You are on page 1of 19

European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

Contents lists available at ScienceDirect

European Journal of Pharmaceutics and Biopharmaceutics


journal homepage: www.elsevier.com/locate/ejpb

Potential drug delivery nanosystems for improving tumor penetration T



Feifei Peng, Ruirui Li, Fang Zhang, Li Qin, Guixia Ling, Peng Zhang
Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China

A R T I C LE I N FO A B S T R A C T

Keywords: Nanosystems, as one of the most important drug delivery systems, play a crucial rule in tumor therapy. However,
Potential the deep tumor penetration is retarded by the tumor physiological factors and nanomedicine properties. In this
Delivery review, we firstly elaborate the factors which impact tumor penetration, including the tumor physiological
Nanosystems factors and nanomedicine properties. Then, the latest and potential drug delivery nanosystems for improving
Penetration
tumor penetration are summarized and analyzed in detail. Moreover, recent combination therapies for im-
Therapies
proving penetration are described to enhance penetration. Finally, we summarize the typical clinical therapies of
potential drug delivery nanosystems.

1. Introduction past decades. There are a variety of treatments for anti-cancer, in-
cluding chemotherapy [1], immunotherapy [2], radiotherapy [3] and
Cancer has been the deadliest disease in many countries over the surgical resection [4]. In terms of chemotherapy, chemical drugs have

Abbreviations: AuNC@CBSA, small-sized renal-clearable red emission cationic bovine serum albumin-protected gold nanocluster; AuNCs, gold nanoclusters; AuNPs,
gold nanoparticles; AuNRs, gold nanorods; Bcl-2, a kind of antiapoptotic genes; Br-MSN, bromelain-conjugated mesoporous silica nanoparticle; BSA, bovine serum
albumin; CAF, cancer-associated fibroblast; CCM, cancer cell membrane; Ce6, chlorine e6; CD13, the receptor of iNGR which improves the capacity of tumor vessel
targeting; CD44, cluster determinant 44; CED, convection-enhanced delivery; CT, computed tomography; CUR, curcumin; DACHPt, 1,2-diaminocyclohexane-pla-
tinum(II); DC101, antivascular endothelial growth factor receptor-2 antibody; DGL, dendrigraft poly-lysine; DMA, dimethylmaleic anhydride; DOX, doxorubicin;
DpNG, biodegradable cationic paclitaxel-loaded nanogel; DSPE, 1,2-distearoyl-sn-glycero −3-phosphoethanolamine; DTX, docetaxel; DTX-pHPM, DTX-pH polymeric
micelle; ECM, extracellular matrix; EGFR, epidermal growth factor receptor; EPR, enhanced permeability and retention; ESMSV, enzyme-stimulated multistage
vector; FAP, fibroblast activation protein; FDA, Food and Drug Administration; FITC, fluorescein isothiocyanate; GBM, glioblastoma multiforme; GEM, gemcitabine;
GNPs, gelatin nanoparticles; GSH, glutathione; GSSG, glutathione disulfide; HA, Hyaluronan; HAase, hyaluronidase; HCC, hepatocellular carcinoma; HIFU, high
intensity focused ultrasound; HMSNs, hollow mesoporous silica nanoparticles; HN, NIR laser sensitive nitric oxide donor; HSA, human serum albumin; HSC, he-
matopoietic stem cell; HT, hyperthermia; ICG, indocyanine green; IDD, small-sized dendrimer prodrug; IFP, interstitial fluid pressure; IL-24, interleukin-24; ILP,
isolated limb perfusion; iNGR, a type of tumor penetrating peptides, which harbors NGR and CendR motifs; iNGR-SSL, iNGR-modified sterically stabilized liposome;
IP, intraperitoneal; iRGD, a type of tumor penetrating cyclic peptides, which harbors the RGD αv-integrin-binding motif and the RGDK cryptic CendR motif; iRNP,
iRGD nanoparticle; KLAK, cytotoxic peptide; Lac, lactose; LCP-QP NP, lipid/calcium /phosphate nanoparticle; MC, merocyanine; MCSs, multicellular spheroids;
MMPs, matrix metalloproteases; MNC, micelle nanocluster; MPEG, methoxy poly (ethylene glycol); MRI, magnetic resonance imaging; Ms-DDS, multistage drug
delivery systems; m-SR-XRF, m-synchrotron radiation X-ray fluorescence; MTX, methotrexate; NDDs, nano-drug delivery system; NIR, near infrared; NLSC, N-lysinal-
N, -succinyl chitosan; NP, nanoparticle; nRGD, one of the iRGD derivatives; NRP-1, neuropilin-1; ODN, oligodeoxynucleotides; AP, acetylated pullulan; PA, pho-
toacoustic; PAE, poly (β-amino ester); PAEMA, poly (2-azepane ethyl methacrylate; PCL-b-PVP, poly (epsilon-caprolactone)-b-poly (N-vinylpyrrolid one); PCL-CDM-
PAMAM, poly (amidoamine)-graft-polycap rolactone; PDGF, platelet-derived growth factor; pDNA, plasmid DNA; PDNP, prodrug nanoparticle; PDT, photodynamic
therapy; PEG, polyethylene glycol; PEI, polyethyleneimine; PLA, poly lactide; PLGA, poly (D,L-lactide-co-glycolide); PLGLAG, MMP2-sensitive peptide; PNIPAM, poly
(N-isopro pylacrylamide); PPMEMA, poly 2-(penta methyleneimino) ethylmethacrylate; PS, photosensitizer; Pt, platinum; PTT, photothermal therapy; PTX, pacli-
taxel; QDs, quantum dots; RBC, red blood cell; RES, reticular endothelial system; rHuPH20, recombinant human hyaluronidase PH20; ROS, reactive oxygen species;
RRGD, a tandem peptide of RGD; Sf, sorafenib; siRNA, small interfering RNA; SNP, shell-stacked nanoparticle; SP, spiropyran; TAM, tumor-associated macrophage;
TAT, cell penetrating peptide; TGF-β, transforming growth factor beta; TGF-β-I, transforming growth factor-β inhibitor; THNC, “Trojan Horse” nanocarriers; TME,
tumor microenvironment; TNF-α, tumor necrosis factor-α; TPGS, tocopheryl polyethylene glycol succinate; TSL, thermosensitive liposome; UCL, up-conversion
luminescence; UCNPs, up-conversion luminescent nanoparticles; UCPZ, up-conversion nanoparticles-platinum-ZnFe2O4; VEGF, vascular endothelial growth factor;
Wnt16, a member of the wingless type MMTV integration site family

Corresponding author.
E-mail addresses: 1134592416@qq.com (F. Peng), 836840641@qq.com (R. Li), 745707048@qq.com (F. Zhang), 1639625350@qq.com (L. Qin),
ajps2006@163.com (G. Ling), zhangpengspu@163.com (P. Zhang).

https://doi.org/10.1016/j.ejpb.2020.04.009
Received 14 June 2019; Received in revised form 2 March 2020; Accepted 11 April 2020
Available online 18 April 2020
0939-6411/ © 2020 Elsevier B.V. All rights reserved.
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

been widely used to prevent the proliferation, infiltration, and metas- normalize tumor blood vessels, the application of antiangiogenic drugs
tasis of cancer cells until eventually killing the cancer cells. However, is necessary to resist the abnormal angiogenesis.
the low selectivity and poor water solubility of chemical drugs restrict In contrast to the healthy vessels, there are too many leakages in
their further clinical applications [5]. Recently, a myriad of nanosys- tumor vessels, which are caused by large pores in blood vessels. In the
tems have attracted extensive attention for cancer treatment, which are system circulation, nanoscale formulations tend to accumulate in the
achieved by the passive targeting (enhanced permeability and retention leaked areas of the tumor. It is commonly described as “EPR effect”
(EPR) effect) and/or active targeting (surface modification of na- which is magnified by the increased retention of tumor lymphatic
noagents with targeting moieties) principles. There exist three phases in drainage [13]. It is worth mentioning that, it guides nanotherapeutics
drug-loaded nanoparticles delivery: blood circulation, tumor penetra- to enter tumor vasculature more rapidly than other organs including
tion, and interaction with the targeted tumor cells. To maximize the the liver, spleen, and lungs. Although the abnormal tumor vasculature
therapeutical efficiency, nanoparticles (NPs) must spread throughout confers an advantage for the accumulation of NPs, the transshipment
the entire tumor from the peripheral to the central regions. Un- ability of the emerging nanomaterials still poses a significant challenge
fortunately, solid tumors are characterized by abnormal tumor vascu- to diffuse into the surrounding tumor tissues and deep areas.
lature, high density of cells and extracellular matrix (ECM), the ele-
vated tumor interstitial fluid pressure (IFP) and so on [6]. The 2.2. Elevated interstitial fluid pressure (IFP)
components of tumor microenvironment together form a big biological
barrier for nanoparticle deep penetration and subsequently weaken the Besides, factors that hinder the penetration of nanodrugs include the
potency of drugs used in the tumor parenchyma. elevated IFP. The negative IFP, which is required to promote fluids flow
On the other hand, the penetration efficiency of nanodrugs is sig- and transport nutrients from blood vessels into the cells of normal tis-
nificantly affected by the physicochemical properties of nanoparticles sues [14]. In terms of some solid tumors, however, there is a sig-
(size, shape, surface charge as well as ligand conjugation). The nano- nificantly elevated IFP which is resulted from the abnormal perme-
particle intratumoral transport depend on the slow diffusion, so its size ability, along with high cell density and the insufficient lymphatic
is identified as a critical design parameter that impacts their distribu- drainage in the tumor interstitial space [15]. As a result, the transport
tion [7]. It was believed that smaller nanoparticles were delivered to of therapeutic agents is largely discouraged by the unfavorable pressure
the core areas of the tumor mass and showed higher performance on the gradient between the tissues and blood vessels. Even worse, elevated
aspect of intratumoral penetration ability [8]. But too small particles IFP may cause substances in the tumor to flow back into the blood
(their diameters are less than 5 nm) were cleared from the renal, thus capillaries, including NPs, cancer cells and growth factors, thus accel-
shortening blood circulation time and compromising tumor targeting erating tumor growth and metastasis [16]. Recently, antagonists of
efficiency [9]. Therefore, the choice of size is involved with seeking platelet-derived growth factor (PDGF), transforming growth factor beta
equilibrium between the extended blood circulation time in the normal (TGF-β), vascular endothelial growth factor (VEGF) and a multikinase
tissues and the enhanced penetration in tumor tissues. To overcome the inhibitor have been used to decrease IFP and improve the intratumor
above difficulties and improve the delivery efficiency, multistage drug transport of nanomedicines.
delivery systems (Ms-DDSs) which are endowed with switchable size
have been exploited by many researchers to enhance tumor penetra- 2.3. Dense extracellular matrix (ECM)
tion. In other words, these nanoassemblies have appropriate char-
acteristics that ensure the prolonged circulation and accumulation in ECM is mainly comprised of a crosslinked net of collagen, elastin
blood vessels, and convert to smaller particles for better penetration fiber, proteoglycan and hyaluronic acid (HA) [17]. It is one of the
capability in tumors. The stimuli utilized to trigger the size transfor- biological barriers to penetrate malignant cells. In tumors, the collagen
mation of nanoagents can be endogenous factors or external factors. content is obviously higher than that in healthy tissues, which forms
In this review, we firstly elaborate tumor physiological factors and relatively compact extracellular spaces. The dense collagen network
nanomedicine properties which impact tumor penetration. Then, the renders nanodrugs immediately localize in the surrounding vascular
recent strategies to fight against cancers through overcoming tumor areas and severely impairs their mobility [18]. In addition, the charged
penetration barriers are described in detail. Finally, the combination substrates, such as collagen and HA, impede the delivery of positively
therapies and typical clinical therapies are also discussed. charged nanomedicines owing to electrostatic interactions [19–20]. By
degrading or normalizing ECM, some researchers have successfully
2. Tumor physiological factors that impact tumor penetration improved the penetration of nanomedicines.

Compared with normal tissues, tumors usually have unique struc- 2.4. Other factors
tural characteristics. Besides, different cancer tissues vary in biological
characteristics, vasculature and stroma. Some can be reached well by The tumor microenvironment (TME) is also characterized by the
nanomedicines (such as Kaposi sarcoma), others are challenging (such overexpression of cancer-associated fibroblasts (CAFs), tumor-asso-
as pancreatic cancer [10]). The latter particularly require strategies to ciated macrophages (TAMs) and hypoxia. CAFs and TAMs, the major
increase penetration. Factors which hinder the in-depth transport of cell types in tumor stroma, prevent the nanomedicine delivery and lead
nanomedicines include abnormal tumor vasculature; elevated IFP; to restricted response of solid tumors. Hypoxia, a prominent feature of
dense ECM; cancer-associated fibroblasts (CAFs); tumor-associated TME, is ascribed to rapid cell proliferation and abnormality in tumor
macrophages (TAMs) and hypoxia (Fig. 1). blood vessel structure. In addition, inadequate oxygen supply occurs in
the center of the tumor away from the microvessels, hindering the NP
2.1. Abnormal tumor vasculature penetration into deep tumor regions [21].

In the development of healthy tissues and cells, the formation of 3. Nanomedicine properties that impact tumor penetration
blood vessels is carefully regulated to ensure that all cells can get ample
oxygen and nutrients by molecular diffusion [11]. Malignant tumors are 3.1. Size
formed irregularly. They must acquire their own blood supply to
achieve rapid proliferation through angiogenesis. Therefore, the struc- Particle size of nanodrugs is one of the most critical features which
ture of tumor vasculature is poorly-organized, directly leading to the govern their vivo fate, including blood circulation kinetics, tumor ac-
more penetration capacity of tumor tissues than healthy ones [12]. To cumulation and deep penetration. NPs whose size are smaller than

221
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

Fig. 1. Overview of tumor unique structural characteristics.

