dyes and pigments - bntl.sogang.ac.krbntl.sogang.ac.kr/bntl/research/documents/dyes and... ·...

6
Contents lists available at ScienceDirect Dyes and Pigments journal homepage: www.elsevier.com/locate/dyepig Molecular theranostic based on esterase-mediated drug activation for hepatocellular carcinoma Amit Sharma a,1 , Eun-Joong Kim b,1 , Seokgyu Mun c , Myung Sun Ji a , Bong Geun Chung d,∗∗ , Jong Seung Kim a,a Department of Chemistry, Korea University, Seoul, 02841, South Korea b Research Center, Sogang University, Seoul, 04107, South Korea c Department of Biomedical Engineering, Sogang University, Seoul, 04107, South Korea d Department of Mechanical Engineering, Sogang University, Seoul, 04107, South Korea ARTICLE INFO Keywords: Cancer Hepatocellular carcinoma Theranostic Targeted therapy Drug delivery system ABSTRACT In past few years, cancer specic targeted drug formulations have shown some hope of improving the ther- apeutics with minimized associated side eects of chemotherapy. However, the benets has proven modest due to lack of systems that can be assessed easily from parent drugs while maintaining the same features of cancer associated prodrug activation. We developed a small molecule-based, carboxylesterase responsive theranostic, EDOX, with tumor-specic enzymatic activation for targeted delivery of the anticancer drug, Doxorubicin (Dox), to hepatocellular carcinoma (HCC) cells. Easy synthetic access, physiological stability, tumor-selective activa- tion, and sustained drug release make EDOX an excellent candidate for use as a next-generation drug delivery system for HCC. 1. Introduction Cancer includes a range of diseases related to the dysregulated growth of malignant cells, often with the ability to invade distant tis- sues and organs [1]. According to an estimate by the World Health Organization (WHO), cancer-related mortality has witnessed a steep increase in the last decades and over 13.1 million cancer-related annual deaths are expected by 2030 [2]. In the last few years, a better un- derstanding of cancer biology, as well as improved diagnostic and therapeutic tools, have led to a noticeable decrease in mortality rates [3]. Currently, available cancer treatments include surgical interven- tion, radiation therapy, photodynamic therapy, chemotherapy or a combination of these options [47]. Most conventional chemother- apeutics interfere with DNA synthesis and mitosis, causing the death of proliferating cancer cells [8]. However, these agents are usually non- selective and ultimately result in the damage of healthy cells and tis- sues, with severe undesired side eects including hair loss, appetite loss, and nausea. It has been calculated that the side eects of che- motherapeutics strongly contribute to high mortality in cancer patients [9]. Hepatocellular carcinoma (HCC), the fth most frequently diagnosed cancer type worldwide, is second cause of cancer-associated death in males and the sixth in females [10,11]. Due to the long latency of HCC, most patients are already in advanced stages of disease at the time of rst diagnosis, and chemotherapy is the only available option. It was observed that chemotherapeutic agents accumulating in the tumor may only be 5%10% of the dosage reaching normal organs [12]. Due to such inecient delivery, the concentration of active drugs at the HCC site is too low to be eective. Additionally, intrinsic and acquired multidrug resistance (MDR) rates are high in HCC, resulting in cancer reoccurrence with poor survival and overall life quality [13]. Thus, new strategies to improve HCC response to treatment are strongly desired. Carboxylesterase (CE) is an enzyme involved in the hydrolytic me- tabolism of various drugs including carboxylic acid esters, amides, and thioesters. There are two main types of human carboxylesterases, hCE1, and hCE2, with distinct roles in the hydrolysis of specic drugs [14,15]. Particularly, CE is known to be highly expressed in the liver and com- monly related to several liver-associated diseases, including hepatic steatosis, hyperlipidemia, and HCC [1620]. In past few years, extensive eorts have been made to pursue ver- satile drug carriers such as small molecules, nanoparticles, polymers, inorganic materials, polymeric hydrogels, lipids and https://doi.org/10.1016/j.dyepig.2018.12.026 Received 24 October 2018; Received in revised form 20 November 2018; Accepted 15 December 2018 Corresponding author. ∗∗ Corresponding author. E-mail addresses: [email protected] (B.G. Chung), [email protected] (J.S. Kim). 1 These authors contributed equally. Dyes and Pigments 163 (2019) 628–633 Available online 17 December 2018 0143-7208/ © 2018 Elsevier Ltd. All rights reserved. T

Upload: others

Post on 01-Jun-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Dyes and Pigments - bntl.sogang.ac.krbntl.sogang.ac.kr/bntl/Research/Documents/Dyes and... · Biosciences, UK) as the substrate, and then expose the X-ray film in the dark room

Contents lists available at ScienceDirect

Dyes and Pigments

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

Molecular theranostic based on esterase-mediated drug activation forhepatocellular carcinoma

