Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Colloids and Surfaces B: Biointerfaces 146 (2016) 482–489

Contents lists available at ScienceDirect

Colloids and Surfaces B: Biointerfaces


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

The application of prodrug-based nano-drug delivery strategy in


cancer combination therapy
Yanxiu Ge, Yakun Ma, Lingbing Li ∗
Department of Pharmaceutics, School of Pharmacy, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China

a r t i c l e i n f o a b s t r a c t

Article history: Single drug therapy that leads to the multidrug resistance of cancer cells and severe side-effect is a thing
Received 30 May 2016 of the past. Combination therapies that affect multiple signaling pathways have been the focus of recent
Received in revised form 24 June 2016 active research. Due to the successful development of prodrug-based nano-drug delivery systems (P-N-
Accepted 27 June 2016
DDSs), their use has been extended to combination therapy as drug delivery platforms. In this review,
Available online 28 June 2016
we focus specifically on the P-N-DDSs in the field of combination therapy including the combinations
of prodrugs with different chemotherapeutic agents, other therapeutic agents, nucleic acid or the com-
Keywords:
bination of different types of therapy (e.g. chemotherapy and phototherapy). The relevant examples of
Combination therapy
Drug delivery
prodrug-based nanoparticulate drug delivery strategy in combination cancer therapy from the recent
Prodrug literature are discussed to demonstrate the feasibilities of relevant technology.
Prodrug-based nano-drug delivery system © 2016 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 482
2. Different chemotherapy drugs combinations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 483
2.1. Prodrug + free drug . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 483
2.2. Prodrug + prodrug . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 484
3. Combination therapy of prodrug with other therapeutic agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 485
4. Combination therapy of prodrug with nucleic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 486
5. Combination chemotherapy and phototherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 487
6. Conclusion and future perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 487
Declaration of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 487
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 487

1. Introduction side effects and improved therapeutic profile has drawn more and
more attention from pharmaceutical researchers [6].
Cancer is a complex disease that represents one of the lead- Combination therapy, generally refers to either the co-
ing causes of death in developed countries [1,2]. Although a large administration of two or more therapeutic agents achieving a
number of potent chemotherapeutic anticancer agents have been synergistic antiproliferative effect or to the combination of dif-
successfully used in clinical practice, there has been no significant ferent types of therapy (e.g. chemotherapy and radiotherapy) [7].
progress in cancer treatment due to the lack of selectivity against Multi-agent therapy can generate synergistic anticancer effects by
cancer cells and associated toxic side effects [1,3,4]. Besides, the focus on different signaling pathways in tumor cells, overcome
multidrug resistance of cancer cells and limited regime of clinical mechanisms of resistance and minimize side effects [8,9]. Actu-
uses of single drug becomes the major limitations in anti-tumor ally, it is routinely used for the treatment of cancer and indeed
treatment [5]. Therefore, the combination therapy with decreased improves the therapeutic effect [10]. For instance, the combination
therapy of doxorubicin (DOX) and paclitaxel (PTX) is used as first-
line treatment for metastatic breast cancer [11–13]. The efficacy
∗ Corresponding author. of hypoxia-activated prodrug TH-302 in combination with dox-
E-mail address: cnlilingbing@yahoo.com (L. Li). orubicin for use in first-line advanced soft tissue sarcoma (STS)

http://dx.doi.org/10.1016/j.colsurfb.2016.06.051
0927-7765/© 2016 Elsevier B.V. All rights reserved.
Y. Ge et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 482–489 483

Table 1
Examples of recently developed P-N-DDSs for combination of prodrug plus free drug.

Name Agent 1 type (name) Nanoplatform Refs.


Agent 2 type (name)

PluronicF127-CS-DOX + PTX Doxorubicin (DOX) Polymeric micelle [35]


Paclitaxel (PTX)
PEGylated Taxol + DOX Doxorubicin (DOX) Janus nanogels [66]
Paclitaxel (PTX)
PEG-C6-AZO-CA4 + DOX Combretastatin A-4 (CA4) Micelle [67]
Doxorubicin (DOX)
Herparin-DEX + DOX Dexamethasone(DEX) Polymeric micelle [68]
Doxorubicin(DOX)
PLGA-PEG-CBP + PTX Carboplatin (CBP) Nanoparticles [36]
Paclitaxel (PTX)
5-FU-stearic acid + CDDP 5-FU Nanostructured lipid carriers (NLC) [69]
Cisplatin (CDDP)
TPGS-cisplatin + docetaxel Cisplatin Nanoparticles [37]
Docetaxel
PLA-Pt (IV) + docetaxel Cisplatin (Pt) Polymeric nanoparticle [18]
Docetaxel
Pluronic P105-DOX + PTX Doxorubicin (DOX) Mixed micelles [31]
Paclitaxel (PTX)
mPEG-b-PLG-g-PTX + DOX Doxorubicin (DOX) Nanoparticles [70]
Paclitaxel (PTX)

CS: chitosan; PEG: polyethylene glycol; C6: hexanethiol; AZO: azobenzene; PLGA: Poly (dl-lactide-co-glycolide); TPGS: d-alpha-tocopheryl polyethylene glycol 1000 succi-
nate; PLA: polyactide; mPEG-b-PLG: methoxypoly(ethylene glycol)-block-poly(l-glutamic acid).