10 nm are easily excreted by the kidneys, whereas larger particles are 3.4. Targeting moieties
usually captured by reticular endothelial system (RES) during circula-
tion [22]. In a pioneering research conducted by Kataoka and cow- The actively-targeted nanovehicles are a landmark discovery in
orkers, 18 nm small interfering RNA (siRNA) complexes, which abro- cancer therapy. After ligand-modified NPs go through several barriers
gated the need for larger NPs, were engineered for the delivery to which are generated by TME, cell internalization and therapeutical
stroma-rich fibrotic pancreatic cancers and orthotopic brain tumors. efficiency are consequently enhanced by ligand-receptor binding. It
The particles with appreciably small size comparable to antibodies, seems that high ligand affinity is beneficial to tumor penetration.
combined stable systemic circulation with enhanced tumor penetration However, Lee et al. investigated the distribution of 25 nm epidermal
[23]. Furthermore, the similar connection between particle size and growth factor receptor (EGFR)-targeted block copolymer micelles and
tumor penetration had also been validated in a series of NPs e.g. non-targeted micelles in tumors. The penetration depth of non-targeted
polymeric NPs [24], mesoporous organosilica NPs [25], ultrasmall gold nanomedicine was longer than the active targeting delivery systems
NPs (AuNPs) [26], and PAMAM dendrimers [27]. All results jointly [35]. It might be attributed to the “binding site barrier” effect.
indicate that smaller NPs have more potential in deep tumor penetra-
tion. 4. Potential drug delivery nanosystems for improving tumor
penetration

3.2. Surface charge


4.1. Tumor microenvironment programming

Besides particle size, surface charge is also an influential element for


The tumor matrix presents a transport barrier which has significant
improved tumor penetration and tumor therapy. Because the cationic
negative effect on the penetration of macromolecules within tumor
NPs have the tendency to interact with negative-charged matrix poly-
tissues, thereby limiting the anticancer efficiency of nanoscale delivery
mers such as collagen and proteoglycans, cationic materials penetrate
systems. As mentioned above, the tumor extracellular matrix includes
more deeply than the anionic counterparts [28]. It was demonstrated
abnormal vascular structures, elevated IFP, tight ECM, CAFs, TAMs, and
that positively charged poly (D, L-lactide-co-glycolide) (PLGA) NPs
hypoxia. It has been demonstrated that the method of regulating the
achieved better tumor penetration than the negative ones (-13 mV) in
TME can be a feasible approach to improve the permeability.
the xenografts [29]. However, the cationic ones were prone to be op-
sonized and cleared by RES during blood circulation. Compared with
4.1.1. Collagenase
the positive or neutral NPs, the negative ones were more effective on
Nanomedicine may be entrapped in complex collagen networks
the aspect of penetration in the liposomes [30] and gold nanorods
which severely restrict deep penetration. The ECM barriers could be
(AuNRs) [31]. In short, the relationship between surface charge and
overcome by collagenase-linked super paramagnetic NPs. There was a
nanomedicine penetration is too complex to elaborate fully.
more efficient localization in collagenase- modified NPs in deep tumor
region than non-modified NPs [36]. Goodman et al. systematically as-
3.3. Shape sessed the penetration depth of collagenase-coated carboxylate poly-
styrene NPs. The number of collagenase-pretreated nanodrugs which
Particle shape is also a crucial parameter that influence blood cir- were delivered to the spheroid core was four times higher than that of
culation and tumor penetration. More and more attention has been paid nanoagents with collagenase non-immobilized [37]. In another study,
to nonspherical vehicles, which penetrate deeper than spherical mate- the pretreatment, which was based on the collagenase liposomes, dis-
rials [32]. In the latest study, the cancer cell membrane (CCM)-coated assembled the collagen stroma and enhanced tissue permeability into
NRs displayed more rapid extracellular matrix penetration than sphe- pancreatic tumors [38]. That is, the NP delivery is significantly pro-
rical particles and shipped doxorubicin (DOX) to the nucleus [33]. The moted through the site-specific degradation of ECM protein.
unique yolk-shell CCM-camouflaged NPs frequently transformed into
ellipsoidal shape particles, facilitating the penetration throughout 4.1.2. Hyaluronidase (HAase)
multicellular spheroids and yielding 95% tumor-inhibition rate [34]. HA (Fig. 2), one of the major ECM elements, binds with collagen to
Taken together, rational shape design is essential to improve antitumor maintain the integrity of extracellular tumor matrix. The content of HA
therapy. in tumor is much higher than that in the normal tissues. This

222
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

breast tumors [44]. In the meanwhile, the interstitial transport of


plasmid DNA (pDNA) was deeper after recombinant relaxin treatment
in 4 T1 tumor cell lines. These observations were correlated with the
results from pulsed electric field experiment, enhancing electric field-
mediated gene delivery [45]. Inducement of collagen remolding with
relaxin treatment reduce ECM protein level in collagen-rich tumors and
create more pores for nanomaterials to deeply penetrate tumor stroma.

4.1.5. Losartan
High density of collagen I network prevents the deep tumor pene-
tration of nanomaterials, leading to the impaired anti-tumor efficiency.
Losartan was recognized by Food and Drug Administration (FDA) as
angiotensin II receptor antagonist with notable antifibrotic activity for
hypertension in clinic. It could enhance the penetration and therapeutic
efficiency of nanomedicines [46]. The much better penetration ability
was acquired in the application of losartan-pretreated NP in HSTS26T
and Mu89 mice models, which was explained by the losartan-induced
collagen fiber disruption. Furthermore, the similar efficiency of losartan
had been also revealed in a pioneer study which employed pegylated
liposomal DOX as the drug carrier [47]. In addition, free losartan was
injected before the treatment to decrease the content of collagen.
Afterwards, the pH-responsible liposome was injected to kill cancer
cells. Tumor distribution and target site accumulation were con-
siderably improved by losartan pretreatment [48]. The recent animal
experiment had also clarified the enhanced tumor penetration ability of
losartan-loaded NPs in synergistic of chemotherapy/photo thermal
therapy (PTT) [49].

4.1.6. Regulation of CAFs, TAMs and hypoxia (Table 1)


Fig. 2. Chemical structure of hyaluronic acid (HA). Based on the abundant CAFs, TAMs in tumors, CAF-targeted lipo-
somes and carboxymethylcellulose NPs were exploited to specifically
eliminate tumor stroma to promote tumor penetration [50–51]. TME
phenomenon makes the efficient penetration of NPs difficult. HAase-
was reprogrammed to favor the delivery of the therapeutic NPs in
modified micelles, which were loaded with photosensitizer chlorine e6
desmoplastic cancers [52–53]. The enhanced cancer immunotherapy
(Ce6), were prepared by Liu et al. to facilitate tumor penetration. It was
was also achieved by the successful TAMs repolarization [54–55]. In
found that the conjugation of HAase resulted in a remarkable increase
addition, the remission of the hypoxic environment had been confirmed
in perfusion and tumor oxygenation, thus greatly improving the effi-
to achieve deep penetration in the combination of multiple treatments
ciency of photodynamic therapy (PDT) [39]. Moreover, in order to
[56–57].
prolong nanocarrier blood circulation, a relatively low-density poly-
ethylene glycol (PEG) layer was covered in the DOX-loaded PEG-PLGA
4.1.7. Growth inhibitors
NPs. The surface was conjugated with recombinant human hyalur-
In normal tissues, angiogenic activators and inhibitors are in dy-
onidase PH20 (rHuPH20) for deep tumor penetration in aggressive 4 T1
namic equilibrium to ensure blood vasculature growth. However, the
tumors [40]. In addition, HAase also promoted the development of
balance is disrupted by the abnormal regulation of growth factors in
nanodrug carriers in convection-enhanced delivery (CED). The tumor
pathological circumstances like tumors.
distribution volume of 54 nm monodisperse polystyrene particles was
increased by 64%, which was higher than the control ones without
4.1.7.1. Transforming growth factor beta (TGF-β)
HAase pre-infusion [41].
inhibitors. Transforming growth factor beta (TGF-β) plays a critical
role during cancer progression regulation. In the later stage of tumor
4.1.3. Bromelain growth, it acts as a promoter for tumor metastasis. The employment of
Bromelain, a crude enzymatic complex, is one of papain peptidase short-acting, small molecule TGF-β inhibitors decreased pericyte
family and purified from pineapple stalks [42]. Bromelain-conjugated coverage of vessels and fibrotic components in the neovasculature of
mesoporous silica nanoparticle (Br-MSN) was synthesized to increase pancreatic tumors, thus promoting the accumulation of 65 nm micelles
nanomedicines uptake in endothelial cells, macrophages and cancer and 90 nm Doxil liposomes [58]. Hence, the introduction of TGF-β
cells. Since bromelain was able to degrade tumor ECM components, Br- inhibitors was significant to treat intractable solid cancers including
MSN showed much faster diffusion and digest ability in vitro as well as pancreatic adenocarcinoma and diffuse-type gastric cancer. Besides, the
in vivo. The bromelain pretreatment extended penetration depth in damaged extravasation of 70 nm 1,2-diamino cyclohexane-platinum
matrigel by twofold (from 500 to 1000 μm), thus improving tumor (II) (DACHPt) micelles was overcome through administering TGF-β
distribution after injection in 4 T1 tumors [43]. inhibitors (TGF-β-I) in BxPC3 tumors. Fluorescence microscopic
assessment of BxPC3 tumor pieces confirmed that the accumulation
4.1.4. Relaxin of 70 nm micelles raised to a comparable level with that of the 30 nm
In previous reviews, relaxin, a peptide hormone of insulin family, countparts. The result was consistent with the m-synchrotron radiation
had been validated to inhibit collagen production and existing collagen X-ray fluorescence (m-SR-XRF) measurement, which demonstrated the
overexpression. Therapeutic potential of relaxin was tested through facilitated intracellular delivery and the enhanced tumoricidal activity
hematopoietic stem cell (HSC)-based approaches. As a result, the pep- of DACHPt micelles by TGF-β-I [59]. In a study of Huang et al., upon
tide hormone relaxin expression decreased collagen amount in the ex- the down regulation of the Wnt16 (a member of the wingless type
tracellular matrix to improve the trastuzumab therapy outcome in MMTV integration site family) levels by quercetin, targeted lipid/

223
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

calcium/phosphate NPs (LCP-QP NPs) were designed to remodel the 4.1.8. Soy protein
TME to promote the penetration of second-wave NPs into TGF-β In addition, different size soy protein NPs could also normalize TME
activated NIH3T3 cells [60]. Specifically, the NIH3T3 cells were pre- such as the elevated IFP and higher tumor stress, improving tumor
treated with 10 μM cisplatin and then activated by 10 ng/mL TGF-β. penetration and impeding tumor growth in living mice. Among dif-
The treatment of 10 μM quercetin was confirmed to downregulate ferent size NPs, DOX-loaded soy protein NPs with the size of 30 nm
Wnt16 level and then inhibit the effect of TGF-β. This strategy exhibited the best cytotoxicity, the best TME modulation efficiency as
significantly improved the penetration of cisplatin NPs. well as the deepest penetration depth [73].

4.1.9. Tumor-penetrating peptides


4.1.7.2. Tumor necrosis factor-α (TNF-α) and derivatives. Tumor necrosis
4.1.9.1. iRGD and nRGD. Tumor-specific tissue penetration systems,
factor-α (TNF-α) played a key role in apoptosis, cell survival and
which were based on iRGD peptide, were discovered to reverse the
inflammation. The extravasation of erythrocytes and lymphocytes led
ineffective therapy and drug resistance [74]. iRGD, a type of tumor
to hemorrhagic necrosis in tumors during isolated limb perfusion (ILP)
penetrating cyclic peptides, harbored the RGD αv-integrin-binding
cancer treatment [61]. In addition, TNF-α had potential in decreasing
motif and the RGDK cryptic CendR motif. After being cleaved by cell
IFP and modulating ECM for better transport [62]. Administration of
surface associated proteases, the CendR motif mediated binding to
TNF-α augmented Doxil regimen rendered the liposomal drugs enter
neuropilin-1 receptor to achieve efficient tumor penetration [75]. The
tumor interstitium, explaining the improved anticancer efficiency [63].
combination of active and passive processes extravasated more
Furthermore, the emergence of TNF derivatives had been already
efficiently and facilitated the intracellular delivery of cytotoxic
proved to avoid TNF toxicity and replace TNF for the clinical
payloads. Poly (epsilon-caprolactone)-b-poly (N-vinylpyrrolidone)
application. Vascular targeting with low concentrations of NGR-TNF,
(PCL-b- PVP) NPs were modified with iRGD and exhibited superior
a combination of TNF derivative and CNGRC peptide, was investigated
tumor penetration in H22 tumor-bearing mice. However, the mon-
[64]. It enhanced the tumor penetration of chemical drugs without
targeted nanomedicines distributed primarily at the periphery of tumor
obvious toxicity.
vessels at 24 h after injection [76]. In addition, the penetration of dual
targeting polymeric NPs, which were functionalized with iRGD and
4.1.7.3. Platelet-derived growth factor (PDGF) antagonists. Platelet- interleukin-13 peptide, was obviously better than that of interleukin-13
derived growth factor (PDGF) receptors are highly expressed in many single-modified NPs in C6 cells. It clearly showed that iRGD NP (iRNP)
tumors, increasing IFP in dermis after anaphylaxis-induced IFP targeted both tumor neovasculature and glioblastoma multiforme
lowering. One of the most vital reasons leading to elevated IFP and (GBM) cells through receptor labeling in vitro and in vivo. Similar
complex collagen network is the existence of myofibroblasts. PDGF iRGD penetration promotion phenomenon had been also reported in the
antagonists decreased the contractile function of new-formed study of iRGD-decorated prodrug NP self-aggregated hydrogel (PDNP-
myofibroblasts in tumor tissues to loosen the ECM [65]. When serine gel) [77] and interleukin-24 (IL-24)-iRGD recombinant protein [78]. In
protease fibroblast activation protein (FAP) on the cell surface was another study, nRGD, which covalently conjugated the alanine-alanine-
specifically overexpressed on tumor fibroblasts, the genetic deletion asparagine “tail” residual with iRGD peptide, was described to enhance
and pharmacologic inhibition of FAP were investigated to control tumoricidal effect of DOX. The nRGD not only retained tumor
myofibroblasts content in tumor ECM. Besides, tumor-associated penetration effect of iRGD but also modulated TME with the
fibroblast, the major source of collagen type I, contributed to the depletion of TAMs. Compared with PEG-modified DOX liposomes and
decreased uptake of nanodrugs. Oral DNA vaccines targeting FAP were nRGD physical mixtures, the nRGD grafted DOX-loaded liposomes
constructed to kill tumor-involved fibroblasts. The tumor growth was showed the dramatic improvements in antitumor efficiency [79].
markedly suppressed through the decrease of collagen type I content, Apart from promoting tumor penetration, the high affinity between
opening a new venue in penetration-promoting strategies which RGD and αv integrin also increased the cell uptake of NPs, which
combined immunotherapy with chemotherapy in the future [66]. broadened RGD application in the delivery of biotherapeutics.

4.1.9.2. iNGR. The tumor-penetrating peptide iNGR included three


4.1.7.4. Vascular endothelial growth factor (VEGF) receptor
independent sections: a vascular homing motif, a R/KXXR/K tissue
inhibitors. Vascular endothelial growth factor (VEGF), one of the most
penetration part and a protease recognition site. It provided a solution
vital angiogenic molecules, is involved with binding to tyrosine kinase
to the poor tumor penetration. It had the dual targeting capacity to
receptors and results in cell proliferation as well as new blood vessel
tumor vessels based on CD13 receptors (the receptor of iNGR) and to
formation [67]. Therefore, anti-angiogenic therapies which normalize
tumor cells based on the neuropilin-1 (NRP-1) receptors [80]. Here,
abnormal tumor vessels can improve the delivery of nanomedicines by
iNGR-modified PEG-PLGA NPs were expected to overcome the leading-
making them less leaky. The method of blocking anti-VEGF- receptor-2
edge difficulty in anti-glioma nanodrug delivery. The acquired iNGR-
antibody (DC101) had been employed to repair the abnormal
NPs were observed to achieve excellent penetration at the glioma sites
vasculature in mammary tumors, allowing smaller NPs,
by initially binding to amino peptidase N, with iNGR hydrolyzed and
macromolecular protein and bovine serum albumin (BSA) to enter
cleaved to CRNGR, and then binding to NRP-1 for the deep penetration.
tumors more rapidly. When the size of pores in the vessels were reduced
Convincingly, compared with cNGR-NP, paclitaxel (PTX)-loaded iNGR-
by the normalization, IFP gradually dropped in the tumor mathematical
NPs exhibited the increased uptake of human umbilical vein endothelial
model. The limited penetration of larger NPs (125 nm) was attributed
cells, thus inducing the strengthened anti-proliferation and anti-tube
to the increased steric and hydrodynamic hindrance [68]. At present,
formation efficiency with IC50 of 16 ng/mL [81]. Currently, iNGR-
anti-VEGF antibodies, such as bevacizumab [69,70], have been used in
functionalized sterically stabilized liposome (iNGR-SSL) was also
the treatment of non-small-cell lung cancer brain metastases,
proved to promote the penetration across the tumor vessel wall, the
glioblastomas and triple negative breast cancers.
GBM tissue and further into tumor cells. From the in vivo imaging and
the immunofluorescence staining analysis, the uptake and cytotoxicity
4.1.7.5. A multikinase inhibitor. Sorafenib (Sf), a multikinase inhibitor, of DOX-loaded iNGR-SSL in HUVEC and U87MG cells were
blocked VEGF receptors and PDGF receptors simultaneously for comparatively higher than that of unmodified SSL/DOX. It was
advanced hepatocellular carcinoma (HCC) therapy [71]. Sf and VEGF- attributed to the increased tumor accumulation and penetration
siRNA co-loaded liposomes were also constructed for the synergistic mediated by iNGR [82]. In summary, the iNGR modification was an
treatment of HCC [72]. effective strategy to improve the tumor curative effect and increase the

