Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Biomaterials 155 (2018) 145e151

Contents lists available at ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Development of a theranostic prodrug for colon cancer therapy by


combining ligand-targeted delivery and enzyme-stimulated activation
Amit Sharma a, 1, Eun-Joong Kim b, 1, Hu Shi c, 1, Jin Yong Lee c, ***, Bong Geun Chung b, **,
Jong Seung Kim a, *
a
Department of Chemistry, Korea University, Seoul 02841, South Korea
b
Department of Mechanical Engineering, Sogang University, Seoul 04107, South Korea
c
Department of Chemistry, Sungkyunkwan University, Suwon 16419, South Korea

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

Article history: The high incidence of colorectal cancer worldwide is currently a major health concern. Although con-
Received 24 August 2017 ventional chemotherapy and surgery are effective to some extent, there is always a risk of relapse due to
Received in revised form associated side effects, including post-surgical complications and non-discrimination between cancer
13 November 2017
and normal cells. In this study, we developed a small molecule-based theranostic system, Gal-Dox, which
Accepted 17 November 2017
Available online 20 November 2017
is preferentially taken up by colon cancer cells through receptor-mediated endocytosis. After cancer-
specific activation, the active drug Dox (doxorubicin) is released with a fluorescence turn-on response,
allowing both drug localization and site of action to be monitored. The therapeutic potency of Gal-Dox
Keywords:
Targeted drug delivery
was also evaluated, both in vivo and ex vivo, thus illustrating the potential of Gal-Dox as a colorectal
Colon cancer cancer theranostic with great specificity.
Theranostic © 2017 Elsevier Ltd. All rights reserved.
b-Galactosidase
Doxorubicin

1. Introduction complications, including the formation of blood clots in the legs,


bleeding at the surgical sites, and damage to nearby organs [7,8].
Colorectal cancer (CRC) is the third leading cause of cancer- Hence, there is an urgent need for the development of new ther-
related deaths worldwide, with over one million new cases in apeutic strategies for CRC, with improved clinical outcomes.
Europe and the US every year [1,2]. It is the second most common Conventional therapeutic strategies, involving the systemic de-
cancer affecting women, after breast cancer, and the third most livery of antitumor drugs, cannot distinguish between normal cells
common in men, after prostate and lung cancers, with the overall and proliferating cancerous cells, causing collateral damage to
risk for developing CRC being approximately 1 in 20 [3]. Typically, healthy tissue. To overcome such a formidable challenge, several
cancers confined to the colon are curable; however, if left un- targeted delivery systems have been developed, including small
treated, they spread to the regional lymph nodes and metastasize to molecule-based drug delivery systems (DDS), liposomes, polymeric
distant organs. Usually, the initial stages of the disease are curable systems, aptamers, and inorganic nanoparticles [9e16]. Ideally,
with surgical excision and combined chemotherapy. However, an certain criteria must be fulfilled for the successful development of a
appreciable proportion of CRC patients in early stage treatment drug delivery formulation. These include preferential targeting of
remain clinically remissive for a prolonged period of time, followed tumors, maximal accumulation in tumors in vivo, and finally, an
by approximately 50% chances of tumor recurrence with later efficient drug release profile within the tumor, with minimal
metastasis [4e6]. Surgery is accompanied by various leakage to contiguous normal cells. Nanoformulations usually take
advantage of leaky vasculature within the tumor mass for prefer-
ential accumulation, termed as enhanced permeability and reten-
* Corresponding author. tion effects (EPR). However, this significantly depends upon the
** Corresponding author. state of angiogenesis and vascularization of solid tumors [17e20].
*** Corresponding author. In past years, the design of nanomedicines in cancer therapeutics
E-mail addresses: jinylee@skku.edu (J.Y. Lee), bchung@sogang.ac.kr has been upgraded, by the incorporation of ligands that actively
(B.G. Chung), jongskim@korea.ac.kr (J.S. Kim).
1
target overexpressed receptors on tumors. However, there are still
These authors contributed equally to this work.

