27
2256 Chem. Soc. Rev., 2012, 41, 2256–2282 This journal is c The Royal Society of Chemistry 2012 Cite this: Chem. Soc. Rev., 2012, 41, 2256–2282 Gold nanoparticles in biomedical applications: recent advances and perspectives Lev Dykman a and Nikolai Khlebtsov* ab Received 19th June 2011 DOI: 10.1039/c1cs15166e Gold nanoparticles (GNPs) with controlled geometrical, optical, and surface chemical properties are the subject of intensive studies and applications in biology and medicine. To date, the ever increasing diversity of published examples has included genomics and biosensorics, immunoassays and clinical chemistry, photothermolysis of cancer cells and tumors, targeted delivery of drugs and antigens, and optical bioimaging of cells and tissues with state-of-the-art nanophotonic detection systems. This critical review is focused on the application of GNP conjugates to biomedical diagnostics and analytics, photothermal and photodynamic therapies, and delivery of target molecules. Distinct from other published reviews, we present a summary of the immunological properties of GNPs. For each of the above topics, the basic principles, recent advances, and current challenges are discussed (508 references). 1. Introduction Gold was one of the first metals discovered by humans, and the history of its study and application is estimated to be a minimum of several thousand years old. The first information on colloidal gold can be found in tracts by Chinese, Arabic, and Indian scientists, who obtained colloidal gold as early as in the fifth and fourth centuries B.C. and used it, in particular, for medical purposes (the Chinese ‘‘gold solution’’ and the Indian ‘‘liquid gold’’). In the Middle Ages in Europe, colloidal gold was studied and employed in alchemists’ laboratories. Specifically, Paracelsus wrote about the therapeutic properties of quinta essentia auri, which he prepared by reducing auric chloride with alcohol or oil plant extracts. He used ‘‘potable gold’’ to treat some mental a Institute of Biochemistry and Physiology of Plants and Microorganisms, RAS, 13 Pr. Entuziastov, Saratov 410049, Russian Federation. E-mail: [email protected] b Saratov State University, 83 Ul. Astrakhanskaya, Saratov 410012, Russian Federation Lev Dykman Dr Lev A. Dykman, leading researcher of the Immuno- chemistry Lab at the Institute of Biochemistry and Physiol- ogy of Plants and Micro- organisms Russian Academy of Sciences. He has published more than 200 scientific works including one monograph on colloidal gold nanoparticles. His current scientific interests include immunochemistry, fabrication of gold nano- particles and their applications to biological and medical studies. In particular, his research is aimed at interaction of nanoparticles and conjugates with immune-responsible cells and at the delivery of engineered particles to target organs, tissues, and cells. Nikolai Khlebtsov Professor Nikolai G. Khlebtsov, head of the Nanobiotechnology Lab at the Institute of Bio- chemistry and Physiology of Plants and Microorganisms Russian Academy of Sciences. He also holds a Biophysics Chair at the Saratov State University. He has published more than 300 scientific works, including two mono- graphs. His current scientific interests include biophotonics and nanobiotechnology of plasmon-resonant particles, biomedical applications of metal nanoparticles, static and dynamic light scattering by small particles and clusters, programming and computer simulation of light scattering and absorption by various metal and dielectric nanostructures. Prof. Khlebtsov also serves as an Associate Editor of the Journal of Quantitative Spectroscopy and Radia- tive Transfer. Chem Soc Rev Dynamic Article Links www.rsc.org/csr CRITICAL REVIEW Published on 30 November 2011. Downloaded by Universidade Tecnologica Federal do Parana on 25/03/2015 12:57:35. View Article Online / Journal Homepage / Table of Contents for this issue

Gold Nanoparticles c1cs15166e

Embed Size (px)

DESCRIPTION

From Elsevier

Citation preview

  • 2256 Chem. Soc. Rev., 2012, 41, 22562282 This journal is c The Royal Society of Chemistry 2012

    Cite this: Chem. Soc. Rev., 2012, 41, 22562282

    Gold nanoparticles in biomedical applications: recent advancesand perspectives

    Lev Dykmana and Nikolai Khlebtsov*ab

    Received 19th June 2011

    DOI: 10.1039/c1cs15166e

    Gold nanoparticles (GNPs) with controlled geometrical, optical, and surface chemical properties

    are the subject of intensive studies and applications in biology and medicine. To date, the ever

    increasing diversity of published examples has included genomics and biosensorics, immunoassays

    and clinical chemistry, photothermolysis of cancer cells and tumors, targeted delivery of drugs

    and antigens, and optical bioimaging of cells and tissues with state-of-the-art nanophotonic

    detection systems. This critical review is focused on the application of GNP conjugates to

    biomedical diagnostics and analytics, photothermal and photodynamic therapies, and delivery

    of target molecules. Distinct from other published reviews, we present a summary of the

    immunological properties of GNPs. For each of the above topics, the basic principles, recent

    advances, and current challenges are discussed (508 references).

    1. Introduction

    Gold was one of the rst metals discovered by humans, and

    the history of its study and application is estimated to be a

    minimum of several thousand years old. The rst information

    on colloidal gold can be found in tracts by Chinese, Arabic,

    and Indian scientists, who obtained colloidal gold as early as

    in the fth and fourth centuries B.C. and used it, in particular,

    for medical purposes (the Chinese gold solution and the

    Indian liquid gold).

    In the Middle Ages in Europe, colloidal gold was studied

    and employed in alchemists laboratories. Specically, Paracelsus

    wrote about the therapeutic properties of quinta essentia auri,

    which he prepared by reducing auric chloride with alcohol or

    oil plant extracts. He used potable gold to treat some mental

    a Institute of Biochemistry and Physiology of Plants andMicroorganisms, RAS, 13 Pr. Entuziastov, Saratov 410049,Russian Federation. E-mail: [email protected]

    b Saratov State University, 83 Ul. Astrakhanskaya, Saratov 410012,Russian Federation

    Lev Dykman

    Dr Lev A. Dykman, leadingresearcher of the Immuno-chemistry Lab at the Instituteof Biochemistry and Physiol-ogy of Plants and Micro-organisms Russian Academyof Sciences. He has publishedmore than 200 scientic worksincluding one monograph oncolloidal gold nanoparticles.His current scientic interestsinclude immunochemistry,fabrication of gold nano-particles and their applicationsto biological and medicalstudies. In particular, his

    research is aimed at interaction of nanoparticles and conjugateswith immune-responsible cells and at the delivery of engineeredparticles to target organs, tissues, and cells.

    Nikolai Khlebtsov

    Professor Nikolai G. Khlebtsov,head of the NanobiotechnologyLab at the Institute of Bio-chemistry and Physiology ofPlants and MicroorganismsRussian Academy of Sciences.He also holds a BiophysicsChair at the Saratov StateUniversity. He has publishedmore than 300 scienticworks, including two mono-graphs. His current scienticinterests include biophotonicsand nanobiotechnology ofplasmon-resonant particles,biomedical applications of

    metal nanoparticles, static and dynamic light scattering by smallparticles and clusters, programming and computer simulation oflight scattering and absorption by various metal and dielectricnanostructures. Prof. Khlebtsov also serves as an AssociateEditor of the Journal of Quantitative Spectroscopy and Radia-tive Transfer.

    Chem Soc Rev Dynamic Article Links

    www.rsc.org/csr CRITICAL REVIEW

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online / Journal Homepage / Table of Contents for this issue

  • This journal is c The Royal Society of Chemistry 2012 Chem. Soc. Rev., 2012, 41, 22562282 2257

    disorders and syphilis. Paracelsus once proclaimed that chemistry

    is for making medicines, not for making gold out of metals.

    His contemporary Giovanni Andrea applied aurum potabile to

    the treatment of lepra, ulcer, epilepsy, and diarrhea. In 1583,

    alchemist David de Planis-Campy, surgeon to the King of

    France Louis XIII, recommended his elixir of longevityan

    aqueous colloidal gold solutionas a means of life prolongation.

    The rst book on colloidal gold preserved to our days was

    published by philosopher and doctor of medicine Francisco

    Antonii in 1618.1 It contains information on the preparation

    of colloidal gold and on its medical applications, including

    practical suggestions.

    In 1880, a method was put forward to treat alcoholism by

    intravenous injection of a colloidal gold solution (gold

    cure).2 In 1927, the use of colloidal gold was proposed to

    ease the suering of inoperable cancer patients.3 Colloidal

    gold in color reactions toward spinal-uid and blood-serum

    proteins has been taken advantage of since the rst half of the

    twentieth century.4 Colloidal solutions of the 198Au gold

    isotope (half-life time, 65 h) were therapeutically successful

    at cancer care facilities.5 More recent examples of colloidal

    gold applications include catalytic processes and electron

    transport in biomacromolecules,6 transport of substances into

    cells by endocytosis,7 investigation of cell motility,8 and improve-

    ment of PCR eciency.9

    Despite the centuries-old history, a revolution in immuno-

    chemistry,10 associated with the use of gold particles in

    biological research, took place in 1971, when British researchers

    W. P. Faulk and G. M. Taylor published an article titled An

    immunocolloid method for the electron microscope.11 In that

    article, a technique was described to conjugate antibodies with

    colloidal gold for direct electron microscopic visualization of

    Salmonella surface antigens, representing the rst time that a

    colloidal gold conjugate served as an immunochemical marker.

    From this point on, the use of colloidal-gold biospecic

    conjugates in various elds of biology and medicine became

    very active. There has been a wealth of reports dealing with the

    application of functionalized gold nanoparticles (GNPs; conju-

    gates with recognizing biomacromolecules, e.g., antibodies, lectins,

    enzymes, or aptamers)1214 to the studies of biochemists, micro-

    biologists, immunologists, cytologists, physiologists, morphologists,

    and many other specialist researchers.

