You are on page 1of 17

This is an open access article published under an ACS AuthorChoice License, which permits

copying and redistribution of the article or any adaptations for non-commercial purposes.

Outlook

Cite This: ACS Cent. Sci. 2020, 6, 100−116

Size-Tunable Strategies for a Tumor Targeted Drug Delivery System


Wenqi Yu,# Rui Liu,# Yang Zhou, and Huile Gao*
Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering
Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of
Pharmacy, Sichuan University, Chengdu 610041, China

ABSTRACT: Nanoparticles have been widely used in tumor


targeted drug delivery, while the antitumor effects are not
always satisfactory due to the limited penetration and
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

retention. As we all know, there is a paradox that nanoparticles


with large sizes tend to distribute around tumor blood vessels
rather than penetrate into tumor parenchyma, while smaller
sizes can penetrate deeply but with poor tumor retention. In
Downloaded via 176.236.99.167 on October 4, 2022 at 09:52:15 (UTC).

recent days, an intelligent, size-tunable strategy provided a


solution to determine the size problem of nanoparticles and
exhibited good application prospects. In this review, we
summarize series of stimuli-induced aggregation and shrinkage
strategies for tumor targeted drug delivery, which can
significantly increase the retention and penetration of
nanodrugs in tumor sites at the same time, thus promoting treatment efficacy. Internal (enzymes, pH, and redox) and
external (light and temperature) stimuli are introduced to change the morphology of the original nanodrugs through
protonation, hydrophobization, hydrogen bond, π−π stacking and enzymolysis-resulted click reactions or dissociation, etc.
Apart from applications in oncotherapy, size-tunable strategies also have a great prospect in the diagnosis and real time
bioimaging fields, which are also introduced in this review. Finally, the potential challenges for application and future directions
are thoroughly discussed, providing guidance for further clinical transformation.

1. INTRODUCTION consideration, which is a very tricky problem to keep in


After years of research and development, nanoparticles have balance. Because of the special structure and environment of
tumor tissues, there is a contradictory effect of a nanoparticle’s
been widely used in antitumor research because of their high
size on drug delivery. That is, nanoparticles with large sizes
specific surface area, easy modification, and strong targeting
tend to be more capable of retention in tumor tissue than
properties.1,2 To passively deliver nanoparticles to tumor sites,
those with smaller sizes.11−13 As for the permeability, things
the enhanced permeability and penetration (EPR) effect is the
become reversed, smaller sizes have a better penetration ability
strategy that is mostly used, which is specific only in tumors in tumor tissues.14 To fully utilize the existing paradox,
due to the rapid proliferation of tumor cells and the abnormal researchers have designed a series of nanoparticles with
tumor vasculature system.3−5 However, more and more studies intelligent tunable sizes, including intelligent size aggregation,
found that only delivering nanodrugs to target sites is far from size shrinkage, and reversible size-changing strategies, which
enough, and accumulation and penetration problems still are systematically discussed in this review.
influence the intratumoral delivery efficacy to a great extent.6
Therefore, scientists have tried to design nanodrugs with both
good accumulation and penetration capacity in tumor tissues Among all the strategies, design-
to achieve in situ therapeutic concentrations and good ing nanoparticles with tunable
treatment efficacy. sizes is the most intuitive and
Among all the strategies, designing nanoparticles with controllable approach.
tunable sizes is the most intuitive and controllable approach.
Many studies have found that there is a close correlation
between the antitumor effect and the size of nanodrugs.7,8 In this review, we will summarize intelligent size-tunable
Usually, the diameter of nanodrugs is designed according to strategies including size aggregation, size shrinkage, as well as
the pore size of a leaky tumor vasculature.9 Though differences reversible size changes. Each section is divided through
may occur owing to the variety of tumor models, subcutaneous different stimuli such as enzyme, pH, redox, light, temperature,
tumors always exhibit a characteristic pore cutoff size ranging etc. In addition to the enhanced retention and penetration, we
from 200 nm to 1.2 μm, and the size is further reduced in
tumors that grow in the cranium such as glioma.10 Then, size- Received: November 6, 2019
related accumulation and penetration abilities are taken into Published: January 21, 2020

© 2020 American Chemical Society 100 DOI: 10.1021/acscentsci.9b01139


ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

Scheme 1. Brief Illustration of Stimuli-Induced Size-Tunable Strategies with Their Potential Applications

also focus on other potential applications in different ways. required for effective passive targeting. Generally, after entering
Aggregation strategies can be used in enhanced cellular uptake, the tumor region, nanoparticles with a large size are capable of
antimetastasis, and tumor diagnosis (photoacoustic imaging being well retained in the tumor surroundings but it is hard for
(PA), positron emission computed tomography (PET), them to penetrate deeply in the dense matrix,22,23 while the
surface-enhanced Raman scattering, and enhanced magnetic small-sized ones remain sufficiently penetrated in the tumor,
resonance imaging (MRI)), while shrinkage strategies exhibit whereas they can be easily pumped back into the bloodstream
advantages in nuclear delivery, drug release (Scheme 1), etc. In by the high interstitial fluid pressure of the tumor.24−26
the end, we conclude with the future application of size- Although there are investigations exploring strategies to
tunable nanoparticles and existing problems that need to be modulate the tumor microenvironment, consequently improv-
solved for better treatment. ing nanoparticles’ intratumoral distribution,27,28 optimizing the
nanoparticles’ size is deemed as a critical solution. Also,
2. SIZE IMPACT ON DELIVERY EFFICACY regarding the principle of the size-dependent tumor distribu-
As one of the most important characteristics of nanoparticles, tion, it is defined that the unique and ideal size remains exactly
size greatly influences the efficiency of tumor targeted drug the equilibrium point between penetration and retention. To
delivery in many ways, including circulation, biodistribution, optimize the situation, size-tunable nanoparticles show their
tumor accumulation and penetration, as well as cellular uptake extraordinary talent. Compared with nanoparticles that are
and subcellular distribution. A thorough understanding of size unique in size, size-tunable nanoparticles can reach both better
will be introduced first to help better elucidate the importance penetration and retention, so long as they change their sizes at
of size-tunable strategies. the proper time.
After entering into the body, the circulation time of
nanodrugs basically determines the efficacy of tumor targeting Nanoparticles with large sizes
as the clearance by mononuclear phagocytic system (MPS) or tend to be more capable of
filtration by the liver and spleen happens very quickly and
sequesters the majority of nanodrugs. There is a correlation retention in tumor tissue than
between the circulation and particle size. The MPS clearance those with smaller sizes. As for
exhibits a size-dependent behavior such that nanoparticles with the permeability, things become
small sizes are less likely to be taken up by macrophages than
large ones.15,16 The biodistribution is also greatly influenced by
reversed, and smaller sizes have
the size of nanodrugs because of the different cutoff size of better penetration ability in
organs. The renal filtration cutoff size is 5.5 nm,17 and the tumor tissues.
vascular fenestrations in liver are 50−100 nm. Particles lower
than 5.5 nm are much more easily excreted through urine, and
sizes smaller than 50 nm could easily penetrate the endothelial After nanoparticles enter into tumor sites, their size also
and get trapped in the liver.18,19 influences the following cellular uptake process in which
To effectively accumulate in tumor sites, there is one more nanoparticles with varied sizes exhibit different internalization
barrier to cross: the leaky tumor vasculature. As for the rapid rates.29−31 Sizes were found to be the determining factor for
growth of tumor cells, the pore cutoff size of the tumor vessel the pathway of cellular internalization. Most of the nano-
ranges from 200 nm to 1.2 μm, depending on the type of particles were internalized into cells through two pathways:
tumor.20,21 Diameters of nanoparticles below the cutoff size are phagocytosis and pinocytosis. The phagocytosis pathway was
101 DOI: 10.1021/acscentsci.9b01139
ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

only in some special cells such as macrophages, neutrophils, or Table 1. Mainly Used Aggregation Strategies
dendritic cells, or particles larger than 500 nm in diameter.
stimuli mechanism ref
Also, the pinocytosis pathway can be further divided into four
different types based on the mediated proteins: caveolae- enzyme click reaction (cyano and 1,2-thiolamino, azide 51−56
and alkyne, amine and acyl)
mediated endocytosis, clathrin-mediated endocytosis, non-
self-assembly (amphiphilic block copolymers) 60−63
caveolin nonclathrin mediated endocytosis, and macropinocy-
pH electrostatic interactions (zwitterionic 65−71
tosis.32 Nanoparticles between 20 and 100 nm are internalized compounds, hemoglobin)
through caveolae-mediated endocytosis,33 120 to 150 nm are light C−C, C−H, O−H, X−H insertions (azobenzene, 73−75
mediated by clathrin,34 and macropinocytosis occurs in the diazirine, triphenylmethane, cinnamenyl)
internalization process of nanoparticles larger than 1 μm.35 As temperature phase transition (PPCs, PNIPAm, PDEAm, PEO, 81−92
a reverse mechanism of endocytosis, exocytosis also plays an PPO, polyphosphoesters)
important role in enhanced cellular accumulation of nano- redox click reaction and host−guest interaction 99, 101
(disulfide bond, Fc+)
particles. Decreased exocytosis avoids nanoparticles secretion
to the extracellular space and therefore improves the
therapeutic efficacy.36 Quantities of research studies found in tumor tissues such as matrix metalloproteinase (MMP),
that there was a negative correlation between the size of legumain, hyaluronidase (HAase), gelatinase,48 furin,49 caspase
nanoparticles and the exocytosis process, and nanoparticles 3/7,50 etc. Click reactions are commonly used in this strategy
with relatively large sizes exhibit decreased exocytosis and because of their easy and quick procedure, which is perfectly
enhanced intracellular retention.37,38 suitable for in vivo synthesis. Many functional groups can be
As with a brain for a human, the nucleus in the cell plays the utilized such as azide and terminal alkynes,51 sulfhydryl and
most important role among all the organelles and is responsible maleimide,52 1,2-thiolamino and cyano groups,53 etc. By
for controlling genetics and metabolism.39−41 Various therapies decorating the above corresponding chemical groups on the
aim at the nucleus to achieve an anticancer effect, such as surface of nanoparticles, aggregation will happen very quickly
anthraquinone derivatives and short hairpin RNA, which once the chemical groups are exposed and interact with each
restrictively exhibit activities inside the nucleus. However, the other. Rao et al. first used the click reaction between the cyano
major obstacle to nuclear delivery is the nuclear membrane, in group of 2-cyano-benzothiazole (CBT) and the 1,2-aminothiol
which nuclear pores are generally ∼9 nm in size.42,43 Even group of cysteine (Cys), which was the last step of the
though there are other strategies to promote the nuclear synthesis of D-luciferin and displayed biological friendliness.54
delivery of nanoparticles with relatively larger sizes, such as Our group utilized the substrate of legumain (Ala-Ala-Asn↓
shape modulation,44 ligand modification for active targeting,45 Cys-Lys) and codelivered 2-cyano-6-amino-benzothiazole
or artificially opening the nuclear envelope by singlet (CABT) to tumor sites where legumain was found overex-
oxygen,46,47 the simplest and most effective way yet is to pressed,55 which generated aggregations of GNPs from 35.6 to
control the nanoparticles’ size below 9 nm. However, the 309.6 nm in 12 h (Figure 1A) and endowed GNPs not only
ultrasmall size ensuring the passage across nuclear pores is the prolonged tumor accumulation but also bioimaging of
comparable with the renal filtration cutoff, risking nano- multispectral optoacoustic tomography (MSOT). In addition
particles’ circulation and distribution at the organ level. To to enzymes that are upregulated in tumor sites, there are
address the contradiction, size-tunable nanoparticles arise at circumstances when the triggered enzymes of click reactions
the appropriate time, gathering both good blood circulation are not or are lowly expressed in tumors. In that case, enzymes
and nuclear distribution. can be delivered together with nanocarriers. Gu et al.
codelivered transglutaminase (TG) in a hyaluronic acid
3. AGGREGATION STRATEGIES (HA)-made nanogel shell on the surface of a carrier (Figure
As a contradictory behavior of varied sizes described above, 1B).56 The protected nanogel shell collapsed after entering
small-sized nanoparticles tend to have good penetration but into the tumor and released sufficient TG to catalyze the
are cleared away rapidly, while nanoparticles with large sizes formation of an isopeptide bond between free amine group
have enhanced retention time but cannot penetrate deeply into from lysine and the acyl group from glutamine, which led to
tumors. Researchers proposed an aggregation strategy to aggregation from 10 to 120 nm and prolonged tumor retention
enhance the accumulation and penetration of nanoparticles in time to more than 72 h.
the meantime. The initial small-sized nanoparticles were first Unlike click reactions which need to modify other functional
used for deep penetration, and form large agglomerates for chemical groups to make aggregation happen, the enzyme-
enhanced retention once deep into the tumor after specific induced self-assembly makes full use of the character of the
stimulations. The stimulations can be divided into several types nanocarrier itself. Such nanocarriers are usually composed of
due to the tumor heterogeneity, such as hypoxia, slightly acidic amphiphilic block copolymers, which can lead to formation,
microenvironment, and specifically upregulated enzymes. In destruction, or morphological transformation after changes in
addition, some external stimuli such as light and temperature the chemical of physical nature.57−59 Gianneschi et al. designed
are also utilized to design the intelligent, size-aggregatable a polymer−peptide block copolymer amphiphiles system
nanoparticles (Table 1). Under external or internal stimuli, containing substrates for four different cancer-associated
initial nanoparticles with relatively small sizes interact with enzymes: protein kinase A (PKA), protein phosphatases-1
each other through click reaction, self-assembly, electrostatic (PP1), MMP-2, and MMP-9 (Figure 1C).60 Phosphorylation
interaction, or phase transition to form aggregations. by PKA caused a 50-fold increase in hydrodynamic diameter
3.1. Enzyme-Induced Aggregations. The enzyme- together with the appearance change of amorphous structure,
induced strategy has great potential because of its highly and the aggregation could be reversed after being successively
specific substrate selectivity and varied expression of enzyme in treated with PP1 for dephosphorylation, providing feasibility
different organs. There are many specific upregulated enzymes for enzymatically size-switchable strategy. Alkaline phospha-
102 DOI: 10.1021/acscentsci.9b01139
ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