assessed in clinical studies was favorably with outcomes achieved bility, charge and molecular weight [26]. It presents a challenge
compared with other first-line chemotherapies for advanced STS for conventional nanocarriers to co-encapsulate two drugs with
[14]. Besides, the combination of gemcitabine and TH-302 signifi- different chemical and physical characters and precisely keep the
cantly improved tumor response compared with gemcitabine alone relative dosages of the two drugs, as most nanoparticle-based drug
in previously untreated, locally advanced or metastatic pancreatic combinations are comprised of bioactive compounds with simi-
cancer [15]. lar water solubility [16,24,27]. For instance, polymeric micelles are
Though appropriate drug combinations can produce signifi- not suitable for loading well water-soluble drugs because of their
cant benefits in cancer treatment, current combination approach hydrophobic core, not to mention relative combination therapy
through the cocktail administration leaves plenty of room for [16]. Liposomes are capable of carrying hydrophilic and hydropho-
improvements [16]. One major challenge is how to ensure the cor- bic drugs in its inner core and bilayer membrane, respectively.
rect radio of the combined drugs to the target, as the optimized dose But this formulation has disadvantages, such as a lower localized
ratio complicated by dissimilar pharmacokinetics and biodistribu- hydrophobic drug concentration in the lipid bilayer and a higher
tion is hard to be maintained due to different rates of metabolism initial burst, as well as thermodynamic instability [28].
within the body [7]. Besides, the combination of small molecule Prodrugs are defined as chemically modified, biologically inert
drugs would accompany with unfavorable toxic side effects. From compounds that are metabolized in vivo to regenerate the parent
this point of view, the application of nanotechnology has opened bioactive components [3]. The main advantages of prodrugs com-
up unprecedented opportunities in controlled drug delivery and pared to the parent free drugs are: (a) improved solubility in water
novel combination strategies [16–18]. A myriad of nanoscale drug or lipid membrane [29]; (b) decreased adverse effects [30,31]; (c)
delivery systems including micelles, liposomes, nanoparticles and improved cell uptake [1,3]. By conjugating a polymer or a tumor-
nanogels, have endowed encapsulated drugs more favorable phar- specific ligand to chemotherapy drugs via a cleavable bond, they
macokinetic profiles compared to free small-molecule drugs [5,19]. can also be designed to target specific antigens or enzymes that are
They solve the shortcomings of most anticancer drugs such as low over-expressed on tumor cells compared with other normal cells
solubility, fast blood clearance, multidrug resistance, reduce the [1,32]. Recently, their applications have been extended to combina-
side effects by the selective accumulation at tumor sites over nor- tion therapy. Prodrug-based nano-drug delivery systems in the field
mal tissues, via both passive and active mechanisms [20–23]. They of combination chemotherapy to facilitate more efficient delivery
can also improve the efficacy of multimodality treatment by their of anticancer drugs are still advanced and pioneering [33].
ability to effectively delivering multiple therapeutic agents simul- The aim of this paper is to review the use of prodrug-based
taneously and to synchronize their delivery to the target site [24]. nano-drug delivery system strategy in combination therapy with
For example, Zhen Li et al. constructed hollow silica nanoparti- a focus on cancer treatment. We focus separately on each type of
cles sealed with ZnO quantum dots to co-deliver camptothecin systems, including combination therapy of prodrugs with different
(CPT) and doxorubicin (DOX· HCl) [25]. Only when the nanopar- chemotherapy agents, other therapeutic agents, nucleic acid and
ticles entered into the acid intracellular compartments of cancer the combination of chemotherapy with phototherapy. We also dis-
cells, could the ZnO quantum dots be dissolved and trigger the cuss some interesting examples from the recent literature on the
release of the drug. The pH-sensitive drug release minimized the development of P-N-DDS strategy in combination cancer therapy.
toxicity for normal tissues. More important, in vitro cellular assays
revealed that the two drugs combinations with different anticancer 2. Different chemotherapy drugs combinations
mechanism highly improved chemotherapeutic effect [25].
However, in a single combinatorial nanoparticle, drug co- 2.1. Prodrug + free drug
encapsulation through non-covalent physical entrapment leads to
poor stability and batch-to-batch inconsistency in drug loading and In this approach, a prodrug is administered in combination with
release kinetics, especially when two drugs have different solu- a low molecular weight drug. It means one free antitumor drug
484 Y. Ge et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 482–489

Table 2
Examples of recently developed P-N-DDSs for combination of prodrug plus prodrug.

Name Agent 1 type (name) Nanoplatform Refs.


Agent 2 type (name)

PLL-PTX + HA-GEM Paclitaxel (PTX) Muti-prodrug nanocarriers [28]


Gemcitabine(GEM)
SSP-CPT + SSP-DOX Camptothecin (CPT) Nonporous silica prodrug nanoparticles [19]
Doxorubicin (DOX)
MPEG-b-P(LA-co-MCC/PTX) + Cisplatin (Pt) Composite micelles [41]
MPEG-b-P(LA-co-MCC/CisPt(IV)) Paclitaxel (PTX)
P1-DRB + P2-Oxa(IV) Oxaliplatin (Oxa) Composite micelle [42]
Daunorubicin (DRB)
DOX-PLA + CPT-PLA Doxorubicin (DOX) Polymeric nanoparticles [40]
Camptothecin (CPT)
HA-lysine (L)-BCL + Baicalein (BCL) Nanoparticles [33]
FA-valine (V)-PTX Paclitaxel (PTX)
ADD-DOX Adjudin (ADD) Micelle [43]
Doxorubicin (DOX)
Ir-Cb Chemotherapy (Ir) Nanoparticles [44]
Chemotherapy (Cb)

PLL: poly (l-lysine); HA: hyaluronic acid; SSP: stimuli-responsive silane prodrug; PLA: poly-l-lactide; HA: hyaluronic acid; FA: folate; P1: MPEG-b-P(LA-co-MCC)-COOH; P2:
MPEG-b-P(LA-co-MCC)-OH; MPEG-b-P(LA-co-MCC): methoxyl-poly (ethylene glycol)-block-poly (lactide-co-2-methyl-2-carboxyl-propylene carbonate).

