drug delivery applications of gold...

6
BIOTECHNOLOGY, MOLECULAR BIOLOGY AND NANOMEDICINE VOL.1 NO.1 OCTOBER 2013 ISSN: 2330-9318 (Print) ISSN: 2330-9326 (Online) http://www.researchpub.org/journal/bmbn/bmbn.html 1 AbstractDuring the last decade, due to advances in functionalization chemistry, novel nano-biomaterials with applications in the therapy of various human diseases have been developed. Nanotechnology might be crucial for drug delivery and nanoparticles (NPs) are of high importance, with many potential applications in clinical medicine and research. These novel materials with their unique physical and chemical properties are bioactive hierarchical structures that hold great hopes as support for future development of human diseases. NPs emerge as the future of drug delivery technology as they might be future crucial diagnostic and therapeutic tools. Of these, gold nanoparticles with their unique chemical and physical properties properties have emerge as promising carrier for delivery of various molecules with therapeutic properties. This paper illustrates the latest achievements in the applications of gold nanoparticles as drug delivery tools for the therapy of human diseases. Keywords Gold; Therapy; Drug delivery; Nanoparticles; Target; Release I. INTRODUCTION here has been immense interest in targeted drug delivery as it is one of the main drawbacks of pharmaceutics and biotechnology.[1] In a few words, drug delivery is the release of bioactive agents at specific rates and specific sites, but the use of these new therapeutics is limited by toxicity issues.[2] Biotechnology advances help discovering and designing many new drug classes, as it is very important to improve specific drug-delivery methods to turn them into clinical realities. Most drugs are limited by their pharmacodynamics properties as well as cytotoxicity and aggregation due to poor solubility, nonspecific delivery, in vivo short circulating half-lives.[3, 4] Drug delivery has also drew attention to biopharmaceutical companies as these new delivery nanomediated systems could represent a feasabile replacing therapy for traditional drugs thus inhibiting competition from generics.[5] These novel nanomediated drug delivery systems can yield a more effective drug accumulation in tissues and body fluids than classic ones, with minimal side effects, increasing patient quality of life with high socio-economic benefits.[3, 4, 6] Moreover, novel drug-delivery systems would offer protection and would enhance the pharmacokinetics of easily degradable peptides and proteins that often have short half-lives in vivo.[7] Thus, today, drug research aims to improve those techniques that could selectively deliver drugs to the pathological sites.[8] Nanotechnology might be crucial for drug delivery and nanoparticles (NPs) are of high importance, with many potential applications in clinical medicine and research. NPs emerge as the future of drug delivery technology as they might be future crucial diagnostic and therapeutic tools. Nanotechnology implies the design and use of materials, tools and systems through the manipulation of matter on nanometric scale that is atomic, molecular and supramolecular scales. Nanotechnology applications in multiple fields and mostly are becoming to be tested in clinical trials on humans.[9] Drug delivery nanomediated systems are based on biocompatible nanocarriers, such as gold nanoparticles, carbon nanotubes, nanovesicles, micellar systems and dendrimers.[10, 11] Additionally, one of the major benefits of nanotechnology is the targeted drug delivery at the site of the disease by passive targeting of drugs to the site of action or by selective active targeting of the active pharmaceutic agent. Nanotechnology also tries to find potential applications of nanoparticles for efficient drug delivery. Of these, gold nanoparticles with their unique chemical and physical properties properties have emerge as promising carrier for delivery of various molecules with therapeutic properties. This paper illustrates the latest achievements in the applications of gold nanoparticles as drug delivery tools for the therapy of human diseases. Peptide delivery using GNPs Various methods employing carriers for drug delivery have been developed during the past one and a half decade in order to ease the uptake of plasmids, peptides, short oligonucleotides and proteins. The discovery of the Antennapedia home domain[12] brought great enthusiasm regarding the ability of short cationic peptides to translocate cell membrane without disruption. These were called cell-penetrating peptides (CPPs).[13] Various CPPs have been detailed since then, including Tat, Sweet Arrow peptide, transportan and polyarginines, together with their controversed method of internalization.[14-16] Drug Delivery Applications of Gold Nanoparticles Lucian Mocan, MD, PhD, T

Upload: phungcong

Post on 20-Mar-2018

219 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Drug Delivery Applications of Gold Nanoparticlesresearchpub.org/journal/bmbn/number/vol1-no1/vol1-no… ·  · 2014-06-21Drug Delivery Applications of Gold ... passivating agents

BIOTECHNOLOGY, MOLECULAR BIOLOGY AND NANOMEDICINE VOL.1 NO.1 OCTOBER 2013

ISSN: 2330-9318 (Print) ISSN: 2330-9326 (Online) http://www.researchpub.org/journal/bmbn/bmbn.html