Figure 1. (A) Diagram depicting the legumain-triggered aggregation and composition of GNPs-DOX-A&C. Copyright 2016, American Chemical
Society. (B) Schematic of tumor microenvironment-mediated construction of combination drug-delivery depots for sustained drug release using
CS-NG. Copyright 2016 American Chemical Society. (C) A variety of morphologies of polymeric amphiphile aggregates depending on the design
of the peptide substrate and enzymes added. Copyright 2011, American Chemical Society.

Figure 2. (A) Schematic illustration of pH-responsive “smart” gold nanoparticles (SANs). Copyright 2013, Royal Society of Chemistry. (B) TEM
images of the Hb-IR780 complex at pH 7.4 (top, the red arrows highlight the Hb-IR780 nanoparticles) and at pH 6.5 (bottom). (C) Increase in
hydrodynamic size of the Hb-IR780 complex from normal tissue pH 7.4 to tumor acidic pH 6.5. Copyright 2018 American Chemical Society.

tases (ALPs) is another enzyme widely used to instruct the around pH 5.0−5.5, while blood and normal tissues are
self-assembly of nanofibers in vivo,61 which was found to be maintained at 7.4.64 Different from enzyme-induced aggrega-
highly expressed on the cell membrane or secreted out of some tions, pH-triggered reactions possess the advantages of quick
cancer cells with the ability to responsively cleave the response and ultrasensitivity. Upon the electrostatic inter-
phosphate groups. Xue et al. and Liang et al. both utilized actions of decorated pH-sensitive surface molecules65,66 or
ALPs to induce nanofibers through π−π stacking.62,63 natural pH-sensitive proteins,67 the nanocarriers will aggregate
3.2. pH-Triggered Aggregations. Different parts of the and prolong retention in tumors due to the increased
human body exhibit different pH levels as we all know. Because sedimentation-driven uptake and decreased extracellular efflux.
of the rapid proliferation of tumor cells, the microenvironment pH-responsive molecules usually are zwitterionic com-
of tumors is slightly acidic at around pH 6.5, and after being pounds, such as hydrolysis-susceptible citraconic amide,68,69
internalized into cells, some cytoplasmic organoids such as 11-mercaptoundecanoic acid, and (10-mercaptodecyl)-
endosomes and lysosomes exhibit an even lower pH degree at trimethylammonium bromide).14 Citraconic amides are rapidly
103 DOI: 10.1021/acscentsci.9b01139
ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

Figure 3. (A) Schematic illustration of light-triggered assembles of dGNPs. (B) TEM images of dGNPs before and after illuminated with 405 nm
laser in different periods of time. Copyright 2016, John Wiley and Sons.

Figure 4. (A) LCST profiles of F1−F3 before and after treatment with GSH, caspase-3, and Atg4B. (B) Specific responsive nanoaggregation in
cells. Confocal images and bio-TEM images of MCF7 cells treated with PPCs and modulated agents. Copyright 2017, American Chemical Society.

converted to positively charged primary amines by hydrolysis responsive polymers are supplied with photoactive groups
at a mildly acidic pH, which would in turn react with the such as azobenzene, spirobenzopyran,73 triphenylmethane,74 or
negatively charged carboxyl groups through electrostatic cinnamenyl that can undergo reversible structural changes
attraction and lead to aggregation. However, it should be under UV−Vis light (Figure 3A,B).75 However, the application
noted that mixed charged ligands with the same strong of this strategy is only limited in experiments on the cellular
electrolytes would not aggregate and remain stable over the level due to the weak penetration ability of UV−vis and blue
entire pH range.70 Therefore, disrupting the balanced electro- light. Upconversion nanoparticles (UCNPs) are novel
lytes was a feasible way to form aggregation (Figure 2A),71 and materials consisting of rare-earth elements, which can convert
the commonly used combination is a weak acid and strong a near-infrared (NIR) light to UV−vis radiation via two
base such as a weak electrolytic 11-mercaptoundecanoic acid photons or a multiphoton mechanism.76 The application of
and strong electrolytic (10-mercaptodecyl)trimethyl- UCNP has been well developed so far and has aroused great
ammonium bromide. interest of researchers as the extraordinary tissue penetration of
Though the commonly used pH-sensitive ligands are long-wavelength NIR light exactly provides a solution to solve
synthesized artificially, natural proteins with acidic or basic the long-standing problem of limited tissue penetration of
amino acid residues are deemed to be pH-sensitive nanoplat- visible light.77 Liu et al. observed an enhanced photodynamic
forms. It is generally known that proteins remain stable at treatment efficacy when loaded with a photosensitizer in
different pH values, while aggregating around the isoelectric UCNP.78 Zhao et al. reported that UCNP can trigger the
point (pI).72 If the protein is properly designed with pI around photoisomerization of azobenzene,79 which is a commonly
the physiological pH of tumor, the natural protein could be used chemical group for light-induced aggregation as
chosen as the carrier for enhanced tumor accumulation. Li et mentioned above. Therefore, the further application of light-
al. employed hemoglobin as a smart pH-sensitive nanocarrier induced aggregation in vivo can be achieved with great
for near-infrared dye IR780 (Hb-IR780).67 Dynamic light prospects by utilizing UCNP as the carrier.
scattering (DLS) results showed that the Hb-IR780 was well- 3.4. Temperature-Triggered Aggregations. To con-
dispersed as a singular protein at pH 7.4 while aggregated struct temperature-responsive nanoparticles, the balance
severely after incubation at pH 6.5 (Figure 2B,C). between segment−segment interactions and segment-solvent
3.3. Light-Induced Aggregations. The light-induced intermolecular interactions can be reversed by temperature
strategy has shown great advantages in noninvasiveness, changes. Some thermosensitive polymers can be designed to
remote manipulation, and easy operation. The photothermal typically undergo the coil−helix transition upon decreasing
therapy even shows an extraordinary antitumor effect and is temperature below the upper critical solution temperature
being used widely. In the aggregation strategies, light- (UCST), while others specifically respond to the increasing
104 DOI: 10.1021/acscentsci.9b01139
ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

Figure 5. (A) Schematic drawing of the GSH-responsive antiphagocytosis 99mTc-labeled Fe3O4 nanoparticles for forming particle aggregates
through the interparticle cross-linking reaction. (B) Temporal hydrodynamic size profiles of nonresponsive probe and (C) responsive probe in
reaction with the GSH treatment. Copyright 2017, John Wiley and Sons. (D) Illustration of intracellular host−guest assembly of gold nanoparticles
triggered by GSH. Copyright The Royal Society of Chemistry 2016.

temperature above the lower critical solution temperature As an important antioxidant and free radical scavenger in the
(LCST). Mostly used functional pairs are polymer−peptide body,100 GSH could not only reduce disulfide but also other
conjugates (PPCs) (Figure 4A,B),80 proteins, polylactic acid species at an oxidative state. Therefore, Wang et al. used GSH
(PLA), and polysaccharides.81−83 Polymers with the merit of as a reducing agent to specifically respond to reductive
thermoresponsive behaviors such as poly(N-isopropylacryla- aggregation.101 They modified GNPs with β-CD and incubated
mide) (PNIPAm),84 polyphosphoesters,85 poly(N-diethylacry- them with Fc+-PEG-Fc+, and aggregation was seen in HepG2
lamide) (PDEAm),86 copolymers of poly(ethylene oxide) cells after 12 h (Figure 5D). The GSH triggered aggregation
(PEO) and poly(propyleneoxide) (PPO),87 and poly(N- caused significant apoptosis of cancer cells, and because of the
vinylcaprolactam)88 have been widely used. However, this specific express site of GSH, this strategy was also promising in
strategy always needs the combination with other stimuli such reducing unexpected side effects induced by traditional
as enzymes or pH to destroy the inherent interactions and chemotherapy. However, the aggregation efficacy was detected
expose the thermosensitive parts so as to respond to different only at the cellular level as it remained difficult to codeliver β-
temperatures.89−91 In other cases, an additional local temper- CD-GNPs and Fc+-PEG-Fc+ to tumor sites.
ature increase was given through introducing photothermal 3.6. Salt-Induced Aggregation. As is known to all, the
molecules for specific responsiveness.92 fluid in vivo can be seen as a buffer salt system, and a great
3.5. Redox-Induced Aggregation. Attributed to the high majority of nanoparticles are unstable and easy to aggregate
proliferation nature of invasive tumors, tumor tissues and cells irreversibly in a high concentration of aqueous salts due to the
show elevated oxidative stress, resulting in a defined high level disruption of the shielding layer on the surface. Researchers
of reactive oxygen species (ROS).93 As a consequence of tried every possible way to prolong the circulation time of
handling the oxidative stress, the redox environment of tumor nanocarriers in vivo such as PEGylation and biomimetic cell
tissues and cells increases in a way. For example, the membrane coating,102,103 etc. However, Sun et al. first took this
intracellular GSH level is elevated in tumors for increased defect as a strategy to design salt-induced GNPs aggrega-
antioxidant capacity and resistance against oxidative stress, as tion.104 After intratumoral injection, GNPs formed irregular
well as regulating cell differentiation, proliferation, and aggregation, while PEG-GNPs did not aggregate at all. The
apoptosis.94 According to research data, the concentration of salt-induced aggregation got rid of complicated surface
GSH in tumor regions, especially the intracellular part, reaches modifications and was completed instantaneously. Yet this
2−10 mM, whereas that in normal tissue remains 2−20 strategy was only applied in superficial solid tumors treatment
μM.95−97 The disulfide bond is mostly used GSH-specific and needed precise operation because of the specific
chemical groups that can be reduced to sulfhydryl, and requirement of intratumoral injection.
therefore is widely used in redox environment responsive drug
delivery.98 Gao et al. utilized the dysregulation of GSH for
aggregation strategies and tumor imaging.99 After entering into
4. SIZE-SHRINKAGE STRATEGIES
tumor sites, the self-peptide was cleaved by GSH, and the With regard to the relationship between nanoparticles’ size and
exposed sulfhydryl interacted with maleimide on the end of intratumoral behaviors mentioned before, the high fluid
PEG of the adjacent nanoparticles according to the click pressure and dense matrix inside solid tumors always impede
reaction (Figure 5A). The reaction rate between sulfhydryl and the deep penetration and consequently homogeneous dis-
maleimide happened extremely rapidly so that the nano- tribution of nanoparticles inside the tumor;105 therefore, it is
particles aggregated from 7.5 to 295 nm in 5 h (Figure 5B,C). essential for nanoparticles to shrink their size and enhance
105 DOI: 10.1021/acscentsci.9b01139
ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

