You are on page 1of 13

Journal of Drug Delivery Science and Technology 55 (2020) 101348

Contents lists available at ScienceDirect

Journal of Drug Delivery Science and Technology


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

Delivery of erlotinib for enhanced cancer treatment: An update review on T


particulate systems
Duy Hieu Truonga,1, Vu Khanh Hoa Leb,1, Tung Thanh Phamc, Anh Hoang Daod,
Thi Phuong Dung Phame, Tuan Hiep Tranf,g,∗
a
Institute of Research and Development, Duy Tan University, Da Nang, Viet Nam
b
NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Viet Nam
c
College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
d
National Institute of Medicinal Materials, Hanoi, Viet Nam
e
Faculty of Pharmacy, Dai Nam University, Hanoi, Viet Nam
f
Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Viet Nam
g
Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Viet Nam

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

Keywords: Erlotinib is an epidermal growth factor receptor (EGFR) inhibitor that has been approved for metastatic non-
Erlotinib small cell lung cancer and highly aggressive pancreatic cancer. Although erlotinib contributes progressively to
EGFR cancer treatment, there is still room to develop a better platform for erlotinib-based therapy. The progression of
Nanoparticles the materials science field has offered many options to incorporate drugs with additional advantages. In this
Delivery systems
review, we summarize various strategies, including polymeric, lipidic, inorganic and hybrid-based carriers, to
Efficacy enhancement
Cancer
improve drug pharmacokinetics, reduce interpatient variability and minimize unwanted side effects. Moreover,
the combination of erlotinib with other therapeutic agents or modalities is further discussed as a potential
paradigm for realizing better efficacy and/or lower systemic toxicity in clinical practice.

1. Introduction poor solubility restricts the dissolution of the drug in the gastro-
intestinal fluid that can limit its absorption, thereby causing high
Erlotinib (ERL), a quinazoline derivative with the chemical name N- fluctuation in its bioavailability and large interpatient variability
(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine, is an [10,11]. Although the commercial tablet dosage form (Tarceva®) shows
FDA-approved small-molecule tyrosine kinase inhibitor for the treat- an acceptable bioavailability of approximately 60% in healthy volun-
ment of metastatic non-small cell lung cancer (NSCLC) and locally ad- teers upon oral administration of 150 mg [12], similar to most of the
vanced, unresectable or metastatic pancreatic cancer in combination compounds used for chemotherapy, patients receiving this therapy still
with gemcitabine [1]. By selectively occupying the adenosine tripho- suffer from drug resistance and severe toxicities, i.e., skin rash, diar-
sphate (ATP) binding sites of epidermal growth factor receptor (EGFR), rhea, gastrointestinal perforations and Stevens-Johnson syndrome, that
this molecule reversely inhibits EGFR activation, resulting in the sup- might hamper its clinical application [13]. These obstacles necessitate
pression of further downstream signaling processes, mostly through the the careful design of suitable delivery systems for not only improved
phosphatidylinositol 3-kinase (PI3K) and mitogen activated protein efficacy but also reduced side effects.
kinase (MAPK) pathways, and consequent inhibition of cellular pro- Recently, nanotechnology has been widely used to improve the
liferation, angiogenesis and metastases [2,3]. To date, the potential of physicochemical and pharmacokinetic properties of poorly water-so-
this substance has been expanded to treat other types of cancer, such as luble drugs, such as solubility, dissolution, absorption and bioavail-
ovarian cancer, head and neck cancer, and glioblastoma [4–8]. ability [14–16]. There have been many different techniques or strate-
As a substance with low aqueous solubility and high permeability gies that can be exploited for these purposes, e.g., spray drying, freeze
(logP of 2.7), ERL is categorized into class II in the biopharmaceutical drying, wet milling and high-pressure homogenization, to fabricate
classification system [9]. Regarding the oral route of administration, its various nanoformulations, such as solid dispersions, drug nanocrystals


Corresponding author. Ton Duc Thang University, Ho Chi Minh City, Viet Nam.
E-mail addresses: truongduyhieu@gmail.com (D.H. Truong), trantuanhiep@tdt.edu.vn (T.H. Tran).
1
These authors contributed equally to this manuscript.

https://doi.org/10.1016/j.jddst.2019.101348
Received 19 August 2019; Received in revised form 11 October 2019; Accepted 22 October 2019
Available online 23 October 2019
1773-2247/ © 2019 Elsevier B.V. All rights reserved.
D.H. Truong, et al.

Table 1
Nanoparticles for delivery of erlotinib as a single therapeutic agent.
Type Materials Experiments Results Ref.

1. Polymer-based particles poly(D,L-lactic-co-glycolic acid) (PLGA) in vitro Decrease of toxicity and damage to internal organs [24]
polycaprolactone - polyethylene glycol - poly caprolactone (PCEC) in vitro Increase of cytotoxicity in A549 NSCLC cells [25]
sulfobutyl ether β-cyclodextrin in vitro, in vivo Increase of dissolution and bioavailability by 2-fold and 3.6-fold, respectively [29]
cyclodextrin-based nanosponge in vitro, in vivo Increase of dissolution and bioavailability by 2-fold, and cytotoxic effects in MIA PaCa-2 and PANC-1 [30]
pancreatic cell lines
glutathione-nanosponge in vitro, in vivo Increase of cytotoxicity in A549 NSCLC cells and inhibition of tumor growth, and decrease distribution in [31]
vital organs
albumin-bound nanoparticles in vitro Increase of cytotoxicity in ASPC-1 and PANC-1 pancreatic cancer cells [33]
albumin-bound nanoparticles in vitro, in vivo Decrease of proliferation of A549 NSCLC cells and inhibition of tumor growth [34]
polyvinylpyrrolidone in vitro, in vivo Increase of dissolution, bioavailability and antitumor effect in A549 xenograft model, and decrease of [35]
fasted/fed variability

2. Lipid-based particles self-emulsifying drug delivery system in vitro, in vivo Increase of dissolution and bioavailability [36]
liposomes in vitro, in vivo Increase of cytotoxicity in A549 NSCLC cells and bioavailability [37]
solid lipid nanoparticles in vitro Increase of cytotoxicity in A549 NSCLC cells [38]
solid lipid nanoparticles in vitro Increase of cytotoxicity in A549 NSCLC cells [39]

2
molecular complex in vitro, in vivo Increase of cytotoxicity in MIA PaCa-2 and PANC-1 pancreas adenocarcinoma cells, solubility and [40]
bioavailability

3. Inorganic particles carbon-coated, iron magnetic nanoparticles in vitro Decrease of proliferation of PANC-1 pancreatic cells, especially under radio frequency [44]
ultra-small superparamagnetic iron oxide nanoparticles in vitro, in vivo Suppression of migration and invasion of CL1-5-F4 lung adenocarcinoma cells, and inhibition of tumor [46]
growth
ultra-small superparamagnetic iron oxide nanoparticles in vitro, in vivo Dose-dependent cytotoxicity in CL1-5-F4 NSCLC, and inhibition of tumor growth and NF-κB activity [47]
iron oxide-gold nanoclusters in vitro Increase of uptake and cytotoxicity in PANC-1 pancreatic cells [48]

4. Hybrid particles polymeric polycaprolactone; phospholipid in vitro Decrease of cellular viability in A549 NSCLC cells [49]
aptamer-conjugated chitosan; liposome in vitro Increase of antiproliferation, induction of cell cycle arrest and apoptosis in EGFR-mutated H1975 and PC- [50]
9 cells
poly(L-aspartic acid); poly(ethylene glycol)-b-poly(L-aspartic acid) (PEG-b- in vitro, in vivo Increase of uptake and cytotoxicity in HCC-827 and NCI-H358 lung cancer cells, and inhibition of tumor [57]
PAsp) growth
polymer-gold hybrid nanomaterial in vitro Increase of uptake and cytotoxicity in A549 NSCLC cells [58]
thermosensitive micelles in vitro Increase of cytotoxicity in OVCAR-3 ovarian carcinoma cells [59]
poly(acrylic acid)-cystamine-oleic acid (PAA-ss-OA) in vitro, in vivo Increase of cytotoxicity in A549 and NCI-H460 NSCLC cells, and antitumor efficacy in a lung cancer [60]
xenograft model
magnetic mesoporous silica nanoparticles; folic acid-poly(ethyleneimine) Increase of drug release at acidic pH by 2-fold and cytotoxicity in HeLa cervical carcinoma cells [61]
conjugate
myofibroblast-targeting nanoparticles in vitro, in vivo Sustained drug release over 12 days, and decrease of tumor burden by 7.3-fold and hepatocyte damage [62]
Journal of Drug Delivery Science and Technology 55 (2020) 101348
D.H. Truong, et al.

Table 2
Nanoparticles for delivery of erlotinib and other therapeutic agent/modalities.
Type Material Other active agents/modalities Experiments Results Ref.