1

Abstract—During the last decade, due to advances in

functionalization chemistry, novel nano-biomaterials with

applications in the therapy of various human diseases have

been developed. Nanotechnology might be crucial for drug

delivery and nanoparticles (NPs) are of high importance,

with many potential applications in clinical medicine and

research. These novel materials with their unique physical

and chemical properties are bioactive hierarchical

structures that hold great hopes as support for future

development of human diseases. NPs emerge as the future

of drug delivery technology as they might be future crucial

diagnostic and therapeutic tools. Of these, gold

nanoparticles with their unique chemical and physical

properties properties have emerge as promising carrier for

delivery of various molecules with therapeutic properties.

This paper illustrates the latest achievements in the

applications of gold nanoparticles as drug delivery tools for

the therapy of human diseases.

Keywords — Gold; Therapy; Drug delivery; Nanoparticles;

Target; Release

I. INTRODUCTION

here has been immense interest in targeted drug delivery as

it is one of the main drawbacks of pharmaceutics and

biotechnology.[1] In a few words, drug delivery is the

release of bioactive agents at specific rates and specific sites,

but the use of these new therapeutics is limited by toxicity

issues.[2] Biotechnology advances help discovering and

designing many new drug classes, as it is very important to

improve specific drug-delivery methods to turn them into

clinical realities. Most drugs are limited by their

pharmacodynamics properties as well as cytotoxicity and

aggregation due to poor solubility, nonspecific delivery, in vivo

short circulating half-lives.[3, 4] Drug delivery has also drew

attention to biopharmaceutical companies as these new delivery

nanomediated systems could represent a feasabile replacing

therapy for traditional drugs thus inhibiting competition from

generics.[5] These novel nanomediated drug delivery systems

can yield a more effective drug accumulation in tissues and

body fluids than classic ones, with minimal side effects,

increasing patient quality of life with high socio-economic

benefits.[3, 4, 6] Moreover, novel drug-delivery systems would

offer protection and would enhance the pharmacokinetics of

easily degradable peptides and proteins that often have short

half-lives in vivo.[7] Thus, today, drug research aims to

improve those techniques that could selectively deliver drugs to

the pathological sites.[8] Nanotechnology might be crucial for

drug delivery and nanoparticles (NPs) are of high importance,

with many potential applications in clinical medicine and

research. NPs emerge as the future of drug delivery technology

as they might be future crucial diagnostic and therapeutic tools.

Nanotechnology implies the design and use of materials, tools

and systems through the manipulation of matter on nanometric

scale that is atomic, molecular and supramolecular scales.

Nanotechnology applications in multiple fields and mostly are

becoming to be tested in clinical trials on humans.[9] Drug

delivery nanomediated systems are based on biocompatible

nanocarriers, such as gold nanoparticles, carbon nanotubes,

nanovesicles, micellar systems and dendrimers.[10, 11]

Additionally, one of the major benefits of nanotechnology is

the targeted drug delivery at the site of the disease by passive

targeting of drugs to the site of action or by selective active

targeting of the active pharmaceutic agent. Nanotechnology

also tries to find potential applications of nanoparticles for

efficient drug delivery. Of these, gold nanoparticles with their

unique chemical and physical properties properties have

emerge as promising carrier for delivery of various molecules

with therapeutic properties. This paper illustrates the latest

achievements in the applications of gold nanoparticles as drug

delivery tools for the therapy of human diseases.

Peptide delivery using GNPs

Various methods employing carriers for drug delivery have

been developed during the past one and a half decade in order to

ease the uptake of plasmids, peptides, short oligonucleotides

and proteins. The discovery of the Antennapedia home

domain[12] brought great enthusiasm regarding the ability of

short cationic peptides to translocate cell membrane without

disruption. These were called cell-penetrating peptides

(CPPs).[13] Various CPPs have been detailed since then,

including Tat, Sweet Arrow peptide, transportan and

polyarginines, together with their controversed method of

internalization.[14-16]

Drug Delivery Applications of Gold

Nanoparticles

Lucian Mocan, MD, PhD,

T

Page 2: Drug Delivery Applications of Gold Nanoparticlesresearchpub.org/journal/bmbn/number/vol1-no1/vol1-no… ·  · 2014-06-21Drug Delivery Applications of Gold ... passivating agents

BIOTECHNOLOGY, MOLECULAR BIOLOGY AND NANOMEDICINE VOL.1 NO.1 OCTOBER 2013

ISSN: 2330-9318 (Print) ISSN: 2330-9326 (Online) http://www.researchpub.org/journal/bmbn/bmbn.html