    The range of uses of GNPs in current medical and biological

    research is extremely broad. In particular, it includes genomics;

    biosensorics; immunoassay; clinical chemistry; detection and

    photothermolysis of microorganisms and cancer cells; targeted

    delivery of drugs, peptides, DNA, and antigens; and optical

    bioimaging and monitoring of cells and tissues with the use of

    state-of-the-art nanophotonic recording systems. GNPs have

    been proposed for use in practically all medical applications,

    including diagnostics, therapy, prophylaxis, and hygiene

    (e.g., in water purication15). Extensive information on the most

    important aspects of preparation and use of colloidal gold in

    biology and medicine can be found elsewhere.1630 Such a

    broad range of application is based on the unique physical and

    chemical properties of GNPs. Specically, the optical properties

    of GNPs are determined by their plasmon resonance, which is

    associated with the collective excitation of conduction electrons

    and is localized in a wide region (from visible to infrared,

    depending on particle size, shape, and structure).31 Scheme 1

    shows a simplied scheme for the current biomedical applications

    of GNPs, which reects the structure of this review. However,

    since biodistribution and toxicity have been extensively reviewed

    and discussed in a number of recent publications (see, e.g., ref. 32

    and references therein), we restrict ourselves to a short comment

    in the Conclusions section.

    Considering the great body of existing information and the

    high speed of its renewal, we chose in this review to generalize

    the data that have accumulated during the past few years for

    the most promising directions in the use of GNPs in current

    medical and biological research.

    2. GNPs in diagnostics

    2.1 Visualization and bioimaging methods

    GNPs have been actively used in various visualization and

    bioimaging methods to identify chemical and biological

    agents.33,34 Historically, electron microscopy (mainly its trans-

    mission variant, TEM) has for a long time (starting in 197111)

    been the principal method to detect biospecic interactions

    with the help of colloidal gold particles (owing to their high

    electron density). Although GNPs can intensely scatter and

    emit secondary electrons, they have not received equally wide

    acceptance in scanning electron microscopy.35 It is no mere

    chance that the rst three-volume book on the use of colloidal

    gold36 was devoted mostly to the application of GNPs in

    TEM. A peculiarity of current use of the electron microscopic

    technique is the application of high-resolution transmission

    electron microscopes and systems for the digital recording and

    processing of images.37 The major application of immunoelectron

    microscopy in present-day medical and biological research is

    the identication of infectious agents and their surface antigens3840

    (Fig. 1a). The techniques often employed for the same purposes

    include scanning atomic-force41 (Fig. 1b), scanning electron,42

    and uorescence43 microscopies.

    Alongside the use of classic colloidal gold with quasi-

    spherical particles (nanospheres) as labels for microscopic

    studies, the past few years have seen the application of

    nonspherical cylindrical particles (nanorods), nanoshells, nano-

    cages, nanostars, and other types of particles, referred to by

    Scheme 1 Generalized scheme for the biomedical application of

    GNPs. Along with basic applications in diagnostics and therapy, this

    review briey discusses the immunological properties of GNPs.

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online

  • 2258 Chem. Soc. Rev., 2012, 41, 22562282 This journal is c The Royal Society of Chemistry 2012

    the broad term noble-metal plasmon-resonant particles25

    (Fig. 2). Table 1 illustrates some plasmonic properties and

    possible biomedical applications of the gold nanomaterials

    shown in Fig. 2. This is by no means a detailed description of

    geometrical or optical parameters (size, shape, structure,

    absorption and scattering cross sections, their spectra, etc.).

    In fact, we only provide typical ranges for the major plasmon

    wavelength and its sensitivity to the dielectric environment in

    terms of plasmonic shift per refractive index unit (RIU).

    Accordingly, Fig. 2 and Table 1 should be considered as an

    attempt to show a cross section of the work in this area, rather

    than a comprehensive list.

    Recently, the popularity of visualization methods employing

    GNPs and optical microscopy,55 in particular confocal laser

    microscopy, has also been on the rise. Confocal microscopy is

    a technique for detecting microobjects with the aid of an

    optical system ensuring that light emission is recorded only

    when it comes from objects located in the systems focal point.

    This allows one to scan samples according to height and,

    ultimately, to create their 3D images by superposition of

    scanograms. In this method, the use of GNPs and their

    antibody conjugates permits real-time detection of gold penetration

    into living (e.g., cancerous) cells at the level of a single particle

    and even estimation of their number.5659

    Confocal images can be obtained with, e.g., detection of

    uorescence emission (confocal uorescence microscopy) or

    resonance elastic or two-photon (multiphoton) light scattering

    by plasmonic nanoparticles (confocal resonance-scattering or

    two-photon luminescence microscopy). These techniques are

    based on detecting microobjects with the optical microscope, in

    which the luminescence of an object is excited owing to simulta-

    neous absorption of two (or more) photons, the energy of each of

    which is lower than that needed for uorescence excitation. The

    basic advantage of this method is the increase in contrast through

    a strong reduction in the background signal. Specically, two-

    photon luminescence of GNPs enables molecular markers of

    cancer to be visualized on or inside cells,6063 Bacillus spores,64

    and the like. Fig. 3a shows an example of combined bioimaging

    of cancer cells with the help of adsorption, uorescence, and

    luminescence plasmon resonance labels.

    Dark-eld microscopy remains one of the most popular GNP-

    aided bioimaging methods. It is based on light scattering by

    microscopic objects, including those whose sizes are smaller than

    the resolution limit for the light microscope (Fig. 3b and c). In

    dark-eld microscopy, the light entering the objective lens is solely

    that scattered by the object at side lighting (similarly to the Tyndall

    eect); therefore, the scattering object looks bright against a dark

    background.

    Compared with uorescent labels, GNPs have greater potential

    to reveal biospecic interactions with the help of dark-eld

    microscopy,65 because the particle scattering cross section is three

    to ve orders of magnitude greater than the uorescence cross

    section for a single molecule. This principle was for the rst time

    employed byMostafa El-Sayeds group66 for their new method of

    simple and reliable diagnosis of cancer with the use of GNPs. The

    method is founded on the preferential binding of GNPs con-

    jugated with tumor-antigen-specic antibodies to the surface

    of cancerous cells, as compared with binding to healthy cells.

    With dark-eld microscopy, therefore, it is possible to map

    out a tumor with an accuracy of several cells (Fig. 3b and c).

    Subsequently, gold nanorods,67 nanoshells,68 nanostars,69 and

    nanocages70 were used for these purposes.

    The use of self-assembled monolayers or island lms, as well

    as nonspherical and/or composite particles, opens up fresh

    opportunities for increased sensitivity of detection of bio-

    molecular binding on or near the nanostructure surface. The

    principle of amplication of a biomolecular binding signal

    depends on the strong local electromagnetic elds arising near

    nanoparticles with spiky surface sites or in narrow (on the

    order of 1 nm or smaller) gaps between two nanoparticles.

    This determines the increased plasmon-resonance sensitivity to

    the local dielectric environment71 and the high scattering

    intensity as compared to the sensitivity and intensity of

    equivolume spheres. Therefore, such nanostructures show

    considerable promise for use in biomedical diagnostics assisted

    by dark-eld microscopy.7274

    In dark-eld microscopy, GNPs are employed for detection

    of microbial cells and their metabolites,75 bioimaging of

    cancerous cells7678 and revelation of receptors on their surface,79,80

    study of endocytosis,81 and other purposes. In most biomedical

    applications, the eectiveness of conjugate labeling of cells is

    assessed qualitatively. One of the few exceptions is the work of

    Khanadeev et al.,82 in which a method was suggested for the

    quantitative estimation of the eectiveness of GNP labeling of

    cells and its use was illustrated with the specic example of

    labeling of pig embryo kidney cells with gold nanoshell con-

    jugates. Another approach was developed by Fu et al.83

    Apart from the just mentioned ways to record biospecic

    interactions with the help of diverse versions of optical micro-

    scopy and GNPs, the development of other state-of-the-art

    detection and bioimaging methods is currently active. These are

    referred to collectively as biophotonic methods.31,84 Biophotonics

    combines all studies related to the interaction of light with

    biological cells and tissues. Existing biophotonic methods include

    optical coherence tomography,8587 X-ray and magnetic

    resonance tomography,88,89 photoacoustic microscopy90 and

    tomography,91 uorescence correlation microscopy,92 and other

    techniques. These methods also successfully use variously sized

    and shaped GNPs. In our opinion, biophotonic methods

    employing nonspherical gold particles may hold particular

    promise for bioimaging in vivo.34,93,94

    Fig. 1 TEM image of a Listeria monocytogenes cell labeled with an

    antibodycolloidal gold conjugate (a) and a scanning atomic-force

    microscopy image of tobacco mosaic virus labeled with an antibody

    colloidal gold conjugate (b). Adapted from Bunin et al.39 (a) and from

    Drygin et al.41 (b) by permission from Springer.

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online

  • This journal is c The Royal Society of Chemistry 2012 Chem. Soc. Rev., 2012, 41, 22562282 2259

    Fig. 2 Various types of plasmon-resonant nanoparticles: 16 nm nanospheres (a);25 gold nanorods (b);44 gold bipyramids (c);45 gold nanorods

    surrounded by silver nanoshells (d);25 nanorice (gold-coated Fe2O3 nanorods) (e);46 SiO2/Au nanoshells (f)

    25 (the inset shows a hollow

    nanoshell47); nanobowls with bottom cores (g);48 spiky SiO2/Au nanoshells (h)49 (the inset shows a gold nanostar50); gold tetrahedra, octahedra,

    and cubooctahedra (i);51,52 gold nanocubes (j);51 silver nanocubes and goldsilver nanocages obtained from them (in the insets) (k);53 and gold

    nanocrescents (l).54 Adapted from the data of the cited papers by permission from The Royal Society of Chemistry, Elsevier, IOP Publishing,

    Springer, Wiley Interscience, and The American Chemical Society.

