You are on page 1of 12

Annals of Biomedical Engineering, Vol. 34, No.

1, January 2006 (
C 2006) pp. 314

DOI: 10.1007/s10439-005-9000-9

Engineering Luminescent Quantum Dots for In Vivo Molecular


and Cellular Imaging
ANDREW M. SMITH, GANG RUAN, MATTHEW N. RHYNER, and SHUMING NIE
Departments of Biomedical Engineering and Chemistry, Emory University and Georgia Institute of Technology, Atlanta, Georgia

(Received 16 January 2005; accepted 30 June 2005; published online: 1 February 2006)

AbstractSemiconductor quantum dots are luminescent and is currently under intense study across many disci-
nanoparticles that are under intensive development for use as a plines. For examples, electronics engineers aspire to fab-
new class of optical imaging contrast agents. Their novel proper-
ricate chips with nanometer-scale parts, physicists hope to
ties such as optical tunability, improved photostability, and mul-
ticolor light emission have opened new opportunities for imaging investigate the intersection between classical physics and
living cells and in vivo animal models at unprecedented sensitivity quantum mechanics, and chemists aim to study the inter-
and spatial resolution. Combined with biomolecular engineering actions of colloidal macromolecules. This broad interest
strategies for tailoring the particle surfaces at the molecular level, arises from the fact that many materials on the nanometer
bioconjugated quantum dot probes are well suited for imaging
length scale have unique properties that are not available
single-molecule dynamics in living cells, for monitoring protein
protein interactions within specific intracellular locations, and for from either discrete molecules or bulk solids. This fascinat-
detecting diseased sites and organs in deep tissue. In this article, ing length scale has also been exploited by nature, because
we describe the engineering principles for preparing high-quality proteins, nucleic acids, and other components of cells all
quantum dots and for conjugating the dots to biomolecular ligands. have nanoscale dimensions. It is thus not surprising that
We also discuss recent advances in using quantum dots for in vivo
the life sciences have been one of the first fields to ben-
molecular and cellular imaging.
efit from nanoscience and nanotechnology. For example,
semiconductor quantum dots (QDs) have been developed
KeywordsNanoparticles, Nanotechnology, Fluorescence, Liv-
as fluorescent labels for bioimaging applications. In this
ing cells, Living animals, Molecular imaging, Cytotoxicity,
review article, we examine the engineering principles of
Cationic peptides, Bioconjugation, Dynamic light scattering.
quantum dot synthesis and the use of quantum dot probes
in biomolecular imaging at a variety of length scales, from
the nanometer to micrometer resolution of living cells up
ABBREVIATIONS
to the centimeter to meter scale of entire organisms.
Quantum dots are nearly spherical semiconductor par-
FRET fluorescence resonance energy transfer
ticles with diameters of the order of 210 nanometers, or
PEG polyethylene glycol
roughly 20010,000 atoms. Their semiconducting nature
QD quantum dot
and their size-confinement properties have made them very
RES reticuloendothelial system
attractive for use in optoelectronic devices, biological detec-
TOP trioctylphosphine
tion, and also as fundamental prototypes for the study of col-
TOPO trioctylphosphine oxide
loids and the size-dependent properties of nanomaterials.
Bulk semiconductors are characterized by a composition-
INTRODUCTION dependent bandgap energy, which is the minimum energy
Scientists and engineers have developed devices and required to excite an electron to an energy level above its
technologies spanning a wide range of scales, from tow- ground state, commonly through the absorption of a photon
ering skyscrapers to microfabricated electronic circuits. A of energy greater than the bandgap energy. Relaxation of
notable gap, however, exists between micro-scale engineer- the excited electron back to its ground state may be accom-
ing and molecular-scale chemistry. This gap occurs in a size panied by photon emission. Because the bandgap energy is
range from a few nanometers to hundreds of nanometers, dependent on the particle size, the optical characteristics of
a QD can be tuned by adjusting its size.43 Figure 1 shows
the optical properties of CdSe QDs at four different sizes
Address correspondence to Shuming Nie , Departments of Biomedi-
cal Engineering and Chemistry, Emory University and Georgia Institute (2.2, 2.9, 44.1, and 7.3 nm in diameter).
of Technology, 1639 Pierce Drive, Suite 2001, Atlanta, Georgia 30322. In comparison with organic dyes and fluorescent pro-
Electronic mail: snie@emory.edu teins, QDs are about 20 times brighter, mainly due to their
3
0090-6964/06/0100-0003/0 
C 2006 Biomedical Engineering Society
4 SMITH et al.

between QDs and proteins also makes them biologically


relevant, and one of the first biological applications was the
use of QDs for protein detection.38

QD SYNTHESIS AND ENGINEERING

QD synthesis was first described in 1982 by Efros and


Ekimov,17,18 who grew nanocrystals and microcrystals of
semiconductors in glass matrices. Since their work, a wide
variety of synthetic methods for QDs have been described,
including preparation in aqueous solution at room tempera-
ture, synthesis at elevated temperature and pressure in an au-
toclave, and vapor-phase deposition on a solid substrate.4,11
Most syntheses yielding colloidal suspensions of QDs in-
volve the introduction of semiconductor precursors under
conditions that thermodynamically favor crystal growth, in
the presence of semiconductor-binding agents, which func-
tion to kinetically control crystal growth to maintain their
size within the nanoscale.
Because the size-dependent properties of QDs are most
pronounced when the nanoparticles are monodispersed,
there has been a drive to produce QDs with narrow size
distributions. Major progress toward this goal was made
in 1993 by Bawendis group, with the introduction of a
synthetic method for monodisperse QDs (<5% root-mean-
square in diameter) made from cadmium sulfide (CdS),
cadmium selenide (CdSe), or cadmium telluride (CdTe).42
Since this seminal report, the synthesis of CdSe QDs has
progressed rapidly, generating QDs that can span the visible
spectrum. As a result, CdSe has become the most common
chemical composition for QD synthesis, especially for bi-
FIGURE 1. Size-dependent optical properties of cadmium se- ological applications. Many techniques have been imple-
lenide QDs dispersed in chloroform, illustrating quantum con- mented to post-synthetically modify QDs for various pur-
finement and size tunable fluorescence emission. (A) Fluores-
cence image of four vials of monodisperse QDs with sizes
poses, such as coating with a protective inorganic shell,12,27
ranging from 2.2 to 7.3 nm in diameter. This image was ob- surface modification to render colloidal stability,23,25 and
tained with ultraviolet lamp illumination at 365 nm. (B) Flu- direct linkage to biologically active molecules.8,9 QD pro-
orescence spectra of the same four QD samples, excited at
400 nm. Narrow emission bands (2326 nm FWHM or full-width
duction has now progressed from a chemical science to an
at half-maximum) indicate narrow particle size distributions. elaborate molecular engineering process. The most com-
(C) Absorption spectra of the same four QD samples. Notice mon scheme involves four steps: (1) synthesis of the QD
that the onset of absorption is slightly blue-shifted from the
emission peak for each QD sample, and that the absorption
core, most often CdSe, in a high-temperature organic sol-
spectra are very broad, so that a wide spectrum can be used vent; (2) growth of an inorganic shell (usually zinc sulfide,
for excitation. Both the absorption and emission intensities ZnS) epitaxially on the core to protect the optical properties
are plotted in arbitrary units (AU).
of the QD; (3) phase transfer of the QD from organic liquid
phase to aqueous solution; and (4) linkage of biologically
large absorption cross sections, 1001000 times more stable active molecules to the QD surface to render functional-
against photobleaching, and show narrower and more sym- ity, or linkage of biologically inert polymers to minimize
metric emission spectra. In addition, a single light source biological activity. Each of these four steps is discussed
can be used to excite multicolor QDs, and the emission later.
wavelength can be tuned from the ultraviolet,64 throughout
the visible and near-infrared spectra,5,28,32,52 and even into
Core Synthesis
the mid-infrared.50 On the other hand, QDs are macro-
molecules that are an order of magnitude larger than or- The most effective and reproducible synthesis pro-
ganic dyes. For this reason, small dyes will be more ap- cedure for monodisperse QDs involves the addition of
propriate biological labels when the size of the fluorescent semiconductor precursors to a liquid coordinating solvent at
label must be minimized. However, the similarity in size high temperature, first described by Murray et al.42 The co-
Engineering Luminescent Quantum Dots for In Vivo Molecular and Cellular Imaging 5

