Quantum Dot Therapeutics A New Class of
Quantum Dot Therapeutics A New Class of
Quantum Dot Therapeutics A New Class of
Abstract
Traditional therapeutics and vaccines represent the bedrock of modern medicine, where isolated biochemical
molecules or designed proteins have led to success in treating and preventing diseases. However, several adaptive
pathogens, such as multidrug-resistant (MDR) superbugs, and rapidly evolving diseases, such as cancer, can evade
such molecules very effectively. This poses an important problem since the rapid emergence of multidrug-
resistance among microbes is one of the most pressing public health crises of our time—one that could claim
more than 10 million lives and 100 trillion dollars annually by 2050. Several non-traditional antibiotics are now
being developed that can survive in the face of adaptive drug resistance. One such versatile strategy is redox
perturbation using quantum dot (QD) therapeutics. While redox molecules are nominally used by cells for
intracellular signaling and other functions, specific generation of such species exogenously, using an
electromagnetic stimulus (light, sound, magnetic field), can specifically kill the cells most vulnerable to such species.
For example, recently QD therapeutics have shown tremendous promise by specifically generating superoxide
intracellularly (using light as a trigger) to selectively eliminate a wide range of MDR pathogens. While the efficacy of
such QD therapeutics was shown using in vitro studies, several apparent contradictions exist regarding QD safety
and potential for clinical applications. In this review, we outline the design rules for creating specific QD therapies
for redox perturbation; summarize the parameters for choosing appropriate materials, size, and capping ligands to
ensure their facile clearance; and highlight a potential path forward towards developing this new class of radical
QD therapeutics.
Keywords: Radical antimicrobials, Multidrug-resistant superbugs, Quantum dot therapeutic, Reactive oxygen species
the cell, the nanoparticle or QD therapy can make use of superoxide was found to be a potent bactericidal at low
the presence of oxygen, water, and if required, an exter- nanomolar doses—killing a range of multidrug-resistant
nal trigger. Therefore a wide range of reactive oxygen (MDR) pathogens without affecting the viability or growth
species (ROS, e.g. superoxide O2●-, hydroxyl OH●, sing- of host mammalian cells in in vitro measurements [19, 20,
let oxygen 1O2, and hydrogen peroxide H2O2) and react- 24, 29]. This difference in nanotherapeutic toxicity be-
ive nitrogen species (RNS, e.g. nitric oxide NO●, tween host and the targeted pathogen is important for de-
peroxynitrite ONOO−) can be formed intracellularly signing the safest possible treatment. Biological
using redox chemistry. Since these species are respon- specificity enables a treatment to effectively clear in-
sible for a broad range of physiology and pathology in fections while preserving the host cells. Although the
living organisms [22, 23], they have been investigated for superoxide anion has a high thermodynamic capacity
such potential applications as cancer therapies and novel to be a strong oxidant, its lack of reactivity with cel-
antimicrobials. Therefore, specific intracellular gener- lular components at physiological pH (largely due to
ation of these species can drastically affect the specificity electrostatic repulsion with negatively charged biomo-
of ROS/RNS therapy using the proposed redox lecules)--except for the inactivation of biosynthetic
perturbation. enzymes containing labile iron-sulfur clusters--is key
Recently, our group assessed these different ROS and to its selectivity [25, 30]. Further, the role of iron se-
RNS species as potential therapeutics [24]. Using these questration in host colonization makes pathogenic
species intracellularly, we determined their respective bacteria particularly vulnerable to superoxide com-
minimum inhibitory concentration (MIC) values. We pared to hosts [31, 32]. Therefore, while several ROS
found a bactericidal effect for several species at high species like hydroxyl radicals are indiscriminate oxi-
threshold concentrations (singlet oxygen: 1 mM; peroxide: dants and can readily oxidize proteins, lipids, and nu-
10 mM; hydroxyl radical > 10 mM; nitric oxide > 1 mM, cleic acids [8, 33], prior studies and our experiments
Fig. 1) [24, 25], where these redox species would be toxic indicate specificity in the of superoxide anions [19,
even for host mammalian cells [26–28]. However, 20, 24, 29, 34, 35]. Given the specificity of
Fig. 1 Identifying pathogen vulnerability using redox perturbation with different ROS. Compared to singlet oxygen and hydroxyl radicals,
superoxide and peroxide have much longer diffusion lengths and half-lives in the cellular environment (red circles, not to scale) [24, 82]. Singlet
oxygen and hydroxyl radicals are also more nonselective – they react rapidly with an abundance of endogenous biomolecules. Superoxide is
more selective, partly due to its negative charge, and it reacts with very particular consequential cellular targets such as iron-sulfur clusters [25].
The endogenous bacterial defense against superoxide is less abundant than the defense against nonselective ROS. This leads to a drastically
lower observed toxicity threshold when compared to other species [24]. Unlike other ROS, superoxide offers a large window of dosage that yields
toxicity in pathogens and nontoxicity in hosts [19, 24]
Levy et al. Journal of Biological Engineering (2019) 13:48 Page 3 of 12
superoxide’s mechanism of action, the low MIC value nominal (bulk) bandgap values, we obtained a list of ‘fa-
for pathogens and higher tolerance in host mamma- vorable’ materials for the proposed QD nanotherapy.