carried by a P-N-DDS formed by the amphiphilic polymer conju- Furthermore, by exploiting selective tumor targeting through
gate of another antitumor drug, or by the prodrug-encapsulated conjugating a tumor-specific ligand to the polymer, the P-N-DDS
nanosystems, in which prodrugs are encapsulated in nanocarriers. resulted in enhanced delivery efficiency to target cells. For exam-
The prodrug-based nanocarriers offer a new strategy to create ideal ple, Yu Mi et al. have conjugated herceptin onto the D-␣-tocopherol
combination cancer therapy systems by noncovalently encapsulat- polyethylene glycol 1000 succinate-cisplain (TPGS-cisplatin) pro-
ing a low molecular weight drug simultaneously. They combined drug nanoparticles to co-deliver both cisplatin and docetaxel [37].
the advantages of both prodrugs and co-delivery systems to facil- Herceptin, a humanized monoclonal antibody, can not only target
itate synergistic therapeutic effects via simultaneous multi-drug the extracellular domain of human epidermal growth factor recep-
accumulation [28]. Recently developed P-N-DDSs for combination tor 2 (HER2) but also be used as a biological therapeutic agent for
of prodrug plus free drug are summarized in Table 1. HER2-overexpressing breast cancer [37–39]. The in vitro cytotoxic-
The P-N-DDSs can be designed to improve the selectivity and ity showed the nanoparticles achieved the highest efficacy for HER2
efficacy by conjugating a polymer to the chemotherapy drug via positive cancer cells and much better therapeutic efficacy than each
the bonds (e.g. ester, amide or disulfide bond) which were cleaved of the two original drugs alone [37].
under triggerable conditions such as a lowered interstitial pH,
high intracellular glutathione levels in tumors, or enzymes over- 2.2. Prodrug + prodrug
expressed by tumors [1]. Based on this strategy, the P-N-DDSs for
combination therapy can enhance drug bioavailability for advanced In this approach, two prodrugs each carrying a single therapeu-
drug delivery [34]. For example, Yakun Ma et al. conjugated DOX tic agent are administered in combination. It means the co-delivery
to Pluronic F127-chitosan (F127-CS) copolymer via acid-sensitive of two polymer-drug conjugates or drug-drug conjugate in one P-
amide linkage, which ensure drug release in cancer cellular acid N-DDS. Recently developed P-N-DDSs for combination of prodrug
environment [35]. The results showed that the release of DOX and plus prodrug are summarized in Table 2.
PTX from polymeric micelles exhibited a pH-dependent manner. Although nanoparticle-based combination therapies unify the
And the pH-sensitive polymeric micelle system based on the con- pharmacokinetics and biodistribution of different drug molecules
jugated prodrug (F127-CS-DOX) for co-delivery of DOX and PTX and thus enable the dosage optimization in vivo, it presents a chal-
exhibited a better bioavailability than the PTX plus DOX solution lenge for controlling the ratios of different types of drugs in one
in vivo pharmacokinetic study [35]. nanoparticle [40]. Steric hindrance between the polymer back-
Most anticancer drugs have difficulties in formulation due bones and different drug molecules, variability in drug-to-polymer
to their problems in solubility. With the help of prodrug-based and drug-to-drug interactions, and batch-to-batch inconsistency
nanocarriers, the difficulty in co-encapsulating hydrophilic and in conjugation chemistry are the influencing factors [40]. One
hydrophobic drugs can be overcome, as the prodrug strategy can approach to address these issues is the combination of two
improve drug molecule’s compatibility with carrier platforms by polymer-drug conjugates, each carrying a single therapeutic agent,
chemical modifications, and hence balance the pharmacokinet- and the final loading yield of the therapeutic agents follows the
ics of hydrophilic and hydrophobic drugs [16,36]. For example, same molar ratio as the drug-polymer conjugates used during the
hydrophilic carboplatin (CBP) in combination with hydrophobic nanoparticle preparation [16,40]. For example, Haihua Xiao and
PTX was used for non-small cell lung cancer (NSCLC) clinically colleagues synthesized a cisplatin (IV) conjugate and a paclitaxel
with severe side effects [36]. Designing nanocarriers platform to conjugates starting with the same biodegradable and amphiphilic
co-encapsulate the two drugs to increase therapeutic efficacy and block copolymer. Composite micelles with desirable drug contents
decrease toxic effect is urgent [36]. Wen Zhang et al. conjugated and ratio can be obtained by simply mixing the two conjugates at
the hydrophilic CBP to PLGA-PEG-COOH to synthesized PLGA-PEG- desirable molar ratios of Pt/PTX and co-assembling them. More-
CBP. Then the combination delivery of CBP and hydrophobic PTX over, the synergistic effects have been demonstrated in vitro and
was achieved through encapsulating PTX into nanoparticles self- the combination therapy in micellar dosage-form led to safer
assembled by PLGA-PEG-CBP. The synergistic anti-tumor efficacy and better antitumor efficacy in vivo than the small molecules
in NCL-H460 lung cancer cells and mice-bearing lung cancer cells based combination [41]. Applying the same principle, the same
model were observed [36]. group also evaluated the therapeutic effect of the composite
Y. Ge et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 482–489 485

Fig. 1. Schematic illustration of multi-prodrug nanocarriers consisting of PLL-PTX and HA-GEM.

micelles co-assembled by polymer-daunorubicin (DRB) conjugate PEG2000 into micelles. ADD could trigger mitochondrial dysfunction
and polymer-Oxaliplatin (IV) conjugate, which has the similar car- in cancer cells and restore the chemosensitization of DOX-related
rier polymer as in the Polymer-DRB conjugate. The relative drug multidrug resistance (MDR) cancer cells. The rapidly release of
doses in the micelles can be modulated easily by simply adjusting free DOX and ADD within the acidic endolysosomes of MCF-7/ADR
the mixed ratios of polymer-DRB conjugate and polymer-Oxa (IV) cancer cells, enhanced the anti-MDR capability of ADD-DOX (M)
conjugate. Treatment with the combination significantly reduced micelles [43].
systematic toxicity and improved synergistic effect compared with Though nanocarriers have lots of advantages in enhancing ther-
combination of small molecules of the two anticancer drugs both apeutic efficacy, in these delivery systems, carriers almost have no
in vitro and in vivo [42]. therapeutic efficacy by themselves or even cause side-effects to
The low delivery efficiency to target cells can be changed tissues and organs in the course of degradation, metabolism, and
as a result of exploiting selective tumor targeting. For example, excretion [44]. To avoid the problems from carriers, Ping Huang
Ilkoo Noh and colleagues developed multi-prodrug nanocarriers et al. designed a drug self-delivery system. Instead of modifying
consisting of poly (l-lysine)-carboxylate paclitaxel (PLL-PTX) and the hydrophilic compound with a hydrophobic polymer deriva-
hyaluronic acid-conjugated gemcitabine (HA-GEM) (Fig. 1). HA has tive, the amphiphilic drug-drug conjugate (ADDC) composing of
a strong affinity for cell surface receptors of cluster determinant the hydrophilic anticancer drug irinotecan (Ir) and the hydrophobic
44 (CD44), which is only over-expressed in malignancy. In vitro anticancer drug chlorambucil (Cb) via a hydrolysable ester link-
and in vivo studies showed that this combination enhanced cellular age can self-assembles into nanoparticles in water with an average
uptake of PTX and GEM into CD44-expressing human biliary cancer hydrodynamic diameter of 88.3 nm. By passive accumulation via
cells compared to free drugs. And this system exhibited synergistic enhanced permeability and retention (EPR) effect, the accumula-
cancer therapeutic efficacy with different treatment pathways of tion of Ir-Cb conjugate in the tumor tissue increased. After the
the dual drugs [28]. hydrolysis of the ester bond in the tumor cells, both free Ir and
Another type of the P-N-DDSs in the field of combination Cb can be released to kill the cancer cells with synergetic cytotox-
chemotherapy is the self-assembling conjugates composed of two icity, leading to an excellent anticancer activity in vitro and in vivo
different antitumor drugs, each of which is conjugated with differ- [44]. Though the ratio between two drugs with different properties
ent polymers separately to achieve dual targets [33]. The polymer can be maintained, the linker of drug-drug conjugates also limited
prodrugs in this kind of P-N-DDS have a dual-targeting effect, which clinical applicability by constraining the molar ratio [40].
means better synergistic effect compared with a single target [33].
For example, Wei Wang et al. developed self-assembled nanopar-
3. Combination therapy of prodrug with other therapeutic
ticles (NPs) for the co-delivery of hyaluronic acid-lysine-baicalein
agents
(HA-l-BCL) and folate-valine-paclitaxel (FA-V-PTX). The PTX-BCL
NPs which have dual-targeted ligands of HA and FA displayed
The P-N-DDSs for co-delivery of anticancer drugs and other
remarkably better antitumor activity and lower systemic toxic-
therapeutic agents can also be used for enhancing chemothera-
ity than their free drug counterparts in mice bearing an A549/PTX
peutic effects furtherly, such as the combination of chemotherapy
drug-resistant lung cancer xenograft [33].
drugs and anticancer synergists [45]. For example, P-N-DDS for
Another strategy to maintain the radio between two drugs with
treating MDR cancer was emerged by the combination of DOX
drastically different properties is to covalently conjugate dual drugs
with disulfiram (DSF), a P-gp inhibitor as well as an apoptosis
using a hydrolysable bond and then encapsulate the drug-drug con-
inducer. DSF was encapsulated into the micelles formed by the self-
jugates into nanoparticles for co-delivery [40]. For example, Xu
assembly of SMA-ADH-DOX (SAD) conjugate, which was obtained
Li and colleagues has conjugated Adjudin (ADD) to DOX via an
by conjugating DOX to poly(styrene-co-maleic anhydride) (SMA)
acid-sensitive hydrazone bond, and then encapsulated by DSPE-
derivative with adipic dihydrazide (ADH) through an acid-cleavable
486 Y. Ge et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 482–489