Amit Sharmaa,1, Eun-Joong Kimb,1, Seokgyu Munc, Myung Sun Jia, Bong Geun Chungd,∗∗,Jong Seung Kima,∗

a Department of Chemistry, Korea University, Seoul, 02841, South Koreab Research Center, Sogang University, Seoul, 04107, South Koreac Department of Biomedical Engineering, Sogang University, Seoul, 04107, South KoreadDepartment of Mechanical Engineering, Sogang University, Seoul, 04107, South Korea

A R T I C L E I N F O

Keywords:CancerHepatocellular carcinomaTheranosticTargeted therapyDrug delivery system

A B S T R A C T

In past few years, cancer specific targeted drug formulations have shown some hope of improving the ther-apeutics with minimized associated side effects of chemotherapy. However, the benefits has proven modest dueto lack of systems that can be assessed easily from parent drugs while maintaining the same features of cancerassociated prodrug activation. We developed a small molecule-based, carboxylesterase responsive theranostic,EDOX, with tumor-specific enzymatic activation for targeted delivery of the anticancer drug, Doxorubicin (Dox),to hepatocellular carcinoma (HCC) cells. Easy synthetic access, physiological stability, tumor-selective activa-tion, and sustained drug release make EDOX an excellent candidate for use as a next-generation drug deliverysystem for HCC.

1. Introduction

Cancer includes a range of diseases related to the dysregulatedgrowth of malignant cells, often with the ability to invade distant tis-sues and organs [1]. According to an estimate by the World HealthOrganization (WHO), cancer-related mortality has witnessed a steepincrease in the last decades and over 13.1 million cancer-related annualdeaths are expected by 2030 [2]. In the last few years, a better un-derstanding of cancer biology, as well as improved diagnostic andtherapeutic tools, have led to a noticeable decrease in mortality rates[3]. Currently, available cancer treatments include surgical interven-tion, radiation therapy, photodynamic therapy, chemotherapy or acombination of these options [4–7]. Most conventional chemother-apeutics interfere with DNA synthesis and mitosis, causing the death ofproliferating cancer cells [8]. However, these agents are usually non-selective and ultimately result in the damage of healthy cells and tis-sues, with severe undesired side effects including hair loss, appetiteloss, and nausea. It has been calculated that the side effects of che-motherapeutics strongly contribute to high mortality in cancer patients[9].

Hepatocellular carcinoma (HCC), the fifth most frequently

diagnosed cancer type worldwide, is second cause of cancer-associateddeath in males and the sixth in females [10,11]. Due to the long latencyof HCC, most patients are already in advanced stages of disease at thetime of first diagnosis, and chemotherapy is the only available option. Itwas observed that chemotherapeutic agents accumulating in the tumormay only be 5%–10% of the dosage reaching normal organs [12]. Dueto such inefficient delivery, the concentration of active drugs at theHCC site is too low to be effective. Additionally, intrinsic and acquiredmultidrug resistance (MDR) rates are high in HCC, resulting in cancerreoccurrence with poor survival and overall life quality [13]. Thus, newstrategies to improve HCC response to treatment are strongly desired.

Carboxylesterase (CE) is an enzyme involved in the hydrolytic me-tabolism of various drugs including carboxylic acid esters, amides, andthioesters. There are two main types of human carboxylesterases, hCE1,and hCE2, with distinct roles in the hydrolysis of specific drugs [14,15].Particularly, CE is known to be highly expressed in the liver and com-monly related to several liver-associated diseases, including hepaticsteatosis, hyperlipidemia, and HCC [16–20].

In past few years, extensive efforts have been made to pursue ver-satile drug carriers such as small molecules, nanoparticles, polymers,inorganic materials, polymeric hydrogels, lipids and

https://doi.org/10.1016/j.dyepig.2018.12.026Received 24 October 2018; Received in revised form 20 November 2018; Accepted 15 December 2018

∗ Corresponding author.∗∗ Corresponding author.E-mail addresses: [email protected] (B.G. Chung), [email protected] (J.S. Kim).

1 These authors contributed equally.

Dyes and Pigments 163 (2019) 628–633

Available online 17 December 20180143-7208/ © 2018 Elsevier Ltd. All rights reserved.

T

Page 2: Dyes and Pigments - bntl.sogang.ac.krbntl.sogang.ac.kr/bntl/Research/Documents/Dyes and... · Biosciences, UK) as the substrate, and then expose the X-ray film in the dark room

biomacromolecular motifs for the selective delivery of chemother-apeutic agents at tumor sites [21–28]. These systems may result inimproved drug specificity and reduced side effects, minimizing multi-drug resistance (MDR). Although several strategies for passive drugtargeting have been developed, only a few of them have made their wayinto clinical trials [29]. Recently, active targeting strategies in nano-medicine have shown interesting results, albeit physiological barrierssuch as tumor heterogeneity, deep penetration, hypoxia, and en-dosomal escape are still major hurdles to their optimal therapeuticapplication [30,31]. From a biopharmaceutical standpoint, the idealsystem should preserve the drug from undesired chemical or enzymaticdegradation for a sufficient time to allow for payload delivery at thedesired target site. Small molecule-based systems hold the potential toovercome the above-mentioned drawbacks as they exhibit several ad-vantages over nano-based systems, including active targeting, easiersynthetic access, and deep tumor penetration [21,24].