https://doi.org/10.1016/j.biomaterials.2017.11.019
0142-9612/© 2017 Elsevier Ltd. All rights reserved.
146 A. Sharma et al. / Biomaterials 155 (2018) 145e151

certain hurdles to be overcome to realize the final goal of precision analysis was a water-acetonitrile gradient (0e30 min; acetonitrile
medicine, including cost-effectiveness, pharmacokinetics, and from 5% to 85%) with a 1.0 mL/min flow rate.
disease-driven formulations [20]. This is where a small molecule-
based combination diagnosis and therapy can play a significant 2.2. Cellular uptake studies by fluorescence imaging and flow
role. In the past 20 years, several small molecule-based DDS have cytometry
been designed and developed, for use in early diagnosis, bio-
imaging, and therapeutics [10,11,15]. DDS are usually decorated To evaluate the intracellular delivery of Gal-Dox, confocal laser
with specific cancer targeting units, along with a drug activation scanning microscopy (CLSM) and flow cytometry were both
trigger moiety, linked to the chemotherapy drug. For drug activa- employed on HT-29, HepG2, and HeLa cells. Cells were seeded and
tion, several stimuli-responsive modes have been reported, incubated in m-slide 8 well chamber (ibidi, Munich, Germany) at a
including pH, elevated enzyme activity, redox status, light, and density of 1  104 cells per well. After overnight incubation, 10 mM
temperature [14,15,21,22]. From a pharmaco-economic perspective, Gal-Dox was treated into the culture media of well for various time
suitable targeting units and drug activation modes can be chosen points. All cells were fixed with 4% paraformaldehyde in PBS for
according to the cancer subtype, and synthetically designed. This 20 min at RT, following washing with phosphate-buffered saline
requires the simultaneous development of new DDS with easier (PBS) for three times. Nuclei and F-actin were counterstained with
accessibility and promising pharmacokinetics that can also be DAPI (Invitrogen, Molecular Probe, Eugene, OR, USA) and Alexa
translated to establish superior therapies for patients. Fluor 488 phalloidin (Invitrogen, Molecular Probe) diluted in 3%
The lysosomal enzyme, b-galactosidase (b-gal) is known to be BSA for 20 min, respectively. Furthermore, Lysotracker (Invitrogen,
upregulated in various cancer subtypes, including liver, lung, and Molecular Probe) was added to the HT-29 cells at a concentration of
ovarian cancers [23e25]. Several efforts have been made to visu- 50 nM for 30 min for the examination of co-localization with Gal-
alize the upregulated activity of b-gal real time in various preclin- Dox and lysosome. The fluorescence images were acquired using a
ical cancer models [24e26]. b-gal activity has also been utilized to CLSM (LSM 710, Carl Zeiss, and Germany). To further verify the Gal-
activate various cancer drug delivery formulations [27] and thera- Dox being targeted to ASGP receptor positive cell lines, the cellular
nostic agents [28]. Considering cancer as a robust system, the uptake was analyzed by flow cytometry (FACS Calibur, BD Bio-
preferential targeting and cellular uptake behavior of each sciences, USA). HT-29 and HeLa cells were incubated in 12-well
designed DDS can be influenced by several factors, including plate at a density of 4  105/well and pretreated with 1 mM of D-
complex structural construct, inherent molecular features, and galactose (Sigma-Aldrich, St. Louis, MO, USA). After incubation for
variation in expression levels of both targeting and trigger modules 4 h, the culture media were discarded, and the harvested cells were
[29e31]. Moreover, the overall safety and efficacy of complex DDS examined by a flow cytometry, following the treatment of 10 mM
can be influenced by several parameters including biodistribution, Gal-Dox for 2 h. The histogram plots were performed using FlowJo
potential immune toxicities and intended targeting that need to be software (TreStar, Olten, Switzerland) and mean fluorescence in-
addressed carefully in preclinical and clinical stages. tensity values were shown in the bar graph.
In exploring a new direction for the advancement of antitumor
chemotherapeutics, it is critical to understand the in vivo uptake 2.3. In vitro anti-cancer assay
and drug activation behavior of a novel DDS. This is because the
optimal drug availability within a tumor is governed by DDS- HT-29 and HeLa cells were plated to 96 well plates at a density of
associated features, which can alter the overall therapeutic effi- 1  104 cells per well for 12 h, and then treated with Dox and Gal-
cacy and toxicity profile of the incorporated drug [30,31]. As Dox at various concentrations for 24 h. Then, the culture media
detailed below, we focus on the b-gal enzyme as a target to both were replaced with fresh medium and the anti-cancer effect was
deliver and activate the DDS (Gal-Dox) for targeted drug delivery evaluated by a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazo-
in vitro and in vivo, in CRC tumor models. The galactose moiety of lium bromide (MTT, Roche Diagnostics GmbH, Mannheim, Ger-
Gal-Dox serves as an excellent targeting ligand for asialoglyco- many) colorimetric assay. MTT was added and incubated for an
protein (ASGP) receptors, which play a significant role in tran- additional 2 h at 37  C. The MTT absorbance was measured at
scriptional regulation at both cellular and molecular levels [32,33]. 570 nm using a microplate reader (EL800, Bio-Tek Instruments,
Doxorubicin (Dox) was chosen as a model anticancer drug. Gal-Dox Winooski, VT, USA) after the treatment of solubilization buffer
showed a significant ability to target ASGP receptors on colon (Roche Diagnostics) to dissolve formazan crystals. All experiments
cancer cells, and enhanced activation behavior via lysosomal b-gal were carried out with three replicates.
enzyme. We also validated the potential of Gal-Dox in a mouse
tumor model and demonstrated its potential for effective cancer 2.4. In vivo tumor targeting and anti-cancer effects
therapy.
For in vivo tumor-targeted imaging and anti-tumor activity,
2. Material and methods colorectal cancer xenografts were established by subcutaneously
inoculating six-week-old male BALB/c nude mice (Orient Bio,
2.1. UV/vis, fluorescence spectroscopy and high performance liquid Sungnam, Korea) with HT-29 cells. All in vivo experimental pro-
chromatography methods cedures involving animals were approved by the Korea University
Institutional Animal Care and Use Committee (IACUC). A total of
A stock solution of Gal-Dox was prepared in DMSO and for 1  106 viable HT-29 cells were injected into the flanks of nude
preliminary solution studies, 10 mM working solution in phosphate mice after anesthetization. When the tumors reached about
saline buffer (PBS) buffer solution (pH ¼ 7.4, 37  C) containing 1% 5e10 mm in diameter, Dox or Gal-Dox was injected intravenously
DMSO was used. Excitation was done at 480 nm with all excitation into the tail vein in a single dose of 5 mg/kg. Whole-body fluores-
and emission slit widths at 5 nm. The total volume for b-Galacto- cence images were continuously monitored and the dissected
sidase response and pH-dependent response tests was fixed at major organs (i.e, liver, kidney, heart, lung, tumor) were visualized
3.0 mL. For HPLC analyses, a reverse-phase column (C18, 5 mm, or analyzed after sacrificing at 6 h, 24 h, 48 h, using an in vivo
Waters) equipped YL9101S (YL-Clarity) instrument was used. A imaging system (Maestro, CRi Inc., Woburn, MA, USA). Moreover,
UVevis detector (480 nm) was used. The eluent used for each tumors were fixed in 4% paraformaldehyde solution, embedded
A. Sharma et al. / Biomaterials 155 (2018) 145e151 147

