Smart nanocarrier-based drug delivery systems for cancer therapy and toxicity studies: A review
Tóm tắt
Từ khóa
Tài liệu tham khảo
American Cancer Society, 2017, Cancer facts and figures 2017, Genes Dev, 21, 2525
Zhang, 2011, The application of carbon nanotubes in target drug delivery systems for cancer therapies, Nanoscale Res Lett, 6, 555, 10.1186/1556-276X-6-555
Ahmad, 2016, Anticancer chemotherapy in teenagers and young adults: managing long term side effects, BMJ, 354, i4567, 10.1136/bmj.i4567
Gillet, 2010
Alfarouk, 2015, Resistance to cancer chemotherapy: failure in drug response from ADME to P-gp, Cancer Cell Int, 15, 71, 10.1186/s12935-015-0221-1
Nooter, 1996, Molecular mechanisms of multidrug resistance in cancer chemotherapy, Pathol Res Pract, 192, 768, 10.1016/S0344-0338(96)80099-9
Gupta, 1990, Drug targeting in cancer chemotherapy: a clinical perspective, J Pharm Sci, 79, 949, 10.1002/jps.2600791102
Kreuter, 2007, Nanoparticles-a historical perspective, Int J Pharm, 331, 1, 10.1016/j.ijpharm.2006.10.021
Khanna, 1970, Bead polymerization technique for sustained-release dosage form, J Pharm Sci, 59, 614, 10.1002/jps.2600590508
Matsumura, 1986, A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs, Cancer Res, 46, 6387
Bae, 2011, Targeted drug delivery to tumors: myths, reality and possibility, J Control Release, 153, 198, 10.1016/j.jconrel.2011.06.001
Ding, 2016, Recent advances in stimuli-responsive release function drug delivery systems for tumor treatment, Molecules, 21, 1715, 10.3390/molecules21121715
Kreyling, 2010, A complementary definition of nanomaterial, Nano Today, 5, 165, 10.1016/j.nantod.2010.03.004
Peer, 2007, Nanocarriers as an emerging platform for cancer therapy, Nat Nanotechnol, 2, 751, 10.1038/nnano.2007.387
Lee, 2015, Smart nanoparticles for drug delivery: boundaries and opportunities, Chem Eng Sci, 125, 158, 10.1016/j.ces.2014.06.042
Liu, 2016, The smart drug delivery system and its clinical potential, Theranostics, 6, 1306, 10.7150/thno.14858
Abuchowski, 1977, Effect of covalent attachment of polyethylene glycol on immunogenicity and circulating life of bovine liver catalase, J Biol Chem, 252, 3582, 10.1016/S0021-9258(17)40292-4
Moghimi, 2003, Stealth liposomes and long circulating nanoparticles: critical issues in pharmacokinetics, opsonization and protein-binding properties, Prog Lipid Res, 42, 463, 10.1016/S0163-7827(03)00033-X
Moghimi, 2001, Long-circulating and target-specific nanoparticles: theory to practice, Pharmacol Rev, 53, 283
Knop, 2010, Poly(ethylene glycol) in drug delivery: pros and cons as well as potential alternatives, Angew Chemie Int Ed, 49, 6288, 10.1002/anie.200902672
Verhoef, 2013, Questioning the use of PEGylation for drug delivery, Drug Deliv Transl Res, 3, 499, 10.1007/s13346-013-0176-5
Xu, 2014, Nanocarriers in gene therapy: a review, J Biomed Nanotechnol, 10, 3483, 10.1166/jbn.2014.2044
Qi, 2017, Co-delivery nanoparticles of anti-cancer drugs for improving chemotherapy efficacy, Drug Deliv, 24, 1909, 10.1080/10717544.2017.1410256
Kang, 2015, Nanocarrier-mediated co-delivery of chemotherapeutic drugs and gene agents for cancer treatment, Acta Pharm Sin B, 5, 169, 10.1016/j.apsb.2015.03.001
Janib, 2010, Imaging and drug delivery using theranostic nanoparticles, Adv Drug Deliv Rev, 62, 1052, 10.1016/j.addr.2010.08.004
Srinivasan, 2015, Multifunctional nanomaterials and their applications in drug delivery and cancer therapy, Nanomaterials, 5, 1690, 10.3390/nano5041690
Parvanian, 2017, Multifunctional nanoparticle developments in cancer diagnosis and treatment, Sens Bio-Sensing Res, 13, 81, 10.1016/j.sbsr.2016.08.002
Bangham, 1965, The action of steroids and streptolysin S on the permeability of phospholipid structures to cations, J Mol Biol, 13, 253, 10.1016/S0022-2836(65)80094-8
Akbarzadeh, 2013, Liposome: classification, preparation, and applications, Nanoscale Res Lett, 8, 102, 10.1186/1556-276X-8-102
Sharma, 1997, Liposomes in drug delivery: progress and limitations, Int J Pharm, 154, 123, 10.1016/S0378-5173(97)00135-X
Huang, 2014, Progress involving new techniques for liposome preparation, Asian J Pharm Sci, 9, 176, 10.1016/j.ajps.2014.06.001
Carugo, 2016, Liposome production by microfluidics: potential and limiting factors, Sci Rep, 6, 25876, 10.1038/srep25876
Bangham, 1978, Properties and uses of lipid vesicles: an overview, Ann N Y Acad Sci, 308, 2, 10.1111/j.1749-6632.1978.tb22010.x
Szoka, 1978, Procedure for preparation of liposomes with large internal aqueous space and high capture by reverse-phase evaporation, Proc Natl Acad Sci U S A, 75, 4194, 10.1073/pnas.75.9.4194
Deamer, 1978, Preparation and properties of ether-injection liposomes, Ann N Y Acad Sci, 308, 250, 10.1111/j.1749-6632.1978.tb22027.x
Zumbuehl, 1981, Liposomes of controllable size in the range of 40 to 180 nm by defined dialysis of lipid/detergent mixed micelles, BBA, 640, 252, 10.1016/0005-2736(81)90550-2
Lesoin, 2011, Preparation of liposomes using the supercritical anti-solvent (SAS) process and comparison with a conventional method, J Supercrit Fluids, 57, 162, 10.1016/j.supflu.2011.01.006
Otake, 2006, Preparation of liposomes using an improved supercritical reverse phase evaporation method, Langmuir, 22, 2543, 10.1021/la051654u
