Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

Chapter 15

Quantum Dots
Tyler Maxwell1, 2, Maria Gabriela Nogueira Campos2, 3, Stephen Smith1, 2, Mitsushita Doomra2, 4,
Zon Thwin1, 2 and Swadeshmukul Santra1, 2, 4, 5
1
Department of Chemistry, University of Central Florida, Orlando, FL, United States; 2NanoScience Technology Center, University of Central
Florida, Orlando, FL, United States; 3Institute of Science and Technology, Federal University of Alfenas, Poços de Caldas, Minas Gerais, Brazil;
4
Burnett School of Biomedical Science, University of Central Florida, Orlando, FL, United States; 5Department of Materials Science and
Engineering, University of Central Florida, Orlando, FL, United States

15.1 Introduction to quantum dots PbS) elements in the periodic table due to their semi-
conductor properties. Impurities can be purposely doped
Quantum dots (QDs) are ultrasmall size semiconductor into the crystal lattice to alter their optical properties.
nanocrystals made up of 100e10,000 atoms,1 within the Transition metal dopants, such as Mn2þ, Cu2þ, Fe2þ, Cr2þ,
size range of 1.5e10 nm. QDs exhibit size unique optical and Co2þ, are widely studied to modulate QD’s magnetic
properties due to changes in band gap energy caused by and optical properties.9 Advantages of doping can include
quantum confinement effects. On absorption of light, improved quantum yield (QY)10 and longer excited state
electrons are promoted from the valence band (lower lifetime.11 High QY gives better limits of detection of QDs
electronic energy state) to the conduction band (upper for imaging. Longer excited state lifetimes in doped QDs
electronic energy state), producing an electronehole pair, are beneficial for eliminating background fluorescence in
called an “exciton.” When the electron and hole recombine, biological systems.
energy is released in the form of a photon (radiative QDs have a high surface-to-volume ratio, which means
recombination). In bulk materials, the exciton can spread a large percentage of the atoms are located on the particle
out over the delocalized lattice. However, when the particle surface. For example, a 5 nm diameter particle would have
size falls under the Bohr radius, the energy required to w20% of its atoms on the surface, whereas a 20 nm par-
create an exciton increases. This effect is termed “quantum ticle would have w5% surface atoms.12 The surface
confinement,” and it is typically observed in ultrasmall size, dangling bonds or defects can serve as a nonradiative
crystalline, semiconductor materials. Smaller QDs possess recombination centers and reduce QY. This is due to
larger band gap energy thereby emitting photons of higher entrapment of excited state electrons and holes which
energy (blue shifted) and vice versa. Tunability of optical migrated to the nonpassivated QD surface. Nonradiative
properties of QDs is shown in Fig. 15.1. QDs have been recombination can be minimized by passivating the QD
synthesized with emissions ranging from near ultraviolet2 surface with an inorganic shell layer having a wide band
to infrared.3 gap (such as ZnS or ZnSe) to produce a core/shell (CeS)
QDs were first described by Ekimov and Onushenko in QD structure. In CeS QDs, excitons are more effectively
1981.4 In 1982, Efros and Efros5 postulated that quantum confined within the core, thereby improving QY.13,14
size effects cause the change in optical and optoelectronic However, mismatches in the lattice spacing of the core with
properties of nanoparticles. In 1984, Louis Brus pioneered shell material can result in an unstable structure. QDs have
the field of colloidal semiconductor nanocrystals6 for which been designed with multiple shell layers such as CdSe/CdS/
he was awarded the Kavli Prize in Nanotechnology in ZnS to minimize the lattice mismatch.15
2008. QDs were first introduced as biological probes in
1988.7 Their broad absorption spectrum, narrow tunable
emission, and increased photostability compared with
15.2 Quantum dot synthesis
organic dyes made them attractive materials for One of the most commonly studied techniques to produce
bioimaging. highly fluorescent QDs is known as the hot injection
QDs are synthesized from group IIeVI (CdSe, CdS, synthesis. In hot injection synthesis,16 a metal precursor,
ZnO, ZnS), IIIeV (GaN, GaP, InP), and IVeVI (PbSe, such as cadmium oleate or CdO, is dissolved in a

Nanoparticles for Biomedical Applications. https://doi.org/10.1016/B978-0-12-816662-8.00015-1 243


Copyright © 2020 Elsevier Inc. All rights reserved.
244 Nanoparticles for Biomedical Applications

surface defects by the thick layer of organic coating on the


surface of the QD. A limitation of this synthesis technique
is the requirement of expensive high-purity organic sol-
vents and stringent synthesis steps such as precise heating
and argon flow settings and recovery of QDs through
purification process. QDs produced by this method also
tend to have larger hydrodynamic diameters from the
thick hydrophobic coatings imparted by the coordinating
solvent.
Water-in-oil (W/O) microemulsion (also known as
reverse micelle) method is an alternative QD synthesis
method.18 A reverse microemulsion is the dispersion of
water in oil in which a surfactant is added to create
extremely small water droplets. W/O microemulsion pa-
rameters such as water-to-surfactant molar ratio are varied
to control the water droplet size.19 The size of the water
droplet has an effect on the particle size. Usually, particle
size increases with the increase of water droplet size but
other factors such as reaction time, intermicellular ex-
change rate, and concentration of reagents also play a
role.20,21 For QD microemulsion synthesis, water-soluble
salts are dissolved in two separate microemulsions, one
for the anion and one for the cation. The ions are not sol-
uble in the bulk oil phase and are therefore confined to the
dispersed water droplets. QD crystals are produced when
the two microemulsion are slowly mixed together at room
temperature (Fig. 15.3A). During mixing, water droplets
undergo collision and coalescence processes, resulting in
the formation of QDs within the water droplet. The W/O
microemulsion synthesis requires large amounts of high
purity oil and surfactant, making it expensive and not
environmentally friendly.
Solegel is another process for making metal oxide or
metal sulfideebased QDs at room temperature. This
method involves the formation of a colloidal solution called
FIGURE 15.1 (A) Image of different sized CdSe quantum dots (QDs)
excited by an ultraviolet lamp demonstrating the quantum confinement a “sol” from a metal precursor dispersed in a solvent, fol-
effect. (B) Fluorescence emission spectra of the same QD samples. (C) lowed by formation of a continuous network or “gel”
Absorption spectra of the QD samples. From Smith, AM, et al., Bio- (Fig. 15.3B). A stabilizing agent such as glutathione
conjugated quantum dots for in vivo molecular and cellular imaging. Adv (GSH), mercaptopropionic acid (MPA), or mercaptounde-
Drug Deliv Rev 2008;60(11):1226e1240.
canoic acid is incorporated in the reaction mix to control
the growth of QD.22 Inexpensive solvents such as ethanol23
coordinating organic solvent such as trioctyl phosphine or water can be used for this method, making it cost-
oxide (TOPO) or trioctyl phosphine. Coordinating sol- effective. Another advantage of using solegel synthesis is
vents are used to control the QD nucleation and growth as that less waste is produced making it more environmentally
they bind to the surface of the nucleating particles. An friendly than hot injection or microemulsion methods.
organic solution containing an anion source (for example, Major drawbacks for this method include broad size dis-
Se2 or S2) is then injected, followed by heating for a set tribution and high level of surface defects.
amount of time, with a longer growth time resulting in Pyrolysis and hydrothermal processes can be used to
larger QDs (Fig. 15.2). QD growth is halted by injection prepare inorganic or carbon QDs (CQDs). These methods
of cold acetone or methanol to precipitate the QDs. are one-pot synthesis procedures where the reactant pre-
Because the reagents are air sensitive, the whole reaction cursors are dissolved in water and heated at high temper-
is performed under argon flow. QDs produced by this ature (and high pressure for hydrothermal process). This
method are more fluorescent than those synthesized by heating partially decomposes chemicals and facilitates
other techniques due to the reduction and passivation of collisions, which cause QD formation (Fig. 15.3C). To
Quantum Dots Chapter | 15 245

FIGURE 15.2 Hot injection synthesis of CdSe/ZnS quantum dots. TOPO, Trioctyl phosphine oxide; TOP, Trioctyl phosphine. From Bailey, RE, A.M.
Smith, and S. Nie, Quantum dots in biology and medicine. Physica E (Amsterdam, Neth.), 2004;25(1):1e12.

prepare CQDs, organic precursors such as ammonium cit- 15.3 Quantum dot surface modification
rate,24,25 glucose,26 chitosan,27 or other carbon sources are
and bioconjugation
used. Nitric acid is commonly added during synthesis to
increase water dispersibility by oxidizing surface groups of After synthesis, many QDs have a hydrophobic coating
the CQDs to carboxyl groups. These methods are scalable, which must be replaced or further modified to give them
low-cost, and environmentally friendly.28 However, the aqueous dispersibility. A few common strategies for this
effect of the heat source on particle formation is still poorly include coating with hydrophilic thiols, silica, amphiphilic
understood.29 polymers, and polyethylene glycol (PEG) (Fig. 15.4).

FIGURE 15.3 Schematic representation of (A) microemulsion synthesis, (B) solegel synthesis, and (C) pyrolysis and hydrothermal syntheses.
246 Nanoparticles for Biomedical Applications

FIGURE 15.4 Types of coating materials used to functionalize quantum dot surface for future conjugation. PEG, polyethylene glycol.
Modified from Smith AM, et al., Bioconjugated quantum dots for in vivo molecular and cellular imaging. Adv Drug Deliv Rev,
2008;60(11):1226e1240.

Thiol groups can bind with both sulfur and metal atoms, help maintain repulsive forces between particles, which
such as zinc and cadmium on the QD surface. Small increase their stability during conjugation.
molecule coating agents containing thiol and carboxyl Sulfhydryl cross-linkers, such as maleimides and
groups are the most commonly used surface capping for pyridyl disulfides, can also be used for biomolecule
QDs. Thiols bind to the QD surface, while the carboxyl conjugation. The maleimideethiol reaction is selective to
groups are exposed to the solvent. At neutral pH, most thiol groups which are present on naturally occurring
carboxyl groups remain deprotonated, producing a nega- cysteine residues (Fig. 15.5A). The availability of
tively charged QD surface. Therefore, QDs in aqueous cysteine residue on the protein can be a limiting factor for
solution are stabilized via electrostatic repulsion. Zeta this conjugation strategy. Thiol groups can also be pur-
potential (z) is a measurement of overall particle surface posely introduced into a protein’s chain by the reaction of
charge (positive or negative). If the z value is more than primary amine groups with sulfhydryl reagents (i.e.,
20 mV, one can expect stable colloidal solution (no Traut’s reagent).
settlement over days) and therefore minimal aggregation Histidine tagging is another method for conjugating
of particles. However, one can still expect some aggre- proteins a QD. In this method, nickelenitrilotriacetate
gation at the particle level, which can be detected using complexes act as a bifunctional agent, which covalently
sensitive light scatteringebased measurements such as binds to the QD surface, while histidine-tagged proteins
dynamic light scattering. Surface chemistry plays an are chelated by the nickel ion. Coordination of histidine
important role in determining how the QD will interact residues to other metals is also possible due to histidine’s
with a biological system. For example, heavy-metal ion strong affinity to metals. For instance, zinc ions on the
toxicity of QD can be masked either by the shell layer or QD surface can serve as a histidine chelator for direct
by ligand capping with biomolecules. Conjugation with attachment of the biomolecule to the QD surface31
biomolecules can not only provide colloidal stability but (Fig. 15.5B).
also improve biocompatibility.8 Noncovalent-based conjugation of biomolecules to
One of the most common methods for attaching bio- QD surface is primarily based on Coulombic (electro-
molecules to QD surface is EDC (1-ethyl-3-(3- static) interactions. Charged QD surfaces can be electro-
dimethylaminopropyl) carbodiimide hydrochloride) statically assembled with oppositely charged ligands to
covalent coupling. In this method, carboxyl-modified QDs create stable conjugates. Usually, QDs are surface-
are activated with water-soluble EDC in buffer to generate modified with negatively charged molecules such as N-
a reactive EDC ester on the particle surface. This ester can acetyl cysteine (NAC) that minimizes particleeparticle
then react with the amine groups on the biomolecule to aggregation and produces a stable QD dispersion in
form a stable amide (peptide) bond between the QD and aqueous medium. This charged QD can interact with
the biomolecule. The EDC ester can also be reacted with positively charged proteins or modified immunoglobulins,
N-hydroxy succinimide (NHS) or sulfo-NHS to form an which act as a bridge to couple the QD to the biomolecule
NHS ester before reacting with amine-containing bio- of interest32 (Fig. 15.5C). Another noncovalent-based QD
molecules in a two-step conjugation. The advantage of coupling method takes advantage of the specific biomol-
using the two-step conjugation is that the NHS ester is ecular interaction between the protein streptavidin and the
more stable and gives higher yields than the initial EDC biotin molecule. The streptavidinebiotin system is one of
ester.30 In addition, the negatively charged sulfo-NHS can the strongest noncovalent biological proteineligand
Quantum Dots Chapter | 15 247