    Table 1 Properties and possible biomedical applications of plasmonic nanoparticles

    Particle Major resonances/nm Plasmonic shift/RIU/nm Possible biomedical applications

    Colloidal Au nanospheres (3100 nm) 510570 4590 EM, OI, HA, SA, PPT,a DCAu nanorods (thickness, 1020 nm; aspect ratio, 210) 6501200 150290 OI, OA, PPT, HIA,Au bipyramids 6501100 150540 OI, BSAu(core)/Ag(shell) NRs 5501000 OI, SERSFe2O3 (core)/Au (shell) nanorice 11001300 790810 SERS, BSSiO2(core)/Au(shell) 6001100 160

    b

    315c OI, SA, PPTHollow Au shell 125 OI, PPTNanobowls 5601000 SERSSpiky SiO2 nanoshells and Au nanostars 600850,

    675770 240665 OI, SERSAu polyhedralsd 550750 EM, OIAu cubes 550700 83 EM, OIAuAg nanocages 4501000 410620 OIA, BS, SERS, PPTAu nanocrescentse 9802600 240880 BS

    Designations: RIUrefractive index unit, EMelectron microscopy, OIoptical imaging, HAhomogeneous assays, SAsolid phase assays,

    PPTplasmonic photothermal therapy, DCdrug carriers, OAoptoacoustical applications, BSbiosensing, SERSsurface enhanced Raman

    scattering. a PPT applications of clusterized Au nanospheres. b Monolayer data. c Suspension data. d Tetrahedra, octahedra, and cubooctahedra.e Nanolithography array.

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online

  • 2260 Chem. Soc. Rev., 2012, 41, 22562282 This journal is c The Royal Society of Chemistry 2012

    2.2 Analytical diagnostic methods

    2.2.1 Homophase techniques. Beginning in 1980s, colloidal

    gold conjugated with recognizing biomacromolecules was com-

    ing into use in various analytical methods in clinical diagnostics.

    In 1980, Leuvering et al.95 put forward a new immunoanalysis

    method that they called sol particle immunoassay (SPIA). A new

    SPIA version was advanced by Mirkin et al.96 for the colori-

    metric detection of DNA. Both versions (protein and DNA

    SPIA) use two principles: (1) The sol color and absorption

    spectrum change little when biopolymers are adsorbed on

    individual particles.25 (2) When particles move closer to each

    other to distances smaller than 0.1 of their diameter, the red color

    of the sol changes to purple or gray and the absorption spectrum

    broadens and red-shifts.97 This change in the absorption spectrum

    can easily be detected spectrophotometrically or visually

    (Fig. 4a and b98,99).

    The authors employed an optimized variation of SPIA (by

    using larger gold particles and monoclonal antibodies to various

    antigenic sites) to detect human chorionic gonadotropin.100

    Subsequently, the assay was used for the immunoanalysis of

    Shistosoma101 and Rubella102 antigens; estimation of immuno-

    globulins,103,104 thrombin (by using aptamers),105 and glucose;106

    direct detection of cancerous cells;107 detection of Leptospira cells

    in urine;108 detection of Alzheimers disease markers;109 determi-

    nation of protease activity;110 and other purposes. The simulta-

    neous use of antibody conjugates of gold nanorods and

    nanospheres for the detection of tumor antigens was described

    by Liu et al.111 Wang et al.112 provided data on the detection of

    hepatitis B virus in blood with gold nanorods conjugated to

    specic antibodies.

    All SPIA versions proved easy to implement and were highly

    sensitive and specic. However, investigators came up against the

    fact that antigenantibody reactions on sol particles do not

    necessarily lead to system destabilization (aggregation). Some-

    times, despite the obvious complementarity of the pair, changes

    in solution color (and, correspondingly, in absorption spectra)

    were either absent or slight. Dykman et al.113 presented a model

    for the formation of a second protein layer on gold particles

    without loss of sol aggregative stability. The spectral changes

    arising from biopolymer adsorption on the surface of metallic

    particles are comparatively small114 (Fig. 4c and d). However, as

    shown by Englebienne,115 even such minor changes in absorption

    spectra, resulting from a change in the structure of the biopolymer

    layer (specically, its average refractive index) near the GNP

    surface, could be recorded and used for assay in biological

    applications.

    For increasing the sensitivity of the analytical reaction, new

    interaction-recording techniques have been proposed, including

    photothermal spectroscopy,116 laser-based double beam absorption

    spectroscopy,117 hyper-Rayleigh scattering,118 dierential light-

    scattering spectroscopy,119 and dynamic light scattering.120 In

    addition, two vibrational spectroscopymethodssurface enhanced

    infrared absorption spectroscopy121 and surface enhanced Raman

    spectroscopy122have been suggested for use in SPIA.

    The ability of gold particles interacting with proteins to

    aggregate with a solution color change served as a basis for the

    development of a colorimetric method for protein determination.123

    A new SPIA version using microplates and an ELISA reader,

    with colloidal-gold-conjugated trypsin as a specic agent for

    proteins, was devised by Dykman et al.124

    Fig. 3 Confocal image of HeLa cells in the presence of GNPs (a). Blue, the nuclei stained with Hoechst 33258; red, the actin cytoskeleton labeled

    with Alexa Fluor 488 phalloidin; green, unlabeled GNPs. The image was taken by two-photon microscopy.63 Dark-eld microscopy of cancerous

    (b) and healthy (c) cells by using GNPs conjugated with antibodies to epidermal growth factor.66 Adapted from the data of the cited papers by

    permission from The American Chemical Society.

    Fig. 4 Sol particle immunoassay. (a) Scheme for the aggregation of

    conjugates as a result of binding by target molecules and (b) the

    corresponding changes in the spectra and in sol color. (c) Scheme for

    the formation of a secondary layer without conjugate aggregation and

    (d) the corresponding dierential extinction spectra at 600 nm.

    Adapted from the data of Khlebtsov et al.,97 Wu et al.,98 and

    Englebienne115 by permission from The American Chemical Society

    and The Royal Society of Chemistry.

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online

  • This journal is c The Royal Society of Chemistry 2012 Chem. Soc. Rev., 2012, 41, 22562282 2261

    At present, colorimetric DNA detection includes two strategies:

    (1) The use of GNPs conjugated with thiol-modied

    ssDNA96,125130 or aptamers.131 (2) The use of unmodied

    GNPs132135 (Table 2).

    The rst strategy is based on the aggregation of conjugates

    of 1030 nm GNPs with thiol-modied ssDNA probes after

    the addition of target polynucleotides to the system. A cross-

    linking variant of the rst strategy uses two types of probes

    complementary to both terminal target sites. Hybridization of

    the targets and probes leads to the formation of GNP aggre-

    gates, which is accompanied by a change in the absorption

    spectrum of the solution and can readily be detected visually,

    photometrically,137 or by dynamic light scattering.130,138

    In contrast to the cross-linking aggregation, Maeda and

    coworkers139,140 developed a non-cross-linking diagnostic system

    involving GNP conjugates of only one type, with 30 or 50 thiol-modied probes. The aggregation of GNPs occurred at high

    ionic strength (1 M NaCl) and only with complementary

    probes and targets, whereas noncomplementary targets prevented

    aggregation. Contrary to the observations of the Maeda group,139

    Baptista et al.129,141 reported enhanced colloidal stability after the

    addition of complementary targets to a DNA conjugate solution

    even at high ionic strength (2 M NaCl), whereas noncomple-

    mentary targets did not prevent aggregation at 2 M NaCl. The

    apparent contradictions between these data were explained by

    Song et al.142 through the dierence in surface functionalization

    density.

    Consider now the second DNA-sensing strategy, which

    utilizes unmodied GNPs. This approach is based on the

    observation by Li and Rothberg133 that at high ionic strength,

    ssDNA protects unmodied gold nanoparticles from aggregation,

    whereas dsDNA does not. This method was employed by

    Shawky et al.143 to detect hepatitis C virus. Recently, Xia et al.144

    described another variant of the second strategy, which uses

    ssDNA, unmodied GNPs, and a cationic polyelectrolyte.

    All the above-cited reports on DNA detection used citrate-

    stabilized anionic GNPs. He et al.135 described a new version

    of unmodied-particle assay employing as-prepared positively

    charged CTAB-coated gold nanorods. After mixing nanorods

    with the probe and target ssDNA under appropriate hybridi-

    zation conditions, the authors observed particle aggregation,

    as determined by absorption and scattering spectra. In contrast,

    the addition of noncomplementary targets did not cause any spectral

    changes. According to He et al.,135 the detection limit (DL) of

    a 21-mer ssDNA was about 0.1 nM, whereas Ma et al.145

    reported a 0.1 pM DL for an optimized version of He et al.s

    assay135 and 30-mer ssDNA. Quite recently, Pylaev et al.44

    applied CTAB-coated positively charged GNPs in combination

    with spectroscopic and dynamic scattering methods for the

    detection of DNA targets. Thus, there exists a more than

    Table 2 Detection of DNA with the use of gold nanoparticles

    Particles Probe Target Detection method Detection limit Ref.