penetration for homogeneous delivery.106 Besides, the size 2,3-Dimethylmaleicanhydride (DMA) reacts with many
(reported to be ∼10 nm and up to 39 nm when amplified)107 amines to form acid amine, which can further respond to
of nuclear pores limits the nuclear-targeting nanoparticles to be slightly acidic condition and break into amine and 2,3-
in an engaged size. The postshrinkage nanoparticles not only dimethylmaleic acid.116−118 Wang’s group further constructed
retain a small size for enhanced penetration, but also a PCL-CDM-PAMAM/Pt system with a size shrinking from
contribute to many other properties, such as drug release,108 100 to 5 nm under intratumoral acidic pH (Figure 7A).119 In
rapid renal clearance,109 secondary distribution,110 etc. By now, addition to directly decomposing contents to reduce size,
plenty of nanoparticles have been investigated based on the DMA also showed another property, charge reversal, for the
endogenous acidic pH, overexpressed enzymes, redox con- design of size-shrinkable nanoparticles (Figure 7B,C).120 The
ditions, and exogenous physicochemical stimuli, which are dramatic size shrinkage and charge reversal were essential for
summarized in Table 2 and discussed in the following. the penetration and cellular internalization inside the tumor.
Schiff base is another widely used pH-sensitive chemical group,
Table 2. Mainly Used Shrinkage Strategies characterized by a double bond formed between carbon and
nitrogen atoms (-CN-),which is unstable under acidic pH
stimuli responsive chemical groups refs and easily broken by hydrolysis.121 Although the critical acidic
pH amino polymers, DMA, Schiff base 114, 115, condition and pervasive proton make a pH-triggered strategy
119−121 sensitive and specific, the acidic and hypoxic regions are
enzyme MMPs, HAase, amylase, thrombin 126−130, commonly far from blood vessels,122 which becomes a major
136−141
redox disulfide bond 142, 143
concern for pH-triggered shrinkage.
ROS thioketal, thioester, polypropylene sulfide, 148−151
4.2. Size Shrinkage Triggered by Overexpressed
phenylboronic ester Enzyme. MMPs and HAase are the most common tumor-
specific enzymes used as recognizing markers in responsive
drug delivery. MMPs include a large family of proteinases, in
4.1. Size Shrinkage Triggered by Acidic pH. Amino which the MMP-2 and MMP-9 are the most known, playing an
polymers are a kind of polymer functionalized by amino important role in tissue remodeling associated with various
groups, which are generally nonprotonated and exhibit physiological or pathological processes.123−125 The MMP-2
hydrophobicity.111−113 However, the amino groups in amino and MMP-9 are generally overexpressed and secreted by
polymers change into hydrophilic when protonated at acidic tumors for matrix digestion, recognizing plenty of substrates
pH, resulting in the disassembly of the hydrophobic core. The involving gelatin and collagens, which retain MMPs’
dramatic protonation of histamine during pH decreasing responsiveness when designed as a component of a drug
provides poly(histamine) with the same property.66 Yuan et al. carrier. The very first size-shrinkable nanovehicle based on
reported a kind of nanoassembly composed of nanomicelles MMPs was designed by Wong and co-workers.126 They
and nanogel,114 which were both self-assembled by amphi- covered the large gelatin nanoparticles (GelNPs) with small
philic copolymers (PDPA30-b-PAMA15 and P(EGMA-GMA- quantum dots (QDs) to form QDGelNPs, which rapidly
PDSEMA), respectively, in aqueous solution (Figure 6A). The shrank from 100 nm to 9.7 nm when responding to
micelle retained a hydrophobic core made up of PDPA, which intratumoral MMP-2 and MMP-9, as a consequence of
was able to respond to intratumoral acidic pH and GelNPs dissociation (Figure 8A). And taking advantage of
consequently transferred to a hydrophilic moiety (Figure 6B) the gelatin nanoparticles, our group functionalized small-sized
with the size apparently shrinking from 35 nm to about 10 nm dendrigraft polylysine (DGL) on the surface of gelatin
(Figure 6C). Another similar work recently reported by Ray et nanoparticles (GelNP), 127−129 with DOX loaded and
al. is much more suitable to address the penetration problem, angiopep-2 decorated on DGL, 130 resulting in deep
since the size shrinkage is from 100 to 150 nm to 2−5 nm.115 penetration and homogeneous tumor therapy (74.1% growth

Figure 6. (A) The polymerization of PDPA30-b-PAMA15 and P(EGMA-GMA-PDSEMA), and consequent construction of micelle and nanogel. (B)
Illustration of nanosystem fabrication and responsive shrinkage. (C) TEM images of nanosystem before and after shrinkage, scale bars: 100 nm.
Copyright The Royal Society of Chemistry 2014.

106 DOI: 10.1021/acscentsci.9b01139


ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

Figure 7. (A) Illustration of construction and pH-responsive size shrinkage of DMA-based nanomicelle. Copyright 2016 National Academy of
Sciences. (B) Schematic illustration of DMA-based PNV, with charge reversal for dissociation. (C) The dissociation of PNV and release of
polymer-Dox inside the tumor. Copyright 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.

Figure 8. (A) Schematic of 100 nm QDGelNPs shrinking size to 10 nm QDs by cleaving away the gelatin scaffold with MMP-2. Copyright 2011
National Academy of Sciences. (B) Schematic illustration of the design and synergistic effects for deep tumor penetration and therapy effects of
IDDHN. Copyright 2018 Elsevier Ltd.

inhibition). Unlike MMPs with a range of substrates, HAase amylase-digested hydroxyethyl starch140 and thrombin-induced
exclusively hydrolyzes HA to short chains or modified glucose depegylation.141 The enzyme responsiveness is of super-
units.131 The natural negative charge on HA makes itself easy selectivity owing to exclusive enzyme−substrate recognition,
to conjugate positively charged materials, and the specific which is essential for improving the therapeutic efficacy and
combination between HA and CD44 also provides HA with reducing the adverse effects of cancer therapy. However,
active-targeting ability.132−135 For application of superior HA, uneven levels of enzyme expression in different tumors restrict
our group cross-linked HA with positively charged DGL to the application scope of certain responsive nanoparticles.
form size-shrinkable HAase-responsive nanoparticles (Figure 4.3. Size Shrinkage Triggered by Redox Condition.
8B) that exhibited extraordinary penetration ability with the Since the GSH level is critical in tumor tissue and cytoplasm,
further assistance of an NO donor on an HA shell.136,137 its responsiveness is also evident in size shrinkage strategies.
As instanced, the enzyme-triggered size shrinkage is Guo et al. constructed a nanomicelle composed of pegylated
commonly achieved by shell dissociation of core−shell polylactide (PEG−PLA) and DMA-modified polythylenimine
nanoparticles and detachment of small-sized decorations (PEI−DMA),142 which was linked by a disulfide bond, forming
from large nanoparticles.138 The rule is always applicable, PEG−PLA−S−S-PEI−DMA (PELEss-DA, see structures in
and a recent-report nanosystem is also representative, which Figure 9A). The high level of intracellular GSH reacted with
can release small-sized cargo nanoparticles when lactate the disulfide bond and deshielded PEI shell. Also, the dramatic
oxidase digests its shell.139 Other enzymes-dependent nano- size shrinkage allowed the remaining nanoparticles’ entry into
particles also obeyed this designing principle, such as α- nucleus (Figure 9B) and consequent DOX release for DNA
107 DOI: 10.1021/acscentsci.9b01139
ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

Figure 9. (A) Schematic design of the nucleus entry of size-shrinkable polymer micelles (PELEss-DA) to overcome MDR. (B) Cellular uptake and
intracellular distribution analysis. Copyright 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim. (C) Schematic illustration of the
cooperative dimensional strategy for anticancer drug delivery mediated by hybrid micelle PSPD/P123-Dex. (D) Evaluation of HeLa cells
internalizing different micelles, the number represents: 1. Free DOX; 2. PPD/DOX; 3. PSPD/DOX; 4. PPD/P123-Dex/DOX; 5. PSPD/P123-Dex/
DOX. Copyright 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.

interruption. The nucleus delivery ensured the drug’s activity cancer cells (∼10 μM) compared with normal tissues (∼10
toward the target, avoiding drug resistant transporters’ nM),144 it is still not sufficient enough to trigger hydrolysis of
function, which is essential for nucleus-targeting chemo- ROS-responsive chemical groups instantly. NIR light retains
therapeutic agents. A similar investigation was found in better tissue penetration compared with ultraviolet and visible
Wang’s work, in which a PSPD/P123-Dex nanomicelle was light.145 Importantly, plenty of photosensitizers are able to
designed (see structures in Figure 9C).143 The two materials convert NIR light into ROS, which has been widely explored as
formed 120 nm PSPD/P123-Dex and dramatically shrank into photodynamic therapy for surficial cancer treatment.146,147 A
approximately 30 nm P123-Dex under an intracellular GSH- thioketal linker is easy to synthesize and respond to ROS,
abundant condition. Regarding the suitable small size and resulting in two broken parts. Cao et al. designed a PCL−PEG
assistance of dexamethasone, P123-Dex pithily delivered nanomicelle with poly(thioketal phosphoester) (TK-PPE)
encapsulated DOX into the nucleus for pronounced cytotox- mixed core.148 The chlorin e6 (Ce6) loaded in the micelle
icity (Figure 9D). could transfer NIR irradiation into ROS, further cleaving the
4.4. Size Shrinkage Triggered by ROS. Although it is TK-PPE and dissociating the core of micelle (Figure 10A). As
widely known that there is a relatively high level of ROS in a consequence, the nanomicelle shrank its size from 154 to 72
108 DOI: 10.1021/acscentsci.9b01139
ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

Figure 10. (A) Preparation and function of the light-activated shrinkable nanoparticle TK-PPE@NPCe6/DOX. (B) TEM images of TK-PPE@
NPCe6 after 660 nm laser irradiation at different times. (C) Plasma DOX concentration versus time after intravenous injection of free DOX,
TKPPE@NPCe6/DOX, and TK-PPE@NPCe6/TK-PPE@NPDOX. (D) Quantification of DOX content in tumors by HPLC. Tumors were
excised 12 and 24 h after intravenous injection. (E) MDA-MB-231 tumor growth curves of various groups after intravenous administration, *p <
0.05; **p < 0.01. Copyright 2017 American Chemical Society.

nm (Figure 10B), with DOX released. By comparison to free reaction, pH-induced protonation and GSH-dependent redox
drug, the preshrinkage large micelle ensured its prolonged may suffer from the protein corona a lot, while the external
circulation (Figure 10C), while the postshrinkage small one stimuli such as light and temperature would not be bothered.
promoted its distribution and deep penetration inside the Though PEGylation has been confirmed to facilitate nano-
tumor (Figure 10D). Together with the transferred ROS and particles with the “stealth” property, which can reduce the
released DOX facilitated by NIR irradiation, the shrinkable protein adsorption and decrease the effect of corona, there
nanomicelle performed chemo- photodynamic therapy in deep comes a new question whether the enzymatic reaction would
tumor region, resulting in remarkable antitumor effect (Figure also be affected by PEGylation, as the nature of enzyme is
10E). Other ROS-responsive structure or materials, such as protein as well. The response sensitivity may be affected in the
polypropylene sulfide,149 phenylboronic ester150 and thioest- enzyme-induced size change after PEGylation.
er151 are also of talent in designing NIR/ROS-triggered 5.2. Accuracy of Measurement. As we are doing research
shrinkage. Unlike other endogenous stimuli, ROS triggered by studies about size changing, the detection of nanoparticles’ size
external NIR irradiation is desirable and controllable, as well as undoubtably requires very high accuracy. However, the size
tumor-specificity. Meanwhile, no activity remains in the measured by DLS always displays a relatively wide peak.153
nonirradiation or nondrug region, ensuring the safety and Nanoparticles with an average DLS size of 100 nm probably
avoiding adverse-effect aroused by cancer therapy. Besides, have a peak ranging from 50 to 400 nm. It is not that
dual stimuli seem like one case with two locks, ensuring the convincing to confirm the success of size shrinkage (from 100
specificity of unfolding. to 50 nm) or size aggregation (from 100 to 400 nm) as there is
an overlap of size during the size change process. The
5. MAJOR CHALLENGES OF SIZE-TUNABLE corresponding distribution intervals can be compared sepa-
STRATEGIES FOR APPLICATION rately instead of just comparing the average number, which is
Though great progress has been made in size-tunable meaningless. On the other hand, nanoparticles with a good
nanoparticles, there are still many challenges that hinder the polydispersity index are highly recommended in the study of
efficacy, which need to be further optimized and solved. size change, such as silica nanoparticles, gold nanoparticles,
5.1. Protein Corona May Shield the Responsive and polystyrene nanoparticles, as these nanoparticles have
Ligand. Protein corona acts as the main issue to affect the already been well industrialized and have quite uniform size,
targeting efficiency. The corona is quickly formed once and the narrow size range would partly solve the problem in a
nanoparticles are introduced into biological fluid. As we have way.
discussed previously, the composition, size, shape, and surface 5.3. Off-Target Effect. The location accuracy of the
chemistry of nanoparticles, and even the different fluid responsive reaction needs to be precisely designed. Nano-
environment in vivo, influence the formation of protein particles are designed to specifically target tumor sites and
corona.152 In addition to acting as opsonin to mark a nanodrug enhance the accumulation. However, the off-target effects
and alter its biodistribution, the adsorbed protein corona on always attenuate the antitumor efficacy and cause damage to
the surface may also shield the responsive ligand from stimuli normal organs. For example, P-aminophenyl-α-D-mannopyr-
and affect the sensitivity of size changing. Among which, the anoside (MAN) is a widely used glucose analogue modified on
mechanism that need direct contact such as enzymatic nanoparticles to help cross the blood−brain barrier and target
109 DOI: 10.1021/acscentsci.9b01139
ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

