Paclitaxel enhances tumoricidal potential of TRAIL via inhibition of MAPK in resistant gastric cancer cells

  • Authors:
    • Lin Li
    • Xian-Zi Wen
    • Zhao-De Bu
    • Xiao-Jing Cheng
    • Xiao-Fang Xing
    • Xiao-Hong Wang
    • Lian-Hai Zhang
    • Ting Guo
    • Hong Du
    • Ying Hu
    • Biao Fan
    • Jia-Fu Ji
  • View Affiliations

  • Published online on: March 9, 2016     https://doi.org/10.3892/or.2016.4666
  • Pages: 3009-3017
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) holds promise for cancer therapy due to its unique capacity to selectively trigger apoptosis in cancer cells. However, TRAIL therapy is greatly hampered by its resistance. A preclinical successful strategy is to identify combination treatments that sensitize resistant cancers to TRAIL. In the present study, we fully assessed TRAIL sensitivity in 9 gastric cancer cell lines. We found combined administration of paclitaxel (PTX) markedly enhanced TRAIL-induced apoptosis in resistant cancer cells both in vitro and in vivo. The sensitization to TRAIL was accompanied by activation of mitochondrial apoptotic pathway, upregulation of TRAIL receptors and downregulation of anti-apoptotic proteins including C-IAP1, C-IAP2, Livin and Mcl-1. Noticeably, we found PTX could suppress the activation of mitogen-activated protein kinases (MAPKs). Inhibition of MAPKs using specific inhibitors (ERK inhibitor U0126, JNK inhibitor SP600125 and P38 inhibitor SB202190) facilitated TRAIL-mediated apoptosis and cytotoxicity. Additionally, SP600125 upregulated TRAL receptors as well as downregulated C-IAP2 and Mcl-1 suggesting the anti-apoptotic role of JNK. Thus, PTX-induced suppression of MAPKs may contribute to restoring TRAIL senstitivity. Collectively, our comprehensive analyses gave new insight into the role of PTX on enhancing TRAIL sensitivity, and provided theoretical references on the development of combination treatment in TRAIL-resistant gastric cancer.

Introduction

Gastric cancer (GC) is the fourth most frequent malignancy and the second leading cause of cancer mortality worldwide (1). In China, most of the patients are identified at the advanced stage which leads to dismal prognosis. Radical surgery is the only curative therapy and chemotherapy serves as common strategy for late stage patients. Patients with GC, however, are not particularly sensitive to conventional chemotherapeutic drugs. Thus, it is of great interest to find new approaches, and targeted manipulation of apoptosis is a research hot spot (2).

TRAIL, a member of tumor necrosis factor (TNF) super family, selectively triggers cell death in transformed cells while causing virtually no toxic towards normal cells (3). TRAIL initiates apoptotic signals via binding to two cell surface death receptors (DRs), DR4 (also known as TRAIL-R1) and DR5 (also known as TRAIL-R2), leading to receptor aggregation and recruitment of Fas-associated death domain (FADD) followed by procaspase-8 (4). The apoptotic process then follows two signaling pathways. In type I cells, activated caspase-8 enables the autocatalytic cleavage of caspase cascade triggers apoptosis. While in type II cells, caspase-8 additionally triggers mitochondrial apoptosis pathway by activating cleavage of Bid, which induces the release of cytochrome c from mitochondria and in turn activates caspase-9 to finally execute cell death (5). Drugs targeting TRAIL pathway, including recombinant TRAIL and agonistic antibodies have been demonstrated with robust anticancer activity in a number of preclinical studies (68).