routinely in paraffin, sectioned at 5 mm thickness, mounted using intermediates and products were well characterized by 1H/13C
with DAPI containing Vecta shield (Vector Laboratories, CA, USA). nuclear magnetic resonance (NMR) and electrospray ionization
To validate the therapeutic efficacy of Gal-Dox, HT-29-inoculated mass spectrometry (Figs. S14e25).
xenograft mice were subcutaneously injected with Saline, and To obtain a deeper insight into binding sites at the atomic level,
3 mg/kg dose of free Dox and Gal-Dox (xenografts, n ¼ 5) four times docking and molecular dynamics (MD) simulations were studied,
every other day. Tumor volumes and body weights were measured including b-gal and Gal-Dox interactions. The molecular binding
every 3 day using a caliper and terminated at 36 days. For in vivo site between Gal-Dox and b-gal, and the decomposed binding free
toxicity study, blood samples were collected from all the mice for energies, are shown in Fig. 1B and S2, respectively. The calculated
the measurement of serum biochemical parameters including binding free energy between Gal-Dox and b-gal was found to
blood urea nitrogen (BUN), lactate dehydrogenase (LDH), aspartate be 20.22 kcal/mol. According to the binding interaction calcula-
aminotransferase (AST), and alanine aminotransferase (ALT) at day tion, four significant interacting residues in b-galactosidase were
30 after tumor inoculation. Also, one mouse from each group was observed, specified as N90 (red), P501 (orange), H528 (blue) and
sacrificed, and main organs such as heart, lung, liver, spleen, and W987 (pink). Considering the calculated binding free energies
kidney, were submitted for routine haematoxylin and eosin (H&E) among whole residues, W987 showed the strongest interaction
staining. with the Gal-Dox ligand. Details of the simulation are provided in
the supporting information (Figs. S2e3).
3. Results and discussion To obtain insight into the preliminary stability and activation
mode of Gal-Dox, the compound was incubated in phosphate
3.1. Synthesis, fluorescence response, and mechanism of Gal-Dox buffered saline (PBS, pH 7.4, 37  C) for 24 h. No significant
activation decomposition was observed during this period (determined by
fluorescence microscopy and high performance liquid chromatog-
Gal-Dox was synthesized by following a series of steps in good raphy, HPLC), suggesting sufficient chemical stability (Fig. 1 and S4).
to excellent yield (Fig. 1A and S1, detailed in the supporting infor- Gal-Dox exhibited a weak fluorescence emission at 590 nm (exci-
mation). Following a reported procedure, galactosidase linker in- tation wavelength lexc. ¼ 480 nm). Interestingly, exposure of Gal-
termediate 2 was synthesized. Further alcohol activation by p- Dox (10 mM) to b-gal (1 U/mL) resulted in cleavage of the galacto-
nitrobenzyl chloroformate and base-mediated substitution with sidase moiety, followed by the 1,6-elimination route to release free
free Dox resulted in the formation of intermediate 4. The acetyl Dox with simultaneous fluorescence enhancement at 590 nm
groups were de-protected with sodium methoxide in anhydrous (Fig. 1C and D). Consequently, we used the fluorescence enhance-
methanol and treated with cation exchange resin, resulting in the ment at 590 nm as an activation response, for the observation of
formation of the final product, Gal-Dox. All synthesized direct Dox release in further studies. The Gal-Dox activation and