224
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

survival time of GBM animals. acoustic pressure increased from 1.7 to 6.9 MPa, the penetration depth
of micro-RNA loaded NPs increased by 2-fold into human colon cancer
xenografts [89]. To our knowledge, ultrasound-induced microvesicle
4.1.10. Sonoporation with microbubbles
cavitation in Phase I clinical trial was conducted to evaluate the safety
Microbubbles are usually filled with hydrophobic gas, which is
against pancreatic cancers. No additional toxicity and prolonged sur-
coated with 10–100 nm layer made of polymers, proteins, and lipids.
vival time were observed. The survival period of patient cohorts ex-
Ultrasound causes microbubble cavitation, i.e., periodically shrink and
tended from 8.9 to 17.6 months, paving the way towards further clin-
expand. It will lead to the formation of temporary pores in the cellular
ical researches [90].
membrane, increasing permeabilization of surrounding cells to drug
molecules. This process is called sonoporation [83]. The existing re-
search papers verified that ultrasound-activated microbubbles in- 4.1.11. Hyperthermia (HT)
creased the penetration of antitumor drugs for optimal nanoagents Hyperthermia (HT) with thermosensitive delivery systems had been
delivery. The combination of DOX and Vevo Micromarker microbubbles applied to enhance tumor penetration. After mild HT, we could easily
had been evaluated to increase cancer cell death ratio. The improve- observe the larger gaps in the endothelial lining of 10 µm and deeper
ment was attributed to the deeper penetration of DOX into human U- penetration into the interstitial space to at least 27.5 µm in radius from
87MG glioblastoma and MDA-MB-231 breast cancer cells. Preliminary the vessel walls [91]. For the more efficacious drug delivery, the
data in vitro showed the therapeutical potential in vivo studies [84]. In coadministration of DOX thermosensitive liposome (DOX-TSL) and
the presence of approximately 25 × 106 microbubbles per ml, pulsed local HT were characterized to initiate the improvement of tumor
ultrasound exposure enhanced the penetration into the breast cancer vasculature permeability and TSL extravasation [92]. The effect of DOX
spheroids (300–500 µm diameter) [85]. Therefore, particles with dif- formulation and mild HT were also investigated to enhance penetration
ferent electrical properties and different particle sizes all achieved into 2D and 3D cell culture model among multiple human ovarian
higher penetration. The tumor penetration efficiency, which was pro- cancer cells [93]. Novel temperature-sensitive liposomes were designed
moted by the above microbubbles, was also observed in sonicated tu- to effectively deliver DOX to tumor nuclei. In vitro results demonstrated
mors to hinder growth [86,87]. And recently, synergistic therapy with that the local release of DOX from heat-responsive liposomes was ac-
sonoporation effect was developed by DOX-loaded human serum al- celerated under high intensity focused ultrasound (HIFU). After HIFU-
bumin (HSA) NPs and Ce6 encapsulated microbubbles (DOX-NPs/Ce6- stimulated heat treatment, anticancer drugs rapidly penetrated into
MBs) complex nanosystem (Fig. 3). Through the employment of ultra- tumor vascular cells and overcome the barriers of limited penetration.
sound, the excellent co-delivery nanocarriers penetrated deeper into More efficacious anti-tumor effect in vivo was verified by the 65.2%
cancerous tumors, exhibiting higher delivery efficiency [88]. As the tumor inhibition rate in the groups of HIFU-treated mice [94].

Fig. 3. A schematic of efficient delivery of drug and photosensitizer through sonoporation effect. Reprinted with permission from [88].

225
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

Fig. 4. Chemical structure of acrylate poly (β-amino ester).

Numerical modeling of intraperitoneal (IP) chemotherapy was estab- agents in the treatment of tumors. Similar designs were also found in
lished to examine the impact of HIFU frequency on the penetration the study of Sha et al. Size-switching micelle nanocluster (MNC), which
depth of DOX in thermosensitive liposomes. An improvement in drug was based on cross-linked and pH-sensitive frameworks between
penetration depth was observed by changing the HIFU frequency from polyethyleneimine (PEI) and tocopheryl polyethylene glycol succinate
0.5 to 1.0 MHz [95]. The phase I study of ultrasound-mediated HT of (TPGS), was validated to enhance tumor targeting and penetration
lyso-thermosensitive liposomal DOX was perfectly carried out in adult [101]. Moreover, owing to the acid-mediated charge-reversal and
oncology patients with incurable liver tumors [96]. dissociation, the versatile hollow mesoporous silica NPs (HMSNs)
were found to sequentially overcome multiple drug gemcitabine
4.2. Multistage drug delivery platforms (GEM) deliver barriers for the enhanced penetration and the
programmed tumor therapy. The enhanced tumor therapy was also
One of successful methods for enhancing drug uptake and pene- verified in three-dimensional multicellular spheroids and A549 tumor
tration is the application of multistage drug delivery platforms. Even xenografted mouse model. The well-designed pH-sensitive multistage
though NPs with larger size were prone to achieve superior accumu- polymeric micelleplexes maintained structural stability at physiological
lation which was gained by EPR effect [97], their penetration had been pH (pH 7.4). Once reached to tumor sites, they presented the triggered
also hampered by the physiological barriers in the TME. Even though release of small PAMAM-Pt dendrimers due to acid-labile characteristic
NPs with smaller size improved tumor penetration and therapeutic ef- of the carboxylic amides of CS component (dimethylmaleic anhydride,
ficacy, NPs whose size was less than 5 nm were rapidly cleared by DMA), following by the release of cisplatin in intracellular reducing
kidney filtration, which resulted in the ineffective killing of cancer cells. environment for better chemotherapy efficacy [102].
Recently, multistage drug delivery platforms, which could retain com-
paratively large size to ensure the prolonged blood circulation and 4.2.1.2. The ionization of chemical groups in low pH. The ultrasensitive
switched to smaller particle to promote extravasation, had been envi- size-tuning systems containing tertiary amine groups had been further
sioned as a promising strategy to meet the contradictory requirements constructed to surmount multiple obstacles in drug delivery. Poly (β-
on particle size. The stimuli utilized to trigger the size alternation of amino ester) (PAE) (Fig. 4) was a kind of pH-responsive biodegradable
NPs could be low pH, overexpression of enzymes, glutathione (GSH), polymers. It was insoluble at higher pH (pH 7.4) and dissolved at a
light and chemical reactions. lower pH due to the protonation of tertiary amino groups. Wang and
colleagues reported a variety of amphiphilic pH-responsive NP
4.2.1. Low pH structures including ionizable tertiary amine groups which rapidly
The pH value of normal tissues and blood (~7.4) was higher than responded to different pH. At neutral pH, these NPs were prepared by
that of TME (~6.5) [98]. The lower pH could be used to trigger the PEG-b-poly (2-azepane ethyl methacrylate)-modified PAMAM
degradation of acid-cleavable linkers [99–103] or ionization of che- dendrimers (PEG-b-PAEMA-PAMAM) and had an original size of
mical groups for better penetration. 80 nm for prolonged circulation. When they were transferred to the
acidic TME, the polarity of PAEMA changed from hydrophobic to
4.2.1.1. The degradation of acid-cleavable linkers in low pH. In a pioneer hydrophilic, resulting in the instantaneous disintegration into the
research, stimuli-responsive size-switching clustered NPs were prepared dendrimer building blocks and the effective tumor penetration. Both
through molecular self-assembly of platinum (Pt) prodrug-conjugated in vitro and in vivo results showed that pH-triggered size switching was
poly (amidoamine)-graft-poly caprolactone (PCL-CDM-PAMAM/Pt), a viable method to improve drug penetration in the poorly permeable
PCL and PEG-b -PCL. After accumulating at tumor tissues with lower BxPC-3 pancreatic tumors [10]. Moreover, intelligent size/charge-
pH, 5 nm PAMAM/Pt was discharged from the smart iCluster with changing micellar systems were developed for enhanced drug
approximately 100 nm via a pH sensitive degradable linker. It also delivery. After extravasation from the leakage of tumor vasculatures,
demonstrated that iCluster significantly improved tumor penetration methoxy poly ethylene glycol poly lactide (MPEG-PLA)-PAE micelles
and delivered threefold drugs to tumor cells [99]. Kim and colleagues penetrated deeper than MPEG-PLA copolymer micelles. That was
functionalized the surface of gold NPs (~13 nm) with Bcl-2 antisense attributed to the protonation of PAE chains in the mildly acidic TME
oligodeoxynucleotides (ODNs). Clustering of AuNPs (~150 nm), which (Fig. 5). Thus, the occurrence of protonation phenomenon caused size
were motivated by the hybridization of i-motif sequence, provided and charge conversions to increase tumor cellular uptake of curcumin
better tumor targeting efficiency and drug loading capability. Results (CUR) and improve cytoplasmic distribution in human breast cancer
showed that apoptosis was accelerated by the disassembly of clustered MCF-7 cells [104]. What’s more, the docetaxel (DTX)-loaded MPEG-
AuNPs into single NPs in response to the lower pH [100]. In addition, PLA-PAE polymeric micelles were manipulated to illustrate the effect of
the down-regulation of Bcl-2 proteins was observed in the antisense- particle size. When the pH reduced to 6.5 through the addition of
grafted size-tunable NPs, which provided multifunctional therapeutic hydrochloric acid (0.1 M), the smaller particles (27.99 nm) percentage

226
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

Fig. 5. A schematic diagram to show the size/charge transformation of MPEG-P LA-PAE micelles in response to the tumor acidic microenvironment. Reprinted with
permission from [104].

increased to 73%, enhancing permeability on 3D KB MCTS model and comprised of N-lysinal-N, -succinyl chitosan (NLSC) with protonatable
showing size-dependent antitumor effect against KB tumors. Taken side groups and poly (N-isopropylacrylamide) (PNIPAM) with BSA
together, DTX-pH polymeric micelles (DTX-pHPMs) showed the best shells. At the pH of 4.5, the protonation caused swelling to 2 µm.
antitumor efficiency with the highest tumor inhibition rate of 84.4% in Afterwards, the swollen nanogels caused endo-lysosomal bursting and
oral epidermoid carcinomas [105]. Such size and charge dual-shifting drug release. Subsequent shrinking at higher pH enabled transfer to
NPs were also observed in the shell-stacked NPs (SNPs), which neighboring tumor cells [109]. These results showed the repeated
underwent substantial particle size reduction to 40 nm and surface swelling-shrinking process ensured the deeper penetration of nanome-
charge reversal to 8.2 mV for efficient penetration. More importantly, dicines in tumors.
DOX-loaded NPs penetrated deeper than the nontransformable ones,
showing significant anticancer efficiency and eradicating the
4.2.2. Overexpression of enzymes
xenografted A549 lung carcinoma cells in mice [106]. Similarly,
Generally, the overexpression of enzymes occurs in a variety of
deeper penetration and higher tumor suppression rates were obtained
tumors. This phenomenon has been applied to realize size tuning of NPs
by the disassociation of long-circulating polymer-prodrug hybrid NPs.
to improve tumor penetration. The enzymes used to trigger the event
The protonation of poly (2-(pentamethyleneimino) ethyl methacrylate)
can be matrix metalloproteases (MMPs) and hyaluronidase (HAase).
(PPMEMA) core at pH 6.8 induced the disassociation of long-circulating
polymer-prodrug hybrid NPs to small-size siRNA composites. The
improved tumor penetration was confirmed by the 5-fold higher cell 4.2.2.1. Overexpression of matrix metalloproteases (MMPs)
uptake rate at pH 6.8 than pH 7.4 [107]. Penetration-promoting 4.2.2.1.1. Gelatin nanoparticles based on the overexpression of
phenomenon had been also reported in the study of Wang et al. MMPs. Being a substrate of MMP-2, gelatin can be degraded by
When NPs were delivered to the tumor tissues, the inner poly (2- MMP-2 and has been employed to construct size tunable NPs. For
azepane ethyl methacrylate) (PAEMA) segments converted from example, AuNPs coated gelatin nanoparticles (GNPs) were prepared as
hydrophobic to hydrophilic. The polar transformation of PAEMA was drug delivery systems. The size reduction from 186.5 nm to 59.3 nm in
attributed to the acidic extracellular TME, thereby shrinking micellar 24 h was observed owing to the presence of MMP-2 (Fig. 6). Therefore,
size for the deeper penetration [108]. the obtained G-AuNPs-DOX-PEG with initial larger size possessed better
Another strategy for deep penetration in response to pH variation transvascular and interstitial penetrating effectiveness, showing
was developed by reversibly switchable size nanogels, which were admirable antitumor effects in 4 T1 and B16F10 tumors [110]. In
addition, quantum dots (QDs) were assembled onto the surface of GNPs.

227
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

Fig. 6. A) Schematic design procedure of G-AuNPs-DOX-PEG. B) Intracellular delivery process of G-AuNPs-DOX-PEG. Reprinted with permission from [110].

Hybrid nanotherapeutics with a larger size of 100 nm were verified to 4.2.2.2. Overexpression of hyaluronidase (HAase). Strong cationic
exhibit the increased accumulation in the leaky areas of tumor biomaterials possessed several undesirable properties including the
vasculature due to EPR effect. The shrunken NPs with small size of aggregation of erythrocytes and platelets. Hence, Na et al. designed
10 nm could more readily diffuse throughout the tumor interstitial biodegradable cationic PTX-loaded nanogels (DpNGs), which were
regions owing to the gelatin degradation by MMP-2. In vivo study, the composed of acetylated pullulan (AP) and low molecular weight PEI
penetration depth of multistage system reached 300 μm from the (1.8 kDa). The surface of DpNGs was modified by anionic HA. The HA/
injection site, displaying the enhanced tumor extravasation and DpNG-PTX nanoagents exhibited significant cytotoxicity in
homogenous distribution in HT-1080 tumors [111]. heterogeneous cancer cells via HA degradation by HAase (Fig. 7).
Besides, by conjugating the gelatin with dendrigraft poly-lysine Correspondingly, the IC50 value was 100 times less than free drug and
(DGL) and anchoring with angiopep-2 targeting, Gao and co-workers the invasive distance of HA/DpNG-PTX increased by 2 times than that
had fabricated dendrimer-GNPs. After degraded by MMP-2, the particle of PA-PTX, enhancing the limited tumor penetration [121].
size of DOX-DGL-GNPs shrank to 50 nm, showing much wider dis- Additionally, Qi laboratory constructed bio-stimuli-sensitive
tribution of as far as 130 μm from the edge [112]. Moreover, the cell multistage methotrexate (MTX)-loaded HA NPs which were modified
penetrating peptide TAT and cytotoxic peptide KLAK decorated den- with PAMAM dendrimers. The initial HA coated PAMAM NPs showed
drimers were synthesized to induce the entrance of nanodrugs into low cytotoxicity and prolonged systematic circulation. However, they
tumor cells through both membrane and endocytosis routes [113]. were easier to be degraded by HAase to release inner PAMAM
MMP2-over expressed TME caused the deprotection of PEG chains, dendrimers for deep drug penetration. As expected, the fluorescein
leading to the obvious decrease of dendrimers diameter and exposure of isothiocyanate (FITC) labeled HA/PAMAM NPs penetrated deeply into
KLAK and TAT. It not only improved solid tumor penetrability but also A549 3D tumor spheroids [122].
disrupted mitochondria effectively through the internalization of In addition, photothermal therapy (PTT) can kill cancer cells by the
bioactive peptides into human primary glioblastoma (U87) tumors. heat generated by photothermal formulations under the irradiation of
Further, based on the targeting of lactose [114], through combining near infrared (NIR) light [123,124]. In recent years, many scientists
GNPs with small molecule prodrug DOX-Lactose (DOX-Lac), optimized combined PTT with chemotherapy and gained better achievements. For
drug delivery systems (GNPs-DOX-Lac) were developed for enhanced example, HAase-motivated size shrinking occurred at the HAase-rich
HCC penetration to overcome a class of pathophysiological barriers TME in the intelligent NPs, which were decorated with NIR laser re-
[115]. sponsive nitric oxide donor (HN), small size dendrimer prodrug (IDD).
4.2.2.1.2. MMPs sensitive bond-linked nanoparticles based on the DOX and indocyanine green (ICG) acted as chemotherapy agent and
overexpression of MMPs. The size-switchable systems could be also photothermal agent respectively (Fig. 8). The novel NPs not only in-
constructed by using MMPs sensitive linker to attach smaller particles duced strong HT effect for PTT but also promoted the NO release to
onto larger size particles. Herein, MMP-2/pH dual-responsive NPs trigger deep penetration [125]. Moreover, based on the alternation of
(DGL/DOX@NPs) were established to break enzyme-sensitive bonds the percentage of HA and AuNC@CBSA (small, kidney-clearable, red-
in the TME and rapidly release DOX. The process of particle size emitting, cationic bovine serum albumin-protected gold nanoclusters),
alteration rendered the systems take advantage of the enhanced Gao et al. screened out and evaluated a well-chosen size of 200 nm NPs.
permeability into both multicellular spheroids (MCSs)and solid The size-reducible nanoplatform performed excellent combinational
tumors [116]. Analogously, comparing MMP2-sensitive peptide therapy by co-delivering PTX and ICG. It was worth mentioning that HA
(PLGLAG) conjugated macromolecules (HA-pep-PAMAM) with MMP2- could serve both as a shielding agent and a ligand of differentiation 44
insensitive macromolecules (HA-PAMAM), the former could penetrate (CD44). In the study, HA shielded the NPs with tumor-specific cluster of
tumor tissues due to the degradation by overexpressed MMP-2 in CD44 targeting and could be degraded by HAase to reduce size, facil-
multicellular spheroids [117]. PLGA-PEG NPs were functionalized itating the penetration effect [126]. HAase-triggered size reduction and
with MMP-2 sensitive linker AGFSGPLGMWSAGSFG. These NPs were deeper penetration were found in the as-prepared red blood cell (RBC)
followed by the modification into porous silicon disk to prepare membrane-coated biomimetic NPs. HAase specifically hydrolyzed the
enzyme-stimulated multistage vector (ESMSV). After intravenously HA layers, followed by the exposure of small size gold nanoclusters
injected to A375 melanoma lung metastasis bearing mice, more (AuNCs) for antitumor combination therapy [127].
polymer NPs were released from the ESMSV and 64.6% of the Potential drug delivery nanosystems, which are based on the effect
melanoma cells were polymer NPs positive [118]. As a result, the of pH and enzymes, are listed in Table 2.
enzyme-sensitive size reduction was a reliable strategy to enhance
tumor penetration [119–120].
4.2.3. High-concentration glutathione (GSH)
Photodynamic therapy (PDT) had been tremendously explored for