Despite the attractive tumoricidal potential, TRAIL-based therapy is greatly hampered by its resistance, a major obstacle to clinical application (810). The most basic cause is dysfunction of death receptors due to hyper-methylation (11), mutation (12) and loss of cell surface expression (13). The defects in caspase protein (14) and overexpression of pro-survival proteins, such as IAP family and anti-apoptotic Bcl-2 family members (1518), are also linked to TRAIL sensitivity. In addition, more recent findings suggested that cell survival signals, including mitogen-activated protein kinase (MAPK) pathway, phosphatidylinositol 3-kinase/Akt pathway, and transcription factor NF-κB played pivotal roles in regulation of TRAIL signaling (1922). Previous studies demonstrated that TRAIL resistance could be alleviated or even reversed by combination therapy (23,24). PTX, one of the cytoskeletal drugs targeting tubulin, is commonly used in advanced GC treatment. It inhibits tumor cell proliferation through stabilizing microtubule network and inhibiting microtubule dynamics (25). PTX has been explored as sensitizing agent towards TRAIL in some cancer types (2628), yet, the molecular mechanisms were not fully elucidated.

In the present study, we demonstrate that combined treatment of PTX and TRAIL significantly boosted apoptosis of TRAIL-resistant GC cells both in vitro and in vivo. The molecular mechanisms underlying the synergism involved enhanced activation of caspases, upregulation of DRs and downregulation of anti-apoptotic proteins. Moreover, we present herein the inhibitory effect of MAPKs in TRAIL-induced apoptosis in GC and reveal for the first time that TRAIL-augmenting effect of PTX was partly due to suppression of the MAPK pathway.

Materials and methods

Cell culture and reagents

GC cell lines AGS, NCI-N87, SNU-1 and SNU-16 were purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA). MKN28 and NUGC3 were purchased from Health Science Research Resources Bank (Tokyo, Japan), and SGC-7901, BGC-823 and MGC-803 were obtained from Cell Research Institute (Shanghai, China). All cells were maintained in Dulbecco's modified Eagle's medium (DMEM; Gibco-BRL, Carlsbad, CA, USA) containing 10% fetal bovine serum (FBS) and penicillin/streptomycin at 37°C with 5% CO2. Recombinant human TRAIL was purchased from PeproTech EC Ltd. (London, UK). The small molecule inhibitors U0126 (#S1102), SP600125 (#S1460) and SB202190 (#S1077) were obtained from Selleck Chemicals (Houston, TX, USA). The chemotherapeutic drug PTX was from Beijing Union Pharmaceutical Factory (Beijing, China).

Cell viability assay

Cell viability was assessed using Cell Counting kit-8 assay (CCK-8) (Dojindo Molecular Technologies, Kumamoto, Japan). Briefly, cells were seeded onto 96-well plates at a density of 3–5×103 cells/well and allowed to attach for 24 h. The cells were incubated with indicated concentrations of TRAIL and/or PTX for 24 h. After incubation, CCK-8 solution (10 µl) was added to each well and absorbance was measured at 450 nm using multiscan spectrum.

Flow cytometry for apoptosis analysis and expression of cell surface DRs

For cell apoptosis assay, cells (3×105) were seeded onto 35-mm2 culture dishes and treated with TRAIL and/or PTX for 24 h as indicated in the figure legends. Then, the cells were stained using an Annexin V/PI double staining kit (Dojindo Molecular Technologies) according to the manufacturer's protocol.

For cell surface expression analysis, cells (1×106) treated with PTX or vehicle for 24 h were incubated with allophycocyanin (APC)-conjugated anti-DR4 and anti-DR5 antibody, or isotype control (BioLegend, Inc., San Diego, CA, USA) for 30 min at 4°C, and analyzed by flow cytometery (BD Accuri C6; BD Biosciences, San Jose, CA, USA). Positive cells were identified and measured by subtracting relative isotype control values.