FIGURE 15.5 Schematic representation of different methods of conjugating biomolecules to quantum dots (QDs). (A) Covalent attachment of bio-
molecules through the amine, thiol, or carboxyl groups displayed on the QD surface. (B) Direct attachment (zero-length coupling) of biomolecules to
atoms on the QD surface, by either dative thiol bonding or metal affinityedriven coordination. (C) Electrostatic (noncovalent) interactions between QD
surfaces and oppositely charged proteins or other biomolecules. Modified from Sapsford KE, et al., Biosensing with luminescent semiconductor quantum
dots. Sensors 2006;6(8):925e953.

interactions known to date and is a widely used tool for versatility.34 Detection probes, immunoprobes, biosensors,
bioconjugation. and drug-delivery systems are some of the several appli-
cations of QDs in the biomedical area. Moreover, conju-
gation of QDs with ABs has recently captured interest due
15.4 Quantum dotebased antibody to the potential for targeted delivery, disease diagnosis, and
probes monitoring therapy.
Antibodies (ABs) or immunoglobulins are glycoproteins
mainly produced by plasma cells. ABs are “Y”-shaped 15.4.1 Antibody conjugation
proteins composed of four polypeptide chains: two light
Selection of bioconjugate chemistry has impact on the
chains and two heavy chains. Both heavy and light chains
stability and functional activity of the QD-AB probe.
are composed of antigen recognition domains. AB frag-
Recognition sites of ABs need to be preserved during
ments (nanobodies), such as the heavy chain and single
conjugation to maintain the specificity of binding to the
variable domain, can also be used as therapeutics, for site
target molecules, such as antigens. Molecular conformation
recognition and/or target detection due to their high spec-
is an important parameter that affects the specificity of the
ificity to the antigen. These properties make ABs suitable
reaction between antigen and AB, as well as the success of
for several biological applications, such as immunoassays
for diagnoses of diseases, bioimaging, biosensing, and for conjugation between QD and ABs. Direct attachment of
AB to QD surface using “zero-length” coupling chemistry
detection of toxins, biomarkers, and other molecules of
(such as cyanogen bromide based) increases the chances of
interest.
blocking the antigen-binding site, thus compromising AB
To prepare AB-based probes, a reporting molecule
performance.
(fluorophore) is attached to the AB. Organic dyes have
Han et al. synthesized norbornene-QD/tetrazine-AB
been used for this purpose, but QDs have been proposed as
conjugates for in vivo cytometric imaging in mice.35 Nor-
alternatives due to their attractive photophysical properties.
bornene, which is an uncharged bridging cycling hydro-
The use of QDs in probes for biological and clinical
applications has been considered advantageous because carbon, was incorporated to the QD to avoid nonspecific
binding to cells or biomolecules due to surface charge. The
of their interesting optical properties, small size, and
248 Nanoparticles for Biomedical Applications

authors reported passivation of the norbornene-QD surface maleimide-activated QDs with single-domain antibodies
with PEG-polyimidazole ligands; the copolymer enhanced (SDABs) through their cysteine residues.37 Tiwari et al.
the QY in aqueous solution before conjugation by cyclo- also studied the effect of conjugation chemistry on the
addition with tetrazine-modified ABs.35 Some of the ad- detection of HER2 expression in cancer cells, using anti-
vantages of their technique on synthesizing conjugates HER2 AB conjugated to CdSe/CdZnS QD.38 The authors
suitable for in vivo single cell imaging include high sta- modified the QD surface with GSH, a natural thiol com-
bility and QYs, minimal nonspecific interaction with cells pound that is present in most cells, to improve QD dis-
and serum, and narrow emission for multiplex cytometric persibility in aqueous medium. The thiol group of GSH
imaging.35 Fernandez-Arguelles et al. reported a simple binds to the QD surface, while its carboxyl group remains
bioconjugation of amphiphilic polymer-coated QD with deprotonated at neutral pH conditions. Negatively charged
ABs for immunoassays.36 The authors synthesized a QDs remain stable through electrostatic repulsion. Three
polymer based on poly(maleic anhydride) functional different coupling agents were tested, and the resulting
groups to improve stability and brightness of polymer- probes were evaluated in their ability to stain HER2 in
coated QD. Then, the carboxylic groups of the polymeric breast cancer cells (Fig. 15.6). The first and second con-
coating were cross-linked with the amine groups of ABs by jugations were nonselective covalent coupling reactions. In
water-soluble EDC chemistry.36 The results showed the the first method, the primary amines of anti-HER2 AB were
ability of a highly fluorescent, polymer-coated QD-AB to reacted with the carboxyl groups of GSH-coated QD via
selectively recognize an antigen/toxin. This approach can EDC/sulfo-NHS coupling. The second conjugation
be used to develop multiplexed immunoassays using occurred by the reaction of maleimide groups of succini-
several types of ABs conjugated with different sized QDs midyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate
that emit at different wavelengths.36 (SMCC)ecoupled GSH-QD and the iminothiolane-
Sensitivity and selectivity are important for early modified anti-HER2 ABs. Third conjugation was specif-
detection of diseases, such as cancer. Human epidermal ically between sulfhydryl groups of the reduced ABs and
growth factor receptor 2 (HER2) is a widely used the maleimide groups of SMCC-coupled GSH-QD. The
biomarker for the detection of several types of cancer, specific conjugation of the QD with the thiol group of ABs
including lung and breast cancers. Several studies reported resulted in a more stable conjugate with proper orientation.
the conjugation of anti-HER2 ABs to QDs for cancer di- A desired orientation is one in which the active site of the
agnostics. Rakovich et al. prepared highly sensitive, ul- AB is pointed out and away from the QD, so it is accessible
trasmall, and bright nanoprobes for detection of HER2 to the antigen. If the antigen-binding site is coupled to the
biomarker in lung and breast cancer cells by conjugating QD, it may become inaccessible due to steric effects.

FIGURE 15.6 Schematic representation for the coupling reactions between glutathione-quantum dots and antiehuman epidermal growth factor receptor
2 antibodies. (A) EDC/sulfo-N-hydroxy succinimide, (B) iminothiolate/sulfo-SMCC, and (C) sulfo-SMCC coupling. Modified from Tiwari DK, et al.,
Synthesis and characterization of anti-HER2 antibody conjugated CdSe/CdZnS quantum dots for fluorescence imaging of breast cancer cells. Sensors
2009;9(11):9332e9354.
Quantum Dots Chapter | 15 249

Orientation of the AB is controlled through the functional interaction between surficial negative charges of QD and
groups used for attachment to the QD. HER2 may contain positive charges of the leucine zipper attached to the IgG.32
many amine groups near its binding site and thus when According to the authors, synthesis of QD-IgG is simple,
these groups are coupled to the QD, the antigen binding is reproducible, and readily formed. They also claimed QD-
compromised. With the AB coupled via thiol groups, the IgG conjugates can be used for several immunoassays
probe demonstrated superior detection of HER2 expression due to their optical properties, such as resistance to pho-
in cancer cells.38 Therefore, conjugation chemistry plays an tobleaching and wide emission wavelengths.32
important role in the specificity and sensitivity of AB-QD
conjugates. 15.4.3 Factors controlling antibody loading to
quantum dot surface
15.4.2 Compact quantum dotdantibody probe
The number of ABs attached to the surface of a QD will
design
depend on the QD surface functionalization, bioconjugation
Direct conjugation of an AB to a QD surface produces a chemistry, orientation of ABs, and size of the QD-AB
compact probe. Wu et al. used the direct conjugation conjugate.
approach to immobilize anti-HER2 ABs to the ZnS QD Recently, Umakoshi et al. reported for the first time
surface. Histidine-modified ADAPTs (albumin-binding, the visualization of QD-AB conjugation using high-speed
domain-derived affinity proteins) were self-assembled on atomic force microscopy.41 They were also able to count
Zn on the QD surface. This method allows for a very close the number of ABs bound to a QD (Fig. 15.7), which in
proximity between the QD and AB by self-assembly of the past was only possible by indirect methods or esti-
ADAPTs on the QD surface, resulting in smaller mative calculations.42 Conjugation was achieved by
conjugates.39 reacting a maleimide-functionalized QD with thiol groups
Liu et al. studied the conjugation of biotin-coated QD of ABs. To expose thiol groups before conjugation, the
with single-domain AB-rhizavidin (RZ) fusions. RZ is a AB molecules were treated using a reducing agent tris(2-
protein with affinity for biotin, as well as streptavidin. carboxyetheyl)phosphine (TCEP). This methodology
Fusion of SDABs with biotin-affinity proteins is another leads to small size structures and well-oriented QD-AB
strategy for direct conjugation of ABs to biotin-coated conjugation. The authors observed ABs split in two
QDs. The authors reported that RZ-SDABs conjugate steps, as well as the formation of an intermediate state for
structure is smaller than the streptavidin one, because RZ is conjugation with QD.41
organized as a homodimer. Streptavidin is a larger quater- Pathak et al. reported the use of quantitative electro-
nary protein (tetramer). The advantages of using RZ- phoresis experiments to derive the number of IgG ABs
SDABs fusion include oriented binding to biotin-coated conjugated to the QD.42 The authors used both covalent
QD and efficient packing. This improves the active sur- (maleimide based) and noncovalent (streptavidinebiotin
face without compromising antigen recognition.40 based) coupling methods to prepare QD-IgG.42 Interest-
Goldman et al. reported the conjugation of QD with ingly, the authors found very low number of ABs per QD,
immunoglobulin G (IgG) using another type of engineered and the results were much lower for covalent conjugation.
adaptor protein. The authors added a positively charged It was suggested that orientation of the AB molecule plays
leucine zipper to the C-terminus of a two-domain recom- an important role in bioconjugation. Moreover, covalent
binant protein. The leucine zipper is a heptad repetition of conjugation using maleimide chemistry requires pretreat-
leucine residues that facilitates proteineprotein interactions ment of ABs to expose bioavailable thiol groups, which
by forming parallel helixehelix association. These residues may compromise the AB functionality. However, IgG
were introduced into the IgG by genetic fusion for further attachment to the QD surface using streptavidinebiotin
conjugation with the DHLA-coated QD by electrostatic interaction increased the yield of functional ABs.42