2

Fig 1.Schematic illustration of potential applications of gold nanoparticles in Biology and Medicine

Briefly, CPPs use more mechanisms of endocytosis in order to

translocate across the plasma membrane. Vizualization of Tat

peptide-conjugated quantum dots in live cells revealed that

peptide-conjugated nanomaterials penetrate cells via

macropinocytosis. [13, 17] Data records show that the quantum

dot-loaded vesicles were actively carried by molecular vehicles

to an asymmetric perinuclear region.[18] Even if this study

employed quantum dots, the same mechanisms may occur

when using gold nanoparticles. Direct membrane translocation

of CPPs was also indicated when they appear in larger

concentrations and in association with endocytotic inhibitors.

CPPs may be employed in transfection as they only require

incubation, without any other physical disruption and

manipulation of the membrane.

CPP functionalized GNPs have been supplied, examining their

efficiency for delivery. CPPs are believed to enhance

intracellular uptake, but their subcellular location and efflux out

of endosomes is contradictory. Electron microscopy indicated

the presence of Tat peptide functionalized gold nanoparticles (3

nm) in the nucleus and cytoplasm.[19] On the other hand,

studies employing Tat, penetratin peptide (10 nm gold

nanoparticles) or amphipathic proline-rich peptide (12 nm gold

nanoparticles, SAP) functionalized gold nanoparticles

indicated that nanoparticles are trapped in endosomes.[19] The

inconsistency of these studies might be related to the type of

cell lines used, the cargo size[20, 21] the chemical content of

the cover layers, and can also be due to artifacts resulted from

the fixation imposed by electron microscopy.[22] Cell shape

and cell viability must also be strongly considered. Several

studies assert intracellular absorption and nuclear localization,

but visualize dead or dying cells. Therefore, these studies are

not notable, as membrane permeability clearly indicates cell

death.

Feldherr et al. successfully achieved nuclear localization using

the microinjection of gold nanoparticles (∼ 20 nm) covered

with nucleoplasmin (containing NLS), but observed no nuclear

import with the NLS sequence alone.[23] Tkachenko et al.

detected endosomal trapping, without nuclear localization,

using the same NLS sequence from the SV40 large T antigen

with 20 nm GNPs nurtured on HepG2 cells.[24] Using

video-enhanced color differential interference contrast

(VEC-DIC) microscopy, the authors could visualize nuclear

delivery of gold nanoparticles with an adenoviral fiber protein

exhibiting receptor-mediated endocytosis (RME) and nuclear

localization sequences (NLS). The single cell showing

evidence of successful delivery does not seem very healthy and

it might be involved in a death program. This stresses one more

time the importance of assessing cell shape and cell viability in

such experiments, in order to prevent the appearance of

artifacts. Altogether, except several studies observing nuclear

localization in dead cells, NLS could not achieve nuclear

localization under any conditions, proving that endosomal

escape is mandatory for a successful intracellular delivery.[25]

Transferrin-receptor mediated drug delivery

Vectors enabling gold nanoparticle delivery also include

proteins. Transferrin-receptor interactions have represented

potential drug and gene uptake pathways.[26-28] Transferrin is

of major importance in iron transport for the synthesis of

hemoglobin. It was successfully used for a better internalization

of gold nanoparticles, 20 nm in size, covered with transferrin

(96 nm hydrodynamic radius) as imaged by atomic force

microscopy (AFM). The absorption of 25 nm

transferrin-coupled gold nanoparticles was also assessed by

laser scanning confocal microscopy. AFM indicated receptor

mediated endocytosis and confocal microscopy observed a

spotty fluorescent signal, suggesting endosomal localization. In

a third study employing differently sized and shaped particles,

TEM and fluorescence imaging also exhibited endosomal

localization.[29]

Nowadays, various functional molecular linkers and

passivating agents are used in functionalized gold nanoparticles

employed in biomedical applications. Nevertheless, the main

groups used for molecule-gold conjugation usually include:

thiolate [30, 31], [21, 29, 32] dithiocarbamate amine[33],

carboxylate, isothiocyanate or phosphine [34-37] moieties.