228
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

Fig. 7. Elucidation of drug delivery procedure of HA/DpNG-PTX. Reprinted with permission from [121].

the treatment of cancer, which was achieved by gathering the photo- was attributed to the special requirements of tumor abnormal growth.
sensitizer (PS) to mediate the incomplete reduction of oxygen upon the Herein, multifunctional nanocomposites had been reported with GSH-
laser light irradiation. High level of reactive oxygen species (ROS) was mediated size-tunable characteristic for bioimaging guided synergetic
generated to induce potent vascular damage and direct tissue destruc- therapy, including chemotherapy, PDT and Fenton reaction. The initial
tion [128]. In normal tissues and cells, the interconversion of glu- size of PEG-decorated up-conversion NPs (UCNPs)-Pt (IV)-ZnFe2O4 was
tathione (GSH) and glutathione disulfide (GSSG) ensured cellular redox around 100 nm, which was beneficial for tumor retention. The Pt (IV)
homeostasis, which was necessary for cell growth [129]. However, the prodrug, which linked UCNPs with ZnFe2O4, was broken owing to
GSH concentration range was from 1 × 10-3 to 15 × 10-3 mol/L in higher GSH in tumor cells. Meanwhile, the nanocomposites were di-
tumor cells, which was significantly higher than normal tissues [130]. It vided into UCNPs (about 25 nm), ZnFe2O4 (about 7 nm) and high

Fig. 8. A) Schematic design procedure of IDDHN. B) Schematic illustration of the synergistic treatments for deep tumor penetration and therapy outcome. Reprinted
with permission from [125].

229
Table 1
Potential drug delivery nanosystems through regulating CAFs and TAMs and hypoxia.
Factor Mechanism Nanocarrier Active substance Type of tumor Therapy Ref
F. Peng, et al.

CAFs Depletion Liposomal navitoclax DOX Hep G2 Chemotherapy [50]


CAFs Depletion Carboxymethylcellulose NPs DTX PAN 02, OCIP 19, OCIP 23 Chemotherapy [51]
CAFs Reprogram Lipid-coated protamine DNA complexes sTRAIL plasmids UMUC3, NIH3T3 Chemotherapy [52]
CAFs The sensitiveness to FAP-α protein overexpressed Cationic PAMAM dendrimers cross-linked by Asp-Ala-Thr-Gly- Pro-Ala DOX PC-3, CAF Chemotherapy [53]
on CAFs peptide
TAMs Reprogram β-cyclodextrin nanoparticles Resiquimod MC38 Immunotherapy [54]
TAMs Reprogram HA-poly(ethylenimine) (HA-PEI) NPs MicroRNA-125b KRASG12D, P53GEM Immunotherapy [55]
Hypoxia Produce oxygen and relieve hypoxia Albumin–MnO2 NPs Ce6 4 T1 Chemotherapy in conjugation with PDT [56]
Hypoxia Produce oxygen and relieve hypoxia Mesoporous silica nanoparticles Catalase MDA-MB231 Chemotherapy in conjugation with [57]
ultrasound

230
Table 2
Potential drug delivery nanosystems based on the effect of pH and enzymes.
Strategy Carriers Active substance Type of Therapy Ref
tumor

The degradation of acid-cleavable linkers in HMSNs and small-sized Pt- PAMAM GEM A549 Chemotherapy [102]
low pH
The ionization of chemical groups in low pH Polymer − prodrug nanocomplexes SiRNA, MTX MDA-MB-231 Chemotherapy in conjugation with gene [107]
therapy
The ionization of chemical groups in low pH Triblock prodrug polymer micelles Capecitabine 4 T1 Chemotherapy in conjugation with two- [108]
photon AIE imaging
The ionization of chemical groups in low pH Reversible swelling–shrinking nanogel DOX Heps Chemotherapy [109]
Gelatin nanoparticles GNPs and prodrug Dox-Lac complex DOX HepG2 Chemotherapy [115]
MMPs sensitive bond-linked nanoparticles HA-pep-PAMAM macromolecules DOX A549, MCF-7 Chemotherapy [117]
MMPs sensitive bond-linked nanoparticles PEG–MMP2-cleavable peptide–phosphatidylethanolamie (PEG-pp-PE) DOX, dasatinib 4 T1 Chemotherapy [119]
copolymer micelles
MMPs sensitive bond-linked nanoparticles RGD-PEI-SS-PLA/PTX@ MMP-2-sensitive nanoclusters PTX MCF-7 Chemotherapy [120]
Overexpression of hyaluronidase (HAase) Nitric Oxide Donor and HA modified dendrimeric prodrug DOX, ICG 4 T1 Chemotherapy in conjugation with PTT [125]
Overexpression of hyaluronidase (HAase) Red blood cell membrane coated NPs Pheophorbide A, PTX, anti-PD-L1 4 T1 Chemotherapy, PDT and immunotherapy [127]
peptide dPPA
European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

Fig. 9. A schematic of DSPE-PEG based


micelles for inhibiting primary tumors
and lymphatic metastasis. Owing to the
small sizes, the drug delivery system
accumulated in the lymph nodes to cure
metastatic tumors. Moreover, copper(I)
triggered CuAAC between azide-func-
tionalized micelles and alkyne-functio-
nalized ones caused the aggregation,
greatly improving tumor accumulation.
Reprinted with permission from [140].

Fig. 10. Schematic representation of the delivery process of M−DOX / Wtmn (+) to tumor cells. M−Dox / Wtmn (+) was made up of ALK-M−DOX/Wtmn and N3-
M−DO X /Wtmn. Along with the injection of the catalysts in the tumor, the size increased to enhance drug accumulation, thus showing outstanding cytotoxicity as
well as autophagy inhibitory efficiency. Reprinted with permission from [141].

virulent Pt (II). The size transformation had been verified to promote intelligent drug delivery systems. However, the employment of light-
tumor distribution, penetration. In addition, the core–shell structured responsive materials of constructing size switchable nanoagents was
UCNPs- Pt -ZnFe2O4 (UCPZ) -PEG acted as the UV source to motivate rarely reported. Kohane and colleagues described UV light-triggered
PDT effect and endowed the energy for ZnFe2O4 to yield highly toxic size adjustable NPs, which provided spatiotemporal controlled release
·OH, taking the place of direct UV illumination. Moreover, based on the of drugs and the enhanced tissue penetration under 365 nm ultraviolet
inherent up-conversion luminescence (UCL) and the adopted Yb3+ and irradiation. In this system, the spiropyran (SP)-C9 transformed from
Fe3+, UCPZ-PEG was employed as a multimodality imaging contrast hydrophobic cores to zwitterionic merocyanine (MC)-C9 upon UV
agent. This research was involved with multiple therapies, including irradiation. In the meanwhile, the size of SP-C9 NPs reduced from
cancer diagnosis (UCL, X-ray computed tomography (CT)), photo- 150 nm to 40 nm due to the solubility change of SP-C9. ICG-modified
acoustic (PA) as well as magnetic resonance imaging (MRI) [131]. SP NPs penetrated about 8 nm in a collagen gel model without UV
illumination, whereas penetrated 12 nm and 16 nm with 10 and 20 s
irradiation [132]. In a following study, the photoswitchable DTX-
4.2.4. Light loaded NPs exhibited better therapeutic efficiency than free DTX as
4.2.4.1. UV light. Light had been repeatedly utilized to prepare

231
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

Fig. 11. Elucidation of the combined strategy: A) losartan pretreatment depleted the collagen I and enhanced penetration of DOX-AuNPs-GNPs. B) MMP2-triggered
size reduction and pH-mediated drug release. Reprinted with permission from [142].

well as DTX-loaded NPs without UV triggering [133]. Another type of sulfate, where ascorbic acid was used as the reducing agent). The azide
nanoassemblies with light-responsive property was constructed from and alkynyl reacted to increase the particle size and promoted tumor
linear flexible polyelectrolytes (poly (diallyl dimethylammonium tissue retention for the treatment of primary tumors. As a result, the
chloride or quartered poly (4-vinylpyridine)) and ionic photo- tumor lymph node metastasis dropped by 66.7%, showing the deeper
isomerize azo dyes (Acid Yellow 38). After UV irradiation, the penetration and better inhibitory effect in BALB/C mice [140]. And
exposure of UV-light caused the size decrease from a hydrodynamic recently, similar size-adjustable and azide/alkyne-modified micelles
radius of 94 nm to 62 nm. It was attributed to trans–cis isomerization of were employed through Cu (I)-catalyzed click reaction for synergistic
the dyes [134]. treatment of primary tumor and metastatic cancer (Fig. 10). Un-
doubtedly, through co-delivering DOX and autophagy inhibitor wort-
4.2.4.2. Near infrared (NIR) light. It was necessary to mention that the mannin (Wtmn), the smaller size DSPE-PEG micelles had outstanding
above UV-triggered formulations could really change size, thus cytotoxicity and obvious suppression effect in melanomas [141]. More
combining the advantages of larger size for extended blood importantly, this novel strategy provided a promising preclinical out-
circulation with smaller size for deep drug penetration. But UV light look for drug delivery to metastatic tumors.
was restricted due to the poorer tissue penetration and the damage to
normal tissues [135]. The employment of NIR light with deeper tissue 4.3. Combination strategies (Table 3)
penetration and higher biocompatibility to control size should be
regarded as a better option. Upon NIR light illumination, the Up to now, combination strategies have achieved very good results
personalized NIR light-activated UCNPs absorbed NIR light and in various laboratories. In Gao et al. lab, a dual strategy of adminis-
selectively released the Pt prodrug complex, which displayed higher trating MMP2-sensitive size shrinkable DOX-AuNPs-GNPs with losartan
cytotoxicity against human ovarian carcinoma A2780 cells and human was elaborated for high intertumoral DOX concentration and high
cisplatin-resistant variant A2780cis cells. Besides, the highly activated tumor apoptosis ratio (Fig. 11). Therefore, the combined NPs, in which
caspases effectively cleaved the probe peptide from NP surface, thereby the losartan depleted the collagen I, exhibited the optimal drug delivery
serving as personalized tumor markers to image in real time in living efficiency and noticeable penetration efficiency in breast tumor bearing
cells [136]. mice, thus providing a highly attractive way for breast cancer therapy
[142]. In the study of Chen et al., size-changeable “Trojan Horse” na-
4.2.5. Chemical reactions nocarriers (THNCs), which were consisted of PTX-loaded Greek soliders
Breast tumors, melanoma and other malignancies spread through (~20 nm), were assembled in amphiphilic gelatin matrix with the ad-
the lymphatic system to lead to the metastasis [137]. The conventional dition of hydrophilic losartan. As expected, after gelatin was cleaved by
nanodrug delivery systems (NDDs) with the fixed size could not address MMP-2, the THNCs exhibited the controllable drug release, higher pe-
this dilemma between primary tumor and lymph node metastasis. Not netration depth and higher cytotoxicity in 3D tumor spheroid models
surprisingly, in Kataoka et al. study, it was verified that smaller NPs [143]. In addition, the enhanced tumor penetration was acquired by
effectively inhibited the growth of lymph node metastasis [138]. Based shape transformation as well as RGD, where the shape transformation
on the successful application of Cu (I)-mediated plasmonic nanosensors was attributed to the overexpression of MMP 9 [144]. Tumor perme-
in the real clinical practice [139], He et al. designed size-transformable ability was improved by NIR-induced size shrinkage. UCNPs could
polymeric micelles for targeted delivery of anti-tumor drugs in primary transform NIR to UV–vis and remove PEG for deep penetration [145].
tumors and lymph node metastatic tumors. They were modified with In another study, hydrophobic agents (CPI-613) were conjugated with
reaction substrate (alkyne group or azide group) on the surface of 1,2- hydrophilic polymers via MMP 2 sensitive linker. The simultaneous
distearoyl-sn-glycero-3-phosphoethanolamine (D SPE)-PEG micelles targeting of tumor cells and stroma was achieved by the co-delivery of
(Fig. 9). The nanocarriers underwent Cu (I) triggered click cycloaddi- TGF-β receptor I/II inhibitor and CPI-613 for deeper treatment of
tions: firstly, the small size micelles (~25 nm) easily delivered drugs to pancreatic cancers [146]. In the study of Jiang et al, TAMs polarization
the lymph nodes to cure metastases; secondly, the size increased to and acid-triggered size transformation reduced IFP and loosened ECM.
120 nm along with the injection of catalysts (ascorbic acid and copper As a result, dandelion-like NPs led to longer circulation and deeper

232
Table 3
Examples of combination strategies for improving tumor penetration.
Strategy Drug Type of Tumor Highlight of the research Therapy Ref
F. Peng, et al.