    ModiedGNSs(GNSs withchemicallyattachedprobes)

    50-HS(CH2)6N13N15-30 ssDNA Cross-linking aggregation,UVAS, spot test

    10 fmol 125, 1997

    50-SH(dT)10-30 Biotinylated PCRproduct

    Hybridization of targets with poly-Aand streptavidin followed by a striptest with GNSs

    2 fmol (500 copiesof PSA cDNA)

    126, 2003

    50-N15(CH2)10SH-30a;

    50-SH(CH2)10N15-30b

    ssDNA Bio-bar-code assay, scanometricdetection of silver-enhanced spots

    500 zmol (10 copiesin 30 mL)

    127, 2004

    50-N15C-30(CH2)3SH;HS(CH2)65

    0-N18-30PCR product Non-cross-linking aggregation in

    1 M NaCl, VE, spot test100250 nM;12 nM

    128, 2006;140, 2005

    50-HSN16-30 1-round PCRproduct

    Stabilization of nanoprobes bytargets in 2M NaCl, VE, UVAS

    300 nM 129, 2006

    50-N12C3SS-30;50-SSC6N15-30

    ssDNA Aggregation, DLS 5 pM 130, 2008

    50-SH(CH2)6N15-30;50-RoxN15(CH2)3SH-30

    ssDNA SERS 10 zM 136, 2010

    UnmodiedGNSs

    Rhodamine red50-N15-30 ssDNA Fluorescence quenching/retainingwith nontarget/target ssDNA

    0.5 pM 133, 2004

    50-N15-30 ssDNA, PCRproduct

    Stabilization/aggregation withnontarget/target ssDNA, UVAS

    2 pM 134, 2004

    ssDNA, aptamers +conjugated polyelectrolytec

    ssDNA, thrombin,cocaine, Hg

    Aggregation in the case of nontargetmolecules, VE, UVAS

    1 pM (DNA);10 nM (thrombin);10 mM (cocaine);50 mM (Hg)

    144, 2010

    50-N21-30, 50-N23-30 ssDNA Aggregation of positively chargedGNSs, UVAS, DLS

    10 pM 44, 2011

    UnmodiedGNRs

    50-N21-30 ssDNA Aggregation, UVLS 1.7 nM 135, 2008

    50-N30-30 ssDNA Aggregation (optimized), UVAS 0.1 pM 145, 2010

    Designations: Nmm-bases oligonucleotide; GNSsgold nanospheres (colloidal gold nanoparticles with a roughly spherical shape); UVASUVvis

    absorption spectroscopy; VEvisual evaluation; UVLSUVvis light scattering spectroscopy; PSAprostate-specic antigen; DLSdynamic light

    scattering; GNRsgold nanorods. a Particle probe. b Substrate probe. c Poly [(9,9-bis (60-N,N,N-trimethylammonium)hexyl)uorene-alt-1,4-phenylene]bromide.

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online

  • 2262 Chem. Soc. Rev., 2012, 41, 22562282 This journal is c The Royal Society of Chemistry 2012

    three-order dierence in the reported estimations of the detection

    sensitivity of colorimetric methods (0.1100 pM). Further work

    is called for, as the existing aggregation models are inconsistent

    with the detection limits of about 0.11 pM DNA.44

    The above-mentioned SPIA formats have served for the

    detection of the DNA of mycobacteria,129,146,147 staphylococci,148

    streptococci,149 and chlamydiae150 in clinical samples.

    The sensitivity of DNA detection can be increased with

    optical methods that utilize plasmonic enhancement of the

    local electromagnetic eld near particle-cluster hotspots. For

    example, Hu et al.136 developed a sensitive DNA biosensor

    based on multilayer metalmoleculemetal nanojunctions and

    the SERS technique. With regard to an HIV-1 DNA sequence,

    the sensor could detect a concentration as low as 1019 M(1023 mol) with the ability of single base mismatch discrimination.Another way to reach PCR-like sensitivity involves a bio-bar-code

    approach combined with a silver-enhanced spot test.127

    2.2.2 Dot immunoassay. At the early stages of immuno-

    assay development, preference was given to liquid-phase techni-

    ques, in which bound antibodies were precipitated or unbound

    antigen was removed by adsorption with dextran-coated activated

    charcoal. Currently, the most popular techniques are solid-

    phase ones (rst used for protein radioimmunoassay), because

    they permit the analysis to be considerably simplied and the

    background signal to be reduced. Themost widespread solid-phase

    carriers are polystyrene plates and nitrocellulose membranes.

    Membrane immunoassays (dot and blot assays) commonly

    employ radioactive isotopes (125I, 14C, 3H) and enzymes

    (peroxidase, alkaline phosphatase, etc.) as labels. In 1984, four

    independent reports were published151154 in which colloidal

    gold was proposed as a label in a solid-phase immunoassay.

    The use of GNP conjugates in solid-phase assays is based on

    the fact that the intense red coloration of a gold-containing

    marker allows the results of a reaction run on a solid carrier to

    be determined visually. Immunogold methods in a dotblot

    assay outperform other techniques (e.g., enzyme immunoassay)

    in terms of sensitivity (Table 3), rapidity, and cost.155,156 After

    an appropriate immunochemical reaction is run, the sizes of

    GNPs can be increased by enhancement with salts of silver157

    or gold (autometallography),158 considerably expanding the

    application limits of the method. An optimized solid-phase

    assay using a densitometry system aorded a dynamic detection

    range of 1 mM to 1 pM159 with a detection limit of 100 aM,which was lowered to 100 zM by silver enhancement. The use

    of state-of-the-art instrumental detection methods, such as

    photothermal deection of a probing laser beam, caused by

    heating of the local environment near absorbing particles

    subjected to light pulses from a heating laser (LISNA assay),160

    also ensures a very broad detection range (within three orders of

    magnitude, to the extent of several individual particles on a spot).

    In specic staining, a membrane with applied material under

    study is incubated in a solution of antibodies (or other

    biospecic probes) labeled with colloidal gold.18 As probes,

    gold dot or blot assays use immunoglobulins, Fab and scFv

    antibody fragments, staphylococcal protein A, lectins, enzymes,

    avidin, aptamers, and other probes. Sometimes, several labels

    are used simultaneously (e.g., colloidal gold and peroxidase or

    alkaline phosphatase) for the detection of multiple antigens on a

    membrane.161

    Colloidal gold in membrane assays has served for the diagnosis

    of parasitic,162166 viral,167170 and fungal171,172 diseases, tuber-

    culosis,173 melioidosis,174 syphilis,175 brucellosis,176 shigellosis,177

    E. coli infections,178 salmonellosis,179 and early pregnancy;180

    blood group determination;181 dotblot hybridization;182 detection

    of antibiotics;183 diagnosis of myocardial infarction184 and

    hepatitis B;185 and other purposes.

    The immunodot assay is one of the simplest methods developed

    to analyze membrane-immobilized antigens. In some cases, it

    permits quantitative determination of antigens. Most commonly,

    the immunodot assay is employed to study soluble antigens.186

    However, there have been several reports in which corpuscular

    antigens (whole bacterial cells) served as a research object in dot

    assays with enzyme labels.187 Bogatyrev et al.188,189 were the rst

    to perform a dot assay of whole bacterial cells, with the reaction

    products being visualized with immunogold markers (cellgold

    immunoblotting). This technique was applied to the serotyping

    of nitrogen-xing soil microorganisms of the genus Azospirillum

    and subsequently to the rapid diagnosis of enteric infections.190

    Gas et al.191 used a dot assay with GNPs to detect whole cells of

    the toxic phytoplankton Alexandrium minutum.

    Khlebtsov et al.192 rst presented experimental results for the

    use of gold nanoshells as biospecic labels in dot assays. Three

    types of gold nanoshells were examined that had silica core

    diameters of 100, 140, and 180 nm and a gold shell thickness of

    about 15 nm (Fig. 5, data for 140/15 nm nanoshells not shown).

    The biospecic pair was normal rabbit serum (target molecules)

    and sheep antirabbit immunoglobulins (target-recognizing

    molecules). When the authors performed a standard protocol

    for a nitrocellulose-membrane dot assay, with 15 nm gold

    nanospheres as labels, the minimum detectable quantity of

    rabbit IgG was 15 ng. Replacing colloidal gold conjugates with

    nanoshells increased the assay sensitivity to 0.2 ng for 180/15 nm

    gold nanoshells and to 0.4 ng for 100/15 and 140/15 nm

    nanoshells. Such noticeable increases in sensitivity with nano-

    shells, as against nanospheres, can be explained by the dierent

    optical properties of the particles.193

    A very promising analytical approach is the use of colloidal gold

    to analyze large arrays of antigens in micromatrices (immuno-

    chips).194,195 These enable an analyte to be detected in 384

    samples simultaneously at a concentration of 6070 ng L1 or,with account taken of the microlitre amounts of sample and

    detecting immunogold marker, with a detection limit of lower

    than 1 pg.

    The sensitivity of protein dot-analysis can be greatly improved

    with a combination of activated glass slides and a CCD

    camera196,197 or a atbed scanner.198 In the case of very dilute

    Table 3 Sensitivity limits for immunodot/blot methods implementedon nitrocellulose lters by using various labels (according to ref. 155)

    LabelSensitivity limit(pg of protein per fraction)

    125I 5Horseradish peroxidase 10Alkaline phosphatase 1Colloidal gold 1Colloidal gold + silver 0.1Fluorescein isothiocyanate 1000

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online

  • This journal is c The Royal Society of Chemistry 2012 Chem. Soc. Rev., 2012, 41, 22562282 2263

    samples, a dot-immunogold ltration assay183 has been suggested.

    Finally, the sensitivity of a standard ELISA can be enhanced

    up to the single-molecule detection limit199 by using GNPs in

    colorimetric analysis of ELISA samples.200,201

    2.2.3 Immunochromatographic assays. In 1990, several

    companies began to manufacture immunochromatographic

    test systems for hand-held diagnostics. Owing to their high

    specicity and sensitivity, these strip tests have found a wide

    utility in the detection of narcotics, toxins, highly dangerous

    infections, and urogenital diseases.202208 Methods have been

    developed for the diagnosis of tuberculosis,209 helicobacteriosis,210

    staphylococcal infection,211 hepatitis B,212 prostatitis,213 and early

    pregnancy,214 for DNA hybridization;215 for the detection of

    pesticides,216 aatoxin,217 hexoestrol,218 and cephalexin219 in

    environmental constituents; and for other purposes.

    The immunochromatographic assay is based on eluent

    movement along the membrane (lateral diusion), giving rise

    to specic immune complexes at dierent membrane sites; the

    complexes are visualized as colored bands.220 As labels, these

    systems use enzymes, colored lattices, but mostly GNPs.221,222

    The sample being examined migrates along the test strip at

    the cost of capillary forces. If the sample contains the sought-

    for substance or immunochemically related compounds, there

    occurs a reaction with colloidal-gold-labeled specic antibodies

    as the sample passes through the absorber. The reaction is

    accompanied by the formation of an antigenantibody

    complex. The colloidal preparation enters into a competitive

    binding reaction with the antigen immobilized in the test zone

    (as a rule, the detection of low-molecular-weight compounds

    employs conjugates of haptens with protein carriers for

    immobilization). If the antigen concentration in the sample

    exceeds the threshold level, the conjugate does not possess free

    valences for interaction in the test zone and the colored band

    corresponding to the formation of the complex is not revealed.

    When the sample does not contain the sought-for substance or

    when the concentration of that substance is lower than the

    threshold level, the antigen immobilized in the test zone of the

    strip reacts with the antibodies on the surface of colloidal gold,

    causing a colored band to develop.

    When the liquid front moves on, the gold particles with

    immobilized antibodies that have not reacted with the antigen

    in the strip test zone bind to antispecies antibodies in the

    control zone of the test strip. The appearance of a colored

    band in the control zone conrms that the test was done

    correctly and that the systems components are diagnostically

    active. Otherwise, the test is invalid. A negative test resultthe

    appearance of two colored bands (in the test zone and in

    the control zone)indicates that the antigen is absent from

    the sample or that its concentration is lower than the threshold

    level. A positive test resultthe appearance of one colored

    band in the control zoneshows that the antigen concen-

    tration exceeds the threshold level (Fig. 6).220

    Studies have shown that such assay systems are highly

    stable, their results are reproducible, and they correlate with

    alternative methods. Densitometric characterization of the

    dissimilarity degree for detected bands yields values ranging

    from 5 to 8%, allowing reliable visual determination of the

    analysis results. These assays are very simple and convenient

    to use.

    Nowadays, GNP-assisted immunochromatographic analysis is

    used actively in such elds as the rapid diagnostics of pseu-

    dorabies,223 tuberculosis,224 and botulism225 and the detection

    of pesticides,226 antibiotics,227 and toxins,228 in biological

    liquids and the environment.

    In summary, being eective diagnostic tools, rapid tests

    allow qualitative and quantitative determination, in a matter

    of minutes, of antigens, antibodies, hormones, and other

    Fig. 5 Dot assay of normal rabbit serum (1) by using suspensions of

    conjugates of 15 nm GNPs and SiO2/Au nanoshells (100 and 180 nm

    silica core diameters and 15 nm Au shell) with sheep antirabbit

    antibodies. The amount of IgG in the rst square of the top row is

    1 mg and decreases from left to right in accordance with twofolddilutions. The lower rows (2) correspond to the application of 10 mg ofbovine serum albumin to each square as a negative control. The

    detected analyte quantity is 15 ng for 15 nm GNPs and 0.4 and 0.2 ng

    for 100/15 and 180/15 nm nanoshells, respectively. Adapted from

    ref. 193 by permission from IOP Publishing.

    Fig. 6 Results of an immunochromatographic assay: positive, negative,

    and invalid determination because of the absent coloration in the

    control zone. Adapted from ref. 219 by permission from The American

    Chemical Society.

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online

  • 2264 Chem. Soc. Rev., 2012, 41, 22562282 This journal is c The Royal Society of Chemistry 2012

    diagnostically important substances in humans and animals.

    Rapid tests are highly sensitive and accurate, as they can

    detect more than 100 diseases (including tuberculosis, syphilis,

    gonorrhea, chlamydiosis, various types of viral hepatitis, etc.)

    and the whole gamut of narcotic substances used, with the

    reliability of detection being high. An important advantage of

    these tests is their use in diagnostics in vitro, which does not

    require a patients presence.

    However, immunochromatographic test strips are not devoid

    of weak points, related to reliability, sensitivity, and cost-eectiveness.

    Reliability and sensitivity depend, rst, on the quality of monoclonal

    antibodies used in a test and, second, on the antigen concen-

    tration in a biomaterial. The quality of monoclonal antibodies

    depends on the methods of their preparation, purication, and

    xation on a carrier. The antigen concentration depends on

    the disease state and the biomaterial quantity. For increasing

    the analysis sensitivity, it has been proposed to employ the

    silver enhancement procedure229 or gold nanorods as labels.230

    In addition, semiquantitative and quantitative instrumental

    formats of immunochromatographic analysis have been developed

    that use special readers for recording the intensity of a labels

    signal in the test zone of a test strip.220

    2.2.4 Plasmonic biosensors. Collective oscillations of

    conductive electron plasma in metals are called plasmons.231

    Depending on the boundary conditions, these oscillations can be

    categorized into three types:232 bulk plasmons (3D plasma);

    propagating surface plasmons (PSP), or surface plasmon polaritons

    (2D dielectric/metal interfaces); and localized surface plasmons

    (LSP), excited in nanoparticles (Fig. 7).25 Bulk plasmons cannot

    be excited by visible light, as their energy is about 10 eV for noble

    metals.231

    The term surface plasmon polaritons designates collective

    charge density oscillations at the metal/dielectric interface,

    propagating in a waveguide-like fashion along the surface

    (Fig. 7b). In the normal direction, the exciting electromagnetic

    eld is rapidly falling o with distance. At a given photon

    energy ho, the wave vector ho/c should be increased to ensureeective coupling for the transformation of incident photons

    into propagating surface plasmons.232 Therefore, direct excitation

    of surface plasmon polaritons with freely propagating light is not

    possible. However, this problem can be solved by two approaches:

    the grating couplingmethod and the attenuated reectionmethod,

    which use a lattice waveguide structure or total reection inside a

    prism, respectively.232

    In metal nanoparticles, the electron plasma is restricted in

    all three dimensions. Accordingly, localized surface plasmons

    dier from propagating surface plasmons because of the

    dierent boundary conditions to the Maxwell electromagnetic

    equations. In a small metal nanoparticle, the incident optical

    eld exerts a force on conductive electrons and displaces them

    from their equilibrium positions to create uncompensated

    charges at the nanoparticle surface (Fig. 7c). As the main

    eect producing the restoring force is the polarization of the

    particle surface, the excited nanoparticle behaves like a resonance

    oscillator possessing a localized plasmon resonance (LSPR).

    The principal dierence between the propagating and localized

    plasmons is that the former can be directly excited by light

    waves without any additional coupling set up. On the other

    hand, in both LSP and PSP cases, the excited plasmons feel

    the dielectric environment. It is the property that is the basis

    for the LSP and the PSP.

    The optical response of nanoparticles or their aggregates

    (especially ordered ones) depends on particle size, shape, and

    composition71,233 interparticle distance,234,235 and the properties

    of the particles local dielectric environment,236,237 which

    enables sensor tuning to be controlled. The theory behind

    the creation of LSP biosensors and their use in practice have

    been considered in ref. 238254. In general, all sensing strate-

    gies are based on the change in the intensity of the LSPR and

    its spectral shift caused by a change in the local dielectric

    environment owing to biospecic interactions near the particle

    surface. A unique local-sensing property of the LSPR is

    related to the rapid decay of its local eld, which only probes

    a nanoscale volume around the particle. In particular, the local

    nature of the LSPR allows one to detect single-molecular

    interactions near the particle surface by using various single-

    particle detecting schemes.254 Fig. 8a illustrates a sensitive

    detection of about 18 streptavidin molecules after their binding

    to a single biotin-functionalized gold nanorod (74 33 nm).255Nusz et al.255 also reported a 1 nM low detection limit (Fig. 8b)

    and discussed several ways to approach single-molecular detection.

    In recent years, gold and silver nanoparticles and their

    composites have served widely as eective optical transducers

    of biospecic interactions.256 In particular, the resonant

    optical properties of nanometre-sized metallic particles have been

    successfully used to develop plasmonic biochips and bio-

    sensors belonging to a broad family of sensors, viz. colorimetric,

    refractometric, electrochemical, and piezoelectric.220,257,258

    Such devices are of much interest in biology (determination

    of nucleic acids, proteins, and metabolites), medicine (screening

    of drugs, analysis of antibodies and antigens, diagnosis of

    Fig. 7 Schematic representation of the bulk (a), propagating surface

    (b), and localized surface (c) plasmons.

    Fig. 8 (a) Single-gold-nanorod light scattering spectra in water (1),

    with biotin, and after binding of about 18 streptavidin molecules.

    (b) LSPR peak shift as a function of the streptavidin concentration.

    The low detection limit is below 1 nM. Adapted from ref. 255 by

    permission from The American Chemical Society.

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online

  • This journal is c The Royal Society of Chemistry 2012 Chem. Soc. Rev., 2012, 41, 22562282 2265

    infections), and chemistry (rapid environmental monitoring,

    assays of solutions and disperse systems). Of particular signi-

    cance is the detection of specied nucleic acid (gene) sequences

    and the construction of new materials that relies on the formation

    of 3D ordered structures during hybridization in solutions of

    complementary oligonucleotides attached covalently to metallic

    nanoparticles.259

    For more than 10 years, biospecic interactions have been

    studied in systems in which GNPs are represented as ordered

    structures: self-assembled (thin lms)260 or as part of polymer

    matrices.261 Such structures have been actively employed for

    detection of biomolecules and infectious agents, development

    of DNA chips, and other purposes. In this case, investigators

    directly implement the possibility, in principle, of using the

    strong enhancement of the optical signal from the probe

    (GNPs conjugated to biospecic macromolecules) resulting

    from the strengthening of the exciting local eld in the

    aggregate formed from gold nanoclusters. At present, biosensors

    are built with novel, unique technologies, including monolayer

    self-assembly of metallic particles,262 ber-based LSPR sensing,263

    and nanolithography.264 The reported types of LPR studies

    of biospecic interactions include biotinstreptavidin, antigen

    antibody, lectincarbohydrate, toxinreceptor, aptamerprotein,

    and DNA hybridization.254 For further information about LPR

    sensing, the readers are referred to a recent excellent review by

    Mayer and Hafner.254

    In experimental work with SPR biosensors, three stages can

    be singled out:239 (1) one of the reagents (target-recognizing

    molecules) is covalently attached to the sensor surface. (2) The

    other reagent (target molecules) is added at a denite concen-

    tration to the sensor surface along with the ow of the buer.

    The process of complex formation is then recorded. (3) The

    sensor is regenerated (dissociation of the formed complexes).

    As this takes place, the following conditions should be met:

    Reagent immobilization on the substrate should not leadto a critical change in the conformation of native molecules.

    The relatively small dierence between the refractiveindices of most biological macromolecules forces one to use

    a high local concentration of binding sites on the sensor

    surface (10100 mM). The reagent being added should be vigorously agitated to

    achieve eective binding to the immobilized molecules. Unbound

    reagent should be promptly removed from the sensor surface to

    avoid nonspecic sorption.

    Apart from that, the sensitivity, stability, and resolution of a

    sensor depend directly on the characteristics of the optical

    system being used for recording. The most popular sensor

    system of this type is BIAcoret.265,266 The measurement

    principle in planar, prismatic, or mirror biosensors is similar

    to the principle of the method of frustrated total internal

    reection, traditionally employed to measure the thickness and

    refractive index of ultrathin organic lms on metallic (reecting)

    surfaces.257 The excitation of the plasmon resonance in a

    planar gold layer occurs when polarized light is incident on

    the surface at a certain angle. The excited electromagnetic

    waves and charged density waves propagate along the metal/

    dielectric interface. These propagating electromagnetic elds

    are localized near the interface because of the exponential

    decrease in amplitude perpendicularly to the dielectric with a

    typical attenuation length of up to 200 nm (the eect of total

    internal reection, Fig. 9). The reection coecient at a

    certain angle and light wavelength depends on the dielectric

    properties of the thin layer at the interface, which are

    ultimately determined by the mass of the captured target

    molecules at the sensing surface.

    Various types of GNP-aided biosensors have been developed

    for the immunodiagnostics of tick-borne encephalitis,268 the

    papilloma269 and HIV270 viruses, and Alzheimers disease;271,272

    the detection of organophosphorus substances and pesticides,273

    antibiotics,274 allergens,275 cytokines,276 carbohydrates,277 and

    immunoglobulins;278 the detection of tumor279 and bacterial280

    cells; the detection of brain cell activity;281 and other purposes.

    GNP-based biosensors are used not only in immuno-

    assay281284 but also for the supersensitive detection of nucleotide

    sequences.96,259,285 In their pioneering works, Raschke et al.286

    and McFarland et al.287 obtained record-high sensitivity of

    such sensors in the zeptomolar range, and they showed the

    possibility of detecting spectra of resonance scattering from

    individual particles as an analytical tool. This opens up the

    way to the recording of intermolecular interactions at the level

    of individual molecules.288,299 To make the response stronger,

    investigators often use avidinbiotin, barnasebarstar, and

    other systems.290 In addition, GNPs are applied in other

    analytical methods (diverse versions of chromatography, electro-

    phoresis, and mass spectrometry).291

    SPR and LSPR biosensors were compared in side-by-side

    experiments by Yonzon et al.292 for concanavilin A binding to

    monosaccharides and by Svedendahl et al.293 for biotinstrep-

    tavidin binding. It was found that both techniques demon-

    strate similar performance. As the bulk refractive index

    sensitivity is known to be higher for SPR, the above similarity

    was attributed to the long decay length of propagating plasmons,

    as compared to localized ones. The overall comparison of SPR

    and LSPR sensors can be found in ref. 251 and 254.

    Future development of low-cost SPR and LSPR biomedical

    sensors needs increasing the detection sensitivity and creating

    substrates that can operate in biological uids and can be easy

    to functionalize with probing molecules, to clean, and to reuse.

    Fig. 9 Typical setup for analyte detection in a BIAcoret-type SPR

    biosensor. The instrument detects changes in the local refractive index

    near a thin gold layer coated by a sensor surface with probing

    molecules. SPR is observed as a minimum in the reected light

    intensity at an angle dependent on the mass of captured analyte.

    The minimum SPR angle shifts from A to B when the analyte binds to

    the sensor surface. The sensogram is a plot of resonance angle versus

    time that allows for real-time monitoring of an association/dissociation

    cycle. Adapted from ref. 267 by permission from Springer.

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online

  • 2266 Chem. Soc. Rev., 2012, 41, 22562282 This journal is c The Royal Society of Chemistry 2012

    3. GNPs in therapy

    3.1 Plasmonic photothermal therapy

    Photothermal damage to cells is currently one of the most

    promising research avenues in the treatment of cancer and

    infectious diseases.294 The essence of this phenomenon is as

    follows: GNPs have an absorption maximum in the visible or

    near-IR region and get very hot when irradiated with corres-

    ponding light. If, in this case, they are located inside or around

    the target cells (which can be achieved by conjugating gold

    particles to antibodies or other molecules), these cells die.

    The thermal treatment of cancerous cells has been known in

    tumor therapy since the 18th century, employing both local

    heating (with microwave, ultrasonic, and radio radiation) and

    general hyperthermia (heating to 4147 1C for 1 h).295 Forlocal heating to 70 1C, the heating time may be reduced to34 min. Local and general hyperthermia leads to irreversible

    damage to the cells, caused by disruption of cell membrane

    permeability and protein denaturation. Naturally, the process

    also injures healthy tissues, which imposes serious limitations

    on the use of this method.

    The revolution in thermal cancer therapy is associated with

    laser radiation, which enabled controlled and limited injury to

    tumor tissues to be achieved.296 Combining laser radiation

    with ber-optic waveguides produced excellent results and was

    named interstitial laser hyperthermia.297 The weak point of

    laser therapy is its low selectivity, related to the need for high-

    powered lasers for eective stimulation of tumor cell death.298

    A variant of photothermal therapy was also proposed in which

    photothermal agents help to achieve selective heating of the

    local environment.299 Selective photothermal therapy relies on

    the principle of selective photothermolysis of a biological tissue

    containing a chromophorea natural or articial substance with

    a high coecient of light absorption.

    GNPs were rst used in photothermal therapy in 2003.300,301

    Subsequently, it was suggested to call this method plasmonic

    photothermal therapy (PPTT).295 Pitsillides et al.302 rst described

    a new technique for selective damage to target cells that is founded

    on the use of 20 and 30 nm gold nanospheres irradiated with 20 ns

    laser pulses (532 nm) to create local heating. For pulse photo-

    thermia in a model experiment, those authors were the rst to

    employ a sandwich technology for labeling T-lymphocytes with

    GNP conjugates.

    In a particularly promising method, GNPs have found

    application in the photothermal therapy of chemotherapy-resistant

    cancers.303 Unlike photosensitizers (see below), GNPs are unique in

    being able to keep their optical properties in cells for a long time

    under certain conditions. Successive irradiation with several laser

    pulses allows control of cell inactivation by a nontraumatic means.

    The simultaneous use of the scattering and absorbing properties of

    GNPs permits PPTT to be controlled by optical tomography.68

    Fig. 10 shows an example of successful therapy of an implanted

    tumor in a model experiment with mice.304

    The further development of PPTT and its acceptance in

    actual clinical practice305 depends on success in solving many

    problems, the most important ones being (1) the choice of

    nanoparticles with optimal optical properties, (2) the enhancement

    of nanoparticle accumulation in tumors and the lowering of

    total potential toxicity, and (3) the development of methods

    for the delivery of optical radiation to the targets and the

    search for alternative radiation sources combining high permeability

    with a possibility of heating GNPs.

    The rst requirement is determined by the concordance of the

    spectral position of the absorption plasmon resonance peak with

    the spectral window for biological tissues in the 700900 nm near-

    IR region.306 Khlebtsov et al.234 made a resumptive theoretical

    analysis of the photothermal eectiveness of GNPs, depending

    on their size, shape, structure, and aggregation extent. They

    showed that although gold nanospheres themselves are

    Fig. 10 Scheme and the results of an experiment on the photothermal destruction of an implanted tumor in a mouse (23 weeks after injection of

    MDA-MB-435 human cancer cells). Laser irradiation (a, b; 810 nm, 2 W cm2, 5 min) was performed at 72 h after injection of gold nanorodsfunctionalized with poly(ethylene glycol) (PEG) (a, c; 20 mg Au per kg) or of buer (b, d). It can be seen that the tumor continued developing after

    particle-free irradiation (control b), as it did after particle or buer administration without irradiation (controls a and d), and that complete

    destruction was obtained only in the experiment (a). Designations: NIR, near-IR region; NRs, nanorods. Adapted in part from ref. 304 by

    permission from The American Association for Cancer Research.

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online

  • This journal is c The Royal Society of Chemistry 2012 Chem. Soc. Rev., 2012, 41, 22562282 2267

    ineective in the near-IR region, aggregates formed from them

    can be very eective at suciently short interparticle distances

    (shorter than 0.1 of the particle diameter). Such clusters can be

    formed both on the surface of and inside cells.307 Experimental

    data indicating an enhancement of PPTT through clusterization

    have been presented in ref. 308310. Specically, Huang et al.308

    demonstrated that small aggregates composed of 30 nm particles

    enable cancerous cells to be destroyed at a radiation power

    20-fold lower than that in the particle-free control.

    The gold nanoshell and nanorod parameters optimal for PPTT

    have also been dened.234,311 By now, there have been quite a few

    publications dealing with the application in PPTT of gold nano-

    rods;67,312314 nanoshells;301,315320 and a comparatively new particle

    class, goldsilver nanocages.93,321,322 Experimental data com-

    paring the heating eciencies of nanorods, nanoshells, and

    nanocages have been reported in ref. 53, 323 and 324.