Recent studies show that direct gold–C binding may be

provided by trimethyltin leaving group. Still, there is need for

further tests in order to use it in biomedical or

nanoparticle-based applications. Chosing a particular

molecular anchor depends on the desired molecule adaptability

to a specific application, with trends in bonding strength

usually following hard–soft acid–base (HSAB) theory

developed by Pearson for soft gold surfaces. Non-adaptable

applications most often involve thiol-based anchoring groups

while adaptable applications employ amine or carboxylate

surface anchors. Burda et al. demonstrated the importance of

using more adaptable amino linkers than stronger thiol groups

Page 3: Drug Delivery Applications of Gold Nanoparticlesresearchpub.org/journal/bmbn/number/vol1-no1/vol1-no… ·  · 2014-06-21Drug Delivery Applications of Gold ... passivating agents

BIOTECHNOLOGY, MOLECULAR BIOLOGY AND NANOMEDICINE VOL.1 NO.1 OCTOBER 2013

ISSN: 2330-9318 (Print) ISSN: 2330-9326 (Online) http://www.researchpub.org/journal/bmbn/bmbn.html

3

for the therapeutic results obtained after gold

nanoparticle-mediated delivery of photodynamic therapy

agents.

GNP as anti-infective agents

In addition to antitumor substances, antibiotics and other

antibacterial agents are also used for GNP delivery. [26-28] Gu

et al. [38] prepared stable water soluble vancomycin

(Van)-covered gold nanoparticles as polyvalent inhibitors and

demonstrated their effectiveness towards various

enteropathogenic strains (including vancomycin-resistant ones),

such as vancomycin-resistant strains (VRE) and

Gram-negative bacteria. In this experiment, gold nanoparticles

(4-5 nm, obtained in toluene) were functionalized with

bis(vancomycin) cystamide (in H2O), under dynamic stirring

for 12 hours, to generate Gold-S bonds that link Van to gold. A

small excess of gold nanoparticles was employed to secure

complete consumption of all Van molecules. When the reaction

was completed, Gold-Van nanoparticles were dissolved in the

aqueous solution and then removed from the organic phase.

Rosemary et al.[39] obtained similar results using a complex

made of ciprofloxacin and gold nanoshells with high

antibacterial activity against E. coli. Drug delivery research

regarding Ciprofloxacin@SiO2 (cip@SiO2) was conducted

employing E. coli DH5R and L. lactis MG 1363 using the agar

dilution method, results being compared to those of free

ciprofloxacin. Hydrophobicity studies on cip@SiO2 presented

a different penetration pathway compared to the free drug.

FITC@SiO2 helped perform nanoshell/bacteria interaction

studies. The fluorescence image indicated shell internalization

into the microorganism. Studies regarding transmission

electron microscopy were carried out on cip@SiO2-treated E.

Coli and showed that bacterial morphology is unharmed by

nanoshell therapy, even if the shells were not observed.

In a study conducted by Selvaraj and Alagar,[40] a colloidal

gold conjugate of the antileukemic drug 5-fluorouracil

exhibited observable antibacterial and antifungal activities

against Staphylococcus aureus, Micrococcus luteus,

Pseudomonas aeruginosa, E. coli, Aspergillus fumigatus and A.

niger. The 5FU-colloidal gold complex, obtained by means of

the imino (–NH) group, was assessed using different analytical

techniques (UV–vis spectroscopy, FT-IR, cyclic voltammetry,

transmission electron microscopy, fluorescence). The type of

interaction was determined using fluorescence and

electrochemical studies. The results indicated that the

association between anticancer drugs and gold nanoparticles

determines a much powerful reaction against Gram-negative

bacterial infections. The mechanisms involved in this improved

ativity are still not clear.

In the experiment carried out by Burygin et al.[41] free

gentamicin and its combination with GNPs proved not to differ

in their antimicrobial activity in assays on solid and liquid

nutrient media. They examined the influence of gold NPs

(16-nm in size) on the antibacterial activity exhibited by

gentamicin. No differences appeared within experimental error

limits between the antibacterial activity of gentamicin and that

of a gentamicin–gold NP mixture in different gentamicin and

particle concentrations. GNPs sedimented from the conjugates

exhibited no antibacterial activity, but the supernatant liquids

from gentamicin–NP complexes and free gentamicin displayed

the same activity. Electron microscopy and extinction spectra

changes demonstrated the presence of NP aggregates that were

unable to enter the gel. This explains the absence of growth

inhibition following addition of NP sediment. Moreover, the

same degree of activity of free gentamicin and the mixture

indicates the small amount of antibiotic that could bind to the

particles. The CFU method indicated that the bacteria-killing

action of a gentamicin–NP complex is not different from that of

free gentamicin within error limits. The parameters inhibiting

bacterial growth in liquid bacterial culture (MIC and MTC)

were also similar for gentamicin and for the gentamicin–NP

mixture. There were no significant differences between the free

antibiotic and the mixture in terms of antibacterial activity, on

neither solid nor liquid nutrient medium, in any of the studies.