Losartan + gelatin NPs DOX 4 T1 Size-shrinkable gelatin NPs with collagen depletion by losartan Chemotherapy [142]
Losartan + gelatin NPs PTX HeLa Size-changeable gelatin NPs with collagen depletion by losartan Chemotherapy [143]
RGD + MMP-9 sensitive linker DOX H22, 4 T1 NP shape transformation with enhanced penetration by RGD Chemotherapy [144]
NIR + RGD DOX MCF-7 NIR-triggered size shrinkage and RGD activation Chemotherapy [145]
TGF-β receptor I/II inhibitor + MMP-2 sensitive linker CPI-613 PANC-1 Normalization of the stroma and cleavage of MMP-2-responsive linker Chemotherapy [146]
TAMs modulation and low pH PTX 4 T1 Normalization of the stroma and acid-triggered size variation Chemotherapy [147]
CAFs regulation + MMP-2 sensitive linker GEM 4 T1 CAFs-targeted regulation and cleavage of MMP-2-responsive linker Chemotherapy [148]
VEGF inhibitor + low pH Etoposide Luc-A549 The protonation of imidazole group and downregulation of VEGF Chemotherapy [149]
RGD + hyperthermia / 4 T1 Magnetic induction hyperthermia and RGD activation tumor imaging and heat treatment [150]

233
Table 4
Recent and typical clinical therapies.
Pathway Operating conditions Tumors Main conclusion Clinical trial Ref

MMP Inhibitor MMP activity was determined before and after Advanced and refractory solid tumors It is well tolerated and obviously inhibited gelatinase Phase I [151]
S-3304 injection activity in volunteers
Degradation of HA PEGPH20 Metastatic pancreatic ductal Higher drug delivery efficiency than drug alone Randomized phase II [152]
adenocarcinoma study
Losartan Losartan for 8 cycles Locally advanced pancreatic cancer Neoadjuvant therapy with losartan prolonged survival Phase II [153]
rate in patients
Targeting TAMs Emactuzumab every 2 or 3 weeks Advanced/metastatic solid tumors Specific depletion of immunosuppressive macrophages Phase I [154]
VEGF inhibitors Bevacizumab or cetuximab Resected pancreatic carcinoma Feasible and safe Phase II [155]
VEGF inhibitors + TNF-α Lenalidomide Advanced pancreatic cancer Feasible and safe Phase I [156]
TNF-α + NGR A peptide-tumor necrosis factor-a fusion Malignant pleural mesothelioma Stroma normalization and targeted drug deliveryuncit Randomize phase III [64]
protein
Microbubbles in combination with pulsed Acoustic pressure increased from 0.5 to Recurrent GBM Blood-brain barrier disruption and optimal carboplatin Phase 1/2a [157]
ultrasound 1.1 MPa delivery
Hyperthermia Microwave spiral strip applicators operating at Superficial tumors Adjuvant HT improves efficacy in radiation-treated Randomized Trial [158]
433 MHz. patients
European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

tumor penetration in tumor tissues in vivo [147]. Similarly, deep drug Nanomed Nanobiotechnol. 11 (2019) e1519, , https://doi.org/10.1002/wnan.
delivery was achieved through CAFs-targeted regulation and the clea- 1519.
[8] V.P. Chauhan, T. Stylianopoulos, J.D. Martin, Z. Popovic, O. Chen, W.S. Kamoun,
vage of MMP 2 responsive linker in the treatment of 4 T1 breast cancer M.G. Bawendi, D. Fukumura, R.K. Jain, Normalization of tumour blood vessels
models [148]. Based on the protonation of imidazole groups under improves the delivery of nanomedicines in a size-dependent manner, Nat
acidic conditions, the co-delivery of VEGF inhibitor and etoposide had Nanotechnol. 7 (2012) 383–388, https://doi.org/10.1038/nnano.2012.45.
[9] S.D. Perrault, C. Walkey, T. Jennings, H.C. Fischer, W.C. Chan, Mediating tumor
also shown great potential in promoting penetration and inhibiting targeting efficiency of nanoparticles through design, Nano Lett. 9 (2015)
tumor proliferation [149]. In a recent study, the penetration depth of 1909–1915, https://doi.org/10.1021/nl900031y.
nanoprobes was also promoted by magnetic induction hyperthermia [10] H.J. Li, J.Z. Du, J. Liu, X.J. Du, S. Shen, Y.H. Zhu, X. Wang, X. Ye, S. Nie, J. Wang,
Smart superstructures with ultrahigh pH-sensitivity for targeting acidic tumor
and RGD activation [150]. microenvironment: instantaneous size switching and improved tumor penetration,
ACS Nano. 10 (2016) 6753–6761, https://doi.org/10.1021/acsnano.6b02326.
4.4. Clinical therapies (Table 4) [11] M.R. Dreher, W. Liu, C.R. Michelich, M.W. Dewhirst, F. Yuan, A. Chilkoti, Tumor
vascular permeability, accumulation, and penetration of macromolecular drug
carriers, J Natl Cancer Inst. 98 (2016) 335–344, https://doi.org/10.1093/jnci/
At present, penetration-promoting therapies have been put into djj070.
clinical trials in cancer treatment. Nevertheless, combination therapies [12] R. Misra, S. Acharya, S.K. Sahoo, Cancer nanotechnology: application of nano-
have been rarely applied to inhibit tumor growth in clinical. In earlier technology in cancer therapy, Drug Discov Today. 15 (2016) 842, https://doi.org/
10.1016/j.drudis.2010.08.006.
years, the application of MMP inhibitor and HT was proved to be safe, [13] T. Stylianopoulos, EPR-effect: utilizing size-dependent nanoparticle delivery to s
effective and well-tolerated for a phase I trial and randomized trial. olid tumors, Ther Deliv. 4 (2013) 421–423, https://doi.org/10.4155/tde.13.8.
Other recent strategies were also shown to optimize drug delivery at [14] B.L. Hylander, A. Sen, S.H. Beachy, R. Pitoniak, S. Ullas, J.F. Gibbs, J. Qiu,
J.D. Prey, G.J. Fetterly, E.A. Repasky, Tumor priming by Apo2L/TRAIL reduces
different stages of clinical trials. It turned out that penetration-pro- interstitial fluid pressure and enhances efficacy of liposomal gemcitabine in a
moting strategies played a vital role in clinical trials (Table 4). patient derived xenograft tumor model, J Control Release. 217 (2015) 160–169,
https://doi.org/10.1016/j.jconrel.2015.08.047.
[15] S. Stapleton, M. Milosevic, I.F. Tannock, C. Allen, D.A. Jaffray, The intra-tumoral
5. Conclusion and future prospective relationship between microcirculation, interstitial fluid pressure and liposome
accumulation, J Control Release. 211 (2015) 163–170, https://doi.org/10.1016/j.
In this review, we elaborate the penetration obstacles which exist in jco nrel.2015.06.008.
[16] J. Liu, M. Li, Z. Luo, L. Dai, X. Guo, K. Cai, Design of nanocarriers based on
tumor regions, and summarize the important properties of NPs which complex biological barriers in vivo for tumor therapy, Nano Today. 15 (2017)
hinder deep penetration. Then, the latest and potential drug delivery 56–90, https://doi.org/10.1016/j.nantod.2017.06.010.
nanosystems for improving tumor penetration are analyzed in detail. At [17] R.K. Jain, J.D. Martin, T. Stylianopoulos, The role of mechanical forces in tumor
growth and therapy, Annu Rev Biomed Eng. 16 (2014) 321–346, https://doi.org/
last, recent combination therapies and clinical therapies are introduced
10.1146/annurev -bioeng-071813-105 259.
to enhance tumor penetration and antitumor efficiency. These therapies [18] L. Li, J. Sun, Z. He, Deep penetration of nanoparticulate drug delivery systems into
show great potential in the aspect of chemotherapy, PDT, PTT, bioi- tumors: challenges and solutions, Curr Med Chem. 20 (2013) 2881–2891, https://
maging therapy and immunotherapy. However, as our ultimate purpose doi.org/10.2174/092986731132099 90004.
[19] G. Wang, Y. Chen, P. Wang, Y. Wang, H. Hong, Y. Li, J. Qian, Y. Yuan, B. Yu,
is to treat human diseases, the precise doses, suitable time and drug C. Liu, Preferential tumor accumulation and desirable interstitial penetration of
safety are needed to be highly concerned. In other words, each therapy poly (lactic-co-glycolic acid) nanoparticles with dual coating of chitosan oligo-
requires further testing to define its therapeutic applicability and clin- saccharide a nd polyethylene glycol-poly (D, L-lactic acid), Acta Biomater. 29
(2016) 248–260, https://doi.org/10.1016/j.actbio.2015.10.017.
ical utility. [20] S. Suzuki, S. Itakura, R. Matsui, K. Nakayama, T. Nishi, A. Nishimoto, S. Hama,
K. Kogure, Tumor microenvironment-sensitive liposomes penetrate tumor tissue
Conflicts of interest via attenuated interaction of the extracellular matrix and tumor cells and ac-
companying actin depolymerization, Biomacromolecules. 18 (2017) 535–543,
https://doi.org/10.1021/acs.bio mac.6b01688.
The authors declare no conflict of interest. [21] Q.V. Le, J. Suh, Y.K. Oh, Nanomaterial-based modulation of tumor micro en-
vironments for enhancing chemo/immunotherapy, AAPS J. 21 (2019) 64, https://
doi.org/10.1208/s12248-019-0333-y.
Acknowledgements
[22] Y. Anraku, A. Kishimura, A. Kobayashi, M. Oba, K. Kataoka, Size-controlled long-
circulating PICsome as a ruler to measure critical cut-off disposition size into
This work was supported by the National Natural Science normal and tumor tissues, Chem Commun (Camb). 47 (2011) 6054–6056, https://
doi.org/10.1039/c1cc11465d.
Foundation of China (81202480, 81302723); the Natural Science
[23] S. Watanabe, K. Hayashi, K. Toh, H.J. Kim, X. Liu, H. Chaya, S. Fukushima,
Foundation of Liaoning Province (2015020749). K. Katsushima, Y. Kondo, S. Uchida, S. Ogura, T. Nomoto, H. Takemoto, H. Cabral,
H. Kinoh, H.Y. Tanaka, M.R. Kano, Y. Matsumoto, H. Fukuhara, S. Uchida,
References M. Nangaku, K. Osada, N. Nishiyama, K. Miyata, K. Kataoka, In vivo rendezvous of
small nucleic acid drugs with charge-matched block catiomers to target cancers,
Nat Commun. 10 (2019) 1894, https://doi.org/10.1038/s41467-019-09856-w.
[1] S. Ferrari, M. Serra, An update on chemotherapy for osteosarcoma, Expert Opin [24] J. Wang, W. Mao, L.L. Lock, J. Tang, M. Sui, W. Sun, H. Cui, D. Xu, Y. Shen, The
Pharmaco. 16 (2015) 2727–2736, https://doi.org/10.1517/14656566.2015. role of micelle size in tumor accumulation, penetration, and treatment, ACS Nano.
1102226. 9 (2015) 7195–7206, https://doi.org/10.1021/acsnano.5b02017.
[2] Y. Ma, J.M. Pitt, Q. Li, H. Yang, The renaissance of anti-neoplastic immunity from [25] J. Zhang, X. Wang, J. Wen, X. Su, L. Weng, C. Wang, Y. Tian, Y. Zhang, J. Tao,
tumor cell demise, Immunol Rev. 280 (2017) 194–206, https://doi.org/10.1111/ P. Xu, G. Lu, Z. Teng, L. Wang, Size effect of mesoporous organosilica nano-
imr.12586. particles on tumor penetration and accumulation, Biomater Sci. 7 (2019)
[3] E. Vlashi, F. Pajonk, Cancer stem cells, cancer cell plasticity and radiation therapy, 4790–4799, https://doi.org/10.1039/c9bm01164a.
Semin Cancer Biol. 31 (2015) 28–35, https://doi.org/10.1016/j.semcancer.2014. [26] J. Bugno, M.J. Poellmann, K. Sokolowski, H.J. Hsu, D.H. Kim, S. Hong, Tumor
07.001. penetration of sub-10 nm nanoparticles: effect of dendrimer properties on their
[4] L.J. Solin, R. Gray, L.L. Hughes, W.C. Wood, M.A. Lowen, S.S. Badve, F.L. Baehner, penetration in multicellular tumor spheroids, Nanomedicine. 21 (2019) 102059, ,
J.N. Ingle, E.A. Perez, A. Recht, J.A. Sparano, N.E. Davidson, Surgical excision https://doi.org/10.1016/j.nano.2019.102059.
without radiation for ductal carcinoma in situ of the breast: 12-year results from [27] H.J. Li, J. Liu, Y.L. Luo, S.B. Chen, R. Liu, J.Z. Du, J. Wang, Intratumor perfor-
the ECOG-ACRIN E5194 study, J Clin Oncol. 33 (2015) 3938–3944, https://doi. mance and therapeutic efficacy of PAMAM dendrimers carried by clustered na-
org/10.1200/JCO.2015.60.8588. noparticles, Nano Lett. 19 (2019) 8947–8955, https://doi.org/10.1021/acs.
[5] B.N. Ho, C.M. Pfeffer, A.T.K. Singh, Update on nanotechnology-based drug de- nanolett.9b03913.
livery systems in cancer treatment, Anticancer Res. 37 (2017) 5975–5981, https:// [28] H.X. Wang, Z.Q. Zuo, J.Z. Du, Y.C. Wang, R. Sun, Z.T. Cao, X.D. Ye, J.L. Wang,
doi.org/10.21873/anticanres.12044. K.W. Leong, J. Wang, Surface charge critically affects tumor penetration and
[6] M. Wu, H.B. Frieboes, S.R. McDougall, M.A. Chaplain, V. Cristini, J. Lowengrub, therapeutic efficacy of cancer nanomedicines, Nano Today. 11 (2016) 133–144,
The effect of interstitial pressure on tumor growth: coupling with the blood and https://doi.org/10.1016/j.nantod.2016.04.008.
lymphatic vascular systems, J Theor Biol. 320 (2013) 131–151, https://doi.org/ [29] G. Wang, Y. Chen, P. Wang, Y. Wang, H. Hong, Y. Li, J. Qian, Y. Yuan, B. Yu,
10.1016/j.jtbi.2 012.11.031. C. Liu, Preferential tumor accumulation and desirable interstitial penetration of
[7] Y.R. Zhang, R. Lin, H.J. Li, W.L. He, J.Z. Du, J. Wang, Strategies to improve tumor poly (lactic-co-glycolic acid) nanoparticles with dual coating of chitosan oligo-
penetration of nanomedicines through nanoparticle design, Wiley Interdiscip Rev saccharide and polyethylene glycol-poly (D, L-lactic acid), Acta Biomater. 29