Table 3
Examples of recently developed P-N-DDSs for combination of prodrug with nucleic acid.

Name Agent 1 type (name) Nanoplatform Refs.


Agent 2 type (name)

DSS-BEN + DNA N1 ,N11 -bisethylnorspermine (BEMSpm) Polyplexes [71]


TNF␣ plasmid DNA
CPTssR5H5 + siRNA Camptothecin(CPT) Vesicles [47]
MAP3K7 siRNA
CPT-PCB + siPlk1 Camptothecin(CPT) Liposomes [49]
siPlk1 siRNA
TPGS-siRNA + docetaxel Docetaxel Micelle [58]
siPlk1 siRNA
PC-DOX + wtP53 plasmid Doxorubicin (DOX) Nanocomplex [53]
wt P53 plasmid

R5H5: oligo-peptide RRRRRHHHHHC; PCB: poly(carboxybetaine); TPGS: ␣-tocopheryl polyethylene glycol 1000 succinate; PC: ␤-cyclodextrin-polyethylenimine.

hydrazone bond. Encapsulated DSF was released faster than DOX prepared by grafting Dox to PC through peptide bonds linkages.
to inactivate P-gp and subsequently inhibits the pumping out of PC-Dox functions dually acted as a gene delivery vector as well
the chemotherapeutic drug. The smart co-delivery system showed as an anticancer prodrug. The prodrug-based drug delivery system
increased tumor accumulation of DOX and excellent antitumor (DDS) formed by PC-Dox could co-deliver Dox and wt p53 plasmid.
effect in MDR tumor [26]. The tumor suppressor gene p53 can not only induces apoptosis of
cells under DNA damage but also enhances the chemosensitivity of
tumor cells to chemotherapeutic drugs through the inhibition P-gp
4. Combination therapy of prodrug with nucleic acid
expression. The PC-Dox/p53 nanocomplexes efficiently deliver pro-
drug and gene to the same cancer cells simultaneously and increase
Due to severe side effects and gradually developed resistance of
the anticancer activity with synergistic therapeutic effects in vitro
those traditional anticancer drugs, nucleic acid therapeutics such
and in vivo [53]. The non-viral gene carriers offer great advan-
as small interfering RNA (siRNA), antisense oligonucleotides (AON),
tages, such as easy chemical modification and functionalization,
small hairpin RNA (shRNA), and plasmid DNA (pDNA) have been
easy preparation, high stability and good safety [46,54].
developed based on the increasing knowledge on molecular biol-
P-N-DDSs can also achieve in controlling the rate and sequence
ogy and biochemistry and shown promise in cancer treatment [46].
of release of the individual agents [46,49]. For instance, Yan Li
The efficacy of siRNA for the treatment of cancer in human has
et al. constructed CPT prodrug based cationic liposomes for the co-
been proven in a phase I clinical trial [47,48]. The combination
delivery siPlk1. The two drugs could be simultaneously delivered to
of chemotherapy drugs and nucleic acid-based therapy targeting
tumor cells and the siRNA targeting Plk1 was released earlier and
multiple disease pathways can cooperatively enhance anticancer
faster than CPT to enhance the sensitivity of CPT to cancer cells by
effect through synergistic or combined effect [45,47,49]. Therefore,
down-regulating the Plk1 expression, while CPT was released in a
considerable efforts have been made to develop safe and effec-
sustained and pH and esterase-dependent manner. As a result, this
tive vehicles to spatiotemporally co-localize anticancer drugs and
co-delivery system exhibited a synergistic effect in vitro and in vivo
nucleic acids in cancer cells [50,51]. Recently developed P-N-DDSs
[49].
for combination of prodrug with nucleic acid are summarized in
As widely investigated in co-encapsulation of nucleic acid
Table 3.
and hydrophobic chemotherapeutic drug are self-assembled block
A large amount of cationic lipid- and polymer-based nanocarri-
copolymers consisting of a hydrophobic block and a cationic block
ers have been used to deliver both nucleic acids and chemotherapy
incorporating the hydrophobic drug and nucleic acid, respectively
drugs [46]. However, the complexes formed by nucleic acids with
[46,55,56]. As the nucleic acids were absorbed on the cationic sur-
cationic polymers or lipids do not allow for easy co-encapsulation
face by electrostatic interaction, the release of them may not be well
of small molecule drugs [46]. One approach to the issue is the
controlled. They could dissociate from the surface and conjugate to
utilization of P-N-DDS, directly conjugating the drug to the poly-
other molecules such as cholesterol or peptide, upon changing in
meric vector via a biodegradable linker [52]. For example, cationic
pH or salt concentration, thus the stability can be improved, and
␤-cyclodextrin-polyethylenimine-Dox (PC-Dox) conjugates were

Fig. 2. Schematic illustration of TCAD@Ce6 and their application in vitro.