The increased antibacterial activity due to the presence of NPs

indicates that there must be at least two conditions (still not

enough) to detect such effects. Firstly, antibiotic–NP

complexes need to be stabilized and their spectrum and color

should correlate with those of single-particle nonaggregated

colloids. Secondly, the amount of antibiotic capping the

particle surface should be large enough to guarantee an increase

in local antibiotic concentration at the site of bacterium–

particle contact. Therefore, even though gold NPs alone do not

employ any antimicrobial activity, they may behave as drug

curriers. In conclusion, GNPs increase the surface area and

carry large amounts of drug on it.

Rai et al.[42] particularly suggested the direct use of cefaclor

antibiotic in the synthesis of GNPs. They have defined a simple

way to synthesise 52 to 22 nm spherical gold nanoparticles

using cefaclor (a second-generation cephalosporin antibiotic) at

different temperatures. Additionally, they acquired robust

antimicrobial coatings on glass slides that maintain their

antimicrobial activity even under unfavorable conditions. The

rate of gold ion reduction in solution was essential in

determining synthesized gold nanoparticle size. The authors

demonstrated that the amino group on cefaclor behaves as both

reducing and coating agent and thus, the antibacterial activity

of cefaclor is retained because of the presence of free blactam

rings available on nanoparticle surface. Moreover, cefaclor

reduced GNPs proved to have effective antimicrobial activity

on both Gram-positive (S. aureus) and Gram-negative (E. coli)

bacteria, compared to cefaclor and gold nanoparticles alone.

Cefaclor inhibited peptidoglycan synthesis resulting in porous

cell walls. As a result, gold nanoparticles produced holes in the

cell wall, followed by leakage of cell contents and cell death.

The authors demonstrated that gold nanoparticles attach to

bacterial DNA and inhibit DNA uncoiling and transcription,

thus supporting bacterial death.

The high antioxidant activity of GNPs conjugated with

salvianic acid A was demonstrated by Du et al. who proposed

potential applications using this complex. The synthesis of a

novel salvianic acid A-coated gold nanoparticle (Au@PEG3SA)

was achieved using a layer-by-layer self-assembly method. The

DPPH radical-scavenging assay showed that the constant value

for DPPH radical - Au@PEG3SA reaction was about nine

Page 4: Drug Delivery Applications of Gold Nanoparticlesresearchpub.org/journal/bmbn/number/vol1-no1/vol1-no… ·  · 2014-06-21Drug Delivery Applications of Gold ... passivating agents

BIOTECHNOLOGY, MOLECULAR BIOLOGY AND NANOMEDICINE VOL.1 NO.1 OCTOBER 2013

ISSN: 2330-9318 (Print) ISSN: 2330-9326 (Online) http://www.researchpub.org/journal/bmbn/bmbn.html

4

times higher than with salvianic acid A monomer. The authors

also showed that the mechanism for the kinetic enhancement of

Au@PEG3SA resulted from the transition state variation in

DPPH radical-scavenging reaction and possibly, from the pi–pi

stacking interactions among adjacent phenolic groups located

on the surface of GNPs. Another major discovery was the

dramatic increase in the kinetics for the ROS scavenging

process in living cells, as well as in vivo, enhancing cellular

antioxidant status.[43]

Data provided by Bowman et al. showed that GNP complexes

involving[44] TAK-779 display a more pronounced activity

against HIV than the native preparation at the cost of the high

local concentration. The synthesis of SDC-1721, a potent HIV

inhibitor TAK-779 fragment, was accompanied by its

conjugation to gold nanoparticles of 2.0 nm in diameter.

Phytohemagglutinin (PHA)-stimulated peripheral blood

mono-nuclear cells (PBMCs) were infected with the

CCR5-tropic HIV-1 clone JR-CSF, in the presence or absence

of test compounds, in order to assess the antiviral activity of

nanoparticle conjugates. On the 7th day after infection,

supernatants were collected and HIV-1 capsid p24 antigen was

assessed using ELISA test. TAK-779 proved to inhibit the

replication of HIV-1 with an IC50 of 10 nM. The IC50 for

TAK-779 against four different CCR5-tropic viral isolates

varied between 1.6 and 3.7 nM. Nevertheless, JR-CSF was not

one of the analyzed viruses. With a similar virus, JR-FL,

TAK-779 indicated a 20 nM IC50. Responsiveness to CCR5

penetration inhibitors is influenced by receptor expression

levels and HIV-1 envelope/receptor affinity, mediated by both

cellular and viral determinants. Thus, JR-CSF appears to be

naturally less sensitive to TAK-779.

GNPS as drug delivery platform for the therapy of

metabolic diseases

Joshi et al. noted hormone[45] insulin functionalized gold

nanoparticles and their application in transmucosal delivery for

diabetes mellitus treatment. Insulin was load onto bare gold

nanoparticles and aspartic acid-coated gold nanoparticles and

administered in diabetic Wistar rats by means of both oral and

intranasal (transmucosal) administration. Much lower blood

glucose levels (postprandial hyperglycemia) were noticed when

insulin was delivered using gold nanoparticle carriers by

transmucosal administration. Moreover, the management of the

intranasal delivery protocol for postprandial hyperglycemia

was similar to that obtained by the conventional subcutaneous

administration used for type I diabetes mellitus.

Chamberland et al.[46] noted the therapeutic effect of

etanercept, an antirheumatic drug, conjugated to gold nanorods.

The authors have estimated the potential of a developing hybrid

imaging technology for use in noninvasive monitoring of

anti-TNF drug delivery (photoacoustic tomography). After

preparing the contrast agent made of etanercept-conjugated

gold nanorods, ELISA tests were performed in order to validate

the conjugation and to indicate that the conjugated anti-TNF-α

drug was biologically active. PAT of ex vivo rat tail joints with

the joint connective tissue increased by intra-articularly

injected contrast agent was carried out to analyze PAT

performance in visualizing the distribution of the

gold-nanorod-conjugated drug in articular tissues. This system

helped visualize gold nanorods of <1 pM concentration in

phantoms or 10 pM concentration in biological tissues, with

good signal-to-noise ratio and high spatial resolution,

indicating the feasibility of binding TNF antagonist

pharmaceutical compounds to gold nanorods, the preservation

of the tumor necrosis factor antagonist mechanism of action

together with the preliminary assessment of the new PAT

technology for the detection of optical contrast agents

conjugated with antirheumatic drugs.

REFERENCES

[1] Giljohann D A, Seferos DS, Daniel WL, Massich MD, Patel

PC, Mirkin CA. Gold nanoparticles for biology and medicine.

Angewandte Chemie International Edition 2010;

49:3280-3294.

[2] Mishra B, Patel BB, Tiwari S. Colloidal nanocarriers: a

review on formulation technology, types and applications

toward targeted drug delivery. Nanomedicine: Nanotechnology,

Biology and Medicine 2010; 6:9-24.

[3] Liang X J, Chen C, Zhao Y, Jia L, Wang PC.

Biopharmaceutics and therapeutic potential of engineered

nanomaterials. Curr Drug Metab 2008; 9:697-709.

[4] Parveen S, Misra R, Sahoo SK. Nanoparticles: a boon to

drug delivery, therapeutics, diagnostics and imaging.

Nanomedicine: Nanotechnology, Biology and Medicine 2012;

8:147-166.

[5] Jain P K, Huang X, El-Sayed IH, El-Sayed MA. Noble

metals on the nanoscale: optical and photothermal properties

and some applications in imaging, sensing, biology, and

medicine. Acc Chem Res 2008; 41:1578-1586.

[6] Chambers E, Mitragotri S. Long circulating nanoparticles

via adhesion on red blood cells: mechanism and extended

circulation. Exp Biol Med 2007; 232:958-966.

[7] Mocan T, Clichici S, Agoşton-Coldea L, Mocan L, Şimon Ş,

Ilie IR et al. Implications of oxidative stress mechanisms in

toxicity of nanoparticles (review). Acta Physiol Hung 2010;

97:247-255.

[8] Ilie I, Ilie R, Mocan T, Bartos D, Mocan L. Influence of

nanomaterials on stem cell differentiation: designing an

appropriate nanobiointerface. International Journal of

Nanomedicine 2012; 7:3011-3025.

[9] Iancu ,Cornel, Ilie I,R., Georgescu C,E., Ilie ,Razvan, Biris

A,R., Mocan ,Teodora et al. Applications of Nanomaterials in

Cell Stem Therapies and the Onset of Nanomedicine.

[10] Bharali D J, Khalil M, Gurbuz M, Simone TM, Mousa SA.

Nanoparticles and cancer therapy: A concise review with

emphasis on dendrimers. International journal of nanomedicine

2009; 4:1.

[11] Ting G, Chang CH, Wang HE. Cancer nanotargeted

radiopharmaceuticals for tumor imaging and therapy.

Anticancer Res 2009; 29:4107-4118.

[12] Chaudhary S, Smith CA, del Pino P, de la Fuente, Jesus M,

Mullin M, Hursthouse A et al. Elucidating the Function of

Penetratin and a Static Magnetic Field in Cellular Uptake of

Magnetic Nanoparticles. Pharmaceuticals 2013; 6:204-222.