234
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

(2016) 248–260, https://doi.org/10.1016/j.actbio.2015.10.017. D. Hedley, S.D. Li, Targeting of metastasis-promoting tumor-associated fibroblasts
[30] S. Krasnici, A. Werner, M.E. Eichhorn, M. Schmitt-Sody, S.A. Pahernik, B. Sauer, and modulation of pancreatic tumor-associated stroma with a carboxymethyl
B. Schulze, M. Teifel, U. Michaelis, K. Naujoks, M. Dellian, Effect of the surface cellulose-docetaxel nanoparticle, J Control Release. 206 (2015) 122–130 10.10
charge of liposomes on their uptake by angiogenic tumor vessels, Int J Cancer. 105 16/j.jconrel.2015.03.023.
(2003) 561–567, https://doi.org/10.1002/ijc.11108. [52] L. Miao, Q. Liu, C.M. Lin, C. Luo, Y. Wang, L. Liu, W. Yin, S. Hu, W.Y. Kim,
[31] S. Jin, X. Ma, H. Ma, K. Zheng, J. Liu, S. Hou, J. Meng, P.C. Wang, X. Wu, L. Huang, Targeting tumor-associated fibroblasts for therapeutic delivery in des-
X.J. Liang, Surface chemistry-mediated penetration and gold nanorod thermo- moplastic tumors, Cancer Res. 77 (2017) 719–731, https://doi.org/10.1158/
therapy in multicellular tumor spheroids, Nanoscale. 5 (2013) 143–146, https:// 0008-5472.CA N-16-0866.
doi.org/10.1039/c2nr318 77f. [53] Lin Hou, Dandan Chen, Lisha Hao, Chunyu Tian, Yingshan Yan, Ling Zhu,
[32] L. Zhang, Y. Wang, D. Yang, W. Huang, P. Hao, S. Feng, D. Appelhans, T. Zhang, Huijuan Zhang, Yi Zhang, Zhenzhong Zhang, Transformable nanoparticles trig-
X. Zan, Shape effect of nanoparticles on tumor penetration in monolayers versus gered by cancer-associated fibroblasts for improving drug permeability and effi-
spheroids, Mol Pharm. 16 (2019) 2902–2911, https://doi.org/10.1021/acs. cacy in desmoplastic tumors, Nanoscale 11 (42) (2019) 20030–20044, https://doi.
molpharmaceut.9b001 07. org/10.1039/C9NR06438A.
[33] W. Zhang, M. Yu, Z. Xi, D. Nie, Z. Dai, J. Wang, K. Qian, H. Weng, Y. Gan, L. Xu, [54] C.B. Rodell, S.P. Arlauckas, M.F. Cuccarese, C.S. Garris, R. Li, M.S. Ahmed,
Cancer cell membrane-camouflaged nanorods with endoplasmic reticulum tar- R.H. Kohler, M.J. Pittet, R. Weissleder, TLR7/8-agonist-loaded nanoparticles
geting for improved antitumor therapy, ACS Appl Mater Interfaces. (2019 Dec 5), promote the polarization of tumour-associated macrophages to enhance cancer
https://doi.org/10.1021/acsami.9b18388. immunotherapy, Nat Biomed Eng. 2 (2018) 578–588, https://doi.org/10.1038/
[34] D. Nie, Z. Dai, J. Li, Y. Yang, Z. Xi, J. Wang, W. Zhang, K. Qian, S. Guo, C. Zhu, s41551-018-0236-8.
R. Wang, Y. Li, M. Yu, X. Zhang, X. Shi, Y. Gan, Cancer cell membrane-coated [55] N.N. Parayath, A. Parikh, M.M. Amiji, Repolarization of tumor-associated mac-
nanoparticles with a yolk-shell structure augment cancer chemotherapy, Nano rophages in a genetically engineered nonsmall cell lung Cancer model by in-
Lett. (2019 Nov 5), https://doi.org/10.1021/acs.nano lett.9b03817. traperitoneal administration of hyaluronic acid-based nanoparticles encapsulating
[35] H. Lee, H. Fonge, B. Hoang, R.M. Reilly, C. Allen, The effects of particle size and microRNA-125b, 3571 2579, Nano Lett. 18 (2018), https://doi.org/10.1021/acs.
molecular targeting on the intratumoral and subcellular distribution of polymeric nanolett.8b0 0689.
nanoparticles, Mol Pharm. 7 (2010) 1195–1208, https://doi.org/10.1021/ [56] Q. Chen, L. Feng, J. Liu, W. Zhu, Z. Dong, Y. Wu, Z. Liu, Intelligent albumin–MnO2
mp100038h. nanoparticles as pH−/H2O2-responsive dissociable nanocarriers to modulate
[36] Q. Sun, T. Ojha, F. Kiessling, T. Lammers, Y. Shi, Enhancing tumor penetration of tumor hypoxia for effective combination therapy, Adv Mater. 28 (2016)
nanomedicines, Biomacromolecules. 18 (2017) 1449–1459, https://doi.org/10. 7129–7136, https://doi.org/10.1002/adma.2 01601902.
1021/acs.bio mac.7b00068. [57] G. Song, Y. Chen, C. Liang, X. Yi, J. Liu, X. Sun, S. Shen, K. Yang, Z. Liu, Catalase-
[37] T.T. Goodman, P.L. Olive, S.H. Pun, Increased nanoparticle penetration in col- loaded TaOx nanoshells as bio-nanoreactors combining high-Z element and en-
lagenase-treated multicellular spheroids, Int J Nanomedicine. 2 (2007) 265–274. zyme delivery for enhancing radiotherapy, Adv Mater. 28 (2016) 7143–7148,
[38] A. Zinger, L. Koren, O. Adir, M. Poley, M. Alyan, Z. Yaari, N. Noor, N. Krinsky, https://doi.org/10.1002/adma.201602111.
A. Simon, H. Gibori, M. Krayem, Y. Mumblat, S. Kasten, S. Ofir, E. Fridman, [58] M.R. Kano, Y. Bae, C. Iwata, Y. Morishita, M. Yashiro, M. Oka, T. Fujii, A. Komuro,
N. Milman, M.M. Lübtow, L. Liba, J. Shklover, J. Shainsky-Roitman, K. Kiyono, M. Kaminishi, K. Hirakawa, Y. Ouchi, N. Nishiyama, K. Kataoka,
Y. Binenbaum, D. Hershkovitz, Z. Gil, T. Dvir, R. Luxenhofer, R. Satchi-Fainaro, K. Miyazono, Improvement of cancer-targeting therapy using nanocarriers for
A. Schroeder, Collagenase nanoparticles enhance the penetration of drugs into intractable solid tumors by inhibition of TGF-beta signaling, Proc Natl Acad Sci U S
pancreatic tumors, ACS Nano. 13 (2019) 11008–11021, https://doi.org/10.1021/ A. 104 (2007) 3460–3465, https://doi.org/10.1073/pnas.0611660104.
acs nano.9b02395. [59] H. Cabral, Y. Matsumoto, K. Mizuno, Q. Chen, M. Murakami, M. Kimura,
[39] H. Gong, Y. Chao, J. Xiang, X. Han, G. Song, L. Feng, J. Liu, G. Yang, Q. Chen, Y. Terada, M.R. Kano, K. Miyazono, M. Uesaka, N. Nishiyama, K. Kataoka,
Z. Liu, Hyaluronidase to enhance nanoparticle-based photodynamic tumor Accumulation of sub-100 nm polymeric micelles in poorly permeable tumours
therapy, Nano Lett 16 (2016) 2512–2521, https://doi.org/10.1021/acs.nanolett. depends on size, Nat nanotechnol. 6 (2011) 815–823, https://doi.org/10.1038/
6b00068. nnano.2011.166.
[40] H. Zhou, Z. Fan, J. Deng, P.K. Lemons, D.C. Arhontoulis, W.B. Bowne, H. Cheng, [60] K. Hu, L. Miao, T.J. Goodwin, J. Li, Q. Liu, L. Huang, Quercetin remodels the
Hyaluronidase embedded in nanocarrier PEG shell for enhanced tumor penetra- tumor microenvironment to improve the permeation, retention, and antitumor
tion and highly efficient antitumor efficacy, Nano Lett. 16 (2016) 3268–3277, effects of nanoparticles, ACS Nano. 11 (2017) 4916–4925, https://doi.org/10.
https://doi.org/10.1021/acs.nanolett.6b00820. 1021/acsnano.7b01522.
[41] K.B. Neeves, A.J. Sawyer, C.P. Foley, W.M. Saltzman, W.L. Olbricht, Dilation and [61] R. Van Horssen, T.L. Ten Hagen, A.M. Eggermont, TNF-alpha in cancer treatment:
degradation of the brain extracellular matrix enhance penetration of infused molecular insights, antitumor effects, and clinical utility, Oncologist. 11 (2016)
polymer nanoparticles, Brain Res. 1180 (2007) 121–132, https://doi.org/10. 397–408, https://doi.org/10.1634/theon cologist.11-4-397.
1016/j.brain res.2007.08.050. [62] F. Curnis, A. Sacchi, A. Corti, Improving chemotherapeutic drug penetration in
[42] Z.I. Arshad, A. Amid, F. Yusof, I. Jaswir, K. Ahmad, S.P. Loke, Bromelain: an tumors by vascular targeting and barrier alteration, J Clin Invest. 110 (2002)
overview of industrial application and purification strategies, Appl Microbiol 475–482, https://doi.org/10.1172/JCI 15223.
Biotechnol. 98 (2014) 7283–7297, https://doi.org/10.1007/s00253-014-5889-y. [63] T.L. Ten Hagen, A.H. Van Der Veen, P.T. Nooijen, S.T. Van Tiel, A.L. Seynhaeve,
[43] A. Parodi, S.G. Haddix, N. Taghipour, S. Scaria, F. Taraballi, A. Cevenini, A.M. Eggermont, Low-dose tumor necrosis factor-alpha augments antitumor ac-
I.K. Yazdi, C. Corbo, R. Palomba, S.Z. Khaled, J.O. Martinez, B.S. Brown, tivity of stealth liposomal doxorubicin (DOXIL) in soft tissue sarcoma-bearing rats,
L. Isenhart, E. Tasciotti, Bromelain surface modification increases the diffusion of Int J Cancer. 87 (2000) 829–837.
silica nanoparticles in the tumor extracellular matrix, ACS Nano. 8 (2014) [64] A. Corti, F. Pastorino, F. Curnis, W. Arap, M. Ponzoni, R. Pasqualini, Targeted drug
9874–9883, https://doi.org/10.1021/nn502807n. delivery and penetration into solid tumors, Med Res Rev. 32 (2012) 1078–1091,
[44] I. Beyer, Z. Li, J. Persson, Y. Liu, R. van Rensburg, R. Yumul, X.B. Zhang, https://doi.org/10.1002/med.20238.
M.C. Hung, A. Lieber, Controlled extracellular matrix degradation in breast cancer [65] K. Pietras, A. Ostman, M. Sjöquist, E. Buchdunger, R.K. Reed, C.H. Heldin,
tumors improves therapy by trastuzumab, Mol Ther. 19 (2011) 479–489, https:// K. Rubin, Inhibition of platelet-derived growth factor receptors reduces interstitial
doi.org/10.1038/mt.2 010.256. hypertension and increases transcapillary transport in tumors, Cancer Res. 61
[45] J. Henshaw, B. Mossop, F. Yuan, Relaxin treatment of solid tumors: effects on (2001) 2929–2934.
electric field-mediated gene delivery, Mol Cancer Ther. 7 (2008) 2566–2573, [66] A.M. Santos, J. Jung, N. Aziz, J.L. Kissil, E. Puré, Targeting fibroblast activation
https://doi.org/10.1158/1535-71 63.MCT-08-0435. protein inhibits tumor stromagenesis and growth in mice, J Clin Invest. 119 (2009)
[46] H. Shen, Q. Gao, Q. Ye, S. Yang, Y. Wu, Q. Huang, X. Wang, Z. Sun, Peritumoral 3613–3625, https://doi.org/10.1530/ERC-10-0224.
implantation of hydrogel-containing nanoparticles and losartan for enhanced na- [67] Q. Feng, C. Zhang, D. Lum, J.E. Druso, B. Blank, K.F. Wilson, A. Welm,
noparticle penetration and antitumor effect, Int J Nanomedicine. 13 (2018) M.A. Antonyak, R.A. Cerione, A class of extracellular vesicles from breast cancer
7409–7426, https://doi.org/10.2147/IJN.S 178585. cells activates VEGF receptors and tumour angiogenesis, Nat Commun. 8 (2017)
[47] B. Diop-Frimpong, V.P. Chauhan, S. Krane, Y. Boucher, R.K. Jain, Losartan inhibits 14450, https://doi.org/10.1038/ncomms14450.
collagen I synthesis and improves the distribution and efficacy of nanotherapeutics [68] V.P. Chauhan, T. Stylianopoulos, J.D. Martin, Z. Popović, O. Chen, W.S. Kamoun,
in tumors, Proc Natl Acad Sci USA 108 (2011) 2909–2914, https://doi.org/10. M.G. Bawendi, D. Fukumura, R.K. Jain, Normalization of tumour blood vessels
1073/pnas.1018892108. improves the delivery of nanomedicines in a size-dependent manner, Nat
[48] L. Zhang, Y. Wang, Y. Yang, Y. Liu, S. Ruan, Q. Zhang, X. Tai, J. Chen, T. Xia, Nanotechnol. 7 (2017) 383–388, https://doi.org/10.1038/nnano.2012.45.
Y. Qiu, H. Gao, Q. He, High tumor penetration of paclitaxel loaded pH sensitive [69] C. Masuda, M. Sugimoto, D. Wakita, M. Monnai, C. Ishimaru, R. Nakamura,
cleavable liposomes by depletion of tumor collagen I in breast cancer, ACS Appl M. Kinoshita, K. Yorozu, M. Kurasawa, O. Kondoh, K. Yamamoto, Bevacizumab
Mater Interfaces. 7 (2015) 9691–9701, https://doi.org/10.1021/acsami.5b01473. suppresses the growth of established non-small-cell lung cancer brain metastases
[49] Y. Zhang, Y. Liu, X. Gao, X. Li, X. Niu, Z. Yuan, W. Wang, Near-infrared-light in a hematogenous brain metastasis model, Clin Exp Metastasis. (2019 Nov 25),
induced nanoparticles with enhanced tumor tissue penetration and intelligent https://doi.org/10.1007/s10585-019-10008-z.
drug release, Acta Biomater. 90 (2019) 314–323, https://doi.org/10.1016/j. [70] Z. Zhao, Y. Li, R. Shukla, H. Liu, A. Jain, A. Barve, K. Cheng, Development of a
actbio.2019.04.022. biocompatible copolymer nanocomplex to deliver VEGF siRNA for triple negative
[50] B. Chen, W. Dai, D. Mei, T. Liu, S. Li, B. He, B. He, L. Yuan, H. Zhang, X. Wang, breast cancer, Theranostics. 9 (2019) 4508–4524, https://doi.org/10.7150/thno.
Q. Zhang, Comprehensively priming the tumor microenvironment by cancer-as- 34314.
sociated fibroblast-targeted liposomes for combined therapy with cancer cell-tar- [71] L. Liu, Y. Cao, C. Chen, X. Zhang, A. McNabola, D. Wilkie, S. Wilhelm, M. Lynch,
geted chemotherapeutic drug delivery system, J Control Release. 241 (2016) C. Carter, Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angio-
68–80, https://doi.org/10.1016/j.jconrel.2016.09.014. genesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/
[51] M.J. Ernsting, B. Hoang, I. Lohse, E. Undzys, P. Cao, T. Do, B. Gill, M. Pintilie, PRF/5, Cancer Res. 66 (2006) 11851–11858, https://doi.org/10.1158/0008-