1. Hybrid particles solid lipid core nanocapsules; polymer shell paclitaxel in vitro Increase of cytotoxicity in NCI-H23 cells [65]
aptamer-conjugated chitosan; liposomes perfluorooctylbromide in vitro, in vivo Increase of proliferation inhibition and induction of apoptosis of [67]
NSCLC cells, and significant decrease of tumor growth
aptamer-conjugated chitosan; liposomes survivin-recombinant short hairpin RNA- in vitro, in vivo Increase of gene transfection efficiency and toxicity in EGFR mutated [68]
expressing plasmid; chloroquine NSCLC cells, and inhibition of tumor growth
liposomes; hydrophilic thermally oxidized porous silicon doxorubicin; in vitro Synergistic effects on killing DOX-resistant MCF-7 breast cancer cells [70]
nanoparticles 17-AAG; DNA nanostructures; nano-magnetite;
gold nanorods
amino-functionalized mesoporous silica nanoparticles; 1,5- doxorubicin in vitro, in vivo Increase of proliferation and apoptosis in A549 NSCLC cells, and [82]
dioctadecyl-L-glutamyl 2-histidyl-hexahydrobenzoic acid inhibition of tumor growth
(HHG2C18)
gold nanocages; poly(acrylic acid); poly(N-isopropylacrylamide- doxorubicin in vitro, in vivo Synergistic tumor-killing effect in vitro and in vivo with stronger [84]

3
co-acrylamide) efficacy in highly EGFR-expressing A431 cells than in weakly EGFR-
expressing MCF-7 cells

2. Lipid-based liquisolid formulation valproic acid in vitro, in vivo Increase of cytotoxicity in ERL-resistant HCC827 lung adenocarcinoma [66]
particles cells and bioavailability
liposomes doxorubicin in vitro, in vivo Increase of apoptosis, decrease of EGFR expression in A549 NSCLC and [81]
BT-20 triple-negative breast cancer cells, and inhibition of tumor
growth

3. Polymer-based chitosan a heptamethine cyanine derivative in vitro, in vivo Increase of reactive oxygen species levels and tumor suppressor p53, [69]
particles and decrease of survivin expression in EGFR mutated NSCLC cells and
tumor growth
poly(L-lactide)-b-polyethylene glycol fedratinib in vitro, in vivo Increase of cytotoxicity in two ERL-resistant lung adenocarcinoma cell [75]
lines and tumor growth suppression in H1650 tumor-bearing mice
poly(L-lactide) DAPT in vitro, in vivo Increase of cytotoxicity in MDA-MB-231 triple-negative breast cancer [76]
cells, and inhibition of tumor growth
poly(L-lactide)-b-polyethylene glycol doxorubicin in vitro, in vivo Increase of cytotoxicity in MDA-MB-468 breast cancer cells and [83]
accumulation in tumor sites
Journal of Drug Delivery Science and Technology 55 (2020) 101348
D.H. Truong, et al. Journal of Drug Delivery Science and Technology 55 (2020) 101348

and solid lipid nanoparticles (SLNs) [17–20]. What if these technologies inhibition in a xenograft mouse model of lung cancer with significantly
can enhance the properties of ERL? What is the outcome of these lower biodistribution in the spleen, heart, kidney and liver resulting
changes? What is the potential of those interventions for clinical ap- from the targeting ability of glutathione. Taken together, these results
plication? To answer these questions, in this review, we summarize suggested that complexation with β-cyclodextrin derivatives could be
several approaches to improve the therapeutic efficacy of ERL as a an effective method to enhance the efficacy of ERL with reduced side
therapeutic agent alone (Table 1) and in combination with other ther- effects.
apeutic compounds or modalities (Table 2) using various drug delivery Albumin has been proven as a safe vehicle that is approved in many
systems, mainly focusing on nanocarriers. clinical products for drug delivery and bioimaging purposes, the most
notable of which is Abraxane® (paclitaxel albumin-bound particles for
2. Drug delivery systems containing erlotinib alone injectable suspension) [32]. By using the same concept, Noorani and
colleagues synthesized ERL-loaded albumin NPs by desolvation fol-
2.1. Polymer-based systems lowed by a thermal cross-linking method [33]. The use of an aqueous
solution of albumin at pH 5 and an acetone/dimethyl sulfoxide mixture
With the advance of chemical engineering, polymers, a large group with a 5:1 (v/v) ratio as the organic solvent resulted in a high drug
of compounds with versatile physical and chemical properties, can be loading capacity (approximately 27%) of ERL in the albumin NPs with
tuned to make nanocarriers for controlling the temporal or spatial re- hydrophobic interactions and hydrogen bonds between the two com-
lease of therapeutic agents [21]. Among the different types of polymers, ponents. In both the ASPC-1 and PANC-1 pancreatic cancer cell lines,
biodegradable polymers, such as poly(D,L-lactic-co-glycolic acid) the ERL-loaded albumin NPs demonstrated higher cytotoxicity than
(PLGA) and polycaprolactone-derived copolymers, are preferable to use that of free drug. In another work, Shen and Li coated ERL albumin-
when designing delivery systems because of their high safety profiles bound NPs with a hyaluronic acid layer to specifically target CD44-
and controlled release capability of cargo [22,23]. To evaluate the overexpressing tumor cells [34]. Due to the presence of the coating
safety of PLGA for delivering ERL, this drug was loaded into PLGA layer, the NPs showed excellent stability after 3 months and slower
nanoparticles (NPs), and then the toxicity of these NPs following oral drug release than their uncoated counterparts. Additionally, compared
administration in Wistar rats was assessed [24]. Compared to free ERL, to the uncoated NPs and free ERL, the NPs whose surface was modified
these ERL-encapsulated NPs showed significantly lower toxicity and by hyaluronic acid exhibited significantly enhanced uptake into A549
damage to internal organs, as confirmed by hematological and biolo- lung cancer cells, thereby further suppressing their proliferation. Im-
gical analyses and histopathological examination. In another study, portantly, the coated NPs demonstrated remarkable tumor growth in-
polycaprolactone - polyethylene glycol - polycaprolactone (PCEC) tri- hibition in tumor-bearing mice without relapse within 30 days post-
block copolymers were used to prepare polymeric NPs in which ERL treatment.
was incorporated into their lipophilic cores [25]. The results showed In another effort to enhance the oral bioavailability of ERL and
that the molar ratios of the polycaprolactone and polyethylene glycol reduce the variation in bioavailability between the fasted and fed states,
monomers defined the characteristics of these NPs. In particular, the a novel nanoparticulation method using fat and supercritical fluid
higher the ratio was, the larger the NP size and encapsulation efficacy (NUFS™) technology was applied [35]. In this method, ERL was melted
of ERL, and the lower release rate of ERL from the NPs. Drug-en- with a mixture of myristyl alcohol and polyvinylpyrrolidone at 130 °C
capsulated NPs showed remarkably higher cytotoxicity than that of the before being rapidly cooled to 15 °C for the preparation of nanoparti-
free drug, especially at higher doses and longer incubation periods, in culated ERL. Then, solidified myristyl alcohol was removed by super-
A549 lung cancer cells. critical CO2 to obtain the formulation as a dried powder. This for-
For a long period of time, cyclodextrins have been used to make mulation showed higher dissolution rates under both fasted and fed
inclusion complexes with various drug compounds to improve their conditions and a 5.5-fold higher AUC in the fasted state than did Tar-
solubility and bioavailability [26–28]. However, natural cyclodextrins ceva. Additionally, the ratio of fasted AUC to fed AUC for the for-
are not highly soluble in aqueous solutions, which limits their practical mulation was 1:1.8, which was significantly lower than that of Tarceva
applications as delivery vehicles. To overcome this hurdle, Suresh's (1:5.8). In particular, compared to Tarceva, the formulation markedly
group employed one of the more hydrophilic cyclodextrin derivatives, improved antitumor efficacy in the A549 xenograft tumor model, which
sulfobutyl ether β-cyclodextrin, to formulate an inclusion complex with likely resulted from the enhanced dissolution and potent inhibition of
ERL by the freeze drying method [29]. By using the design of experi- EGFR signaling.
ments technique, the molar ratio of 1.05:1 between ERL and sulfobutyl
ether β-cyclodextrin was found to be the optimal ratio to easily prepare 2.2. Lipid-based systems
an inclusion complex. Compared to free ERL, this formulation demon-
strated more than 2-fold higher dissolution at 60 min and 3.6-fold Lipid-based delivery carriers have demonstrated great promise in
higher AUC0-∞. Later, this group utilized a β-cyclodextrin-based na- enhancing the delivery of poorly water-soluble drugs, with numerous
nosponge, which was prepared by cross-linking a β-cyclodextrin successful commercial products, such as Neoral® (self-emulsifying for-
monomer and the cross-linker carbonyldiimidazole, for complexing mulations of cyclosporine), Onivyde > ® (liposomal irinotecan) and
with ERL [30]. After performing some preliminary solubility experi- AmBisome® (liposomal amphotericin B). Given the similarity in solu-
ments, the formulation with the optimum ERL and nanosponge ratio of bility between these drugs and ERL, the abovementioned products
1:4 was prepared and characterized. In comparison to pure ERL, this suggest several potential options of lipid carriers that can be used to
optimized formula approximately doubled the dissolution rate and oral develop ERL-containing formulations with improved profiles.
bioavailability and exhibited more cytotoxic effects on the MIA PaCa-2 Our work sought to utilize a self-emulsifying formulation to deliver
and PANC-1 pancreatic cancer cell lines. For lung-specific delivery of ERL [36]. By using the ternary phase diagram, we chose the optimized
ERL, Momin et al. synthesized glutathione nanosponges through a high- formulation, consisting of 5% w/w of oil (linoleoyl polyoxyl-6 glycer-
yielding reaction among β-cyclodextrin, 2-hydroxyethyl disulfide and ides or Labrafil® M2125CS), 65% w/w of surfactant (caprylocaproyl
the cross-linker pyromellitic dianhydride [31]. ERL-loaded glutathione polyoxyl-8 glycerides or Labrasol®) and 30% w/w of cosolvent (die-
nanosponges were able to show a sustained release pattern of ERL (up thylene glycol monoethyl ether or Transcutol® HP), to dissolve ERL and
to 7 days), which was proportionally dependent on the concentrations then solidified the resultant solution by spray-drying with dextran 40 or
of glutathione in the surrounding environment. The drug-loaded glu- Aerosil® 200 as the inert carrier. The incorporation of ERL into these
tathione nanosponges not only were markedly more cytotoxic in A549 formulations in the amorphous form or as a molecular dispersion re-
NSCLC cells than free ERL but also demonstrated higher tumor growth markably improved its dissolution and bioavailability compared to

4
D.H. Truong, et al. Journal of Drug Delivery Science and Technology 55 (2020) 101348

Fig. 1. (A) Dual functional FeDC-E NPs may serve both therapeutic effect and diagnostic utility in the same time in NSCLC. Reprinted with permission from Ref. [47].
(B) Schematic representation of the formation of Fe3O4@AuNCs@ERL nanocomposite. Reprinted with permission from Ref. [48].