In this study, we have developed a carboxylesterase responsive bi-functional drug delivery system, EDOX, for delivery of an anticancerdrug, doxorubicin (Dox), and tested its effects on various cancer celllines. Our preliminary results showed that EDOX was preferentiallyactivated by the intracellular carboxylesterase of HCC-derived cells, incomparison with normal fibroblasts and other cancer cell lines. Fig. 1summarizes the overall chemical design strategy.

2. Materials and procedures

2.1. General information and materials

All chemicals and solvents used for synthetic purposes were pur-chased from Sigma-Aldrich, Alfa, or TCI-Korea. Moisture sensitive re-actions were generally carried out under a blanket of argon gas. NMRspectral analyses were performed on Bruker NMR (500MHz for 1H,125MHz for 13C) instruments at room temperature using CDCl3, MeOD,and DMSO‑d6 as the solvents with tetramethylsilane (TMS) as the in-ternal standard. Chemical shifts (δ) are recorded in parts per million(ppm) and coupling constants are given in Hz. High-resolution massspectra were recorded on an Ion Spec Hi-Res ESI mass spectrometer.HPLC analyses were performed on a YL9101S (YL-Clarity) instrumentequipped with a reverse-phase (RP) column (C18, 5mm, Waters).Eluent used in the analysis was a water-acetonitrile (ACN) gradient(acetonitrile from 5% to 85%; 0–30min) with a flow rate of 1.0 mL/minand UV–Vis detector (475 nm).

2.2. Preliminary solution and interference studies

A stock solution of EDOX (1 mM) was prepared in the DMSO solu-tion. The working solution (10uM) of EDOX was prepared by diluting inphosphate saline buffered (PBS, pH 7.4, 37 °C). The carboxylesterase(Sigma Aldrich, E2884-1KU) was used in this study for preliminarysolution tests. For interference studies, all the stock solution of bio-analytes were prepared in PBS and used in concentrations as mentioned

in studies.

2.3. Cell culture, fluorescence imaging, and flow cytometry analysis

The HepG2 and HT-29 were maintained in RPMI1640 medium(Gibco, Invitrogen. Co., Carlsbad, CA, USA) while HeLa, MCF7, andNIH3T3 cells were cultured with the Dulbecco's Modified Eagle Medium(DMEM), Gibco, supplemented with fetal bovine serum (10% FBS,Gibco) and penicillin/streptomycin (1%, Gibco) in an incubator (5%CO2, 37 °C). To screen for the most proper cancer cell line response toEDOX among various cells, we observed the fluorescence in EDOX-treated cells using confocal laser scanning microscopy (CLSM) and theflow cytometry. All cells were seeded at 1×105 cells/mL suspension inμ-slide, 8 well (ibidi, Munich) and incubated for 12 h at 37 °C. Next,5 μM of EDOX was added to cells for 2 h, followed by washing in PBS.After fixing with 4% paraformaldehyde for 30min at room tempera-ture, the cells were stained by fluorophore Alexa Fluor 488 phalloidin(Invitrogen, Molecular Probe, Eugene, OR, USA) for 20min and 4′ 6-diamidino-2-phenylindole (DAPI) for 5min. The fluorescent cell imageswere obtained by a CLSM (LSM 710, Carl Zeiss, Germany). In addition,the cellular fluorescence was analyzed by flow cytometry (FACS, BDBiosciences, USA) for quantitative evaluation of the fluorescence inEDOX-treated cells, all cells were incubated in 12-well plate at a densityof 4× 105/well and treated with 5 μM EDOX for 2 h. After washingwith PBS 3 times, the harvested cells were examined by the flow cy-tometry. The histogram plots were performed using the FlowJo soft-ware (TreStar, Switxìzerland).