brain tumor cells,154 while its receptor, the mannose receptor, requirement of tumor volume. In tumor-bearing mice, it is
is also expressed on Kupffer cells of liver.155 The majority of until the tumor grows to 200 mm3 that it exhibit good EPR
nanoparticles are trapped in the liver, limiting the effective effect, which is nearly impossible for a human to have a such
delivery to tumor sites. On the other hand, since the pH and big tumor. Recently, scientists are more prone to intervene in
enzyme environments are somehow different on both sides of tumors at an early stage after inoculation in mice or choose
the biological barrier, it remains to be explored whether the active-targeted strategies to treat cancers. Moreover, there are
responsiveness still exists after crossing barriers, or the still other problems that limit the clinical translation such as
responsiveness is crippled by prereacting with enzyme on the the biocompatibility of nanodrugs and scalable manufacturing
barriers. Furthermore, the most commonly used targeting for industry, which is a long way to go.
strategy is ligand−receptor binding; ligands are modified on
nanoparticles to help actively target the tumor sites. However, 6. OUTLOOK AND FUTURE DIRECTIONS
the chemical modification would destroy the innate structure In addition to the above strategies, there are many other novel
of ligands and attenuate the binding efficiency to some extent. approaches to make nanocarriers aggregate or shrink in tumor
The recently rising bio-orthogonal reactions improve the tissues, such as using enzyme-instructed self-assembly to
targeting specificity very well.156,157 By first giving a primary generate intracellular supramolecular nanofibers to achieve
exogenous tag to label target cells, the secondary tag could the aggregation goal,164−167 or using the change of shape to
specifically recognize the labeled cells and avoid the off-target achieve the purpose of aggregation in a round-about way.168,169
effect.158−160 However, the existing primary tag delivery still Intracellular aggregation will decrease the efflux of nanodrugs
depends on the interaction of ligand and receptor, and relevant in a way, which has potential in overcoming multidrug
studies are only at the cellular level; further work needs to be resistance.49 Except prolonging the retention of nanocarriers
done to make it better applied in vivo. after aggregation, properties of aggregated nanodrugs tend to
5.4. Complicated Tumor Microenvironment. Because be changed, and most of them exhibit a good photothermal,
of the heterogeneous nature of tumor sites, the tumor photoacoustic effect and enhanced contract of MRI, which
microenvironment should be taken into consideration. The provide a possibility for real time imaging and photothermal
dense ECM and high interstitial pressure of the tumor tissue therapy leading to better antitumor treatment prospects.170
attenuate nanoparticle delivery efficiency. However, the Aggregation-induced emission (AIE) fluorophores are the
intelligent size change of nanoparticles together with application of aggregation strategies to solve the aggregation-
remodeling of the tumor microenvironment could synergisti- caused quenching (ACQ) problems and give a new long-term
cally improve the accumulation and penetration of nano- tracer for in vitro or in vivo imaging.171,172 Also, because of the
particles at the tumor sites, such as degradation of ECM and good results achieved in antitumor therapy, aggregation
normalization of tumor vasculature system, which could strategies are increasingly being used in other areas, such as
enhance the penetration ability of nanoparticles and signifi- brain tumor surgery guidance173 and myocardial infarction.174
cantly reduce the reflux of nanoparticles leading to promoted
accumulation at tumor sites. However, there also exists With more and more applications
unpredicted adverse effects. Disruption of ECM may also
lead to tumor migration and metastasis, and untimely
of size aggregation and size-
normalization of blood vessels may make it difficult for shrinkage strategies, there comes
nanoparticles to passively target tumor tissues through an EPR a question of which strategy is
effect. The extent and timing of remodeling of microenviron- better.
ment need to be carefully balanced.
5.5. Sensitivity of Transformation. Because of the high
interstitial pressure of tumor sites, nanoparticles that enter Moreover, the shrinkage strategy also shows capabilities
tumor sites are easily pumped back to vessels, and the more than penetration promotion and nucleus delivery. The
retention time of the original nanoparticles will not be too shrinking procedure always accompanies shell dissociation or
long. Therefore, the sensitivity and response time with stimuli volume squeezing of nanoparticles, which have potential for
are a key factor in designing size-tunable nanoparticles. As specific drug release. Besides, nanoparticles’ size influences
demonstrated above, pH-triggered reactions possess the their distribution not only in tumor tissue, but also in normal
advantages of quick response and ultrasensitivity, while the organs. Small-sized nanoparticles tend to accumulate in organs
reaction time between enzyme and substate is not that fast. such as kidney, spleen, and lung, which generate different
The enzyme-induced aggregations are usually observed at the potential functions and strategies for nanoparticle-based
12 or 24 h time point, and a majority of the original therapy, such as secondary distribution in spleen for immune
nanoparticles may be eliminated before aggregations happen. activation, rapid renal clearance for elimination of toxic
5.6. Possibility of Clinical Translation. In the last few materials, and lung-targeting function for alveolus remodeling.
decades, an EPR effect was considered to be the basic principle With more and more applications of size aggregation and
for passive drug delivery to tumor sites.161 However, there are size-shrinkage strategies, there comes a question of which
more and more controversies and doubts about the EPR effect strategy is better. In fact, though the two strategies are totally
because of the unsatisfactory therapeutic efficacy in clinical opposite from the methodology, the ultimate goal and
trials. Also, it has been reported that only 0.7% nanoparticles therapeutic efficacy are the same, which are to prolong the
can be finally delivered to tumor sites.162 One of the most penetration and retention time in tumor sites at the same time.
important reasons is the difference between human and model Besides, the emergence of the size-reversible strategies further
animals,163 and there is no single animal model that can fully optimizes the strategies of size change.175,176 And recently,
reproduce human cancers. In addition, the EPR effect only nanoparticles were produced that could simultaneously change
exhibits good results in animal models, which is because of the the tumor microenvironment along with the size change,177,178
110 DOI: 10.1021/acscentsci.9b01139
ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

such as hypoxia relief,179 normalization of vasculature,180 and (11) Perrault, S. D.; Walkey, C.; Jennings, T.; Fischer, H. C.; Chan,
extracellular matrix modification,181 endowing a better W. C. Mediating tumor targeting efficiency of nanoparticles through
therapeutic effect. design. Nano Lett. 2009, 9 (5), 1909−1915.
In short, it is trendy to carefully design responsive, size- (12) Sykes, E. A.; Chen, J.; Zheng, G.; Chan, W. C. W. Investigating
the Impact of Nanoparticle Size on Active and Passive Tumor
changeable nanoparticles based on a certain tumor, even a
Targeting Efficiency. ACS Nano 2014, 8 (6), 5696−5706.
certain part of the tumor tissue. We look forward the time (13) She, W.; Luo, K.; Zhang, C.; Wang, G.; Geng, Y.; Li, L.; He, B.;
when multiresponsive nanoparticles with multistage size Gu, Z. The potential of self-assembled, pH-responsive nanoparticles
changes are developed and optimize current treatment, of mPEGylated peptide dendron-doxorubicin conjugates for cancer
which seems hard to achieve today but will be of great value. therapy. Biomaterials 2013, 34 (5), 1613−1623.

■ AUTHOR INFORMATION
Corresponding Author
(14) Liu, X.; Chen, Y.; Li, H.; Huang, N.; Jin, Q.; Ren, K.; Ji, J.
Enhanced retention and cellular uptake of nanoparticles in tumors by
controlling their aggregation behavior. ACS Nano 2013, 7 (7), 6244−
6257.
*E-mail: gaohuile@scu.edu.cn; gaohuilescu@163.com. (15) Champion, J. A.; Walker, A.; Mitragotri, S. Role of particle size
ORCID in phagocytosis of polymeric microspheres. Pharm. Res. 2008, 25 (8),
Huile Gao: 0000-0002-5355-7238 1815−1821.
(16) He, C.; Hu, Y.; Yin, L.; Tang, C.; Yin, C. Effects of particle size
Author Contributions and surface charge on cellular uptake and biodistribution of polymeric
#
W.Y. and R.L. contributed equally to this work. nanoparticles. Biomaterials 2010, 31 (13), 3657−3666.
Notes (17) Choi, H. S.; Liu, W.; Misra, P.; Tanaka, E.; Zimmer, J. P.; Itty
Ipe, B.; Bawendi, M. G.; Frangioni, J. V. Renal clearance of quantum
The authors declare no competing financial interest.


dots. Nat. Biotechnol. 2007, 25 (10), 1165−1170.
(18) Liu, D.; Mori, A.; Huang, L. Role of liposome size and RES
ACKNOWLEDGMENTS blockade in controlling biodistribution and tumor uptake of GM1-
This work was supported by National Natural Science containing liposomes. Biochim. Biophys. Acta, Biomembr. 1992, 1104
Foundation of China (31571016), Research Foundation of (1), 95−101.
Sichuan Science and Technology Department (19YYJC2250), (19) Chen, L.-T.; Weiss, L. The Role of the Sinus Wall in the
Passage of Erythrocytes Through the Spleen. Blood 1973, 41, 529−
the Young Elite Scientists Sponsorship Program by CAST
537.
(2017QNR001), the Fundamental Research Funds for the (20) Hobbs, S. K.; Monsky, W. L.; Yuan, F.; Roberts, W. G.; Griffith,
Central Universities, and 111 Project (B18035).