Western blot assay

Total proteins were extracted using RIPA lysis buffer (Beijing Solarbio Science and Technology Co., Ltd., Beijing, China) with protease inhibitors and phosphatase inhibitors (Beijing Solarbio Science and Technology). Protein concentration was quantified using the Bradford method (Pierce, Rockford, IL, USA). Equivalent amount of protein (20 µg) was separated by 10% SDS-PAGE gels and transferred onto PVDF membranes (Millipore, Billerica, MA, USA). The membranes were blocked in 5% non-fat milk in TBS-T for 1 h and incubated with primary antibodies overnight at 4°C, followed by incubation with HRP-conjugated secondary antibodies. Signals were detected using chemiluminescent agents (Pierce). Primary antibodies of DR4 and GAPDH were from Abcam (Cambridge, MA, USA), and DR5 was from Sigma-Aldrich (St. Louis, MO, USA). Other primary antibodies purchased from Cell Signaling Technology (Danvers, MA, USA) were as follows: caspase-3 (#9662), cleaved-caspase-3 (#9664), caspase-7 (#9492), cleaved-caspase-7 (#8438), caspase-8 (#9746), caspase-9 (#9502), cleaved-caspase-9 (#7237), PARP (#9542), cleaved-PARP (#5625), Bid (#2002), C-IAP1 (#7065), C-IAP2 (#3130), XIAP (#2045), Livin (#5471), Bcl-xL (#2764), Bcl-2 (#2870), Mcl-1 (#5453), ERK (#4695), phosphorylated-ERK (#4370), JNK (#9258), phosphorylated-JNK (#4668), p38 (#8690), phosphorylated-p38 (#4511).

siRNA transfection

C-IAP1 siRNA and scrambled siRNA were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Cells (1×105) in 6-well plates were incubated for 24 h and then transfected with siRNA (100 nmol/l) and Lipofectamine 2000 reagent (Invitrogen, Carlsbad, CA, USA) in Opti-MEM without serum, according to the manufacturer's specifications. After 6 h, the medium was replaced with fresh medium. Forty-eight hours after transfection, C-IAP1 knockdown effect was verified by western blotting.

In vivo xenograft study

Animal studies were carried out in strict adherence with institutional guidelines. SGC-7901 cells (2×106/200 µl per mouse) were subcutaneously injected into the right hind legs of 6–8 week-old female nude mice. When tumors volume reached 50–100 mm3, the mice were randomized to 4 groups and dosing was initiated. They were: i) control (vehicle only); ii) TRAIL (100 µg/kg intratumoral injection); iii) PTX (10 mg/kg i.p.); and iv) the sequential administration of PTX (10 mg/kg i.p.) followed by TRAIL (100 µg/kg intratumoral injection) after 24 h. All groups were treated once every 3 days for 18 days. The tumor size and weight were monitored three times a week. Tumor volume (V) was calculated as V = 0.5 × length × width2. Tumor growth inhibition (TGI) was assessed in accordance with the formula [1− (T−T0)/(C−C0)] × 100, where T and T0 were the mean tumor volumes at the end of the drug administration and day 0, respectively for treated group, and C−C0 were those for vehicle control group.

Statistical analysis

All the values are presented as mean ± SD of three independent experiments. The statistical significance among experimental groups was assessed by two-sided Student's t-test. P<0.05 were considered to be significant. All statistical analyses were performed using GraphPad Prism 5.0 software.

Results

Response of GC cells to TRAIL and expression of TRAIL receptors

First, we analyzed the effects of TRAIL on cell viability in 9 GC cells using CCK-8 assay. As shown in Fig. 1A, GC cells had varying degree of sensitivity to TRAIL. SNU-16, NUGC3, NCI-N87 and SNU-1 were defined as sensitive cells, in which >50% growth inhibition was achieved when treated with TRAIL (200 ng/ml) for 24 h. By contrast, the growth inhibition rates of TRAIL-resistant cells (BGC-823, MKN28, MGC-803, SGC-7901 and AGS) were <50% even under condition of elevated concentration or prolonged exposure time indicating an intrinsic resistance mechanisms in these cells. TRAIL-induced apoptosis was also confirmed by Annexin V/PI double staining in the same cell lines (Fig. 1B). To determine whether TRAIL pathway was intact, we measured cell surface expression of DR4 and DR5 (Fig. 1C and D). Flow cytometric analysis revealed a predominant expression of DR5 in all these cells except SNU-16. DR4, however, exhibited diverse levels in 9 cell lines and the top three sensitive cells (SNU-16, NUGC3 and NCI-N87) expressed the highest levels.