ordinating solvent commonly consists of trioctylphosphine have used diverse molecular precursors for CdSe (cadmium
oxide (TOPO) and trioctylphosphine (TOP), which contain oxide, dimethylcadmium, and cadmium acetate, combined
basic functional groups that can bond to the QD surface with TOP-Se),42,48,53 various coordinating ligands (alky-
during growth to prevent the formation of bulk semicon- lamines and alkanoic acids) for enhanced monodispersity
ductors. The alkyl chains from coordinating ligands extend and quantum yield,53,57 and it has been found that the co-
away from the QD surface, rendering the QDs sterically ordinating solvent may be replaced with a noncoordinating
stable as colloids, dispersible in many nonpolar solvents. In solvent, like octadecene, containing a small amount of coor-
a typical synthesis of CdSe, room-temperature QD precur- dinating ligand.62 The vast number of potential parameter
sors, dimethylcadmium and elemental selenium dissolved combinations has allowed the simple synthesis of CdSe
in liquid TOP, are swiftly injected into hot (290350 C) QDs with diameters between 2 and 8 nm, with the emission
TOPO, immediately initiating nucleation of QD crystals. wavelengths (450650 nm) spanning the entire visible spec-
CdSe nucleation and growth is favored thermodynamically, trum with just one composition. By also adjusting the QD
because the precursors are introduced at concentrations well composition (ZnS, CdS, CdSe, CdTe, PbS, PbSe, and their
above the solubility of the resulting semiconductor. How- alloys), it is now possible to span the wavelength range
ever, crystal growth is kinetically controlled by monomer 4004000 nm.5,28,32,50,52,64 Adjusting the solvent charac-
diffusion, due to the high viscosity of the solvent (14 cp at teristics and initial precursor concentration has further re-
the melting point of TOPO, 50 C), and is also controlled sulted in nanocrystals with diverse shapes, like rods and
through the reaction rate of monomers at the QD surface, tetrapods.47
due to strong binding of the coordinating solvent with semi- When designing QD cores for a specific wavelength re-
conductor precursors and QD surfaces. A high temperature gion, the first choice is to select a chemical composition,
at injection overcomes the steric, kinetic barrier, allow- since high-quality QDs can only be obtained for a certain
ing precursor association and nucleation. The swift drop in wavelength range for each composition. For example, CdSe
temperature, combined with the drop in monomer concen- QDs may be tuned to emit between 450 and 650 nm, while
tration (due to the nucleation of many small QD crystals), CdTe QDs may be tuned to emit from 500 to 750 nm.
stops nucleation within seconds after injection, allowing The QD diameter is then chosen to determine the specific
even and homogeneous growth on similarly sized nuclei. wavelength of emission, and the QDs are then generated
This separation of nucleation and growth is responsible for through focused particle growth using the synthesis pa-
the monodispersity of the final QDs. Also the use of a hot rameters described earlier. The resulting QDs are coated
solvent yields semiconductor nanoparticles that are highly in coordinating ligands and suspended in a crude mix-
crystalline, while minimizing thermodynamically unfavor- ture of the coordinating solvent and molecular precursors.
able lattice defects. The QDs are highly hydrophobic, and can be isolated and
The initial step of nucleation is so rapid that it is difficult purified from the reaction mixture, either through liquid
to study experimentally, but the ensuing growth process liquid extraction (a mixture of hexane and methanol),63 or
is controllable and proceeds slowly, and distinct stages of through precipitation from a polar solvent (methanol or ace-
growth have been identified. After nucleation, remaining tone) that dissolves the reactants and coordinating ligands,
monomers grow epitaxially on the nuclei, and the QDs but not the QDs.42 The pure core QDs are then used as
reach a size-focusing point, at which the size distribution is substrates for further modification to generate biological
the narrowest.49 Interestingly, the quantum yield of the QDs probes.
also reaches a high value, at which the QDs in the reaction
reach a bright point, and afterward decrease in photo-
Shell Growth
luminescence efficiency.52 After this size-focusing point,
the monomers become depleted, and the size distribution Because QDs have high surface area to volume ratios,
widens (defocusing). A process known as Ostwald ripening a large fraction of the constituent atoms are exposed to the
also starts to occur, in which smaller particles are dissolved surface, and therefore have atomic or molecular orbitals
to grow on large particles.49 These growth phases are depen- that are not completely bonded. These dangling orbitals
dent on the monomer concentration in solution, and many may form bonds with organic ligands such as TOPO. This
parameters (temperature, each precursors initial concen- leads to an electrically insulating monolayer that serves
tration, and solvent composition) can be tuned to adjust the to passivate the QD surface by maintaining the inter-
size at which the QDs are focused. At the focusing point, it is nal lattice structure and protecting the inorganic surface
desirable to quench the reaction, usually by decreasing the from external effects.3 However, the bond strength between
temperature until crystal growth is negligible. This synthe- the organic ligand and the semiconductor surface atom is
sis procedure has been found to be the most versatile and re- typically much lower than the internal bond strength of
producible for QDs composed of CdSe, although QDs with the semiconductor lattice, and desorption of ligands makes
other compositions have similarly been synthesized in co- the core physically accessible. For this reason, it is ad-
ordinating solvents.5,58,65 New variations on this synthesis vantageous to grow a shell of another semiconductor on
6 SMITH et al.