L.; Torchilin, V. P.; Jain, R. K. Regulation of transport pathways in
tumor vessels: role of tumor type and microenvironment. Proc. Natl.
REFERENCES Acad. Sci. U. S. A. 1998, 95 (8), 4607−4612.
(1) Pan, L.; He, Q.; Liu, J.; Chen, Y.; Ma, M.; Zhang, L.; Shi, J. (21) Yuan, F.; Dellian, M.; Fukumura, D.; Leunig, M.; Berk, D. A.;
Nuclear-targeted drug delivery of TAT peptide-conjugated mono- Torchilin, V. P.; Jain, R. K. Vascular permeability in a human tumor
disperse mesoporous silica nanoparticles. J. Am. Chem. Soc. 2012, 134 xenograft: molecular size dependence and cutoff size. Cancer Res.
(13), 5722−5725. 1995, 55 (17), 3752−3756.
(2) Pan, L.; Liu, J.; He, Q.; Shi, J. MSN-mediated sequential (22) Poh, S.; Chelvam, V.; Low, P. S. Comparison of nanoparticle
vascular-to-cell nuclear-targeted drug delivery for efficient tumor penetration into solid tumors and sites of inflammation: studies using
regression. Adv. Mater. 2014, 26 (39), 6742−6748. targeted and nontargeted liposomes. Nanomedicine 2015, 10 (9),
(3) Gao, H. Shaping Tumor Microenvironment for Improving 1439−1449.
Nanoparticle Delivery. Curr. Drug Metab. 2016, 17 (8), 731−736. (23) Wang, R.; Shen, Q.; Li, X.; Xie, C.; Lu, W.; Wang, S.; Wang, J.;
(4) Miao, L.; Lin, C. M.; Huang, L. Stromal barriers and strategies Wang, D.; Liu, M. Efficacy of inverso isomer of CendR peptide on
for the delivery of nanomedicine to desmoplastic tumors. J. Controlled tumor tissue penetration. Acta Pharm. Sin. B 2018, 8 (5), 825−832.
Release 2015, 219, 192−204. (24) Tang, L.; Yang, X.; Yin, Q.; Cai, K.; Wang, H.; Chaudhury, I.;
(5) Setyawati, M. I.; Tay, C. Y.; Bay, B. H.; Leong, D. T. Gold Yao, C.; Zhou, Q.; Kwon, M.; Hartman, J. A.; Dobrucki, I. T.;
Nanoparticles Induced Endothelial Leakiness Depends on Particle Dobrucki, L. W.; Borst, L. B.; Lezmi, S.; Helferich, W. G.; Ferguson,
Size and Endothelial Cell Origin. ACS Nano 2017, 11 (5), 5020− A. L.; Fan, T. M.; Cheng, J. Investigating the optimal size of
5030. anticancer nanomedicine. Proc. Natl. Acad. Sci. U. S. A. 2014, 111
(6) Li, C.; Wang, J.; Wang, Y.; Gao, H.; Wei, G.; Huang, Y.; Yu, H.; (43), 15344−15349.
Gan, Y.; Wang, Y.; Mei, L.; Chen, H.; Hu, H.; Zhang, Z.; Jin, Y. (25) Maman, S.; Witz, I. P. A history of exploring cancer in context.
Recent progress in drug delivery. Acta Pharm. Sin. B 2019, 9 (6), Nat. Rev. Cancer 2018, 18 (6), 359−376.
1145−1162. (26) Danhier, F. To exploit the tumor microenvironment: Since the
(7) Carnovale, C.; Bryant, G.; Shukla, R.; Bansal, V. Size, shape and EPR effect fails in the clinic, what is the future of nanomedicine? J.
surface chemistry of nano-gold dictate its cellular interactions, uptake Controlled Release 2016, 244, 108−121.
and toxicity. Prog. Mater. Sci. 2016, 83, 152−190. (27) Carpenter, A. W.; Schoenfisch, M. H. Nitric oxide release: part
(8) Talamini, L.; Violatto, M. B.; Cai, Q.; Monopoli, M. P.; Kantner, II. Therapeutic applications. Chem. Soc. Rev. 2012, 41 (10), 3742−
K.; Krpetic, Z.; Perez-Potti, A.; Cookman, J.; Garry, D.; Silveira, C. P.; 3752.
Boselli, L.; Pelaz, B.; Serchi, T.; Cambier, S.; Gutleb, A. C.; Feliu, N.; (28) Papageorgis, P.; Stylianopoulos, T. Role of TGFβ in regulation
Yan, Y.; Salmona, M.; Parak, W. J.; Dawson, K. A.; Bigini, P. Influence of the tumor microenvironment and drug delivery (review). Int. J.
of Size and Shape on the Anatomical Distribution of Endotoxin-Free Oncol. 2015, 46 (3), 933−943.
Gold Nanoparticles. ACS Nano 2017, 11 (6), 5519−5529. (29) Gratton, S. E.; Ropp, P. A.; Pohlhaus, P. D.; Luft, J. C.;
(9) Hashizume, H.; Baluk, P.; Morikawa, S.; McLean, J. W.; Madden, V. J.; Napier, M. E.; DeSimone, J. M. The effect of particle
Thurston, G.; Roberge, S.; Jain, R. K.; McDonald, D. M. Openings design on cellular internalization pathways. Proc. Natl. Acad. Sci. U. S.
between Defective Endothelial Cells Explain Tumor Vessel Leakiness. A. 2008, 105 (33), 11613−11618.
Am. J. Pathol. 2000, 156 (4), 1363−1380. (30) Jiang, W.; Kim, B. Y.; Rutka, J. T.; Chan, W. C. Nanoparticle-
(10) Moghimi, S. M.; Hunter, A. C.; Murray, J. C. Nanomedicine: mediated cellular response is size-dependent. Nat. Nanotechnol. 2008,
current status and future prospects. FASEB J. 2005, 19 (3), 311−330. 3 (3), 145−150.

111 DOI: 10.1021/acscentsci.9b01139


ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

(31) Huang, J.; Bu, L.; Xie, J.; Chen, K.; Cheng, Z.; Li, X.; Chen, X. (51) Wang, Z.; An, H. W.; Hou, D.; Wang, M.; Zeng, X.; Zheng, R.;
Effects of nanoparticle size on cellular uptake and liver MRI with Wang, L.; Wang, K.; Wang, H.; Xu, W. Addressable Peptide Self-
polyvinylpyrrolidone-coated iron oxide nanoparticles. ACS Nano Assembly on the Cancer Cell Membrane for Sensitizing Chemo-
2010, 4 (12), 7151−7160. therapy of Renal Cell Carcinoma. Adv. Mater. 2019, 31 (11),
(32) Doherty, G. J.; McMahon, H. T. Mechanisms of endocytosis. e1807175.
Annu. Rev. Biochem. 2009, 78, 857−902. (52) Liu, Y.; Yu, Y.; Lam, J. W.; Hong, Y.; Faisal, M.; Yuan, W. Z.;
(33) Wang, Z., Caveolae-mediated Delivery of Therapeutic Nano- Tang, B. Z. Simple biosensor with high selectivity and sensitivity:
particles across Blood-endothelial Barrier. Austin J. Anal Pharm. Chem. thiol-specific biomolecular probing and intracellular imaging by AIE
2014, 1 (4). fluorogen on a TLC plate through a thiol-ene click mechanism. Chem.
(34) Chithrani, B. D.; Ghazani, A. A.; Chan, W. C. W. Determining - Eur. J. 2010, 16 (28), 8433−8438.
the size and shape dependence of gold nanoparticle uptake into (53) Ai, X.; Ho, C. J. H.; Aw, J.; Attia, A. B. E.; Mu, J.; Wang, Y.;
mammalian cells. Nano Lett. 2006, 6 (4), 662−668. Wang, X.; Wang, Y.; Liu, X.; Chen, H.; Gao, M.; Chen, X.; Yeow, E.
(35) Swanson, J. A. Shaping cups into phagosomes and macro- K. L.; Liu, G.; Olivo, M.; Xing, B. In vivo covalent cross-linking of
pinosomes. Nat. Rev. Mol. Cell Biol. 2008, 9 (8), 639−649. photon-converted rare-earth nanostructures for tumour localization
(36) Wang, T.; Wang, L.; Li, X.; Hu, X.; Han, Y.; Luo, Y.; Wang, Z.; and theranostics. Nat. Commun. 2016, 7, 10432.
Li, Q.; Aldalbahi, A.; Wang, L.; Song, S.; Fan, C.; Zhao, Y.; Wang, M.; (54) Liang, G.; Ren, H.; Rao, J. A biocompatible condensation
Chen, N. Size-Dependent Regulation of Intracellular Trafficking of reaction for controlled assembly of nanostructures in living cells. Nat.
Polystyrene Nanoparticle-Based Drug-Delivery Systems. ACS Appl. Chem. 2010, 2 (1), 54−60.
Mater. Interfaces 2017, 9 (22), 18619−18625. (55) Ruan, S.; Hu, C.; Tang, X.; Cun, X.; Xiao, W.; Shi, K.; He, Q.;
(37) Chithrani, B. D.; Chan, W. C. Elucidating the mechanism of Gao, H. Increased Gold Nanoparticle Retention in Brain Tumors by
cellular uptake and removal of protein-coated gold nanoparticles of in Situ Enzyme-Induced Aggregation. ACS Nano 2016, 10 (11),
different sizes and shapes. Nano Lett. 2007, 7 (6), 1542−1550. 10086−10098.
(38) Mei, L.; Rao, J.; Liu, Y.; Li, M.; Zhang, Z.; He, Q. Effective (56) Hu, Q.; Sun, W.; Lu, Y.; Bomba, H. N.; Ye, Y.; Jiang, T.;
treatment of the primary tumor and lymph node metastasis by Isaacson, A. J.; Gu, Z. Tumor Microenvironment-Mediated
polymeric micelles with variable particle sizes. J. Controlled Release Construction and Deconstruction of Extracellular Drug-Delivery
2018, 292, 67−77. Depots. Nano Lett. 2016, 16 (2), 1118−1126.
(39) Gorlich, D.; Kutay, U. Transport between the cell nucleus and (57) Chien, M. P.; Rush, A. M.; Thompson, M. P.; Gianneschi, N. C.
the cytoplasm. Annu. Rev. Cell Dev. Biol. 1999, 15, 607−660. Programmable shape-shifting micelles. Angew. Chem., Int. Ed. 2010, 49
(40) Rippe, K. Dynamic organization of the cell nucleus. Curr. Opin. (30), 5076−5080.
Genet. Dev. 2007, 17 (5), 373−380. (58) Jain, S.; Bates, F. S. On the origins of morphological complexity
(41) Penman, S. RNA metabolism in the HeLa cell nucleus. J. Mol. in block copolymer surfactants. Science 2003, 300 (5618), 460−464.
Biol. 1966, 17 (1), 117−130. (59) Sundararaman, A.; Stephan, T.; Grubbs, R. B. Reversible
(42) Huang, K.; Ma, H.; Liu, J.; Huo, S.; Kumar, A.; Wei, T.; Zhang, restructuring of aqueous block copolymer assemblies through
X.; Jin, S.; Gan, Y.; Wang, P. C.; He, S.; Zhang, X.; Liang, X. J. Size- stimulus-induced changes in amphiphilicity. J. Am. Chem. Soc. 2008,
dependent localization and penetration of ultrasmall gold nano- 130 (37), 12264−12265.
particles in cancer cells, multicellular spheroids, and tumors in vivo. (60) Ku, T.-H.; Chien, M.-P.; Thompson, M. P.; Sinkovits, R. S.;
ACS Nano 2012, 6 (5), 4483−4493. Olson, N. H.; Baker, T. S.; Gianneschi, N. C. Controlling and
(43) Huo, S. D.; Jin, S. B.; Ma, X. W.; Xue, X. D.; Yang, K. N.; Switching the Morphology of Micellar Nanoparticles with Enzymes. J.
Kumar, A.; Wang, P. C.; Zhang, J. C.; Hu, Z. B.; Liang, X. J. Am. Chem. Soc. 2011, 133 (22), 8392−8395.
Ultrasmall Gold Nanoparticles as Carriers for Nucleus-Based Gene (61) Feng, Z.; Zhang, T.; Wang, H.; Xu, B. Supramolecular catalysis
Therapy Due to Size-Dependent Nuclear Entry. ACS Nano 2014, 8 and dynamic assemblies for medicine. Chem. Soc. Rev. 2017, 46 (21),
(6), 5852−5862. 6470−6479.
(44) Hinde, E.; Thammasiraphop, K.; Duong, H. T.; Yeow, J.; (62) Zhou, J.; Du, X.; Yamagata, N.; Xu, B. Enzyme-Instructed Self-
Karagoz, B.; Boyer, C.; Gooding, J. J.; Gaus, K. Pair correlation Assembly of Small D-Peptides as a Multiple-Step Process for
microscopy reveals the role of nanoparticle shape in intracellular Selectively Killing Cancer Cells. J. Am. Chem. Soc. 2016, 138 (11),
transport and site of drug release. Nat. Nanotechnol. 2017, 12 (1), 3813−3823.
81−89. (63) Zheng, Z.; Chen, P.; Xie, M.; Wu, C.; Luo, Y.; Wang, W.; Jiang,
(45) Pan, L. M.; Liu, J. A.; He, Q. J.; Wang, L. J.; Shi, J. L. J.; Liang, G. Cell Environment-Differentiated Self-Assembly of
Overcoming multidrug resistance of cancer cells by direct intranuclear Nanofibers. J. Am. Chem. Soc. 2016, 138 (35), 11128−11131.
drug delivery using TAT-conjugated mesoporous silica nanoparticles. (64) Engin, K.; Leeper, D. B.; Cater, J. R.; Thistlethwaite, A. J.;
Biomaterials 2013, 34 (11), 2719−2730. Tupchong, L.; Mcfarlane, J. D. Extracellular Ph Distribution in
(46) Zhu, Y. X.; Jia, H. R.; Pan, G. Y.; Ulrich, N. W.; Chen, Z.; Wu, Human Tumors. Int. J. Hyperthermia 1995, 11 (2), 211−216.
F. G. Development of a Light-Controlled Nanoplatform for Direct (65) Wang, Y.; Lin, Y. X.; Qiao, Z. Y.; An, H. W.; Qiao, S. L.; Wang,
Nuclear Delivery of Molecular and Nanoscale Materials. J. Am. Chem. L.; Rajapaksha, R. P.; Wang, H. Self-assembled autophagy-inducing
Soc. 2018, 140 (11), 4062−4070. polymeric nanoparticles for breast cancer interference in-vivo. Adv.
(47) Pan, L.; Liu, J.; Shi, J. Cancer cell nucleus-targeting Mater. 2015, 27 (16), 2627−2634.
nanocomposites for advanced tumor therapeutics. Chem. Soc. Rev. (66) Yang, P.; Luo, Q.; Qi, G.; Gao, Y.; Li, B.; Zhang, J.; Wang, L.;
2018, 47 (18), 6930−6946. Wang, H. Host Materials Transformable in Tumor Microenvironment
(48) Zhang, D.; Qi, G. B.; Zhao, Y. X.; Qiao, S. L.; Yang, C.; Wang, for Homing Theranostics. Adv. Mater. 2017, 29 (15), 1605869.
H. In Situ Formation of Nanofibers from Purpurin18-Peptide (67) Li, H.; Chen, Y.; Li, Z.; Li, X.; Jin, Q.; Ji, J. Hemoglobin as a
Conjugates and the Assembly Induced Retention Effect in Tumor Smart pH-Sensitive Nanocarrier To Achieve Aggregation Enhanced
Sites. Adv. Mater. 2015, 27 (40), 6125−6130. Tumor Retention. Biomacromolecules 2018, 19 (6), 2007−2013.
(49) Yuan, Y.; Wang, L.; Du, W.; Ding, Z.; Zhang, J.; Han, T.; An, (68) Ju, K. Y.; Kang, J.; Pyo, J.; Lim, J.; Chang, J. H.; Lee, J. K. pH-
L.; Zhang, H.; Liang, G. Intracellular Self-Assembly of Taxol Induced aggregated melanin nanoparticles for photoacoustic signal
Nanoparticles for Overcoming Multidrug Resistance. Angew. Chem., amplification. Nanoscale 2016, 8 (30), 14448−14456.
Int. Ed. 2015, 54 (33), 9700−9704. (69) Song, J.; Kim, J.; Hwang, S.; Jeon, M.; Jeong, S.; Kim, C.; Kim,
(50) Julien, O.; Wells, J. A. Caspases and their substrates. Cell Death S. ″Smart″ gold nanoparticles for photoacoustic imaging: an imaging
Differ. 2017, 24 (8), 1380−1389. contrast agent responsive to the cancer microenvironment and signal