PTX and TRAIL act synergistically to inhibit GC cell growth

PTX exhibited growth inhibiting effect in TRAIL-resistant cell lines (Fig. 2A). We attempted to investigate whether PTX augments TRAIL-inducing apoptosis and found a significant decrease on cell viability in combination group compared with TRAIL or PTX alone in all 5 resistant cell lines (P<0.05) (Fig. 2B). To confirm this synergism, SGC-7901 and MGC-803 cells were cultured with different combinations of TRAIL (100 and 200 ng/ml) and PTX (5 and 10 nM). The results indicated that PTX promoted TRAIL-mediated cytotoxic effect in a dose-dependent manner (Fig. 2C). Considering changes in the order of drug exposure could enhance cell death (29) and sequential application of PTX followed by other anticancer chemicals is common in GC treatment (30), we tested the possibility of sequential administration of PTX and TRAIL. Cells were pre-incubated with PTX alone for 12 h and subsequently treated with TRAIL for an additional 12 h. Reduction of cell viability was more distinct in the sequential exposure group compared with simultaneous administration group (Fig. 2D), indicating that pre-treatment of PTX rendered cancer cell more vulnerable to TRAIL.

PTX sensitizes GC cells to TRAIL-induced apoptosis by activating caspase-dependent mitochondrial apoptotic pathway

To ascertan whether cell growth inhibition was due to apoptosis, Annexin V/PI double staining was performed. A significant increase in apoptosis was observed in the combination treatment group compared with either agent alone (Fig. 3A and B). Taking SGC-7901 cell line as an example, the mean apoptotic rate was 2.8 and 8% treated with PTX and TRAIL alone, respectively, but as much as 22.4% after combination treatment. The expression of full length and cleaved caspase-3, -7, -8 and -9 and PARP was measured by western blotting (Fig. 3C). Combination of TRAIL and PTX markedly enhanced cleavage of all the caspase proteins compared with single agent alone. Noteworthy, cleavage of caspase-9, but not caspase-8, was induced by PTX, indicating the effect of PTX on activation of mitochondrial pathway. We further tested expression of Bid, another key component in mitochondrial pathway, which was significantly decreased in combination group.

PTX upregulated TRAIL receptors and downregulated anti-apoptotic proteins

Given the therapeutic potential of combined regimen, we focused the following studies on unraveling the molecular mechanism of the synergism. As shown in Fig. 4A, PTX induced DR4 and DR5 expression in both SGC-7901 and MGC-803 cells, albeit the trend was not distinct in MGC-803 cells. Additionally, enhanced cell surface expression of DR4, not DR5, was also examined after PTX treatment by flow cytometric analysis (Fig. 4B). To further uncover the role of PTX on modulating cell death, apoptosis-related proteins were investigated. Notably, combination treatment significantly inhibited expression of anti-apoptotic regulators including C-IAP1, C-IAP2, Livin and Mcl-1 (Fig. 4C). Consistent with these findings, decreased expression of these proteins was also observed in SGC-7901 and MGC-803 cells treated with PTX alone in a dose- and time-dependent manner (Fig. 4D). Moreover, a representative protein C-IAP1 was chosen for functional verification. Silencing of C-IAP1 by siRNA enhanced TRAIL-mediated cytotoxicity and amplified the TRAIL-sensitizing effect of PTX (Fig. 4E). All these results suggested that PTX sensitized TRAIL-induced apoptosis via upregulating DRs and downregulating anti-apoptotic proteins.