those of the free drug. were approximately 10% and 4.5–6.7 μm, respectively, similar to those
Zhou and coworkers incorporated ERL into the hydrophobic layer of of commercial products, demonstrating the suitability of these inhal-
a PEGylated liposomal formulation by the thin-film hydration method able formulations for lung cancer treatment.
[37]. In contrast to non-PEGylated ERL-loaded liposomes, the PEGy- In addition to using polymers to improve ERL's efficacy [29,30],
lated ERL-loaded liposomes exhibited slightly higher cellular cytotoxi- Suresh's group also cooperated with Jain's group to employ phospha-
city and a 2-fold increase in bioavailability without causing any ap- tidyl choline for developing a molecular complex with ERL [40]. A
parent damage to tissues of vital internal organs owing to the enhanced drug-to-phospholipid ratio of 1–2 promoted stable complexation where
permeation and retention effect. the drug was engulfed by the phospholipid, thereby transforming the
The most well-known indication of ERL is to treat NSCLC; hence, drug into a less crystalline phase and improving its hydrophilicity,
direct delivery of ERL to lung cancer tissues and cells is highly desir- which led to 5.4-fold and 1.7-fold increases in the aqueous solubility
able. For that purpose, a microparticulate formulation of inhalable dry and oral bioavailability of ERL, respectively. Along with exhibiting
powder (IDP) containing ERL-encapsulated SLNs was prepared [38]. higher cytotoxicity and apoptotic index in two human pancreas ade-
The optimal SLNs, composed of Compritol® 888 ATO, Tween® 80 and nocarcinoma cell lines (i.e., MIA PaCa-2 and PANC-1) owing to efficient
Poloxamer 407 at a weight ratio of 24:1:20, were spherical particles internalization, the ERL-phospholipid complex also demonstrated
with sizes of less than 100 nm. The ERL-encapsulated SLNs showed better safety profiles in blood and normal tissues than those of free ERL,
enhanced cytotoxic effects of ERL on A549 human NSCLC cells. For the which might result from the protective and free radical scavenging ef-
delivery of ERL to deep lung tissues, the ERL-loaded SLNs were spray- fects of the phospholipid.
dried into mannitol, forming microparticles (1–5 μm) with suitable
flowability and aerodynamic properties, as confirmed by Hausner ratio, 2.3. Inorganic systems
Carr's index and the results of the pharmacopoeial Next Generation
Impactor® (NGI) test. Another SLN-loaded IDP for the pulmonary de- Magnetic NPs have versatile applications in cancer therapy, such as
livery of ERL was developed by Naseri et al. using different components diagnosis, drug delivery and theranostics [41–43]. Xu et al. prepared
[39]. The oily phase, which was composed of glycerol monostearate, carbon-coated iron magnetic NPs to combine the effect of hyperthermal
Transcutol®, poloxamer 188 and ERL, was mixed in distilled water to therapy induced by radio frequency with the effect of ERL attached
form SLNs. Similar to the abovementioned study, these drug-loaded onto the carbon shells [44]. Because of the strong affinity of the drug
SLNs showed higher cytotoxicity than that of the free drug in the for the magnetic NPs, as shown by the high loading capacity, only 30%
A549 cell line. The fine particle fraction (FPF) and mass median aero- of the drug was released at pH 7.4, with a slight increase to approxi-
dynamic diameter (MMAD) values of three mannitol-based powders mately 41% at pH 5.5 resulting from its protonation. Under a radio

5
D.H. Truong, et al. Journal of Drug Delivery Science and Technology 55 (2020) 101348

frequency of 350 kHz, the release of ERL from the NPs was also am- cells, and the effects on drug-resistant H1975 cells were much more
plified, especially in an acidic pH environment. In PANC-1 pancreatic pronounced than those on drug-sensitive PC-9 cells, supporting the
cells treated with the magnetic NPs, although radio frequency did not usage of the selected aptamer to overcome erlotinib resistance caused
significantly increase apoptosis, it halved cell proliferation to 25.4%. by EGFR mutation.
Theranostics, which integrate diagnostics and therapeutics into a Recently, tumor microenvironment-responsive NPs have attracted
single platform, have become increasingly important in clinical on- great attention in cancer therapy due to their capability to specifically
cology and personalized medicine [45]. Theranostic agents can si- target cancer sites by exploiting the different signals and conditions in
multaneously provide the results of diagnostic tests and monitor drug the tumor microenvironment compared to healthy tissues, such as
effects in real time for early disease detection and optimization of acidic pH and abnormal levels of enzymes, glutathione, and reactive
personal treatment. Considering the dual role of ERL as a therapeutic oxygen species [51–56]. Kim et al. used lipid nanocapsules with a PE-
and EGFR-targeting agent, Chen's laboratory designed a theranostic Gylated polypeptide shell, which can prolong systemic circulation, and
platform based on ERL-conjugated ultrasmall superparamagnetic iron a pH-responsive poly(L-aspartic acid) core, which can accelerate the
oxide nanoparticles (FeDC-E NPs) [46]. With ERL on the surface, the release of encapsulated cargo in acidic environments, for ERL delivery
FeDC-E NPs preferentially accumulated inside EGFR-overexpressing [57]. As expected, the lipid nanocapsules exhibited efficient cellular
cells, intracellularly released ERL and productively eradicated these uptake into cancer cells and higher drug release at pH 5.0 than at pH
cells. These conjugated NPs showed remarkable suppression of the 7.4. In addition, in comparison with the untreated control and free ERL,
EGFR-extracellular signal-regulated kinase-nuclear factor kappa B the ERL-encapsulated nanocapsules were dose-dependently cytotoxic to
(EGFR-ERK–NF–κB) signaling pathways, which subsequently resulted HCC-827 and NCI-H358 lung cancer cells and significantly retarded
in inhibition of the migration and invasion of CL1-5-F4 lung adeno- tumor growth in mice bearing an HCC-827 xenograft tumor.
carcinoma cells. Notably, in a xenograft-bearing mouse model, the Regarding the application of thermoresponsive NPs, Fathi et al.
FeDC-E NPs markedly decreased tumor growth and reduced the nor- exploited a widely used thermosensitive polymer, poly(N-iso-
malized T2-weighted MRI signal intensities within the tumor. Recently, propylacrylamide) (PNIPAAm), and chemosensitivity-induced oleic
this group combined the MRI technique with a nuclear factor kappa-B acid for developing a polymer-gold hybrid nanomaterial in which bio-
(NF-κB) reporter gene system to further evaluate the FeDC-E NP treat- degradable chitosan served as the reducing agent of auric salt (HAuCl4)
ment response for the potential clinical application of these NPs against and steric stabilizer for the resulting NPs [58]. In addition to demon-
NSCLC and to understand their roles in the apoptotic mechanisms strating excellent stability in the pH range from 3 to 8 for several
(Fig. 1A) [47]. They found that the cellular uptake and intratumoral months, the polymer-gold hybrid nanomaterials exhibited temperature-
distribution of this probe could be noninvasively monitored by using dependent release of ERL, in which drug release was clearly faster
ΔR2* values from MRI. Furthermore, because of the NF-κB reporter above the lower critical solution temperature. The efficient inter-
gene system, the apoptotic mechanism of the FeDC-E NPs was shown to nalization of the hybrid NPs into A549 cells (approximately 86% over
effectively suppress antiapoptotic effects through NF-κB signaling and 3 h of incubation) led to greater cytotoxicity in this cell line. To further
significantly induce intrinsic and extrinsic apoptotic pathways in vitro enhance the active targeting ability of thermosensitive micelles based
and in vivo. These results demonstrated the potential of this theranostic on PNIPAAm, this group modified the surface of these NPs with folic
probe for clinical translation in the treatment of lung cancer. acid [59]. As expected, the folate-modified micelles were significantly
To enhance the targeting to EGFR-overexpressing pancreatic cancer more cytotoxic to the folate receptor-positive OVCAR-3 ovarian carci-
cells and combine multimodal imaging techniques for early cancer di- noma cells than to the folate receptor-negative A549 lung adenocarci-
agnosis and treatment, ERL was conjugated to iron oxide-gold na- noma cells, which was consistent with enhanced internalization into the
noclusters to form a Fe3O4@AuNCs@ERL nanocomposite (Fig. 1B) OVCAR-3 cells. Additionally, drug-loaded surface-modified NPs de-
[48]. The core-shell nanocomposite improved the uptake and cyto- monstrated higher cytotoxicity than the corresponding free drug, sug-
toxicity of ERL in PANC-1 pancreatic cells owing to the appearance of gesting that receptor-mediated endocytosis of the targeted NPs might
ERL while still preserving the intense fluorescence property of the gold help the P-glycoprotein substrate ERL bypass the efflux mechanism
nanoclusters, which can be employed to monitor these cells before through this transporter.
surgery by confocal laser scanning microscopy imaging. To enhance ERL delivery to cancer sites using multiple stimuli, Tan
et al. prepared pH-sensitive and redox-responsive NPs (PAA-ERL-NPs)
2.4. Hybrid systems based on poly(acrylic acid)-cystamine-oleic acid (PAA-ss-OA), which
was formed through the conjugation among pH-sensitive poly(acrylic
Mandal et al. developed core-shell-type lipid–polymer hybrid NPs in acid), oleic acid and cystamine [60]. The outer layer composed of poly
which an ERL-incorporated polymeric polycaprolactone core was cov- (acrylic acid) decelerated drug release from the PAA-ERL-NPs, which
ered by a phospholipid monolayer [49]. Optimization of the formula- likely resulted from the fluid-phase endocytosis/pinocytosis of these
tion and process parameters resulted in an optimized formula with a NPs into endosomal compartments before releasing ERL and inducing
size of 170 nm, making it suitable for intravenous injection. The Cell- more cytotoxicity than that of free ERL and ERL-loaded nonlayered NPs
Titre-Glo® luminescent cell viability assay showed that in comparison in A549 and NCI-H460 NSCLC cells. Additionally, the PAA-ERL-NPs
with ERL solution, the ERL-loaded hybrid NPs remarkably reduced the exhibited the highest antitumor effect in a lung cancer xenograft mouse
viability of A549 cells with a 25-fold lower IC50 value (100 nM), which model, supporting the efficacy of the dual-targeting approach in lung
was consistent with the result of a colony formation assay. cancer treatment. In another study, a folic acid-poly(ethyleneimine)
As a strategy for active targeting of ERL to lung cancer cells with conjugate was attached to the surface of magnetic mesoporous silica
EGFR gene mutation—a phenomenon that can cause drug resistance NPs to prepare a pH-sensitive nanocarrier [61]. After four days, the
and limit long-term efficacy—Li et al. anchored EGFR-binding aptamer- percentage of ERL released from the coated nanocarrier at pH 5.5
conjugated chitosan onto liposomes, forming multifunctional liposomal (approximately 63%) was nearly double that at pH 7.4, confirming the
complexes [50]. The aptamer-modified complexes showed enhanced pH-responsive properties of the nanocarrier. Moreover, despite ex-
recognition and internalization by EGFR-mutated H1975 and PC-9 cells hibiting excellent biocompatibility with red blood cells, these coated
compared to EGFR-negative Helf cells and an increased drug release NPs demonstrated a significant cytotoxic effect on HeLa cervical car-
percentage in the acidic tumor microenvironment compared to that of cinoma cells owing to the overexpression of the folate receptor on this
their unmodified counterparts. These results likely contributed to the cell line.
observably increased antiproliferation and induction of cell cycle arrest To repurpose the use of the potent but toxic kinase inhibitor ERL
and apoptosis by the aptamer-modified complexes in these lung cancer from therapy to safer prevention of liver cancer, versatile