FIGURE 15.7 High-speed AFM


analysis of the number of attached anti-
bodies (ABs) on quantum dots (QDs). (A)
e(C) H-S AFM images of QD-ABs con-
jugates. (D) Histogram for the number of
attached ABs on QDs. Modified from
Umakoshi T., et al., Quantum-dot antibody
conjugation visualized at the single-
molecule scale with high-speed atomic
force microscopy. Colloids Surfaces B
Biointerfaces 2018;167:267e274.
250 Nanoparticles for Biomedical Applications

QD-AB conjugates have some advantages for in vivo biomolecules.33,61,65e71 Typically, QD biosensors are
cell-labeling studies, such as broad absorption, narrow designed with a fluorescence quencher bound to the QD in
emission, and large multiphoton cross sections. Long-term, a way that the analyte of interest can remove or change the
deep tissue tracking and sensing experiments may benefit quencher so that the QD fluorescence is restored. When the
from the use of near-infrared (NIR)eemitting QDs.35 quencher is bound to the QD, it exhibits low fluorescence
Organic NIR dyes have limited QY and photostability intensity or “OFF” state. When the quencher is removed,
compared with QDs.43 Controlling AB orientation on the the system is shifted to a high fluorescence or “ON” state
QD surface remains a challenge because the surface prop- (Fig. 15.8). This “OFF/ON” design is advantageous over
erties of QDs vary based on their preparation. The size of “always ON” probes because these probes have higher
QD-AB probes may also impact their usefulness as larger signal-to-noise ratio, lower detection limit, and are capable
probes may not be able to reach desired target due to steric of real-time detection in living organisms.72,73
effects. On the other hand, for ex vivo assays, where size of
probe is not a concern, QD-ABs conjugates can be 15.5.1 Quantum dot pH sensors
designed using different types of ABs for simultaneous
detection of biomarkers for a number of diseases, and/or for Many diseases such as cancer and Alzheimer’s can cause
more accurate/sensitive detection of diseases at early stage. unidirectional changes in pH to more acidic levels of the
cellular environment and/or organelles. Change in pH is
observed in inflamed tissue (e.g., cancer tissue) due to
15.5 Quantum dot biosensors increased rates of glycolysis of those cells.74,75 As this pH
Biosensors are systems engineered to produce a distin- change is universal trend for these diseases, sensing pH
guishable signal in response to a specific analyte in a changes at the cellular level has been studied for diagnostic
complex biological matrix. Fluorescence probes are useful purposes.
tools for biosensing due to their fast response times and QD-based pH sensors can be synthesized by attachment
in situ nondestructive sample analysis. There has been of a ligand with pH-dependent redox properties to the QD to
extensive research using fluorescent dyes and nano- control the fluorescence. Mattoussi et al. accomplished this
materials as biosensors, but more sensitive biosensing by binding dopamine (neurotransmitter) to the surface of
probes are still needed to help understand complex bio- CdSe/ZnS QDs. Dopamine isothiocyanate was attached to
logical interactions. QDs have been employed to improve amine-terminated PEG on the QD surface. Dopamine exists
the sensitivity and selectivity of the detection of bio- in its reduced catechol form at low pH and oxidized quinone
molecules. QDs offer advantages over other nanomaterials, at high pH. The quinone state can accept electrons from the
in that their emission can be tuned to allow better spectral excited QD to quench the QD’s fluorescence. The authors
overlap with quencher molecules for improved sensitivity. show the QD-dopamine probe was sensitive to pH changes
Improved photostability over organic dyes and proteins is between 4 and 10 (Fig. 15.9). The QD probe was injected
another attractive QD property because it facilitates long- into COS-1 (fibroblast) cells to test intracellular pH-sensing
term biosensing or tracking applications. capabilities. An increase in fluorescence over time was
QD biosensors have been developed to sense various observed as the pH of the endosomes decreased.46,47
biologically relevant stimuli, such as pH changes,44e54 Ratiometric fluorescence sensors combine an analyte-
reactive oxygen species (ROS),55e59 metal ions,60e64 and sensitive fluorophore and an analyte-insensitive fluorophore

FIGURE 15.8 Scheme showing


the design of quantum dotebased
biosensors.
Quantum Dots Chapter | 15 251

FIGURE 15.9 Schematic design of pH-


responsive quantum dotedopamine probe
(top) and a photograph of probe fluorescence
emission in the pH range of 4e10 (bottom).
From Ji X, et al., On the pH-dependent
quenching of quantum dot photo-
luminescence by redox active dopamine. J
Am Chem Soc 2012;134(13):6006e6017.

(usually the QD) to obtain information on the analyte. Infor- into cell. Transport of QDs to areas outside the endosome is
mation is obtained based on the fluorescence intensity ratio of still a challenge limiting their usefulness.
the two fluorophores. Ratiometric sensors have advantages
over redox-based sensors, in that they provide probes with a 15.5.2 Quantum dot metal ion sensors
self-calibration ability to improve their sensitivity and selec-
tivity. The concentration of the probe can be calculated from The sensing of metal ions is vital to understanding many
the insensitive fluorophore, and the pH-sensitive fluorophore different diseases and metabolic processes. To obtain sub-
would report on pH change. For sensing pH in the range from cellular information on metal ions, it is necessary to use
6 to 8, Miyazaki et al. attached fluorescein isothiocyanate either metal ion selective electrodes or dyes. Electrodes are
(FITC) to GSH-capped CdTe/ZnS QDs.76 The QY of fluo- limited by their large size which results in poor spatial
rescein is pH sensitive, it decreases as the carboxyl groups resolution. Optical-based probes are well suited for sub-
(pKa 6.4) are protonated. cellular visualization of metal ions with fluorescent detec-
Sensing pH in the endosome and lysosome compart- tion being the most utilized. Organic dyes modified with
ments requires probes to have a working pH range metal-binding groups are widely available to report on
between 4 and 6. Todd et al. designed an acid reactive intracellular metal ion concentrations. QDs with metal-
probe by attaching a thiol-modified dye (2-(dimethyla- binding ligands can be used in place of these organic
mino) ethyl) aminonaphthalimide to CdSe/ZnS QDs. dyes resulting in probes that are more photostable. Cyclic
The tertiary amine group on the dye shifts the fluores- ethers (crown ethers) and azamacrocycles (aza-crown) have
cence spectra when protonated. Energy transfer from the been attached to QDs to create metal ion sensors. These
dye to QD occurs at acidic pH enhancing QD cyclic compounds are electron rich and thus quench the QD
fluorescence.51 fluorescence through electron transfer (Fig. 15.10). When
In conclusion, QD pH sensors have been synthesized the crown ether is bound to a metal cation, its ability to
with the capability of measuring physiologically relevant quench the QD is reduced.77
pHs at the cellular level such as those in the endosome. A zinc ion sensor was synthesized by Hall et al. by
Because of their good photostability, these QDs may be linking an azamacrocycle (cyclam or cyclen) to MPA-
useful for longer duration studies. Currently, cell membrane capped CdSe/ZnS QDs by carbodiimide coupling.
permeable pH-reactive dyes are the most effective at Cyclam was able to quench the QD fluorescence through
quantifying intercellular pH as they can easily penetrate electron transfer. When bound to a Zn2þ ion, cyclam’s

FIGURE 15.10 Mechanism of quantum dot zinc ion sensor. Based on Ruedas-Rama MJ, Hall EAH. Azamacrocycle activated quantum dot for zinc ion
detection. Anal Chem (Washington, DC, U S), 2008;80(21):8260e8268.
252 Nanoparticles for Biomedical Applications

molecular orbitals were no longer available for electron dependent. On protonation (pH < 6) of the imidazole group,
transfer. The authors showed that QD fluorescence intensity its interaction with the particle’s surface is destabilized.
responded linearly to Zn2þ ion concentration, and fluores- The polyphosphate backbone of DNA can be modified
cence was again quenched after applying strong chelating to phosphorothioates, where a nonbridging oxygen atom is
agent (EDTA) which would remove bound Zn2þ ions. This replaced with sulfur (Fig. 15.12). When interacting with
shows that the Zn2þ binding was reversible and the sensor cadmium-containing QDs, the affinity of sulfur for Cdþ2 is
can be reused. There was little interference from Group 1 approximately 3000 times greater than that of oxygen.
and 2 metals (Na, K, Ca, Mg); however, copper, cobalt, and Successful conjugation of phosphorothioate-modified
iron all bind very strongly to the ZnS shell causing DNA molecules to many types of QDs has been
interference.64 reported.83e85 However, the conjugated DNA will no
longer hybridize complementary DNA due to conforma-
tional distortion.83 To overcome this limitation, a DNA
15.6 Quantum dotdDNA probes overhang with a phosphodiester backbone which contains
In clinical diagnostics, the ability to rapidly detect or the target sequence may be added to the phosphorothioate-
sequence low concentrations of DNA is critical for the early modified DNA to preserve functionality in biosensing
diagnosis of many diseases such as cancer. QD biosensors applications.86
have reported improved detection limits compared with Phospholipids self-assemble on the surface of the QDs,
traditional molecular assays. These low limits are achieved allowing polar groups to promote water solubility of the
due to the QDs’ very high QY and their capacity to QDs. The resulting lipid encapsulated QDs may be opti-
immobilize multiple recognition biomolecules. The hy- mized to display functional groups capable of conjugating
bridization of a single target DNA molecule at the surface is DNA by controlling the composition of encapsulating
capable of recruiting multiple reporter probe molecules. phospholipids used (Fig. 15.13).87,88 This conjugation
QD-DNA/RNA constructs have been demonstrated to strategy requires the QD to possess a hydrophobic shell
successfully monitor the delivery of therapeutic agents like (e.g., QD coated with TOPO). These hydrophobic QDs may
small interfering RNA (siRNA) to cells.78e80 then be encapsulated by an amphiphilic compound such as
1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC). Lipid-
15.6.1 Quantum DoteDNA noncovalent conjugated oligonucleotides are then added to amphiphilic
DOPCecoated QD. Oligonucleotides are thus immobilized
conjugation strategies
onto QDs through hydrophobicehydrophobic interaction,
The method of conjugation of the nucleic acid to the QD is forming a stable conjugate.89 This method does suffer from a
important to consider when developing biosensors. Strate- lack of control regarding loading efficiency. In addition,
gies for conjugating DNA to QDs via noncovalent coupling these phospholipid formulations are generally poor at
may be categorized into two groups: affinity and biological delivering cargo to the cytoplasm and crossing the cellular
interactions. lipid bilayer.90e92
Imidazoles carry high affinity toward the surface of QDs The negatively charged phosphate backbone of DNA
and can be employed for anchoring groups onto the QD may also adsorb QDs with positively charged surfaces by
surface. To utilize this affinity in conjugation strategies, electrostatic interactions. QDs with positively charged
DNA may be tagged with polyhistidine peptides coatings can be loaded with DNA by simply mixing the
(Fig. 15.11).81,82 However, the affinity of the imidazole two together. However, interactions of the DNA with
functional group toward the particle surface is pH the QD in this manner may distort the conformation of the

FIGURE 15.11 Imidazole-mediated conjugation. From Medintz IL, et al., A reactive peptidic linker for self-assembling hybrid quantum DotDNA
bioconjugates. Nano Lett 2007;7(6):1741e1748.
Quantum Dots Chapter | 15 253