2.4. Anti-proliferation assay and western blotting

Anti-cancer effects of EDOXwere evaluated in HepG2, HT-29, HeLa,MCF-7 and NIH3T3 cells by using MTT assays and western blot analysis.All cells were seeded into each well of 96 well plates at 1×104 cells perwell, and then, they were treated with various concentrations of EDOXfor 48 h, followed by performing a 3-(4,5-dimethylthiazol-2-yl)-2,5-di-phenyl tetrazolium bromide (MTT, Roche Diagnostics GmbH, Germany)colorimetric analysis. After replacing the culture media, MTT reagentwas added into each well and incubated for an additional 4 h. Followingthe treatment of solubilization buffer (Roche Diagnostics) to each well,the absorbance of formazan product produced by MTT was measured at570 nm using a microplate reader (EL800, Bio-Tek Instruments,Winooski, USA). For western blot analysis, HepG2 cells were cultured ina 6 well plate and treated with 5 μM of EDOX for 12 h. Then, theharvested cells were lysed with cell lysis solution (GenDEPOT, TX, USA)including protease inhibitor cocktail solution (Xpert Protease InhibitorCocktail Solution, GenDEPOT). Total protein was quantified by usingthe BCA protein assay, and the equal amount (50 μg) of protein wasseparated by 10% (w/v) SDS-polyacrylamide gel electrophoresis. Afterthe proteins were transferred to polyvinylidene fluoride (PVDF) mem-brane, they were blocked by 5% skimmed milk for 1 h. The membraneswere incubated with the following antibodies: anti-PARP-1 (1:1000;abcam, UK), anti-Caspase-3 (1:1000; Cell Signaling Technology, USA),and β-actin (Santa Cruz, USA). Then, The HRP-conjugated secondaryantibodies were incubated for 1 h at room temperature. After washing,the membrane was incubated with ECL reagent (AmershamBiosciences, UK) as the substrate, and then expose the X-ray film in thedark room.

3. Results and discussion

3.1. Design, synthesis, and characterization of EDOX

To prevent any undesired chemical or enzymatic degradation of theprodrug conjugate, intermediate 1 (Scheme S1) was chosen for EDOXsynthesis. On the basis of the difference in electron donor ability be-tween amides and carbamates (weak donors, Hammett constant,Fig. 1. Schematic representation of the EDOX conjugate.

A. Sharma et al. Dyes and Pigments 163 (2019) 628–633

629

Page 3: Dyes and Pigments - bntl.sogang.ac.krbntl.sogang.ac.kr/bntl/Research/Documents/Dyes and... · Biosciences, UK) as the substrate, and then expose the X-ray film in the dark room

σp=0.00) and amines (strong donors, σp=−0.66), we predicted ahigher stability and a more efficient drug release profile of this con-jugate upon enzymatic hydrolysis [31]. Moreover, esterase-mediatedhydrolysis of aromatic amides is well-documented in the literature[32,33]. Additionally, the presence of a covalent bond between the Doxamino group and the chemical linker may attenuate the drug action,until unmasking takes place [24,25,34,35]. The synthetic route forEDOX is shown in Scheme S1. First, we evaluated drug stability andactivation mechanism. Upon incubation of EDOX in phosphate salinebuffered (PBS, pH 7.4, 37 °C) for 4 h, no degradation was observed.EDOX showed an intrinsic weak fluorescence emission at 560 nm(Fig. 2A). Likewise, EDOX exhibited similar kind of stability in culturemedia up to 12 h (Fig. S1). However, exposure to esterase (3 U/mL) ledto cleavage of the aniline amide bond, followed by self-immolation via1,6-elimination and release of the active drug, Dox. This phenomenonwas accompanied by a concomitant enhancement of the emission bandat 560 nm, corresponding to free Dox. EDOX activation was furtherconfirmed by combined fluorescence experiments and HPLC analysis.Exposure of EDOX to esterase resulted in a time-dependent cumulativerelease of active Dox up to 4 h (Fig. 2B). We reasoned that fluorescencerestoration at 560 nm could be a turn-on response potentially suitablefor the diagnostic assessment of drug distribution. Under the detectionconditions specified in the supplementary information, the reverse-phase HPLC chromatograms showed a peak at retention time of 2060 scorresponding to EDOX (Fig. 2C). Notably, the Dox standard peaks at1975 s, indicating that EDOX is more hydrophobic than Dox itself. Thisproperty may result particularly convenient in the design of self-as-sembled nanomedicine systems based on PEGylation, for the optimi-zation of drug pharmacokinetics (PK) [36]. Upon carboxylesterase ac-tion, EDOX was exclusively converted to active Dox within 4 h by aproposed route shown in Fig. 2D. Possible interference with EDOX re-sponse of various bioanalytes including amino acids, biomolecules, andenzymes was also tested. None of these molecules induced significantEDOX fluorescence change, that was solely activated by esterase, in-dicating that the esterase-EDOX interaction was highly specific. (Fig.S2).