lian cells, its long lifetime and large diffusion length Keeping in mind the extinction of light as it enters the
make it an ideal candidate for selective redox therapy. skin considering the most common constituents as
water, hemoglobin, melanin, etc. [20, 36, 37], there is a
Selective redox activation using quantum states window of nominal biological transparency (~ 800–1300
QDs, or semiconductor nanocrystals, have size-, shape-, nm wavelength), which narrows the material and band-
and composition-tunable quantum states for reduction gap considerations further (Fig. 2b-d). Materials which
and oxidation reactions. These states can be triggered by absorb violet and UV light are less suitable for QD
external electromagnetic radiation like light, and have nanotherapy—such short wavelengths of light will be
demonstrated a promising role in non-traditional redox quickly scattered or absorbed near the surface of animal
therapy [17–20, 24, 29]. Precise control over their tissue. This lack of penetration would make it extremely
photogenerated electron and hole states provides a challenging to use wide-bandgap materials to treat sys-
unique ability to tailor their photochemistry in the cellu- temic infections. Red and near-infrared absorbing QDs
lar environment, thereby providing control over intracel- would be far less susceptible to this issue. Therefore,
lular redox species. As the first step towards designing near-infrared materials like cadmium telluride (CdTe)
an effective QD therapeutic, we assessed a range of dif- [19, 20, 24, 29], copper indium sulfide (CIS2) [19], in-
ferent materials, along with their corresponding (bulk) dium phosphide (InP), and gallium arsenide (GaAs)
reduction and oxidation states (Fig. 2a). To select for could serve as good candidates for selective antimicro-
specific intracellular generation of superoxide, the reduc- bial, material stability, cytotoxicity, and surfaces [17, 18].
tion potential should exceed − 0.33 V on normal hydro- Using dopants and bandgap engineering, it is also pos-
gen electrode (NHE) scale. Simultaneously, to avoid the sible to improve the suitability of some other materials.
formation of other non-specific ROS species that can For instance, carbon QDs and silicon QDs have many
cause indiscriminate cell damage, the oxidation potential reported biological applications in bio-imaging, cancer
should be less than 1.8 V NHE. Using this metric as a se- therapy, as well as some reports describing ROS-medi-
lection criterion for selective redox antimicrobial ther- ated therapy [38–41]. Depending on particle size and
apy, and classifying the materials on the basis of their dopants,the optical properties of these materials can be
Fig. 2 Criteria for material selection for QD therapeutic against MDR superbugs. a) Conduction band (red) and valence band (blue) positions for
select semiconductors in bulk, according to references cited in reviews herein [83, 84]. Band edge positions shown in relation to thresholds for
superoxide (green) and hydroxyl radical (red) generation. Many of these materials only absorb ultraviolet light or are unable to generate superoxide. b)
By applying rational constraints to this list of materials, we can narrow this (non-exhaustive) list of candidates. c) These candidates could potentially
generate therapeutic superoxide using visible or NIR light, which penetrates deeper through tissue than UV (d), reproduced with permission from the
American Chemical Society37
Levy et al. Journal of Biological Engineering (2019) 13:48 Page 4 of 12
specifically tailored to decrease the energy of light re- present issues of colloidal stability or inherent cytotoxicity.
quired for photoactivation [42]. The same approach can A good solution to enable using such materials could be
also be extended to metal oxide materials. to use that material as a redox-active core, covered by a
Due to several contradictory reports of ROS gener- thin shell of biocompatible material [29]. Further, material
ation and potential “therapeutic” action of two cytotoxicity is often tied to a nanoparticle’s physical size,
FDA-approved materials, zinc oxide (ZnO) and titanium hydrodynamic radius, and surface charge (zeta potential).
dioxide (TiO2) QDs and nanoparticles [43–49], we eval- These factors can directly affect a nanoparticle’s affinity
uated their redox properties and demonstrated bandgap for surrounding biomolecules and tissue. For example,
and redox state engineering approach proposed in this worsened zeta potential can hinder colloidal stability and
review. First, unmodified or undoped ZnO and TiO2 potentially result in particle aggregation. This could lead
nanoparticles were tested using electron paramagnetic to selective accumulation of QDs in some organs like kid-
resonance (EPR) spectroscopy technique. In order to ney, spleen, and liver [17, 18], where the host cells have
evaluate the short-lived radical species formed from small pore sizes, but are quickly cleared through the rest
redox chemistry, we used a spin-trapping method to of the organs and blood circulation. Nominally, QDs with
form more stable adducts. Both ZnO and TiO2 nanopar- a hydrodynamic diameter below ~ 10 nm can be cleared
ticles showed only hydroxyl radical formation upon exci- from the body in in vivo animal tests. After accumulating
tation with ultraviolet light above their bandgap in organs, due to lack of typical metabolism as seen in
(Fig. 3a,b). This would be problematic for potential small molecules, one hypothesis suggests the potential for
nanotherapy because both ultraviolet light and hydroxyl surface material leaching to occur, causing the QDs to
radicals are indiscriminately toxic to all cells. Careful “shrink” in size and clear away. This release of metal ele-
electrochemical measurements revealed that, while the ments could present unpredictable and undesired host
ZnO reduction potential is too low for superoxide for- toxicity issues. Therefore, the choice of biocompatible ma-
mation, its oxidation potential is very high—leading to terial, at least on the QD surface, can be critical for suc-
hydroxyl generation upon light activation (Fig. 3c,e). cessful application, reducing potential toxicity concerns
Therefore, even if ZnO nanoparticles were doped with a for host cells. Evidence in support of this hypothesis from
cation (to reduce the nominal reduction potential) or literature can be seen in Table 1 [85–104], where different
anion (to reduce the oxidation potential), the visible light core materials (like CdSe and CdTe) when coated with
absorbing nanoparticles still could not form superoxide other more benign/biocompatible materials, display a sig-
(Fig. 3e,g). This was further confirmed via electrochem- nificant reduction in toxicity. We reviewed a number of
ical measurements by removing oxygen, where direct such studies in literature with a wide range of sizes, hydro-
hole-injection into water leads to the formation of hy- dynamic radius, and in vitro and in vivo studies. As a re-
droxyl radical (Fig. 3c). Evaluation of TiO2 nanoparticles sult, we found that even large QDs with significant
showed more promising results, however. While the retention made of/coated with less toxic elements, dis-
oxidation potential of undoped TiO2 was too high, the played much lower cytotoxicity. Further, materials that do
reduction potential was suitably matched for superoxide not create any toxic ROS, such as hydroxyl or singlet oxy-
formation (Fig. 3d, f ). Therefore, anion-doped TiO2 gen, also displayed low cytotoxicity to the host cells. These
nanoparticles, in principle, should form therapeutic materials could be used either as stable single-material
superoxide. However, the presence of oxygen vacan- QDs, or as coating/shell for QDs with a different core ma-
cies and resulting Ti3+ ions nominally present on this terial better suited for redox perturbation.