15.6.2 Quantum DoteDNA covalent


conjugation strategies
Covalent conjugation strategies are also used to function-
alize QDs with DNA. Covalent conjugation not only in-
creases the QD-DNA stability but also provides flexibility
for controlling the orientation of DNA on the particle sur-
face. Limitations of covalent binding of DNA include
tedious sample preparation and purification processes that
often involve multiple steps. Carboxyl-modified QDs can
be linked to amine-functionalized DNA via carbodiimide
coupling chemistry which forms a stable amide bond be-
tween the QD and DNA. The conjugation strategy gener-
ally results in minimal size increase of the QDs. However,
when the surface carboxyl groups esterify with EDC-NHS,
FIGURE 15.12 Phosphorothioate DNA conjugation. Unmodified DNA colloidal stability of the particle can be compromised.97
(A) will coordinate to the particle’s surface via N7 position interactions Covalent conjugation of DNA to QDs may also be
of adenine (circled in yellow) and nonbridging oxygen (circled in accomplished using commercially available bifunctional
blue). Phosphorothioate DNA (B) produces stronger complexes by
coordinating on the particle’s surface via the higher-affinity sulfur
linking reagents. The linker sulfosuccinimidyl-4-(N-
(circled in blue) interactions. From Zhou W, et al., Tandem phosphor- maleimido-methyl) cyclohexane-1-carboxylate (sSMCC)
othioate Modifications for DNA adsorption Strength and polarity is well-suited use for this purpose. After labeling DNA
control on gold nanoparticles. ACS Appl Mater Interfaces molecules with amine groups, reaction with terminal NHS
2014;6(17):14795e14800. groups of sSMCC produces DNA-maleimide (Fig. 15.14).
Polymer coatings on the QDs may be reduced with TCEP
DNA and are pH dependent.93 DNA attached electrostati- to produce free thiol groups at the particle’s surface. The
cally is also subject to premature release by displacement thiol displaying QDs and DNA-maleimide are then reacted
by biomolecules. to produce DNA-conjugated QDs98
QDs functionalized with streptavidin can bind bio- Copper-free click chemistry can be used to conjugate
tinylated DNA. This strategy has been widely used and azide functionalized biomolecules to ligands with alkyne
offers a simple “mix and shake” route for conjugation.94 moieties via cycloaddition. Originally, the reaction
However, with an additional layer of protein, the resulting employed copper ions as catalysts, but copper is known to
QD-conjugate size may exceed the desired range for many quench QD photoluminescence.99 Recently, copper-free
biological applications. D’Agata et al. reported a hydro- alternatives have been demonstrated to successfully con-
dynamic diameter of 31.1 nm when 19 nm particles were jugate the DNA while preserving high QYs.100 Zhang
functionalized with streptavidin95. On binding of bio- et al. reported DHLA-PEG-N3-coated QDs added to
tinylated DNA in the study, the hydrodynamic diameter cyclooctyne-functionalized DNA resulted in nearly two-
further increased to 40 nm. In addition, the quantity of third of the DNA to be successfully conjugated. The
DNA molecules loaded per QD in this method is not loading efficiency in conjunction with the commercial
homogenous.96 availability of these linkers makes this conjugation

FIGURE 15.13 Lipid oligonucleotide


conjugation. Lipid oligonucleotide conju-
gates formed from 1,2-dioleoyl-sn-glycero-
3-phosphocholine (DOPC), palmitic uridine
phosphocholine (PUPC), and palmitic uri-
dine (PUOH). From Aimé A, et al., Quantum
dot lipid oligonucleotide bioconjugates: to-
ward a new anti-micro RNA nanoplatform.
Bioconjug Chem 2013;24(8):1345e1355.
254 Nanoparticles for Biomedical Applications

FIGURE 15.14 Amine to thiol-reactive linker (sulfo-SMCC)emediated conjugation.

strategy one of the most popular for producing QD-DNA enhanced green fluorescent protein (EGFP) expression,
conjugates. was covalently attached to QDs using amineethiol reac-
tive linkers. After cellular internalization, delivery of the
15.6.3 Quantum DoteDNA sensor plasmid to the nucleus was tracked by monitoring the QD
fluorescence signal over time. Simultaneous time-
applications
dependent monitoring of the GFP signal allows confir-
QD-DNA conjugates employing FRET-based biosensing mation of successful delivery and expression of the
have been reported to detect single point mutations in plasmid in the nucleus.
clinical samples with lower detection limits than molecular Successful siRNA-mediated gene silencing has been
beacon probes.101 The system reported by Zhang et al. reported using QD-siRNA conjugates. Derfus et al. pro-
labeled reporter probes with a fluorescent dye and the duced QDs with targeting F3 peptides and siRNA against
capture probes with biotin. In the presence of target DNA, EGFP covalently attached using thioleamine reactive
these probes form a sandwiched hybrid (Fig. 15.15). When linkers.78 The ability of these siRNA delivery vehicles to
streptavidin-functionalized QDs are introduced, the sand- escape the endosome is a limiting factor for their useful-
wiched hybrid forms an FRET donoreacceptor nano- ness. To promote endosomal escape, cationic liposomes
structure via interactions between the biotinylated capture were incubated with cells having already internalized the
probe and the streptavidin-functionalized QD. Now in QD-siRNA conjugates. These liposomes were internalized
proximity with the QD, and on selective excitation of the by new endosomes, which then fused with the QD-
QD, the dye labeled reporter probe generates fluorescence siRNAecontaining endosomes. The acidic environment
signal through FRET from excited QD to dye. Displaying induced osmotic lysis, resulting in release of the trapped
multiple biotinebinding sites, the QD can act as a QD-siRNA conjugates. On endosomal escape, mRNA ho-
concentrator to amplify signals by confining multiple dye mologous to the siRNA was degraded and EGFP knock-
molecules on a single probe. down was observed. Other genes have been silenced
Utilizing QDs in lieu of traditional fluorophores offers successfully with similar design strategies.78e80
comparatively higher signal-to-noise ratios. Capable of
detecting target nucleic acid molecules at concentrations
less than 1 femtomol,102 the utility of QD-FRETebased
15.7 Quantum dot drug delivery
biosensors in molecular diagnostics is clear. The primary systems for cancer therapy
limitation of these biosensors exists in controlling the Quantum Dots are designed to perform multiple tasks such
number of DNA molecules associated with each QD. as disease diagnosis and delivery of therapeutics, thus
Future success of these sensors in molecular diagnostics serving as a multifunctional (theranostic) nanoparticle
depends on the ability to produce homogenously labeled system. As a therapeutic delivery system, QDs can be
conjugates.98 surface loaded with anticancer drugs and targeted to cancer
QD-DNA conjugates have also demonstrated success tissue, while QD fluorescence will allow for imaging of
in delivering therapeutics to cells. These therapeutic ap- cancer tissue.
plications are the regulation of gene expression or delivery Theranostic CdSe and CdTe-based QDs have been
of gene therapies. One of the earliest demonstrations of incorporated into several composite drug delivery systems
using QD-DNA conjugates to regulate gene expression such as hydrogels,104,105 silica, polymers,106,107 or lipo-
was reported in 2006.80,103 Plasmid DNA, coded for somes.108 Kang et al. reported a multifunctional system in
Quantum Dots Chapter | 15 255

FIGURE 15.15 (A) Scheme showing the formation of the nanosensor by the sandwiching of the target DNA between the capture probe and reporter
probe. (B) Florescence emission of the dye-labeled (Cy5) reporter probe by FRET from the QD to Cy5. As FRET is distance dependent unbound reporter
probes do not participate in FRET with QD and are not fluorescent (C) Confocal microscopy experimental setup utilizing a continuous-flow of nanosensor
to minimize photobleaching. From Zhang CY, et al., Single-quantum-dot-based DNA nanosensor. Nat Mater 2005;4(11):826e831.

which core/shell CdSe/ZnS QDs and paclitaxel (PTX, While many studies have shown Cd-based QDs when
anticancer drug) were encapsulated in lipid micelles con- properly coated are nontoxic, over time they do carry
sisting of phosphatidylethanolamine (DSPE) and some risk of toxicity due to heavy metal ion leakage from
maleimide-functionalized PEG (Fig. 15.16). The micelle the core.110 Thus, noneheavy metal-containing QDs have
was targeted to cancer cells with antiepidermal growth been explored as nontoxic alternatives to Cd-containing
factor receptor (EGFR, receptor commonly overexpressed QDs. InP QDs have been shown to be less toxic
in cancer) ABs.109 The AB was thiolated with Traut’s re- than Cd-containing QDs.111,112 InP QDs can also be
agent and then reacted with maleimide-functionalized PEG. tailored to have emissions in the NIR range. Wang et al.
In the low pH environment of tumor tissue, the micelle encapsulated InP/ZnS QDs and aminoflavone (AF,
structure was destabilized via protonation of amine groups anticancer compound) in a micelle composed of PEG-
on the phospholipid. This response gives the micelle an PLA block copolymer.113 First, the InP/ZnS QDs were
activatable release mechanism where the drug would be coated with mercaptoacetic acid through ligand exchange,
released when the micelle encounters the low pH envi- then carbodiimide coupling (DMF/DMAP) was used to
ronment of tumor tissue. In vivo fluorescence imaging re- link hydroxyl groups on the polymer (PLA-PEG-
sults confirmed successful entry and delivery of QD maleimide and PLA-PEG-OCH3) to carboxyl groups on
micelles into tumor tissue, LS174T-xenograft BALB/c the QD. An anti- (EGFR) nanobody, 7D12 Nb, was
mice after 24 h. The QD signal remained elevated in tumor employed for targeting and was attached to the micelle
tissue with the EGFR-targeted treatment as opposed to by maleimideethiol reaction (Fig. 15.17A). It was
nontargeted micelles. It was also observed that the targeted shown that the nanobody increased micelle uptake on
micelles were more effective in reducing tumor growth than EGFR-overexpressing cells and improved its cytotoxic
nontargeted micelles and free PTX. effect in vitro. Both targeted and nontargeted QD
256 Nanoparticles for Biomedical Applications

micelles inhibited tumor growth in vivo with the targeted Nanocarriers that incorporate multiple modes of im-
treatment performing significantly better at reducing tu- aging, i.e., fluorescence and magnetic resonance imaging
mor volume (Fig. 15.17B). The presence of the QD (MRI) or positron emission tomography, can provide
allowed the tumors to be imaged by NIR fluorescence more information on biodistribution and biological
in vivo and could be used to monitor tumor progression fate of the particles than those with only a single mo-
(Fig. 15.17C). dality. Iron oxide NPs (IONP, a T2 contrast agent) or

FIGURE 15.16 Synthesis of paclitaxel


(PTX) and quantum dot (QD)eloaded lipid
micelles. Based on Kang SJ, et al., Anti-
EGFR lipid micellar nanoparticles co-
encapsulating quantum dots and paclitaxel
for tumor-targeted theranosis. Nanoscale
2018;10(41):19338e19350.