Y. Ge et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 482–489 487

the biodistribution is changed [57]. In response to this issue, Jing optimization strategies to arrive at the identification of appropri-
Zhao et al. have conjugated siPlk1 to TPGS via disulfide bond to ate drug combinations, correct choice of dosages, the optimal ratio
form TPGS-siPlk1, which could prevent premature release of siRNA of the co-loaded therapeutics to achieve synergy, accurate drug
and facilitates triggering of its release in cell cytoplasm due to ele- release and the critical features like relative safety and stability.
vated glutathione concentration. The micelles consisted of TPGS Prodrug-based nano-drug delivery systems synchronized the
and TPGS-siRNA conjugates for co-delivery docetaxel and siPlk1, advantages of prodrugs and nanotechnology, and thus able to
further enhances the efficacy to the HER2 positive breast cancer achieve enhanced antitumor efficiency. What’s more, numerous
through herceptin conjugation. The synergistic effects and the tar- nanoparticles based on self-assembling polymer-drug conjugates
geting effects are assessed in vitro by IC50 [58]. are under clinical trials. This shows the great potential of these P-
N-DDSs in the clinical application in the future but at the same time
poses new challenges, such as the industrial production of chemi-
5. Combination chemotherapy and phototherapy
cal synthesis, the correction of in vitro studies with behavior in vivo
and the kinetics of drug release [45].
Photodynamic therapy (PDT) refers to the cancer treatment
The successful development of P-N-DDSs has inspired the inter-
through the selective uptake of a light-sensitive agent, a photosen-
est of scientists to utilize them for combination therapy, which
sitizer (PS), which can produce highly reactive oxygen species (ROS)
combined the advantages of P-N-DDSs and combination therapy.
followed by exposure to a specific wavelength that irreversibly
This kind of nanoprepatrations can seamlessly integrate not just
induce cell apoptosis or necrosis in the targeted tissue [59–61]. As
multiple therapeutic payloads with improved stability and drug
the activating light is non-ionizing and the PS itself is minimally
loading, but also co-delivery of hydrophobic drugs and hydrophilic
toxic in the absence of light, only those cells that are simultane-
drug. Besides, it has been served as the effective delivery platforms
ously exposed to the PS, light and oxygen are exposed to cytotoxic
for drug-nucleic acid combination. Sophisticated nanoparticles,
effect [62–64]. Thus PDT displayed the possibility of dual-selectivity
which have active molecular targeting and respond to multi-
for cancer treatment by the preferential accumulation of PS in the
ple stimuli, could achieve more efficient therapy. Although, such
tumor through developing technologies and light irradiation selec-
nanopreparations aroused great interest of scientists, they are
tively on the tumor [62]. Based on the attractive tumor therapy
largely limited to research laboratories for now. While few anti-
properties of PDT, the combination therapy of phototherapy and
cancer prodrugs have actually reached the market, numerous
chemotherapy may have synergistic therapeutic effects [59].
prodrugs of cytotoxic anticancer agents are currently under clinical
For example, a P-N-DDS based on chemo-photodynamic combi-
trials.
nation was prepared by co-delivering the photosensitizer chlorine
This review article provides an overview of recent advances of
e6 (Ce6) and DOX in TCAD@Ce6 nanoparticles. TCAD was obtained
prodrug-based nano-drug delivery systems in the field of combi-
by conjugating TPGS to acid-sensitive cis-aconitic anhydride-
nation therapy against cancer. The benefits of combination therapy
modified doxorubicin (CAD), and Ce6 was loaded into the prodrug
for improved cancer therapies are clearly established. The future of
nanoparticle. The system could be easily hydrolyzed in the acid
these systems is optimism and the need for designing multifunc-
microenvironment of tumors (Fig. 2). Compared with the free form
tional P-N-DDSs for combination therapy will keep growing in the
of DOX and Ce6, TCAD@Ce6 nanoparticles exhibited higher uptake
future.
of DOX and Ce6 in vitro experiments, and induced higher inhibi-
tion of A549 tumor cells under near-infrared laser irradiation both
in vitro and in vivo [59]. Declaration of interest
Instead of polymer conjugates, chemotherapy drugs conju-
gates with PS can also be utilized for the combination therapy of The authors gratefully acknowledge financial support from Nat-
chemotherapy and phototherapy. For example, fullerenes, which ural Science Foundation of Shandong Province (ZR2015HM070).
also have photoexcitation properties, covalently incorporated DOX The authors have no relevant affiliations or financial involvement
via a cleavable bond. The formed water-dispersible fullerene with any organization or entity with a financial interest in or finan-
aggregates with targeting ligands onto fullerenes could not only cial conflict with the subject matter or materials discussed in the
exhibit a photodynamic effect but also enhanced cytotoxicity manuscript.
towards folate receptor-positive cancer cells. The combined effect
of chemotherapy and PDT increases the therapeutic efficacy of the References
DOX-fullerene aggregate prodrug. The P-N-DDS successfully com-
bined pH-responsible chemotherapeutic, active targeting, and PDT [1] R. Mahato, W. Tai, K. Cheng, Prodrugs for improving tumor targetability and
efficiency, Adv. Drug Deliv. Rev. 63 (2011) 659–670.
properties into one system [65]. [2] S.-S. Feng, Chemotherapeutic engineering: concept, feasibility, safety and
prospect—a tribute to Shu Chien’s 80th birthday, Cell. Mol. Bioeng. 4 (2011)
708–716.
6. Conclusion and future perspective [3] L. Bildstein, C. Dubernet, P. Couvreur, Prodrug-based intracellular delivery of
anticancer agents, Adv. Drug Deliv. Rev. 63 (2011) 3–23.
[4] H. He, Y. Wang, H. Wen, X. Jia, Dendrimer-based multilayer nanocarrier for
Due to the molecular complexity of many diseases, the mul-
potential synergistic paclitaxel–doxorubicin combination drug delivery, RSC
tidrug resistance of cancer cells and associated toxicity of clinical Adv. 4 (2014) 3643–3652.
uses of single drug, successful cancer therapies need to be based on [5] H. Wang, Y. Zhao, Y. Wu, Y.L. Hu, K. Nan, G. Nie, H. Chen, Enhanced anti-tumor
combination approaches, especially there is a significant process efficacy by co-delivery of doxorubicin and paclitaxel with amphiphilic
methoxy PEG-PLGA copolymer nanoparticles, Biomaterials 32 (2011)
in our understanding of various molecular basis and signal trans- 8281–8290.
duction pathways in cancer. Because of the advances in polymer [6] X. Zhao, Q. Chen, Y. Li, H. Tang, W. Liu, X. Yang, Doxorubicin and curcumin
chemistry, nanopreparations have evolved for combination ther- co-delivery by lipid nanoparticles for enhanced treatment of
diethylnitrosamine-induced hepatocellular carcinoma in mice, Eur. J. Pharm.
apy, such as the incorporation of different chemotherapy drugs Biopharm. 93 (2015) 27–36.
and the combination of chemotherapy and phototherapy. Besides, [7] F. Greco, M.J. Vicent, Combination therapy: opportunities and challenges for
availability of advanced genomic technologies spurred co-delivery polymer-drug conjugates as anticancer nanomedicines, Adv. Drug Deliv. Rev.
61 (2009) 1203–1213.
of chemotherapy drugs and nucleic acid. However, it is more chal- [8] J.L. Wu, C.Q. Wang, R.X. Zhuo, S.X. Cheng, Multi-drug delivery system based
lenging to develop combination therapies than mono-therapies. on alginate/calcium carbonate hybrid nanoparticles for combination
The combination within a nanopreparation requires sophisticated chemotherapy, Colloids Surf. B Biointerfaces 123 (2014) 498–505.
488 Y. Ge et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 482–489