oxide material surface [50–52] catalyzes rapid Fenton
chemistry to dismutate superoxide, converting it into QD ligands, size, and clearance
toxic hydroxyl radicals [53]. To prevent such un- Comparing data from identical QDs/nanoparticles with
wanted dismutation, we coated the surface of TiO2 different ligands, charge, and hence resulting different
nanoparticles with a zinc sulfide (ZnS) shell, and saw hydrodynamic radius, we observed significant differences
significant superoxide formation (in visible light) in their retention and cytotoxicity (Table 2) [18–20],
using anion-doped (N-doped) TiO2 core/ZnS shell [105–109]. Notably, even across materials with different
nanoparticles (Fig. 3f, h). toxicity, e.g. CdSe, CdTe, and Au, surface ligands clearly
influence retention and cytotoxicity. At identical QD/
Choosing the right material(s) for QD therapeutic nanoparticle core sizes, positively charged ligands (cyste-
While the choice of an appropriate redox-active material amine) show indiscriminate adhesion to different nega-
is important for selective therapeutic action, as shown tively charged biomolecules, creating a protein “corona”
above, the QD surface plays a key role in cellular photo- that increases its hydrodynamic radius significantly. This
chemistry and biocompatibility. Many materials have ap- effectively increases QD retention and resulting cytotox-
propriate redox properties to enable QD therapy, but icity [17, 18, 29, 54, 55]. Switching to negatively-charged
Levy et al. Journal of Biological Engineering (2019) 13:48 Page 5 of 12
Fig. 3 Bandgap and redox state engineering for therapeutic radicals. a-b) EPR spectra for UV illuminated ZnO and TiO2 nanoparticles, respectively,
showing DMPO-OH peaks corresponding to the spin-trapped adduct of hydroxyl radicals. c-d) cyclic voltammograms for ZnO and TiO2, respectively, in
deoxygenated water. Without a source of oxygen, TiO2 generates no radical signal but ZnO shows a peak corresponding to hydroxyl
radicals – indicating superoxide-generating ability from TiO2 but not ZnO. e-f) reduction and oxidation state positions for ZnO and TiO2,
respectively, as well as the effects of doping. Anionic doping shifts the VB and cationic doping shifts the reduction potential. g-h) EPR
spectra for engineered ZnO and TiO2, respectively. Anionic and cationic doping of ZnO, as well as anionic doping of TiO2, yield hydroxyl
production with visible light. Cationic doping of TiO2 shows no radical signal – indicating reliance on the reduction potential for superoxide generation.
EPR spectra for N-TiO2/ZnS shows clear DMPO-OOH peaks corresponding to the superoxide radical adduct
ligands (mercaptopropionic acid) at same/similar core retention, with low/moderate toxicity. However, a simi-
size meanwhile reduces indiscriminate biomolecule at- larly sized zwitterionic-ligand (cysteamine) results in low
tachment and lowers/eliminates toxicity. This ligand still hydrodynamic radius and toxicity. These findings can be
results in higher hydrodynamic radius and higher explained by the lack of formation of a protein corona
Levy et al. Journal of Biological Engineering (2019) 13:48 Page 6 of 12
Table 1 Review of in vitro and in vivo toxicity reported for relevant core/shell QDs [85–104]
and higher rates of renal clearance in in vivo animal (zwitterionic) to maintain a low-hydrodynamic radius,
studies. This points to a clear strategy of controlling the high rates of clearance, and low toxicity, can be
QD core/shell size, along with ligand and charge, so that employed for a suitable therapeutic bottom-up design
the total hydrodynamic diameter remains below 10–15 strategy for redox QD therapies.
nm. Taken together, this 3-layer design approach con-
sists of: 1) QD made with core material with tuned Future outlook/approaches
reduction-oxidation potentials for selective generation of Addressing host toxicity
superoxide for as antimicrobial for MDR superbugs; 2) There are three major potential sources of toxicity for
non-toxic and biocompatible shell core or shell material, the host mammalian cells that the current and future
resulting in high chemical stability and low material non-traditional QD therapeutics need to address: 1)
leaching and cytotoxicity; and 3) ligand design Acute material toxicity leading to loss of host cell
Levy et al. Journal of Biological Engineering (2019) 13:48 Page 7 of 12
Table 2 Review of common charged ligands and reported effects on biodistribution and toxicity [18–20], [105–109]
viability or growth; 2) Oxidative stress; and 3) DNA cytotoxicity concerns in in vitro screenings, and only ad-
damage and carcinogenesis. To address these concerns, vance candidates that show clear differences in MIC
the QD therapeutics first need to address acute values for the host and pathogen cells. This can be
Levy et al. Journal of Biological Engineering (2019) 13:48 Page 8 of 12
established by designing the mechanism of action after adjuvant nanoparticles (< 20–50 nm) which can evoke a
careful consideration of potential vulnerabilities in the stronger immune response, aiding the QD nanothera-
pathogen’s cellular environment and metabolism. This, peutic [56, 57], by acting as “Nano-Immunotherapy”
therefore, leads to a more directed and dynamic (Fig. 4a); and b) ~ 20–50 nm nanoparticles coated with
approach to counter the adaptive resistance in these [Fe-S] cluster [58] complexes as countermeasures, for
MDR pathogens. Similarly, the use of specific ROS like size-selective uptake in host cells (Fig. 4b), to reduce the
superoxide–which can be selectively toxic to superoxide concentrations and ROS stress in the host
iron-sequestering pathogens–creates a clear window for and protect them against any potential toxic mechanism
differences in MIC values between host and pathogens. (Fig. 4a).