the QD surface after synthesis. By using a shell of wider the hydrophobic QDs with amphiphilic polymers.16,21,61
bandgap than the underlying core, strong electronic insu- These methods yield QDs that can be dispersed in aqueous
lation results in enhanced photoluminescence efficiency, solution and remain stable for long periods of time due to
and a stable shell provides a physical barrier to degrada- a protective hydrophobic bilayer encapsulating each QD
tion or oxidation. As an example, to passivate CdSe QDs through hydrophobic interactions. No matter what method
with ZnS, the cores are purified to remove unreacted cad- is used to suspend the QDs in aqueous buffers, they should
mium or selenium precursors, and then resuspended in a be purified from residual ligands and excess amphiphiles
coordinating solvent.12 Molecular precursors of the shell, before use in biological assays, using ultracentrifugation,
usually diethylzinc and hexamethyldisilathiane dissolved dialysis, or filtration. Also, when choosing a water solu-
in TOP, are then slowly added at elevated temperatures. bilization method, it should be noted that many biological
The temperature for growth of ZnS on CdSe is chosen such and physical properties of the QDs may be affected by
that it is high enough to favor epitaxial crystalline growth, the surface coating, and the overall physical dimensions
but low enough to prevent nucleation of ZnS crystals and of the QDs are dependent on the coating thickness (Fig.
Ostwald ripening of CdSe cores. Normally, this is a tem- 2). Typically, the QDs are much larger when coated with
perature around 160220 C. The (core)shell (CdSe)ZnS amphiphiles, compared with those with a monolayer of
nanocrystals may then be purified just like the cores. Al- ligand.
though shell growth is a common procedure, uncapped
CdSe cores are also of great interest, especially for fluo-
rescence resonance energy transfer (FRET) applications, Bioconjugation
in which physical access to charge carriers in the core is Most water solubilization methods result in QDs covered
important. with carboxylic acid groups, and the QDs are negatively
charged in neutral or basic buffers. Many schemes used to
prepare QD bioconjugates rely on covalent bond forma-
Aqueous Solubilization tion between carboxylic acids and biomolecules (Fig. 3).9
Since the QD surface has a net negative charge, positively
Because the resulting QDs are coated with alkyl chains charged molecules can also be used for electrostatic bind-
that render solubility only in nonpolar organic solvents, ing, a technique that has been used to coat QDs with cationic
phase transfer is an essential and nontrivial step for the avidin proteins and recombinant maltose-binding proteins
QDs to be useful as biological reporters. Alternatively, fused with positively charged peptides.24,40 Alternatively,
QD syntheses have been performed directly in aqueous biomolecules containing basic functional groups, such as
solution generating QDs ready to use in biological envi- amines or thiols, may interact directly with the QD sur-
ronments,22 but these protocols rarely achieve the level of face as ligands.20 If biomolecules do not innately contain
monodispersity, crystallinity, and fluorescent efficiency as groups for direct QD binding, they may be modified to
the QDs produced in high-temperature coordinating sol- add this functionality. For example, nucleic acids and pep-
vents. Two general strategies have been used to make hy- tides have been modified to add thiol groups for binding
drophobic QDs soluble in aqueous solution (Fig. 2): lig- to QDs.1,41 Surface modification has also become modu-
and exchange, and coating with an amphiphilic polymer. lar through high-affinity streptavidinbiotin binding. QD
For ligand exchange, a suspension of TOPO-coated QDs streptavidin conjugates are convenient for indirect bind-
are mixed with a solution containing an excess of a heter- ing to a broad range of biotinylated biomolecules.61 If the
obifunctional ligand, which has one functional group that QDs are intended as nonfunctional probes, then they can be
binds to the QD surface, and another functional group that coated with inert hydrophilic polymers, such as polyethy-
is hydrophilic. Thereby, hydrophobic TOPO ligands are lene glycol (PEG), which act to reduce nonspecific adsorp-
displaced from the QD through mass action, as the new tion and to increase colloidal stability.7 Biocompatible QDs
bifunctional ligand adsorbs to render water solubility. Us- are now commercially available, conjugated to a variety of
ing this method, (CdSe)ZnS QDs have been coated with functional biological molecules, like streptavidin, biotin, or
mercaptoacetic acid and (3-mercaptopropyl) trimethoxysi- monoclonal antibodies.
lane, both of which contain basic thiol groups to bind to
the QD surface atoms, yielding QDs displaying carboxylic
acids or silane monomers, respectively.8,9 These methods APPLICATIONS IN CELLULAR AND
generate QDs that are useful for biological assays, but MOLECULAR IMAGING
ligand exchange is commonly associated with decreased
fluorescent efficiency and a propensity to aggregate and Much of our understanding of human physiology and
precipitate in biological buffers. More recently it has been pathology has resulted from a strict reductionist approach
shown that these problems can be alleviated by retaining to biology. Purified biological molecules are used to predict
the native TOPO molecules on the surface, and covering their behaviors in complex living cells, which are then stud-
Engineering Luminescent Quantum Dots for In Vivo Molecular and Cellular Imaging 7

FIGURE 2. Molecular engineering to modify the physical dimensions and chemical characteristics of QDs, using common surface
coatings. The first column shows QDs with a core diameter of roughly 5.5 nm, coated with different organic compounds, drawn
to scale to demonstrate the relative sizes of the QDs and their surface coatings. The second column shows a magnified view
of each surface to clarify the molecular interactions. The third column shows dynamic light scattering data from (CdSe)ZnS QDs
coated with each of the corresponding ligands, to show their effective hydrodynamic diameters. (A) QDs coated with TOPO ligands,
which bind to the QD surface via phosphine oxide functional groups.42 These hydrophobic QDs were dissolved in chloroform. (B)
TOPO-coated QDs were ligand exchanged with mercaptoacetic acid, which binds to the QDs via thiol functional groups, causing
carboxylic acid groups to extend outward from the surface.9 These anionic QDs were dissolved in phosphate buffered saline
(PBS). (C) TOPO-coated QDs were covered with an amphiphilic anionic polymer, 40% octylamine-modified poly(acrylic acid), which
wrapped around the hydrophobic surface, intercalating with the TOPO molecules, stabilizing the surfaces through hydrophobic
interactions.61 The resulting anionic QDs were then dispersed in PBS. (D) TOPO-coated QDs were encapsulated in amphiphilic
phospholipids, a mixture of 40% 1,2-dipalmitoyl-sn-diglycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000], and
60% 1,2-dipalmitoyl-glycero-3-phosphocholine.16 This QD coating was stabilized by similar effects as the polymer-coated QDs, but
after dissolution in PBS, steric stabilization and hydrophilicity of the resulting colloids was provided by long polyethylene glycol
(PEG) arms that extended outward from the QD surface. Data provided by A.M. Smith, M.N. Rhyner, and S. Nie. AU: arbitrary units.

ied in isolation to deduce their physiological roles in living cellular locations, which normally requires the extraction
multicellular organisms. This research paradigm has been of cells from organismal tissues, growth of these cells in
successful, but it is also limited because purified molecules culture, followed by sacrifice of the cells to test cellular ex-
may function differently in complex cells. For this reason, it tracts in standard and sometimes quantitative assays (e.g.,
is desirable to study molecules of interest within their native using gel electrophoresis or immunoassays) to determine
8 SMITH et al.