112 DOI: 10.1021/acscentsci.9b01139


ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

amplification via pH-induced aggregation. Chem. Commun. (Cam- (89) Klaikherd, A.; Nagamani, C.; Thayumanavan, S. Multi-stimuli
bridge, U. K.) 2016, 52 (53), 8287−8290. sensitive amphiphilic block copolymer assemblies. J. Am. Chem. Soc.
(70) Li, S. D.; Huang, L. Pharmacokinetics and biodistribution of 2009, 131 (13), 4830−4838.
nanoparticles. Mol. Pharmaceutics 2008, 5 (4), 496−504. (90) Roy, D.; Cambre, J. N.; Sumerlin, B. S. Triply-responsive
(71) Nam, J.; Ha, Y. S.; Hwang, S.; Lee, W.; Song, J.; Yoo, J.; Kim, S. boronic acid block copolymers: solution self-assembly induced by
pH-responsive gold nanoparticles-in-liposome hybrid nanostructures changes in temperature, pH, or sugar concentration. Chem. Commun.
for enhanced systemic tumor delivery. Nanoscale 2013, 5 (21), (Cambridge, U. K.) 2009, No. 16, 2106−2108.
10175−10178. (91) Agut, W.; Brulet, A.; Schatz, C.; Taton, D.; Lecommandoux, S.
(72) Krebs, M. R.; Domike, K. R.; Donald, A. M. Protein pH and temperature responsive polymeric micelles and polymersomes
aggregation: more than just fibrils. Biochem. Soc. Trans. 2009, 37 by self-assembly of poly[2-(dimethylamino)ethyl methacrylate]-b-
(Pt 4), 682−686. poly(glutamic acid) double hydrophilic block copolymers. Langmuir
(73) Kundu, P. K.; Samanta, D.; Leizrowice, R.; Margulis, B.; Zhao, 2010, 26 (13), 10546−10554.
H.; Borner, M.; Udayabhaskararao, T.; Manna, D.; Klajn, R. Light- (92) Liu, F. H.; Cong, Y.; Qi, G. B.; Ji, L.; Qiao, Z. Y.; Wang, H.
controlled self-assembly of non-photoresponsive nanoparticles. Nat. Near-Infrared Laser-Driven in Situ Self-Assembly as a General
Chem. 2015, 7 (8), 646−652. Strategy for Deep Tumor Therapy. Nano Lett. 2018, 18 (10),
(74) Pieroni, O.; Fissi, A.; Angelini, N.; Lenci, F. Photoresponsive 6577−6584.
polypeptides. Acc. Chem. Res. 2001, 34 (1), 9−17. (93) Wu, W. S. The signaling mechanism of ROS in tumor
(75) Cheng, X.; Sun, R.; Yin, L.; Chai, Z.; Shi, H.; Gao, M. Light- progression. Cancer Metastasis Rev. 2007, 25 (4), 695−705.
Triggered Assembly of Gold Nanoparticles for Photothermal Therapy (94) Lee, F. Y.; Vessey, A.; Rofstad, E.; Siemann, D. W.; Sutherland,
and Photoacoustic Imaging of Tumors In Vivo. Adv. Mater. 2017, 29 R. M. Heterogeneity of glutathione content in human ovarian cancer.
(6), 1604894. Cancer Res. 1989, 49 (19), 5244−5248.
(76) Kuk, S.; Lee, B. I.; Lee, J. S.; Park, C. B. Rattle-Structured (95) Liu, T.; Lai, L.; Song, Z.; Chen, T. A Sequentially Triggered
Upconversion Nanoparticles for Near-IR-Induced Suppression of Nanosystem for Precise Drug Delivery and Simultaneous Inhibition of
Alzheimer’s beta-Amyloid Aggregation. Small 2017, 13 (11), Cancer Growth, Migration, and Invasion. Adv. Funct. Mater. 2016, 26
1603139. (43), 7775−7790.
(77) Zhao, M.; Wang, R.; Li, B.; Fan, Y.; Wu, Y.; Zhu, X.; Zhang, F. (96) Shimura, N.; Musya, A.; Hashimoto, T.; Kojima, S.; Kubodera,
Precise In Vivo Inflammation Imaging Using In Situ Responsive A.; Sasaki, T. Usefulness of 99m Tc-d,l-HMPAO for estimation of
Cross-linking of Glutathione-Modified Ultra-Small NIR-II Lanthanide GSH content in tumor tissues. Nucl. Med. Biol. 2000, 27 (6), 577−
Nanoparticles. Angew. Chem., Int. Ed. 2019, 58 (7), 2050−2054. 580.
(78) Xu, J.; Xu, L.; Wang, C.; Yang, R.; Zhuang, Q.; Han, X.; Dong, (97) Yuan, D.; Ding, L.; Sun, Z.; Li, X. MRI/Fluorescence bimodal
Z.; Zhu, W.; Peng, R.; Liu, Z. Near-Infrared-Triggered Photodynamic amplification system for cellular GSH detection and tumor cell
imaging based on manganese dioxide nanosheet. Sci. Rep. 2018, 8,
Therapy with Multitasking Upconversion Nanoparticles in Combina-
No. 1747, DOI: 10.1038/s41598-018-20110-z.
tion with Checkpoint Blockade for Immunotherapy of Colorectal
(98) Bardwell, J. C. A.; Mcgovern, K.; Beckwith, J. Identification of a
Cancer. ACS Nano 2017, 11 (5), 4463−4474.
protein required for disulfide bond formation in vivo. Cell 1991, 67
(79) Zhao, T.; Wang, P.; Li, Q.; Al-Khalaf, A. A.; Hozzein, W. N.;
(3), 581−589.
Zhang, F.; Li, X.; Zhao, D. Near-Infrared Triggered Decomposition of
(99) Gao, Z.; Hou, Y.; Zeng, J.; Chen, L.; Liu, C.; Yang, W.; Gao, M.
Nanocapsules with High Tumor Accumulation and Stimuli
Tumor Microenvironment-Triggered Aggregation of Antiphagocyto-
Responsive Fast Elimination. Angew. Chem., Int. Ed. 2018, 57 (10), sis (99m) Tc-Labeled Fe3 O4 Nanoprobes for Enhanced Tumor
2611−2615. Imaging In Vivo. Adv. Mater. 2017, 29 (24), 1701095.
(80) Qiao, S. L.; Ma, Y.; Wang, Y.; Lin, Y. X.; An, H. W.; Li, L. L.; (100) Li, F.; Cui, L.; Yu, D.; Hao, H.; Liu, Y.; Zhao, X.; Pang, Y.;
Wang, H. General Approach of Stimuli-Induced Aggregation for Zhu, H.; Du, W. Exogenous glutathione improves intracellular
Monitoring Tumor Therapy. ACS Nano 2017, 11 (7), 7301−7311. glutathione synthesis via the gamma-glutamyl cycle in bovine zygotes
(81) Schmaljohann, D. Thermo- and pH-responsive polymers in and cleavage embryos. J. Cell. Physiol. 2019, 234 (5), 7384−7394.
drug delivery. Adv. Drug Delivery Rev. 2006, 58 (15), 1655−1670. (101) Wang, Y.; Li, H.; Jin, Q.; Ji, J. Intracellular host-guest assembly
(82) Schmaljohann, D.; Beyerlein, D.; Nitschke, M.; Werner, C. of gold nanoparticles triggered by glutathione. Chem. Commun.
Thermo-reversible swelling of thin hydrogel films immobilized by (Cambridge, U. K.) 2016, 52 (3), 582−585.
low-pressure plasma. Langmuir 2004, 20 (23), 10107−10114. (102) Gao, W.; Hu, C. M.; Fang, R. H.; Luk, B. T.; Su, J.; Zhang, L.
(83) Jeong, B.; Kim, S. W.; Bae, Y. H. Thermosensitive sol−gel Surface functionalization of gold nanoparticles with red blood cell
reversible hydrogels. Adv. Drug Delivery Rev. 2012, 64, 154−162. membranes. Adv. Mater. 2013, 25 (26), 3549−3553.
(84) Jones, S. T.; Walsh-Korb, Z.; Barrow, S. J.; Henderson, S. L.; (103) Suk, J. S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L. M.
del Barrio, J.; Scherman, O. A. The Importance of Excess Poly(N- PEGylation as a strategy for improving nanoparticle-based drug and
isopropylacrylamide) for the Aggregation of Poly(N-isopropylacryla- gene delivery. Adv. Drug Delivery Rev. 2016, 99 (Pt A), 28−51.
mide)-Coated Gold Nanoparticles. ACS Nano 2016, 10 (3), 3158− (104) Sun, M.; Liu, F.; Zhu, Y.; Wang, W.; Hu, J.; Liu, J.; Dai, Z.;
3165. Wang, K.; Wei, Y.; Bai, J.; Gao, W. Salt-induced aggregation of gold
(85) Wang, Y. C.; Tang, L. Y.; Li, Y.; Wang, J. Thermoresponsive nanoparticles for photoacoustic imaging and photothermal therapy of
block copolymers of poly(ethylene glycol) and polyphosphoester: cancer. Nanoscale 2016, 8 (8), 4452−4457.
thermo-induced self-assembly, biocompatibility, and hydrolytic (105) McKee, T. D.; Grandi, P.; Mok, W.; Alexandrakis, G.; Insin,
degradation. Biomacromolecules 2009, 10 (1), 66−73. N.; Zimmer, J. P.; Bawendi, M. G.; Boucher, Y.; Breakefield, X. O.;
(86) Idziak, I.; Avoce, D.; Lessard, D.; Gravel, D.; Zhu, X. X. Jain, R. K. Degradation of fibrillar collagen in a human melanoma
Thermosensitivity of Aqueous Solutions of Poly(N,N-diethylacryla- xenograft improves the efficacy of an oncolytic herpes simplex virus
mide). Macromolecules 1999, 32 (4), 1260−1263. vector. Cancer Res. 2006, 66 (5), 2509−2513.
(87) Loh, X. J.; Goh, S. H.; Li, J. New biodegradable thermogelling (106) Minchinton, A. I.; Tannock, I. F. Drug penetration in solid
copolymers having very low gelation concentrations. Biomacromole- tumours. Nat. Rev. Cancer 2006, 6 (8), 583−592.
cules 2007, 8 (2), 585−593. (107) Panté, N.; Kann, M. Nuclear pore complex is able to transport
(88) Vihola, H.; Laukkanen, A.; Hirvonen, J.; Tenhu, H. Binding and macromolecules with diameters of about 39 nm. Mol. Biol. Cell 2002,
release of drugs into and from thermosensitive poly(N-vinyl 13 (2), 425−434.
caprolactam) nanoparticles. Eur. J. Pharm. Sci. 2002, 16 (1−2), 69− (108) Chen, J. J.; Ding, J. X.; Wang, Y. C.; Cheng, J. J.; Ji, S. X.;
74. Zhuang, X. L.; Chen, X. S. Sequentially Responsive Shell-Stacked