Fig. 1. Chemical structure, fluorescence response and activation mode for Gal-Dox. A, Chemical structure of Gal-Dox. B, Molecular binding of Gal-Dox with the b-gal enzyme. C,
Fluorescence enhancement of Gal-Dox (10 mM) upon exposure to the b-gal enzyme (1 U/mL) (excitation wavelength lexc. ¼ 480 nm). D, Time-dependent fluorescence enhancement
upon b-gal action (10 mM, 1 U/mL enzyme, 37  C). Fluorescence enhancement is directly related to cumulative Dox release. E, RP-HPLC curves of Gal-Dox (10 mM) treated with b-gal
(1 U/mL) at 37  C for 2 h. F, Mode of activation of Gal-Dox upon b-gal action.
148 A. Sharma et al. / Biomaterials 155 (2018) 145e151

drug release mechanisms were also determined using mass spec- same time. Fluorescence in Gal-Dox-treated cells was reached a
troscopy (MS) and HPLC (Fig. 1E and S7). The HPLC profile showed a plateau after 6 h (Fig. S8). To further verify the ASGP receptor-
peak with an elution time of 20.4 min, corresponding to Gal-Dox. targeted cellular uptake of Gal-Dox, a competition assay was con-
Upon treatment with b-gal, a new peak appeared at 15.3 min, ducted by flow cytometry analysis. Initially, HT-29 cells were pre-
corresponding to free Dox. Altogether, treatment of Gal-Dox with treated 100 times with free galactose as a competitor, followed by
b-gal resulted in the complete release of active Dox, over a time the addition of Gal-Dox at a concentration of 10 mM with incubation
period of 2 h. Given the disparities in pH and the presence of other for 2 h. There was no significant difference in fluorescence intensity
bio-analytes between cancerous and normal healthy cells, we next between HeLa cells treated with excess galactose and control un-
evaluated the fluorescence behavior of Gal-Dox, in the presence of treated cells, whereas the fluorescence intensity in HT-29 cells
various bio-analytes, and at a range of pHs. No appreciable change pretreated with galactose remarkably decreased (Fig. S10). Alto-
in the fluorescence signal of Gal-Dox was observed at different pHs, gether, these results support that the cellular uptake of Gal-Dox can
or in the presence of various bio-analytes, including reductase, be specifically achieved by ASGP receptor-mediated endocytosis. To
GSH, H2O2, and amino acids (Figs. S5e6). These results suggested the best of our knowledge, this is the first report citing the specific
that Gal-Dox should exhibit high stability under biological condi- targeting properties of a small molecule-based prodrug system to
tions. Gal-Dox exhibited distinct fluorescence enhancement ASGP receptor-overexpressing colon cancer cells, through cellular
(590 nm) only upon treatment with the b-gal enzyme. uptake via receptor-mediated endocytosis.