Sercombe, 2015, Advances and challenges of liposome assisted drug delivery, Front Pharmacol, 6, 286, 10.3389/fphar.2015.00286
Allen, 2013, Liposomal drug delivery systems: from concept to clinical applications, Adv Drug Deliv Rev, 65, 36, 10.1016/j.addr.2012.09.037
Noble, 2014, Ligand-targeted liposome design: challenges and fundamental considerations, Trends Biotechnol, 32, 32, 10.1016/j.tibtech.2013.09.007
Sapra, 2003, Ligand-targeted liposomal anticancer drugs, Prog Lipid Res, 42, 439, 10.1016/S0163-7827(03)00032-8
Sawant, 2012, Challenges in development of targeted liposomal therapeutics, AAPS J, 14, 303, 10.1208/s12248-012-9330-0
Ruoslahti, 2012, Peptides as targeting elements and tissue penetration devices for nanoparticles, Adv Mater, 24, 3747, 10.1002/adma.201200454
Lee, 2017, Stimuli-responsive liposomes for drug delivery, Wiley Interdiscip Rev Nanomed Nanobiotechnol, 9, e1450, 10.1002/wnan.1450
Huang, 2004, Acoustically active liposomes for drug encapsulation and ultrasound-triggered release, Biochim Biophys Acta – Biomembr, 1665, 134, 10.1016/j.bbamem.2004.07.003
Jin, 2016, Microwave-triggered smart drug release from liposomes co-encapsulating doxorubicin and salt for local combined hyperthermia and chemotherapy of cancer, Bioconjug Chem, 27, 2931, 10.1021/acs.bioconjchem.6b00603
Ogihara-Umeda, 1996, Optimal radiolabeled liposomes for tumor imaging, J Nucl Med, 37, 326
Petersen, 2012, Liposome imaging agents in personalized medicine, Adv Drug Deliv Rev, 64, 1417, 10.1016/j.addr.2012.09.003
Li, 2012, Novel multifunctional theranostic liposome drug delivery system: construction, characterization, and multimodality MR, near-infrared fluorescent, and nuclear imaging, Bioconjug Chem, 23, 1322, 10.1021/bc300175d
Muthu, 2013, Theranostic liposomes for cancer diagnosis and treatment: current development and pre-clinical success, Expert Opin Drug Deliv, 10, 151, 10.1517/17425247.2013.729576
Samson, 2018, Liposomal co-delivery-based quantitative evaluation of chemosensitivity enhancement in breast cancer stem cells by knockdown of GRP78/CLU, J Liposome Res, 1, 10.1080/08982104.2017.1420081
Zununi Vahed, 2017, Liposome-based drug co-delivery systems in cancer cells, Mater Sci Eng C, 71, 1327, 10.1016/j.msec.2016.11.073
Shin, 2016, Polymeric micelle nanocarriers in cancer research, Front Chem Sci Eng, 10, 348, 10.1007/s11705-016-1582-2
Cagel, 2017, Polymeric mixed micelles as nanomedicines: achievements and perspectives, Eur J Pharm Biopharm, 113, 211, 10.1016/j.ejpb.2016.12.019
Trivedi, 2010, Nanomicellar formulations for sustained drug delivery: strategies and underlying principles, Nanomedicine, 5, 485, 10.2217/nnm.10.10
Kataoka, 2001, Block copolymer micelles for drug delivery: design, characterization and biological significance, Adv Drug Deliv Rev, 47, 113, 10.1016/S0169-409X(00)00124-1
Chen, 2017, Reverse micelle-based water-soluble nanoparticles for simultaneous bioimaging and drug delivery, Org Biomol Chem, 15, 3232, 10.1039/C7OB00169J
Tang, 2009, Shell-detachable micelles based on disulfide-linked block copolymer as potential carrier for intracellular drug delivery, Bioconjug Chem, 20, 1095, 10.1021/bc900144m
Deng, 2014, PEG-b-PCL copolymer micelles with the ability of pH-controlled negative-to-positive charge reversal for intracellular delivery of doxorubicin, Biomacromolecules, 15, 4281, 10.1021/bm501290t
Sutton, 2007, Functionalized micellar systems for cancer targeted drug delivery, Pharm Res, 24, 1029, 10.1007/s11095-006-9223-y
Letchford, 2007, A review of the formation and classification of amphiphilic block copolymer nanoparticulate structures: micelles, nanospheres, nanocapsules and polymersomes, Eur J Pharm Biopharm, 65, 259, 10.1016/j.ejpb.2006.11.009
Liu, 2004, Polymer–drug compatibility: a guide to the development of delivery systems for the anticancer agent, ellipticine, J Pharm Sci, 93, 132, 10.1002/jps.10533
Kohori, 2002, Process design for efficient and controlled drug incorporation into polymeric micelle carrier systems, J Control Release, 78, 155, 10.1016/S0168-3659(01)00492-8
Cajot, 2013, In vitro investigations of smart drug delivery systems based on redox-sensitive cross-linked micelles, Macromol Biosci, 13, 1661, 10.1002/mabi.201300250
Husseini, 2002, Investigating the mechanism of acoustically activated uptake of drugs from Pluronic micelles, BMC Cancer, 2, 20, 10.1186/1471-2407-2-20
Seo, 2015, Tumor-targeting co-delivery of drug and gene from temperature-triggered micelles, Macromol Biosci, 15, 1198, 10.1002/mabi.201500137
Blanco, 2009, Multifunctional micellar nanomedicine for cancer therapy, Exp Biol Med, 234, 123, 10.3181/0808-MR-250
Rapoport, 2007, Multifunctional nanoparticles for combining ultrasonic tumor imaging and targeted chemotherapy, JNCI J Natl Cancer Inst, 99, 1095, 10.1093/jnci/djm043
Palmerston Mendes, 2017, Dendrimers as nanocarriers for nucleic acid and drug delivery in cancer therapy, Molecules, 22, 1401, 10.3390/molecules22091401
Jackson, 1998, Visualization of dendrimer molecules by transmission electron microscopy (TEM): staining methods and cryo-TEM of vitrified solutions, Macromolecules, 31, 6259, 10.1021/ma9806155
Nanjwade, 2009, Dendrimers: emerging polymers for drug-delivery systems, Eur J Pharm Sci, 10.1016/j.ejps.2009.07.008
Majoros, 2008, New dendrimers: synthesis and characterization of POPAM-PAMAM hybrid dendrimers, Macromolecules, 41, 8372, 10.1021/ma801843a