113 DOI: 10.1021/acscentsci.9b01139


ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

Nanoparticles for Deep Penetration into Solid Tumors. Adv. Mater. (126) Cliff, W.; Triantafyllos, S.; Jian, C.; John, M.; Chauhan, V. P.;
2017, 29 (32), 1701170. Wen, J.; Zoran, P.; Jain, R. K.; Bawendi, M. G.; Dai, F. Multistage
(109) Choi, H. S.; Liu, W.; Misra, P.; Tanaka, E.; Zimmer, J. P.; Ipe, nanoparticle delivery system for deep penetration into tumor tissue.
B. I.; Bawendi, M. G.; Frangioni, J. V. Renal Clearance of Proc. Natl. Acad. Sci. U. S. A. 2011, 108 (6), 2426−2431.
Nanoparticles. Nat. Biotechnol. 2007, 25 (10), 1165. (127) Ruan, S.; Cao, X.; Cun, X.; Hu, G.; Zhou, Y.; Zhang, Y.; Lu,
(110) Chen, K.-H.; Lundy, D. J.; Toh, E. K.-W.; Chen, C.-H.; Shih, L.; He, Q.; Gao, H. Matrix metalloproteinase-sensitive size-shrinkable
C.; Chen, P.; Chang, H.-C.; Lai, J. J.; Stayton, P. S.; Hoffman, A. S.; nanoparticles for deep tumor penetration and pH triggered
Hsieh, P. C.-H. Nanoparticle distribution during systemic inflamma- doxorubicin release. Biomaterials 2015, 60, 100−110.
tion is size-dependent and organ-specific. Nanoscale 2015, 7 (38), (128) Ruan, S.; He, Q.; Gao, H. Matrix metalloproteinase triggered
15863−15872. size-shrinkable gelatin-gold fabricated nanoparticles for tumor micro-
(111) Zhou, K.; Wang, Y.; Huang, X.; Luby-Phelps, K.; Sumer, B. environment sensitive penetration and diagnosis of glioma. Nanoscale
D.; Gao, J. Tunable, ultrasensitive pH-responsive nanoparticles 2015, 7 (21), 9487−9496.
targeting specific endocytic organelles in living cells. Angew. Chem., (129) Cun, X.; Chen, J.; Ruan, S.; Zhang, L.; Wan, J.; He, Q.; Gao,
Int. Ed. 2011, 50 (27), 6109−6114. H. A Novel Strategy through Combining iRGD Peptide with Tumor-
(112) Zhu, Z.; Armes, S. P.; Liu, S. pH-Induced Micellization Microenvironment-Responsive and Multistage Nanoparticles for
Kinetics of ABC Triblock Copolymers Measured by Stopped-Flow Deep Tumor Penetration. ACS Appl. Mater. Interfaces 2015, 7 (49),
Light Scattering. Macromolecules 2005, 38 (23), 9803−9812. 27458−27466.
(113) Ma, X.; Wang, Y.; Zhao, T.; Li, Y.; Su, L. C.; Wang, Z.; Huang, (130) Hu, G.; Chun, X.; Wang, Y.; He, Q.; Gao, H. Peptide
G.; Sumer, B. D.; Gao, J. Ultra-pH-Sensitive Nanoprobe Library with mediated active targeting and intelligent particle size reduction-
Broad pH Tunability and Fluorescence Emissions. J. Am. Chem. Soc. mediated enhanced penetrating of fabricated nanoparticles for triple-
2014, 136 (31), 11085−11092. negative breast cancer treatment. Oncotarget 2015, 6 (38), 41258−
(114) Yuan, C.; Raghupathi, K.; Popere, B. C.; Ventura, J.; Dai, L.; 41274.
Thayumanavan, S. Composite supramolecular nanoassemblies with (131) Menzel, E. J.; Farr, C. Hyaluronidase and its substrate
independent stimulus sensitivities. Chem. Sci. 2014, 5 (1), 229−234. hyaluronan: biochemistry, biological activities and therapeutic uses.
(115) Ray, P.; Alhalhooly, L.; Ghosh, A.; Choi, Y.; Banerjee, S.; Cancer Lett. 1998, 131 (1), 3−11.
Mallik, S.; Banerjee, S.; Quadir, M. Size-Transformable, Multifunc- (132) Arpicco, S.; De Rosa, G.; Fattal, E. Lipid-Based Nanovectors
tional Nanoparticles from Hyperbranched Polymers for Environment- for Targeting of CD44-Overexpressing Tumor Cells. J. Drug Delivery
Specific Therapeutic Delivery. ACS Biomater. Sci. Eng. 2019, 5 (3), 2013, 2013 (4), 860780.
1354−1365. (133) Liu, R.; Hu, C.; Yang, Y.; Zhang, J.; Gao, H. Theranostic
(116) Kaneda, Y.; Yamamoto, Y.; Kamada, H.; Tsunoda, S.; nanoparticles with tumor-specific enzyme-triggered size reduction and
Tsutsumi, Y.; Hirano, T.; Mayumi, T. Antitumor activity of tumor
drug release to perform photothermal therapy for breast cancer
necrosis factor alpha conjugated with divinyl ether and maleic
treatment. Acta Pharm. Sin. B 2019, 9 (2), 410−420.
anhydride copolymer on solid tumors in mice. Cancer Res. 1998, 58
(134) Yu, W.; He, X.; Yang, Z.; Yang, X.; Xiao, W.; Liu, R.; Xie, R.;
(2), 290−295.
Qin, L.; Gao, H. Sequentially responsive biomimetic nanoparticles
(117) Mehrishi, J. N. Positively charged amino groups on the surface
with optimal size in combination with checkpoint blockade for
of normal and cancer cells. Eur. J. Cancer 1970, 6 (2), 127−137.
cascade synergetic treatment of breast cancer and lung metastasis.
(118) Kozin, S. V.; Shkarin, P.; Gerweck, L. E. The cell
transmembrane pH gradient in tumors enhances cytotoxicity of Biomaterials 2019, 217, 119309.
specific weak acid chemotherapeutics. Cancer Res. 2001, 61 (12), (135) Luo, Z.; Dai, Y.; Gao, H. Development and application of
4740−4743. hyaluronic acid in tumor targeting drug delivery. Acta Pharm. Sin. B
(119) Li, H.-J.; Du, J.-Z.; Du, X.-J.; Xu, C.-F.; Sun, C.-Y.; Wang, H.- 2019, 9 (6), 1099−1112.
X.; Cao, Z.-T.; Yang, X.-Z.; Zhu, Y.-H.; Nie, S.; Wang, J. Stimuli- (136) Hu, C.; Cun, X.; Ruan, S.; Liu, R.; Xiao, W.; Yang, X.; Yang,
responsive clustered nanoparticles for improved tumor penetration Y.; Yang, C.; Gao, H. Enzyme-triggered size shrink and laser-
and therapeutic efficacy. Proc. Natl. Acad. Sci. U. S. A. 2016, 113 (15), enhanced NO release nanoparticles for deep tumor penetration and
4164−4169. combination therapy. Biomaterials 2018, 168, 64−75.
(120) Xuan, M.; Shao, J.; Dai, L.; He, Q.; Li, J. Macrophage Cell (137) Hu, C.; Yang, X.; Liu, R.; Ruan, S.; Zhou, Y.; Xiao, W.; Yu,
Membrane Camouflaged Mesoporous Silica Nanocapsules for In Vivo W.; Yang, C.; Gao, H. Coadministration of iRGD with Multistage
Cancer Therapy. Adv. Healthcare Mater. 2015, 4 (11), 1645−1652. Responsive Nanoparticles Enhanced Tumor Targeting and Pene-
(121) Cordes, E. H.; Jencks, W. P. On the Mechanism of Schiff Base tration Abilities for Breast Cancer Therapy. ACS Appl. Mater.
Formation and Hydrolysis. J. Am. Chem. Soc. 1962, 84 (5), 832−837. Interfaces 2018, 10 (26), 22571−22579.
(122) Helmlinger, G.; Yuan, F.; Dellian, M.; Jain, R. K. Interstitial (138) Niu, Y.; Zhu, J.; Li, Y.; Shi, H.; Gong, Y.; Li, R.; Huo, Q.; Ma,
pH and pO2 gradients in solid tumors in vivo: High-resolution T.; Liu, Y. Size shrinkable drug delivery nanosystems and priming the
measurements reveal a lack of correlation. Nat. Med. 1997, 3 (2), tumor microenvironment for deep intratumoral penetration of
177−182. nanoparticles. J. Controlled Release 2018, 277, 35−47.
(123) Coussens, L. M.; Tinkle, C. L.; Hanahan, D.; Werb, Z. MMP- (139) Tseng, S. J.; Kempson, I. M.; Huang, K. Y.; Li, H. J.; Fa, Y. C.;
9 supplied by bone marrow-derived cells contributes to skin Ho, Y. C.; Liao, Z. X.; Yang, P. C. Targeting Tumor Microenviron-
carcinogenesis. Cell 2000, 103 (3), 481−490. ment by Bioreduction-Activated Nanoparticles for Light-Triggered
(124) Vu, T. H.; Shipley, J. M.; Bergers, G.; Berger, J. E.; Helms, J. Virotherapy. ACS Nano 2018, 12 (10), 9894−9902.
A.; Hanahan, D.; Shapiro, S. D.; Senior, R. M.; Werb, Z. MMP-9/ (140) Li, Y.; Hu, H.; Zhou, Q.; Ao, Y.; Xiao, C.; Wan, J.; Wan, Y.;
Gelatinase B Is a Key Regulator of Growth Plate Angiogenesis and Xu, H.; Li, Z.; Yang, X. alpha-Amylase- and Redox-Responsive
Apoptosis of Hypertrophic Chondrocytes. Cell 1998, 93 (3), 411− Nanoparticles for Tumor-Targeted Drug Delivery. ACS Appl. Mater.
422. Interfaces 2017, 9 (22), 19215−19230.
(125) Oh, J.; Takahashi, R.; Kondo, S.; Mizoguchi, A.; Adachi, E.; (141) Guo, X.; Deng, G.; Liu, J.; Zou, P.; Du, F.; Liu, F.; Chen, A.
Sasahara, R. M.; Nishimura, S.; Imamura, Y.; Kitayama, H.; Alexander, T.; Hu, R.; Li, M.; Zhang, S.; Tang, Z.; Han, L.; Liu, J.; Sheth, K. N.;
D. B.; Ide, C.; Horan, T. P.; Arakawa, T.; Yoshida, H.; Nishikawa, S.- Chen, Q.; Gou, X.; Zhou, J. Thrombin-Responsive, Brain-Targeting
i.; Itoh, Y.; Seiki, M.; Itohara, S.; Takahashi, C.; Noda, M. The Nanoparticles for Improved Stroke Therapy. ACS Nano 2018, 12 (8),
membrane-anchored MMP inhibitor RECK is a key regulator of 8723−8732.
extracellular matrix integrity and angiogenesis. Cell 2001, 107 (6), (142) Guo, X.; Wei, X.; Jing, Y.; Zhou, S. Size Changeable
789−800. Nanocarriers with Nuclear Targeting for Effectively Overcoming