3.2. ASGP receptor-targeted delivery of Gal-Dox into colorectal 3.3. b-Galactosidase mediated activity of Gal-Dox
cancer cells
As Gal-Dox exhibited b-gal-triggered Dox release, we next
The specific interaction between the galactose residue and ASGP confirmed enzyme-specific Gal-Dox activation via the reduction of
receptors has been used for hepatocyte-targeted DDS via receptor- b-gal activity within the cells. To knock down the expression of b-
mediated endocytosis [34]. In this study, Gal-Dox was employed as gal, an siRNA strategy was attempted to investigate the activity of
a theranostic prodrug against the intestinal ASGP receptors in colon Gal-Dox in the presence of the low level of endogenous b-gal
cancer [35]. To confirm the selective delivery of Gal-Dox to colon within HT-29 cells. For this, HT-29 cells were treated with various
cancer cells overexpressing ASGP receptors, we evaluated its tar- concentrations of b-gal siRNA for 48 h, and the optimal siRNA
geting ability both in HT-29 cells, a colon adenocarcinoma cell line, concentration (50 nM) was determined by western blot analysis for
and in HepG2 cells, a well-known cancer cell line bearing the ASGP further studies (Fig. 3A). After b-gal expression interference by
receptor. As shown in Fig. 2, HT-29 and HepG2 cells showed pro- pretreated siRNA (50 nM) for 48 h, HT-29 cells were incubated with
nounced intracellular fluorescence, while relatively low fluores- 10 mM of Gal-Dox for 0.5 h and 12 h. While fluorescence decreased
cence signals were detected in HeLa cells, an ASGP receptor- in HT-29 cells with low b-gal expression, we observed relatively
negative cell line (control) [36,37], even after 2 h incubation. Co- strong fluorescence in control siRNA-untreated cells (Fig. 3B and
localization experiments confirmed that Gal-Dox was specifically Fig. S11). Enzyme-specific activation of Gal-Dox was further
localized with lysosomes of HT-29 cells, in which b-gal activity confirmed by flow cytometric analysis, which provided a compar-
occurs (Figs. S8e9), Especially, intracellular fluorescence was ison between the mean fluorescence intensity of Gal-Dox-treated
clearly observed 30 min after treatment because of its targeting HT-29 cells, with and without siRNA knockdown (Fig. 3C). Collec-
ability for ASGP receptors, while free Dox was not detectable at the tively, these siRNA silencing results suggest that the activation of

Fig. 2. Targeted cellular uptake of Gal-Dox through ASGP receptor-mediated endocytosis. HT-29, HepG2 and HeLa cells were incubated with 10 mM Gal-Dox for 2 h, fixed with 4%
paraformaldehyde, and counterstained with DAPI and Alexa Fluor 488-Phalloidin to visualize nuclei (blue) and F-actin (green), respectively. Confocal microscopy images of Gal-Dox
(red) were obtained with a laser using an excitation wavelength of 480 nm and an emission wavelength of 560e590 nm. Scale bar: 20 mm. (For interpretation of the references to
colour in this figure legend, the reader is referred to the web version of this article.)
A. Sharma et al. / Biomaterials 155 (2018) 145e151 149

Fig. 3. Gal-Dox activation through b-galactosidase-mediated cleavage. (A) Western blot analysis of b-gal knock-down by siRNA. HT-29 cells were transfected with b-gal siRNA
(25 nM and 50 nM) for 48 h. The silencing effect of siRNA was analyzed by western blot, using an anti-b-gal antibody and b-actin antibody as a loading control. (B) After transfection
of siRNA (50 nM) directed against b-gal for 48 h, HT-29 cells were treated with 10 mM Gal-Dox for 0.5 h, and examined with confocal microscopy. Filter sets: Doxorubicin (lex: 480/
lem: 560e590), F-actin (lex: 495/lem: 518 and DAPI (lex: 358/lem: 461) Scale bar: 20 mm. (C) Flow cytometry analysis was conducted with HT-29 cells treated with 10 mM Gal-Dox
after transfection of 50 nM of b-gal siRNA.

Gal-Dox could be achieved by upregulated b-gal-responsive trig- Gal-Dox at concentrations ranging from 1 to 200 mM, for 24 h.
gering. This sophisticated system permits simultaneous Dox release Although Gal-Dox did not significantly reduce viability even at the
and fluorescence activation-based imaging, for greater precision in maximum dose in both cells, similar behavior was observed for
monitoring the drug activation event and its location. both free Dox- and Gal-Dox-treated HT-29 cells (Fig. 4A). However,
a remarkable difference in cytotoxicity was detected between free
Dox- and Gal-Dox-treated HeLa cells (Fig. 4B). The comparison of
3.4. Anticancer effects of Gal-Dox with cell selectivity
IC50 values indicated that Gal-Dox-treated HT-29 cells possessed
approximately 3-fold more potent therapeutic effects than HeLa
The cytotoxicity of Gal-Dox was evaluated in HT-29 and HeLa
cell (Table S1). These results demonstrate that targeted Gal-Dox
cells using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazo-
delivery via the overexpressed ASGP receptors in colon cancer cells
lium bromide (MTT) assay, as its anti-proliferative activity can be
not only improved cellular uptake, but also enhanced its anticancer
determined by selective cellular uptake via ASGP receptor-
effect.
mediated endocytosis. Both cells were treated with free Dox and

Fig. 4. Anticancer effects of Gal-Dox in HT-29 cancer cells (A) and HeLa (B) cells. Both cells were treated with Gal-Dox and Dox as a positive control, at various concentrations for
24 h. Cell viability was then determined using the MTT assay.
150 A. Sharma et al. / Biomaterials 155 (2018) 145e151