Richardt, 2009, 81
Tomalia, 1985, A new class of polymers: starburst-dendritic macromolecules, Polym J, 17, 117, 10.1295/polymj.17.117
Hawker, 1990, Preparation of polymers with controlled molecular architecture. A new convergent approach to dendritic macromolecules, J Am Chem Soc, 112, 7638, 10.1021/ja00177a027
Buhleier, 1978, “Cascade”- and “nonskid-chain-like” syntheses of molecular cavity topologies, Synthesis (Stuttg), 1978, 155, 10.1055/s-1978-24702
Bosman, 1999, About dendrimers: structure, physical properties, and applications, Chem Rev, 99, 1665, 10.1021/cr970069y
Esfand, 2001, Poly(amidoamine) (PAMAM) dendrimers: from biomimicry to drug delivery and biomedical applications, Drug Discov Today, 6, 427, 10.1016/S1359-6446(01)01757-3
Newkome, 1985, Micelles. Part 1. Cascade molecules: a new approach to micelles. A [27]-arborol, J Org Chem, 50, 2003, 10.1021/jo00211a052
Abbasi, 2014, Dendrimers: synthesis, applications, and properties, Nanoscale Res Lett, 9, 247, 10.1186/1556-276X-9-247
Jain, 2010, Dendrimer toxicity: let’s meet the challenge, Int J Pharm, 394, 122, 10.1016/j.ijpharm.2010.04.027
Bugno, 2015, Tweaking dendrimers and dendritic nanoparticles for controlled nano-bio interactions: potential nanocarriers for improved cancer targeting, J Drug Target, 23, 642, 10.3109/1061186X.2015.1052077
Wang, 2016, Stimuli-responsive dendrimers in drug delivery, Biomater Sci, 4, 375, 10.1039/C5BM00532A
Pandita, 2014, Dendrimers in drug delivery and targeting: drug-dendrimer interactions and toxicity issues, J Pharm Bioallied Sci, 6, 139, 10.4103/0975-7406.130965
Ye, 2013, Targeted biodegradable dendritic MRI contrast agent for enhanced tumor imaging, J Control Release, 169, 239, 10.1016/j.jconrel.2013.01.034
Brühwiler, 2010, Postsynthetic functionalization of mesoporous silica, Nanoscale, 2, 887, 10.1039/c0nr00039f
Watermann, 2017, Mesoporous silica nanoparticles as drug delivery vehicles in cancer, Nanomaterials, 7, 189, 10.3390/nano7070189
Roggers, 2014, The practicality of mesoporous silica nanoparticles as drug delivery devices and progress toward this goal, AAPS PharmSciTech, 15, 1163, 10.1208/s12249-014-0142-7
Nandiyanto, 2009, Synthesis of spherical mesoporous silica nanoparticles with nanometer-size controllable pores and outer diameters, Microporous Mesoporous Mater, 120, 447, 10.1016/j.micromeso.2008.12.019
Slowing, 2008, Mesoporous silica nanoparticles as controlled release drug delivery and gene transfection carriers, Adv Drug Deliv Rev, 60, 1278, 10.1016/j.addr.2008.03.012
Asefa, 2012, Biocompatibility of mesoporous silica nanoparticles, Chem Res Toxicol, 25, 2265, 10.1021/tx300166u
Lin, 2009, Synthesis and characterization of biocompatible and size-tunable multifunctional porous silica nanoparticles, Chem Mater, 21, 3979, 10.1021/cm901259n
Popat, 2012, A pH-responsive drug delivery system based on chitosan coated mesoporous silica nanoparticles, J Mater Chem, 22, 11173, 10.1039/c2jm30501a
Tang, 2012, Mesoporous silica nanoparticles: synthesis, biocompatibility and drug delivery, Adv Mater, 24, 1504, 10.1002/adma.201104763
Yamamoto, 2014, Preparation of size-controlled monodisperse colloidal mesoporous silica nanoparticles and fabrication of colloidal crystals, Chem Mater, 26, 2927, 10.1021/cm500619p
He, 2010, The effect of PEGylation of mesoporous silica nanoparticles on nonspecific binding of serum proteins and cellular responses, Biomaterials, 31, 1085, 10.1016/j.biomaterials.2009.10.046
Paris, 2015, Polymer-grafted mesoporous silica nanoparticles as ultrasound-responsive drug carriers, ACS Nano, 9, 11023, 10.1021/acsnano.5b04378
Yanes, 2012, Development of mesoporous silica nanomaterials as a vehicle for anticancer drug delivery, Ther Deliv, 3, 389, 10.4155/tde.12.9
Nadrah, 2014, Stimulus-responsive mesoporous silica particles, J Mater Sci, 49, 481, 10.1007/s10853-013-7726-6
Song, 2016, Mesoporous silica nanoparticles for stimuli-responsive controlled drug delivery: advances, challenges, and outlook, Int J Nanomed, 12, 87, 10.2147/IJN.S117495
Hergt, 2007, Magnetism in medicine, J Appl Phys, 404, 550
Hsiao, 2017, Preparation and characterization of multifunctional mesoporous silica nanoparticles for dual magnetic resonance and fluorescence imaging in targeted cancer therapy, Microporous Mesoporous Mater, 250, 210, 10.1016/j.micromeso.2017.04.050
Conde, 2012, Noble metal nanoparticles applications in cancer, J Drug Deliv, 2012, 751075, 10.1155/2012/751075
Chithrani, 2006, Determining the size and shape dependence of gold nanoparticle uptake into mammalian cells, Nano Lett, 6, 662, 10.1021/nl052396o
Applications, 2017, Unique roles of gold nanoparticles in drug delivery, targeting and imaging applications, Molecules, 22, 1445, 10.3390/molecules22091445
Noguez, 2007, Surface plasmons on metal nanoparticles: the influence of shape and physical environment, J Phys Chem C, 111, 3806, 10.1021/jp066539m
El-Sayed, 2005, Surface plasmon resonance scattering and absorption of anti-EGFR antibody conjugated gold nanoparticles in cancer diagnostics: applications in oral cancer, Nano Lett, 5, 829, 10.1021/nl050074e
Kimling, 2006, Turkevich method for gold nanoparticle synthesis revisited, J Phys Chem B, 110, 15700, 10.1021/jp061667w
Mafune, 2002, Full physical preparation of size-selected gold nanoparticles in solution: laser ablation and Laser induced size control, J Phys Chem B, 106, 7575, 10.1021/jp020577y
Song, 2009, Biological synthesis of gold nanoparticles using Magnolia kobus and Diopyros kaki leaf extracts, Process Biochem, 44, 1133, 10.1016/j.procbio.2009.06.005
Khan, 2014, Gold nanoparticles: synthesis and applications in drug delivery, Trop J Pharm Res, 13, 1169, 10.4314/tjpr.v13i7.23
Clark, 2015, Increased brain uptake of targeted nanoparticles by adding an acid-cleavable linkage between transferrin and the nanoparticle core, Proc Natl Acad Sci U S A, 112, 12486, 10.1073/pnas.1517048112
Dreaden, 2012, Size matters: gold nanoparticles in targeted cancer drug delivery, Ther Deliv, 3, 457, 10.4155/tde.12.21
Qian, 2011, Highly efficient and controllable PEGylation of gold nanoparticles prepared by femtosecond laser ablation in water, J Phys Chem C, 115, 23293, 10.1021/jp2079567
Yang, 2005, Transferrin-mediated gold nanoparticle cellular uptake, Bioconjug Chem, 16, 494, 10.1021/bc049775d
Han, 2007, Functionalized gold nanoparticles for drug delivery, Nanomedicine, 2, 113, 10.2217/17435889.2.1.113
Dixit, 2006, Synthesis and grafting of thioctic acid-PEG-folate conjugates onto Au nanoparticles for selective targeting of folate receptor-positive tumor cells, Bioconjug Chem, 17, 603, 10.1021/bc050335b
Yao, 2016, Gold nanoparticle mediated phototherapy for cancer, J Nanomater, 2016, 1
Tian, 2016, Stimuli-responsive gold nanoparticles for cancer diagnosis and therapy, J Funct Biomater, 7, 19, 10.3390/jfb7030019
Mendes, 2017, Gold nanoparticle approach to the selective delivery of gene silencing in cancer—the case for combined delivery?, Genes (Basel), 8, 94, 10.3390/genes8030094
Tiwari, 2011, Functionalized gold nanoparticles and their biomedical applications, Nanomaterials, 1, 31, 10.3390/nano1010031
Wahajuddin, 2012, Superparamagnetic iron oxide nanoparticles: magnetic nanoplatforms as drug carriers, Int J Nanomed, 7, 3445, 10.2147/IJN.S30320
Cano, 2016, Synthesis and characterization of multifunctional superparamagnetic iron oxide nanoparticles (SPION)/C 60 nanocomposites assembled by fullerene–amine click chemistry, RSC Adv, 6, 70374, 10.1039/C6RA14047E
Kandasamy, 2015, Recent advances in superparamagnetic iron oxide nanoparticles (SPIONs) for in vitro and in vivo cancer nanotheranostics, Int J Pharm, 496, 191, 10.1016/j.ijpharm.2015.10.058
Cole, 2011, Cancer theranostics: the rise of targeted magnetic nanoparticles, Trends Biotechnol, 29, 323, 10.1016/j.tibtech.2011.03.001
Patra, 2015, Dual-responsive polymer coated superparamagnetic nanoparticle for targeted drug delivery and hyperthermia treatment, ACS Appl Mater Interfaces, 7, 9235, 10.1021/acsami.5b01786
Mok, 2013, Superparamagnetic iron oxide nanoparticle-based delivery systems for biotherapeutics, Expert Opin Drug Deliv, 10, 73, 10.1517/17425247.2013.747507
Laurent, 2014, Superparamagnetic iron oxide nanoparticles for delivery of therapeutic agents: opportunities and challenges, Expert Opin Drug Deliv, 11, 1449, 10.1517/17425247.2014.924501
Santhosh, 2013, Multifunctional superparamagnetic iron oxide nanoparticles: promising tools in cancer theranostics, Cancer Lett, 336, 8, 10.1016/j.canlet.2013.04.032
Liu, 2011, Carbon materials for drug delivery & cancer therapy, Mater Today, 14, 316, 10.1016/S1369-7021(11)70161-4
Cantoro, 2006, Catalytic chemical vapor deposition of single-wall carbon nanotubes at low temperatures, Nano Lett, 6, 1107, 10.1021/nl060068y
Eatemadi, 2014, Carbon nanotubes: properties, synthesis, purification, and medical applications, Nanoscale Res Lett, 1
Bianco, 2005, Applications of carbon nanotubes in drug delivery, Curr Opin Chem Biol, 9, 674, 10.1016/j.cbpa.2005.10.005
Li, 2017, Functionalized single-walled carbon nanotubes: cellular uptake, biodistribution and applications in drug delivery, Int J Pharm, 524, 41, 10.1016/j.ijpharm.2017.03.017
Lay, 2011, Functionalized carbon nanotubes for anticancer drug delivery, Expert Rev Med Devices, 8, 561, 10.1586/erd.11.34
Schmaljohann, 2006, Thermo- and pH-responsive polymers in drug delivery, Adv Drug Deliv Rev, 58, 1655, 10.1016/j.addr.2006.09.020
Wang, 2015, Functionalized carbon nanotubes: revolution in brain delivery, Nanomedicine, 10, 2639, 10.2217/nnm.15.114
Kafa, 2015, The interaction of carbon nanotubes with an in vitro blood-brain barrier model and mouse brain in vivo, Biomaterials, 53, 437, 10.1016/j.biomaterials.2015.02.083
Son, 2016, Carbon nanotubes as cancer therapeutic carriers and mediators, Int J Nanomed, 11, 5163, 10.2147/IJN.S112660
Seifalian, 2011, A new era of cancer treatment: carbon nanotubes as drug delivery tools, Int J Nanomed, 6, 2963, 10.2147/IJN.S16923
Chen, 2017, The advances of carbon nanotubes in cancer diagnostics and therapeutics, J Nanomater, 2017, 1
Matea, 2017, Quantum dots in imaging, drug delivery and sensor applications, Int J Nanomed, 12, 5421, 10.2147/IJN.S138624
Zrazhevskiy, 2010, Designing multifunctional quantum dots for bioimaging, detection, and drug delivery, Chem Soc Rev, 39, 4326, 10.1039/b915139g
Ghasemi, 2009, Quantum dot: magic nanoparticle for imaging, detection and targeting, Acta Biomed, 80, 156
Qi, 2008, Emerging application of quantum dots for drug delivery and therapy, Expert Opin Drug Deliv, 5, 263, 10.1517/17425247.5.3.263
Nakata, 2000, Molecular beam epitaxial growth of InAs self-assembled quantum dots with light-emission at 1.3μm, J Cryst Growth, 208, 93, 10.1016/S0022-0248(99)00466-2
Bertino, 2007, Quantum dots by ultraviolet and X-ray lithography, Nanotechnology, 18, 315603, 10.1088/0957-4484/18/31/315603
Valizadeh, 2012, Quantum dots: synthesis, bioapplications, and toxicity, Nanoscale Res Lett, 7, 480, 10.1186/1556-276X-7-480
Gui, 2014, Ratiometric and time-resolved fluorimetry from quantum dots featuring drug carriers for real-time monitoring of drug release in situ, Anal Chem, 86, 5211, 10.1021/ac501293e
Zhang, 2008, Quantum dots for cancer diagnosis and therapy: biological and clinical perspectives, Nanomedicine (Lond), 3, 83, 10.2217/17435889.3.1.83
Iannazzo, 2017, Graphene quantum dots for cancer targeted drug delivery, Int J Pharm, 518, 185, 10.1016/j.ijpharm.2016.12.060
Zhao, 2016, The research and applications of quantum dots as nano-carriers for targeted drug delivery and cancer therapy, Nanoscale Res Lett, 11, 207, 10.1186/s11671-016-1394-9
Kamal, 2012, Nanotechnology-based approaches in anticancer research, Int J Nanomed, 7, 4391, 10.2147/IJN.S33838
Senapati, 2018, Controlled drug delivery vehicles for cancer treatment and their performance, Signal Transduct Target Ther, 3, 7, 10.1038/s41392-017-0004-3
Gao, 2004, In vivo cancer targeting and imaging with semiconductor quantum dots, Nat Biotechnol, 22, 969, 10.1038/nbt994
Danhier, 2010, To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery, J Control Release, 148, 135, 10.1016/j.jconrel.2010.08.027
Mohanty, 2011, Receptor mediated tumor targeting: an emerging approach for cancer therapy, Curr Drug Deliv, 8, 45, 10.2174/156720111793663606
Matsumura, 1986, A new concept for macromolecular therapeutics in cancer-chemotherapy – mechanism of tumoritropic acumulation of proteins and the antitumor agent Smancs, Cancer Res, 46, 6387
Nakamura, 2016, Nanodrug delivery: is the enhanced permeability and retention effect sufficient for curing cancer?, Bioconjug Chem, 27, 2225, 10.1021/acs.bioconjchem.6b00437
Jain, 1987, Transport of molecules across tumor vasculature, Cancer Metastasis Rev, 6, 559, 10.1007/BF00047468
Heldin, 2004, High interstitial fluid pressure—an obstacle in cancer therapy, Nat Rev Cancer, 4, 806, 10.1038/nrc1456
Blanco, 2015, Principles of nanoparticle design for overcoming biological barriers to drug delivery, Nat Biotechnol, 33, 941, 10.1038/nbt.3330
Villiers, 2010, Analysis of the toxicity of gold nano particles on the immune system: effect on dendritic cell functions, J Nanoparticle Res, 12, 55, 10.1007/s11051-009-9692-0
Liu, 2017, Internal stimuli-responsive nanocarriers for drug delivery: design strategies and applications, Mater Sci Eng C, 71, 1267, 10.1016/j.msec.2016.11.030
Vander Heiden, 2009, Understanding the Warburg effect: the metabolic requirements of cell proliferation, Science, 324, 1029, 10.1126/science.1160809
Gerweck, 1996, Cellular pH gradient in tumor versus normal tissue: potential exploitation for the treatment of cancer, Cancer Res, 56, 1194
Vaupel, 1989, Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: a review, Cancer Res, 49, 6449
Engin, 1995, Extracellular pH distribution in human tumours, Int J Hyperthermia, 11, 211, 10.3109/02656739509022457
Nilsson, 2003, Analysis of cytosolic and lysosomal pH in apoptotic cells by flow cytometry, Methods Cell Sci, 25, 185, 10.1007/s11022-004-8228-3
Gamcsik, 2012, Glutathione levels in human tumors, Biomarkers, 17, 671, 10.3109/1354750X.2012.715672
Meng, 2009, Reduction-sensitive polymers and bioconjugates for biomedical applications, Biomaterials, 30, 2180, 10.1016/j.biomaterials.2009.01.026
Wen, 2014, Redox sensitive nanoparticles with disulfide bond linked sheddable shell for intracellular drug delivery, Med Chem (Los Angeles), 4, 748, 10.4172/2161-0444.1000225
Cheng, 2011, Glutathione-responsive nano-vehicles as a promising platform for targeted intracellular drug and gene delivery, J Control Release, 152, 2, 10.1016/j.jconrel.2011.01.030
Andresen, 2010, Enzyme-triggered nanomedicine: drug release strategies in cancer therapy, Mol Membr Biol, 27, 353, 10.3109/09687688.2010.515950
Andresen, 2005, Advanced strategies in liposomal cancer therapy: problems and prospects of active and tumor specific drug release, Prog Lipid Res, 44, 68, 10.1016/j.plipres.2004.12.001
Meers, 2001, Enzyme-activated targeting of liposomes, Adv Drug Deliv Rev, 53, 265, 10.1016/S0169-409X(01)00205-8
Yao, 2016, External-stimuli responsive systems for cancer theranostic, Asian J Pharm Sci, 11, 585, 10.1016/j.ajps.2016.06.001
Hua, 2011, The effectiveness of a magnetic nanoparticle-based delivery system for BCNU in the treatment of gliomas, Biomaterials, 32, 516, 10.1016/j.biomaterials.2010.09.065
Plassat, 2011, Anti-estrogen-loaded superparamagnetic liposomes for intracellular magnetic targeting and treatment of breast cancer tumors, Adv Funct Mater, 21, 83, 10.1002/adfm.201001450
Bringas, 2012, Triggered release in lipid bilayer-capped mesoporous silica nanoparticles containing SPION using an alternating magnetic field, Chem Commun, 48, 5647, 10.1039/c2cc31563g
Hu, 2008, Core/single-crystal-shell nanospheres for controlled drug release via a magnetically triggered rupturing mechanism, Adv Mater, 20, 2690, 10.1002/adma.200800193
Hu, 2012, Multifunctional nanocapsules for simultaneous encapsulation of hydrophilic and hydrophobic compounds and on-demand release, ACS Nano, 6, 2558, 10.1021/nn205023w
Shao, 2011, The application of thermosensitive nanocarriers in controlled drug delivery, J Nanomater, 2011, 1, 10.1155/2011/389640
Kost, 2012, Responsive polymeric delivery systems, Adv Drug Deliv Rev, 64, 327, 10.1016/j.addr.2012.09.014
Yokoyama, 1996, Targetable drug carriers: present status and a future perspective, Adv Drug Deliv Rev, 21, 77, 10.1016/S0169-409X(96)00439-5
Topp, 1997, Thermosensitive micelle-forming block copolymers of poly(ethylene glycol) and poly(N-isopropylacrylamide), Macromolecules, 30, 8518, 10.1021/ma9710803
Klouda, 2008, Thermoresponsive hydrogels in biomedical applications, Eur J Pharm Biopharm, 68, 34, 10.1016/j.ejpb.2007.02.025
Lu, 2008, Light-activated nanoimpeller-controlled drug release in cancer cells, Small, 4, 421, 10.1002/smll.200700903
Yuan, 2012, Detection of serum human epididymis secretory protein 4 in patients with ovarian cancer using a label-free biosensor based on localized surface plasmon resonance, Int J Nanomed, 7, 2921, 10.2147/IJN.S32641
Yan, 2012, Functional mesoporous silica nanoparticles for photothermal-controlled drug delivery in vivo, Angew Chemie Int Ed, 51, 8373, 10.1002/anie.201203993
Yang, 2016, Near-infrared-light responsive nanoscale drug delivery systems for cancer treatment, Coord Chem Rev, 320–321, 100, 10.1016/j.ccr.2016.04.004
Rapoport, 2009, Controlled and targeted tumor chemotherapy by ultrasound-activated nanoemulsions/microbubbles, J Control Release, 138, 268, 10.1016/j.jconrel.2009.05.026
Dromi, 2007, Pulsed-high intensity focused ultrasound and low temperature-sensitive liposomes for enhanced targeted drug delivery and antitumor effect, Clin Cancer Res, 13, 2722, 10.1158/1078-0432.CCR-06-2443
Schroeder, 2009, Ultrasound, liposomes, and drug delivery: principles for using ultrasound to control the release of drugs from liposomes, Chem Phys Lipids, 162, 1, 10.1016/j.chemphyslip.2009.08.003
Geers, 2012, Crucial factors and emerging concepts in ultrasound-triggered drug delivery, J Control Release, 164, 248, 10.1016/j.jconrel.2012.08.014
Ge, 2012, Drug release from electric-field-responsive nanoparticles, ACS Nano, 6, 227, 10.1021/nn203430m
George, 2006, Electrically controlled drug delivery from biotin-doped conductive polypyrrole, Adv Mater, 18, 577, 10.1002/adma.200501242
Im, 2010, The effect of carbon nanotubes on drug delivery in an electro-sensitive transdermal drug delivery system, Biomaterials, 31, 1414, 10.1016/j.biomaterials.2009.11.004
Murdan, 2003, Electro-responsive drug delivery from hydrogels, J Control Release, 92, 1, 10.1016/S0168-3659(03)00303-1
Abidian, 2006, Conducting-polymer nanotubes for controlled drug release, Adv Mater, 18, 405, 10.1002/adma.200501726
Panzner, 1979, Control of in vitro cytotoxicity of positively charged liposomes, J Cancer Res Clin Oncol, 95, 29, 10.1007/BF00411106
Parnham, 1993, Toxicity screening of liposomes, Chem Phys Lipids, 64, 263, 10.1016/0009-3084(93)90070-J
Knudsen, 2015, In vivo toxicity of cationic micelles and liposomes, Nanomed Nanotechnol Biol Med, 11, 467, 10.1016/j.nano.2014.08.004
Filion, 1997, Toxicity and immunomodulatory activity of liposomal vectors formulated with cationic lipids toward immune effector cells, BBA, 1329, 345, 10.1016/S0005-2736(97)00126-0
Szebeni, 2009, Liposome triggering of innate immune responses: a perspective on benefits and adverse reactions, J Liposome Res, 19, 85, 10.1080/08982100902792855
Haber, 2010, Route of administration-dependent anti-inflammatory effect of liposomal alendronate, J Control Release, 148, 226, 10.1016/j.jconrel.2010.08.030
Goldsmith, 2011, Grand challenges in modulating the immune response with RNAi nanomedicines, Nanomedicine, 6, 1771, 10.2217/nnm.11.162
Dokka, 2000, Oxygen radical-mediated pulmonary toxicity induced by some cationic liposomes, Pharm Res, 17, 521, 10.1023/A:1007504613351
Mozafari, 2007, Cytotoxicity evaluation of anionic nanoliposomes and nanolipoplexes prepared by the heating method without employing volatile solvents and detergents, Pharmazie, 62, 205
Landesman-Milo, 2012, Altering the immune response with lipid-based nanoparticles, J Control Release, 161, 600, 10.1016/j.jconrel.2011.12.034
Roursgaard, 2016, In vitro toxicity of cationic micelles and liposomes in cultured human hepatocyte (HepG2) and lung epithelial (A549) cell lines, Toxicol In Vitro, 36, 164, 10.1016/j.tiv.2016.08.002
Kawaguchi, 2009, Histological study on side effects and tumor targeting of a block copolymer micelle on rats, J Control Release, 136, 240, 10.1016/j.jconrel.2009.02.011
Liu, 2017, Evaluation of in vitro toxicity of polymeric micelles to human endothelial cells under different conditions, Chem Biol Interact, 263, 46, 10.1016/j.cbi.2016.12.014
Kumar, 2012, In vitro evaluation of theranostic polymeric micelles for imaging and drug delivery in cancer, Theranostics, 2, 714, 10.7150/thno.3927
Gupta, 2015, Polymeric micelles and nanoemulsions as drug carriers: therapeutic efficacy, toxicity, and drug resistance, J Control Release, 212, 70, 10.1016/j.jconrel.2015.06.019
Li, 2009, Self-assembled polymeric micellar nanoparticles as nanocarriers for poorly soluble anticancer drug ethaselen, Nanoscale Res Lett, 4, 1502, 10.1007/s11671-009-9427-2
Knudsen, 2013, Differential toxicological response to positively and negatively charged nanoparticles in the rat brain, Nanotoxicology, 8, 1, 10.3109/17435390.2013.829589
Chung, 2015, In vivo biodistribution and clearance of peptide amphiphile micelles, Nanomed Nanotechnol Biol Med, 11, 479, 10.1016/j.nano.2014.08.006
Leroueil, 2008, Wide varieties of cationic nanoparticles induce defects in supported lipid bilayers, Nano Lett, 8, 420, 10.1021/nl0722929
Asthana, 2005, Poly(amidoamine) (PAMAM) dendritic nanostructures for controlled sitespecific delivery of acidic anti-inflammatory active ingredient, AAPS PharmSciTech, 6, E536, 10.1208/pt060367
Agrawal, 2007, Glycoconjugated peptide dendrimers-based nanoparticulate system for the delivery of chloroquine phosphate, Biomaterials, 28, 3349, 10.1016/j.biomaterials.2007.04.004
Ziemba, 2011, In vivo toxicity of poly(propyleneimine) dendrimers, J Biomed Mater Res Part A, 99A, 261, 10.1002/jbm.a.33196
Sadekar, 2012, Transepithelial transport and toxicity of PAMAM dendrimers: implications for oral drug delivery, Adv Drug Deliv Rev, 64, 571, 10.1016/j.addr.2011.09.010
Pereira, 2011, In vivo biodistribution of carboxymethylchitosan/poly(amidoamine) dendrimer nanoparticles in rats, J Bioact Compat Polym, 26, 619, 10.1177/0883911511425567
Albertazzi, 2013, In vivo distribution and toxicity of PAMAM dendrimers in the central nervous system depend on their surface chemistry, Mol Pharm, 10, 249, 10.1021/mp300391v
Naha, 2018, Toxicology of engineered nanoparticles: focus on poly(amidoamine) dendrimers, Int J Environ Res Public Health, 15, 338, 10.3390/ijerph15020338
Padilla De Jesús, 2002, Polyester dendritic systems for drug delivery applications: in vitro and in vivo evaluation, Bioconjug Chem, 13, 453, 10.1021/bc010103m
Di Pasqua, 2008, Cytotoxicity of mesoporous silica nanomaterials, J Inorg Biochem, 102, 1416, 10.1016/j.jinorgbio.2007.12.028
Napierska, 2009, Size-dependent cytotoxicity of monodisperse silica nanoparticles in human endothelial cells, Small, 5, 846, 10.1002/smll.200800461
Ye, 2010, In vitro toxicity of silica nanoparticles in myocardial cells, Environ Toxicol Pharmacol, 29, 131, 10.1016/j.etap.2009.12.002
Heidegger, 2016, Immune response to functionalized mesoporous silica nanoparticles for targeted drug delivery, Nanoscale, 8, 938, 10.1039/C5NR06122A
Bibi, 2012, Trigger release liposome systems: local and remote controlled delivery?, J Microencapsul, 29, 262, 10.3109/02652048.2011.646330
So, 2008, Effect of micro/nano silica particle feeding for mice, J Nanosci Nanotechnol, 8, 5367, 10.1166/jnn.2008.1347
He, 2011, Mesoporous silica nanoparticle based nano drug delivery systems: synthesis, controlled drug release and delivery, pharmacokinetics and biocompatibility, J Mater Chem, 21, 5845, 10.1039/c0jm03851b
Ivanov, 2012, In vivo toxicity of intravenously administered silica and silicon nanoparticles, Materials (Basel), 5, 1873, 10.3390/ma5101873
Pan, 2007, Size-dependent cytotoxicity of gold nanoparticles, Small, 3, 1941, 10.1002/smll.200700378
Connor, 2005, Gold nanoparticles are taken up by human cells but do not cause acute cytotoxicity, Small, 1, 325, 10.1002/smll.200400093
Shukla, 2005, Biocompatibility of gold nanoparticles and their endocytotic fate inside the cellular compartment: a microscopic overview, Langmuir, 21, 10644, 10.1021/la0513712
Goodman, 2004, Toxicity of gold nanoparticles functionalized with cationic and anionic side chains, Bioconjug Chem, 15, 897, 10.1021/bc049951i
Alkilany, 2010, Toxicity and cellular uptake of gold nanoparticles: what we have learned so far?, J Nanoparticle Res, 12, 2313, 10.1007/s11051-010-9911-8
Chen, 2009, Assessment of the in vivo toxicity of gold nanoparticles, Nanoscale Res Lett, 4, 858, 10.1007/s11671-009-9334-6
De Jong, 2008, Particle size-dependent organ distribution of gold nanoparticles after intravenous administration, Biomaterials, 29, 1912, 10.1016/j.biomaterials.2007.12.037
Zhang, 2009, Influence of anchoring ligands and particle size on the colloidal stability and in vivo biodistribution of polyethylene glycol-coated gold nanoparticles in tumor-xenografted mice, Biomaterials, 30, 1928, 10.1016/j.biomaterials.2008.12.038
Balasubramanian, 2010, Biodistribution of gold nanoparticles and gene expression changes in the liver and spleen after intravenous administration in rats, Biomaterials, 31, 2034, 10.1016/j.biomaterials.2009.11.079
Jia, 2017, The in vitro and in vivo toxicity of gold nanoparticles, Chin Chem Lett, 28, 691, 10.1016/j.cclet.2017.01.021
Fratoddi, 2015, How toxic are gold nanoparticles? The state-of-the-art, Nano Res, 8, 1771, 10.1007/s12274-014-0697-3
Singh, 2010, Potential toxicity of superparamagnetic iron oxide nanoparticles (SPION), Nano Rev, 1, 5358, 10.3402/nano.v1i0.5358
Edge, 2016, Pharmacokinetics and bio-distribution of novel super paramagnetic iron oxide nanoparticles (SPIONs) in the anaesthetized pig, Clin Exp Pharmacol Physiol, 43, 319, 10.1111/1440-1681.12533
Jarockyte, 2016, Accumulation and Toxicity of superparamagnetic iron oxide nanoparticles in cells and experimental animals, Int J Mol Sci, 17, 1193, 10.3390/ijms17081193
Patil, 2018, Comprehensive cytotoxicity studies of superparamagnetic iron oxide nanoparticles, Biochem Biophys Reports, 13, 63, 10.1016/j.bbrep.2017.12.002