114 DOI: 10.1021/acscentsci.9b01139


ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

Multidrug Resistance in Cancer Therapy. Adv. Mater. 2015, 27 (41), (161) Li, H.; Li, J.; He, X.; Zhang, B.; Liu, C.; Li, Q.; Zhu, Y.;
6450−6456. Huang, W.; Zhang, W.; Qian, H.; Ge, L. Histology and antitumor
(143) Wang, H.; Li, Y.; Bai, H.; Shen, J.; Chen, X.; Ping, Y.; Tang, G. activity study of PTX-loaded micelle, a fluorescent drug delivery
A Cooperative Dimensional Strategy for Enhanced Nucleus-Targeted system prepared by PEG-TPP. Chin. Chem. Lett. 2019, 30 (5), 1083−
Delivery of Anticancer Drugs. Adv. Funct. Mater. 2017, 27 (24), 1088.
1700339. (162) Wilhelm, S.; Tavares, A. J.; Dai, Q.; Ohta, S.; Audet, J.;
(144) Pei, Q.; Hu, X.; Zheng, X.; Liu, S.; Li, Y.; Jing, X.; Xie, Z. Dvorak, H. F.; Chan, W. C. W. Analysis of nanoparticle delivery to
Light-Activatable Red Blood Cell Membrane-Camouflaged Dimeric tumours. Nature Reviews Materials 2016, 1 (5), 16014.
Prodrug Nanoparticles for Synergistic Photodynamic/Chemotherapy. (163) Shi, J.; Kantoff, P. W.; Wooster, R.; Farokhzad, O. C. Cancer
ACS Nano 2018, 12 (2), 1630−1641. nanomedicine: progress, challenges and opportunities. Nat. Rev.
(145) Luo, S.; Zhang, E.; Su, Y.; Cheng, T.; Shi, C. A review of NIR Cancer 2017, 17 (1), 20−37.
dyes in cancer targeting and imaging. Biomaterials 2011, 32 (29), (164) Li, J.; Kuang, Y.; Shi, J.; Zhou, J.; Medina, J. E.; Zhou, R.;
7127−7138. Yuan, D.; Yang, C.; Wang, H.; Yang, Z.; Liu, J.; Dinulescu, D. M.; Xu,
(146) Patel, N.; Pera, P.; Joshi, P.; Dukh, M.; Tabaczynski, W. A.; B. Enzyme-Instructed Intracellular Molecular Self-Assembly to Boost
Siters, K. E.; Kryman, M.; Cheruku, R. R.; Durrani, F.; Missert, J. R.; Activity of Cisplatin against Drug-Resistant Ovarian Cancer Cells.
Watson, R.; Ohulchanskyy, T. Y.; Tracy, E. C.; Baumann, H.; Pandey, Angew. Chem., Int. Ed. 2015, 54 (45), 13307−13311.
R. K. Highly Effective Dual-Function Near-Infrared (NIR) Photo- (165) Huang, P.; Gao, Y.; Lin, J.; Hu, H.; Liao, H. S.; Yan, X.; Tang,
sensitizer for Fluorescence Imaging and Photodynamic Therapy Y.; Jin, A.; Song, J.; Niu, G.; Zhang, G.; Horkay, F.; Chen, X. Tumor-
(PDT) of Cancer. J. Med. Chem. 2016, 59 (21), 9774−9787. Specific Formation of Enzyme-Instructed Supramolecular Self-
(147) Lu, T.; Shao, P.; Mathew, I.; Sand, A.; Sun, W. Synthesis and Assemblies as Cancer Theranostics. ACS Nano 2015, 9 (10),
photophysics of benzotexaphyrin: a near-infrared emitter and 9517−9527.
photosensitizer. J. Am. Chem. Soc. 2008, 130 (47), 15782−15783. (166) Son, J.; Kalafatovic, D.; Kumar, M.; Yoo, B.; Cornejo, M. A.;
(148) Cao, Z.; Ma, Y.; Sun, C.; Lu, Z.; Yao, Z.; Wang, J.; Li, D.; Contel, M.; Ulijn, R. V. Customizing Morphology, Size, and Response
Yuan, Y.; Yang, X. ROS-Sensitive Polymeric Nanocarriers with Red Kinetics of Matrix Metalloproteinase-Responsive Nanostructures by
Light-Activated Size Shrinkage for Remotely Controlled Drug Systematic Peptide Design. ACS Nano 2019, 13 (2), 1555−1562.
Release. Chem. Mater. 2018, 30 (2), 517−525. (167) Li, L. L.; Qiao, S. L.; Liu, W. J.; Ma, Y.; Wan, D.; Pan, J.;
(149) Thomas, S. N.; van der Vlies, A. J.; O’Neil, C. P.; Reddy, S. T.; Wang, H. Intracellular construction of topology-controlled polypep-
Yu, S. S.; Giorgio, T. D.; Swartz, M. A.; Hubbell, J. A. Engineering tide nanostructures with diverse biological functions. Nat. Commun.
complement activation on polypropylene sulfide vaccine nano- 2017, 8 (1), 1276.
particles. Biomaterials 2011, 32 (8), 2194−2203. (168) Liu, Y.; He, J.; Yang, K.; Yi, C.; Liu, Y.; Nie, L.; Khashab, N.
(150) Zhang, T.; Chen, X.; Xiao, C.; Zhuang, X.; Chen, X. Synthesis M.; Chen, X.; Nie, Z. Folding Up of Gold Nanoparticle Strings into
of a phenylboronic ester-linked PEG-lipid conjugate for ROS-
Plasmonic Vesicles for Enhanced Photoacoustic Imaging. Angew.
responsive drug delivery. Polym. Chem. 2017, 8 (40), 6209−6216.
Chem., Int. Ed. 2015, 54 (52), 15809−15812.
(151) Zhao, W.; Qiao, Z.; Duan, Z.; Wang, H. Synthesis and self-
(169) Liu, R.; Yu, M.; Yang, X.; Umeshappa, C. S.; Hu, C.; Yu, W.;
assembly of pH and ROS dual responsive Poly (beta-thioester) s.
Qin, L.; Huang, Y.; Gao, H. Linear Chimeric Triblock Molecules Self-
Huaxue Xuebao 2016, 74 (3), 234−240.
Assembled Micelles with Controllably Transformable Property to
(152) Xiao, W.; Gao, H. The impact of protein corona on the
Enhance Tumor Retention for Chemo-Photodynamic Therapy of
behavior and targeting capability of nanoparticle-based delivery
system. Int. J. Pharm. 2018, 552 (1−2), 328−339. Breast Cancer. Adv. Funct. Mater. 2019, 29 (23), 1808462.
(153) Bhattacharjee, S. DLS and zeta potential - What they are and (170) Xing, R.; Liu, K.; Jiao, T.; Zhang, N.; Ma, K.; Zhang, R.; Zou,
what they are not? J. Controlled Release 2016, 235, 337−351. Q.; Ma, G.; Yan, X. An Injectable Self-Assembling Collagen-Gold
(154) Ruan, S.; Qin, L.; Xiao, W.; Hu, C.; Zhou, Y.; Wang, R.; Sun, Hybrid Hydrogel for Combinatorial Antitumor Photothermal/Photo-
X.; Yu, W.; He, Q.; Gao, H. Acid-Responsive Transferrin Dissociation dynamic Therapy. Adv. Mater. 2016, 28 (19), 3669−3676.
and GLUT Mediated Exocytosis for Increased Blood-Brain Barrier (171) Wang, Z.; Yong, T. Y.; Wan, J.; Li, Z. H.; Zhao, H.; Zhao, Y.;
Transcytosis and Programmed Glioma Targeting Delivery. Adv. Funct. Gan, L.; Yang, X. L.; Xu, H. B.; Zhang, C. Temperature-sensitive
Mater. 2018, 28 (30), 1802227. fluorescent organic nanoparticles with aggregation-induced emission
(155) Hirata, K.; Maruyama, T.; Watanabe, H.; Maeda, H.; Nakajou, for long-term cellular tracing. ACS Appl. Mater. Interfaces 2015, 7 (5),
K.; Iwao, Y.; Ishima, Y.; Katsumi, H.; Hashida, M.; Otagiri, M. 3420−3425.
Genetically engineered mannosylated-human serum albumin as a (172) Yang, L.; Fang, W.; Ye, Y.; Wang, Z.; Hu, Q.; Tang, B. Z.
versatile carrier for liver-selective therapeutics. J. Controlled Release Redox-responsive fluorescent AIE bioconjugate with aggregation
2010, 145 (1), 9−16. enhanced retention features for targeted imaging reinforcement and
(156) Oliveira, B. L.; Guo, Z.; Bernardes, G. J. L. Inverse electron selective suppression of cancer cells. Materials Chemistry Frontiers
demand Diels-Alder reactions in chemical biology. Chem. Soc. Rev. 2019, 3 (7), 1335−1340.
2017, 46 (16), 4895−4950. (173) Gao, X.; Yue, Q.; Liu, Z.; Ke, M.; Zhou, X.; Li, S.; Zhang, J.;
(157) Devaraj, N. K. The Future of Bioorthogonal Chemistry. ACS Zhang, R.; Chen, L.; Mao, Y.; Li, C. Guiding Brain-Tumor Surgery via
Cent. Sci. 2018, 4 (8), 952−959. Blood-Brain-Barrier-Permeable Gold Nanoprobes with Acid-Trig-
(158) Taichi, M.; Nomura, S.; Nakase, I.; Imamaki, R.; Kizuka, Y.; gered MRI/SERRS Signals. Adv. Mater. 2017, 29 (21), 1603917.
Ota, F.; Dohmae, N.; Kitazume, S.; Taniguchi, N.; Tanaka, K. In Situ (174) Nguyen, M. M.; Carlini, A. S.; Chien, M. P.; Sonnenberg, S.;
Ligation of High- and Low-Affinity Ligands to Cell Surface Receptors Luo, C.; Braden, R. L.; Osborn, K. G.; Li, Y.; Gianneschi, N. C.;
Enables Highly Selective Recognition. Adv. Sci. (Weinh) 2017, 4 (11), Christman, K. L. Enzyme-Responsive Nanoparticles for Targeted
1700147. Accumulation and Prolonged Retention in Heart Tissue after
(159) Steen, E. J. L.; Edem, P. E.; Norregaard, K.; Jorgensen, J. T.; Myocardial Infarction. Adv. Mater. 2015, 27 (37), 5547−5552.
Shalgunov, V.; Kjaer, A.; Herth, M. M. Pretargeting in nuclear (175) Liang, H.; Ren, X.; Qian, J.; Zhang, X.; Meng, L.; Wang, X.; Li,
imaging and radionuclide therapy: Improving efficacy of theranostics L.; Fang, X.; Sha, X. Size-Shifting Micelle Nanoclusters Based on a
and nanomedicines. Biomaterials 2018, 179, 209−245. Cross-Linked and pH-Sensitive Framework for Enhanced Tumor
(160) Westerlund, K.; Vorobyeva, A.; Mitran, B.; Orlova, A.; Targeting and Deep Penetration Features. ACS Appl. Mater. Interfaces
Tolmachev, V.; Karlstrom, A. E.; Altai, M. Site-specific conjugation of 2016, 8 (16), 10136−10146.
recognition tags to trastuzumab for peptide nucleic acid-mediated (176) Su, Y.; Yu, T.; Chiang, W.; Chiu, H.; Chang, C.; Chiang, C.;
radionuclide HER2 pretargeting. Biomaterials 2019, 203, 73−85. Hu, S. Hierarchically Targeted and Penetrated Delivery of Drugs to

115 DOI: 10.1021/acscentsci.9b01139


ACS Cent. Sci. 2020, 6, 100−116
ACS Central Science Outlook

Tumors by Size-Changeable Graphene Quantum Dot Nanoaircrafts


for Photolytic Therapy. Adv. Funct. Mater. 2017, 27 (23), 1700056.
(177) Dong, X.; Liu, H. J.; Feng, H. Y.; Yang, S. C.; Liu, X. L.; Lai,
X.; Lu, Q.; Lovell, J. F.; Chen, H. Z.; Fang, C. Enhanced Drug
Delivery by Nanoscale Integration of a Nitric Oxide Donor To Induce
Tumor Collagen Depletion. Nano Lett. 2019, 19 (2), 997−1008.
(178) Yang, S.; Gao, H. Nanoparticles for modulating tumor
microenvironment to improve drug delivery and tumor therapy.
Pharmacol. Res. 2017, 126, 97−108.
(179) Chen, Q.; Feng, L. Z.; Liu, J. J.; Zhu, W. W.; Dong, Z. L.; Wu,
Y. F.; Liu, Z. Intelligent Albumin-MnO2 Nanoparticles as pH-/
H2O2-Responsive Dissociable Nanocarriers to Modulate Tumor
Hypoxia for Effective Combination Therapy. Adv. Mater. 2016, 28
(33), 7129−7136.
(180) Xiao, W.; Ruan, S.; Yu, W.; Wang, R.; Hu, C.; Liu, R.; Gao, H.
Normalizing Tumor Vessels To Increase the Enzyme-Induced
Retention and Targeting of Gold Nanoparticle for Breast Cancer
Imaging and Treatment. Mol. Pharmaceutics 2017, 14 (10), 3489−
3498.
(181) Hu, X.; He, P.; Qi, G.; Gao, Y.; Lin, Y.; Yang, C.; Yang, P.;
Hao, H.; Wang, L.; Wang, H. Transformable Nanomaterials as an
Artificial Extracellular Matrix for Inhibiting Tumor Invasion and
Metastasis. ACS Nano 2017, 11 (4), 4086−4096.

116 DOI: 10.1021/acscentsci.9b01139


ACS Cent. Sci. 2020, 6, 100−116

You might also like