3.2. Cellular uptake and activation of EDOX

Inspired by these findings, we next evaluated EDOX activity withvarious cell lines, including cells derived from human hepatocyte car-cinoma (HepG2), human colorectal adenocarcinoma (HT-29), humancervix adenocarcinoma (HeLa), human breast adenocarcinoma (MCF7),and mouse embryo fibroblasts (NIH3T3). To assess EDOX activation bycellular carboxylesterase, confocal laser scanning microscopy (CLSM)was employed for the detection of intracellular fluorescence arisingfrom Dox. As shown in Fig. 3, only HCC-derived HepG2 cells showed asignificant fluorescence enhancement, corresponding to active Dox,upon treatment with 5 μM EDOX for 2 h. These results were also con-firmed by flow cytometry analysis (Fig. S3) and indicated that EDOXinternalization and/or activation did not occur (or occurred at negli-gible rates) in HT-29, HeLa, MCF7 and NIH3T3 cells as its quenchedfluorescence emission remain intact. Consistently, previous studieshave reported that human carboxylesterase is mainly expressed in theliver and is highly detectable in HepG2 cell lysates by western blottinganalysis [37–39]. Moreover, carboxylesterase activity has been de-tected in HepG2 cells by highly specific fluorescent reporter assays[19]. To evaluate the dynamics of the EDOX-cell interaction, we mea-sured the time-dependent fluorescence enhancement in HepG2 cellsexposed to the conjugate (Fig. S4). A significant enhancement influorescent emission was observed within 30min and reached satura-tion in 24 h. Furthermore, we observed that, upon EDOX enzymaticcleavage, active Dox was initially localized in the cytosol and succes-sively translocated to the nucleus over an incubation time of 36 h (Fig.S4). Collectively, these findings suggested that EDOX hydrolysis re-quires the presence of elevated carboxylesterase activity and, therefore,the complex is preferentially activated by the HCC-derived HepG2 cells,resulting in fluorescence enhancement.

3.3. Cell viability of EDOX towards various cancer cell lines and apoptosismechanism

Next, we evaluated the anticancer activity of EDOX in different celllines by using a tetrazolium dye, 3-(4,5-dimethylthiazol-2-yl)-2,5-

Figure 2. (A) Fluorescence intensity response of EDOX (10 μM) upon incubation with esterase (3 U/mL) in phosphate buffered solution (PBS) at 37 °C (Excitationwavelength= 470 nm). (B) Time-dependent fluorescence enhancement was observed upon incubation of EDOX with esterase (3 U/mL) in PBS at 37 °C. The changein fluorescence intensity is directly related to the release of active Dox from EDOX. (C) Reverse phase high-performance liquid chromatography (RP-HPLC) curves ofEDOX in the presence of esterase at 37 °C for 0 h, 1 h, and 4 h, and Dox standard. (D) Proposed mode of EDOX activation upon exposure to esterase.

A. Sharma et al. Dyes and Pigments 163 (2019) 628–633

630

Page 4: Dyes and Pigments - bntl.sogang.ac.krbntl.sogang.ac.kr/bntl/Research/Documents/Dyes and... · Biosciences, UK) as the substrate, and then expose the X-ray film in the dark room

diphenyltetrazolium bromide (MTT) assay. Cell viability was measuredafter a 48 h incubation with different concentrations of EDOX (Fig. 4A).EDOX exerted a HepG2 cell-specific anticancer activity, even at con-centrations below 10 μM, and was remarkably effective up to 50 μM,whereas no significant impact on the viability of the other cell lines wasobserved in this range of concentrations. The results of the MTT assaywere in agreement with those obtained by confocal microscopy.

Although EDOX-induced cytotoxicity was lower than that exerted byfree Dox under these conditions, its cell specificity could help minimizeDox-related side effects (Fig. S5). Moreover, the comparison of 50%growth inhibitory concentration (IC50) values exhibited that EDOX-treated HepG 2 cells presented more potent therapeutic effects thanother cell lines (Table S1). To further characterize EDOX effects, theexpression of apoptosis-related proteins was evaluated by western blot

Fig. 3. Confocal laser scanning microscopy (CLSM) images of intracellular fluorescence of EDOX in various cell lines. HepG2, HT-29, HeLa, MCF7, and NIH3T3 cellswere incubated with EDOX (5 μM) for 2 h, fixed with paraformaldehyde (4%), and counterstained with the DAPI (blue) and Alexa Fluor 488-Phalloidin (green) tovisualize nucleus and F-actin, respectively. CLSM images of EDOX (red) were obtained by using an excitation wavelength of 488 nm and an emission wavelength560–590 nm. Scale bars: 20 μm. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

Fig. 4. Anticancer effects of EDOX in various cell lines. (A)HepG2, HT-29, HeLa, MCF7, and NIH3T3 cells were treatedwith EDOX at various concentrations for 48 h. Then, cellviability was determined by MTT assay. (B) Western blotanalysis in Dox- and EDOX-treated HepG2 cells. Cells weretreated with Dox or EDOX (5 μM) for 12 h. Apoptosis wasassessed by western blot using anti-PARP-1 and anti-Caspase-3 monoclonal antibodies, and a β-actin antibody asa loading control.