6
D.H. Truong, et al. Journal of Drug Delivery Science and Technology 55 (2020) 101348

Fig. 2. Myofibroblast-targeting nanoparticles. (A) Schematic presentation of erlotinib-loaded nanoparticles that target PDGFRB-expressing myofibroblasts (PPB-NP-
erlotinib). Erlotinib is released after cellular internalization and inhibits intracellular kinase domain of EGFR protein. In the 3D structural modeling, PPB fits the
extracellular domain of PDGFRB with close proximity to the site of PDGF ligand binding. (B) Morphology of the nanoparticles in rat plasma over time by TEM
imaging. Scale bar indicates 200 nm. (C) Size of nanoparticles in rat plasma over time measured on TEM images (median of 59 nanoparticles at each time point).
Boxes represent 75th and 25th percentile, horizontal line is median, and whiskers mark lowest and highest values. Outliers outside 1.5 × of inter-quartile range are
shown as open circles. (D) Concentration of erlotinib released from the nanoparticles over time in vitro in rat plasma. (E) Cumulative amount (%) of erlotinib released
from the nanoparticles over time in vitro in rat plasma. PDGFRB: platelet-derived growth factor receptor-β, PPB: PDGFRB-binding peptide, NP: nanoparticle, GMBS:
N–maleimidobutyryl-oxysuccinimide ester, PEG: polyethylene glycol, EGFR: epidermal growth factor receptor. Reprinted with permission from Ref. [62].

myofibroblast-targeting NPs were fabricated (Fig. 2) [62]. The proof of delivery to NSCLC cells [65]. The authors found that these two drugs
concept was to conjugate ERL-loaded NPs with a short peptide that can could be released in a pH-dependent and controlled manner. Ad-
specifically bind to platelet-derived growth factor receptor β (PDGFRB) ditionally, by using this delivery system, both drugs are efficiently
uniquely expressed on the surface of hepatic myofibroblasts. Due to the taken up by NCI-H23 cells, resulting in remarkably higher cytotoxicity
polyethylene glycol layer coating, the NPs showed a sustained release than that of the free drugs individually or in combination.
pattern of ERL with stable drug concentrations in rat plasma over the To prevent or delay acquired resistance to ERL, a liquisolid for-
period of 12 days (Fig. 2D and E), which is much longer than the half- mulation containing ERL and valproic acid was prepared by using
life of ERL (approximately 36 h) [1]. Compared to non-PGFRB-expres- polyethylene glycol 400 as the solvent [66]. With the assistance of
sing Hep G2 hepatoma cells, two types of PGFRB-expressing myofi- calcium silicate as a microporous adsorbent, the optimized formula, in
broblasts, TWNT-4 and LX-2 cells, demonstrated higher uptake of the which the molar ratio between ERL and valproic acid was 1:2, de-
targeted NPs and dose-dependent phospho-EGFR inhibition. In parti- monstrated a significant enhancement in the dissolution and bioavail-
cular, by using a cirrhosis-driven liver cancer model in rats, the authors ability of ERL owing to the ionic interaction between the components.
observed a significant decrease in tumor size by 7.3-fold and reduced Importantly, this formulation exhibited substantially higher cytotoxi-
hepatocyte damage in the group treated with the myofibroblast-tar- city and apoptosis percentage in ERL-resistant HCC827 lung adeno-
geting NPs compared with the group systemically treated with the free carcinoma cells than those of ERL alone by downregulating the ex-
drug at an equivalent dose. pression of survivin.
To reverse hypoxia-induced drug resistance in lung cancer, Gao's
laboratory codelivered ERL and perfluorooctylbromide—a high-capa-
3. Combination with other therapeutic agents or modalities city oxygen carrier—in aptamer-conjugated chitosan-anchored lipo-
somal complexes [67]. Owing to the presence of the anti-EGFR ap-
Combination therapies that combine two or more therapeutic agents tamer, these multifunctional NPs were specifically recognized by EGFR-
or modalities, such as photodynamic or photothermal therapies, possess positive A549, H1975 and PC-9 NSCLC cells, and compared to EGFR-
several advantages over monotherapies in cancer treatment. They are negative Helf cells, these EGFR-positive cells exhibited enhanced up-
able to enhance efficacy and reduce tumor resistance by simultaneously take of the NPs. While ERL-loaded complexes showed a lower total
targeting various anticancer mechanisms, decrease costs and effort to- percentage of ERL release under hypoxic conditions than under nor-
ward the development of new therapeutic compounds, minimize side moxic conditions, these coloaded complexes still demonstrated similar
effects, etc. [63,64]. release rates under both conditions, which might result from the pro-
Gupta et al. coloaded paclitaxel and ERL into the solid lipid core of motion of internal pressure by oxygen. In addition, the coloaded NPs
nanocapsules having a PEGylated block copolymer shell for dual-drug

7
D.H. Truong, et al. Journal of Drug Delivery Science and Technology 55 (2020) 101348

revealed strong effects on the inhibition of proliferation and induction distribution of the NPs in the lung.
of apoptosis of NSCLC cells and a significant inhibitory effect on the To overcome the multidrug resistance to DOX and other che-
expression of EGFR, p-EGFR and hypoxia-inducible factor-1α (HIF-1α), motherapeutics in cancer cells and to reduce drug side effects, giant
especially under hypoxic conditions. In vivo experiments showed pro- liposomes that are coloaded with three drugs (DOX, ERL and 17-AAG),
found tumor growth suppression and relatively lower distribution in the DNA nanostructures, nanomagnetite and gold nanorods were as-
spleen and heart in the groups of mice treated with the coloaded NPs sembled with hydrophilic thermally oxidized porous silicon NPs on a
compared to the other groups. Later, this laboratory attempted to microfluidic chip to generate a nano-in-micro platform [70]. The use of
overcome ERL resistance in EGFR mutation-positive NSCLC by sy- porous silicon NPs allowed high drug loading and sustained release of
nergizing the effects of chloroquine on normalizing the disturbed tumor the hydrophobic drug ERL as well as control over the drug ratio in the
vascular structure and downregulating the anti-apoptotic protein sur- liposomes, whereas the nanomagnetite and gold nanorods rendered the
vivin [68]. In this study, the authors coloaded survivin-recombinant platform magnetically and photothermally responsive, which might
short hairpin RNA-expressing plasmid (survivin-shRNA) with ERL into facilitate targeted drug delivery at tumor sites. In particular, the com-
anti-EGFR aptamer-modified polyamidoamine dendrimers. Chlor- bination of DNA nanostructures and the three drugs achieved sy-
oquine was shown to markedly amplify the intracellular uptake and nergistic effects on killing DOX-resistant MCF-7 breast cancer cells with
gene transfection efficiency of these coloaded nanocomplexes, thereby a significantly lower percentage of cell viability than that achieved with
enhancing their cytotoxicity in EGFR-mutated NSCLC cells. In addition, other combinations.
cotreatment with chloroquine and the nanocomplexes exhibited dra- Considering the possibility that resistance to EGFR inhibition can be
matic tumor suppression, which might partly stem from the decreased induced by the activation of Janus kinase 2 (JAK2)/signal transducer
tumor vessel leakage, improved tumor vessel maturation of and ele- and activator of transcription 3 (STAT3) signaling [71,72] or Notch
vated intratumoral drug concentration. Very recently, these authors signaling [73,74], the combination of ERL and JAK2 inhibitors or Notch
developed a nanotheranostic platform based on chitosan to encapsulate inhibitors might be a rational approach to overcome this resistance. In
ERL and a heptamethine cyanine derivative (Cy7) as a near-infrared one study, Chen et al. loaded both ERL and a JAK2 in-
photosensitizer for photodynamic therapy (Fig. 3) [69]. These NPs hibitor—fedratinib—into poly(L-lactide)-b-polyethylene glycol (PEG-b-
demonstrated high release of these two cargos in an acidic medium PLA) NPs [75]. Compared to single drug-loaded NPs, the dual drug-
with lysozyme and high uptake into PC-9 cells. With the combination of loaded NPs exerted significantly higher cytotoxic effects on two ERL-
near-infrared irradiation and the coloaded NPs, profound generation of resistant lung adenocarcinoma cell lines, H1975 and H1650, as well as
reactive oxygen species, marked upregulation of tumor suppressor p53 stronger tumor growth suppression ability in H1650 tumor-bearing
and downregulation of survivin expression were observed in three mice. In another study, Wan and colleagues coencapsulated ERL and a
subtypes of NSCLC cells, especially in PC-9 cells. Importantly, this sy- gamma secretase inhibitor—DAPT—into poly(L-lactide) (PLA)-based
nergistic combination significantly inhibited tumor growth in a mouse NPs [76]. To enhance their tumor targeting and penetrating abilities,
model implanted with PC-9 cells, which might result from the specific the prepared NPs were functionalized with a conjugate of a tumor-

Fig. 3. Schematic illustration of the preparation and the action of chitosan-based self-assembled nanoparticles CE7Ns. (a) Self-assembly and structural
composition of CE7Ns. The CE7Ns were composed of biodegradable polymer Cs, molecular targeted drug erlotinib, and NIRF dye Cy7 with PDT effects. (b) Schematic
illustration of the self-assembled nanoparticles as a robust nanoplatform for molecular targeted drug-mediated imaging of druggable mutations and combinational
therapy of drug-resistant lung tumor. Reprinted with permission from Ref. [69]. Copyright (2018) American Chemical Society.