FIGURE 15.17 (A) Design of InP/ZnS quantum dot (QD) micelle drug delivery system. (B) Normalized tumor volumes plotted over time of targeted
(T) or nontargeted (NT) QD micelles with anticancer drug (AF). near-infrared imaging of mouse and tumor after injection with targeted QD micelle.
Modified from Wang Y, et al., Quantum-dot-based theranostic micelles conjugated with an anti-EGFR nanobody for triple-negative breast cancer
therapy. ACS Appl Mater Interfaces 2017;9(36):30297e30305.
Quantum Dots Chapter | 15 257

gadolinium ions (Gd3þ, a T1 contrast agent) can be linked of these particles in the animal model (Fig. 15.18
to QDs to provide contrast enhancement for MRI imaging bottom).
capabilities. QD drug delivery platforms incorporating
this can be imaged through both MRI and QD fluores- 15.7.1 Activatable quantum dot drug delivery
cence. MRI is advantageous as it is more tissue pene-
systems
trating than fluorescence, which makes it better for
in vivo imaging, whereas fluorescence is better for Drug delivery systems that incorporate stimuli responsive
in vitro cellular imaging or histological studies. Ye et al. or “activatable” QDs, where QD fluorescence is altered by
used a biodegradable polymer, poly(lactic-co-glycolic a stimulus such as the presence of a biomolecule or change
acid) (PLGA), to link ZnS:Mn QDs to IONPs and in pH, can provide additional information on the drug de-
busulfan (cancer therapeutic) in an oil-in-water emulsion livery event. Activatable drug release can help maximize
(Fig. 15.18 top).114 First, oleate-stabilized IONPs were the bioavailability of a therapeutic by minimizing prema-
mixed with a solution of decanethiol-capped ZnS:Mn ture drug release.
QDs in dichloromethane (DCM). PLGA (15 kDa MW) Multimodal activatable QDs have been synthesized by
was then added to the particles in DCM which was mixed Santra et al. with folic acid (FA) acting as a targeting ligand
with an aqueous solution (1:20 oil/water ratio) of poly and quencher for QD fluorescence.115 First, a micro-
lactic acid (PLA) under sonication to form an emulsion. emulsion was used to synthesize CdS:Mn/ZnS QDs. FA
The oil was then evaporated, and the particles were pu- was bound to NAC using EDC chemistry. The FA-NAC
rified by dialysis. The QD-IONP system could be tracked was attached to the QD via a disulfide bond. FA was able
by fluorescence in vitro. Darker contrast appeared in the to quench QD fluorescence emission by transferring elec-
liver (7 minutes post injection), confirming accumulation trons to the excited QD. The QD fluorescence can be
restored (“OFF” to “ON”) by cleaving the disulfide bond
connecting FA to the QD. When the QD comes in contact
with GSH (antioxidant), it is able to reductively cleave the
disulfide bond (Fig. 15.19).116e118 An anticancer drug (SF-
1-046, STAT3 inhibitor) is also released on activation
because it too is attached to the QD via disulfide bond.
With this design, the QD releases the drug after interacting
with GSH, and it is also able to report on the drug delivery
event through increased fluorescence. Utilizing GSH as an
activator for NP drug delivery systems is attractive as some
cancer types have high concentrations of GSH in their
cytosol.119 This QD was attached to an IONP to give the
ability of MRI imaging.
Zinc oxide (ZnO) is generally regarded as safe (GRAS)
by the United States Food and Drug Administration and is
used in many foods and cosmetics. ZnO QDs can be syn-
thesized in large-scale economically when compared with
other nanomaterials.120 For these reasons, ZnO QDs have
great potential for use in commercial biomedical applica-
tions. The increased solubility of ZnO QDs in slightly
acidic environment (pH 5.0) can be exploited for use in
activatable drug delivery. One of the hallmarks of cancers
is decreased pH of the tumor microenvironment. This
acidic environment can be used to activate or dissolve ZnO-
based drug carriers, thereby releasing the bound drug where
it can kill the cancer. On dissolution in microenvironment,
released zinc ions have also been demonstrated to induce
cytotoxic effect. Studies have shown ZnO QDs themselves
FIGURE 15.18 Scheme showing the applications of quantum dot (QD)eiron can be cytotoxic to some cultured malignant cells.121,122
oxide nanoparticles and T2*-weighted in vivo MR images of a rat before, 7min The release of zinc ions from ZnO QDs has been proposed
after, and 2h 40 min after injection of QD-IONPs. Modified from Ye F, et al.,
Biodegradable polymeric vesicles containing magnetic nanoparticles, quantum
to be used as a mechanism of killing for cancer treat-
dots and anticancer drugs for drug delivery and imaging. Biomaterials ment.123 High concentrations of free zinc ions can bind to
2014;35(12):3885e3894. DNA changing its conformation and potentially disrupting
258 Nanoparticles for Biomedical Applications

FIGURE 15.19 Scheme of an


activatable quantum doteiron oxide
nanoparticle drug delivery platform.
From Mitra RN, et al., An activatable
multimodal/multifunctional nanop-
robe for direct imaging of intracel-
lular drug delivery. Biomaterials
2012;33(5):1500e1508.

its transcription.124,125 ZnO QDs have also been shown to silica (SiO2) is commonly added to the ZnO QDs before
generate damaging ROS as well as cause cell membrane drug loading. This shell increases the stability of the
damage (Fig. 15.20). These intrinsic properties of ZnO QDs QDs and can also provide functional groups (such as
have the potential to work synergistically to enhance the amine) for further derivatization. Amine-derivatized silane,
activity of bound therapeutics.122 ROS generation can (3-aminopropyl)triethoxysilane (APTES), is often used to
make the cancer cells more sensitive to chemotherapy, and provide reactive surface amine groups for the coupling of
membrane disruption could enhance the bioavailability of targeting ligands and/or PEG.
therapeutics.126 Liang et al. loaded ZnO QDs with DOX by a copreci-
One of the most widely used methods of preparing pitation method to make an activatable drug delivery sys-
ZnO-drug conjugates exploits the high reactivity of surface tem.132 The decomposition of the nanoparticles in acidic
Zn to bind to functional groups such as ketone and alco- solution was confirmed through TEM/SEM analysis. The
hols.127,128 This method involves the simple mixing of ZnO particles were stable at pH 7.5, but rapidly decomposed at
QDs with the drug and does not involve any complex cross- pH 5.0. This means that when the nanoparticles are in the
linking reagents. Drugs attached in this way are rapidly acidified endosome, they will start to release drug as they
released when the ZnO QD dissolves, providing an acti- dissolve (Fig. 15.21). The authors used flow cytometry to
vatable release mechanism. ZnO QD-drug conjugates have show increased intracellular uptake of DOX from the ZnO
been synthesized with doxorubicin (DOX),129e139 tanger- QD-DOX treatment than from free DOX. The increased
etin140, paclitaxel,141 and others122,142 to make pH- delivery of DOX was confirmed through MTT (cell pro-
responsive drug delivery vehicles. A protective shell of liferation) assay which showed ZnO QD-DOX was more
effective than free DOX at killing the MCF-7 cells. The
authors showed that the killing was due to enhanced DOX
delivery because the ZnO particles themselves had no
toxicity on MCF-7 cells at the concentrations tested. ZnO
QD-DOX was also effective at killing resistant MCF-7R
cells where free DOX had no effect. This may be due to
the high localized concentration of DOX delivered by the
QDs. Zhu et al. built on this system by showing the syn-
ergistic anticancer activity of ZnO QDs with DOX due to
ROS generation from the ZnO.138
Multimodal imaging capability can be extended to ZnO
drug delivery vehicles by the incorporation of Gd3þ ions.
Polymer-coated ZnO QDs were synthesized with excess
FIGURE 15.20 Scheme showing the different modes of action ZnO carboxyl groups, and they were mixed with Gd3þ and DOX
quantum dots can take to damage cells: (A) membrane damage, (B) reactive to create a fluorescence (600e700 nm) and MRI trackable
oxygen species (ROS) production, and (C) Zn2þ ion release. pH-responsive drug delivery vehicle.133 The authors
Quantum Dots Chapter | 15 259

FIGURE 15.21 (A) Scheme


showing the pH-induced delivery of
doxorubicin (DOX) by ZnO QDs
compared to free DOX. In vitro
cytotoxicity of free DOX and DOX
loaded ZnO QDs (ZD NPs) on
(B) drug resistant cells (MCF-7R)
and (C) drug sensitive cells (MCF-
7S). Modified from Liu J, et al., Zinc
oxide nanoparticles as adjuvant to
facilitate doxorubicin intracellular
accumulation and visualize pH-
responsive release for overcoming
drug resistance. Mol Pharm
2016;13(5):1723e1730.

reported significant antitumor activity of the particles on different kinds of QDs have been used for bacterial
BxPC-3 tumor-bearing nude mice. Fig. 15.22A shows an detection such as CdSe/ZnS,143 CdSe,144 CQDs,145 and
increase in tumor size for the control, whereas no change in ZnO.146
tumor size was observed for DOX, Doxil, or ZnO-Gd-DOX It is possible to reliably detect low levels of bacterial
QD treatments. Hematoxylin and eosin (H&E) staining of cells in a sample using QDs due to their high QY in
the tumor showed that QD-DOX treatment damaged the comparison with organic dyes. Hahn et al. used biotinylated
tumor cells more severely than DOX or Doxil treatments, anti-Escherichia coli O157:H7 ABs that specifically bind to
making it a more effective treatment (Fig. 15.22B). the outer membrane of E. coli O157:H7 cells. Bacterial
cells were then labeled with streptavidin-conjugated CdSe/
ZnS QDs. The authors showed that streptavidin-QD la-
15.8 Imaging of bacteria using beling showed around two orders of magnitude more sen-
fluorescent quantum dots sitive detection of bacterial cells than streptavidin-FITC
The photoluminescent properties of QDs can be employed (organic dye). The QDs were even capable of detecting a
to detect bacteria both in vitro and in vivo. Detection of single pathogenic cell.143
bacteria is important to identify contaminated food or water QDs can also be used for the quantification of bacteria.
and to diagnose infections. For bacterial detection, QDs are CdSe-thioglycolic acid (TGA) QDs were coupled to Sal-
generally conjugated to a molecule such as an AB or sugar monella typhimurium membrane proteins using EDC
moiety in order to provide bacterial recognition. Many coupling chemistry. QD fluorescence intensity responded
260 Nanoparticles for Biomedical Applications

FIGURE 15.22 (A) Images of BxPC-3 tumor-bearing nude mice after 18 and 36 days with different treatments. (B) H&E staining of tumor cross
sections after 36 days treatment. Modified from Ye DX, et al., ZnO-based Nanoplatforms for labeling and treatment of mouse tumors without detectable
toxic Side effects. ACS Nano 2016;10(4):4294e4300.