[9] P. Parhi, C. Mohanty, S.K. Sahoo, Nanotechnology-based combinational drug [33] W. Wang, M. Xi, X. Duan, Y. Wang, F. Kong, Delivery of baicalein and paclitaxel
delivery: an emerging approach for cancer therapy, Drug Discov. Today 17 using self-assembled nanoparticles: synergistic antitumor effect in vitro and
(2012) 1044–1052. in vivo, Int. J. Nanomed. 10 (2015) 3737–3750.
[10] T. Feng, H. Tian, C. Xu, L. Lin, Z. Xie, M.H. Lam, H. Liang, X. Chen, Synergistic [34] V. Delplace, P. Couvreur, J. Nicolas, Recent trends in the design of anticancer
co-delivery of doxorubicin and paclitaxel by porous PLGA microspheres for polymer prodrug nanocarriers, Polym. Chem. 5 (2014) 1529–1544.
pulmonary inhalation treatment, Eur. J. Pharm. Biopharm. 88 (2014) [35] Y. Ma, X. Fan, L. Li, pH-sensitive polymeric micelles formed by doxorubicin
1086–1093. conjugated prodrugs for co-delivery of doxorubicin and paclitaxel, Carbohydr.
[11] D.L. Gustafson, A.L. Merz, M.E. Long, Pharmacokinetics of combined Polym. 137 (2016) 19–29.
doxorubicin and paclitaxel in mice, Cancer Lett. 220 (2005) 161–169. [36] W. Zhang, C. Li, C. Shen, Y. Liu, X. Zhao, Y. Liu, D. Zou, Z. Gao, C. Yue,
[12] W.L. Lee, W.M. Guo, V.H. Ho, A. Saha, H.C. Chong, N.S. Tan, E.Y. Tan, S.C. Loo, Prodrug-based nano-drug delivery system for co-encapsulate paclitaxel and
Delivery of doxorubicin and paclitaxel from double-layered microparticles: carboplatin for lung cancer treatment, Drug Deliv. (2015) 1–6.
the effects of layer thickness and dual-drug vs. single-drug loading, Acta [37] Y. Mi, J. Zhao, S.S. Feng, Targeted co-delivery of docetaxel, cisplatin and
Biomater. 27 (2015) 53–65. herceptin by vitamin E TPGS-cisplatin prodrug nanoparticles for
[13] Y. Liu, J. Fang, Y.J. Kim, M.K. Wong, P. Wang, Codelivery of doxorubicin and multimodality treatment of cancer, J. Control. Release 169 (2013) 185–192.
paclitaxel by cross-linked multilamellar liposome enables synergistic [38] C. Lin, D.R. Chen, K.J. Chang, T.W. Chang, H.C. Wang, A phase II study of
antitumor activity, Mol. Pharm. 11 (2014) 1651–1661. neoadjuvant chemotherapy with docetaxel, cisplatin and trastuzumab for T2
[14] S.P. Chawla, L.D. Cranmer, B.A. Van Tine, D.R. Reed, S.H. Okuno, J.E. Butrynski, breast cancers, Cancer Chemother. Pharmacol. 69 (2012) 1363–1368.
D.R. Adkins, A.E. Hendifar, S. Kroll, K.N. Ganjoo, Phase II study of the safety [39] J. Hurley, P. Doliny, I. Reis, O. Silva, C. Gomez-Fernandez, P. Velez, G. Pauletti,
and antitumor activity of the hypoxia-activated prodrug TH-302 in J.E. Powell, M.D. Pegram, D.J. Slamon, Docetaxel, cisplatin, and trastuzumab as
combination with doxorubicin in patients with advanced soft tissue sarcoma, primary systemic therapy for human epidermal growth factor receptor
J. Clin. Oncol. 32 (2014) 3299–3306. 2-positive locally advanced breast cancer, J. Clin. Oncol. 24 (2006) 1831–1838.
[15] M.J. Borad, S.G. Reddy, N. Bahary, H.E. Uronis, D. Sigal, A.L. Cohn, W.R. [40] S. Aryal, C.M. Hu, L. Zhang, Polymeric nanoparticles with precise ratiometric
Schelman, J. Stephenson Jr., E.G. Chiorean, P.J. Rosen, B. Ulrich, T. Dragovich, control over drug loading for combination therapy, Mol. Pharm. 8 (2011)
S.A. Del Prete, M. Rarick, C. Eng, S. Kroll, D.P. Ryan, Randomized phase II trial 1401–1407.
of gemcitabine plus TH-302 versus gemcitabine in patients with advanced [41] H. Xiao, H. Song, Q. Yang, H. Cai, R. Qi, L. Yan, S. Liu, Y. Zheng, Y. Huang, T. Liu, X.
pancreatic cancer, J. Clin. Oncol. 33 (2015) 1475–1481. Jing, A prodrug strategy to deliver cisplatin(IV) and paclitaxel in nanomicelles
[16] C.M. Hu, L. Zhang, Nanoparticle-based combination therapy toward to improve efficacy and tolerance, Biomaterials 33 (2012) 6507–6519.
overcoming drug resistance in cancer, Biochem. Pharmacol. 83 (2012) [42] H. Xiao, W. Li, R. Qi, L. Yan, R. Wang, S. Liu, Y. Zheng, Z. Xie, Y. Huang, X. Jing,
1104–1111. Co-delivery of daunomycin and oxaliplatin by biodegradable polymers for
[17] S.M. Cohen, R. Mukerji, S. Cai, I. Damjanov, M.L. Forrest, M.S. Cohen, safer and more efficacious combination therapy, J. Control. Release 163 (2012)
Subcutaneous delivery of nanoconjugated doxorubicin and cisplatin for 304–314.
locally advanced breast cancer demonstrates improved efficacy and [43] X. Li, C. Gao, Y. Wu, C.Y. Cheng, W. Xia, Z. Zhang, Combination delivery of
decreased toxicity at lower doses than standard systemic combination Adjudin and Doxorubicin via integrating drug conjugation and nanocarrier
therapy in vivo, Am. J. Surg. 202 (2011) 646–652, discussion 652–643. approaches for the treatment of drug-resistant cancer cells, J. Mater. Chem. B
[18] N. Kolishetti, S. Dhar, P.M. Valencia, L.Q. Lin, R. Karnik, S.J. Lippard, R. Langer, 3 (2015) 1556–1564.
O.C. Farokhzad, Engineering of self-assembled nanoparticle platform for [44] P. Huang, D. Wang, Y. Su, W. Huang, Y. Zhou, D. Cui, X. Zhu, D. Yan,
precisely controlled combination drug therapy, Proc. Natl. Acad. Sci. U. S. A. Combination of small molecule prodrug and nanodrug delivery: amphiphilic
107 (2010) 17939–17944. drug-drug conjugate for cancer therapy, J. Am. Chem. Soc. 136 (2014)
[19] Z. Xu, S. Liu, Y. Kang, M. Wang, Glutathione- and pH-responsive nonporous 11748–11756.
silica prodrug nanoparticles for controlled release and cancer therapy, [45] C. Luo, J. Sun, B. Sun, Z. He, Prodrug-based nanoparticulate drug delivery
Nanoscale 7 (2015) 5859–5868. strategies for cancer therapy, Trends Pharmacol. Sci. 35 (2014) 556–566.
[20] X. Zhang, K. Achazi, D. Steinhilber, F. Kratz, J. Dernedde, R. Haag, A facile [46] J. Li, Y. Wang, Y. Zhu, D. Oupicky, Recent advances in delivery of drug-nucleic