8
D.H. Truong, et al. Journal of Drug Delivery Science and Technology 55 (2020) 101348

Fig. 4. Schematic illustration of preparation of erlotinib/DOX combination co-delivery nanocarriers and synergistic therapy of erlotinib and DOX. The surface of
mesoporous silica nanoparticles (A) were modified with 3-aminopropyltriethoxysilane (APTES) to form MSN-NH2 (B). MSN-NH2 were then loaded with DOX to
obtain DOX-loaded MSN-NH2 (C), followed by supported with erlotinib-loaded lipid bilayer (D) to yield M-HHG2C18-L(E + D) (E). After injection of M-HHG2C18-L
(E + D), the nanoparticles accumulated at the tumor site through enhanced permeability and retention effect in tumor blood vessels. M-HHG2C18-L(E + D) were
positively charged at extracellular environment (F) leading to easy internalization by tumor cells. After cell uptake, as erlotinib was loaded in the exterior lipid
bilayer and the controlled release ability of MSN-NH2, erlotinib released faster than DOX. Additionally, the lipid bilayer was more positive and induced a strong
repulsion with MSN-NH2 in intracellular environment (G), enhancing sequential staggered release of erlotinib and DOX, which was pretreated and staggered to
inhibit kinase domain of EGFR in cell membrane (H) and was targeted to cell nucleus (I) respectively, thus maximized the synergistic therapy. Effect of extracellular
and intracellular pH (pHe and pHi), respectively on the HHG2C18 (J). Reprinted with permission from Ref. [82].

homing peptide and a cell-penetrating peptide. Due to the presence of the uptake of the liposomes and DOX into folate receptor-over-
an acid-sensitive hydrazone linkage between these two peptides, the expressing A549 NSCLC and BT-20 triple-negative breast cancer cell
modified NPs showed the highest cellular uptake at an acidic pH of 6.0 lines. The core-shell design facilitated the release of ERL first and DOX
and the highest cytotoxicity in MDA-MB-231 triple-negative breast later from the dual drug-loaded liposomes, which demonstrated a sig-
cancer cells. Notably, among all mice groups tested, tumor-bearing nificant increase in apoptosis and prolonged inhibition of EGFR in these
mice injected with these modified NPs exhibited the lowest tumor size two cell lines compared to those induced by DOX-loaded liposomes.
without weight loss resulting from the highest tumor accumulation and Notably, although the dual drug combination reduced the tumor
lowest distribution in other organs. burden in A549 and BT-20 xenograft-bearing mice, the two drugs must
Long-term pretreatment with the EGFR inhibitor ERL was found to be packed in the same vehicle to achieve the desired efficacy. In another
significantly enhance or sensitize the efficacy of the DNA-damaging study, He et al. codelivered a combination of ERL and DOX by using a
agent doxorubicin (DOX) in NSCLC cells and triple-negative breast pH-sensitive charge conversion nanocarrier (designated as M-HHG2C18-
cancer cells [77] and in MCF-7 and MDA-MB-468 human breast cancer L) [82]. In this system, DOX was loaded into the core of amino-func-
cells [29]. Similar phenomena were also observed with other combi- tionalized mesoporous silica NPs (MSN-NH2), which were then shielded
nations of drug(s) and/or gene therapy in other cancer models [78–80], by an ERL-incorporated lipid bilayer to form M-HHG2C18-L(E + D)
suggesting that the efficacy of combination therapies does not depend (Fig. 4). Under the low pH in the tumor extracellular and intracellular
simply on their compositions but rather critically on the sequence of environments, the surface potential of the dual drug-loaded NPs shifted
their component's delivery. Bearing that in mind, to deliver both ERL from negative to positive due to the protonation of the zwitterionic
and DOX to the same tumor cells, Morton et al. encapsulated them in oligopeptide lipid in the external layer, thereby enhancing the inter-
the exterior hydrophobic layer and interior hydrophilic compartment of nalization of the NPs into cancer cells and improving the release of both
folate-coated liposomes, respectively [81]. The folate coating enhanced drugs. Notably, the sequential release of the two drugs remarkably

9
D.H. Truong, et al. Journal of Drug Delivery Science and Technology 55 (2020) 101348

Fig. 5. Mechanisms of acquired resistance to gefi-


tinib/erlotinib in EGFR‐mutated NSCLC. EGFR, epi-
dermal growth factor receptor; ErbB3, v‐erb‐b2 avian
erythroblastic leukemia viral oncogene homolog 3;
NSCLC, non‐small‐cell lung cancer; RTK, receptor
tyrosine kinase; MET, met proto‐oncogene; AXL, AXL
receptor tyrosine kinase; mAb, monoclonal antibody;
TKI, tyrosine kinase inhibitor; NF‐κB, nuclear factor
kappa‐light‐chain‐enhancer of activated B cells; AKT,
v‐akt murine thymoma viral oncogene homolog 1;
STAT, signal transducers and activation of tran-
scription; ERK, extracellular signal‐regulated kinase;
BIM, BCL2‐like 11 (apoptosis facilitator). Reprinted
with permission from Ref. [87] under a Creative
Commons Attribution 4.0 International License (CC
BY 4.0).

Table 3
Advantages and disadvantages associated with different types of ERL-loaded delivery systems.
Delivery systems Advantages Disadvantages

Polymeric NPs Enhance bioavailability Poor drug loading capacity


Enhance toxicity to tumor cells and reduce toxicity to internal organs Use of organic solvent in formulation

Lipidic NPs
Solid self-emulsifying formulation Enhance bioavailability Poor drug loading capacity
Improve storage stability
Liposomes Enhance bioavailability Poor storage stability
Ease of functionalization to enhance targeting and stability
Deliver both hydrophilic and hydrophobic agents
Solid lipid nanoparticles (SLNs) Use of biocompatible ingredients Poor storage stability
Ease of scale-up to industrial scale Poor drug loading capacity

Inorganic NPs Versatile applications (e.g. drug delivery, theranostics) Possible metabolism-associated issues
High cellular uptake
Biocompatibility

Hybrid NPs Deliver multiple therapeutic agents Poor storage stability


Ease of functionalization to enhance targeting and control drug release

synergized their antiproliferative and apoptotic effects on A549 cells gold nanocages loosened their shell because of the acidic pH and re-
and dramatically delayed tumor growth in tumor-bearing mice with an leased ERL. Then, after 6 h, under exogenous NIR irradiation, the gold
excellent safety profile. Recently, by using a different one-compartment nanocages generated heat to make the poly(N-isopropylacrylamide-co-
carrier design, Zhou et al. coloaded ERL and the hydrophobic form of acrylamide) shell collapse and release DOX. The combination of che-
DOX into poly(L-lactide)-b-polyethylene glycol (PLA-b-PEG) NPs after motherapy and phototherapy demonstrated a synergistic tumor-killing
ion-pairing of positively charged DOX with a negatively charged lipid, effect with more potent efficacy in highly EGFR-expressing A431 cells
1,2-dioleoyl-sn-glycero-3-phosphate [83]. This complexation sig- than in weakly EGFR-expressing MCF-7 cells in both in vitro and in vivo
nificantly retarded the release rate of DOX while exhibiting burst re- models attributed to the caspase-8 signaling pathway.
lease of ERL (approximately 80% in 4 h) that increased the efficacy of
the coloaded NPs against MDA-MB-468 breast cancer cells. Further- 4. Current challenges
more, an in vivo biodistribution study demonstrated that the polymeric
NPs considerably accumulated inside the tumor with marginal dis- Despite some initial clinical success, acquired (secondary) resistance
tribution in the major organs, confirming their efficacy and safety. To to erlotinib usually develops in most NSCLC cases after 10–13 months
control the time lag of drug administration even more precisely for [85]. The resistance can stem mainly from the acquisition of a sec-
optimal efficacy, two separate gold nanocages with pH-sensitive poly ondary EGFR mutation and amplification of the hepatocyte growth
(acrylic acid) and thermosensitive poly(N-isopropylacrylamide-co-ac- factor receptor (HGFR/c-MET) oncogene (Fig. 5) as well as various
rylamide) shells were developed to carry ERL and DOX, respectively conditions in the tumor microenvironment. First, the gatekeeper T790
[84]. Upon accumulation in the tumor microenvironment via the en- mutation is the most common type of mutation, occurring in approxi-
hanced permeability and retention effect, the poly(acrylic acid)-covered mately 50% of all resistant NSCLC cases, whereas amplification of c-