FIGURE 3. Schematic illustration showing the most common methods to conjugate QDs to biological molecules such as proteins,
peptides, nucleic acids, or small organic molecules. The QD used in this illustration is rendered water-soluble using mercaptoacetic
acid, which is drawn disproportionately large to clarify its role in conjugation. Similar coupling schemes have been used with other
surface modifications as shown in Fig. 2. One exception is that direct coupling to the QD surface is much more difficult if the
coating layer thickness is large, such as for QDs coated with silica or an amphiphilic polymer. Also electrostatic attraction will only
occur if the QD has a net charge. See text for descriptions of these strategies.

the presence and/or abundance of biomolecules. This ap- tivity, and robustness has been fluorescence microscopy.
proach results in the loss of time-dependent data and spatial Using this technique, the signal intensity from fluores-
information, that is, how the analyte under study changes cent molecules is used to determine the locations of fluo-
over time and where it exists within the heterogeneous rophores and their concentrations. Typically, biomolecules
structures of cells and tissues. Performing assays directly on are given a fluorescent tag (an organic dye or a fluorescent
dead cells and tissues, like immunocytochemical staining, protein), which allows detection of the biomolecule using
has been one approach to retain spatial information, but this a fluorescence microscope, or with a fluorescence imag-
technique still lacks the important time element. For this ing system for whole organisms. However, signal intensity
reason, it has been a long-standing goal of the biological from these tags quickly diminishes due to photobleaching,
sciences to study living cells and tissues in real time. As greatly reducing the signal-to-noise ratio over time, which is
spatial information is readily viewed using an imaging tech- why photostable quantum dots could significantly improve
nique, various types of microscopies have been developed real-time fluorescence detection in living cells and organ-
to image living cells with subcellular resolution, such as isms. Also QDs can be tuned to emit in the near-infrared
electron microscopy, fluorescence microscopy, optical ab- spectrum (>650 nm), which could allow unmatched
sorption microscopy, and magnetic resonance imaging. Of sensitivity and depth penetration through biological
these, the best combination of simplicity, resolution, sensi- tissue.
Engineering Luminescent Quantum Dots for In Vivo Molecular and Cellular Imaging 9

Living Cells blinking), demonstrating a remarkable level of sensitiv-


ity of protein detection on living cells. Protein diffusion
In 1998, two research groups simultaneously reported
was fluorescently monitored on the cellular membrane, and
that QDs could be used as fluorescent contrast agents
could even be visualized after the proteins were internal-
for labeling either living or fixed cells in culture.8,9 Fol-
ized. Dahan et al. similarly reported that glycine receptors
lowing these reports, interest in using QDs as reporters
on the membranes of living neurons could be stained with
in immunocytochemical and immunohistochemical assays
QDs coupled to an antibody fragment specific for the re-
quickly grew, due to the realization that QDs were su-
ceptor.13 Single receptors were identified, and tracking of
perior to conventional fluorescent dyes in photostability,
single QDs on the membranes allowed the calculation of
brightness, and spectral linewidth.56,61 Now it has been
protein diffusion constants. These conjugates showed supe-
established that QDantibody conjugates can be used to
rior photostability, lateral resolution, and sensitivity relative
label membrane proteins, cytosolic biomolecules, and nu-
to organic dyes.
clear antigens in fixed cells and tissues (Fig. 4).8,56,61
Detecting cellular membrane molecules on living cells
The use of QDs as live cell imaging agents has also
is very useful for many studies, but the ability to study
progressed, albeit at a slower pace due to the inherently
the complex internal machinery of living cells has been
greater difficulty of maintaining cellular viability during
a long sought-after goal. The primary method to study
staining.
proteins in their native environment has been through the
The main obstacle to QD imaging in living cells has been
expression of the protein of interest fused to a fluores-
the diffusion barrier of the cellular membrane. This is not
cent protein tag. Using this technique, the transgenic pro-
a problem if the antigen of interest resides on the plasma
tein is localized in its innate cellular location, and it may
membrane, as demonstrated by two reports in 2002. Lidke
be monitored through fluorescence microscopy. However,
et al. stained erbB/HER membrane receptors on cultured
photobleaching is commonly a problem, as is the inherent
human cancer cells using red-light emitting (CdSe)ZnS
difficulty in constructing the fusion protein. An alterna-
QDs coupled to epidermal growth factor, a small protein
tive approach is to couple fluorescent dyes to biomolecules
with a specific affinity for this receptor.36 Receptor-bound
of interest in vitro, and then introduce these conjugates
QDs were identified at the single-molecule level (single
into living cells. However, these probes must transverse
QDs may be distinguished from aggregates because the
the plasma membrane before finding their targets, and
fluorescent intensity from discrete dots is intermittent, or
also suffer from photobleaching. Using QDs in the place

FIGURE 4. Immunocytochemical stain of F-actin in fixed fibroblast cells using green quantum dots. Cells were first incubated with a
phalloidinbiotin conjugate, containing the small molecule, phalloidin, that has a specific affinity for F-actin. The biotin associated
with the F-actin was then detected by using QDstreptavidin conjugates emitting green fluorescence. This image was obtained
using an epifluorescence microscope and a 100 oil-immersion objective.
10 SMITH et al.

of dyes or proteins could alleviate photobleaching con-


cerns, and could also allow enhanced detection sensitivity
and spatial resolution. But a major problem still remains,
which is the delivery of QD probes across the plasma
membrane.
QDs have been loaded passively into cells by exploiting
the innate capacity of most cells to uptake their extracel-
lular space through endocytosis.26,31,46 This effect may be
enhanced by coupling the QDs to membrane receptors, in
part due to the higher local concentration of QDs in the
vicinity of the cellular membrane.15,31,36 However, these
methods invariably lead to sequestration of aggregated QDs
in vesicles, which are no longer free to find their intended in-
tracellular targets. Yet this is a simple way to label cells with
QDs, if specific intracellular localization is not necessary,
or if the intent is to target the endocytotic vesicles. Other
methods for internalization of QD-bioconjugates in living
cells were developed originally for the delivery of drugs FIGURE 5. Cellular uptake of QDcationic peptide conjugates.
Monolayers of living human cancer cells were incubated with
and biomolecules, and fall under two broad categories: red fluorescent (CdSe)ZnS QDs conjugated to TAT peptides,
chemical and mechanical delivery. Chemically-mediated which are cationic peptides derived from an HIV-associated
delivery enhances plasma membrane translocation by us- membrane translocation protein. This image was obtained us-
ing confocal microscopy, a 100 oil-immersion objective, with
ing cationic lipids,14,59 membrane-permeating proteins,9 or the focal plane near the middle of the cells. The aggrega-
cationic peptides14,21,34,39 all of which have been shown tion of QDs in internal cellular structures is indicative of their
to increase cellular uptake of many different molecules presence in endocytotic vesicles.
and particles (Fig. 5). Although the delivery mechanisms
are not well understood, these techniques seem to cause
QD aggregation inside of vesicles, similar to delivery by assay.46 The phagokinetic tracks of human breast cancer
endocytosis. Mechanical methods include microinjection cells were monitored through fluorescence microscopy,
of QDs into individual cells and electroporation of cells as the cells transversed a QD-coated substrate. The path
in the presence of QDs. Microinjection is the only tech- traveled by each cell became dark, and the cells increased in
nique that has been reported to deliver QDs homogeneously fluorescence as they took up more QDs. QDs have also been
into the cytoplasms of cells;14,16 however, this method is used to study the particle size dependence of endocytosis,
of low statistical value, as careful manipulation of single using the fact that the surface parameters of nanoparticles
cells prevents the use of large sample sizes. Electropora- may be altered to generate specific nanoparticle sizes
tion makes use of the increased permeability of cellular (Fig. 2).45
membranes under pulsed electric fields to deliver QDs,
but this method was reported to result in aggregation of
Living Animals
QDs in the cytoplasm.14 It is clear that if this field is to
progress, a greater understanding of this technical barrier Compared to studying living cells in culture, new
must be combined with new techniques to overcome this challenges arise with the increase in complexity to
problem. a multicellular organism and with the accompanying
In 2004, Derfus et al. demonstrated that QDs conjugated increase in size scale. Unlike monolayers of cultured cells
to organelle-targeting peptides could specifically stain and thin tissue sections, tissue thickness becomes a major
either cellular mitochondria or nuclei, following mi- concern because biological tissue attenuates most signals
croinjection into fibroblast cytoplasms.14 Similarly, Chen used for imaging. Therefore, the major imaging techniques
et al. targeted peptideQDs conjugates to cellular nuclei, used reliably for imaging whole animals rely on signals that
using electroporation to overcome the plasma membrane can transmit through thick tissue, or signals that transmit
barrier.10 These schemes have resulted in organelle-level differentially through specific types of tissue, using ultra-
resolution of intracellular targets for living cells, yielding sonic waves (ultrasound imaging), X-rays (computed X-ray
fluorescent contrast of vesicles, mitochondria, and nuclei, tomography), gamma rays (positron emission tomography),
but resolution on the scale of intracellular molecules has not or radio waves (magnetic resonance imaging). Optical
been reached. Although QD intracellular targeting is still imaging, especially fluorescence imaging, has been used
elusive, live cell labeling has already generated new assays. in living animal models, but it is still limited by the poor
The innate capacity of cells to uptake QDs via endocytosis transmission of visible light through biological tissue. It has
was utilized by Parak et al. to devise a cellular motility been suggested that there is a near-infrared optical window
Engineering Luminescent Quantum Dots for In Vivo Molecular and Cellular Imaging 11