linearly to bacterial counts over a concentration range of (TGRAKRRMQYNRR, 1693Da) and MPA (NIR dye)
102e106 CFU/mL of bacteria.144 Lai et al. demonstrated using EDC (ZnO@BSA-PEP-MPA). The conjugate was
that mannose CQDs can also be used for the detection of tested in both Staphylococcus aureus and Bacillus subtilis
bacteria in samples of tap water, apple juice, and human infection mouse models. In these models, S. aureus or
urine. The authors employed specific interaction between B. subtilis suspensions were injected into the right axillary
mannose and E. coli bacterial protein FimH for the bacterial fossa, followed by exposure to the QD conjugates through
labeling and showed that this QD probe has the potential to tail-vein injection. QD fluorescence allowed detection of
be used in clinical applications.145 infection site after 6 h of treatment (Fig. 15.23B and
C).146 In a subsequent study, vancomycin (VAN) was
loaded onto the ZnO-PEP-MPA QD conjugates for
15.9 Application of quantum dots as an theranostic activity. Mice infected with S. aureus showed
antimicrobial agent or significant reduction in skin lesions when treated with
antimicrobial delivery system VAN@ZnO-PEP-MPA than the QD or antibiotic alone
(Fig. 15.23D).146 Thus, the authors suggest that
Several studies have demonstrated applications of QDs as
ZnO@BSA-PEP-MPA serves as a nanocarrier for the
antibacterial agents such as CdTe,147,148 ZnSe/ZnS,149 and
targeted delivery of VAN.
ZnO.150,151 Chen et al. also examined ZnO QDs as an
antimicrobial delivery system.146 Although ROS generation
is shown to play a role in antibacterial mechanisms of QDs, 15.10 Conclusions and perspective
the comprehensive understanding of interactions between
QD composition along with a number of physical and
QDs and bacteria remains unclear.147,148,151
chemical properties is important to consider when selecting
It has been also reported that certain QDs when con-
them for biomedical applications. Other than QD compo-
jugated to antibiotics are able to enhance the efficacy of the
sition, particle size, surface charge, surface chemistry, and
antibiotic. The antibiotic ceftriaxone (Rocephin) when
targeting ligands must be carefully selected to make them
conjugated to CdTe QDs demonstrated a synergistic effect
suitable for use in biological systems. Selection of appro-
of killing E. coli.152 Using EDC coupling chemistry, Jijie
priate coupling chemistry for bioconjugation is important
et al. conjugated ampicillin (-carboxyl) to amine-
for efficient loading, stability, and orientation of target
functionalized CQDs. The conjugate was significantly
molecules such as ABs, proteins, antibiotics, and anticancer
more effective in killing E. coli than ampicillin alone.
drugs onto the QD surface.
Authors suggested that the increased efficacy was likely
For biomedical applications, QDs offer many advan-
due to the cooperative antibacterial activity of QD-
tages over traditional organic fluorescent dyes for sensitive
ampicillin as compared with free antibiotic.153
imaging, target detection, targeted delivery of therapeutics,
For diagnostic applications, Chen et al. synthesized
as well as monitoring of therapy due to their high QY,
ZnO QDs coated with BSA and then functionalized
photostability, long lifetime, and large Stokes’ shift
with a cationic antimicrobial peptide (PEP) UBI29-41
Quantum Dots Chapter | 15 261

FIGURE 15.23 Figure shows in vivo detection and treatment of bacterial infection by ZnO QD conjugate. Synthesis of antibiotic-ZnO QD complex (A),
in vivo detection of Bacillus subtilis (B) and Staphylococcus aureus (C) site of infection by ZnO QD conjugate are shown. (D) Reduction of skin lesion in
S. aureus infection mouse model on treatment with antibioticeQD complex compared with QD conjugate or antibiotic alone. Modified from Chen H,
et al., Versatile antimicrobial peptide-based ZnO quantum dots for in vivo bacteria diagnosis and treatment with high specificity. Biomaterials
2015;53:532e544.

(separation of excitation and emission wavelengths). additional benefit from the EPR effect which helps increase
Because of large surface-to-volume ratio, QDs serve as an accumulation at the tumor site.155 The incorporation of
excellent nanoscale payload carrier for therapeutic cargoes. PEG to the surface of NP systems can help to minimize
Several reports have shown that the capability of QDs to nonspecific interactions and serum protein adsorption that
improve the bioavailability therapeutics at the target site as promote circulatory clearance via opsonisation.156 Detailed
well synergistic effect on therapeutic efficacy. Many of study of QD interactions with biological systems leading to
these applications rely on careful control of physiochemical their uptake, mobility, clearance, and degradation will
properties of surface-modified QD that allows for recog- allow for gaining new knowledge based on which more
nition of cell-specific surface receptor proteins. efficient QD systems can be developed.
For successful clinical applications of QD nanotech- QD toxicity and accumulation in off-target tissue also
nology as a drug delivery system, there are many questions must be addressed.157 QDs containing heavy-metals,
that must be answered. Despite careful QD surface engi- despite their biocompatible coating, may not be suitable for
neering, only small fractions of them are able to reach the in vivo applications due to high risk of toxic metal ion
target site.154 This demands further advancement in QD release. However, ex vivo use of QDs for diagnostic pur-
design that will avoid their premature removal from the poses could be feasible. ZnO-based QDs are attractive as
circulatory system by the reticuloendothelial system fol- they can be eventually degraded to Zn2þ ions, entering to
lowed by their excretion through liver or kidney. In general, the metabolic pool. Ultrasmall size QDs (<10 nm size)
particles that have extended circulation times can draw coated with PEGylated silica (SiO2)158 will have the
262 Nanoparticles for Biomedical Applications

advantage of reduced bioaccumulation and could be 18. Kortan AR, et al. Nucleation and growth of CdSe on ZnS quantum
excreted from the body. crystallite seeds, and vice versa, in inverse micelle media. J Am
CQDs have a low toxicity profile which makes them Chem Soc 1990;112(4):1327e32.
19. Bandyopadhyaya R, et al. Modeling of precipitation in reverse
attractive over metal-based QDs.159 Amino acidederived
micellar systems. Langmuir 1997;13(14):3610e20.
as-synthesized CQDs usually have functional groups
20. Malik MA, Wani MY, Hashim MA. Microemulsion method: a novel
available for bioconjugation which makes them industrially route to synthesize organic and inorganic nanomaterials: 1st Nano
attractive. Unfortunately, CQDs are not biodegradable due Update. Arabian J Chem 2012;5(4):397e417.
to their crystalline carbon core. Therefore, CQDs must be 21. Bagwe RP, Khilar KC. Effects of intermicellar exchange rate on the
engineered to be excreted from the body to avoid the risk of formation of silver nanoparticles in reverse microemulsions of AOT.
tissue accumulation, in particular the nontarget tissue. Langmuir 2000;16(3):905e10.
22. Samanta A, Deng Z, Liu Y. Aqueous synthesis of glutathione-
capped CdTe/CdS/ZnS and CdTe/CdSe/ZnS core/shell/shell nano-
References crystal heterostructures. Langmuir 2012;28(21):8205e15.
1. Bera D, et al. Quantum dots and their multimodal applications: a 23. Bera D, et al. Photoluminescence of ZnO quantum dots produced by
review. Materials 2010;3:2260e345. a sol-gel process. Opt Mater 2008;30(8):1233e9.
2. Kwak J, et al. High-power genuine ultraviolet light-emitting diodes 24. Bourlinos AB, et al. Surface functionalized carbogenic quantum
based on colloidal nanocrystal quantum dots. Nano Lett dots. Small 2008;4(4):455e8.
2015;15(6):3793e9. 25. Yang Z, et al. Nitrogen-doped, carbon-rich, highly photoluminescent
3. Pichaandi J, van Veggel FCJM. Near-infrared emitting quantum carbon dots from ammonium citrate. Nanoscale 2014;6(3):1890e5.
dots: recent progress on their synthesis and characterization. Coord 26. Zhu H, et al. Microwave synthesis of fluorescent carbon nano-
Chem Rev 2014;263e264:138e50. particles with electrochemiluminescence properties. Chem Commun
4. Ekimov AI, O AA. Quantum size effect in three-dimensional 2009;(34):5118e20.
microscopic semiconductor crystals. JETP Lett (Engl Transl) 27. Liu X, et al. Simple approach to synthesize amino-functionalized
1981;34(6):345e9. carbon dots by carbonization of chitosan. Sci Rep 2016;6:31100.
5. Efros AL, Efros AL. Interband light absorption in a semiconductor 28. Chiu S-H, et al. Rapid fabrication of carbon quantum dots as
sphere. Fiz Tekh Poluprovodn (Leningrad) 1982;16(7):1209e14. multifunctional nanovehicles for dual-modal targeted imaging and
6. Brus LE. Electron-electron and electron-hole interactions in small chemotherapy. Acta Biomater 2016;46:151e64.
semiconductor crystallites: the size dependence of the lowest excited 29. Zhu Y-J, Chen F. Microwave-assisted preparation of inorganic
electronic state. J Chem Phys 1984;80(9):4403e9. nanostructures in liquid phase. Chem Rev 2014;114(12):6462e555.
7. Lippa A. Fluorescence immunoassay involving energy transfer be- 30. Staros JV. N-hydroxysulfosuccinimide active esters: bis(N-
tween two fluorophores. USA: Ethigen Corp.; 1987. p. 27. hydroxysulfosuccinimide) esters of two dicarboxylic acids are
8. Smith AM, et al. Bioconjugated quantum dots for in vivo molecular hydrophilic, membrane-impermeant, protein cross-linkers.
and cellular imaging. Adv Drug Deliv Rev 2008;60(11):1226e40. Biochemistry 1982;21(17):3950e5.
9. Wu P, Yan X-P. Doped quantum dots for chemo/biosensing and 31. Lao UL, Mulchandani A, Chen W. Simple conjugation and purifi-
bioimaging. Chem Soc Rev 2013;42(12):5489e521. cation of quantum dot-antibody complexes using a thermally
10. Sun L-W, et al. Lanthanum-doped ZnO quantum dots with greatly responsive elastin-protein L scaffold as immunofluorescent agents.
enhanced fluorescent quantum yield. J Mater Chem J Am Chem Soc 2006;128(46):14756e7.
2012;22(17):8221e7. 32. Goldman ER, et al. Conjugation of luminescent quantum
11. Bol AA, Meijerink A. Long-lived Mn2þ emission in nanocrystalline dots with antibodies using an engineered adaptor protein to
ZnS:Mn2þ. Phys Rev B 1998;58(24):R15997e6000. provide new reagents for fluoroimmunoassays. Anal Chem
12. Neus GB, et al. The reactivity of colloidal inorganic nanoparticles. 2002;74(4):841e7.
2012. 33. Sapsford KE, et al. Biosensing with luminescent semiconductor
13. Lee W, et al. An effective oxidation approach for luminescence quantum dots. Sensors 2006;6(8):925e53.
enhancement in CdS quantum dots by H2O2. Nanoscale Res Lett 34. Medintz IL, et al. Quantum dot bioconjugates for imaging, labelling
2012;7(1):672. and sensing. Nat Mater 2005;4(6):435e46.
14. Vela J, et al. Effect of shell thickness and composition 35. Han HS, et al. Quantum dot/antibody conjugates for in vivo cyto-
on blinking suppression and the blinking mechanism in metric imaging in mice. Proc Natl Acad Sci USA
‘giant’ CdSe/CdS nanocrystal quantum dots. J Biophot 2015;112(5):1350e5.
2010;3(10e11):706e17. 36. Fernandez-Arguelles MT, et al. Simple bio-conjugation of polymer-
15. Talapin DV, et al. CdSe/CdS/ZnS and CdSe/ZnSe/ZnS Core- coated quantum dots with antibodies for fluorescence-based immu-
ShellShell nanocrystals. J Phys Chem B 2004;108(49):18826e31. noassays. Analyst 2008;133(4):444e7.
16. Murray CB, Norris DJ, Bawendi MG. Synthesis and characterization 37. Rakovich TY, et al. Highly sensitive single domain antibody-
of nearly monodisperse CdE (E ¼ sulfur, selenium, tellurium) quantum dot conjugates for detection of HER2 biomarker in lung
semiconductor nanocrystallites. J Am Chem Soc and breast cancer cells. ACS Nano 2014;8(6):5682e95.
1993;115(19):8706e15. 38. Tiwari DK, et al. Synthesis and characterization of anti-HER2
17. Bailey RE, Smith AM, Nie S. Quantum dots in biology and medi- antibody conjugated CdSe/CdZnS quantum dots for fluorescence
cine. Physica E 2004;25(1):1e12. imaging of breast cancer cells. Sensors 2009;9(11):9332e54.
Quantum Dots Chapter | 15 263