PTX induced inactivation of the MAPK pathway

Considering MAPKs have been implicated in regulation of TRAIL-resistance (19,31) in conjunction with the reports that PTX could modulate activation of MAPKs (32), we hypothesized that PTX-mediated MAPK activity may contribute to TRAIL sensitivity in GC cells. To this end, we first evaluated the effect of PTX on MAPK activity. Results depicted in Fig. 5A demonstrated ERK, JNK and p38 were strongly suppressed after treatment of PTX alone as well as in combination with TRAIL. Next, we investigated the possible role of MAPK deprivation on the sensitivity to TRAIL. Treatment of U0126 (ERK inhibitor), SP600125 (JNK inhibitor) and SB202190 (p38 inhibitor) respectively, significantly enhanced TRAIL-induced apoptosis and cytotoxicity, as did PTX (Fig. 5B and C). Among the three inhibitors, SP600125 showed the strongest sensitization effect. To further assess the effect of MAPKs on TRAIL signalling, expression of DRs and anti-apoptotic proteins were evaluated after pretreatment with specific inhibitors. Results showed SP600125 increased DR4 and DR5 expression while U0126 suppressed them indicating MAPKs exhibited opposite effect on DRs (Fig. 5D). Moreover, C-IAP2 was suppressed by SP600125 and Mcl-1 was decreased by all three inhibitors (Fig. 5E). Collectively, these results suggested that PTX-mediated inhibition of MAPKs might contribute to facilitate TRAIL potential in GC cells.

PTX enhances the antitumor effect of TRAIL in a tumor xenograft model

To assess the therapeutic effect of PTX and TRAIL, we established nude mouse models bearing SGC-7901 tumor xenografts. The results revealed that TRAIL or PTX alone only slightly suppressed growth of tumors (TGI was 14.8 and 56.0%, respectively), whereas in the combination group, TGI was 85.1% at the end of drug administration (Fig. 6). The data further confirmed that PTX enhanced the tumor-suppressing capacity of TRAIL in resistant GC cells.

Discussion

Identification of new drugs causing tumor specific apoptosis has roused enormous interest. TRAIL legend and its receptors are attractive targets for the selective eradication of tumor cells. Recombinant TRAIL protein and TRAIL receptor agonistic antibodies have been tested in clinical trials, displaying encouraging antitumor activities with mild side-effects. Nevertheless, resistance to TRAIL limits their clinical application. In the present study, we found not all the cell lines underwent significant apoptosis when treated with TRAIL. Numerous chemotherapeutic agents can sensitize tumor cells to TRAIL-mediated apoptosis (2628). However, there are scarce data elucidating the synergistic interaction between PTX and TRAIL in GC cells. In the present study, we assessed the tumoricidal potential of TRAIL combined with PTX both in vitro and in vivo, and analyzed the mechanism by which PTX sensitized TRAIL-resistant GC cells.

One possible mechanism is synergistic activation of caspase. Apoptosis can be induced by two pathways. TRAIL and PTX share two complementary characteristics for performing apoptosis: one is high efficacy of cell death triggered by death receptor-mediated pathway, and the other is activation of mitochondria-controlled signaling. In the present study, we observed that TRAIL alone only slightly activated caspase-3, -8 and PARP in SGC-7901 and MGC-803 cells. In contrast, simultaneous administration of PTX and TRAIL resulted in a dramatic increase in cleavage of all caspase proteins. Notably, the combined administration markedly activated caspase-9 and induced cleavage of Bid, which are key events in mitochondrial apoptosis signaling, suggesting that PTX facilitated sensitivity to TRAIL by reinforcing mitochondria-mediated apoptosis. This observation is supported by others reporting that PTX induces apoptosis via a death receptor-independent, caspase-3/-8-driven mitochondrial amplification loop (33).

Apoptosis signals are initiated when TRAIL binds to DR4 and DR5. Upregulation of DRs can enhance the responsiveness of cancer cells to TRAIL-mediated cell death (26,34). According to our data, high DR5 expression was prevalent and almost undifferentiated in GC cells. Intriguingly, GC cells shared diverse surface expression of DR4 with the tendency that TRAIL-resistant cell lines exhibited relatively low levels. These findings provided novel evidence that susceptibility of GC cells to TRAIL might be mainly ascribed to the surface expression of DR4. As we found PTX could increase DR4 expression, it was plausible that induction of DR4 would be an efficient way to potentiate tumoricidal potential of TRAIL in GC. Another crucial point for increasing cell susceptibility to TRAIL is inhibition of the anti-apoptotic protein. Previous studies supported that TRAIL-resistant cells were re-sensitized by Bcl-2 or IAP antagonist (17,18). Here we revealed that PTX itself markedly inhibited expression of C-IAP1, C-IAP2, Livin and Mcl-1 in both SGC-7901 and MGC-803 cells, and combined application of PTX and TRAIL exhibited similar effects. In addition, knockdown of C-IAP1 by siRNA augmented cytotoxic potential of TRAIL indicating PTX-mediated suppression of anti-apoptotic proteins contributed to the sensitizing effect.