approach for dual-responsive prodrug nanogels based on dendritic acid combinations for cancer treatment, J. Control. Release 172 (2013)
polyglycerols with minimal leaching, J. Control. Release 174 (2014) 209–216. 589–600.
[21] H. Zhang, K. Wang, P. Zhang, W. He, A. Song, Y. Luan, Redox-sensitive micelles [47] Q. Tang, B. Cao, G. Cheng, Co-delivery of small interfering RNA using a
assembled from amphiphilic mPEG-PCL-SS-DTX conjugates for the delivery of camptothecin prodrug as the carrier, Chem. Commun. 50 (2014) 1323–1325.
docetaxel, Colloids Surf. B Biointerfaces 142 (2016) 89–97. [48] M.E. Davis, J.E. Zuckerman, C.H. Choi, D. Seligson, A. Tolcher, C.A. Alabi, Y. Yen,
[22] P. Zhang, H. Zhang, W. He, D. Zhao, A. Song, Y. Luan, Disulfide-linked J.D. Heidel, A. Ribas, Evidence of RNAi in humans from systemically
amphiphilic polymer-docetaxel conjugates assembled redox-sensitive administered siRNA via targeted nanoparticles, Nature 464 (2010)
micelles for efficient antitumor drug delivery, Biomacromolecules (2016). 1067–1070.
[23] S. Li, Q. He, T. Chen, W. Wu, K. Lang, Z.M. Li, J. Li, Controlled co-delivery [49] Y. Li, R. Liu, J. Yang, G. Ma, Z. Zhang, X. Zhang, Dual sensitive and temporally
nanocarriers based on mixed micelles formed from cyclodextrin-conjugated controlled camptothecin prodrug liposomes codelivery of siRNA for high
and cross-linked copolymers, Colloids Surf. B Biointerfaces 123 (2014) efficiency tumor therapy, Biomaterials 35 (2014) 9731–9745.
486–492. [50] X. Xu, K. Xie, X.Q. Zhang, E.M. Pridgen, G.Y. Park, D.S. Cui, J. Shi, J. Wu, P.W.
[24] M.K. liang Ma, Andrew Smith, Nanoparticles for combination drug therapy, Kantoff, S.J. Lippard, R. Langer, G.C. Walker, O.C. Farokhzad, Enhancing tumor
ACS Nano 7 (2013) 9518–9525. cell response to chemotherapy through nanoparticle-mediated codelivery of
[25] Z. Li, H. Li, L. Liu, X. You, C. Zhang, Y. Wang, A pH-sensitive nanocarrier for siRNA and cisplatin prodrug, Proc. Natl. Acad. Sci. U. S. A. 110 (2013)
co-delivery of doxorubicin and camptothecin to enhance chemotherapeutic 18638–18643.
efficacy and overcome multidrug resistance in vitro, RSC Adv. 5 (2015) [51] H. Yu, C. Guo, B. Feng, J. Liu, X. Chen, D. Wang, L. Teng, Y. Li, Q. Yin, Z. Zhang, Y.
77097–77105. Li, Triple-layered pH-responsive micelleplexes loaded with siRNA and
[26] X. Duan, J. Xiao, Q. Yin, Z. Zhang, H. Yu, S. Mao, Y. Li, Smart pH-sensitive and cisplatin prodrug for NF-kappa B targeted treatment of metastatic breast
temporal-controlled polymeric micelles for effective combination therapy of cancer, Theranostics 6 (2016) 14–27.
doxorubicin and disulfiram, ACS Nano 7 (2013) 5858–5869. [52] S. Liu, Y. Guo, R. Huang, J. Li, S. Huang, Y. Kuang, L. Han, C. Jiang, Gene and
[27] J. Park, S.H. Wrzesinski, E. Stern, M. Look, J. Criscione, R. Ragheb, S.M. Jay, S.L. doxorubicin co-delivery system for targeting therapy of glioma, Biomaterials
Demento, A. Agawu, P. Licona Limon, A.F. Ferrandino, D. Gonzalez, A. 33 (2012) 4907–4916.
Habermann, R.A. Flavell, T.M. Fahmy, Combination delivery of TGF-beta [53] X. Lu, Q.Q. Wang, F.J. Xu, G.P. Tang, W.T. Yang, A cationic prodrug/therapeutic
inhibitor and IL-2 by nanoscale liposomal polymeric gels enhances tumour gene nanocomplex for the synergistic treatment of tumors, Biomaterials 32
immunotherapy, Nat. Mater. 11 (2012) 895–905. (2011) 4849–4856.
[28] I. Noh, H.O. Kim, J. Choi, Y. Choi, D.K. Lee, Y.M. Huh, S. Haam, Co-delivery of [54] F.Z. Wang, Z.S. Xie, L. Xing, B.F. Zhang, J.L. Zhang, P.F. Cui, J.B. Qiao, K. Shi, C.S.
paclitaxel and gemcitabine via CD44-targeting nanocarriers as a prodrug with Cho, M.H. Cho, X. Xu, P. Li, H.L. Jiang, Biocompatible polymeric
synergistic antitumor activity against human biliary cancer, Biomaterials 53 nanocomplexes as an intracellular stimuli-sensitive prodrug for type-2
(2015) 763–774. diabetes combination therapy, Biomaterials 73 (2015) 149–159.
[29] V. Bala, S. Rao, B.J. Boyd, C.A. Prestidge, Prodrug and nanomedicine [55] P. Liu, H. Yu, Y. Sun, M. Zhu, Y. Duan, A mPEG-PLGA-b-PLL copolymer carrier
approaches for the delivery of the camptothecin analogue SN38, J. Control. for adriamycin and siRNA delivery, Biomaterials 33 (2012) 4403–4412.
Release 172 (2013) 48–61. [56] N. Cao, D. Cheng, S. Zou, H. Ai, J. Gao, X. Shuai, The synergistic effect of
[30] Q. Guo, H. Wang, Y. Zhao, H. Wang, F. Zeng, H. Hua, Q. Xu, Y. Huang, hierarchical assemblies of siRNA and chemotherapeutic drugs co-delivered
Cell-penetrating albumin conjugates for enhanced doxorubicin delivery, into hepatic cancer cells, Biomaterials 32 (2011) 2222–2232.
Polym. Chem. 4 (2013) 4584. [57] C. Wolfrum, S. Shi, K.N. Jayaprakash, M. Jayaraman, G. Wang, R.K. Pandey, K.G.
[31] Y. Chen, W. Zhang, Y. Huang, F. Gao, X. Sha, X. Fang, Pluronic-based functional Rajeev, T. Nakayama, K. Charrise, E.M. Ndungo, T. Zimmermann, V.
polymeric mixed micelles for co-delivery of doxorubicin and paclitaxel to Koteliansky, M. Manoharan, M. Stoffel, Mechanisms and optimization of
multidrug resistant tumor, Int. J. Pharm. 488 (2015) 44–58. in vivo delivery of lipophilic siRNAs, Nat. Biotechnol. 25 (2007) 1149–1157.
[32] Q. Mou, Y. Ma, X. Zhu, D. Yan, A small molecule nanodrug consisting of [58] J. Zhao, Y. Mi, S.S. Feng, Targeted co-delivery of docetaxel and siPlk1 by
amphiphilic targeting ligand-chemotherapy drug conjugate for targeted herceptin-conjugated vitamin E TPGS based immunomicelles, Biomaterials 34
cancer therapy, J. Control. Release 230 (2016) 34–44. (2013) 3411–3421.
[59] W. Hou, X. Zhao, X. Qian, F. Pan, C. Zhang, Y. Yang, J.M. de la Fuente, D. Cui,
pH-Sensitive self-assembling nanoparticles for tumor near-infrared
Y. Ge et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 482–489 489