10
D.H. Truong, et al. Journal of Drug Delivery Science and Technology 55 (2020) 101348

MET accounts for approximately 20% of mutations [3,86,87]. Several [2] J.D. Moyer, E.G. Barbacci, K.K. Iwata, L. Arnold, B. Boman, A. Cunningham,
second-generation EGFR tyrosine kinase inhibitors (e.g., afatinib and C. DiOrio, J. Doty, M.J. Morin, M.P. Moyer, Induction of apoptosis and cell cycle
arrest by CP-358,774, an inhibitor of epidermal growth factor receptor tyrosine
dacomitinib) and third-generation inhibitors (e.g., osimertinib) that kinase, Cancer Res. 57 (21) (1997) 4838–4848.
bind irreversibly to EGFR and selectively to T790 M EGFR, respectively, [3] V.D. Cataldo, D.L. Gibbons, R. Pérez-Soler, A. Quintás-Cardama, Treatment of
and c-MET inhibitors (e.g., crizotinib) have been developed to over- non–small-cell lung cancer with erlotinib or gefitinib, N. Engl. J. Med. 364 (10)
(2011) 947–955.
come this type of resistance. Second, hypoxia, which refers to the low [4] H. Hirte, A. Oza, K. Swenerton, S. Ellard, R. Grimshaw, B. Fisher, M. Tsao,
oxygen levels in tumor tissues, and disturbed tumor tissue vasculature L. Seymour, A phase II study of erlotinib (OSI-774) given in combination with
are two major factors in the tumor microenvironment that substantially carboplatin in patients with recurrent epithelial ovarian cancer (NCIC CTG IND.
149), Gynecol. Oncol. 118 (3) (2010) 308–312.
limit drug penetration and antitumor activity in tumor tissues [5] E. Despierre, I. Vergote, R. Anderson, C. Coens, D. Katsaros, F.R. Hirsch, B. Boeckx,
[46,88,89]. These factors can be bypassed by delivering oxygen directly M. Varella-Garcia, A. Ferrero, I. Ray-Coquard, Epidermal growth factor receptor
into tumor sites and by using vascular normalization agents to restore (EGFR) pathway biomarkers in the randomized phase III trial of erlotinib versus
observation in ovarian cancer patients with no evidence of disease progression after
the functions of tumor vessels, respectively [67,68]. In addition, the
first-line platinum-based chemotherapy, Target. Oncol. 10 (4) (2015) 583–596.
upregulation and activation of some proteins, such as survivin, STAT3 [6] A.S.M. Anisuzzaman, A. Haque, D. Wang, M.A. Rahman, C. Zhang, Z. Chen,
and phosphoglycerate dehydrogenase (PHGDH), in the tumor micro- Z.G. Chen, D.M. Shin, A.R. Amin, In vitro and in vivo synergistic antitumor activity of
environment were found to be positively associated with drug re- the combination of BKM120 and erlotinib in head and neck cancer: mechanism of
apoptosis and resistance, Mol. Cancer Ther. 16 (4) (2017) 729–738.
sistance and poor prognosis in lung cancer [90–92]. This can be ad- [7] J. Raizer, P. Giglio, J. Hu, M. Groves, R. Merrell, C. Conrad, S. Phuphanich,
dressed by silencing these genes using specific short interfering RNAs or V.K. Puduvalli, M. Loghin, N. Paleologos, A phase II study of bevacizumab and
gene suppressors [68,90,92,93]. erlotinib after radiation and temozolomide in MGMT unmethylated GBM patients,
J. Neuro Oncol. 126 (1) (2016) 185–192.
[8] J.J. Raizer, L.E. Abrey, A.B. Lassman, S.M. Chang, K.R. Lamborn, J.G. Kuhn,
5. Authors’ outlook W.A. Yung, M.R. Gilbert, K.A. Aldape, P.Y. Wen, A phase II trial of erlotinib in
patients with recurrent malignant gliomas and nonprogressive glioblastoma mul-
tiforme postradiation therapy, Neuro Oncol. 12 (1) (2009) 95–103.
In this review, we have discussed several approaches for delivering [9] N. Budha, A. Frymoyer, G. Smelick, J. Jin, M. Yago, M. Dresser, S. Holden, L. Benet,
erlotinib by using numerous carrier systems to enhance its efficacy. J. Ware, Drug absorption interactions between oral targeted anticancer agents and
First, polymer-based, lipid-based, inorganic and hybrid systems were PPIs: is pH‐dependent solubility the Achilles heel of targeted therapy? Clin.
Pharmacol. Ther. 92 (2) (2012) 203–213.
used to improve erlotinib's characteristics, such as its aqueous solubi- [10] F. Thomas, P. Rochaix, M. White-Koning, I. Hennebelle, J. Sarini, A. Benlyazid,
lity, dissolution, bioavailability and toxicity. Second, NPs delivering L. Malard, J.-L. Lefebvre, E. Chatelut, J.P. Delord, Population pharmacokinetics of
combinations of erlotinib and other therapeutic agents or modalities erlotinib and its pharmacokinetic/pharmacodynamic relationships in head and
neck squamous cell carcinoma, Eur. J. Cancer 45 (13) (2009) 2316–2323.
were exploited to prevent the development of tumor resistance and
[11] A. Gruber, M. Czejka, P. Buchner, M. Kitzmueller, N.K. Baroian, C. Dittrich,
improve the effects of erlotinib. The advantages and disadvantages of A.S. Hrgovcic, Monitoring of erlotinib in pancreatic cancer patients during long-
delivery systems of erlotinib are summarized in Table 3. time administration and comparison to a physiologically based pharmacokinetic
These promising results encourage the translation of these nano- model, Cancer Chemother. Pharmacol. 81 (4) (2018) 763–771.
[12] J.R. Johnson, M. Cohen, R. Sridhara, Y.-F. Chen, G.M. Williams, J. Duan,
particulate systems into the clinic. Nevertheless, there are some chal- J. Gobburu, B. Booth, K. Benson, J. Leighton, Approval summary for erlotinib for
lenges that delay their appearance on the market. First, the large-scale treatment of patients with locally advanced or metastatic non–small cell lung
manufacturing of NPs according to GMP guidelines is a laborious and cancer after failure of at least one prior chemotherapy regimen, Clin. Cancer Res. 11
(18) (2005) 6414–6421.
expensive process because of the high costs of materials, low production [13] M. D'Arcangelo, F. Cappuzzo, Erlotinib in the first-line treatment of non-small-cell
yield, incomplete purification of the final products, etc. Second, in the lung cancer, Expert Rev. Anticancer Ther. 13 (5) (2013) 523–533.
preclinical phase, there is a need for developing standardized protocols [14] S. Kumar, N. Dilbaghi, R. Saharan, G. Bhanjana, Nanotechnology as emerging tool
for enhancing solubility of poorly water-soluble drugs, BioNanoSci 2 (4) (2012)
to evaluate the toxicity of NPs in vitro, in vivo and in animal models and 227–250.
to understand their interaction with cells/tissues/organs, etc. Third, [15] P. Chi Lip Kwok, H.-K. Chan, Nanotechnology versus other techniques in improving
due to the complex nature of NPs, there is a lack of appropriate gov- drug dissolution, Curr. Pharmaceut. Des. 20 (3) (2014) 474–482.
[16] M. Sharma, R. Sharma, D.K. Jain, Nanotechnology based approaches for enhancing
ernment regulations for nanoparticulate systems that would help guide
oral bioavailability of poorly water soluble antihypertensive drugs, Sci. Tech. Rep.
the manufacturing practices, quality control and design of optimal 2016 (2016).
clinical trials to evaluate their safety/toxicity and therapeutic efficacy [17] J. Hu, K.P. Johnston, R.O. Williams III, Nanoparticle engineering processes for
enhancing the dissolution rates of poorly water soluble drugs, Drug Dev. Ind.
in humans [94]. Moreover, patents and intellectual properties can be
Pharm. 30 (3) (2004) 233–245.
serious problems that halt the commercialization of nanomaterials be- [18] H. Chen, C. Khemtong, X. Yang, X. Chang, J. Gao, Nanonization strategies for poorly
cause their nomenclature is much more complicated than that of con- water-soluble drugs, Drug Discov. Today 16 (7–8) (2011) 354–360.
ventional products on the market. Therefore, by addressing all the [19] C.M. Keck, R.H. Müller, Drug nanocrystals of poorly soluble drugs produced by high
pressure homogenisation, Eur. J. Pharm. Biopharm. 62 (1) (2006) 3–16.
abovementioned issues, pharmaceutical companies might successfully [20] H. Bunjes, Lipid nanoparticles for the delivery of poorly water‐soluble drugs, J.
develop an oral formulation of erlotinib with improved pharmacoki- Pharm. Pharmacol. 62 (11) (2010) 1637–1645.
netics and toxicity profiles for enhanced clinical efficacy and patient [21] O. Pillai, R. Panchagnula, Polymers in drug delivery, Curr. Opin. Chem. Biol. 5 (4)
(2001) 447–451.
compliance. [22] H.K. Makadia, S.J. Siegel, Poly lactic-co-glycolic acid (PLGA) as biodegradable
controlled drug delivery carrier, Polymers 3 (3) (2011) 1377–1397.
Declaration of competing interest [23] D. Mondal, M. Griffith, S.S. Venkatraman, Polycaprolactone-based biomaterials for
tissue engineering and drug delivery: current scenario and challenges, Int. J. Polym.
Mat. Polym. Biomat. 65 (5) (2016) 255–265.
The authors declare no conflict of interest. [24] G. Marslin, C.J. Sheeba, V. Kalaichelvan, R. Manavalan, P. Neelakanta Reddy,
G. Franklin, Poly (D, L-lactic-co-glycolic acid) nanoencapsulation reduces Erlotinib-
induced subacute toxicity in rat, J. Biomed. Nanotechnol. 5 (5) (2009) 464–471.
Acknowledgements
[25] L. Barghi, D. Asgari, J. Barar, A. Nakhlband, H. Valizadeh, Synthesis, character-
ization and in vitro anti-tumoral evaluation of Erlotinib-PCEC nanoparticles, Asian
This research received no specific grant from any funding agency in Pac. J. Cancer Prev. APJCP 15 (23) (2014) 10281–10287.
[26] W. Saenger, Cyclodextrin inclusion compounds in research and industry, Angew
the public, commercial, or not-for-profit sectors.
Chem. Int. Ed. Engl. 19 (5) (1980) 344–362.
[27] J. Szejtli, Cyclodextrin Inclusion Complexes, Cyclodextrin Technology, Springer,
References 1988, pp. 79–185.
[28] M.E. Davis, M.E. Brewster, Cyclodextrin-based pharmaceutics: past, present and
future, Nat. Rev. Drug Discov. 3 (12) (2004) 1023–1035.
[1] U.S. Food and Drug Administration, Ful prescribing information of erlotinib [29] N. Devasari, C.P. Dora, C. Singh, S.R. Paidi, V. Kumar, M.E. Sobhia, S. Suresh,
(Tarceva), http://www.accessdata.fda.gov/drugsatfda_docs/label/2016/ Inclusion complex of erlotinib with sulfobutyl ether-β-cyclodextrin: preparation,
021743s025lbl.pdf , Accessed date: 25 September 2018. characterization, in silico, in vitro and in vivo evaluation, Carbohydr. Polym. 134