A. Sharma et al. Dyes and Pigments 163 (2019) 628–633

631

Page 5: Dyes and Pigments - bntl.sogang.ac.krbntl.sogang.ac.kr/bntl/Research/Documents/Dyes and... · Biosciences, UK) as the substrate, and then expose the X-ray film in the dark room

in EDOX-treated HepG2 cells. PARP-1 (poly (ADP-ribose) polymerase-1) plays an important role in the recovery of DNA-damaged cells. Thelevel of cleaved PARP-1 is one of the most important markers ofapoptosis and it was reported to be increased after Dox treatment[40–42]. Similarly, the expression level of activated caspase-3, a mi-tochondrial apoptosis-associated protein, is known to be upregulated inDox-treated cells [43]. We found that the expression of both PARP-1and cleaved caspase-3 was strongly enhanced in HepG2 cells upontreatment with Dox or 5 μM EDOX (Fig. 4B). The different ability of Doxand EDOX to induce the expression of apoptotic proteins in HepG2 cellsat 12 h might be due to the additional time required for completion ofDox release upon cleavage by carboxylesterase. Overall, our findingsshowed that EDOX was specifically activated by HepG2 carbox-ylesterase and this led to cell death by the induction of apoptosis.

3.4. Esterase mediated activation of EDOX in HCC cells

Finally, to clearly establish the relationship between EDOX activa-tion and endogenous carboxylesterase, we verified whether EDOXfluorescence enhancement could be prevented by the inhibition ofcarboxylesterase activity [44]. Therefore, HepG2 cells were pretreatedfor 12 h with bis-(4-nitrophenyl)phosphate (BNPP), a carboxylesteraseinhibitor, followed by treatment with 10 μM EDOX for an additional2 h. As shown in Fig. 5, EDOX response was nearly abolished in BNPP-pretreated HepG2 cells, further supporting the key role of carbox-ylesterase in EDOX activation.

4. Conclusion

In conclusion, using the acetamide-chemodrug conjugate, EDOX,we have developed a bifunctional system allowing, on the one hand, forsustained, HCC-specific drug release and, on the other, for real-timemonitoring of prodrug activation and drug distribution. EDOX testingin various cancer cell lines revealed a preferential cytotoxic effect onHCC-derived cells. Thus, we propose EDOX, a stimuli-responsive pro-drug conjugate, as a suitable chemotherapeutic option for HCC.

Acknowledgment

This work was supported by the National Research Foundation ofKorea (CRI 2018R1A3B1052702, J.S.K; 2017R1D1A1B04033453,E.J.K). This research was also supported by BioNano Health-GuardResearch Center funded by the Ministry of Science and ICT(MSIT) ofKorea as Global Frontier Project (Grant number H-

GUARD_2013M3A6B2078950 (2014M3A6B2060302)).

Appendix A. Supplementary data

Supplementary data to this article can be found online at https://doi.org/10.1016/j.dyepig.2018.12.026.

References

[1] Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell2011;144(5):646–74.

[2] Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM.Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid,liver, and pancreas cancers in the United States. Cancer Res 2014;74(11):2913–21.

[3] Maman S, Witz IP. A history of exploring cancer in context. Nat Rev Canc2018;18(6):359–76.

[4] Qiu M, Wang D, Liang WY, Liu LP, Zhang Y, Chen X, Sang DK, Xing CY, Li ZJ, DongBQ, Xing F, Fan DY, Bao SY, Zhang H, Cao YH. Novel concept of the smart NIR-light-controlled drug release of black phosphorus nanostructure for cancer therapy. PNatl Acad Sci USA 2018;115(3):501–6.

[5] Xie ZJ, Wang D, Fan TJ, Xing CY, Li ZJ, Tao W, Liu LP, Bao SY, Fan DY, Zhang H.Black phosphorus analogue tin sulfide nanosheets: synthesis and application asnear-infrared photothermal agents and drug delivery platforms for cancer therapy. JMater Chem B 2018;6(29):4747–55.

[6] Sun ZB, Xie HH, Tang SY, Yu XF, Guo ZN, Shao JD, Zhang H, Huang H, Wang HY,Chu PK. Ultrasmall black phosphorus quantum dots: synthesis and use as photo-thermal agents. Angew Chem Int Ed 2015;54(39):11526–30.

[7] Qiu M, Ren WX, Jeong T, Won M, Park GY, Sang DK, Liu LP, Zhang H, Kim JS.Omnipotent phosphorene: a next-generation, two-dimensional nanoplatform formultidisciplinary biomedical applications. Chem Soc Rev 2018;47(15):5588–601.

[8] Swift LH, Golsteyn RM. Genotoxic anti-cancer agents and their relationship to DNAdamage, mitosis, and checkpoint Adaptation in proliferating cancer cells. Int J MolSci 2014;15(3):3403–31.

[9] Widakowich C, De Castro G, De Azambuja E, Dinh P, Awada A. Review: side effectsof approved molecular targeted therapies in solid cancers. Oncologist2007;12(12):1443–55.

[10] Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancerstatistics, 2012. CA-Canc J Clin 2015;65(2):87–108.

[11] Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, Gores G.Hepatocellular carcinoma. Nat Rev Dis Primers 2016;2:16018.