This therapeutic window of concentration has been suc-
cessfully identified and utilized as a therapeutic in in Nano-Immunotherapeutics
vitro studies to target MDR pathogens, while preserving are a class of new immunomodulatory materials, where
host viability and growth. Further experiments are their physicochemical properties: size, shape, surface
needed to determine the transcriptomic response to charge, molecular weight, roughness, and hydrophobi-
superoxide therapy. More specifically, this is necessary city, are used to mimic normal cellular components and
to understand how oxidative stress from the proposed evade or suppress immune response (immune-evasive or
treatment affects the host. So far, the experimental evi- immune-suppressing), or designed to inflame the host
dence in literature points to non-perturbative stress re- immune response for potential therapeutic effect
sponse of the host to specific ROS like superoxide, and (immune-activating materials) [56, 57]. For instnace,
to a mechanism of action limited primarily to enzyme smaller nanoparticles have higher uptake and trafficking,
deactivation and indiscriminate DNA/RNA damage or allowing them to reach the lymph nodes—evoking
genotoxicity. higher levels of surface maturation markers and inflam-
To further alleviate these concerns, our lab is develop- matory cytokine secretion [59–61]. Further, asymmet-
ing two nanoparticle therapeutic adjuvant and “counter- rical shapes, such as nanorods, show similar trends in
measures”, made from FDA approved materials, to be their immune response: Nanorods with similar radius
supplied with the QD therapeutic: a) larger-sized but smaller length show higher uptake [61]. But longer
Fig. 4 Addressing QD toxicity and future directions. a) Schematic and summary of three proposed types of non-traditional therapeutic, adjuvant,
and countermeasure nanoparticles. QD therapeutics (top) using superoxide generation; Nano-immunotherapy (middle) using 20–50 nm benign
nanorods to trigger an immune response; and a countermeasure (bottom) using large or small benign nanoparticles coordinated with Fe-S to
serve as a host-specific nano-antioxidant. b) Depiction of host-specific protection using the larger nanoparticles coated with [Fe-S] clusters as
countermeasures against the superoxide ROS stress from QD therapeutic. c) Depiction of probiotic-specific protection using transcriptomic/
proteomic targeting with countermeasure nanoparticles
Levy et al. Journal of Biological Engineering (2019) 13:48 Page 9 of 12
nanorods induced a higher inflammatory response hydrophobicity [63, 70, 71]; and 4) have a designed surface
(IL-1α and TNF- α) because of frustrated phagocytic in- charge to tune the inflammatory response (Fig. 4a) [64, 72,
teractions with cells, due to their larger size [62]. In- 73]. These nano-immunotherapeutic nanoparticles could,
creasing the hydrophobicity of the nanomaterials surface reversibly, also be used to downregulate the immune
identifies them as foreign, and potentially dangerous ma- response and inflammation, in case the QD nanotherapeu-
terials, by the immune system. This increases the tic has any adverse/side-effects due to retention of
gene-expression of pro-inflammatory cytokines [63]. Coat- over-activity. Further, the size-selected uptake of nanoparti-
ing the QD or other nanomaterial surfaces with hydro- cles acting as counter-measures for the host cells would be
philic molecules reduces the surface protein adsorption designed to counter any ROS stress, deactivation of super-
and decreases interaction with immune cells, thereby re- oxide in the host, and reduce any potential for genotoxicity
ducing immunomodulatory response. Studies on the effect from the QD therapeutic, using a coating of [Fe-S] clusters
of the surface charge have shown confounding effects with on these nanoparticles (Fig. 4a,b) [58]. Such coatings can be
other dominant physiochemical properties. Gold nanorods easily created using a hydrophobic-hydrophilic surface
with positive surface charge (amine-terminated ligands) interaction, and will be used with a small subset of FDA ap-
exhibit expression of anti-inflammatory surface antigens proved materials, like ZnO, TiO2, or silica nanoparticles.
and negatively-charged (carboxylic acid-terminated li-
gands) surfaces induced expression of pro-inflammatory Improving selective uptake in different cell-types (host
genes [64]. However, other studies have concluded that and pathogen)
negatively-charged amino acid residues can sometimes One future approach to improving QD therapeutics is
prevent uptake of long fibrillized peptide materials by targeting selective uptake between host and pathogen
antigen-presenting cells, and hence prevent presentation (Fig. 4b), as well as between different pathogens (e.g.
of epitope peptides–thereby inhibiting immune function pathogenic vs. probiotics, Fig. 4c). Size can be an im-
[65]. Overall, zwitterionic ligands or surface charges pre- portant factor in tuning uptake between host and patho-
vent accumulation/adsorption of biomolecules like pro- gens [74, 75]. By selecting for the appropriate size, a QD
teins (biofouling), thereby evading foreign-body response therapy can selectively generate therapeutic superoxide
[56, 57, 66]. These elements of immunomodulation were and induce pathogen-killing, while protecting the host
used in our QD design (small size, spherical shape, small cells using nano-countermeasures. For selectivity be-
hydrophilic ligands, and zwitterionic surface charge). By tween different types of bacteria, such as pathogenic
reducing the potential of non-specific inflammation of the strains and gut microbiota, target specificity must be
host immune system, the potential side effects of the QD considered. Reaching such targets would require the
therapeutic nanoparticles could be avoided. identification of the genomic, transcriptomic, or prote-
Other aspects of immune modulation using physio- omic factors that separate the distinct strains. QD thera-
chemical properties of nanoparticles depend on their peutics can be easily coated with peptides or DNA/RNA
molecular weight and surface roughness. The effect of molecules with appropriate target sequences (Fig. 4c)
surface topography at the nanoscale, along with surface [76–81]. Using this methodology, similar sized
chemistry was used to understand the innate immune pathogen-targeted QDs can be selectively uptaken by the
response. While surface acidity has a larger role in pathogens as a QD therapeutic, while similarly sized
immunomodulation, surface roughness directly is corre- countermeasures can be selectively transported into the
lated with enhanced matrix metalloproteinase-9 produc- probiotic bacteria, further protecting them from adverse
tion by primary neutrophils, and a decrease in the effects of the QD therapeutic. This approach can boost
pro-inflammatory cytokine secretion from primary mac- the efficacy of QD therapeutics while reducing potential
rophages [67]. This immunomodulation via surface side-effects. Importantly, the window of QD therapeutic
roughness could be attributed to a reduction in inflam- flux between host and pathogens can be further ex-
mation and increased healing on encountering rough panded to provide more immediate and effective relief
surfaces. to patients.