11
D.H. Truong, et al. Journal of Drug Delivery Science and Technology 55 (2020) 101348

(2015) 547–556. small molecules for effective chemotherapy in solid tumors, Acta Biomater. 48
[30] C.P. Dora, F. Trotta, V. Kushwah, N. Devasari, C. Singh, S. Suresh, S. Jain, Potential (2017) 131–143.
of erlotinib cyclodextrin nanosponge complex to enhance solubility, dissolution [56] T. Ramasamy, H.B. Ruttala, B. Gupta, B.K. Poudel, H.-G. Choi, C.S. Yong, J.O. Kim,
rate, in vitro cytotoxicity and oral bioavailability, Carbohydr. Polym. 137 (2016) Smart chemistry-based nanosized drug delivery systems for systemic applications: a
339–349. comprehensive review, J. Control. Release 258 (2017) 226–253.
[31] M.M. Momin, Z. Zaheer, R. Zainuddin, J.N. Sangshetti, Extended release delivery of [57] J. Kim, T. Ramasamy, J.Y. Choi, S.T. Kim, Y.S. Youn, H.-G. Choi, C.S. Yong,
erlotinib glutathione nanosponge for targeting lung cancer, Artif. Cells Nanomed. J.O. Kim, PEGylated polypeptide lipid nanocapsules to enhance the anticancer ef-
Biotechnol. 46 (5) (2018) 1064–1075. ficacy of erlotinib in non-small cell lung cancer, Colloids Surfaces B Biointerfaces
[32] F.-F. An, X.-H. Zhang, Strategies for preparing albumin-based nanoparticles for 150 (2017) 393–401.
multifunctional bioimaging and drug delivery, Theranostics 7 (15) (2017) [58] M. Fathi, P.S. Zangabad, J. Barar, A. Aghanejad, H. Erfan-Niya, Y. Omidi, Thermo-
3667–3689. sensitive chitosan copolymer-gold hybrid nanoparticles as a nanocarrier for de-
[33] M. Noorani, N. Azarpira, K. Karimian, H. Heli, Erlotinib-loaded albumin nano- livery of erlotinib, Int. J. Biol. Macromol. 106 (2018) 266–276.
particles: a novel injectable form of erlotinib and its in vivo efficacy against pan- [59] M. Fathi, P.S. Zangabad, A. Aghanejad, J. Barar, H. Erfan-Niya, Y. Omidi, Folate-
creatic adenocarcinoma ASPC-1 and PANC-1 cell lines, Int. J. Pharm. 531 (1) conjugated thermosensitive O-maleoyl modified chitosan micellar nanoparticles for
(2017) 299–305. targeted delivery of erlotinib, Carbohyd, Polymers 172 (2017) 130–141.
[34] Y. Shen, W. Li, HA/HSA co-modified erlotinib–albumin nanoparticles for lung [60] S. Tan, G. Wang, Redox-responsive and pH-sensitive nanoparticles enhanced sta-
cancer treatment, Drug Des. Dev. Ther. 12 (2018) 2285–2292. bility and anticancer ability of erlotinib to treat lung cancer in vivo, Drug Des. Dev.
[35] K.M. Yang, I.C. Shin, J.W. Park, K.-S. Kim, D.K. Kim, K. Park, K. Kim, Ther. 11 (2017) 3519–3529.
Nanoparticulation improves bioavailability of Erlotinib, Drug Dev. Ind. Pharm. 43 [61] N. Avedian, F. Zaaeri, M.P. Daryasari, H.A. Javar, M. Khoobi, pH-sensitive bio-
(9) (2017) 1557–1565. compatible mesoporous magnetic nanoparticles labeled with folic acid as an effi-
[36] D.H. Truong, T.H. Tran, T. Ramasamy, J.Y. Choi, H.H. Lee, C. Moon, H.-G. Choi, cient carrier for controlled anticancer drug delivery, J. Drug Deliv. Sci. Technol. 44
C.S. Yong, J.O. Kim, Development of solid self-emulsifying formulation for im- (2018) 323–332.
proving the oral bioavailability of erlotinib, AAPS PharmSciTech 17 (2) (2016) [62] M. Deshmukh, S. Nakagawa, T. Higashi, A. Vincek, A. Venkatesh, M.R. De
466–473. Galarreta, A.P. Koh, N. Goossens, H. Hirschfield, C.B. Bian, Cell type-specific
[37] X. Zhou, H. Tao, K.-H. Shi, Development of a nanoliposomal formulation of erlotinib pharmacological kinase inhibition for cancer chemoprevention, Nanomed.
for lung cancer and in vitro/in vivo antitumoral evaluation, Drug Des. Dev. Ther. 12 Nanotechnol. Biol. Med. 14 (2) (2018) 317–325.
(2018) 1–8. [63] J.A. DiMasi, R.W. Hansen, H.G. Grabowski, The price of innovation: new estimates
[38] Z. Bakhtiary, J. Barar, A. Aghanejad, A.A. Saei, E. Nemati, J. Ezzati Nazhad of drug development costs, J. Health Econ. 22 (2) (2003) 151–185.
Dolatabadi, Y. Omidi, Microparticles containing erlotinib-loaded solid lipid nano- [64] R.B. Mokhtari, T.S. Homayouni, N. Baluch, E. Morgatskaya, S. Kumar, B. Das,
particles for treatment of non-small cell lung cancer, Drug Dev. Ind. Pharm. 43 (8) H. Yeger, Combination therapy in combating cancer, Oncotarget 8 (23) (2017)
(2017) 1244–1253. 38022–38043.
[39] N. Naseri, P. Zakeri-Milani, H. Hamishehkar, Y. Pilehvar-Soltanahmadi, [65] B. Gupta, B.K. Poudel, S. Regmi, S. Pathak, H.B. Ruttala, M. Gautam, G.J. An, J.-
H. Valizadeh, Development, in vitro characterization, antitumor and aerosol per- H. Jeong, H.-G. Choi, C.S. Yong, Paclitaxel and erlotinib-co-loaded solid lipid core
formance evaluation of respirable prepared by self-nanoemulsification method, nanocapsules: assessment of physicochemical characteristics and cytotoxicity in
Drug Res. 67 (2017) 343–348. non-small cell lung cancer, Pharm. Res. 35 (5) (2018) 96.
[40] C.P. Dora, V. Kushwah, S.S. Katiyar, P. Kumar, V. Pillay, S. Suresh, S. Jain, [66] K. Patel, R. Doddapaneni, M. Patki, V. Sekar, A. Bagde, M. Singh, Erlotinib-valproic
Improved oral bioavailability and therapeutic efficacy of erlotinib through mole- acid liquisolid formulation: evaluating oral bioavailability and cytotoxicity in er-
cular complexation with phospholipid, Int. J. Pharm. 534 (1–2) (2017) 1–13. lotinib-resistant non-small cell lung cancer cells, AAPS PharmSciTech 20 (2019)
[41] P. Martinkova, M. Brtnicky, J. Kynicky, M. Pohanka, Iron oxide nanoparticles: in- 135.
novative tool in cancer diagnosis and therapy, Adv. Healthc. Mater. 7 (5) (2018) [67] F. Li, H. Mei, Y. Gao, X. Xie, H. Nie, T. Li, H. Zhang, L. Jia, Co-delivery of oxygen
1700932. and erlotinib by aptamer-modified liposomal complexes to reverse hypoxia-induced
[42] S. Fathi Karkan, M. Mohammadhosseini, Y. Panahi, M. Milani, N. Zarghami, drug resistance in lung cancer, Biomat 145 (2017) 56–71.
A. Akbarzadeh, E. Abasi, A. Hosseini, S. Davaran, Magnetic nanoparticles in cancer [68] T. Lv, Z. Li, L. Xu, Y. Zhang, H. Chen, Y. Gao, Chloroquine in combination with
diagnosis and treatment: a review, Artif. cell. Nanomed. Biotechnol. 45 (1) aptamer-modified nanocomplexes for tumor vessel normalization and efficient er-
(2017) 1–5. lotinib/Survivin shRNA co-delivery to overcome drug resistance in EGFR-mutated
[43] M. Arruebo, R. Fernández-Pacheco, M.R. Ibarra, J. Santamaría, Magnetic nano- non-small cell lung cancer, Acta Biomater. 76 (2018) 257–274.
particles for drug delivery, Nano Today 2 (3) (2007) 22–32. [69] Y. Gao, H. Zhang, Y. Zhang, T. Lv, L. Zhang, Z. Li, X. Xie, F. Li, H. Chen, L. Jia,
[44] Y. Xu, A. Karmakar, W.E. Heberlein, T. Mustafa, A.R. Biris, A.S. Biris, Erlotinib-guided self-assembled trifunctional click nanotheranostics for distin-
Multifunctional magnetic nanoparticles for synergistic enhancement of cancer guishing druggable mutations and synergistic therapy of NSCLC, Mol. Pharm. 15
treatment by combinatorial radio frequency thermolysis and drug delivery, Adv. (11) (2018) 5146–5161.
Healthc. Mater. 1 (4) (2012) 493–501. [70] F. Kong, X. Zhang, H. Zhang, X. Qu, D. Chen, M. Servos, E. Mäkilä, J. Salonen,
[45] S. Luo, X. Yang, C. Shi, Newly emerging theranostic agents for simultaneous can- H.A. Santos, M. Hai, Inhibition of multidrug resistance of cancer cells by
certargeted imaging and therapy, Curr. Med. Chem. 23 (5) (2016) 483–497. Co‐delivery of DNA nanostructures and drugs using porous silicon nanoparticles@
[46] A.A.A. Ali, F.-T. Hsu, C.-L. Hsieh, C.-Y. Shiau, C.-H. Chiang, Z.-H. Wei, C.-Y. Chen, giant liposomes, Adv. Funct. Mater. 25 (22) (2015) 3330–3340.
H.-S. Huang, Erlotinib-conjugated iron oxide nanoparticles as a smart cancer-tar- [71] D. Harada, N. Takigawa, N. Ochi, T. Ninomiya, M. Yasugi, T. Kubo, H. Takeda,
geted theranostic probe for MRI, Sci. Rep. 6 (2016) 36650. E. Ichihara, K. Ohashi, S. Takata, JAK 2‐related pathway induces acquired erlotinib
[47] F.-T. Hsu, H.-S. Liu, A.A.A. Ali, P.-H. Tsai, Y.-C. Kao, C.-F. Lu, H.-S. Huang, C.- resistance in lung cancer cells harboring an epidermal growth factor receptor‐-
Y. Chen, Assessing the selective therapeutic efficacy of superparamagnetic erlotinib activating mutation, Cancer Sci. 103 (10) (2012) 1795–1802.
nanoparticles in lung cancer by using quantitative magnetic resonance imaging and [72] B.D. Looyenga, D. Hutchings, I. Cherni, C. Kingsley, G.J. Weiss, J.P. MacKeigan,
a nuclear factor kappa-B reporter gene system, Nanomed. Nanotechnol. Biol. Med. STAT3 is activated by JAK2 independent of key oncogenic driver mutations in non-
14 (3) (2018) 1019–1031. small cell lung carcinoma, PLoS One 7 (2) (2012) e30820.
[48] J. Nebu, J.A. Devi, R. Aparna, K. Abha, G. Sony, Erlotinib conjugated gold na- [73] R.R. Arasada, J.M. Amann, M.A. Rahman, S.S. Huppert, D.P. Carbone, EGFR
nocluster enveloped magnetic iron oxide nanoparticles–A targeted probe for ima- blockade enriches for lung cancer stem–like cells through Notch3-dependent sig-
ging pancreatic cancer cells, Sensor. Actuators B: Chem. 257 (2018) 1035–1043. naling, Cancer Res. 74 (19) (2014) 5572–5584.
[49] B. Mandal, N.K. Mittal, P. Balabathula, L.A. Thoma, G.C. Wood, Development and [74] A.T. Baker, A. Zlobin, C. Osipo, Notch-EGFR/HER2 bidirectional crosstalk in breast
in vitro evaluation of core–shell type lipid–polymer hybrid nanoparticles for the cancer, Front. Oncol. 4 (2014) 360.
delivery of erlotinib in non-small cell lung cancer, Eur. J. Pharm. Sci. 81 (2016) [75] D. Chen, F. Zhang, J. Wang, H. He, S. Duan, R. Zhu, C. Chen, Y. Chen, L. Yin,
162–171. Biodegradable nanoparticles mediated Co-delivery of erlotinib (ELTN) and fe-
[50] F. Li, H. Mei, X. Xie, H. Zhang, J. Liu, T. Lv, H. Nie, Y. Gao, L. Jia, Aptamer-con- dratinib (FDTN) toward the treatment of ELTN-resistant non-small cell lung cancer
jugated chitosan-anchored liposomal complexes for targeted delivery of erlotinib to (NSCLC) via suppression of the JAK2/STAT3 signaling pathway, Front. Pharmacol.
EGFR-mutated lung cancer cells, AAPS J. 19 (3) (2017) 814–826. 9 (2018) 1214.
[51] R. Mo, Z. Gu, Tumor microenvironment and intracellular signal-activated nano- [76] X. Wan, C. Liu, Y. Lin, J. Fu, G. Lu, Z. Lu, pH sensitive peptide functionalized na-
materials for anticancer drug delivery, Mater. Today 19 (5) (2016) 274–283. noparticles for co-delivery of erlotinib and DAPT to restrict the progress of triple
[52] S. Uthaman, K.M. Huh, I.-K. Park, Tumor microenvironment-responsive nano- negative breast cancer, Drug Deliv. 26 (1) (2019) 470–480.
particles for cancer theragnostic applications, Biomater. Res. 22 (2018) 22. [77] M.J. Lee, S.Y. Albert, A.K. Gardino, A.M. Heijink, P.K. Sorger, G. MacBeath,
[53] H.B. Ruttala, T. Ramasamy, T. Madeshwaran, T.T. Hiep, U. Kandasamy, K.T. Oh, H.- M.B. Yaffe, Sequential application of anticancer drugs enhances cell death by re-
G. Choi, C.S. Yong, J.O. Kim, Emerging potential of stimulus-responsive nanosized wiring apoptotic signaling networks, Cell 149 (4) (2012) 780–794.
anticancer drug delivery systems for systemic applications, Arch Pharm. Res. [78] L. Zhang, Z. Lu, Q. Zhao, J. Huang, H. Shen, Z. Zhang, Enhanced chemotherapy
(Seoul) 41 (2) (2018) 111–129. efficacy by sequential delivery of siRNA and anticancer drugs using PEI‐grafted
[54] H.B. Ruttala, N. Chitrapriya, K. Kaliraj, T. Ramasamy, W.H. Shin, J.-H. Jeong, graphene oxide, Small 7 (4) (2011) 460–464.
J.R. Kim, S.K. Ku, H.-G. Choi, C.S. Yong, Facile construction of bioreducible [79] X. Qian, Y. Ren, Z. Shi, L. Long, P. Pu, J. Sheng, X. Yuan, C. Kang, Sequence-de-
crosslinked polypeptide micelles for enhanced cancer combination therapy, Acta pendent synergistic inhibition of human glioma cell lines by combined temozolo-
Biomater. 63 (2017) 135–149. mide and miR-21 inhibitor gene therapy, Mol. Pharm. 9 (9) (2012) 2636–2645.
[55] T. Ramasamy, H.B. Ruttala, N. Chitrapriya, B.K. Poudal, J.Y. Choi, S.T. Kim, [80] D.B. Pacardo, F.S. Ligler, Z. Gu, Programmable nanomedicine: synergistic and se-
Y.S. Youn, S.K. Ku, H.-G. Choi, C.S. Yong, Engineering of cell microenvironment- quential drug delivery systems, Nanoscale 7 (8) (2015) 3381–3391.
responsive polypeptide nanovehicle co-encapsulating a synergistic combination of [81] S.W. Morton, M.J. Lee, Z.J. Deng, E.C. Dreaden, E. Siouve, K.E. Shopsowitz,