235
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

5472.CAN-06-1 377. [93] S. Eetezadi, R. De Souza, M. Vythilingam, R. Lessa Cataldi, C. Allen, Effects of
[72] Y. Yao, T. Wang, Y. Liu, N. Zhang, Co-delivery of sorafenib and VEGF-siRNA via doxorubicin delivery systems and mild hyperthermia on tissue penetration in 3D
pH-sensitive liposomes for the synergistic treatment of hepatocellular carcinoma, cell culture models of ovarian cancer residual disease, Mol Pharm. 12 (2015)
Artif Cells Nanomed Biotechnol. 47 (2019) 1374–1383, https://doi.org/10.1080/ 3973-3985. doi: 10.1021/acs. molpharmaceut.5b00426.
21691401.20 19.1596943. [94] Z. Deng, Y. Xiao, M. Pan, F. Li, W. Duan, L. Meng, X. Liu, F. Yan, H. Zheng,
[73] X. Qian, L. Ge, K. Yuan, C. Li, X. Zhen, W. Cai, R. Cheng, X. Jiang, Targeting and Hyperthermia-triggered drug delivery from iRGD-modified temperature-sensitive
microenvironment-improving of phenylboronic acid-decorated soy protein nano- liposomes enhances the anti-tumor efficacy using high intensity focused ultra-
particles with different sizes to tumor, Theranostics. 9 (2019) 7417–7430, https:// sound, J Control Release. 243 (2016) 333–341, https://doi.org/10.1016/j.jconrel.
doi.org/10.7150/thno.33470. 2016.10.030.
[74] F. Karandish, J. Froberg, P. Borowicz, J.C. Wilkinson, Y. Choi, S. Mallik, Peptide- [95] M. Rezaeian, A. Sedaghatkish, M. Soltani, Numerical modeling of high-intensity
targeted, stimuli-responsive polymersomes for delivering a cancer stemness in- focused ultrasound-mediated intraperitoneal delivery of thermosensitive lipo-
hibitor to cancer stem cell microtumors, Colloids Surf B Biointerfaces. 163 (2018) somal doxorubicin for cancer chemotherapy, Drug Deliv. 26 (2019) 898–917,
225–235, https://doi.org/10.1016/j.colsurfb.2 017.12.036. https://doi.org/10.1080/10717544.2019.1660435.
[75] H.J. Cho, S.J. Park, Y.S. Lee, S. Kim, Theranostic iRGD peptide containing cisplatin [96] P.C. Lyon, L.F. Griffiths, J. Lee, D. Chung, R. Carlisle, F. Wu, M.R. Middleton,
prodrug: dual-cargo tumor penetration for improved imaging and therapy, J F.V. Gleeson, C.C. Coussios, Clinical trial protocol for TARDOX: a phase I study to
Control Release. 300 (2019) 73–80, https://doi.org/10.1016/j.jconrel.2019.02. investigate the feasibility of targeted release of lyso-thermosensitive liposomal
043. doxorubicin (ThermoDox®) using focused ultrasound in patients with liver tu-
[76] Z. Zhu, C. Xie, Q. Liu, X. Zhen, X. Zheng, W. Wu, R. Li, Y. Ding, X. Jiang, B. Liu, mours, J Ther Ultrasound. 5 (2017) 28, https://doi.org/10.1186/s40349-017-
The effect of hydrophilic chain length and iRGD on drug delivery from poly (ε- 0104-0.
caprolactone)-poly(N-vinylpyrrolidone) nanoparticles, Biomaterials. 32 (2011) [97] A.M. Kallinen, M.P. Sarparanta, D. Liu, E.M. Mäkilä, J.J. Salonen, J.T. Hirvonen,
9525–9535 10.10 16/j.biomaterials.2011.08.072. H.A. Santos, A.J. Airaksinen, In vivo evaluation of porous silicon and porous si-
[77] Huiming Liu, Xiaoguang Shi, Di Wu, Frewein Kahsay Khshen, Liandong Deng, licon solid lipid nanocomposites for passive targeting and imaging, Mol Pharm. 11
Anjie Dong, Weiwei Wang, Jianhua Zhang, Injectable, Biodegradable, (2014) 2876–2886, https://doi.org/10.1021/mp500225b.
Thermosensitive Nanoparticles-Aggregated Hydrogel with Tumor-Specific [98] S. Peppicelli, F. Bianchini, L. Calorini, Extracellular acidity, a “reappreciated” tra
Targeting, Penetration, and Release for Efficient Postsurgical Prevention of Tumor it of tumor environment driving malignancy: perspectives in diagnosis and
Recurrence, ACS Appl. Mater. Interfaces 11 (22) (2019) 19700–19711, https:// therapy, Cancer Metastasis Rev. 33 (2014) 823–832, https://doi.org/10.1007/
doi.org/10.1021/acsami.9b0198710.1021/acsami.9b01987.s001. s10555-0 14-9506-4.
[78] J. Yang, J. Yang, Y. Wei, H. Yin, L. Fang, D. Chai, H. Li, H. Li, Q. Zhang, J. Zheng, [99] H.J. Li, J.Z. Du, X.J. Du, C.F. Xu, C.Y. Sun, H.X. Wang, Z.T. Cao, X.Z. Yang,
Modification of IL-24 by tumor penetrating peptide iRGD enhanced its antitumor Y.H. Zhu, S. Nie, J. Wang, Stimuli-responsive clustered nanoparticles for improved
efficacy against non-small cell lung cancer, Int Immunopharmacol. 70 (2019) tumor penetration and therapeutic efficacy, Proc Natl Acad Sci U S A. 113 (2016)
125–134, https://doi.org/10.1016/j.intim p. 2019.02.027. 4164–4169, https://doi.org/10.1073/p nas.1522080113.
[79] X. Song, Z. Wan, T. Chen, Y. Fu, K. Jiang, X. Yi, H. Ke, J. Dong, L. Yang, L. Li, [100] J. Kim, Y.M. Lee, Y. Kang, W.J. Kim, Tumor-homing, size-tunable clustered na-
X. Sun, T. Gong, Z. Zhang, Development of a multi-target peptide for potentiating noparticles for anticancer therapeutics, ACS Nano. 8 (2014) 9358–9367, https://
chemotherapy by modulating tumor microenvironment, Biomaterials. 108 (2016) doi.org/10.1021/nn503349g.
44–56, https://doi.org/10.1016/j.bio materials.2016.09.001. [101] H. Liang, X. Ren, J. Qian, X. Zhang, L. Meng, X. Wang, L. Li, X. Fang, X. Sha, Size-
[80] S. An, X. Jiang, J. Shi, X. He, J. Li, Y. Guo, Y. Zhang, H. Ma, Y. Lu, C. Jiang, Single- shifting micelle nanoclusters based on a cross-linked and pH-sensitive framework
component self-assembled RNAi nanoparticles functionalized with tumor-tar- for enhanced tumor targeting and deep penetration features, ACS Appl Mater
geting iNGR delivering abundant siRNA for efficient glioma therapy, Biomaterials. Interfaces. 8 (2016) 10136–10146, https://doi.org/10.1021/acsami.6b00668.
3 (2015) 330–340, https://doi.org/10.1016/j.biomaterials.2015.02.084. [102] J. Liu, X. Guo, Z. Luo, J. Zhang, M. Li, K. Cai, Hierarchically stimuli-responsive
[81] T. Kang, X. Gao, Q. Hu, D. Jiang, X. Feng, X. Zhang, Q. Song, L. Yao, M. Huang, nanovectors for improved tumor penetration and programed tumor therapy,
X. Jiang, Z. Pang, H. Chen, J. Chen, iNGR-modified PEG-PLGA nanoparticles that Nanoscale. 10 (2016) 13737–13750, https://doi.org/10.1039/c8nr02971g.
recognize tumor vasculature and penetrate gliomas, Biomaterials. 35 (2014) [103] Q. Chen, H. Liang, Y. Sun, Y. Chen, W. He, X. Fang, X. Sha, J. Li, A carbohydrate
4319–4332, https://doi.org/10.1016/j.bio materials.2014.01.082. mimetic peptide modified size-shrinkable micelle nanocluster for anti-tumor tar-
[82] J.E. Zhou, J. Yu, L. Gao, L. Sun, T. Peng, J. Wang, J. Zhu, W. Lu, L. Zhang, Z. Yan, geting and penetrating drug delivery, Int J Nanomedicine. 14 (2019) 7339–7352,
L. Yu, iNGR-modified liposomes for tumor vascular targeting and tumor tissue https://doi.org/10.2147/IJN.S213455.
penetrating delivery in the treatment of glioblastoma, Mol Pharm. 14 (2017) [104] Y. Yu, X. Zhang, L. Qiu, The anti-tumor efficacy of curcumin when delivered by
1811–1820, https://doi.org/10.1021/acs.molpharm aceut.7b00101. size/charge-changing multistage polymeric micelles based on amphiphilic poly (β-
[83] M. Paškevičiūtė, V. Petrikaitė, Overcoming transporter-mediated multidrug re- amino ester) derivates, Biomaterials. 35 (2014) 3467–3479, https://doi.org/10.
sistance in cancer: failures and achievements of the last decades, Drug Deliv Transl 1016/j.bio materials.2013.12.096.
Res. 9 (2019) 379–393, https://doi.org/10.1007/s13346-018-0584-7. [105] Y. Yu, L. Qiu, Optimizing particle size of docetaxel-loaded micelles for enhanced
[84] J.M. Escoffre, J. Piron, A. Novell, A. Bouakaz, Doxorubicin delivery into tumor treatment of oral epidermoid carcinoma, Nanomedicine. 12 (2016) 1941–1949,
cells with ultrasound and microbubbles, Mol Pharm. 8 (2011) 799–806, https:// https://doi.org/10.1016/j.nano.2016.04.012.
doi.org/10.1021/mp100397p. [106] J. Chen, J. Ding, Y. Wang, J. Cheng, S. Ji, X. Zhuang, X. Chen, Sequentially re-
[85] S.J. Grainger, J.V. Serna, S. Sunny, Y. Zhou, C.X. Deng, M.E. El-Sayed, Pulsed sponsive shell-stacked nanoparticles for deep penetration into solid tumors, Adv
ultrasound enhances nanoparticle penetration into breast cancer spheroids, Mol Mater. 29 (2017), https://doi.org/10.1002/adma.201701170.
Pharm. 7 (2010) 2006–2019, https://doi.org/10.1021/mp100280b. [107] P.E. Saw, H. Yao, C. Lin, W. Tao, O.C. Farokhzad, X. Xu, Stimuli-responsive
[86] C.Y. Lin, J.R. Li, H.C. Tseng, M.F. Wu, W.L. Lin, Enhancement of focused ultra- polymer-prodrug hybrid nanoplatform for multistage siRNA delivery and combi-
sound with microbubbles on the treatments of anticancer nanodrug in mouse tu- nation cancer therapy, Nano Lett. 19 (2019) 5967–5974, https://doi.org/10.
mors, Nanomedicine. 8 (2012) 900–907, https://doi.org/10.1016/j.nano.2011. 1021/acs.nano lett.9b01660.
10.005. [108] B. Ma, W. Zhuang, H. Xu, G. Li, Y. Wang, Hierarchical responsive nanoplatform
[87] C.Y. Lin, H.C. Tseng, H.R. Shiu, M.F. Wu, C.Y. Chou, W.L. Lin, Ultrasound soni- with two-photon aggregation-induced emission imaging for efficient cancer ther-
cation with microbubbles disrupts blood vessels and enhances tumor treatments of anostics, ACS Appl Mater Interfaces. (2019 Dec 9), https://doi.org/10.1021/
anticancer nanodrug, Int J Nanomedicine. 7 (2012) 2143–2152, https://doi.org/ acsami.9b17587.
10.2147/IJN.S 29514. [109] C. Ju, R. Mo, J. Xue, L. Zhang, Z. Zhao, L. Xue, Q. Ping, C. Zhang, Sequential intra-
[88] H. Lee, J. Han, H. Shin, H. Han, K. Na, H. Kim, Combination of chemotherapy and intercellular nanoparticle delivery system for deep tumor penetration, Angew
photodynamic therapy for cancer treatment with sonoporation effects, J Control Chem Int Ed Engl. 53 (2014) 6253–6258, https://doi.org/10.1002/anie.
Release. 283 (2018) 190–199, https://doi.org/10.1016/j.jconrel.2018.06.008. 201311227.
[89] T.Y. Wang, J.W. Choe, K. Pu, R. Devulapally, S. Bachawal, S. Machtaler, [110] S. Ruan, X. Cao, X. Cun, G. Hu, Y. Zhou, Y. Zhang, L. Lu, Q. He, H. Gao, Matrix
S.M. Chowdhury, R. Luong, L. Tian, B. Khuri-Yakub, J. Rao, R. Paulmurugan, metalloproteinase-sensitive size-shrinkable nanoparticles for deep tumor pene-
J.K. Willmann, Ultrasound-guided delivery of microRNA loaded nanoparticles into tration and pH triggered doxorubicin release, Biomaterials. 60 (2015) 100–110,
cancer, J Control Release. 203 (2015) 99–108, https://doi.org/10.1016/j.jconrel. https://doi.org/10.1016/j.biomaterial s.2015.05.006.
2015.02.018. [111] C. Wong, T. Stylianopoulos, J. Cui, J. Martin, V.P. Chauhan, W. Jiang, Z. Popovic,
[90] G. Dimcevski, S. Kotopoulis, T. Bjånes, D. Hoem, J. Schjøtt, B.T. Gjertsen, R.K. Jain, M.G. Bawendi, D. Fukumura, Multistage nanoparticle delivery system
M. Biermann, A. Molven, H. Sorbye, E. McCormack, M. Postema, O.H. Gilja, A for deep penetration into tumor tissue, Proc Natl Acad Sci U S A. 108 (2011)
human clinical trial using ultrasound and microbubbles to enhance gemcitabine 2426–2431, https://doi.org/10.1073/pnas.1018382108.
treatment of inoperable pancreatic cancer, J Control Release. 243 (2016) [112] G. Hu, X. Chun, Y. Wang, Q. He, H. Gao, Peptide mediated active targeting and
172–181, https://doi.org/10.1016/j.j conrel.2016.10.007. intelligent particle size reduction-mediated enhanced penetrating of fabricated
[91] L. Li, T.L. ten Hagen, M. Bolkestein, A. Gasselhuber, J. Yatvin, G.C. van Rhoon, nanoparticles for triple-negative breast cancer treatment, Oncotarget. 6 (4125)
A.M. Eggermont, D. Haemmerich, G.A. Koning, Improved intratumoral nano- (2015) 8–41274, https://doi.org/10.18632/oncotarget.5692.
particle extravasation and penetration by mild hyperthermia, J Control Release. [113] F.H. Liu, C.Y. Hou, D. Zhang, W.J. Zhao, Y. Cong, Z.Y. Duan, Z.Y. Qiao, H. Wang,
167 (2013) 130–137, https://doi.org/10.1016/j.jc onrel.2013.01.026. Enzyme-sensitive cytotoxic peptide-dendrimer conjugates enhance cell apoptosis
[92] L. Li, T.L. ten Hagen, M. Hossann, R. Süss, G.C. van Rhoon, A.M. Eggermont, and deep tumor penetration, Biomater Sci. 6 (2018) 604–613, https://doi.org/10.
D. Haemmerich, G.A. Koning, Mild hyperthermia triggered doxorubicin release 1039/c7bm01182b.
from optimized stealth thermosensitive liposomes improves intratumoral drug [114] A. Ketkar-Atre, T. Struys, T. Dresselaers, M. Hodenius, I. Mannaerts, Y. Ni,
delivery and efficacy, J Control Release. 168 (2013) 142–150, https://doi.org/10. I. Lambrichts, L.A. Van Grunsven, M. De Cuyper, U. Himmelreich, In vivo hepa-
1016/j.jconrel.2013.03.011. tocyte MR imaging using lactose functionalized magnetoliposomes, Biomaterials.