Based on the design rules summarized here, the adjuvant
Nano-Immunotherapeutic will: 1) be larger-size nanoparti- Conclusions
cles than QD therapeutics (< 20–50 nm), but small enough In conclusion, this review summarizes the potential, exist-
that they easily transport to reach lymph nodes [68, 69] and ing, state-of-the-art, and future outlook for an emerging
initiates/upregulates the innate immune response of the class of radical QD therapeutics. Here, we specifically
body [59], to aid the QD therapeutic and fight pathogens; sought to show several aspects of QD design, geared to-
2) be shaped as short nanorods, rather than spherical nano- wards treating MDR superbug infections. By tailoring the
particles, for preferential uptake and stronger immunomo- stimuli-triggered photochemistry, inherent materials, and
dulation [64]; 3) have induced surface roughness and chosen mechanism of action, a bottom-up rational design
Levy et al. Journal of Biological Engineering (2019) 13:48 Page 10 of 12
strategy was outlined for the QD therapeutic. This ap- Ethics approval and consent to participate
proach begins with a mechanism of redox action that tar- Not applicable.
19. Courtney CM, Goodman SM, McDaniel JA, Madinger NE, Chatterjee A, 45. Sirelkhatim A, Mahmud S, Seeni A, Kaus NHM, Ann LC, Bakhori SKM, Hasan
Nagpal P. Photoexcited quantum dots for killing multidrug-resistant H, Mohamad D. Review on Zinc Oxide Nanoparticles: Antibacterial Activity
Bacteria. Nat Mater. 2016;15(5):529–34. and Toxicity Mechanism. Nano-Micro Letters. 2015:219–42.
20. Courtney, C. M.; Goodman, S.; Nagy, T.; Levy, M.; Bhusal, P.; Madinger NE, 46. Heinlaan M, Ivask A, Blinova I, Dubourguier HC, Kahru A. Toxicity of Nanosized
Detweiler CS, Nagpal P, Chatterjee A. Potentiating Clinical Drug Resistant and bulk ZnO, CuO and TiO2 to Bacteria Vibrio Fischeri and crustaceans daphnia
Bacteria via Stimuli-Activated Superoxide Generation. Sci. Adv. 2017, 3 magna and Thamnocephalus Platyurus. Chemosphere. 2008.
(October), e1701776. 47. Song W, Zhang J, Guo J, Zhang J, Ding F, Li L, Sun Z. Role of the dissolved
21. Loo C, Lowery A, Halas N, West J, Drezek R. Immunotargeted Nanoshells for zinc ion and reactive oxygen species in cytotoxicity of ZnO nanoparticles.
integrated Cancer imaging and therapy. Nano Lett. 2005;5(4):709–11. Toxicol Lett. 2010;199(3):389–97.
22. Di Meo, S.; Reed, T. T.; Venditti, P.; Victor, V. M. Role of ROS and RNS Sources 48. Lin X, Li J, Ma S, Liu G, Yang K, Tong M, Lin D. Toxicity of TiO2nanoparticles
in Physiological and Pathological Conditions; 2016; Vol. 2016. to Escherichia coli: effects of particle size, Crystal Phase and Water Chemistry.
23. D’Autréaux B, Toledano MB. ROS as Signalling molecules: mechanisms that PLoS One. 2014;9(10):e110247.
generate specificity in ROS homeostasis. Nat Rev Mol Cell Biol. 2007;8(10): 49. Pagnout C, Jomini S, Dadhwal M, Caillet C, Thomas F, Bauda P. Role of
813–24. electrostatic interactions in the toxicity of titanium dioxide nanoparticles
24. Levy M, Courtney CM, Chowdhury PP, Ding Y, Grey EL, Goodman SM, toward Escherichia Coli. Colloids Surfaces B Biointerfaces. 2012;92:315–21.
Chatterjee A, Nagpal P. Assessing different reactive oxygen species as 50. Singh V, Beltran IJC, Ribot JC, Nagpal P. Photocatalysis deconstructed:
potential antibiotics: selectivity of intracellular superoxide generation using Design of a new Selective Catalyst for artificial photosynthesis. Nano Lett.
quantum dots. Bio Mater: ACS Appl; 2018. 2014;14(2):597–603.
25. Imlay JA. Pathways of oxidative damage. Annu Rev Microbiol. 2003;57(1): 51. Alivov Y, Singh V, Ding Y, Cerkovnik LJ, Nagpal P. Doping of wide-Bandgap
395–418. titanium-dioxide nanotubes: optical, Electronic and Magnetic Properties.
26. Imlay JA, Linn S. DNA damage and oxygen radical toxicity. Science. 1988; Nanoscale. 2014;6(18):10839–49.
240(4857):1302–9. 52. Alivov Y, Funke HH, Singh V, Nagpal P. Air-pressure tunable depletion width,
27. Nakamura J, Purvis ER, Swenberg JA. Micromolar Concentrations of Hydrogen rectification behavior, and charge conduction in oxide nanotubes. ACS Appl
Peroxide Induce Oxidative DNA Lesions More Efficiently than Millimolar Mater Interfaces. 2015;7(4):2153–9.