39. Wu YT, et al. Quantum dot-based FRET immunoassay for HER2 59. Liu R, et al. Design of a new near-infrared ratiometric fluorescent
using ultrasmall affinity proteins. Small 2018;14(35). nanoprobe for real-time imaging of superoxide anions and hydroxyl
40. Liu JL, et al. Conjugation of biotin-coated luminescent quantum dots radicals in live cells and in situ tracing of the inflammation process
with single domain antibody-rhizavidin fusions. Biotechnol Rep in vivo. Anal Chem 2018;90(7):4452e60.
2016;10:56e65. 60. Wang J, et al. Diethyldithiocarbamate functionalized CdSe/
41. Umakoshi T, et al. Quantum-dot antibody conjugation visualized at CdS quantum dots as a fluorescent probe for copper ion
the single-molecule scale with high-speed atomic force microscopy. detection. Spectrochim Acta A Mol Biomol Spectrosc
Colloids Surf., B 2018;167:267e74. 2011;81(1):178e83.
42. Pathak S, Davidson MC, Silva GA. Characterization of the func- 61. Liao S, et al. Novel S, N-doped carbon quantum dot-based "off-on"
tional binding properties of antibody conjugated quantum dots. fluorescent sensor for silver ion and cysteine. Talanta
Nano Lett 2007;7(7):1839e45. 2018;180:300e8.
43. Resch-Genger U, et al. Quantum dots versus organic dyes as fluo- 62. Ma Q, et al. A novel carboxymethyl chitosan-quantum dot-based
rescent labels. Nat Methods 2008;5:763. intracellular probe for Zn2þ ion sensing in prostate cancer cells.
44. Liu Y-S, et al. pH-sensitive photoluminescence of CdSe/ZnSe/ZnS Acta Biomater 2014;10(2):868e74.
quantum dots in human ovarian cancer cells. J Phys Chem C 63. Ren H-B, et al. Silica-coated S2–enriched manganese-doped ZnS
2007;111(7):2872e8. quantum dots as a photoluminescence probe for imaging intracel-
45. Huang X, Li W, He W. pH tunable turn on and turn off quantum lular Zn2þ ions. Anal Chem 2011;83(21):8239e44.
dots conjugated with poly(2-dialkylamino)ethyl methacrylate). Adv 64. Ruedas-Rama MJ, Hall EAH. Azamacrocycle activated quantum dot
Mater Res 2014:998e9 (Advances in Applied Sciences, Engineering for zinc ion detection. Anal Chem 2008;80(21):8260e8.
and Technology II): pp. 75e78, 5 pp. 65. Wang G, Li Z, Ma N. Next-generation DNA-functionalized quantum
46. Medintz IL, et al. Quantum-dot/dopamine bioconjugates function as dots as biological sensors. ACS Chem Biol 2018;13(7):1705e13.
redox coupled assemblies for in vitro and intracellular pH sensing. 66. Ebrahim S, et al. CdTe quantum dots as a novel biosensor for Ser-
Nat Mater 2010;9:676. ratia marcescens and Lipopolysaccharide. Spectrochim Acta, Part A
47. Ji X, et al. On the pH-dependent quenching of quantum dot pho- 2015;150:212e9.
toluminescence by redox active dopamine. J Am Chem Soc 67. Zhu G, Yang K, Zhang C-y. A single quantum dot-based biosensor
2012;134(13):6006e17. for telomerase assay. Chem Commun 2015;51(31):6808e11.
48. Maria Jose R-R, Elizabeth AHH. pH sensitive quantum dote 68. Wu P, et al. Analyte-activable probe for protease based on cyto-
anthraquinone nanoconjugates. Nanotechnology 2014;25(19):195501. chrome C-capped Mn: ZnS quantum dots. Anal Chem
49. Snee PT, et al. A ratiometric CdSe/ZnS nanocrystal pH sensor. J Am 2014;86(20):10078e83.
Chem Soc 2006;128(41):13320e1. 69. Zhou Z-M, Yu Y, Zhao Y-D. A new strategy for the detection of
50. Kurabayashi T, et al. CdSe/ZnS quantum dots conjugated with a adenosine triphosphate by aptamer/quantum dot biosensor based on
fluorescein derivative: a FRET-based pH sensor for physiological chemiluminescence resonance energy transfer. Analyst
alkaline conditions. Anal Sci 2014;30(5):545e50. 2012;137(18):4262e6.
51. Ast S, Rutledge PJ, Todd MH. pH-Responsive quantum dots 70. Yang C, et al. Quantum-dot-based biosensor for simultaneous
(RQDs) that combine a fluorescent nanoparticle with a pH-sensitive detection of biomarker and therapeutic drug: first steps toward an
dye. Phys Chem Chem Phys 2014;16(46):25255e7. assay for quantitative pharmacology. Analyst 2012;137(5):1205e9.
52. Paek K, et al. Fluorescent and pH-responsive diblock copolymer- 71. Geissler D, et al. Quantum dot biosensors for ultrasensitive multi-
coated core-shell CdSe/ZnS particles for a color-displaying, ratio- plexed diagnostics. Angew Chem Int Ed 2010;49(8):1396e401.
metric pH sensor. Chem Commun 2011;47(37):10272e4. S1396/1-S1396/9.
53. Dennis AM, et al. Quantum dot-fluorescent protein FRET probes for 72. Stanisavljevic M, et al. Quantum dots-fluorescence resonance energy
sensing intracellular pH. ACS Nano 2012;6(4):2917e24. transfer-based nanosensors and their application. Biosens Bio-
54. Pratiwi FW, et al. Construction of single fluorophore ratiometric pH electron 2015;74:562e74.
sensors using dual-emission Mn2þ-doped quantum dots. Biosens 73. Bakalova R, Zhelev Z, Ohba H. Quantum dots open new trends in
Bioelectron 2016;84:133e40. biosensor evolution. Sens Lett 2006;4(4):452e4.
55. Yuan J, Guo W, Wang E. Utilizing a CdTe quantum dots-enzyme 74. Yamagata M, et al. The contribution of lactic acid to acidification of
hybrid system for the determination of both phenolic compounds tumors: studies of variant cells lacking lactate dehydrogenase. Br J
and hydrogen peroxide. Anal Chem 2008;80(4):1141e5. Canc 1998;77(11):1726e31.
56. Bhattacharya S, et al. Detection of reactive oxygen 75. Jaehde E, et al. pH in human tumor xenografts and transplanted rat
species by a carbon-dot-ascorbic acid hydrogel. Anal Chem tumors: effect of insulin, inorganic phosphate, and m-
2017;89(1):830e6. iodobenzylguanidine. Cancer Res 1992;52(22):6209e15.
57. Huang XY, et al. Quantum dot-based FRET for sensitive determi- 76. Jin T, et al. A quantum dot-based ratiometric pH sensor. Chem
nation of hydrogen peroxide and glucose using tyramide reaction. Commun 2010;46(14):2408e10.
Talanta 2013;106:79e84. 77. Castanho MARB, Prieto MJE. Fluorescence quenching data inter-
58. Adegoke O, Khene S, Nyokong T. Fluorescence "switch on" of pretation in biological systems: the use of microscopic models for
conjugates of CdTe@ZnS quantum dots with Al, Ni and Zn data analysis and interpretation of complex systems. Biochim Bio-
tetraamino-phthalocyanines by hydrogen peroxide: characterization phys Acta Biomembr 1998;1373(1):1e16.
and applications as luminescent nanosensors. J Fluoresc 78. Derfus AM, et al. Targeted quantum dot conjugates for siRNA de-
2013;23(5):963e74. livery. Bioconjug Chem 2007;18(5):1391e6.
264 Nanoparticles for Biomedical Applications