in most biological tissue that is the key to deep-tissue the lymphatic system is another circulatory system that may
optical imaging.60 The rationale is that Rayleigh scattering be used to deliver QDs to specific tissues.33 Near-infrared
decreases with increasing wavelength, and that the major QDs were intradermally injected in mice and pigs. The
chromophores in mammals, hemoglobin and water, have lo- QDs were engulfed by dendritic cells, which migrated to
cal minima in absorption in this window. Few organic dyes sentinel lymph nodes, generating fluorescent contrast of
are available that emit brightly in this spectral region, and these nodes.
they suffer from the same photobleaching problems as their Cells can also be loaded with QDs in vitro, and then ad-
visible counterparts, although this has not prevented their ministered to an organism. The fluorescent tag can be used
successful use as contrast agents for living organisms.19 to identify the original cells and their progeny within the
One of the greatest advantages of QDs for imaging in organism. This was first demonstrated on a small organism
living tissue is that their emission wavelengths can be tuned scale by Dahan et al. in 2001, by microinjecting QDs into
throughout the near-infrared spectrum by adjusting their the cytoplasms of single frog embryos.16 As the embryos
composition and size, resulting in photostable fluorophores grew, the cells divided, and each cell that descended from
that are stable in biological buffers.33 Because visible QDs the original labeled cell retained a portion of the fluores-
are more synthetically advanced than near-infrared QDs, cent cytoplasm, which could be fluorescently imaged in real
most of the living animal studies implementing QDs have time under continuous illumination. In reports by Hoshino
used (CdSe)ZnS QDs that emit visible light. But even et al.30 and Voura et al.59 cells loaded with QDs were in-
visible QDs have shown great promise, due to their ability jected intravenously into mice, and their distributions in
to remain photostable and brightly emissive in living the animals were later determined through tissue dissec-
organisms. tion, followed by fluorescence imaging. Also Gao et al.
Quantum dots have been used to passively image the loaded human cancer cells with QDs, and injected these
vascular systems of various animal models. In a report by cells subcutaneously in an immune-compromised mouse.21
Larson et al., intravenously injected green-light emitting The cancer cells divided to form a solid tumor, which
QDs remained fluorescent and detectable when they circu- could be visualized fluorescently through the skin of the
lated to capillaries in the adipose tissue and skin of a living mouse. With recent reports that cells may be labeled with
mouse.35 This report made use of two-photon excitation, QDs at a high degree of specificity,34,39 it is foreseeable
in which near-infrared light is used to excite visible QDs, that multiple types of cells may be monitored in living
allowing for deeper penetration of excitation light, despite organisms, and also identified using their distinct optical
strong absorption and scattering of the emitted visible light. codes.
Lim et al. intravenously injected near-infrared (CdTe)CdSe The previous reports have demonstrated the capability
QDs to image the coronary vasculature of a rat heart.37 Both of using QDs in living animals, but have only demonstrated
of these reports utilized the native process of circulation to image contrast on the level of tissues and organs. Molec-
deliver fluorescent probes from a single location (the tail ular imaging is the generation of image contrast due to
vein) throughout the entire vasculature. The circulatory sys- the molecular differences in cells, tissues, and organs. This
tem is a useful means for delivering exogenous molecules, requires a probe that contains a molecular targeting func-
but the delivered substances have a limited lifetime of cir- tionality to generate contrast only in locations specified
culation due to uptake by vascularized organs. QDs, like by the targeted probe. In the past, mice have been geneti-
other nanoparticles, are taken up nonspecifically by the cally altered to express fluorescent proteins in specific cell
reticuloendothelial system (RES), comprising phagocytotic types, and targeted fluorophores have been administrated
cells such as macrophages, mainly located in the spleen, to animals to detect spatial differences in protein or gene
liver, and lymph nodes. Therefore, over time, intravenously expression. Using a similar technique, Akerman et al. used
administered QDs will end up in these organs, unless they QDpeptide conjugates to target specific vascular tissues.1
have some type of targeting functionality. Also long-term Microscopic fluorescence imaging of tissue sections from
studies of the vascular system using QDs must account for the mice demonstrated that the QDs were taken up by their
this finite lifetime in the blood stream, which could require target tissues, and nonspecific RES uptake could be min-
multiple injections of fluorescent probes. It was demon- imized via attachment of PEG to the QD surface. Whole-
strated by Ballou et al. that the lifetime of QDs in the animal imaging of molecular detection was realized by Gao
bloodstream of mice is significantly increased if the QDs et al. in 2004 using red fluorescent QDs conjugated to anti-
are coated with PEG polymer chains,7 an effect that has also bodies specific for antigens on a human cancer induced in a
been documented for other types of nanoparticles and small mouse.21 Tail-vein injection of targeted QD probes resulted
molecules, in part due to decreased nonspecific adsorption in specific uptake of the probes in the cancerous tissue,
of the nanoparticles, and decreased antigenicity.54 In this visualized using whole-body fluorescence imaging of liv-
report, RES uptake of PEG-coated red-fluorescent QDs af- ing mice, generating fluorescent image contrast due to the
ter intravenous injection in living mice was monitored in molecular difference between normal tissue and cancerous
real time using full-body imaging. Kim et al. demonstrated tissue.
12 SMITH et al.