[12] Bosslet K, Straub R, Blumrich M, Czech J, Gerken M, Sperker B, Kroemer HK,Gesson JP, Koch M, Monneret C. Elucidation of the mechanism enabling tumorselective prodrug monotherapy. Cancer Res 1998;58(6):1195–201.

[13] Han KH, Park JY. Chemotherapy for advanced hepatocellular carcinoma. JGastroen Hepatol 2008;23(5):682–4.

[14] Yang J, Shi D, Yang DF, Song XL, Yan BF. Interleukin-6 alters the cellular respon-siveness to clopidogrel, irinotecan, and Oseltamivir by suppressing the expression ofCarboxylesterases HCE1 and HCE2. Mol Pharmacol 2007;72(3):686–94.

[15] Wang J, Williams ET, Bourgea J, Wong YN, Patten CJ. Characterization of re-combinant human carboxylesterases: fluorescein diacetate as a probe substrate forhuman carboxylesterase 2. Drug Metab Dispos 2011;39(8):1329–33.

[16] Na K, Lee EY, Lee HJ, Kim KY, Lee H, Jeong SK, Jeong AS, Cho SY, Kim SA, Song SY,Kim KS, Cho SW, Kim H, Paik YK. Human plasma carboxylesterase 1, a novel ser-ologic biomarker candidate for hepatocellular carcinoma. Proteomics2009;9(16):3989–99.

Fig. 5. Inhibition of cellular EDOX fluorescence by BNPP. After treatment with BNPP (100 μM) for 12 h, HepG2 cells were treated with 10 μM EDOX for 2 h andexamined with CSLM. Scale bars: 20 μm.

A. Sharma et al. Dyes and Pigments 163 (2019) 628–633

632

Page 6: Dyes and Pigments - bntl.sogang.ac.krbntl.sogang.ac.kr/bntl/Research/Documents/Dyes and... · Biosciences, UK) as the substrate, and then expose the X-ray film in the dark room

[17] Quiroga AD, Li L, Trotzmuller M, Nelson R, Proctor SD, Kofeler H, Lehner R.Deficiency of carboxylesterase 1/esterase-x results in obesity, hepatic steatosis, andhyperlipidemia. Hepatology 2012;56(6):2188–98.

[18] Feng Z, Wang H, Zhou R, Li J, Xu B. Enzyme-instructed assembly and disassemblyprocesses for targeting downregulation in cancer cells. J Am Chem Soc2017;139:3950–3.

[19] Liu ZM, Feng L, Ge GB, Lv X, Hou J, Cao YF, Cui JN, Yang L. A highly selectiveratiometric fluorescent probe for in vitro monitoring and cellular imaging of humancarboxylesterase 1. Biosens Bioelectron 2014;57:30–5.

[20] Burkhart DJ, Barthel BL, Post GC, Kalet BT, Nafie JW, Shoemaker RK, Koch TH.Design, synthesis, and preliminary evaluation of doxazolidine carbamates as pro-drugs activated by carboxylesterases. J Med Chem 2006;49:7002–12.

[21] Lee MH, Sharma A, Chang MJ, Lee J, Son S, Sessler JL, Kang C, Kim JS. Fluorogenicreaction-based prodrug conjugates as targeted cancer theranostics. Chem Soc Rev2018;47(1):28–52.

[22] Fan WP, Yung B, Huang P, Chen XY. Nanotechnology for multimodal synergisticcancer therapy. Chem Rev 2017;117(22):13566–638.

[23] Jung HS, Verwilst P, Sharma A, Shin J, Sessler JL, Kim JS. Organic molecule-basedphotothermal agents: an expanding photothermal therapy universe. Chem Soc Rev2018;47(7):2280–97.

[24] Chen GY, Roy I, Yang CH, Prasad PN. Nanochemistry and nanomedicine for na-noparticle-based diagnostics and therapy. Chem Rev 2016;116(5):2826–85.

[25] Sharma A, Kim EJ, Shi H, Lee JY, Chung BG, Kim JS. Development of a theranosticprodrug for colon cancer therapy by combining ligand-targeted delivery and en-zyme-stimulated activation. Biomaterials 2018;155:145–51.

[26] Sharma A, lee M-G, Shi H, Won M, Arambula JF, Sessler JL, Lee JY, Chi S-G, Kim JS.Overcoming drug resistance by targeting cancer bioenergetics with an activatableprodrug. Chem 2018;4:2370–83.

[27] Zhou Y, Maiti M, Sharma A, Won M, Yu L, Miao LX, Shin J, Podder A, Bobba KN,Han J, Bhuniya S, Kim JS. Azo-based small molecular hypoxia responsive ther-anostic for tumor-specific imaging and therapy. J Contr Release 2018;288:14–22.

[28] Kim HS, Sharma A, Ren WX, Han J, Kim JS. COX-2 Inhibition mediated anti-an-giogenic activatable prodrug potentiates cancer therapy in preclinical models.Biomaterials 2018;185:63–72.