236
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

35 (2014) 1015–1024, https://doi.org/10.1016/j.bio materials.2013.10.029. Y. Yatabe, H. Iwata, Occult breast cancer may originate from ectopic breast tissue
[115] Y. Liu, L. Li, L. Li, Z. Zhou, F. Wang, X. Xiong, R. Zhou, Y. Huang, Programmed present in axillary lymph nodes, Breast Cancer Res Treat. 172 (2018) 1–7, https://
drug delivery system based on optimized “size decrease and hydrophilicity/hy- doi.org/10.1007/s10549-018-4898-4.
drophobicity transformation” for enhanced hepatocellular carcinoma therapy of [138] H. Cabral, J. Makino, Y. Matsumoto, P. Mi, H. Wu, T. Nomoto, K. Toh, N. Yamada,
doxorubicin, Nanomedicine. 14 (2018) 1111–1122, https://doi.org/10.1016/j.na Y. Higuchi, S. Konishi, M.R. Kano, H. Nishihara, Y. Miura, N. Nishiyama,
no.2018.02.006. K. Kataoka, Systemic targeting of lymph node metastasis through the blood vas-
[116] G.L. Hu, Y. Wang, Q. He, H.L. Gao, Multistage drug delivery system based on cular system by using size-controlled nanocarriers, ACS Nano. 9 (2018)
microenvironment-responsive dendrimer–gelatin nanoparticles for deep tumor 4957–4967, https://doi.org/10.1021/nn5070259.
penetration, RSC Advances. 5 (2015) 85933–85937, https://doi.org/10.1039/ [139] Y. Xianyu, Z. Wang, X. Jiang, A plasmonic nanosensor for immunoassay via en-
C5RA18833D. zyme-triggered click chemistry, ACS Nano. 8 (2014) 12741–12747, https://doi.
[117] M. Han, M.Y. Huang-Fu, W.W. Guo, N.N. Guo, J. Chen, H.N. Liu, Z.Q. Xie, org/10.1021/nn 505857g.
M.T. Lin, Q.C. Wei, J.Q. Gao, MMP-2-sensitive HA end-conjugated poly (amido [140] L. Mei, J. Rao, Y. Liu, M. Li, Z. Zhang, Q. He, Effective treatment of the primary
amine) dendrimers via click reaction to enhance drug penetration in to solid tumor and lymph node metastasis by polymeric micelles with variable particle
tumor, ACS Appl Mater Interfaces. 9 (2017) 42459–42470, https://doi.org/10. sizes, J Control Release. 292 (2018) 67–77, https://doi.org/10.1016/j.jconrel.20
1021/acsami.7b 10098. 18.04.053.
[118] Y. Mi, J. Wolfram, C. Mu, X. Liu, E. Blanco, H. Shen, M. Ferrari, Enzy me-re- [141] J. Rao, L. Mei, J. Liu, X. Tang, S. Yin, C. Xia, J. Wei, D. Wan, X. Wang, Y. Wang,
sponsive multistage vector for drug delivery to tumor tissue, Pharmacol Res. 113 M. Li, Z. Zhang, Q. He, Size-adjustable micelles co-loaded with a chemo ther-
(2016) 92–99, https://doi.org/10.1016/j.phrs.2016.08.024. apeutic agent and an autophagy inhibitor for enhancing cancer treatment via in-
[119] Q. Yao, Y. Liu, L. Kou, Y. Tu, X. Tang, L. Zhu, Tumor-targeted drug delivery and creased tumor retention, Acta Biomater. 89 (2019) 300–312, https://doi.org/10.
sensitization by MMP2-responsive polymeric micelles, Nanomedicine. 19 (2019) 1016/j.actbio.2 019.03.022.
71–80, https://doi.org/10.1016/j.nano.2019.03.012. [142] X. Cun, S. Ruan, J. Chen, L. Zhang, J. Li, Q. He, H. Gao, A dual strategy to improve
[120] X. Sun, J. Zhang, C. Yang, Z. Huang, M. Shi, S. Pan, H. Hu, M. Qiao, D. Chen, the penetration and treatment of breast cancer by combining shrinking nano-
X. Zhao, Dual-responsive size-shrinking nanocluster with hierarchical disassembly particles with collagen depletion by losartan, Acta Biomater. 31 (2016) 186–196,
capability for improved tumor penetration and therapeutic efficacy, ACS Appl https://doi.org/10.1016/j.actbio.2015.1 2.002.
Mater Interfaces. 11 (2019) 11865–11875, https://doi.org/10.1021/acsami. [143] Y.H. Lai, C.S. Chiang, T.H. Kao, S.Y. Chen, Dual-drug nanomedicine with hydro-
8b21580. philic F127-modified magnetic nanocarriers assembled in amphiphilic gelatin for
[121] H. Yim, S.J. Park, Y.H. Bae, K. Na, Biodegradable cationic nanoparticles loaded enhanced penetration and drug delivery in deep tumor tissue, Int J Nanomedicine.
with an anticancer drug for deep penetration of heterogeneous tumours, 13 (2018) 3011–3026, https://doi.org/10.2147/IJN.S161314.
Biomaterials. 34 (2013) 7674–7682, https://doi.org/10.1016/j.biomaterials. [144] C. Xu, Y. Sun, Y. Yu, M. Hu, C. Yang, Z. Zhang, A sequentially responsive and
2013.06.058. structure transformable nanoparticle with a comprehensively improved ‘CAPIR
[122] M. Huo, W. Li, A.S. Chaudhuri, Y. Fan, X. Han, C. Yang, Z. Wu, X. Qi, Bio-stimuli- cascade’ for enhanced antitumor effect, Nanoscale. 11 (2019) 1177–1194, https://
responsive multi-scale hyaluronic acid nanoparticles for deepened tumor pene- doi.org/10.1039/c8nr08781d.
tration and enhanced therapy, Carbohydr Polym. 171 (2017) 173–182, https:// [145] M. Zhou, H. Huang, D. Wang, H. Lu, J. Chen, Z. Chai, S.Q. Yao, Y. Hu, Light-
doi.org/10.1016/j.carbpo l.2017.05.017. triggered PEGylation/dePEGylation of the nanocarriers for enhanced tumor pe-
[123] J. Dang, H. He, D. Chen, L. Yin, Manipulating tumor hypoxia toward enhanced netration, Nano Lett. 19 (2019) 3671–3675, https://doi.org/10.1021/acs.
photodynamic therapy (PDT), Biomater Sci. 5 (2017) 1500–11511, https://doi. nanolett.9b00737.
org/10.1039/c7bm 00392g. [146] Y. Li, Z. Zhao, H. Liu, J.P. Fetse, A. Jain, C.Y. Lin, K. Cheng, Development of a
[124] A. Oniszczuk, K.A. Wojtunik-Kulesza, T. Oniszczuk, K. Kasprzak, The potential of tumor-responsive nanopolyplex targeting pancreatic cancer cells and stroma, ACS
photodynamic therapy (PDT)-experimental investigations and clinical use, Biomed Appl Mater Interfaces. 11 (2019) 45390–45403, https://doi.org/10.1021/acsami.
Pharmacother. 83 (2016) 912–929, https://doi.org/10.1016/j.biopha.2016.07. 9b15116.
058. [147] Q. Guo, X. He, C. Li, Y. He, Y. Peng, Y. Zhang, Y. Lu, X. Chen, Y. Zhang, Q. Chen,
[125] C. Hu, X. Cun, S. Ruan, R. Liu, W. Xiao, X. Yang, Y. Yang, C. Yang, H. Gao, T. Sun, C. Jiang, Dandelion-like tailorable nanoparticles for tumor micro-
Enzyme-triggered size shrink and laser enhanced NO release nanoparticles for environment modulation, Adv Sci (Weinh). 6 (2019) 1901430, https://doi.org/10.
deep tumor penetration and combination therapy, Biomaterials. 168 (2018) 1002/a dvs.201901430.
64–75, https://doi.org/10.1016/j.biomaterials.2018.03.046. [148] X. Cun, J. Chen, M. Li, X. He, X. Tang, R. Guo, M. Deng, M. Li, Z. Zhang, Q. He,
[126] R. Liu, W. Xiao, C. Hu, R. Xie, H. Gao, Theranostic size-reducible and no donor Tumor-associated fibroblast-targeted regulation and deep tumor delivery of che-
conjugated gold nanocluster fabricated hyaluronic acid nanoparticle with optimal motherapeutic drugs with a multifunctional size-switchable nanoparticle, ACS
size for combinational treatment of breast cancer and lung metastasis, J Control Appl Mater Interfaces. 11 (2019) 39545–39559, https://doi.org/10.1021/acsami.
Release. 278 (2018) 127–139, https://doi.org/10.1016/j.jconrel.2018.04.005. 9b13957.
[127] W. Yu, X. He, Z. Yang, X. Yang, W. Xiao, R. Liu, R. Xie, L. Qin, H. Gao, Sequentially [149] F. Li, Y. Wang, W.L. Chen, D.D. Wang, Y.J. Zhou, B.G. You, Y. Liu, C.X. Qu,
responsive biomimetic nanoparticles with optimal size in combination with S.D. Yang, M.T. Chen, X.N. Zhang, Co-delivery of VEGF siRNA and etoposide for
checkpoint blockade for cascade synergetic treatment of breast cancer and lung enhanced anti-angiogenesis and anti-proliferation effect via multi-functional na-
metastasis, Biomaterials. 217 (2019) 119309, , https://doi.org/10.1016/j. noparticles for orthotopic non-small cell lung cancer treatment, Theranostics. 9
biomater ials.2019.11 9309. (2019) 5886–5898, https://doi.org/10.7150/thno.32416.
[128] L. Yang, S. Zhang, X. Ling, P. Shao, N. Jia, M. Bai, Multilayer photodynamic [150] L. Chen, Y. Wu, H. Wu, J. Li, J. Xie, F. Zang, M. Ma, N. Gu, Y. Zhang, Magnetic
therapy for highly effective and safe cancer treatment, Acta Biomater. 54 (2017) targeting combined with active targeting of dual-ligand iron oxide nanoprobes to
271–280, https://doi.org/10.1016/j.actbio.2017.03.012. promote the penetration depth in tumors for effective magnetic resonance imaging
[129] C.L. Sun, X.Y. Li, X.D. Du, T. Wang, Redox-responsive micelles for triggered drug and hyperthermia, Acta Biomater. 96 (2019) 491–504, https://doi.org/10.1016/j.
delivery and effective laryngopharyngeal cancer therapy, Int J Biol Macromol. 112 actbio.2019.07.017.
(2018) 65–73, https://doi.org/10.1016/j.ijbiomac.2018.01.136. [151] A.A. Chiappori, S.G. Eckhardt, R. Bukowski, D.M. Sullivan, M. Ikeda, Y. Yano,
[130] H. Fan, G. Yan, Z. Zhao, X. Hu, W. Zhang, H. Liu, X. Fu, T. Fu, X.B. Zhang, W. Tan, T. Yamada-Sawada, Y. Kambayashi, K. Tanaka, M.M. Javle, T. Mekhail,
A smart photosensitizer-manganese dioxide nanosystem for enhanced photo- C.L. O'Bryant, P.J. Creaven, A Phase I Pharmacokinetic and Pharmacodynamic
dynamic therapy by reducing glutathione levels in cancer cells, Angew Chem Int Study of S-3304, a Novel Matrix Metalloproteinase Inhibitor, in Patients with
Ed. 55 (2016) 5477–5482, https://doi.org/10.1002/anie.201510748. Advanced and Refractory Solid Tumors, Clinical Cancer Research 13 (7) (2007)
[131] H. Bi, Y. Dai, P. Yang, J. Xu, D. Yang, S. Gai, F. He, B. Liu, C. Zhong, G. An, J. Lin, 2091–2099, https://doi.org/10.1158/1078-0432.CCR-06-1586.
Glutathione mediated size-tunable UCNPs-Pt (IV)-ZnFe2O4 nanocomposite for [152] S.R. Hingorani, L. Zheng, A.J. Bullock, T.E. Seery, W.P. Harris, D.S. Sigal,
multiple bioimaging guided Synergetic Therapy, Small. 14 (2018) e1703809, , F. Braiteh, P.S. Ritch, M.M. Zalupski, N. Bahary, P.E. Oberstein, A. Wang-Gillam,
https://doi.org/10.1002/smll. 2017 03809. W. Wu, D. Chondros, P. Jiang, S. Khelifa, J. Pu, C. Aldrich, A.E. Hendifar, HALO
[132] R. Tong, H.D. Hemmati, R. Langer, D.S. Kohane, Photoswitchable nano particles 202: Randomized phase II study of PEGPH20 plus nab-paclitaxel/gemcitabine
for triggered tissue penetration and drug delivery, J Am Chem Soc. 134 (2012) versus nab-paclitaxel/ gemcitabine in patients with untreated, metastatic pan-
8848–8855, https://doi.org/10.1021/ja211888a. creatic ductal adenocarcinoma, J Clin Oncol. 36 (2018) 359–366, https://doi.org/
[133] R. Tong, H.H. Chiang, D.S. Kohane, Photoswitchable nanoparticles for in vivo 10.1200/JCO.2017.74.9564.
cancer chemotherapy, Proc Natl Acad Sci U S A. 110 (2013) 19048–19053, [153] J.E. Murphy, J.Y. Wo, D.P. Ryan, J.W. Clark, W. Jiang, B.Y. Yeap, L.C. Drapek,
https://doi.org/10.1073/pnas.13 15336110. L. Ly, C.V. Baglini, L.S. Blaszkowsky, C.R. Ferrone, A.R. Parikh, C.D. Weekes,
[134] D. Moldenhauer, F. Gröhn, Nanoassemblies with light-responsive size and density R.D. Nipp, E.L. Kwak, J.N. Allen, R.B. Corcoran, D.T. Ting, J.E. Faris, A.X. Zhu,
from linear flexible polyelectrolytes, J. Polym. Sci. Pol. Phys. 51 (2013) 802–816, L. Goyal, D.L. Berger, M. Qadan, K.D. Lillemoe, N. Talele, R.K. Jain, T.F. DeLaney,
https://doi.org/10.1002/pol b.23276. D.G. Duda, Y. Boucher, C. Fernández-Del Castillo, T.S. Hong, Total neoadjuvant
[135] A. Barhoumi, Q. Liu, D.S. Kohane, Ultraviolet light-mediated drug delivery: therapy with FOLFIRINOX in combination with losartan followed by chemor-
principles, applications, and challenges, J Control Release. 219 (2015) 31–42, adiotherapy for locally advanced pancreatic cancer a phase 2 clinical trial, JAMA,
https://doi.org/10.1016/j.jconrel.20 15.07.018. Oncol. 5 (2019) 1020–1027, https://doi.org/10.1001/jamaoncol.2019.0892.
[136] Y. Min, J. Li, F. Liu, E.K. Yeow, B. Xing, Near-infrared light-mediated photo ac- [154] C.A. Gomez-Roca, A. Italiano, C. Le Tourneau, P.A. Cassier, M. Toulmonde,
tivation of a platinum antitumor prodrug and simultaneous cellular apoptosis ima S.P. D'Angelo, M. Campone, K.L. Weber, D. Loirat, M.A. Cannarile, A.M. Jegg,
ging by upconversion-luminescent nanoparticles, Angew Chem Int Ed Engl. 53 C. Ries, R. Christen, G. Meneses-Lorente, W. Jacob, I. Klaman, C.H. Ooi, C. Watson,
(2014) 1012–1016, https://doi.org/10.1002/anie.2013 08834. K. Wonde, B. Reis, F. Michielin, D. Rüttinger, J.P. Delord, J.Y. Blay, Phase I study
[137] M. Terada, Y. Adachi, M. Sawaki, M. Hattori, A. Yoshimura, G. Naomi, H. Kotani, of emactuzumab single agent or in combination with paclitaxel in patients with
M. Iwase, A. Kataoka, S. Onishi, K. Sugino, M. Mori, N. Horisawa, E. Sasaki, advanced/metastatic solid tumors reveals depletion of immunosuppressive M2-

237
F. Peng, et al. European Journal of Pharmaceutics and Biopharmaceutics 151 (2020) 220–238

like macrophages, Ann Oncol. 30 (2019) 1381–1392. 0121197.


[155] J.D. Berlin, Y. Feng, P. Catalano, J.L. Abbruzzese, P.A. Philip, R.R. McWilliams, A. [157] A. Carpentier, M. Canney, A. Vignot, V. Reina, K. Beccaria, C. Horodyckid,
M. Lowy, AB. III. Benson, A.W. Blackstock, An intergroup randomized phase II C. Karachi, D. Leclercq, C. Lafon, J.Y. Chapelon, L. Capelle, P. Cornu, M. Sanson,
study of bevacizumab or cetuximab in combination with gemcitabine and in K. Hoang-Xuan, J.Y. Delattre, A. Idbaih, Clinical trial of blood-brain barrier dis-
combination with chemoradiation in patients with resected pancreatic carcinoma: ruption by pulsed ultrasound, Sci Transl Med. 8 343re2 (2016), https://doi.org/
a trial of the ECOG-ACRIN cancer research group (E2204), Oncology. 94 (2018) 10.1126/scitranslmed. Aaf 6086.
39-46. doi: 10.1159/000480295. [158] E.L. Jones, J.R. Oleson, L.R. Prosnitz, T.V. Samulski, Z. Vujaskovic, D. Yu,
[156] G.J. Ullenhag, E. Rossmann, M. Liljefors, A phase I dose-escalation study of le- L.L. Sanders, M.W. Dewhirst, Randomized trial of hyperthermia and radiation for
nalidomide in combination with gemcitabine in patients with advanced pancreatic superficial tumors, J Clin Oncol. 23 (2005) 3079–3085, https://doi.org/10.1200/
cancer, PLoS One. 10 (2015) e0121197, , https://doi.org/10.1371/journal.pone. JCO.2005.0 5.520.

238

You might also like