Concentrations in Mammalian Cells. Nucleic Acids Res. 2003:1790–5. 53. Xiong LB, Li JL, Yang B, Yu Y. Ti 3+ in the Surface of Titanium Dioxide: Generation,
28. Li C-Q, Pang B, Kiziltepe T, Trudel LJ, Engelward BP, Dedon PC, Wogan GN. Properties and Photocatalytic Application. J Nanomaterials. 2012;9.
Threshold effects of nitric oxide-induced toxicity and cellular responses in 54. Lewinski N, Colvin V, Drezek R. Cytotoxicity of nanoparticles. Small. 2008;4(1):
wild-type and p53-null human Lymphoblastoid cells. Chem Res Toxicol. 26–49.
2006;19(3):399–406. 55. Hardman R. A Toxicologic Review of Quantum Dots: Toxicity Depends on
29. Goodman SM, Levy M, Li F-F, Ding Y, Courtney CM, Chowdhury PP, Erbse A, Physicochemical and Environmental Factors. Environmental Health
Chatterjee A, Nagpal P. Designing superoxide-generating quantum dots for Perspectives. 2006:165–72.
selective light-activated Nanotherapy. Front Chem. 2018;6:46. 56. Andorko JI, Jewell CM. Designing biomaterials with Immunomodulatory
30. Imlay JA. Iron-Sulphur Clusters and the Problem with Oxygen. Mol properties for tissue engineering and regenerative medicine. Med: Bioeng.
Microbiol. 2006:1073–82. Transl; 2017.
31. Iglewski, B.; Clark V. Molecular Basis of Bacterial Pathogenesis; 1990. 57. Lewis JS, Roy K, Keselowsky BG. Materials That Harness and Modulate the
32. Pan X, Yang Y, Zhang J-R. Molecular basis of host specificity in human Immune System. MRS Bull. 2014;39(1):25–34.
pathogenic Bacteria. Emerg Microbes Infect. 2014;3(3):e23. 58. Li C-B, Li Z-J, Yu S, Wang G-X, Wang F, Meng Q-Y, Chen B, Feng K, Tung C-
33. Winterbourn CC. Reconciling the Chemistry and Biology of Reactive Oxygen H, Wu L-Z. Interface-directed assembly of a simple precursor of [FeFe]–
Species. Nature Chemical Biology. 2008:278–86. H2ase mimics on CdSe QDs for photosynthetic hydrogen evolution in
34. Sawyer DT, Valentine JS. How super is superoxide? Acc Chem Res. 1981; water. Energy Environ Sci. 2013;6(9):2597–602.
14(12):393–400. 59. Fernández TD, Pearson JR, Leal MP, Torres MJ, Blanca M, Mayorga C, Le Guével
35. Sawyer DT, Valentine JS, Fridovich I, Sawyer DT, Valentine JS, Fridovich I. X. Intracellular accumulation and immunological properties of fluorescent gold
How innocuous is superoxide? Comments. Acc Chem Res. 1982;15(7):200. Nanoclusters in human dendritic cells. Biomaterials. 2015;43:1–12.
36. Sun Q-C, Ding YC, Sagar DM, Nagpal P. Photon Upconversion towards 60. Da Silva CA, Chalouni C, Williams A, Hartl D, Lee CG, Elias JA. Chitin is a size-
applications in energy conversion and bioimaging. Prog Surf Sci. 2017;92(4): dependent regulator of macrophage TNF and IL-10 production. J Immunol.
281–316. 2009;182(6):3573–82.
37. Sun QC, Mundoor H, Ribot JC, Singh V, Smalyukh II, Nagpal P. Plasmon- 61. Kumar S, Anselmo AC, Banerjee A, Zakrewsky M, Mitragotri S. Shape and
enhanced energy transfer for improved Upconversion of infrared radiation size-dependent immune response to antigen-carrying nanoparticles. J
in doped-lanthanide Nanocrystals. Nano Lett. 2014;14(1):101–6. Control Release. 2015;220:141–8.
38. Tian Z, Yao X, Hanagata N, Liu J, Zhu Y. Mesoporous silica nanoparticles 62. Padmore T, Stark C, Turkevich LA, Champion JA. Quantitative analysis of the
capped with Graphene quantum dots for potential chemo–Photothermal role of Fiber length on phagocytosis and inflammatory response by alveolar
synergistic Cancer therapy. Langmuir. 2016;33(2):591–9. macrophages. Biochim. Biophys. Acta - Gen. Subj. 2017;1861(2):58–67.
39. Chinnathambi S, Chen S, Ganesan S, Hanagata N. Silicon quantum dots for 63. Moyano DF, Goldsmith M, Solfiell DJ, Landesman-Milo D, Miranda OR, Peer
biological applications. Adv. Healthc. Mater. 2014;3(1):10–29. D, Rotello VM. Nanoparticle hydrophobicity dictates immune response. J
40. Gara PMD, Garabano NI, Portoles MJL, Moreno MS, Dodat D, Casas OR, Am Chem Soc. 2012;134(9):3965–7.
Gonzalez MC, Kotler ML. ROS Enhancement by Silicon Nanoparticles in X- 64. Bartneck M, Keul HA, Singh S, Czaja K, Bornemann J, Bockstaller M, Moeller
Ray Irradiated Aqueous Suspensions and in Glioma C6 Cells. J. Nanoparticle M, Zwadlo-Klarwasser G, Groll J. Rapid uptake of gold Nanorods by primary
Res. 2012;14(3):741. human blood phagocytes and Immunomodulatory effects of surface
41. Hassan M, Gomes VG, Dehghani A, Ardekani SM. Engineering carbon chemistry. ACS Nano. 2010;4(6):3073–86.
quantum dots for Photomediated Theranostics. Nano Res. 2018;11(1):1–41. 65. Wen Y, Waltman A, Han H, Collier JH. Switching the immunogenicity of
42. Abdelhameed M, Martir DR, Chen S, Xu WZ, Oyeneye OO, Chakrabarti S, peptide assemblies using surface properties. ACS Nano. 2016;10(10):
Zysman-Colman E, Charpentier PA. Tuning the optical properties of silicon 9274–86.
quantum dots via surface functionalization with conjugated aromatic 66. Zhang L, Cao Z, Bai T, Carr L, Ella-Menye JR, Irvin C, Ratner BD, Jiang S.