3.5. In vivo/ex vivo diagnosis and chemotherapeutic in xenograft in vivo toxicities were observed during this study (Fig. S13). We
mouse models believe that this remarkable therapeutic response of Gal-Dox re-
flects its efficient tumor uptake behavior by the galactosidase unit,
To investigate the antitumor activity of Gal-Dox, in vivo tumor- which not only serves to improve cancer targeting, but also as an
targeting ability was initially confirmed in HT-29 tumor-bearing efficient prodrug activation mode to deliver the active Dox.
mice. For bio-distribution studies using an in vivo fluorescence
imaging system, saline, free Dox and Gal-Dox were intravenously
injected into mice at a single dose of 3 mg/kg, and real-time whole- 4. Conclusions
body fluorescence imaging was performed 2d after administration.
While the tumor-targeted accumulation of Gal-Dox was continu- In conclusion, a small molecule-based theranostic prodrug, Gal-
ously visualized by enhanced fluorescence signal up to 48 h in the Dox, was successfully developed for colon cancer chemotherapy,
HT-29 tumor, free Dox was observed to slightly clear from the tu- using receptor-mediated targeting and enzyme-responsive activa-
mor site in 24 h post injection (Fig. 5 and S12A). After sacrificing the tion strategies. The imaging properties and therapeutic efficacy of
mice 48 h later, the tumor tissues were harvested and subsequently Gal-Dox in colorectal cancer models have been successfully
subjected to ex vivo fluorescence imaging (Fig. S12B). Collectively, demonstrated both in vitro and in vivo. Gal-Dox is preferentially
whole-body fluorescence imaging results were comparable to taken up by HT-29 cancer cells through ASGP receptor-mediated
those for the dissected tumor tissues (Fig. 5B) and biodistribution endocytosis and was activated by elevated lysosomal b-gal
studies (Fig. S12C and D) from saline-, free Dox-, and Gal-Dox- enzyme. In particular, the theranostic activation event is accom-
treated mice. The in vivo therapeutic efficacy of Gal-Dox was also panied by a simultaneous fluorescence turn-on response. This also
evaluated in nude mice bearing HT-29 cell-inoculated tumors, supports the additional possibility of monitoring both the site of
following intravenous administration of Gal-Dox (5 mg/kg, four drug activation, and the therapeutic response towards the tumor,
times every second day), and controls including saline and free Dox. during early stages of treatment. Compared to the pre-reported
As shown in Fig. 5C, tumor growth was significantly retarded in multi-component DDS systems incorporating Galactosidase as
Dox- and Gal-Dox-injected xenograft mice, compared to saline- trigger, drugs and cancer-specific targeting units altogether in a
injected mice. However, administration of Gal-Dox showed complex design, Gal-Dox with simple design possesses remarkable
remarkable tumor growth inhibition (53.1%) compared to free Dox tumor specific targeting and therapeutic potential in ASGP receptor
treatment (34.9%, see Fig. 5D). Although Dox has been known to positive cell lines. Additionally, complexity of pre-designed DDS as
have several adverse effects such as cardiotoxicity, no significant multi-component requires detailed design and orthogonal analysis
with reproducible scale-up process for achieving a consistent

Fig. 5. Tumor-targeted accumulation and in vivo antitumor activity of Gal-Dox in nude mice bearing HT-29 cancer xenografts. (A) In vivo fluorescence imaging of Gal-Dox-treated
HT-29-xenograft nude mice. Mice bearing HT29 subcutaneous xenografts were intravenously injected with 5 mg/kg of Gal-Dox and in vivo fluorescent images were obtained 48 h
after treatment using the Maestro in vivo imaging system. (B) Fluorescence images in paraffin-embedded tumor tissue. Scale bar: 20 mm (C) Tumor growth inhibition in the various
groups. HT-29-bearing mice were intravenously injected with saline, Dox, or Gal-Dox at a dose of 3 mg/kg, four times every second day. Tumor volume was measured twice per
week. Data are shown as mean ± SE. *: P < 0.05 versus Dox; ***: P < 0.001 versus saline, determined using Student's t-test. (D) Tumor growth inhibition after treatment with Dox
and Gal-Dox at 36 days.
A. Sharma et al. / Biomaterials 155 (2018) 145e151 151

commercial product for intended physiological properties, phar- effects of carboplatin by enhancing tumor vascularization and perfusion,
Theranostics 5 (2015) 905e918.
macological profiles, and biological response. Gal-Dox with a sim-
[18] W. Jiang, Y. Huang, Y. An, B.Y. Kim, Remodeling tumor vasculature to enhance
ple design, can be accessed with ease synthetically from pharmaco- delivery of intermediate-sized nanoparticles, ACS Nano 9 (2015) 8689e8696.
economic point of view also. Hence Gal-Dox shows the potential as [19] R.K. Jain, T. Stylianopoulos, Delivering nanomedicine to solid tumors, Nat. Rev.
an ideal theranostic system in future personalized cancer Clin. Oncol. 7 (2010) 653e664.
[20] J.I. Hare, T. Lammers, M.B. Ashford, S. Puri, G. Storm, S.T. Barry, Challenges and
chemotherapeutics. strategies in anti-cancer nanomedicine development: an industry perspective,
Adv. Drug. Deliv. Rev. 108 (2017) 25e38.
Acknowledgements [21] S. Nowag, R. Haag, pH-responsive micro- and nanocarrier systems, Angew.
Chem. Int. Ed. Engl. 53 (2014) 49e51.
[22] S. Jin, J. Wan, L. Meng, X. Huang, J. Guo, L. Liu, C. Wang, Biodegradation and
This research was supported by the CRI project (No. 2009- toxicity of protease/redox/pH stimuli-eesponsive PEGlated PMAA nano-
0081566, J.S.K.) and the Bio & Medical Technology Development hydrogels for targeting drug delivery, ACS Appl. Mater. Interfaces 7 (2015)
19843e19852.
Program (No. 2015M3A9D7030461, B.G.C.) of the National Research [23] S.K. Chatterjee, M. Bhattacharya, J.J. Barlow, Glycosyltransferase and glycosi-
Foundation funded by the Ministry of Science, ICT and Future dase activities in ovarian cancer patients, Cancer Res. 39 (1979) 1943e1951.
Planning of Korea. [24] D. Asanuma, M. Sakabe, M. Kamiya, K. Yamamoto, J. Hiratake, M. Ogawa,
N. Kosaka, P.L. Choyke, T. Nagano, H. Kobayashi, Y. Urano, Sensitive b-galac-
tosidase-targeting fluorescence probe for visualizing small peritoneal meta-
Appendix A. Supplementary data static tumours in vivo, Nat. Commun. 6 (2015) 6463.
[25] E.J. Kim, R. Kumar, A. Sharma, B. Yoon, H.M. Kim, H. Lee, K.S. Hong, J.S. Kim,
In vivo imaging of b-galactosidase stimulated activity in hepatocellular car-
Supplementary data related to this article can be found at
cinoma using ligand-targeted fluorescent probe, Biomaterials 122 (2017)
https://doi.org/10.1016/j.biomaterials.2017.11.019. 83e90.
[26] L. Liu, R.P. Mason, Imaging beta-galactosidase activity in human tumor xe-
References nografts and transgenic mice using a chemiluminescent substrate, PLoS One 5
(2010), e12024.
[27] T. Legigan, J. Clarhaut, I. Tranoy-Opalinski, A. Monvoisin, B. Renoux,
[1] R.L. Segel, et al., Colorectal cancer statistics, 2017, CA Cancer J. Clin. 67 (2017) M. Thomas, A. Le Pape, S. Lerondel, S. Papot, The first generation of b-galac-
177e193. tosidase-responsive prodrugs designed for the selective treatment of solid
[2] F.A. Haggar, R.P. Boushey, Colorectal cancer epidemiology: incidence, mor- tumors in prodrug monotherapy, Angew. Chem. Int. Ed. Engl. 51 (2012)
tality, survival, and risk factors, Clin. Colon Rectal Surg. 22 (2009) 191e197. 11606e11610.
[3] C.E. Kistler, Colorectal-cancer incidence and mortality after screening, N. Engl. [28] raudeau, P. Poinot, I. Tranoy-Opalinski, J. Clarhaut, B. Renoux,
J. Alsarraf, E. Pe
J. Med. 369 (2009) 2354e2355. S. Papot, A dendritic b-galactosidase-responsive folate-monomethylauristatin
[4] S.D. Markowitz, D.M. Dawson, J. Willis, J.K. Willson, Focus on colon cancer, E conjugate, Chem. Commun. (Camb) 51 (2015) 15792e15795.
Cancer Cell 1 (2002) 233e236. [29] E. Vlashi, L.E. Kelderhouse, J.E. Sturgis, P.S. Low, Effect of folate-targeted
[5] H.G. Welch, D.J. Robertson, Colorectal cancer on the decline e why screening nanoparticle size on their rates of penetration into solid tumors, ACS Nano
can't explain it all, N. Engl. J. Med. 374 (2016) 1605e1607. 7 (2013) 8573e8582.
[6] M. Riihima €ki, A. Hemminki, J. Sundquist, K. Hemminki, Patterns of metastasis
[30] J.H. Jang, W.R. Kim, A. Sharma, S.H. Cho, T.D. James, C. Kang, J.S. Kim, Targeted
in colon and rectal cancer, Sci. Rep. 6 (2016) 29765. tumor detection: guidelines for developing biotinylated diagnostics, Chem.
[7] G. Grosso, A. Biondi, S. Marventano, A. Mistretta, G. Calabrese, F. Basile, Major Commun. 53 (2017) 2154e2157.
postoperative complications and survival for colon cancer elderly patients, [31] A. Ivetac, J.A. McCammon, Mapping the druggable allosteric space of G-pro-
BMC Surg. 12 (Suppl 1) (2012) S20. tein coupled receptors: a fragment-based molecular dynamics approach,
[8] J. Kountouras, C. Zavos, D. Chatzopoulos, Therapy for colorectal cancer, N. Engl. Chem. Biol. Drug. Des. 76 (2010) 201e217.
J. Med. 352 (2005) 1820e1821. [32] P.P. Breitfeld, C.F. Simmons Jr., G.J. Strous, H.J. Geuze, A.L. Schwartz, Cell
[9] P.T. Wong, S.K. Choi, Mechanisms of drug release in nanotherapeutic delivery biology of the asialoglycoprotein receptor system: a model of receptor-
systems, Chem. Rev. 115 (2015) 3388e3432. mediated endocytosis, Int. Rev. Cytol. 97 (1985) 47e95.
[10] M.H. Lee, A. Sharma, M.J. Chang, J. Lee, S. Son, J.L. Sessler, C. Kang, J.S. Kim, [33] M.D. Bider, M. Spiess, Ligand-induced endocytosis of the asialoglycoprotein
Fluorogenic reaction-based prodrug conjugates as targeted theranostics, receptor: evidence for heterogeneity in subunit oligomerization, FEBS Lett.
Chem. Soc. Rev. (2017), https://doi.org/10.1039/C7CS00557A. 434 (1998) 37e41.
[11] M. Srinivasarao, C.V. Galliford, P.S. Low, Principles in the design of ligand- [34] Y. Zou, Y. Song, W. Yang, F. Meng, H. Liu, Z. Zhong, Galactose-installed photo-
targeted cancer therapeutics and imaging agents, Nat. Rev. Drug. Discov. 14 crosslinked pH-sensitive degradable micelles for active targeting chemo-
(2015) 203e219. therapy of hepatocellular carcinoma in mice, J. Control Release 193 (2014)
[12] S. Bhattacharyya, R.A. Kudgus, R. Bhattacharya, P. Mukherjee, Inorganic 154e161.
nanoparticles in cancer therapy, Pharm. Res. 28 (2011) 237e259. [35] J.Z. Mu, M. Gordon, J.S. Shao, D.H. Alpers, Apical expression of functional
[13] X. Wu, J. Chen, M. Wu, J.X. Zhao, Aptamers: active targeting ligands for cancer asialoglycoprotein receptor in the human intestinal cell line HT-29, Gastro-
diagnosis and therapy, Theranostics 5 (2015) 322e344. enterology 113 (1997) 1501e1509.
[14] V.P. Torchilin, Multifunctional, stimuli-sensitive nanoparticulate systems for [36] Y.M. Li, S.C. Xu, J. Li, K.Q. Han, H.F. Pi, L. Zheng, G.H. Zuo, X.B. Huang, H.Y. Li,
drug delivery, Nat. Rev. Drug. Discov. 13 (2014) 813e827. H.Z. Zhao, Z.P. Yu, Z. Zhou, P. Liang, Epithelial-mesenchymal transition
[15] J.H. Jang, H. Lee, A. Sharma, S.M. Lee, T.H. Lee, C. Kang, J.S. Kim, Indomethacin- markers expressed in circulating tumor cells in hepatocellular carcinoma
guided cancer selective prodrug conjugate activated by histone deacetylase patients with different stages of disease, Cell Death Dis. 4 (2013), e831.
and tumour-associated protease, Chem. Commun. 52 (2016) 9965e9968. [37] Y. Tao, J. He, M. Zhang, Y. Hao, J. Liub, P. Ni, Galactosylated biodegradable
[16] M.H. Lee, E.J. Kim, H. Lee, H.M. Kim, M.J. Chang, S.Y. Park, K.S. Hong, J.S. Kim, poly(ε-caprolactone-co-phosphoester) random copolymer nanoparticles for
J.L. Sessler, Liposomal texaphyrin theranostics for metastatic liver cancer, potent hepatoma-targeting delivery of doxorubicin, Polym. Chem. 5 (2014)
J. Am. Chem. Soc. 138 (2016) 16380e16387. 3443e3452.
[17] D. Doleschel, et al., Erythropoietin improves the accumulation and therapeutic

You might also like