Biocompatibility and Detection Specificity than small molecules, which makes it important to minimize
binding to background substrates. Nonspecific binding can
Fluorescent detection of the events in living cells and
often be minimized using hydrophilic polymers with a high
animals would not be very valuable if reporter molecules
entropy of solubilization. Polymers like PEG have been at-
significantly perturb the native biological processes. For
tached to the surface of QD probes for reducing nonspecific
this reason, one of the major questions surrounding the
binding.
use of QDs in living systems is whether they are pertur-
bative or cytotoxic. The most common QD composition,
(CdSe)ZnS, contains a core of cadmium and selenium,
both of which are known to be toxic. However, almost
all studies so far have reported negligible cytotoxicity of FUTURE DIRECTIONS
QDs.10,15,16,21,30,31,35,39,46,51,59 A ZnS shell, combined with Quantum dots have been received as technological
a robust organic coating, can protect the core well enough marvels with characteristics that could greatly improve bio-
to prevent oxidation and release of elemental cadmium logical detection. However, extensive engineering and fun-
and selenium, within the time course of an assay.15 In- damental studies are still needed. First, QD synthesis and
deed, the only reports mentioning moderate cytotoxicity post-synthesis nanoengineering must be perfected so that
in living cells utilized QDs coated with bifunctional cap- reproducible, robust, and highly specific biological probes
ping ligands,29,30,55 which are known to be colloidally un- can be generated for light emission in the visible spec-
stable due to constant desorption/adsorption of ligands.2 trum, and also in the biologically important near-infrared
This equilibrium of surface protective molecules may have spectrum. Second, their use in already existing assays us-
caused exposure of the underlying semiconductor material ing conventional fluorophores should be explored to deter-
and QD degradation, leading to the release of cadmium and mine if they can supplant dyes and fluorescent proteins, and
selenium. Further research on QD surface coatings and on whether they are potentially better labels, especially with
protection of the underlying semiconductor crystal will be respect to their inherently large sizes. Third, their spectral
important to ensure that QDs can be used as viable in vivo characteristics such as narrow emission bands, photostabil-
probes. ity, and multicolor emission, should be tested in biological
Besides cytotoxicity, it is also important that QDs do environments to determine if these unique characteristics
not interfere with other cellular or physiological processes, are truly useful. Once these tasks have been performed, QDs
either through nonspecific binding, by preventing spe- may be regarded as powerful tools for biological detection
cific binding through steric hindrance, or by changing and imaging.
biomolecule functionality or diffusivity. Nonspecific bind- There are also new biological applications of QDs that
ing is a problem that can be addressed through surface are promising. For example, QDs are inherently strong con-
coatings, as hydrophilic polymers such as PEG are known trast agents for electron microscopy due to their electron-
to reduce nonspecific interactions with particles. The fact dense nature, relative to biological organic molecules and
that QDs are on the same size scale as large proteins could water.44 Thereby, QDs are not only useful contrast agents
alter the ability of coupled biomolecules to bind to their for macroscale imaging of entire organisms, and micron-
targets, either by steric hindrance or by altering biomolecu- scale optical resolution using fluorescence microscopy, but
lar functionality. The large size of QDs would also decrease they can even be used to image at atomic-scale resolution
the rate of diffusion of attached biomolecules. If these prob- using electron microscopy. Also QDs have recently been
lems are significant, smaller sized QD probes will need to proposed as energy donors for photodynamic therapy, a pro-
be designed, or small molecule organic dyes may be used cess in which a photosensitizer within a cell absorbs light
instead of QDs. It is possible that the use of QDs as targeted and transfers this absorbed energy to generate a reactive,
imaging agents will become a nanoengineering problem, in cytotoxic oxygen species that is able to kill cancer cells.6
which the surface properties to the QD particles can be en- This concept, combined with the potential innate cytotoxic-
gineered for biological utility, as well as increased clinical ity of QDs, could result in dual-modality QD conjugates that
relevance. are both targeted imaging contrast agents and therapeutic
Detection specificity depends on the specific and non- agents. Furthermore, simple QDs may be combined with
specific interactions of QD probes with target molecules other nanomaterials to assemble small but complex multi-
and the surrounding environment. Specific biomolecular functional machines, much in the same way that biological
interactions are determined by the conjugated ligands such proteins and other macromolecules are assembled in cells
as antibodies, peptides, nucleic acids, or small molecules. to create multifunctional structures such as the ribosome.
By maximizing biomolecular binding affinity and minimiz- These devices could be used as multimodal imaging agents,
ing cross reactivity, the QD probe can have highly specific for instance, for simultaneous image contrast for magnetic
interactions. However, because QDs are nanoparticle col- resonance imaging and optical imaging, or even for combin-
loids, they have a higher degree of nonspecific interactions ing specific therapeutic agents with optical imaging agents.
Engineering Luminescent Quantum Dots for In Vivo Molecular and Cellular Imaging 13

