with cells. Biochimica et Biophysica Acta, 1998, 1390( 2): 119– 133
https://doi.org/10.1016/S0005-2760(97)00169-0 pmid: 9507083
238 C, Passirani J P Benoit. Complement activation by injectable colloidal drug carriers. In: Mahato R I, ed. Biomaterials for Delivery and Targeting of Proteins and Nucleic Acids. Boca Raton, FL, USA: CRC Press, 2005
239 I, Brigger C, Dubernet P Couvreur . Nanoparticles in cancer therapy and diagnosis. Advanced Drug Delivery Reviews, 2002, 54( 5): 631– 651
https://doi.org/10.1016/S0169-409X(02)00044-3
[1] Vahid ALIMARDANI, Ghazal FARAHAVAR, Sepide SALEHI, Saeed TAGHIZADEH, Moosa Rahimi GHIASI, Samira Sadat ABOLMAALI. Gold nanocages in cancer diagnosis, therapy, and theranostics: A brief review[J]. Front. Mater. Sci., 2021, 15(4): 494-511.
[2] Qingni XU, Chaohua LI, Yuqi CHEN, Yueli ZHANG, Bo LU. Metal-organic framework-based intelligent drug delivery systems for cancer theranostic: A review[J]. Front. Mater. Sci., 2021, 15(3): 374-390.
[3] Shalmali BASU, Kamalika SEN. A review on graphene-based materials as versatile cancer biomarker sensors[J]. Front. Mater. Sci., 2020, 14(4): 353-372.
[4] Jiangli XUE, Maosong MO, Zhuming LIU, Dapeng YE, Zhihua CHENG, Tong XU, Liangti QU. A general synthesis strategy for the multifunctional 3D polypyrrole foam of thin 2D nanosheets[J]. Front. Mater. Sci., 2018, 12(2): 105-117.
[5] Zhi-Qin YAN,Wei ZHANG. The development of graphene-based devices for cell biology research[J]. Front. Mater. Sci., 2014, 8(2): 107-122.
[6] Jie MA, Jian-Nong WANG, Chung-Jung TSAI, Buyong MA, Ruth NUSSINOV, . Diameters of single-walled CNTs (SWCNTs) and related nanochemistry and nanobiology[J]. Front. Mater. Sci., 2010, 4(1): 17-28.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed