搜档网
当前位置:搜档网 › 文献翻译原文

文献翻译原文

Hindawi Publishing Corporation

Journal of Nanomaterials

Volume2010,Article ID796303,9pages

doi:10.1155/2010/796303

Review Article

Cancer Therapy Based on Nanomaterials and

Nanocarrier Systems

Weili Qiao,Bochu Wang,Yazhou Wang,Lichun Yang,Yiqiong Zhang,and Pengyu Shao

College of Bioengineering,Chongqing University,Chongqing400030,China

Correspondence should be addressed to Bochu Wang,wangbc2000@https://www.sodocs.net/doc/8e1054715.html,

Received30September2009;Revised1December2009;Accepted21January2010

Academic Editor:Chao Lin

Copyright?2010Weili Qiao et al.This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use,distribution,and reproduction in any medium,provided the original work is properly cited.

Targeted delivery of drug molecules to tumor tissue is one of the most interesting and challenging endeavors faced in pharmaceutical?eld,due to the critical and pharmacokinetically speci?c environment that exists in tumor.Over these years, cancer targeting treatment has been greatly improved by new tools and approaches based on nanotechnology.The review?rstly introduces the speci?c physical and chemical properties of a serial of nanomaterials,such as nanoparticles,micelles,dendrimers, carbon nanotubes,quantum dots,and nano?bers.It then places great emphasis on their application in the?eld of cancer therapy when they are used as nanocarrier systems.Based on the current status,the paper further discusses the unsolved problems and makes a perspective for the future prospects of the nanocarrier systems.

1.Introduction

Cancer occurs at a molecular level when multiple subsets of genes undergo genetic alterations,either activation of oncogenes or inactivation of tumor suppressor genes.Then malignant proliferation of cancer cells,tissue in?ltration, and dysfunction of organs will appear[1].Tumor tissues are characterized with active angiogenesis and high vascular density which keep blood supply for their growth,but with a defective vascular https://www.sodocs.net/doc/8e1054715.html,bined with poor lymphatic drainage,they contribute to what is known as the enhanced permeation and retention(EPR)e?ect[2,3]. Tumor genes are not stable with their development and often show genovariation.The inherent complexity of tumor microenvironment and the existence of P-glycoprotein(P-gp)usually act as barriers to traditional chemotherapy by preventing drug from reaching the tumor mass.Meanwhile, delivery of the therapeutic agents in vivo shares physiological barriers,including hepatic and renal clearance,enzymolysis and hydrolysis,as well as endosomal/lysosomal degradation [4,5].In addition,the e?ciency of anticancer drugs is limited by their unsatisfactory properties,such as poor solu-bility,narrow therapeutic window,and intensive cytotoxicity to normal tissues,which may be the causes of treatment failure in cancer[6].

Accordingly,there is a great need for new therapeutic strategies capable of delivering chemical agents and other therapeutic materials speci?cally to tumor locations[7]. With the development of nanotechnology,the integration of nanomaterials into cancer therapeutics is one of the rapidly advancing?elds.It can revolutionize the treatment of cancer.Nanotechnology is the creation and utilization of materials,devices,and systems through the control of matter on the nanometer(1billionth of a meter)scale[8]. Nanocarrier systems can be designed to interact with target cells and tissues or respond to stimuli in well-controlled ways to induce desired physiological responses.They represent new directions for more e?ective diagnosis and therapy of cancer[9].Therefore,this pape discusses the characteristics of various nanotechnology-based drug delivery systems in details,including nanoparticles,micelles,carbon nanotubes, dendrimers,quantunm dots,and nano?bers,and with more emphasis on their applications in cancer therapy.

2.Different Nanotechnology-Based Nanocarrier Systems

Based on nanotechnology,nanocarriers synthesized from organic and inorganic materials have been developed,such

as nanoparticles,micelles,carbon nanotubes,dendrimers, quantunm dots,and nano?bers[10,11](Figure1).They have shown great potential in cancer therapy by enhancing the performance of medicines and reducing systemic side e?ect in order to gain therapeutic e?ciency.

2.1.Polymeric Nanoparticles.Polymeric nanopaticles are particles of less than1μm diameter that are prepared from natural or synthetic polymers.Depending on the methods of preparation,nanoparticles can be obtained with di?erent

properties and di?erent release characteristics by forming matrix-type or reservoir-type structure,named nanospheres or nanocapsules[12].They have been considered as the promising carriers for drug delivery because they can improve the speci?city of action of drugs by changing their tissue distribution and pharmacokinetics[13].Polymeric nanoparticles have played pivotal roles in delivering antitu-mor drugs in a targeted manner to the malignant tumor cells,thereby reducing the systemic toxicity and increasing their therapeutic e?cacy.Due to the reticulo-endothelial system(RES)and the e?ect of enhanced permeation and retention(EPR),nanoparticles can be formulated for passive delivery to the lymphatic system,brain,arterial walls,lungs, liver,spleen,or made for long-term systemic circulation [2,13,14].Importantly,the critical feature of polymeric nanoparticles as drug carriers is that they are amenable to surface functionalization for active targeting to tumor tissues or cells and for stimulus-responsive controlled release of drug[15].Active targeting of nanoparticles to action sites is based on the pathological state of tumor tissues,such as the angiogenesis and the overexpressed receptors.Thus,varieties of researchers have focused on formulating multifunctional nanoparticles to improve the e?ectiveness of drug delivery and therapy[15].

The receptor-mediated endocytosis(RME)reveals the selective recognition,high-a?nity binding,and immediate internalization for the ligands at a cellular level[16].For this reason,various targeting moieties can be attached to the polymer backbone,which will act as a secondary uptake mechanism following EPR-based primary accumu-lation[3,17](Figure2).Liang et al.developed paclitaxel-loaded nanoparticles with galactosamine conjugated on for targeting to liver cancer cells.The prepared nanoparticles appeared most e?cient in reducing the size of the tumor when injected into hepatoma-tumor-bearing nude mice, through the speci?c interaction between galactosamine and asialoglycoprotein receptors[18].Thermosensitive magne-toliposomes(TMs)encapsulated with methotrexate(MTX) prepared by reverse-phase evaporation can achieve a good magnetic targeting e?ect and rapid drug release in response to hyperthermia,which implies their great potential in cancer therapy[19].

2.2.Micelles.Polymeric micelles are usually formed into core-shell structures by spontaneous assembly when its concentration is above critical micelle concentration(CMC). They have a number of unique features,including nanosize, easy manipulation of surface chemistry,core

functionalities,

Liposome

Magnetic nanoparticles

Micelle

Quantum dot

Dendrimer

Gold nanoparticles

Carbon nanotube

Figure1:Examples of nanomaterials and nanocarrier systems (adapted from[11,15

]).

Drug

Drug

Nanocarriers

Untargeted drug delivery Targeted drug delivery Figure2:Schematic of nanocarrier systems for site-targeted drug delivery(cited from[16]).

as well as ease of fabrication,making them suitable as carriers for encapsulation,and delivery of water insoluble agents[4].The micelles have a solid-like inner core,which serves as a potent nanocontainer of hydrophobic compounds for solubilization of chemotherapeutics,including docetaxel (DOC)[20],paclitaxel(PTX)[21],camptothecin[22],and dequalinium(DQA)[23].While polyionic complex(PIC) micelles and cationic polymer micelles can incorporate and protect anionic gene or protein with low rate of cellular uptake and low physiological environment stability,such as vascular endothelial growth factor(VEGF),siRNA[24],and luciferase reporter gene[25].Thanks to their hydrophilic shell,polymer micelles play an important part in escaping the recognition of RES and prolonging the blood circulation of drugs.The small size(<100nm)allows micelles for e?cient accumulation in pathological tissues with permeabilized vas-culature via the enhanced permeability and retention(EPR)

Aqueous media

T >LCST pH =5~6

T u m o r c e l l

Drug

Hydrophobic block Hydrophilic block

Figure 3:Schematic of drug release from micelles with temperature or pH trigger when they are uptaken by tumor cells (adapted from [35]).

e ?ect [26].However,the physiological factors,such as the density and heterogeneity o

f the vasculature at tumor sites,interstitial ?uid pressure,and transport of macromolecules in the tumor interstitium,are responsible for the extent of micelles extravasations.Mikhail contributed a detailed review in this perspective [27].

Stimuli-responsive polymeric micelles are often designed for controlled release of drug into tumor tissue with external stimuli trigger,like temperature,pH,ultrasound,and special enzymes [28,29](Table 1).Among these stimuli,pH and temperature are of representativeness,because the external pH of cancerous tissue tends to be lower and the temperature is higher compared to the surrounding normal tissue,which are caused by abnormal metabolism of cancer tissues [4].Lower critical solution temperature (LCST)polymers,such as poly (N-isopropylacrylamide)(PNIPAAm)with a cloud point around 32?C or some other poly (N-alkylacrylamide)compounds,were investi-gated as components of temperature-responsive copolymer micelles [29].The micelles exhibited rapid and temperature-responsive drug release in cancer cells,which was caused by the destruction of the hydrophobic-hydrophilic balance with the increase of temperature (Figure 3).Licciardi et al.synthesized novel folic acid-(FA-)functionalized diblock copolymer micelles for target delivery of antitumor drugs.The micelles could be directly targeted delivery via folic acid and rapidly release of drug could be triggered by lowering the solution pH to 5.This strategy combined the targeted delivery of therapeutics and pH-controlled drug release together,providing a tumor-selective nanocarrier for the e ?cient delivery of anticancer drugs [30].

2.3.Dendrimers.Dendrimers are arti?cial macromolecules with tree-like structures in which the atoms are arranged in many branches and subbranches radiate out from a central core [36,37].They are synthesized from branched monomer units in a stepwise manner (Figure 4).Thus it

is possible to control their molecular properties,such as size,shape,dimension,and polarity,which depend on the branched monomer units [37].These highly branched archi-tectures o ?er unique interfacial and functional performance advantages due to their empty internal cavities and surface functional groups [38].So they have an enormous capacity for solubilization of hydrophobic drugs and can be modi?ed or conjugated with various interesting guest molecules [37].Based on the speci?c properties,the dendrimers have shown great promise in the development of anticancer drug delivery systems [39].

To achieve active targeting drugs to tumor tissues,the well-de?ned multivalency of dendrimers are widely exploited for covalent attachment of special targeting moieties,such as sugar [40],folic acid [30],antibody [41],biotin [37],and epidermal growth factors [42].At the same time,ther-apeutic drugs can be encapsulated into or conjugated with dendrimers.For example,Choi and coworkers have prepared generation 5polyamidoamine (G5PAMAM)dendrimers conjugated to ?uorescein and folic acid,and then linked them together using complementary DNA oligonucleotides to produce clustered molecules for targeting cancer cells that overexpress the high-a ?nity folate receptor.In vitro studies indicated the DNA-linked dendrimer clusters could speci?cally bind to KB cells and may be used as imaging agents and therapeutics for cancer therapy [43].

2.4.Carbon Nanotubes.Carbon nanotubes are cylinders of one several coaxial graphite layers with a diameter in the order of nanometers,and they serve as instructive examples of the Janus-like properties of nanomaterials [44].They can be classi?ed into two general categories based on their structure:single-walled carbon nanotubes (SWCNTs)with a single cylindrical carbon wall and multiwalled carbon nan-otubes (MWCNTs)with multiple walls—cylinders nested within other cylinders [45](Figure 5).Thanks to their unique electronic,thermal,and structural characteristics,they can o ?er a promising approach for gene and drug delivery for cancer therapy [16,45].

Heating of organs and tissues by placing multifunctional nanomaterials at tumor sites is emerging as an art of tumor treatment by “nanothermal therapy”[46].Carbon nanotubes have become candidates to kill cancer cells via local hyperthermia,due to their thermal conductivity and optical properties.A research showed that oligonucleotides could be translocated into cell nucleus by nanotubes and cause cell death with continuous near-infrared radiation (NIR)because of excessive local heating of SWCNT in vitro [47].It can a ?ord carbon nanotubes an opportunity to be uptaken only by cancerous cells via functionalization of them with tumor-speci?c ligands and antibody,like folic acid and monoclonal antibody which can act as targeting agents for many tumors [48].Accordingly,a highly e ?ective drug delivery system triggered by pH change has been developed via ?rstly coated with a polysaccharide material and then modi?ed with folic acid [48].The encapsulated doxorubicin was only released from the modi?ed nanotubes at a low pH,resulting in nuclear DNA damage and the Hela

Table1:Stimuli that can be utilized to control the behavior and properties of micelles.

Stimuli Stimuli origin Examples References

pH Decreased pH in cancer site

caused by hypoxia and massive

cell death

Tumoral acidic extracellular pH

targeting of pH-responsive

micelles encapsulated

doxorubicin for cancer therapy

[31]

Temperature Increased temperature in

cancerous tissues owing to their

high metabolic activities.

External applied heating

Self-assembled,thermosensitive

micelles for controlled

doxorubicin delivery

[32]

Magnetic?eld Locally applied magnetic?eld Folate-encoded and

Fe3O4-loaded polymeric micelles

for dual targeting of cancer cells

[33]

Ultrasound Locally applied ultrasound Ultrasonic release of doxorubicin

from pluronic P105micelles

[28]

Redox potential

Increased concentration of

glutathione inside many

cancerous cells

Poly(ethylene glycol)-modied

thiolated gelatin nanoparticles

for glutathione-responsive

intracellular DNA delivery

[34]

3

1step

5

1step

12

1step

24

1step

Figure4:Dendrimers synthesized in a stepwise manner(cited from[38]).

cell proliferation inhibition.However,the pharmacokinetics, biodistribution,toxicity,and activity of novel functional carbon nanotubes in vivo should be further investigated to ensure their future applications[46].

2.5.Quantum Dots(QDs).Quantum dots are inorganic ?uorescent semiconductor nanoparticels composed of10–50atoms with a diameter ranging from2to10nm[11,49]. Their sizes and shapes which determine their absorption and emission properties can be controlled precisely[36].They are widely studied for optical image application in living systems and are stable for months without degradation and alteration [11].Targeted ligands have been attached to QDs in order to achieve speci?c targeting for tumor cell labeling[49].Thus, they are assured to be chosen as long-term,high-sensitivity and multicontrast imaging agents applied for the detection and diagnosis of cancer in vivo[36].

Now,many researchers focus on using quantum dots as carriers for genes delivery to overcome the obstacles of cell membranes.Klein and coworkers have developed functionalized silicon quantum dots(SiQDs)to serve as self-tracking transfection tool for ABCB1siRNA[50].Li et al.investigated glutathione-mediated release of functional plasmid DNA from positively charged CdTe quantum dots, which suggested potential applications of these QDs in selective unpacking of payload in living cells in a visible manner[51].The applicability of quantum dots for cancer therapies based on the mechanisms of photosensitization and radiosensitization has also been investigated.Possessing electronic energy levels in the range of1–5eV,quantum dots can perform as photosentizers applied in photodynamic therapy(PDT),which has recently become an approved treatment modality for some type of cancer.Due to high atom and electron density,quantum dots could absorb high-energy photons acting as radiosensitizers to cause localized and targeted damages to cancer cells,which was reviewed in details by Juzenas[52].

With the help of near-infrared(NIR)optical imaging devices,QDs-based tumor imaging and treatment could allow for application in deeper tissues and may o?er optical guide for surgery on organs.However,the toxicity of QDs cannot be ignored for their applications in vivo,because they are composed of hazardous heavy metals.Accordingly,it is necessary to investigate their toxicity systematically to ensure their security for further applications to human.

2.6.Nano?bers.Electrospinning has gained widespread interest as a potential polymer processing technique to produce ultra?ne polymer?bers for drug delivery appli-cations.It has been proven to be a relatively simple and versatile method for producing polymeric?bers with diameters ranging from tens of nanometers to microns[53].

Protein Gene Chemicals

SWCNT MWCNT

Figure 5:Structure of single-walled carbon nanotubes (SWCNTs)and multiwalled carbon nanotubes (MWCNTs),as well as drug loaded carbon nanotubes (adapted from http://www-ibmc.u-strasbg.fr/ict/vectorisation/nanotubes eng.shtml and http://www-ibmc.u-strasbg .fr/ict/vectorisation/vectorisation eng.shtml

).

solution

Nano?bers

H i g h v o l t a g e p o w e r s u p p l y

Figure 6:Schematic of electrospinning process (adapted from [56]).

Electrospinning process can be brie?y described as follows.A polymer solution is ejected from the capillary outlet of a syringe by strong enough electrostatic forces,and ?nally deposits as a nonwoven fabric mat on a grounded metal screen (counter electrode).As this jet travels through the air,the solvent evaporates,leaving behind ultra?ne polymer ?bers [54,55](Figure 6).Accordingly,the properties of nano?bers can be controlled easily by a number of processing parameters,such as applied voltage,polymer ?ow rate,capillary-collector distance,as well as the surface tension and viscoelasticity of solution [53].

As a ?brous sca ?old,nano?bers are able to entrap drugs with a high loading capacity and high encapsulation e ?-ciency because of their low weight and inherent high surface-to-volume ratio.They have been designed as promising carriers for delivering anticancer drugs,especially in post-operative local chemotherapy via surgical implantation of the sca ?old [57].Xie and Wang develop electrospun PLGA-based micro-and nano?bers as implants for the sustained

delivery of pacilitaxel to treat C6glioma in vitro.With an increased drug loading and prolonged release period,the antitumor e ?ciency was comparable to commercial pacili-taxel formulation Taxol [58].Xu et al.developed implantable BCNU-loaded poly (ethylene glycol)-poly (L-lactic acid)(PEG-PLLA)diblock copolymer ?bers for the controlled release of 1,3-bis (2-chloroethyl)-1-nitrosourea (BCNU)for postoperative chemotherapy of https://www.sodocs.net/doc/8e1054715.html,pared with pristine BCUN,the BCNU released from the ?ber retained its e ?cacy for prolonged period and increased antitumor activity against rat Glioma C6cells [59].Recently,they prepared nano?bers entrapped both paclitaxel (PTX)and doxorubicin hydrochloride (DOX)by means of “emulsion-electrospinning,”providing a new approach for multidrug delivery on combination therapy [60].Although electrospun nano?bers have been investigated greatly and show e ?ective antitumor activity as drug carriers for postoperative local chemotherapy in vitro,further researches on the destiny and antitumor activity of the nano?bers in vivo should be investigated.

2.7.Others.Current nanotechnology platforms for cancer therapeutics also include other nanomaterials.In addition to polymeric nanoparticles,gold nanoparticles (GNPs)and magnetic nanoparticles are being paid more attention because their unique physical,chemical,and biological prop-erties are quite di ?erent from the bulk of their counterparts [7].They can act as active medicine or drug additives for detection and hyperthermia treatment of cancer after laser irradiation [65,66].Nanoshells which consist of a silica core and a thin gold shell are designed to apply in cancer imaging and therapy.Because of the silica core and gold shell,the most useful nanoshells can strongly absorb NIR light and create intense heat that is lethal to cells for thermal ablation therapy [67].Recombinant adenovirus (Ad)can produce a linear,double-stranded viral genome that does not integrate with the genome of the host cell during the replication but induces a DNA double-strand break repair response in the host cell.There are many studies on the

Table2:Advantages of nanotechnology-based on drug delivery systems in cancer therapy.

Features Examples References

Solubilization e?ect Self-assembled polymeric micelles with hydrophobic core can serve as a

potent nanocontainer for increasing the solubility of hydrophobic drugs.

[20]

Protective e?ect Entrapment or conjugation of a drug to polymeric systems may protect the

drug from inactivation and help to store its activity for prolonged duration.

[61]

Passive targeting The enhanced permeability and retention(EPR)e?ect allows for the

accumulation of drug carriers in the interstitial?uid of the diseased tissue.

The drug entrapped or conjugated to the carriers can be retained in the

tumor tissue for a longer time,whereas free drug with low-molecular

weight easily di?use back out.

[62]

Active targeting Active targeting can be achieved via speci?c recognition processes,

including ligand-receptor and antibody-antigen recognition by the surface

modi?cation with various ligands,such as folate,mannose,and galactose.

[27,63,64]

Controlled release of drugs The drug can be controlled release at diseased site when an appropriate

signal is o?ered,such as pH,temperature,ultrasound,or special enzymes.

[28,29]

use of recombinant adenovirus particles as carriers for sen-sitizers to enhance noninvasive cancer therapeutics,such as radiation therapy,photodynamic therapy,and photothermal therapy.They can also be fabricated to combine multiple functions of targeting,imaging,and drug delivery together to increase the sensitivity and speci?city of cancer therapy [68].

3.Summary

Nanotechnology-based drug carriers and materials have yielded more medical bene?ts in the recent years,especially the?eld of cancer https://www.sodocs.net/doc/8e1054715.html,pared to conventional formations,nanocarrier systems have many advantages.For example,they can improve the solubility of poorly soluble drugs[20],protect the recombinant of protein and genes [61],circulate in blood stream for longer time without being recognized by macrophages,as well as controlled release of drugs at an expected rate in the desired area[28](Table2). Due to their various advantages,the nanocarrier systems and nanomaterials have demonstrated comparable or superior anticancer e?ciency to commercial formations and provided new strategies to?ght against cancer.

4.Problems and Future Perspective

The nanoscale platforms have made signi?cant progress in formulation preparations and achieved more precise treatment at a molecular level.There are already novel formations for cancer therapy available commercially,such as Abraxane,Doxil,and Embosphere,et al.[69–71],and they may o?er great opportunities for personalized medicine. Although drug delivery systems and nanomaterials are mostly investigated in preclinical animal models,too few studies are carried out to examine the cellular uptake of delivery systems in human body,because(a)the data from pre-clinical animals cannot exactly re?ect its e?ect on human body because of the complexity of tumor biology and the di?erences between animal and human physiology,(b) the development of new drug delivery systems is based on the advancement of the carrier materials,but there are only few types of materials approved by Food and Drug Administration(FDA),and(c)the security and the long-term e?ects of nanotechnology are unknown,which need research strategies to evaluate the speci?c risks when used in human body.With the development of cancer biology and polymer chemistry,new nanotechnology-based tools and therapeutic strategies will be designed and applied in the ?elds of cancer precautiondiagnosis and treatment.Besides, nanotechnology also plays an important part in delivery of drugs for ocular therapy and in generation of sca?olds for tissue engineering.However,the other major developments will have to take place for them to be permanently established not only in academia but also in industry.The basic theories of drug release from nanocarriers and pharmacokinetic models need to be developed and improved.The cytotoxicity of nanomedicine or their degradation products remains a serious problem,so more attention should be paid to improve the biocompatibility in future investigations.Their toxicity,long-term stability,and degradation pathways may also in?uence the integrity of the environment.It will not be of great bene?t to clinical application and indus-trial production of nanomedicine unless the problems are solved.

Acknowledgment

The authors would like to thank Dr.Gurinder K.Singh for her critical evaluation and helpful suggestions to the paper.

References

[1]F.H.Sarkar,S.Banerjee,and Y.W.Li,“Pancreatic can-

cer:pathogenesis,prevention and treatment,”Toxicology and Applied Pharmacology,vol.224,no.3,pp.326–336,2007. [2]J.D.Byrne,T.Betancourt,and L.Brannon-Peppas,“Active

targeting schemes for nanoparticle systems in cancer thera-peutics,”Advanced Drug Delivery Reviews,vol.60,no.15,pp.

1615–1626,2008.

[3]A.K.Iyer,G.Khaled,J.Fang,and H.Maeda,“Exploiting

the enhanced permeability and retention e?ect for tumor targeting,”Drug Discovery Today,vol.11,no.17-18,pp.812–818,2006.

[4]N.Wiradharma,Y.Zhang,S.Venkataraman,J.L.Hedrick,

and Y.Y.Yang,“Self-assembled polymer nanostructures for delivery of anticancer therapeutics,”Nano Today,vol.4,no.

4,pp.302–317,2009.

[5]L.Jabr-Milane,L.V.Vlerken,H.Devalapally,et al.,“Multi-

functional nanocarriers for targeted delivery of drugs and genes,”Journal of Controlled Release,vol.130,no.2,pp.121–128,2008.

[6]M.Pulkkinen,J.Pikkarainen,T.Wirth,et al.,“Three-

step tumor targeting of paclitaxel using biotinylated PLA-PEG nanoparticles and avidin-biotin technology:formulation development and in vitro anticancer activity,”European Journal of Pharmaceutics and Biopharmaceutics,vol.70,no.1, pp.66–74,2008.

[7]N.R.Panyala,E.M.Pe?n a-M′e ndez,and J.Havel,“Gold and

nano-gold in medicine:overview,toxicology and perspec-tives,”Journal of Applied Biomedicine,vol.7,no.2,pp.75–91, 2009.

[8]K.K.Jain,“Nanotechnology in clinical laboratory diagnos-

tics,”Clinica Chimica Acta,vol.358,no.1-2,pp.37–54,2005.

[9]G.Linazasoro,“Potential applications of nanotechnologies

to Parkinson’s disease therapy,”Parkinsonism and Related Disorders,vol.14,no.5,pp.383–392,2008.

[10]W.Cai,A.R.Hsu,Z.-B.Li,and X.Chen,“Are quantum

dots ready for in vivo imaging in human subjects?”Nanoscale Research Letters,vol.2,no.6,pp.265–281,2007.

[11]W.Cai and X.Chen,“Nanoplatforms for targeted molecular

imaging in living subjects,”Small,vol.3,no.11,pp.1840–1854,2007.

[12]A.K.Bajpai,S.K.Shukla,S.Bhanu,and S.Kankane,

“Responsive polymers in controlled drug delivery,”Progress in Polymer Science,vol.33,no.11,pp.1088–1118,2008. [13]C.Fonseca,S.Simoes,and R.Gaspar,“Paclitaxel-loaded PLGA

nanoparticles:preparation,physicochemical characterization and in vitro anti-tumoral activity,”Journal of Controlled Release,vol.83,no.2,pp.273–286,2002.

[14]M.L.Hans and A.M.Lowman,“Biodegradable nanoparticles

for drug delivery and targeting,”Current Opinion in Solid State and Materials Science,vol.6,no.4,pp.319–327,2002. [15]N.Sanvicens and M.P.Marco,“Multifunctional

nanoparticles—properties and prospects for their use in human medicine,”Trends in Biotechnology,vol.26,no.8,pp.

425–433,2008.

[16]T.Tanaka,S.Shiramoto,M.Miyashita,Y.Fujishima,and Y.

Kaneo,“Tumor targeting based on the e?ect of enhanced permeability and retention(EPR)and the mechanism of receptor-mediated endocytosis(RME),”International Journal of Pharmaceutics,vol.277,no.1-2,pp.39–61,2004.

[17]S.Sandhiya,S.A.Dkhar,and A.Surendiran,“Emerging trends

of nanomedicine—an overview,”Fundamental and Clinical Pharmacology,vol.23,no.3,pp.263–269,2009.

[18]Y.Fukumori and H.Ichikawa,“Nanoparticles for cancer

therapy and diagnosis,”Advanced Powder Technology,vol.17, no.1,pp.1–28,2006.

[19]M.Johannsen,U.Gneveckow, B.Thiesen,et al.,“Ther-

motherapy of prostate cancer using magnetic nanoparticles: feasibility,imaging,and three-dimensional temperature dis-tribution,”European Urology,vol.52,no.6,pp.1653–1662, 2007.[20]B.Liu,M.Yang,R.Li,et al.,“The antitumor e?ect of novel

docetaxel-loaded thermosensitive micelles,”European Journal of Pharmaceutics and Biopharmaceutics,vol.69,no.2,pp.527–534,2008.

[21]K.M.Huh,H.S.Min,S.C.Lee,H.J.Lee,S.Kim,and

K.Park,“A new hydrotropic block copolymer micelle system for aqueous solubilization of paclitaxel,”Journal of Controlled Release,vol.126,no.2,pp.122–129,2008.

[22]O.M.Koo,I.Rubinstein,and H.Onyuksel,“Camp-

tothecin in sterically stabilized phospholipid micelles:a novel nanomedicine,”Nanomedicine:Nanotechnology,Biology,and Medicine,vol.1,no.1,pp.77–84,2005.

[23]C.Lizano,V.Weissig,V.P.Torchilin,P.Sancho, A.I.

Garc′?a-P′e rez,and M.Pinilla,“In vivo biodistribution of ery-throcytes and polyethyleneglycol-phosphatidylethanolamine micelles carrying the antitumour agent dequalinium,”Euro-pean Journal of Pharmaceutics and Biopharmaceutics,vol.56, no.2,pp.153–157,2003.

[24]A.M.Al-Abd,S.H.Lee,S.H.Kim,et al.,“Penetration

and e?cacy of VEGF siRNA using polyelectrolyte complex micelles in a human solid tumor model in-vitro,”Journal of Controlled Release,vol.137,no.2,pp.130–135,2009. [25]N.Wiradharma,Y.W.Tong,and Y.-Y.Yang,“Self-assembled

oligopeptide nanostructures for co-delivery of drug and gene with synergistic therapeutic e?ect,”Biomaterials,vol.30,no.

17,pp.3100–3109,2009.

[26]L.B.Li and Y.B.Tan,“Preparation and properties of mixed

micelles made of Pluronic polymer and PEG-PE,”Journal of Colloid and Interface Science,vol.317,no.1,pp.326–331,2008.

[27]A.S.Mikhail and C.Allen,“Block copolymer micelles for

delivery of cancer therapy:transport at the whole body,tissue and cellular levels,”Journal of Controlled Release,vol.138,no.

3,pp.214–223,2009.

[28]G.A.Husseini,N.Y.Rapoport,D.A.Christensen,J.D.Pruitt,

and W.G.Pitt,“Kinetics of ultrasonic release of doxorubicin from pluronic P105micelles,”Colloids and Surfaces B,vol.24, no.3-4,pp.253–264,2002.

[29]N.Rapoport,“Physical stimuli-responsive polymeric micelles

for anti-cancer drug delivery,”Progress in Polymer Science,vol.

32,no.8-9,pp.962–990,2007.

[30]M.Licciardi,G.Giammona,J.Du,S.P.Armes,Y.Tang,and

A.L.Lewis,“New folate-functionalized biocompatible block

copolymer micelles as potential anti-cancer drug delivery systems,”Polymer,vol.47,no.9,pp.2946–2955,2006. [31]J.Ko,K.Park,Y.-S.Kim,et al.,“Tumoral acidic extracellular

pH targeting of pH-responsive MPEG-poly(β-amino ester) block copolymer micelles for cancer therapy,”Journal of Controlled Release,vol.123,no.2,pp.109–115,2007. [32]M.Nakayama,T.Okano,T.Miyazaki,F.Kohori,K.Sakai,and

M.Y okoyama,“Molecular design of biodegradable polymeric micelles for temperature-responsive drug release,”Journal of Controlled Release,vol.115,no.1,pp.46–56,2006.

[33]X.Yang,Y.H.Chen,R.Yuan,et al.,“Folate-encoded and

Fe3O4-loaded polymeric micelles for dual targeting of cancer cells,”Polymer,vol.49,no.16,pp.3477–3485,2008.

[34]V.Torchilin,“Multifunctional and stimuli-sensitive pharma-

ceutical nanocarriers,”European Journal of Pharmaceutics and Biopharmaceutics,vol.71,no.3,pp.431–444,2009.

[35]C.Chang,H.Wei, C.-Y.Quan,et al.,“Fabrication of

thermosensitive PCL-PNIPAAm-PCL triblock copolymeric micelles for drug delivery,”Journal of Polymer Science A,vol.

46,no.9,pp.3048–3057,2008.

[36]K.J.Morrow Jr.,R.Bawa,and C.Wei,“Recent advances in

basic and clinical nanomedicine,”Medical Clinics of North America,vol.91,no.5,pp.805–843,2007.

[37]W.J.Yang,Y.Y.Cheng,T.W.Xu,X.Y.Wang,and L.P.Wen,

“Targeting cancer cells with biotine-dendrimer conjugates,”

European Journal of Medicinal Chemistry,vol.44,no.2,pp.

862–868,2009.

[38]S.Svenson and D.A.Tomalia,“Dendrimers in biomedical

applications—re?ections on the?eld,”Advanced Drug Deliv-ery Reviews,vol.57,no.15,pp.2106–2129,2005.

[39]E.R.Gillies and J.M.J.Fr′e chet,“Dendrimers and dendritic

polymers in drug delivery,”Drug Discovery Today,vol.10,no.

1,pp.35–43,2005.

[40]D.Bhadra,A.K.Yadav,S.Bhadra,and N.K.Jain,“Glycoden-

drimeric nanoparticulate carriers of primaquine phosphate for liver targeting,”International Journal of Pharmaceutics,vol.

295,no.1-2,pp.221–233,2005.

[41]A.K.Patri,A.Myc,J.Beals,T.P.Thomas,N.H.Bander,and

J.R.Baker Jr.,“Synthesis and in vitro testing of J591antibody-dendrimer conjugates for targeted prostate cancer therapy,”

Bioconjugate Chemistry,vol.15,no.6,pp.1174–1181,2004.

[42]M.Hussain,M.Shchepinov,M.Sohail,et al.,“A novel

anionic dendrimer for improved cellular delivery of antisense oligonucleotides,”Journal of Controlled Release,vol.99,no.1, pp.139–155,2004.

[43]Y.Choi,T.Thomas,A.Kotlyar,M.T.Islam,and J.R.Baker

Jr.,“Synthesis and functional evaluation of DNA-assembled polyamidoamine dendrimer clusters for cancer cell-speci?c targeting,”Chemistry and Biology,vol.12,no.1,pp.35–43, 2005.

[44]A.A.Shvedova,E.R.Kisin,D.Porter,et al.,“Mechanisms of

pulmonary toxicity and medical applications of carbon nan-otubes:two faces of Janus?”Pharmacology and Therapeutics, vol.121,no.2,pp.192–204,2009.

[45]https://www.sodocs.net/doc/8e1054715.html,cerda,A.Bianco,M.Prato,and K.Kostarelos,“Carbon

nanotubes as nanomedicines:from toxicology to pharmacol-ogy,”Advanced Drug Delivery Reviews,vol.58,no.14,pp.

1460–1470,2006.

[46]R.Sharma and C.J.Chen,“Newer nanoparticles in hyper-

thermia treatment and thermometry,”Journal of Nanoparticle Research,vol.11,no.3,pp.671–689,2009.

[47]N.W.S.Kam,M.O’Connell,J.A.Wisdom,and H.Dai,

“Carbon nanotubes as multifunctional biological transporters and near-infrared agents for selective cancer cell destruction,”

Proceedings of the National Academy of Sciences of the United States of America,vol.102,no.33,pp.11600–11605,2005. [48]X.K.Zhang,L.J.Meng,Q.H.Lu,Z.F.Fei,and P.J.Dyson,

“Targeted delivery and controlled release of doxorubicin to cancer cells using modi?ed single wall carbon nanotubes,”

Biomaterials,vol.30,no.30,pp.6041–6047,2009.

[49]K.Y.Kim,“Nanotechnology platforms and physiological

challenges for cancer therapeutics,”Nanomedicine:Nanotech-nology,Biology,and Medicine,vol.3,no.2,pp.103–110,2007.

[50]S.Klein,O.Zolk,M.F.Fromm,F.Schr¨o dl,W.Neuhuber,and

C.Kryschi,“Functionalized silicon quantum dots tailored for

targeted siRNA delivery,”Biochemical and Biophysical Research Communications,vol.387,no.1,pp.164–168,2009.

[51]D.Li,G.P.Li,W.Guo,P.Li, E.Wang,and J.Wang,

“Glutathione-mediated release of functional plasmid DNA from positively charged quantum dots,”Biomaterials,vol.29, no.18,pp.2776–2782,2008.

[52]P.Juzenas,W.Chen,Y.-P.Sun,et al.,“Quantum dots and

nanoparticles for photodynamic and radiation therapies of

cancer,”Advanced Drug Delivery Reviews,vol.60,no.15,pp.

1600–1614,2008.

[53]M.Wang,A.J.Hsieh,and G.C.Rutledge,“Electrospinning

of poly(MMA-co-MAA)copolymers and their layered silicate nanocomposites for improved thermal properties,”Polymer, vol.46,no.10,pp.3407–3418,2005.

[54]X.L.Xu,X.S.Chen,A.X.Liu,Z.K.Hong,and X.B.Jing,

“Electrospun poly(L-lactide)-grafted hydroxyapatite/poly(L-lactide)nanocomposite?bers,”European Polymer Journal,vol.

43,no.8,pp.3187–3196,2007.

[55]M.Wang,H.Singh,T. A.Hatton,and G. C.Rutledge,

“Field-responsive superparamagnetic composite nano?bers by electrospinning,”Polymer,vol.45,no.16,pp.5505–5514, 2004.

[56]E.Jo,S.Lee,K.T.Kim,et al.,“Core-sheath nano?bers

containing colloidal arrays in the core for programmable multi-agent delivery,”Advanced Materials,vol.20,no.9,pp.

968–972,2008.

[57]X.Xu,X.Chen,P.Ma,X.Wang,and X.Jing,“The release

behavior of doxorubicin hydrochloride from medicated?bers prepared by emulsion-electrospinning,”European Journal of Pharmaceutics and Biopharmaceutics,vol.70,no.1,pp.165–170,2008.

[58]J.Xie and C.-H.Wang,“Electrospun micro-and nano?bers

for sustained delivery of paclitaxel to treat C6Glioma in vitro,”

Pharmaceutical Research,vol.23,no.8,pp.1817–1826,2006.

[59]X.Xu,X.Chen,X.Xu,et al.,“BCNU-loaded PEG-PLLA

ultra?ne?bers and their in vitro antitumor activity against Glioma C6cells,”Journal of Controlled Release,vol.114,no.

3,pp.307–316,2006.

[60]X.L.Xu,X.S.Chen,Z.F.Wang,and X.B.Jing,“Ultra?ne

PEG–PLA?bers loaded with both paclitaxel and doxorubicin hydrochloride and their in vitro cytotoxicity,”European Jour-nal of Pharmaceutics and Biopharmaceutics,vol.72,no.1,pp.

18–25,2009.

[61]Y.T.Ko, A.Kale,W. C.Hartner, B.Papahadjopoulos-

Sternberg,and V.P.Torchilin,“Self-assembling micelle-like nanoparticles based on phospholipid-polyethyleneimine conjugates for systemic gene delivery,”Journal of Controlled Release,vol.133,no.2,pp.132–138,2009.

[62]K.F.Pirollo and E.H.Chang,“Does a targeting ligand

in?uence nanoparticle tumor localization or uptake?”Trends in Biotechnology,vol.26,no.10,pp.552–558,2008.

[63]D.J.Betting,X.Y.Mu,K.Ka?,et al.,“Enhanced immune

stimulation by a therapeutic lymphoma tumor antigen vaccine produced in insect cells involves mannose receptor targeting to antigen presenting cells,”Vaccine,vol.27,no.2,pp.250–259, 2009.

[64]E.Gabano,M.Ravera,C.Cassino,S.Bonetti,G.Palmisano,

and D.Osella,“Stepwise assembly of platinum—folic acid conjugates,”Inorganica Chimica Acta,vol.361,no.5,pp.1447–1455,2008.

[65]H.F.Liang,C.T.Chen,S.C.Chen,et al.,“Paclitaxel-

loaded poly(g-glutamic acid)-poly(lactide)nanoparticles as a targeted drug delivery system for the treatment of liver cancer,”

Biomaterials,vol.27,no.9,pp.2051–2059,2006.

[66]L.Zhu,Z.Huo,L.Wang,X.Tong,Y.Xiao,and K.Ni,

“Targeted delivery of methotrexate to skeletal muscular tissue by thermosensitive magnetoliposomes,”International Journal of Pharmaceutics,vol.370,no.1-2,pp.136–143,2009. [67]K.Y.Kim,“Nanotechnology platforms and physiological

challenges for cancer therapeutics,”Nanomedicine:Nanotech-nology,Biology,and Medicine,vol.3,no.2,pp.103–110,2007.

[68]R.Singh and K.Kostarelos,“Designer adenoviruses for

nanomedicine and nanodiagnostics,”Trends in Biotechnology, vol.27,no.4,pp.220–229,2009.

[69]W.J.Gradishar,S.Tjulandin,N.Davidson,et al.,“Phase III

trial of nanoparticle albumin-bound paclitaxel compared with polyethylated castor oil-based paclitaxel in women with breast cancer,”Journal of Clinical Oncology,vol.23,no.31,pp.7794–7803,2005.

[70]T.Chidiac,G.T.Budd,R.Pelley,et al.,“Phase II trial

of liposomal doxorubicin(Doxil )in advanced soft tissue sarcomas,”Investigational New Drugs,vol.18,no.3,pp.253–259,2000.

[71]S.O.Rodiek,A.Stolzle,and C.B.Lumenta,“Preoperative

embolization of intracranial meningiomas with Embosphere microspheres,”Minimally Invasive Neurosurgery,vol.47,no.5, pp.299–305,2004.

Scientifica Corrosion Polymer Science

Volume 2014

Hindawi Publishing Corporation https://www.sodocs.net/doc/8e1054715.html, Ceramics Composites Nanoparticles Volume 2014International Journal of Biomaterials Nanoscience Textiles

Hindawi Publishing Corporation https://www.sodocs.net/doc/8e1054715.html, Volume 2014Journal of Nanotechnology Crystallography Volume 2014The Scientific World Journal Hindawi Publishing Corporation https://www.sodocs.net/doc/8e1054715.html, Volume 2014Coatings Advances in Materials Science and Engineering Hindawi Publishing Corporation https://www.sodocs.net/doc/8e1054715.html, Volume 2014 S mart Materials

Research Hindawi Publishing Corporation https://www.sodocs.net/doc/8e1054715.html,

Volume 201Metallurgy Journal of

BioMed Research International Materials Journal of Hindawi Publishing Corporation https://www.sodocs.net/doc/8e1054715.html, Volume 2014Journal of

Nanomaterials

相关主题