ACKNOWLEDGMENTS 18
Ekimov, A. I., and A. A. Onushchenko. Quantum size effect in
the optical-spectra of semiconductor micro-crystals. Sov. Phys.
This work was supported by grants from the National Semicond. 16:775778, 1982.
19
Institutes of Health (P20 GM072069, R01 CA108468, and Frangioni, J. V. In vivo near-infrared fluorescence imaging. Curr.
R01 GM058173), the U.S. Department of Energy Genomes Opin. Chem. Biol. 7:626634, 2003.
20
Gao, X. H., W. C. W. Chan, and S. M. Nie. Quantum-dot
to Life Program, and the Georgia Cancer Coalition (GCC). nanocrystals for ultrasensitive biological labeling and multicolor
One of the authors (A.M.S.) acknowledges the Whitaker optical encoding. J. Biomed. Opt. 7:532537, 2002.
21
Foundation for generous fellowship support. Gao, X. H., Y. Y. Cui, R. M. Levenson, L. W. K. Chung, and S. M.
Nie. In vivo cancer targeting and imaging with semiconductor
quantum dots. Nat. Biotechnol. 22:969976, 2004.
22
REFERENCES Gaponik, N., D. V. Talapin, A. L. Rogach, K. Hoppe,
E. V. Shevchenko, A. Kornowski, A. Eychmuller, and H.
1
Weller. Thiol-capping of CdTe nanocrystals: An alternative to
Akerman, M. E., W. C. W. Chan, P. Laakkonen, S. N. Bhatia, and organometallic synthetic routes. J. Phys. Chem. B 106:7177
E. Ruoslahti. Nanocrystal targeting in vivo. Proc. Natl. Acad. 7185, 2002.
Sci. U.S.A. 99:1261712621, 2002. 23
Gerion, D., F. Pinaud, S. C. Williams, W. J. Parak, D.
2
Aldana, J., Y. A. Wang, and X. G. Peng. Photochemical insta- Zanchet, S. Weiss, and A. P. Alivisatos. Synthesis and prop-
bility of CdSe nanocrystals coated by hydrophilic thiols. J. Am. erties of biocompatible water-soluble silica-coated CdSe/ZnS
Chem. Soc. 123:88448850, 2001. semiconductor quantum dots. J. Phys. Chem. B 105:88618871,
3
Alivisatos, A. P. Perspectives on the physical chemistry of 2001.
semiconductor nanocrystals. J. Phys. Chem. 100:1322613239, 24
Goldman, E. R., E. D. Balighian, H. Mattoussi, M. K. Kuno, J.
1996. M. Mauro, P. T. Tran, and G. P. Anderson. Avidin: A natural
4
Alivisatos, A. P. Semiconductor clusters, nanocrystals, and bridge for quantum dot-antibody conjugates. J. Am. Chem. Soc.
quantum dots. Science 271:933937, 1996. 124:63786382, 2002.
5
Bailey, R. E., and S. M. Nie. Alloyed semiconductor quantum 25
Guo, W., J. J. Li, Y. A. Wang, and X. G. Peng. Luminescent
dots: Tuning the optical properties without changing the particle CdSe/CdS core/shell nanocrystals in dendron boxes: Superior
size. J. Am. Chem. Soc. 125:71007106, 2003. chemical, photochemical and thermal stability. J. Am. Chem.
6
Bakalova, R., H. Ohba, Z. Zhelev, M. Ishikawa, and Y. Baba. Soc. 125:39013909, 2003.
Quantum dots as photosensitizers? Nat. Biotechnol. 22:1360 26
Hanaki, K., A. Momo, T. Oku, A. Ko-
1361, 2004. moto, S. Maenosono, Y. Yamaguchi, and K.
7
Ballou, B., B. C. Lagerholm, L. A. Ernst, M. P. Bruchez, and Yamamoto. Semiconductor quantum dot/albumin complex is a
A. S. Waggoner. Noninvasive imaging of quantum dots in mice. long-life and highly photostable endosome marker. Biochem.
Bioconjug. Chem. 15:7986, 2004. Biophys. Res. Commun. 302:496501, 2003.
8
Bruchez, M., M. Moronne, P. Gin, S. Weiss, and A. P. Alivisatos. 27
Hines, M. A., and P. Guyot-Sionnest. Synthesis and characteri-
Semiconductor nanocrystals as fluorescent biological labels. zation of strongly luminescing ZnS-capped CdSe nanocrystals.
Science 281:20132016, 1998. J. Phys. Chem.. 100:468471, 1996.
9
Chan, W. C. W., and S. M. Nie. Quantum dot bioconjugates for 28
Hines, M. A., and G. D. Scholes. Colloidal PbS nanocrystals
ultrasensitive nonisotopic detection. Science 281:20162018, with size-tunable near-infrared emission: Observation of post-
1998. synthesis self-narrowing of the particle size distribution. Adv.
10
Chen, F., and D. Gerion. Fluorescent CdSe/ZnS nanocrystal Mater. 15:18441849, 2003.
peptide conjugates for long-term, nontoxic imaging and nuclear 29
Hoshino, A., K. Fujioka, T. Oku, M. Suga, Y. Sasaki, T. Ohta, M.
targeting in living cells. Nano Lett. 4:18271832, 2004. Yasuhara, K. Suzuki, and K. Yamamoto. Physicochemical prop-
11
Crouch, D., S. Norager, P. OBrien, J. H. Park, and N. Pickett. erties and cellular toxicity of nanocrystal quantum dots depend
New synthetic routes for quantum dots. Philos. Trans. R. Soc. on their surface modification. Nano Lett. 4:21632169, 2004.
Lond., Ser. A. 361:297310, 2003. 30
Hoshino, A., K. Hanaki, K. Suzuki, and K. Yamamoto. Appli-
12
Dabbousi, B. O., J. RodriguezViejo, F. V. Mikulec, J. R. Heine, cations of T-lymphoma labeled with fluorescent quantum dots
H. Mattoussi, R. Ober, K. F. Jensen, and M. G. Bawendi. to cell tracing markers in mouse body. Biochem. Biophys. Res.
(CdSe)ZnS core-shell quantum dots: Synthesis and character- Commun. 314:4653, 2004.
ization of a size series of highly luminescent nanocrystallites. J. 31
Jaiswal, J. K., H. Mattoussi, J. M. Mauro, and S. M. Simon.
Phys. Chem. B 101:94639475, 1997. Long-term multiple color imaging of live cells using quantum
13
Dahan, M., S. Levi, C. Luccardini, P. Rostaing, B. Riveau, and dot bioconjugates. Nat. Biotechnol. 21:4751, 2003.
A. Triller. Diffusion dynamics of glycine receptors revealed by 32
Kim, S., B. Fisher, H. J. Eisler, and M. Bawendi. Type-II quan-
single-quantum dot tracking. Science 302:442445, 2003. tum dots: CdTe/CdSe(core/shell) and CdSe/ZnTe(core/shell)
14
Derfus, A. M., W. C. W. Chan, and S. N. Bhatia. Intracellular heterostructures. J. Am. Chem. Soc. 125:1146611467,
delivery of quantum dots for live cell labeling and organelle 2003.
tracking. Adv. Mater. 16:961966, 2004. 33
Kim, S., Y. T. Lim, E. G. Soltesz, A. M. De Grand, J. Lee, A.
15
Derfus, A. M., W. C. W. Chan, and S. N. Bhatia. Probing the Nakayama, J. A. Parker, T. Mihaljevic, R. G. Laurence, D. M.
cytotoxicity of semiconductor quantum dots. Nano Lett. 4:11 Dor, L. H. Cohn, M. G. Bawendi, and J. V. Frangioni. Near-
18, 2004. infrared fluorescent type II quantum dots for sentinel lymph
16
Dubertret, B., P. Skourides, D. J. Norris, V. Noireaux, A. H. node mapping. Nat. Biotechnol. 22:9397, 2004.
Brivanlou, and A. Libchaber. In vivo imaging of quantum dots 34
Lagerholm, B., M. Wang, L. Ernst, D. Ly, H. Liu, M. Bruchez,
encapsulated in phospholipid micelles. Science 298:17591762, and A. Waggoner. Multicolor coding of cells with cationic pep-
2002. tide coated quantum dots. Nano Lett. 4:20192022, 2004.
17
Efros, A. L., and A. L. Efros. Interband absorption of light in a 35
Larson, D. R., W. R. Zipfel, R. M. Williams, S. W. Clark, M.
semiconductor sphere. Sov. Phys. Semicond. 16:772775, 1982. P. Bruchez, F. W. Wise, and W. W. Webb. Water-soluble quan-
14 SMITH et al.