Wei, 2016, Iron overload by superparamagnetic iron oxide nanoparticles is a high risk factor in cirrhosis by a systems toxicology assessment, Sci Rep, 6, 29110, 10.1038/srep29110
Shi Kam, 2004, Nanotube molecular transporters: internalization of carbon nanotube−protein conjugates into mammalian cells, J Am Chem Soc, 126, 6850, 10.1021/ja0486059
Sayes, 2006, Functionalization density dependence of single-walled carbon nanotubes cytotoxicity in vitro, Toxicol Lett, 161, 135, 10.1016/j.toxlet.2005.08.011
Gaillard, 2011, Carbon nanotube-coupled cell adhesion peptides are non-immunogenic: a promising step toward new biomedical devices, J Pept Sci, 17, 139, 10.1002/psc.1290
Jain, 2011, Toxicity of multiwalled carbon nanotubes with end defects critically depends on their functionalization density, Chem Res Toxicol, 24, 2028, 10.1021/tx2003728
Allegri, 2016, Toxicity determinants of multi-walled carbon nanotubes: the relationship between functionalization and agglomeration, Toxicol Rep, 3, 230, 10.1016/j.toxrep.2016.01.011
Hatami, 2017, Toxicity assessment of multi-walled carbon nanotubes on Cucurbita pepo L. under well-watered and water-stressed conditions, Ecotoxicol Environ Saf, 142, 274, 10.1016/j.ecoenv.2017.04.018
Fujita, 2015, Size effects of single-walled carbon nanotubes on in vivo and in vitro pulmonary toxicity, Inhal Toxicol, 27, 207, 10.3109/08958378.2015.1026620
Hardman, 2006, A toxicologic review of quantum dots: toxicity depends on physicochemical and environmental factors, Environ Health Perspect, 114, 165, 10.1289/ehp.8284
Pelley, 2009, State of academic knowledge on toxicity and biological fate of quantum dots, Toxicol Sci, 112, 276, 10.1093/toxsci/kfp188
Wang, 2016, Immunotoxicity assessment of CdSe/ZnS quantum dots in macrophages, lymphocytes and BALB/c mice, J Nanobiotechnol, 14, 10, 10.1186/s12951-016-0162-4
Wu, 2014, Toxicity of quantum dots on respiratory system, Inhal Toxicol, 26, 128, 10.3109/08958378.2013.871762
Salykina YF, Zherdeva VV, Dezhurov SV, Wakstein MS, Shirmanova MV, Zagaynova EV, et al. Biodistribution and clearance of quantum dots in small animals. In: Tuchin VV, Genina EA, editors. 2010. p. 799908.
Libralato, 2017, Toxicity effects of functionalized quantum dots, gold and polystyrene nanoparticles on target aquatic biological models: a review, Molecules, 22, 1439, 10.3390/molecules22091439
Manshian, 2016, Evaluation of quantum dot cytotoxicity: interpretation of nanoparticle concentrations versus intracellular nanoparticle numbers, Nanotoxicology, 10, 1318, 10.1080/17435390.2016.1210691
Zhao, 2017, A comparison between sphere and rod nanoparticles regarding their in vivo biological behavior and pharmacokinetics, Sci Rep, 7, 4131, 10.1038/s41598-017-03834-2
Bednarski, 2015, The influence of the route of administration of gold nanoparticles on their tissue distribution and basic biochemical parameters: in vivo studies, Pharmacol Rep, 67, 405, 10.1016/j.pharep.2014.10.019
Naqvi, 2010, Concentration-dependent toxicity of iron oxide nanoparticles mediated by increased oxidative stress, Int J Nanomed, 5, 983, 10.2147/IJN.S13244
Bahadar, 2016, Toxicity of nanoparticles and an overview of current experimental models, Iran Biomed J, 20, 1
Zhang, 2017, Shape dependent cytotoxicity of PLGA-PEG nanoparticles on human cells, Sci Rep, 7, 1
Wang, 2008, Challenge in understanding size and shape dependent toxicity of gold nanomaterials in human skin keratinocytes, Chem Phys Lett, 463, 145, 10.1016/j.cplett.2008.08.039
Kermanizadeh, 2017, Hepatic toxicity assessment of cationic liposome exposure in healthy and chronic alcohol fed mice, Heliyon, 3, e00458, 10.1016/j.heliyon.2017.e00458
Ferrari, 2005, Cancer nanotechnology: opportunities and challenges, Nat Rev Cancer, 5, 161, 10.1038/nrc1566
Shi, 2017, Cancer nanomedicine: progress, challenges and opportunities, Nat Rev Cancer, 17, 20, 10.1038/nrc.2016.108
Tsoi, 2016, Mechanism of hard-nanomaterial clearance by the liver, Nat Mater, 15, 1212, 10.1038/nmat4718
Oberdörster, 2010, Safety assessment for nanotechnology and nanomedicine: concepts of nanotoxicology, J Intern Med, 267, 89, 10.1111/j.1365-2796.2009.02187.x
Yang, 2017, Toxicity assessment of nanoparticles in various systems and organs, Nanotechnol Rev, 6, 279, 10.1515/ntrev-2016-0047
Huang, 2017, Nanomedicine-based combination anticancer therapy between nucleic acids and small-molecular drugs, Adv Drug Deliv Rev, 115, 82, 10.1016/j.addr.2017.06.004
Das, 2015, Gene therapies for cancer: strategies, challenges and successes, J Cell Physiol, 230, 259, 10.1002/jcp.24791
Bosetti, 2015, Cost–effectiveness of nanomedicine: the path to a future successful and dominant market?, Nanomedicine, 10, 1851, 10.2217/nnm.15.74
Hare, 2017, Challenges and strategies in anti-cancer nanomedicine development: an industry perspective, Adv Drug Deliv Rev, 108, 25, 10.1016/j.addr.2016.04.025
Ventola, 2017, Progress in nanomedicine: approved and investigational nanodrugs, P T, 42, 742
Bobo, 2016, Nanoparticle-based medicines: a review of FDA-approved materials and clinical trials to date, Pharm Res, 33, 2373, 10.1007/s11095-016-1958-5
Pillai, 2014, Nanomedicines for cancer therapy: an update of FDA approved and those under various stages of development, SOJ Pharm Pharm Sci, 1, 1