12
D.H. Truong, et al. Journal of Drug Delivery Science and Technology 55 (2020) 101348

N.J. Shah, M.B. Yaffe, P.T. Hammond, A nanoparticle-based combination che- resistance, Cancer Med 4 (11) (2015) 1621–1632.
motherapy delivery system for enhanced tumor killing by dynamic rewiring of [88] E.C. Finger, A.J. Giaccia, Hypoxia, inflammation, and the tumor microenvironment
signaling pathways, Sci. Signal. 7 (325) (2014) ra44. in metastatic disease, Cancer Metastasis Rev. 29 (2) (2010) 285–293.
[82] Y. He, Z. Su, L. Xue, H. Xu, C. Zhang, Co-delivery of erlotinib and doxorubicin by [89] O. Trédan, C.M. Galmarini, K. Patel, I.F. Tannock, Drug resistance and the solid
pH-sensitive charge conversion nanocarrier for synergistic therapy, J. Control. tumor microenvironment, J. Natl. Cancer Inst. (Bethesda) 99 (19) (2007)
Release 229 (2016) 80–92. 1441–1454.
[83] Z. Zhou, C. Kennell, M. Jafari, J.-Y. Lee, S.J. Ruiz-Torres, S.E. Waltz, J.-H. Lee, [90] K. Okamoto, I. Okamoto, E. Hatashita, K. Kuwata, H. Yamaguchi, A. Kita,
Sequential delivery of erlotinib and doxorubicin for enhanced triple negative Breast K. Yamanaka, M. Ono, K. Nakagawa, Overcoming erlotinib resistance in EGFR
cancer treatment using polymeric nanoparticle, Int. J. Pharm. 530 (1–2) (2017) mutation–positive non–small cell lung cancer cells by targeting survivin, Mol.
300–307. Cancer Ther. 11 (1) (2012) 204–213.
[84] Y. Feng, Y. Cheng, Y. Chang, H. Jian, R. Zheng, X. Wu, K. Xu, L. Wang, X. Ma, X. Li, [91] D. Harada, N. Takigawa, K. Kiura, The role of STAT3 in non-small cell lung cancer,
Time-staggered delivery of erlotinib and doxorubicin by gold nanocages with two Cancers 6 (2) (2014) 708–722.
smart polymers for reprogrammable release and synergistic with photothermal [92] J.-K. Dong, H.-M. Lei, Q. Liang, Y.-B. Tang, Y. Zhou, Y. Wang, S. Zhang, W.-B. Li,
therapy, Biomat 217 (2019) 119327. Y. Tong, G. Zhuang, Overcoming erlotinib resistance in EGFR mutation-positive
[85] M. Zhang, B. Sai, P. Cao, Z. Li, L. Zhang, C. Shuai, X. Wang, J. Wang, G. Li, J. Xiang, lung adenocarcinomas through repression of phosphoglycerate dehydrogenase,
Iron oxide nanoparticles synergize with erlotinib to suppress refractory non-small Theranostics 8 (7) (2018) 1808–1823.
cell lung cancer cell proliferation through the inhibition of ErbB/PI3K/AKT and [93] R. Li, Z. Hu, S.-Y. Sun, Z.G. Chen, T.K. Owonikoko, G.L. Sica, S.S. Ramalingam,
PTEN activation, J. Biomed. Nanotechnol. 13 (4) (2017) 458–468. W.J. Curran, F.R. Khuri, X. Deng, Niclosamide overcomes acquired resistance to
[86] A. Ahsan, Mechanisms of Resistance to EGFR Tyrosine Kinase Inhibitors and erlotinib through suppression of STAT3 in non–small cell lung cancer, Mol. Cancer
Therapeutic Approaches: an Update, Lung Cancer Personal. Med., Springer, 2016, Ther. 12 (10) (2013) 2200–2212.
pp. 137–153. [94] S. Tinkle, S.E. McNeil, S. Mühlebach, R. Bawa, G. Borchard, Y.C. Barenholz,
[87] R.M. Jotte, D.R. Spigel, Advances in molecular‐based personalized non‐small‐cell L. Tamarkin, N. Desai, Nanomedicines: addressing the scientific and regulatory gap,
lung cancer therapy: targeting epidermal growth factor receptor and mechanisms of An. New York Acad. Sci. 1313 (1) (2014) 35–56.

13

You might also like