tum dots for multiphoton fluorescence imaging in vivo. Science infrared emitting colloidal PbSe quantum dots. J. Am. Chem.
300:14341436, 2003. Soc. 126:1175211753, 2004.
36 51
Lidke, D. S., P. Nagy, R. Heintzmann, D. J. Arndt-Jovin, J. Pinaud, F., D. King, H. P. Moore, and S. Weiss. Bioactivation
N. Post, H. E. Grecco, E. A. Jares-Erijman, and T. M. Jovin. and cell targeting of semiconductor CdSe/ZnS nanocrystals with
Quantum dot ligands provide new insights into erbB/HER phytochelatin-related peptides. J. Am. Chem. Soc. 126:6115
receptor-mediated signal transduction. Nat. Biotechnol. 22:198 6123, 2004.
52
203, 2004. Qu, L. H., and X. G. Peng. Control of photoluminescence
37
Lim, Y. T., S. Kim, A. Nakayama, N. E. Stott, M. G. Bawendi, properties of CdSe nanocrystals in growth. J. Am. Chem. Soc.
and J. V. Frangioni. Selection of quantum dot wavelengths for 124:20492055, 2002.
53
biomedical assays and imaging. Mol. Imag. 2:5064, 2003. Qu, L. H., Z. A. Peng, and X. G. Peng. Alternative routes toward
38
Mahtab, R., J. P. Rogers, C. P. Singleton, and C. J. Mur- high quality CdSe nanocrystals. Nano Lett. 1:333337, 2001.
54
phy. Preferential adsorption of a kinked DNA to a neutral Roberts, M., M. Bentley, and J. Harris. Chemistry for peptide
curved surface: Comparisons to and implications for nonspecific and protein PEGylation. Adv. Drug Delivery. Rev. 54:459476,
DNA-protein interactions. J. Am. Chem. Soc. 118:70287032, 2002.
55
1996. Shiohara, A., A. Hoshino, K. Hanaki, K. Suzuki, and K. Ya-
39
Mattheakis, L., J. Dias, Y. Choi, J. Gong, M. Bruchez, J. Liu, mamoto. On the cyto-toxicity caused by quantum dots. Micro-
and E. Wang. Optical coding of mammalian cells using semi- biol. Immunol. 48:669675, 2004.
56
conductor quantum dots. Anal. Biochem. 327:200208, 2004. Sukhanova, A., M. Devy, L. Venteo, H. Kaplan, M. Artemyev,
40
Mattoussi, H., J. M. Mauro, E. R. Goldman, G. P. Anderson, V. V. Oleinikov, D. Klinov, M. Pluot, J. H. M. Cohen, and I.
C. Sundar, F. V. Mikulec, and M. G. Bawendi. Self-assembly of Nabiev. Biocompatible fluorescent nanocrystals for immunola-
CdSeZnS quantum dot bioconjugates using an engineered re- beling of membrane proteins and cells. Anal. Biochem. 324:60
combinant protein. J. Am. Chem. Soc. 122:1214212150, 2000. 67, 2004.
41 57
Mitchell, G. P., C. A. Mirkin, and R. L. Letsinger. Programmed Talapin, D. V., A. L. Rogach, A. Kornowski, M. Haase,
assembly of DNA functionalized quantum dots. J. Am. Chem. and H. Weller. Highly luminescent monodisperse CdSe and
Soc. 121:81228123, 1999. CdSe/ZnS nanocrystals synthesized in a hexadecylamine-
42
Murray, C. B., D. J. Norris, and M. G. Bawendi. Synthesis and trioctylphosphine oxide-trioctylphospine mixture. Nano Lett.
characterization of nearly monodisperse CdE (E = S, Se, Te) 1:207211, 2001.
58
semiconductor nanocrystallites. J. Am. Chem. Soc. 115:8706 Talapin, D. V., A. L. Rogach, I. Mekis, S. Haubold, A.
8715, 1993. Kornowski, M. Haase, and H. Weller. Synthesis and surface
43
Nirmal, M., and L. Brus. Luminescence photophysics in semi- modification of amino-stabilized CdSe, CdTe and InP nanocrys-
conductor nanocrystals. Acc. Chem. Res. 32:407414, 1999. tals. Colloids Surf. A 202:145154, 2002.
44 59
Nisman, R., G. Dellaire, Y. Ren, R. Li, and D. P. Bazett-Jones. Voura, E., J. Jaiswal, H. Mattoussi, and S. Simon. Tracking
Application of quantum dots as probes for correlative fluores- metastatic tumor cell extravasation with quantum dot nanocrys-
cence, conventional, and energy-filtered transmission electron tals and fluorescence emission-scanning microscopy. Nat. Med.
microscopy. J. Histochem. Cytochem. 52:1318, 2004. 10:993998, 2004.
45 60
Osaki, F., T. Kanamori, S. Sando, T. Sera, and Y. Aoyama. A Weissleder, R. A clearer vision for in vivo imaging. Nat. Biotech-
quantum dot conjugated sugar ball and its cellular uptake on the nol. 19:316317, 2001.
61
size effects of endocytosis in the subviral region. J. Am. Chem. Wu, X. Y., H. J. Liu, J. Q. Liu, K. N. Haley, J. A. Treadway, J. P.
Soc. 126:65206521, 2004. Larson, N. F. Ge, F. Peale, and M. P. Bruchez. Immunofluores-
46
Parak, W. J., R. Boudreau, M. Le Gros, D. Gerion, D. Zanchet, cent labeling of cancer marker Her2 and other cellular targets
C. M. Micheel, S. C. Williams, A. P. Alivisatos, and C. Larabell. with semiconductor quantum dots. Nat. Biotechnol. 21:4146,
Cell motility and metastatic potential studies based on quantum 2003.
62
dot imaging of phagokinetic tracks. Adv. Mater. 14:882885, Yu, M. W., and X. G. Peng. Formation of high-quality CdS
2002. and other IIVI semiconductor nanocrystals in noncoordinating
47
Peng, X. G. Mechanisms for the shape-control and shape- solvents: Tunable reactivity of monomers. Angew. Chem. Int.
evolution of colloidal semiconductor nanocrystals. Adv. Mater. Ed. 41:23682371, 2002.
63
15:459463, 2003. Yu, W. W., L. H. Qu, W. H. Guo, and X. G. Peng. Experimental
48
Peng, Z. A., and X. G. Peng. Formation of high-quality CdTe, determination of the extinction coefficient of CdTe, CdSe, and
CdSe, and CdS nanocrystals using CdO as precursor. J. Am. CdS nanocrystals. Chem. Mater. 15:28542860, 2003.
64
Chem. Soc. 123:183184, 2001. Zhong, X. H., Y. Y. Feng, W. Knoll, and M. Y. Han. Alloyed
49
Peng, X. G., J. Wickham, and A. P. Alivisatos. Kinetics of IIVI Znx Cd1x S nanocrystals with highly narrow luminescence spec-
and IIIV colloidal semiconductor nanocrystal growth: "Focus- tral width. J. Am. Chem. Soc. 125:1355913563, 2003.
65
ing" of size distributions. J. Am. Chem. Soc. 120:53435344, Zhong, X. H., M. Y. Han, Z. Dong, T. J. White, and W. Knoll.
1998. Composition-tunable Znx Cd1x Se nanocrystals with high lu-
50
Pietryga, J., R. Schaller, D. Werder, M. Stewart, V. Klimov, minescence and stability. J. Am. Chem. Soc. 125:85898594,
and J. Hollingsworth. Pushing the band gap envelope: Mid- 2003.

You might also like