    Regarding the optimization of particle parameters, one should

    be aware of three matters of principle. First, the absorption cross

    section is not the sole parameter determining the eectiveness of

    PPTT.325 Rapid heating of nanoparticles or aggregates gives rise

    to vapor bubbles,326 which can cause cavitation damage to cells

    irradiated with visible309 or near-IR327 light. The eectiveness of

    vapor bubble formation increases substantially when nanoparticle

    aggregates are formed.301,307 Possibly, it is this eect, and not

    enhanced absorption, that bears responsibility for greater damage

    to cells, with all other factors being the same.325 Finally, particle

    irradiation with high-power resonant nanosecond IR pulses can

    lead to particle destruction as early as after the rst pulse (see, e.g.,

    ref. 328 and 329 and references therein to earlier publications). In

    a series of recent investigations, Lapotko et al.325,330 (see also

    references therein) paid their attention to the fact that the heating

    of GNPs and their destruction can sharply reduce the photo-

    thermal eectiveness of cold particles, which have been tuned to

    the laser wavelength. The use of femtosecond pulses oers no

    solution to this problem because of the low energy supplied, and

    for this reason, it is necessary to exert close control over the

    preservation of nanoparticles properties for the chosen irradiation

    mode. Furthermore, bubble formation strongly depends on the

    media as well as on laser intensity, thus making cellular damage

    poorly controlled.

    We now shift to consider the second question, associated

    with targeted nanoparticle delivery to a tumor. This question

    has two important aspects: increasing the particle concen-

    tration in the target and lowering the side eects caused by

    GNP accumulation in other organs, primarily in the liver and

    spleen (see below). Usually, there are two delivery strategies.

    One is based on the conjugation of GNPs to PEG; the other,

    on the conjugation with antibodies developed to specic

    marker proteins of tumor cells. PEG acts to enhance the

    bioavailability and stability of nanoparticles, ultimately

    prolonging the time of their circulation in the blood stream.

    Citrate-coated gold nanospheres, CTAB-coated nanorods,

    and nanoshells are less stable in saline solutions. When

    nanoparticles are conjugated to PEG, their stability is improved

    considerably and salt aggregation is prevented.

    In vivo PEGylated nanoparticles preferentially accumulate

    in tumor tissue owing to the increased permeability of the

    tumor vessels331 and are retained in it owing to the decreased

    lymph outow. In addition, PEGylated nanoparticles are less

    accessible to the immune system (stealth technologies). This

    delivery method is called passive, as distinct from the active

    version, which uses antibodies332,333 (Fig. 11). The active

    method of delivery is more reliable and eective, employing

    antibodies to specic tumor markers, most often to epidermal

    growth factor receptor (EGFR) and its varieties (e.g.,

    Her2),315,334,335 as well as to tumor necrosis factor (TNF).336

    Particular promise is oered by the simultaneous use of

    GNPantibody conjugates for both diagnosis and PPTT

    (methods of what is known as theranostics).337339 In addition

    to antibodies, active delivery may also use folic acid, which

    serves as a ligand for the numerous folate receptors of tumor

    cells,313,340,341 and hormones.342

    In the very recent past, the eectiveness of targeted nano-

    particle delivery to tumors has again become a subject for

    detailed study and discussion.343 In experiments with liposomes

    labeled with anti-Her2344 and GNPs labeled with transferrin,345

    it was shown that functionalization improves nanoparticle pene-

    tration into cells but produces no appreciable increase in

    particle accumulation in tumors. Huang et al.343 examined

    the biodistribution and localization of gold nanorods labeled

    with three types of probing molecules, including (1) an scFv

    peptide that recognizes EGFR; (2) an amino terminal frag-

    ment peptide that recognizes the urokinase plasminogen acti-

    vator receptor; and (3) a cyclic RGD peptide that recognizes

    the avb3 integrin receptor. The authors showed that wheninjected intravenously, all three ligands induce insignicant

    increases in nanoparticle accumulation in cell models and in

    tumors but greatly aect extracellular distribution and intra-

    cellular localization. They concluded that for PPTT, direct

    administration of particles to the tumor can be more eective

    than intravenous injection.

    The nal important question in current PPTT concerns

    eective delivery of radiation to a biological target. Because

    the absorption of biological tissue chromophores in the visible

    region is lower than that in the near-IR region by two orders

    of magnitude,294 the use of IR radiation radically decreases the

    nontarget heat load and enhances the penetration of radiation

    into the tissue interior. Nevertheless, the depth of penetration

    usually does not exceed 510 mm,84,311 so it is necessary to

    look for alternative solutions. One approach consists in using pulse

    (nanoseconds) irradiation modes in preference to continuous

    ones, which enables irradiation power to be enhanced without

    Fig. 11 Scheme for a PPTT employing active delivery of GNPs to

    cancer cells.

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online

  • 2268 Chem. Soc. Rev., 2012, 41, 22562282 This journal is c The Royal Society of Chemistry 2012

    increasing side eects. Another approach involves ber-optic

    devices for endoscopic or intratissue delivery of radiation.

    The strong and weak points of such an approach are evident.

    Finally, for hyperthermia, it is possible to use radiations with

    greater depths of penetration, e.g., radiofrequency346349 or

    nonthermal air plasma.350

    GNPs conjugated to antibiotics and antibodies have also

    served as photothermal agents for selective damage to protozoa

    and bacteria.351354 Information on certain issues in the use of

    PPTT can be found in several books and reviews.295,355359 Of

    particular note is the most recent comprehensive review by

    Kennedy et al.294

    In summary, gold nanostructures with a plasmon resonance

    oer considerable promise for selective PPTT of cancer and

    other diseases. Without doubt, several questions await further

    study, including the stability and biocompatibility of nano-

    particle bioconjugates, their chemical interaction in physiological

    environments, the period of circulation in blood, penetration

    into the tumor, interaction with the immune system, and

    nanoparticle excretion. We expect that the success of the initial

    stages of nanoparticle use for selective PPTT can be eectively

    enhanced at the clinical stage, provided that further studies are

    made on the optimal procedural parameters. In particular, one

    can mention the eorts of J. Feldmanns group360 related to

    thermoplasmonicsa eld that is not well understood yet and

    that is nowadays is a trend for hyperthermia and delivery upon

    light-to-heat conversion.

    3.2 Photodynamic therapy with GNPs

    The photodynamic method of treating oncological diseases

    and certain skin or infectious diseases is based on the application

    of light-sensitizing agents called photosensitizers (including

    dyes) and, as a rule, of visible light at a specic wavelength.361

    Most often, sensitizers are introduced intravenously, but contact

    and oral administration is also possible. The substances used

    in photodynamic therapy (PDT) can selectively accumulate in

    tumors or other target tissues (cells). The aected tissues are

    irradiated with laser light at a wavelength corresponding to the

    peak of dye absorption. In this case, apart from the usual heat

    emission through absorption,21 an essential role is played by

    another mechanism, related to the photochemical generation

    of singlet oxygen and the formation of highly active radicals,

    which induce necrosis and apoptosis in tumor cells. PDT also

    disrupts the nutrition of the tumor and leads to its death by

    damaging its microvessels. The major shortcoming of PDT is

    that the photosensitizer remains in the organism for a long

    time, leaving patient tissues highly sensitive to light. On the

    other hand, the eectiveness of dye use for selective tissue

    heating21 is low because of the small cross section of chromo-

    phore absorption.

    It is well known362 that metallic nanoparticles are eective

    uorescence quenchers. However, it has been shown recently363,364

    that uorescence intensity can be enhanced by a plasmonic

    particle if the molecules are placed at an optimal distance from

    the metal. In principle, this idea can improve the eectiveness

    of PDT.

    Several investigators have proposed methods for the delivery

    of drugs as part of polyelectrolyte capsules on GNPs

    (which decompose when acted upon by laser radiation and

    deliver the drug to the targets365,366) or by using nanoparticles

    surrounded by a layer of polymeric nanogel.367,368 Apart from

    that, the composition of nanoconjugates includes photoactive

    substances,369peptides (e.g., CALLNN), and proteins (e.g.,

    transferrin), which facilitate intracellular penetration.345,370,371

    Recently, Bardhan et al.372 suggested the use of composite

    nanoparticles, including, in addition to gold nanoshells, magnetic

    particles, a photodynamic dye, PEG, and antibodies. Finally,

    according to the data of Kuo et al.,373,374 nanoparticles

    conjugated with photodynamic dyes can demonstrate a synergetic

    antimicrobial eect, though the absence of such an eect has

    also been reported.375

    3.3 GNPs as a therapeutic agent

    In addition to being used in diagnostics and cell photothermolysis,

    GNPs have been increasingly applied directly for therapeutic

    purposes.29 In 1997, Abraham and Himmel376 reported success

    in colloidal gold treatment of rheumatoid arthritis in humans.

    In 2008, Abraham published a great body of data from a

    decade of clinical trials of Aurasolsan oral preparation for

    the treatment of severe rheumatoid arthritis.377 Tsai et al.378

    described positive results obtained when rats with collagen-

    induced arthritis were intraarticularly injected with colloidal

    gold. The authors explain the positive eect by an enhance-

    ment of antiangiogenic activity resulting from the binding of

    GNPs to vascular endothelium growth factor, which brought

    about a decrease in macrophage inltration and in inammation.

    Similar results were obtained by Brown et al.,379,380 who

    subcutaneously injected GNPs into rats with collagen- and

    pristane-induced arthritis.

    A series of papers by a research team from Maryland

    University have described the application of a colloidal gold

    vector to the delivery of TNF to solid tumors in rats.381384

    When injected intravenously, GNPs conjugated to TNF accu-

    mulated rapidly in tumor cells and could not be detected in

    cells of the liver, spleen, and other healthy animal organs. The

    accumulation of GNPs in the tumor was proven by a record-

    able change in its color, because it became bright red-purple

    (characteristic of colloidal gold and its aggregates), which

    was coincident with the peak of the tumor-specic activity

    of TNF (Fig. 12). The colloidal goldTNF vector was less

    toxic and more eective in tumor reduction than was native

    TNF, because the maximal antitumor reaction was attained

    at lower drug doses. A medicinal preparation based on the

    GNPTNF conjugate, which is called AurImmunet and

    intended for intravenous injection, is already in the third stage

    of clinical trials.