79. Elbakry A, et al. Layer-by-Layer assembled gold nanoparticles for 100. Zhang H, et al. Robust and specific ratiometric biosensing using a
siRNA delivery. Nano Lett 2009;9(5):2059e64. copper-free clicked quantum doteDNA aptamer sensor. Nanoscale
80. Qi L, Gao X. Quantum DotAmphipol nanocomplex for intracel- 2013;5(21):10307e15.
lular delivery and real-time imaging of siRNA. ACS Nano 101. Zhang CY, et al. Single-quantum-dot-based DNA nanosensor. Nat
2008;2(7):1403e10. Mater 2005;4(11):826e31.
81. Sapsford KE, et al. Kinetics of metal-affinity driven self-assembly 102. Liang R-Q, et al. An oligonucleotide microarray for microRNA
between proteins or peptides and CdSeZnS quantum dots. expression analysis based on labeling RNA with quantum dot and
J Phys Chem C 2007;111(31):11528e38. nanogold probe. Nucleic Acids Res 2005;33(2). p. e17-e17.
82. Medintz IL, et al. A reactive peptidic linker for self-assembling 103. Srinivasan C, et al. Labeling and intracellular tracking of function-
hybrid quantum DotDNA bioconjugates. Nano Lett ally active plasmid DNA with semiconductor quantum dots. Mol
2007;7(6):1741e8. Ther 2006;14(2):192e201.
83. Zhou W, et al. Tandem phosphorothioate Modifications for DNA 104. Wu W, et al. In-situ immobilization of quantum dots in
adsorption Strength and polarity control on gold nanoparticles. ACS polysaccharide-based nanogels for integration of optical pH-sensing,
Appl Mater Interfaces 2014;6(17):14795e800. tumor cell imaging, and drug delivery. Biomaterials
84. Liu C, et al. Aptamer-functionalized CdTe:Zn2þ quantum dots for 2010;31(11):3023e31.
the detection of tomato systemin. Analytical Methods 105. Li Z, et al. Quantum dots loaded nanogels for low cytotoxicity, pH-
2015;7(18):7748e52. sensitive fluorescence, cell imaging and drug delivery. Carbohydr
85. Jiang G, et al. Cascaded FRET in conjugated polymer/quantum dot/ Polym 2015;121:477e85.
dye-labeled DNA complexes for DNA hybridization detection. ACS 106. Huang H-K, et al. A novel cancer nanotheranostics system based on
Nano 2009;3(12):4127e31. quantum dots encapsulated by a polymer-prodrug with controlled
86. Farlow J, et al. Formation of targeted monovalent quantum dots by release behaviour. Aust J Chem 2017;70(12):1302e11.
steric exclusion. Nat Methods 2013;10:1203. 107. Glueckert R, et al. Nanoparticle mediated drug delivery of rolipram
87. Carion O, et al. Synthesis, encapsulation, purification and coupling to tyrosine kinase B positive cells in the inner ear with targeting
of single quantum dots in phospholipid micelles for their use in peptides and agonistic antibodies. Front Aging Neurosci
cellular and in vivo imaging. Nat Protoc 2007;2:2383. 2015;7:1e18.
88. Dubertret B, et al. In vivo imaging of quantum dots encapsulated in 108. Olerile LD, et al. Near-infrared mediated quantum dots and pacli-
phospholipid micelles. Science 2002;298(5599):1759. taxel co-loaded nanostructured lipid carriers for cancer theragnostic.
89. Aimé A, et al. Quantum dot lipid oligonucleotide bioconjugates: Colloids Surf B Biointerfaces 2017;150:121e30.
toward a new anti-micro RNA Nanoplatform. Bioconjug Chem 109. Kang SJ, et al. Anti-EGFR lipid micellar nanoparticles co-
2013;24(8):1345e55. encapsulating quantum dots and paclitaxel for tumor-targeted
90. Van Lehn RC, et al. Lipid tail protrusions mediate the insertion of theranosis. Nanoscale 2018;10(41):19338e50.
nanoparticles into model cell membranes. Nat Commun 110. Derfus AM, Chan WCW, Bhatia SN. Probing the cytotoxicity of
2014;5:4482. semiconductor quantum dots. Nano Letters 2004;4(1):11e8.
91. Chen AA, et al. Quantum dots to monitor RNAi delivery and improve 111. Chibli H, et al. Cytotoxicity of InP/ZnS quantum dots related to
gene silencing. Nucleic Acids Res 2005;33(22). p. e190-e190. reactive oxygen species generation. Nanoscale 2011;3(6):2552e9.
92. Nel AE, et al. Understanding biophysicochemical interactions at the 112. Brunetti V, et al. InP/ZnS as a safer alternative to CdSe/ZnS core/
nanoebio interface. Nat Mater 2009;8:543. shell quantum dots: in vitro and in vivo toxicity assessment.
93. Algar WR, Krull UJ. Adsorption and hybridization of oligonucleotides Nanoscale 2013;5(1):307e17.
on mercaptoacetic acid-capped CdSe/ZnS quantum dots and quantum 113. Wang Y, et al. Quantum-dot-based theranostic micelles conjugated
DotOligonucleotide conjugates. Langmuir 2006;22(26):11346e52. with an anti-EGFR nanobody for triple-negative breast cancer
94. Chen Y, et al. Manganese oxide-based multifunctionalized meso- therapy. ACS Appl Mater Interfaces 2017;9(36):30297e305.
porous silica nanoparticles for pH-responsive MRI, ultrasonography 114. Ye F, et al. Biodegradable polymeric vesicles containing magnetic
and circumvention of MDR in cancer cells. Biomaterials nanoparticles, quantum dots and anticancer drugs for drug delivery
2012;33(29):7126e37. and imaging. Biomaterials 2014;35(12):3885e94.
95. D’Agata R, Palladino P, Spoto G. Streptavidin-coated gold nano- 115. Mitra RN, et al. An activatable multimodal/multifunctional nanop-
particles: critical role of oligonucleotides on stability and fractal robe for direct imaging of intracellular drug delivery. Biomaterials
aggregation. Beilstein J Nanotechnol 2017;8:1e11. 2012;33(5):1500e8.
96. Zhang Y, et al. Mapping DNA quantity into electrophoretic mobility 116. Maxwell T, et al. Non-cytotoxic quantum dot-chitosan nanogel
through quantum dot nanotethers for high-resolution genetic and biosensing probe for potential cancer targeting agent. Nanomaterials
epigenetic analysis. ACS Nano 2012;6(1):858e64. 2015;5(4):2359e79.
97. Smith AM, et al. A systematic examination of surface coatings on 117. Li Q-L, et al. pH and glutathione dual-responsive dynamic cross-
the optical and chemical properties of semiconductor quantum dots. linked supramolecular network on mesoporous silica nanoparticles
Phys Chem Chem Phys 2006;8(33):3895e903. for controlled anticancer drug release. ACS Appl Mater Interfaces
98. Banerjee A, et al. Quantum dotseDNA bioconjugates: synthesis to 2015;7(51):28656e64.
applications. Interface Focus 2016;6(6). 118. Banerjee S, et al. Quantum dot-based OFF/ON probe for detection
99. Beaune G, et al. Luminescence of polyethylene glycol coated of glutathione. J Phys Chem C 2009;113(22):9659e63.
CdSeTe/ZnS and InP/ZnS nanoparticles in the presence of copper 119. Calvert P, et al. Clinical studies of reversal of drug resistance based
cations. ChemPhysChem 2011;12(12):2247e54. on glutathione. Chem Biol Interact 1998;111e112:213e24.
Quantum Dots Chapter | 15 265

120. Liu K-K, et al. Large-scale synthesis of ZnO nanoparticles and their 139. Cai X, et al. pH-responsive ZnO nanocluster for lung cancer
application as phosphors in light-emitting devices. Opt Mater Ex- chemotherapy. ACS Appl Mater Interfaces 2017;9(7):5739e47.
press 2017;7(7):2682e90. 140. Roshini A, et al. pH-sensitive tangeretin-ZnO quantum dots exert
121. Ostrovsky S, et al. Selective cytotoxic effect of ZnO nanoparticles apoptotic and anti-metastatic effects in metastatic lung cancer cell
on glioma cells. Nano Res 2009;2(11):882e90. line. Mater Sci Eng C 2018;92:477e88.
122. Guo D, et al. Synergistic cytotoxic effect of different sized 141. Puvvada N, et al. Novel ZnO hollow-nanocarriers containing
ZnO nanoparticles and daunorubicin against leukemia cancer paclitaxel targeting folate-receptors in a malignant pH-
cells under UV irradiation. J Photochem Photobiol B microenvironment for effective monitoring and promoting breast
2008;93(3):119e26. tumor regression. Sci Rep 2015;5:11760.
123. Cory H, et al. Preferential killing of cancer cells and activated human 142. Tripathy N, et al. Enhanced anticancer potency using an acid-
T cells using ZnO nanoparticles. Nanotechnology 2008;19(29):295103. responsive ZnO-incorporated liposomal drug-delivery system.
124. Langlais M, Tajmir-Riahi HA, Savoie R. Raman spectroscopic study Nanoscale 2015;7(9):4088e96.
of the effects of Ca2þ, Mg2þ, Zn2þ, and Cd2þ ions on calf thymus 143. Hahn MA, Tabb JS, Krauss TD. Detection of single bacterial
DNA: binding sites and conformational changes. Biopolymers pathogens with semiconductor quantum dots. Anal Chem
1990;30(7e8):743e52. 2005;77(15):4861e9.
125. Martínez-Balbás MA, Jiménez-García E, Azorín F. Zinc(II) ions 144. Crespo RF, Perez OJP, Ramirez C. Total count of Sal-
selectively interact with DNA sequences present at the TFIIIA monella typhimurium coupled on water soluble CdSe
binding site of the Xenopus 5S-RNA gene. Nucleic Acids Res quantum dots by fluorescence detection. J Electron Mater
1995;23(13):2464e71. 2018;47(8):4379e84.
126. Chakraborty P, et al. Sensitization of cancer cells to cyclophos- 145. Lai IP-J, et al. Solid-state synthesis of self-functional carbon quan-
phamide therapy by an organoselenium compound through ROS- tum dots for detection of bacteria and tumor cells. Sens Actuators B
mediated apoptosis. Biomed Pharmacother 2016;84:1992e9. Chem 2016;228:465e70.
127. Abraham SA, et al. Formation of transition metal-doxorubicin 146. Chen H, et al. Versatile antimicrobial peptide-based ZnO quantum
complexes inside liposomes. Biochim Biophys Acta Biomembr dots for in vivo bacteria diagnosis and treatment with high speci-
2002;1565(1):41e54. ficity. Biomaterials 2015;53:532e44.
128. Fiallo MML, et al. How Fe3þ binds anthracycline antitumour 147. Lu Z, et al. Mechanism of antimicrobial activity of CdTe quantum
compounds. The myth and the reality of a chemical sphinx. J Inorg dots. Langmuir 2008;24(10):5445e52.
Biochem 1999;75(2):105e15. 148. Dumas E-M, et al. Toxicity of CdTe quantum dots in bacterial
129. Barick KC, Nigam S, Bahadur D. Nanoscale assembly of mesoporous strains. IEEE Trans Nanobioscience 2009;8(1):58e64.
ZnO: a potential drug carrier. J Mater Chem 2010;20(31):6446e52. 149. Mir IA, et al. Antimicrobial and biocompatibility of highly fluo-
130. Muhammad F, et al. Acid degradable ZnO quantum dots as a plat- rescent ZnSe core and ZnSe@ ZnS core-shell quantum dots.
form for targeted delivery of an anticancer drug. J Mater Chem J Nanoparticle Res 2018;20(7):174.
2011;21(35):13406e12. 150. Jin T, et al. Antimicrobial efficacy of zinc oxide quantum dots
131. Zhang ZY, et al. Biodegradable ZnO@polymer coreeshell nano- against Listeria monocytogenes, Salmonella enteritidis, and
carriers: pH-triggered release of doxorubicin In Vitro. Angew Chem Escherichia coli O157: h7. J Food Sci 2009;74(1):M46e52.
Int Ed 2013;52(15):4127e31. 151. Dutta RK, et al. Studies on antibacterial activity of ZnO nano-
132. Liu J, et al. Zinc oxide nanoparticles as adjuvant to facilitate particles by ROS induced lipid peroxidation. Colloids Surfaces B
doxorubicin intracellular accumulation and visualize pH- Biointerfaces 2012;94:143e50.
responsive release for overcoming drug resistance. Mol Pharm 152. Luo Z, et al. Cooperative antimicrobial activity of CdTe quantum
2016;13(5):1723e30. dots with rocephin and fluorescence monitoring for Escherichia coli.
133. Ye D-X, et al. ZnO-based Nanoplatforms for labeling and treatment J Colloid Interface Sci 2011;362(1):100e6.
of mouse tumors without detectable toxic Side effects. ACS Nano 153. Jijie R, et al. Enhanced antibacterial activity of carbon dots func-
2016;10(4):4294e300. tionalized with ampicillin combined with visible light triggered
134. Zhang J, et al. Dextran microgels loaded with ZnO QDs: pH- photodynamic effects. Colloids Surfaces B Biointerfaces
triggered degradation under acidic conditions. J Appl Polym Sci 2018;170(1):347e54.
2018;135(6):n/a. 154. Dai Q, et al. Quantifying the ligand-coated nanoparticle delivery to
135. Cai X, et al. pH-sensitive ZnO quantum dots-doxorubicin nano- cancer cells in solid tumors. ACS Nano 2018;12(8):8423e35.
particles for lung cancer targeted drug delivery. ACS Appl Mater 155. Greish K, et al. Macromolecular therapeutics. Clin Pharmacokinet
Interfaces 2016;8(34):22442e50. 2003;42(13):1089e105.
136. Muhammad F, et al. pH-Triggered controlled drug release from 156. Salmaso S, Caliceti P. Stealth properties to improve therapeutic
mesoporous silica nanoparticles via intracelluar dissolution of ZnO efficacy of drug nanocarriers. J Drug Delivery 2013;2013:19.
nanolids. J Am Chem Soc 2011;133(23):8778e81. 157. Park K. Facing the truth about nanotechnology in drug delivery.
137. Kim S, Lee SY, Cho H-J. Doxorubicin-wrapped zinc oxide nano- ACS Nano 2013;7(9):7442e7.
clusters for the therapy of colorectal adenocarcinoma. Nano- 158. Chen F, et al. Ultrasmall targeted nanoparticles with engineered
materials 2017;7(11):354/1e354/13. antibody fragments for imaging detection of HER2-overexpressing
138. Wang J, et al. Exploration of zinc oxide nanoparticles as a multi- breast cancer. Nat Commun 2018;9(1):4141.
target and multifunctional anticancer nanomedicine. ACS Appl 159. Kang Y-F, et al. Carbon quantum dots for zebrafish fluorescence
Mater Interfaces 2017;9(46):39971e84. imaging. Sci Rep 2015;5:11835.

You might also like