Page 5: Drug Delivery Applications of Gold Nanoparticlesresearchpub.org/journal/bmbn/number/vol1-no1/vol1-no… ·  · 2014-06-21Drug Delivery Applications of Gold ... passivating agents

BIOTECHNOLOGY, MOLECULAR BIOLOGY AND NANOMEDICINE VOL.1 NO.1 OCTOBER 2013

ISSN: 2330-9318 (Print) ISSN: 2330-9326 (Online) http://www.researchpub.org/journal/bmbn/bmbn.html

5

[13] Cleal K, He L, D Watson P, T Jones A. Endocytosis,

Intracellular Traffic and Fate of Cell Penetrating Peptide Based

Conjugates and Nanoparticles. Curr Pharm Des 2013;

19:2878-2894.

[14] Nasrolahi Shirazi A, Tiwari R, Chhikara BS, Mandal D,

Parang K. Design and Biological Evaluation of

Cell-Penetrating Peptide–Doxorubicin Conjugates as Prodrugs.

Molecular pharmaceutics 2013; 10:488-499.

[15] Lehner R, Wang X, Marsch S, Hunziker P. Intelligent

Nanomaterials for Medicine: Carrier platforms and targeting

strategies in the context of clinical application. Nanomedicine:

Nanotechnology, Biology and Medicine 2013.

[16] Martin I, Teixido M, Giralt E. Intracellular Fate of

Peptide-Mediated Delivered Cargoes. Curr Pharm Des 2013;

19:2924-2942.

[17] Lévy R, Shaheen U, Cesbron Y, Sée V. Gold nanoparticles

delivery in mammalian live cells: a critical review. Nano

Reviews 2010; 1.

[18] Kairdolf B A, Smith AM, Stokes TD, Wang MD, Young

AN, Nie S. Semiconductor Quantum Dots for Bioimaging and

Biodiagnostic Applications. Annual Review of Analytical

Chemistry 2013.

[19] Ruan G, Agrawal A, Marcus AI, Nie S. Imaging and

tracking of tat peptide-conjugated quantum dots in living cells:

new insights into nanoparticle uptake, intracellular transport,

and vesicle shedding. J Am Chem Soc 2007; 129:14759-14766.

[20] Webster A, Compton SJ, Aylott JW. Optical calcium

sensors: development of a generic method for their introduction

to the cell using conjugated cell penetrating peptides. Analyst

2005; 130:163-170.

[21] Liu Y, Shipton MK, Ryan J, Kaufman ED, Franzen S,

Feldheim DL. Synthesis, stability, and cellular internalization

of gold nanoparticles containing mixed peptide-poly (ethylene

glycol) monolayers. Anal Chem 2007; 79:2221-2229.

[22] Richard J P, Melikov K, Vives E, Ramos C, Verbeure B,

Gait MJ et al. Cell-penetrating peptides A reevaluation of the

mechanism of cellular uptake. J Biol Chem 2003; 278:585-590.

[23] Feldherr C M, Lanford RE, Akin D. Signal-mediated

nuclear transport in simian virus 40-transformed cells is

regulated by large tumor antigen. Proceedings of the National

Academy of Sciences 1992; 89:11002-11005.

[24] Tkachenko A G, Xie H, Liu Y, Coleman D, Ryan J,

Glomm WR et al. Cellular trajectories of peptide-modified gold

particle complexes: comparison of nuclear localization signals

and peptide transduction domains. Bioconjug Chem 2004;

15:482-490.

[25] Chen F, Gerion D. Fluorescent CdSe/ZnS

nanocrystal-peptide conjugates for long-term, nontoxic

imaging and nuclear targeting in living cells. Nano Letters 2004;

4:1827-1832.

[26] Choi C H J, Alabi CA, Webster P, Davis ME. Mechanism

of active targeting in solid tumors with transferrin-containing

gold nanoparticles. Proceedings of the National Academy of

Sciences 2010; 107:1235-1240.

[27] Yang P, Sun X, Chiu J, Sun H, He Q. Transferrin-mediated

gold nanoparticle cellular uptake. Bioconjug Chem 2005;

16:494-496.

[28] Chithrani B D, Chan WC. Elucidating the mechanism of

cellular uptake and removal of protein-coated gold

nanoparticles of different sizes and shapes. Nano letters 2007;

7:1542-1550.

[29] Li J, Wang L, Liu X, Zhang Z, Guo H, Liu W et al. < i> In

vitro</i> cancer cell imaging and therapy using

transferrin-conjugated gold nanoparticles. Cancer Lett 2009;

274:319-326.

[30] Strange M, Lopez-Acevedo O, a kkinen H. Oligomeric

Gold− Thiolate Units Define the Properties of the Molecular

Junction between Gold and Benzene Dithiols. The Journal of

Physical Chemistry Letters 2010; 1:1528-1532.