    In experiments in vitro and in vivo, Bhattacharya et al.385

    and Mukherjee et al.386 demonstrated the antiangiogenic

    properties of GNPs. The particles were found to interact with

    heparin-binding glycoproteins, including vascular permeability

    factor/vascular endothelial growth factor and basic broblast

    growth factor. These substances mediate angiogenesis, including

    that in tumor tissues, and inhibit tumor activity by changing

    the conformation of the molecules.387 Because intense angio-

    genesis (the formation of new vessels in organs or tissues) is a

    major factor in the pathogenesis of tumor growth, the presence

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online

  • This journal is c The Royal Society of Chemistry 2012 Chem. Soc. Rev., 2012, 41, 22562282 2269

    of antiangiogenic properties in GNPs makes them potentially

    promising in oncotherapy.388,389 The same research team has

    shown that GNPs enhance the apoptosis of chronic lympho-

    cytic leukemia cells resistant to programmed death390 and

    inhibit the proliferation of multiple myeloma cells.391

    Quite recently, Wang et al.392 revealed that PEG-coated

    gold nanorods have an unusual property: they can induce

    tumor cell death by accumulating in mitochondria and sub-

    sequently damaging them. Unexpectedly, for normal and stem

    cells, such an eect is either absent or less pronounced.

    4. GNPs as drug carriers

    4.1 Targeted delivery of anticancer drugs

    One of the most promising aspects of GNP use in medicine,

    currently under intense investigation, is targeted drug

    delivery.393395 The most popular objects for targeted delivery

    are antitumor preparations396 and antibiotics.

    GNPs have been conjugated to a variety of antitumor

    substances382411 listed in Table 4.

    Conjugation is done both by simple physical adsorption of

    preparations on GNPs and by using alkanethiol linkers. The

    action of the conjugates is evaluated both in vitro (primarily),

    with tumor cell cultures, and in vivo, with mice bearing implanted

    tumors of various nature and localizations (Lewis lung carcinoma,

    pancreatic adenocarcinoma, etc.). For creation of a delivery

    system, target molecules (e.g., cetuximab) are applied along

    with the active substance so as to ensure better anchoring and

    penetration of the complex into the target cells.399 It was also

    suggested that multimodal delivery systems be used,412 in

    which GNPs are loaded with several drugs (both hydrophilic

    and hydrophobic) and with auxiliary substances (target mole-

    cules, PDT dyes, etc.;413,414 Fig. 13). Most researchers have

    noted the high eectiveness of GNP-conjugated antitumor

    preparations.415

    4.2 Delivery of other substances and genes

    Besides antitumor substances, other objects employed to

    deliver GNPs are antibiotics and other antibacterial agents.

    Gu et al.416 prepared a stable vancomycincolloidal gold

    Fig. 12 Accumulation of the GNPTNF conjugate in the tumor after

    15 h. Diseased mice were intravenously injected with 15 mg of theGNPTNF vector. The belly images were obtained at the indicated

    times and show changes in tumor color within 5 h. The red arrow

    shows vector accumulation in the tumor, and the blue arrows mark the

    accumulation in the tissues around the tumor. Adapted from ref. 382

    by permisson from Wiley Interscience.

    Table 4 Antitumor substances conjugated with GNPs

    Drugs ParticlesMethods offunctionalization Auxiliary substances Cell lines or animals Ref.

    Paclitaxel GNSs, 26 nm PaclitaxelSH PEGSH, TNF MC-38; C57/BL6 mice implantedwith B16/F10; melanoma cells

    382, 2006

    Methotrexate GNSs, 13 nm Physical adsorption LL2, ML-1, MBT-2, TSGH 8301,TCC-SUP, J82, PC-3, HeLa

    397, 2007

    Daunorubicin GNSs, 5 nm,16 nm

    3-Mercaptopropionicacid as a linker

    K562/ADM 398, 2007

    Gemcitabine GNSs, 5 nm Physical adsorption Cetuximab(monoclonalantibodies)

    PANC-1, AsPC-1, MIA Paca2 399, 2008

    6-Mercaptopurine GNSs, 5 nm Physical adsorption K-562 400, 2008Dodecylcysteine GNSs,

    36 nmPhysical adsorption EAC 401, 2008

    5-Fluorouracil GNSs, 2 nm Thiol ligand MCF-7 402, 2009c,c,t-[Pt(NH3)2Cl2(OH)(O2CCH2CH2CO2H)]

    GNSs, 13 nm Amide linkages DNA HeLa, U2OS, PC3 403, 2009

    Cisplatin GNSs, 5 nm PEGSH as a linker Folic acid, PEGSH OV-167, OVCAR-5,HUVEC, OSE

    404, 2010

    Oxaliplatin GNSs, 30 nm PEGSH as a linker PEGSH A549, HCT116, HCT15,HT29, RKO

    405, 2010

    Kahalalide F GNSs, 20,40 nm

    Physical adsorption HeLa 406, 2009

    Tamoxifen GNSs, 25 nm PEGSH as a linker PEGSH MDA-MB-231, MCF-7, HSC-3 407, 2009Herceptin GNRs, lmax=760 nm 11-Mercaptoundecanoic

    acid as a linker BT474, SKBR3, MCF-7 408, 2009

    b-Lapachon GNSs, 25 nm Physical adsorption Cyclodextrin as a drugpocket, anti-EGFR,PEGSH

    MCF-7 409, 2009

    Doxorubicin GNSs, 12 nm Physical adsorption Folate-modied PEG KB 410, 2010Prospidin GNSs, 50 nm Physical adsorption HeLa 411, 2010

    Publ

    ished

    on

    30 N

    ovem

    ber 2

    011.

    Dow

    nloa

    ded

    by U

    nive

    rsid

    ade

    Tecn

    olog

    ica

    Fede

    ral d

    o Pa

    rana

    on

    25/0

    3/20

    15 1

    2:57

    :35.

    View Article Online

  • 2270 Chem. Soc. Rev., 2012, 41, 22562282 This journal is c The Royal Society of Chemistry 2012

    complex and showed its eectiveness toward various (including

    vancomycin-resistant) enteropathogenic strains of Escherichia

    coli, Enterococcus faecium, and Enterococcus faecalis. Similar

    results were presented by Rosemary et al.:417 a complex

    formed between ciprooxacin and gold nanoshells had high

    antibacterial activity against E. coli. Selvaraj and Alagar418

    reported that a colloidal gold conjugate of the antileukemic

    drug 5-uorouracil exhibited noticeable antibacterial and

    antifungal activities against Micrococcus luteus, Staphylococcus

    aureus, Pseudomonas aeruginosa, E. coli, Aspergillus fumigatus,

    and A. niger. Noteworthy is the fact that in all those cases, the

    drugGNP complexes were stable, which could be judged by the

    optical spectra of the conjugates.

    By contrast, Saha et al.419 (antibiotics: ampicillin, streptomycin,

    and kanamycin; bacteria: E. coli,M. luteus, and S. aureus) and

    Grace and Pandian420,421 (aminoglycoside antibiotics: genta-

    micin, neomycin, and streptomycin; quinolone antibiotics:

    ciprooxacin, gatioxacin, and noroxacin; bacteria: E. coli,

    M. luteus, S. aureus, and P. aeruginosa) failed to make stable

    complexes with GNPs. Nevertheless, those authors showed

    that depending on the antibiotic used, the increase in the

    activity of an antibioticcolloidal gold mixture, as compared

    to that of the native drug, ranged from 12 to 40%. From these

    data, it was concluded that the antibacterial activity of the

    antibiotics is enhanced at the cost of GNPs. However, the

    question as to the mechanisms involved in such possible

    enhancement remained unclaried, which was noted by the

    authors themselves. Burygin et al.422 experimentally proved

    that free gentamicin and its mixture with GNPs do not

    signicantly dier in antimicrobial activity in assays on solid

    and in liquid nutrient media. They proposed that a necessary

    condition for enhancement of antibacterial activity is the

    preparation of stable conjugates of nanoparticles coated with

    antibiotic molecules. Specically, Rai et al.423 suggested the

    use of the antibiotic cefaclor directly in the synthesis of GNPs.

    As a result, they obtained a stable conjugate that had high

    antibacterial activity against E. coli and S. aureus.

    Other drugs conjugated to GNPs are referred to much more

    rarely in the literature. However, some of those works deserve

    mention. Nie et al.424 demonstrated high antioxidant activity

    of GNPs complexed with tocoferol and suggested potential

    applications of the complex. Bowman et al.425 provided data

    to show that a conjugate of GNPs with the preparation TAK-

    779 exhibits more pronounced activity against HIV than the

    native preparation at the cost of the high local concentration.

    Joshi et al.426 described a procedure for oral and intranasal

    administration of colloidal-gold-conjugated insulin to diabetic

    rats, and they reported a signicant decrease in blood sugar,

    which was comparable with that obtained by subcutaneous

    insulin injection, Finally, Chamberland et al.427 reported a

    therapeutic eect of the antirheumatism drug etanercept con-

    jugated to gold nanorods.

    In conclusion, it is necessary to mention gene therapy, which

    can be seen as an ideal strategy for the treatment of genetic

    and acquired diseases.428 The term gene therapy is used in

    reference to a medical approach based on the administration,

    for therapeutic purposes, of gene constructs to cells and the

    organism.429 The desired eect is achieved either as a result of

    expression of the introduced gene or through partial or

    complete suppression of the function of a damaged or over-

    expressing gene. There have also been recent attempts at

    correcting the structure and function of an improperly func-

    tioning (sick) gene. In such a case, too, GNPs can serve as

    an eective means