fluorescence imaging and chemo-photodynamic combination therapy, [66] J. Wei, H. Wang, M. Zhu, D. Ding, D. Li, Z. Yin, L. Wang, Z. Yang, Janus nanogels
Nanoscale 8 (2015) 104–116. of PEGylated Taxol and PLGA-PEG-PLGA copolymer for cancer therapy,
[60] S. Verma, G.M. Watt, Z. Mai;1;, T. Hasan, Strategies for enhanced Nanoscale 5 (2013) 9902–9907.
photodynamic therapy effects, Photochem. Photobiol. 83 (2007) 996–1005. [67] H. Liu, R. Zhang, Y. Niu, Y. Li, C. Qiao, J. Weng, J. Li, X. Zhang, Z. Xiao, X. Zhang,
[61] K. Pizova, K. Tomankova, A. Daskova, S. Binder, R. Bajgar, H. Kolarova, Development of hypoxia-triggered prodrug micelles as doxorubicin carriers
Photodynamic Therapy for Enhancing Antitumour Immunity, 156, Biomedical for tumor therapy, RSC Adv. 5 (2015) 20848–20857.
papers of the Medical Faculty of the University Palacky, Olomouc, [68] N.N. Li, J. Lin, D. Gao, L.M. Zhang, A macromolecular prodrug strategy for
Czechoslovakia, 2012, pp. 93–102. combinatorial drug delivery, J. Colloid Interface Sci. 417 (2014) 301–309.
[62] A. Master, M. Livingston, A. Sen Gupta, Photodynamic nanomedicine in the [69] C.Y. Qu, M. Zhou, Y.W. Chen, M.M. Chen, F. Shen, L.M. Xu, Engineering of lipid
treatment of solid tumors: perspectives and challenges, J. Control. Release prodrug-based, hyaluronic acid-decorated nanostructured lipid carriers
168 (2013) 88–102. platform for 5-fluorouracil and cisplatin combination gastric cancer therapy,
[63] J.D. Miller, E.D. Baron, H. Scull, A. Hsia, J.C. Berlin, T. McCormick, V. Colussi, Int. J. Nanomed. 10 (2015) 3911–3920.
M.E. Kenney, K.D. Cooper, N.L. Oleinick, Photodynamic therapy with the [70] Q. Li, S. Lv, Z. Tang, M. Liu, D. Zhang, Y. Yang, X. Chen, A co-delivery system
phthalocyanine photosensitizer Pc 4: the case experience with preclinical based on paclitaxel grafted mPEG-b-PLG loaded with doxorubicin:
mechanistic and early clinical-translational studies, Toxicol. Appl. Pharmacol. preparation, in vitro and in vivo evaluation, Int. J. Pharm. 471 (2014) 412–420.
224 (2007) 290–299. [71] Y. Zhu, J. Li, S. Kanvinde, Z. Lin, S. Hazeldine, R.K. Singh, D. Oupicky,
[64] M. Bio, P. Rajaputra, G. Nkepang, S.G. Awuah, A.M. Hossion, Y. You, Self-immolative polycations as gene delivery vectors and prodrugs targeting
Site-specific and far-red-light-activatable prodrug of combretastatin A-4 polyamine metabolism in cancer, Mol. Pharm. 12 (2015) 332–341.
using photo-unclick chemistry, J. Med. Chem. 56 (2013) 3936–3942.
[65] J. Fan, G. Fang, F. Zeng, X. Wang, S. Wu, Water-dispersible fullerene aggregates
as a targeted anticancer prodrug with both chemo- and photodynamic
therapeutic actions, Small 9 (2013) 613–621.

You might also like