Noteworthy, we found MAPKs were involved in the PTX-mediated sensitization to TRAIL. MAPK pathway, regulating cell proliferation, differentiation, mitosis and apoptosis, mainly consists of ERK, JNK1/2 and p38 MAPK members. They are frequently activated in GC (35). In this study, we found MAPKs (including ERK, JNK and p38) acted as negative regulators in TRAIL-induced apoptosis. PTX, however, suppressed MAPK activation, abolished the inhibition effect, and therefore partly restored TRAIL sensitivity. We also revealed suppression of JNK by the specific inhibitor SP600125, which showed the most effective sensitization effect. Consistent with the results of Mucha et al (19), JNK inhibition sensitised hepatocellular carcinoma cells to TRAIL. Similarly, ERK abrogated TRAIL-induced apoptosis by phosphorylating pro-caspase-8 and inhibited the cleavage of Bid (36,37). However, other groups reported that enforced activation of ERK and p38 conferred sensitization of tumor cells to TRAIL by suppressing expression of FLIP and increasing expression of DRs (34,38). These inconsistent results indicate that MAPK signaling may play various roles in the way the diverse cancer cells react to TRAIL. Moreover, our results showed that MAPKs played opposing roles on modulating DRs and only JNK inhibitor could induce both DR4 and DR5 expression. In agreement with our results, Kim et al (39) have shown SP600125 upregulated DRs surface expression on hepatocellular cancer cells. We also found MAPK inhibitors could suppress expression of anti-apoptotic protein C-IAP2 and Mcl-1. All these results suggested that inhibition of MAPKs was involved in TRAIL-sensitizing effect of PTX.

In summary, the present study suggested that PTX sensitized resistant GC cells to TRAIL-mediated tumoricidal effect both in vitro and in vivo. Potentiation of mitochondrial apoptotic signals, upregulation of death receptors, downregulation of anti-apoptotic proteins and inactivation of MAPKs were all involved in the synergistic interaction. Our findings strongly suggested that the combination of PTX with TRAIL could serve as a new therapeutic strategy for GC. Further study on the potential application in this direction is warranted.

Acknowledgments

The present study was supported in part by the National Nature Science Foundation of China (nos. 81402308 and 81301874), the Natural Science Foundation of Beijing (no. 7132051), the 985 special project sponsored by the Peking University Health Science Center (no. 2013-5-09), and the Ministry of Science and Technology of the People's Republic of China (Nos. 2014AA020603, 2012AA02A203-B01, 2012AA02A504-B01, 2012AA020101 and 2014AA020603).

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Call JA, Eckhardt SG and Camidge DR: Targeted manipulation of apoptosis in cancer treatment. Lancet Oncol. 9:1002–1011. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Pitti RM, Marsters SA, Ruppert S, Donahue CJ, Moore A and Ashkenazi A: Induction of apoptosis by Apo-2 ligand, a new member of the tumor necrosis factor cytokine family. J Biol Chem. 271:12687–12690. 1996. View Article : Google Scholar : PubMed/NCBI

4 

Kischkel FC, Lawrence DA, Chuntharapai A, Schow P, Kim KJ and Ashkenazi A: Apo2L/TRAIL-dependent recruitment of endogenous FADD and caspase-8 to death receptors 4 and 5. Immunity. 12:611–620. 2000. View Article : Google Scholar : PubMed/NCBI