[31] Martin B R, Dermody DJ, Reiss BD, Fang M, Lyon LA,

Natan MJ et al. Orthogonal self‐assembly on colloidal gold‐platinum nanorods. Adv Mater 1999; 11:1021-1025.

[32] Tong M C, Chen W, Sun J, Ghosh D, Chen S.

Dithiocarbamate-capped silver nanoparticles. The Journal of

Physical Chemistry B 2006; 110:19238-19242.

[33] Daniel M, Astruc D. Gold nanoparticles: assembly,

supramolecular chemistry, quantum-size-related properties,

and applications toward biology, catalysis, and nanotechnology.

Chem Rev 2004; 104:293-346.

[34] Martin‐Rendon E, Sweeney D, Lu F, Girdlestone J,

Navarrete C, Watt S. 5‐Azacytidine‐treated human

mesenchymal stem/progenitor cells derived from umbilical

cord, cord blood and bone marrow do not generate

cardiomyocytes in vitro at high frequencies. Vox Sang 2008;

95:137-148.

[35] Martin‐Rendon E, Sweeney D, Lu F, Girdlestone J,

Navarrete C, Watt S. 5‐Azacytidine‐treated human

mesenchymal stem/progenitor cells derived from umbilical

cord, cord blood and bone marrow do not generate

cardiomyocytes in vitro at high frequencies. Vox Sang 2008;

95:137-148.

[36] Martin‐Rendon E, Sweeney D, Lu F, Girdlestone J,

Navarrete C, Watt S. 5‐Azacytidine‐treated human

mesenchymal stem/progenitor cells derived from umbilical

cord, cord blood and bone marrow do not generate

cardiomyocytes in vitro at high frequencies. Vox Sang 2008;

95:137-148.

[37] Mitchell K E, Weiss ML, Mitchell BM, Martin P, Davis D,

Morales L et al. Matrix cells from Wharton's jelly form neurons

and glia. Stem Cells 2003; 21:50-60.

[38] Gu H, Xu K, Xu C, Xu B. Biofunctional magnetic

nanoparticles for protein separation and pathogen detection.

Chemical Communications 2006:941-949.

[39] Rosemary M, MacLaren I, Pradeep T. Investigations of the

antibacterial properties of ciprofloxacin@ SiO2. Langmuir

2006; 22:10125-10129.

[40] Selvaraj V, Alagar M. Analytical detection and biological

assay of antileukemic drug 5-fluorouracil using gold

nanoparticles as probe. Int J Pharm 2007; 337:275-281.

[41] Burygin G, Khlebtsov B, Shantrokha A, Dykman L,

Bogatyrev V, Khlebtsov N. On the enhanced antibacterial

activity of antibiotics mixed with gold nanoparticles.

Nanoscale research letters 2009; 4:794-801.

[42] Rai A, Prabhune A, Perry CC. Antibiotic mediated

synthesis of gold nanoparticles with potent antimicrobial

Page 6: Drug Delivery Applications of Gold Nanoparticlesresearchpub.org/journal/bmbn/number/vol1-no1/vol1-no… ·  · 2014-06-21Drug Delivery Applications of Gold ... passivating agents

BIOTECHNOLOGY, MOLECULAR BIOLOGY AND NANOMEDICINE VOL.1 NO.1 OCTOBER 2013

ISSN: 2330-9318 (Print) ISSN: 2330-9326 (Online) http://www.researchpub.org/journal/bmbn/bmbn.html

6

activity and their application in antimicrobial coatings. Journal

of Materials Chemistry 2010; 20:6789-6798.

[43] Du L, Miao X, Jiang Y, Jia H, Tian Q, Shen J et al. An

effective strategy for the synthesis of biocompatible gold

nanoparticles using danshensu antioxidant: prevention of

cytotoxicity via attenuation of free radical formation.

Nanotoxicology 2012; 7:294-300.

[44] Bowman M, Ballard TE, Ackerson CJ, Feldheim DL,

Margolis DM, Melander C. Inhibition of HIV fusion with

multivalent gold nanoparticles. J Am Chem Soc 2008;

130:6896-6897.

[45] Joshi H M, Bhumkar DR, Joshi K, Pokharkar V, Sastry M.

Gold nanoparticles as carriers for efficient transmucosal insulin

delivery. Langmuir 2006; 22:300-305.

[46] Chamberland D L, Agarwal A, Kotov N, Fowlkes JB,

Carson PL, Wang X. Photoacoustic tomography of joints aided

by an Etanercept-conjugated gold nanoparticle contrast

agent—an ex vivo preliminary rat study. Nanotechnology 2008;

19:095101.