Fluorophores. Sci Rep. 2018;8(1):1–10. Zwitterionic hydrogels implanted in mice resist the foreign-body reaction.
43. Premanathan M, Karthikeyan K, Jeyasubramanian K, Manivannan G. Selective Nat Biotechnol. 2013;31(6):553–6.
toxicity of ZnO nanoparticles toward gram-positive Bacteria and Cancer 67. Christo SN, Bachhuka A, Diener KR, Mierczynska A, Hayball JD, Vasilev K. The
cells by apoptosis through lipid peroxidation. Biol. Med: Nanomedicine role of surface Nanotopography and chemistry on primary neutrophil and
Nanotechnology; 2011. macrophage cellular responses. Adv Healthc Mater. 2016;5(8):956–65.
44. Lakshmi Prasanna V, Vijayaraghavan R. Insight into the mechanism of 68. Bracho-Sanchez E, Xia CQ, Clare-Salzler MJ, Keselowsky BG. Micro and
antibacterial activity of ZnO: surface defects mediated reactive oxygen Nano Material Carriers for Immunomodulation. AM J Transplant. 2016;
species even in the dark. Langmuir. 2015. 16(12):3362–70.
Levy et al. Journal of Biological Engineering (2019) 13:48 Page 12 of 12
69. Banerjee A, Qi J, Gogoi R, Wong J, Mitragotri S. Role of Nanoparticle Size, 94. Yong KT, Roy I, Law WC, Hu R. Synthesis of cRGD-peptide conjugated near-
Shape and Surface Chemistry in Oral Drug Delivery. J. Control. Release. 2016. infrared CdTe/ZnSe Core-Shell quantum dots for in vivo Cancer targeting
70. Seong SY, Matzinger P. Hydrophobicity: An Ancient Damage-Associated and imaging. Chem Commun. 2010;46(38):7136–8.
Molecular Pattern That Initiates Innate Immune Responses. Nature Rev 95. Yong KT, Roy I, Hu R, Ding H, Cai H, Zhu J, Zhang X, Bergey EJ, Prasad
Immunol. 2004:469–78. PN. Synthesis of ternary CuInS2/ZnS quantum dot bioconjugates and
71. Li WA, Lu BY, Gu L, Choi Y, Kim J, Mooney DJ. The effect of surface their applications for targeted Cancer bioimaging. Integr Biol. 2010;2(2–
modification of Mesoporous silica micro-rod scaffold on immune cell 3):121–9.
activation and infiltration. Biomaterials. 2016;83:249–56. 96. Pons, T.; Pic, E.; Lequeux, N.; Cassette, E.; Bezdetnaya, L.; Dubertret, B.
72. Gallorini S, Berti F, Parente P, Baronio R, Aprea S, D’Oro U, Pizza M, Telford Cadmium-Free cuins2/ZnS Quantum Dots for Sentinel Lymph Node
JL, Wack A. Introduction of Zwitterionic motifs into bacterial polysaccharides Imaging with Reduced Toxicity. 2010, XXX (Xx), 4–11.
generates TLR2 agonists able to activate APCs. J Immunol. 2007;179(12): 97. Ayupova D, Dobhal G, Laufersky G, Nann T, Goreham R. An in vitro
8208–15. investigation of cytotoxic effects of InP/Zns quantum dots with different
73. Getts DR, Terry RL, Getts MT, Deffrasnes C, Müller M, Vreden C, Ashhurst TM, surface chemistries. Nanomaterials. 2019;9(2):135.
Chami B, McCarthy D, Wu H, et al. Therapeutic inflammatory monocyte 98. Zhang L, Yang XQ, An J, Zhao SD, Zhao TY, Tan F, Cao YC, Zhao YD. In vivo
modulation using immune-modifying microparticles. Sci Transl Med. 2014; tumor active Cancer targeting and CT-fluorescence dual-modal imaging
10:9274–86. with Nanoprobe based on gold Nanorods and InP/ZnS quantum dots. J
74. Butler KS, Peeler DJ, Casey BJ, Dair BJ, Elespuru RK. Silver nanoparticles: Mater Chem B. 2018;6(17):2574–83.
correlating nanoparticle size and cellular uptake with Genotoxicity. 99. Yaghini E, Turner H, Pilling A, Naasani I, MacRobert AJ. In vivo
Mutagenesis. 2015;30:577–91. biodistribution and toxicology studies of cadmium-free indium-based
75. Shang L, Nienhaus K, Nienhaus GU. Engineered Nanoparticles Interacting quantum dot nanoparticles in a rat model. Nanomedicine Nanotechnology,
with Cells: Size Matters. Journal of Nanobiotechnology. 2014:12:5. Biol Med. 2018;14(8):2644–55.
76. Goodman SM, Siu A, Singh V, Nagpal P. Long-range energy transfer in self- 100. Karlsson HL, Cronholm P, Gustafsson J, Mo L. Copper oxide nanoparticles
assembled quantum dot-DNA cascades. Nanoscale. 2015;7(44):18435–40. are highly toxic: a comparison between metal oxide nanoparticles and
77. Noh H, Goodman SM, Mohan P, Goodwin AP, Nagpal P, Cha JN. Direct carbon nanotubes - chemical research in toxicology (ACS publications).
conjugation of DNA to quantum dots for scalable assembly of photoactive Chem Res Toxicol. 2008:1726–32.
thin films. RSC Adv. 2014;4(16):8064–71. 101. Cho Y, Ivansevic A. In vitro assessment of the biocompatibility of chemically
78. Goodman SM, Singh V, Ribot JC, Chatterjee A, Nagpal P. Multiple energy modified GaAs surfaces. J Med Dent Sci. 2006;2:51–60.