[29] Shi JJ, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: progress,challenges and opportunities. Nat Rev Canc 2017;17(1):20–37.

[30] Anchordoquy TJ, Barenholz Y, Boraschi D, Chorny M, Decuzzi P, DobrovolskaiaMA, Farhangrazi ZS, Farrell D, Gabizon A, Ghandehari H, Godin B, La-Beck NM,Ljubimova J, Moghimi SM, Pagliaro L, Park JH, Peer D, Ruoslahti E, Serkova NJ,Simberg D. Mechanisms and barriers in cancer nanomedicine: addressing chal-lenges, looking for solutions. ACS Nano 2017;11(1):12–8.

[31] Hansch C, Leo A, Taft RW. A survey of Hammett substituent constants and re-sonance and field parameters. Chem Rev 1991;91(2):165–95.

[32] Yoshioka K, Komatsu T, Nakada A, Onagi J, Kuriki Y, Kawaguchi M, Terai T, UenoT, Hanaoka K, Nagano T, Urano Y. Identification of tissue-restricted bioreactionsuitable for in vivo targeting by fluorescent substrate library-based enzyme dis-covery. J Am Chem Soc 2015;137(38):12187–90.

[33] Jin Q, Feng L, Wang DD, Dai ZR, Wang P, Zou LW, Liu ZH, Wang JY, Yu Y, Ge GB,Cui JN, Yang L. A two-photon ratiometric fluorescent probe for imaging carbox-ylesterase 2 in living cells and tissues. ACS Appl Mater Interfaces2015;7(51):28474–81.

[34] Garsky VM, Lumma PK, Feng DM, Wai J, Ramjit HG, Sardana MK, Oliff A, Jones RE,DeFeo-Jones D, Freidinger RM. The synthesis of a prodrug of doxorubicin designedto provide reduced systemic toxicity and greater target efficacy. J Med Chem2001;44(24):4216–24.

[35] Jang JH, Lee H, Sharma A, Lee SM, Lee TH, Kang C, Kim JS. Indomethacin-guidedcancer selective prodrug conjugate activated by histone deacetylase and tumour-associated protease. Chem Commun 2016;52(64):9965–8.

[36] Farokhzad OC, Cheng JJ, Teply BA, Sherifi I, Jon S, Kantoff PW, Richie JP, LangerR. Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo.Proc Natl Acad Sci USA 2006;103(16):6315–20.

[37] Sanghani SP, Sanghani PC, Schiel MA, Bosron WF. Human carboxylesterases: anupdate on CES1, CES2 and CES3. Protein Pept Lett 2009;16(10):1207–14.

[38] Pratt SE, Durland-Busbice S, Shepard RL, Heinz-Taheny K, Iversen PW, Dantzig AH.Human carboxylesterase-2 hydrolyzes the prodrug of gemcitabine (LY2334737) andconfers prodrug sensitivity to cancer cells. Clin Canc Res 2013;19(5):1159–68.

[39] Ye F, Xin ZS, Han W, Fan JJ, Yin B, Wu SZ, Yang W, Yuan JG, Qiang BQ, Sun W,Peng XZ. Quantitative proteomics analysis of the hepatitis C virus replicon high-permissive and low-permissive cell lines. PLoS One 2015;10(11):e0142082.

[40] Park HJ, Bae JS, Kim KM, Moon YJ, Park SH, Ha SH, Hussein UK, Zhang Z, Park HS,Park BH, Moon WS, Kim JR, Jang KY. The PARP inhibitor olaparib potentiates theeffect of the DNA damaging agent doxorubicin in osteosarcoma. J Exp Clin Canc Res2018;37:107.

[41] Wu IJ, Lin RJ, Wang HC, Yuan TM, Chuang SM. TRIB3 downregulation enhancesdoxorubicin-induced cytotoxicity in gastric cancer cells. Arch Biochem Biophys2017;622:26–35.

[42] Choi JR, Shin KS, Choi CY, Kang SJ. PARP1 regulates the protein stability andproapoptotic function of HIPK2. Cell Death Dis 2016;7:e2438.

[43] Wei L, Surma M, Gough G, Shi S, Lambert-Cheatham N, Chang J, Shi JJ. Dissectingthe mechanisms of doxorubicin and oxidative stress-induced cytotoxicity: the in-volvement of actin cytoskeleton and ROCK1. PLoS One 2015;10(7):e0131763.

[44] Eng H, Niosi M, McDonald TS, Wolford A, Chen Y, Simila STM, Bauman JN,Warmus J, Kalgutkar AS. Utility of the carboxylesterase inhibitor bis-para-ni-trophenylphosphate (BNPP) in the plasma unbound fraction determination for ahydrolytically unstable amide derivative and agonist of the TGR5 receptor.Xenobiotica 2010;40(6):369–80.

A. Sharma et al. Dyes and Pigments 163 (2019) 628–633

633