Exciton shelves in quantum-dot − DNA Nanobioelectronics. J Phys Chem 102. Webb DR, Sipes IG, Carter DE. In vitro solubility and in vivo toxicity of
Lett. 2014;5:3909–13. gallium arsenide. Toxicol Appl Pharmacol. 1984;76(1):96–104.
79. Goodman SM, Noh H, Singh V, Cha JN, Nagpal P. Charge Transport through 103. Manzoor K, Johny S, Thomas D, Setua S, Menon D, Nair S. Bio-Conjugated
Exciton Shelves in Cadmium Chalcogenide Quantum Dot-DNA Nano- Luminescent Quantum Dots of Doped ZnS: A Cyto-Friendly System for
Bioelectronic Thin Films. Appl. Phys. Lett. 2015;106(8):083109. Targeted Cancer Imaging. Nanotechnology. 2009;20(6):065102.
80. Chan WCW, Nie S. Quantum Dot Bioconjugates for Ultrasensitive 104. Yang Y, Lan J, Xu Z, Chen T, Zhao T, Cheng T, Shen J, Lv S, Zhang H.
Nonisotopic Detection. Science (80-. ). 1998;281:2016–8. Toxicity and biodistribution of aqueous synthesized ZnS and ZnO quantum
81. Farokhzad OC, Karp JM, Langer R. Nanoparticle–aptamer bioconjugates for dots in mice. Nanotoxicology. 2014;8(1):107–16.
Cancer targeting. Expert Opin Drug Deliv. 2006;3(3):311–24. 105. Wang T, Hsieh H, Hsieh Y, Chiang C, Sun Y, Wang C. The in vivo
82. Gáspár S. Detection of superoxide and hydrogen peroxide from living cells biodistribution and fate of CdSe quantum dots in the murine model: a laser
using electrochemical sensors. Diagnostics, Prevention, and Therapy: ablation inductively coupled plasma mass spectrometry study. Anal Bioanal
Oxidative Stress; 2011. Chem. 2012;404(10):3025–36.
83. Lu, Q.; Yu, Y.; Ma, Q.; Chen, B.; Zhang, H. 2D Transition-Metal- 106. Robe A, Pic E, Lassalle HP, Bezdetnaya L, Guillemin F, Marchal F. Quantum
Dichalcogenide-Nanosheet-Based Composites for Photocatalytic and dots in axillary lymph node mapping: biodistribution study in healthy mice.
Electrocatalytic Hydrogen Evolution Reactions. 2015, No. March 2018. BMC Cancer. 2008;8:1–9.
84. Tamirat AG, Rick J, Dubale AA, Su W, Hwang B. Using Hematite for 107. Nagy A, Steinbrück A, Gao J, Doggett N, Hollingsworth JA, Iyer R.
Photocatalytic Water Splitting: A Review of Current Developments and Comprehensive analysis of the effects of CdSe quantum dot size, surface
Challenges. Nanoscale Horizons. 2016;1(4):243–67. charge, and functionalization on primary human lung cells. ACS Nano. 2012;
6(6):4748–62.
85. Derfus AM, Chan WCW, Bhatia SN. Probing the cytotoxicity of
108. Xiao K, Liu Q, Li H, Xia Q, Liu Y. Role of surface charge in determining
semiconductor quantum dots. Nano Lett. 2004;4(1):11–8.
the biological effects of CdSe/ZnS quantum dots. Int J Nanomedicine.
86. Liu W, Zhang S, Wang L, Qu C, Zhang C, Hong L, Yuan L, Huang Z, Wang Z,
2015;10:7073.
Liu S, et al. CdSe Quantum Dot (QD)-Induced Morphological and Functional
109. Hirn S, Semmler-behnke M, Schleh C, Wenk A, Schäffler M, Takenaka S,
Impairments to Liver in Mice. PLoS One. 2011;6(9):e24406.
Möller W, Schmid G, Kreyling WG. Particle size-dependent and surface
87. Chang E, Thekkek N, Yu WW, Colvin VL, Drezek R. Evaluation of quantum
charge-dependent biodistribution of gold nanoparticles after intravenous
dot cytotoxicity based on intracellular uptake. Small. 2006;2(12):1412–7.
administration. Eur Journal Biopharm. 2012;77(3):407–16.
88. Haque MM, Im HY, Seo JE, Hasan M, Woo K, Kwon OS. Acute
toxicity and tissue distribution of CdSe/CdS-MPA quantum dots after
repeated Intraperitoneal injection to mice. J Appl Toxicol. 2013;33(9):
940–50.
89. Male KB, Lachance B, Hrapovic S, Sunahara G, Luong JHT. Assessment of
cytotoxicity of quantum dots and gold nanoparticles using cell-based
impedance spectroscopy. Anal Chem. 2008;80(14):5487–93.
90. Law WC, Yong KT, Roy I, Ding H, Hu R, Zhao W, Prasad PN. Aqueous-phase
synthesis of highly luminescent CdTe/ZnTe Core/shell quantum dots
optimized for targeted bioimaging. Small. 2009;5(11):1302–10.
91. Cho SJ, Maysinger D, Jain M, Röder B, Hackbarth S, Winnik FM. Long-term
exposure to CdTe quantum dots causes functional impairments in live cells.
Langmuir. 2007;23(4):1974–80.
92. Zhang T, Hu Y, Tang M, Kong L, Ying J, Wu T, Xue Y, Pu Y. Liver toxicity of
cadmium telluride quantum dots (CdTe QDs) due to oxidative stress in vitro
and in vivo. Int J Mol Sci. 2015;16(10):23279–99.
93. Lovrić J, Bazzi HS, Cuie Y, Fortin GRA, Winnik FM, Maysinger D. Differences
in subcellular distribution and toxicity of Green and red emitting CdTe
quantum dots. J Mol Med. 2005;83(5):377–85.