5 

Samraj AK, Keil E, Ueffing N, Schulze-Osthoff K and Schmitz I: Loss of caspase-9 provides genetic evidence for the type I/II concept of CD95-mediated apoptosis. J Biol Chem. 281:29652–29659. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Herbst RS, Eckhardt SG, Kurzrock R, Ebbinghaus S, O'Dwyer PJ, Gordon MS, Novotny W, Goldwasser MA, Tohnya TM, Lum BL, et al: Phase I dose-escalation study of recombinant human Apo2L/TRAIL, a dual proapoptotic receptor agonist, in patients with advanced cancer. J Clin Oncol. 28:2839–2846. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Soria JC, Smit E, Khayat D, Besse B, Yang X, Hsu CP, Reese D, Wiezorek J and Blackhall F: Phase 1b study of dulanermin (recombinant human Apo2L/TRAIL) in combination with paclitaxel, carboplatin, and bevacizumab in patients with advanced non-squamous non-small-cell lung cancer. J Clin Oncol. 28:1527–1533. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Wiezorek J, Holland P and Graves J: Death receptor agonists as a targeted therapy for cancer. Clin Cancer Res. 16:1701–1708. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Dimberg LY, Anderson CK, Camidge R, Behbakht K, Thorburn A and Ford HL: On the TRAIL to successful cancer therapy? Predicting and counteracting resistance against TRAIL-based therapeutics. Oncogene. 32:1341–1350. 2013. View Article : Google Scholar

10 

Prasad S, Kim JH, Gupta SC and Aggarwal BB: Targeting death receptors for TRAIL by agents designed by Mother Nature. Trends Pharmacol Sci. 35:520–536. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Horak P, Pils D, Haller G, Pribill I, Roessler M, Tomek S, Horvat R, Zeillinger R, Zielinski C and Krainer M: Contribution of epigenetic silencing of tumor necrosis factor-related apoptosis inducing ligand receptor 1 (DR4) to TRAIL resistance and ovarian cancer. Mol Cancer Res. 3:335–343. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Bin L, Thorburn J, Thomas LR, Clark PE, Humphreys R and Thorburn A: Tumor-derived mutations in the TRAIL receptor DR5 inhibit TRAIL signaling through the DR4 receptor by competing for ligand binding. J Biol Chem. 282:28189–28194. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Zhang Y and Zhang B: TRAIL resistance of breast cancer cells is associated with constitutive endocytosis of death receptors 4 and 5. Mol Cancer Res. 6:1861–1871. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Grotzer MA, Eggert A, Zuzak TJ, Janss AJ, Marwaha S, Wiewrodt BR, Ikegaki N, Brodeur GM and Phillips PC: Resistance to TRAIL-induced apoptosis in primitive neuroectodermal brain tumor cells correlates with a loss of caspase-8 expression. Oncogene. 19:4604–4610. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Ricci MS, Kim SH, Ogi K, Plastaras JP, Ling J, Wang W, Jin Z, Liu YY, Dicker DT, Chiao PJ, et al: Reduction of TRAIL-induced Mcl-1 and cIAP2 by c-Myc or sorafenib sensitizes resistant human cancer cells to TRAIL-induced death. Cancer Cell. 12:66–80. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Munshi A, Pappas G, Honda T, McDonnell TJ, Younes A, Li Y and Meyn RE: TRAIL (APO-2L) induces apoptosis in human prostate cancer cells that is inhibitable by Bcl-2. Oncogene. 20:3757–3765. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Finlay D, Vamos M, González-López M, Ardecky RJ, Ganji SR, Yuan H, Su Y, Cooley TR, Hauser CT, Welsh K, et al: Small-molecule IAP antagonists sensitize cancer cells to TRAIL-induced apoptosis: Roles of XIAP and cIAPs. Mol Cancer Ther. 13:5–15. 2014. View Article : Google Scholar :

18 

Kocab AJ, Veloso A, Paulsen MT, Ljungman M and Duckett CS: Effects of physiological and synthetic IAP antagonism on c-IAP-dependent signaling. Oncogene. 34:5472–5481. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Mucha SR, Rizzani A, Gerbes AL, Camaj P, Thasler WE, Bruns CJ, Eichhorst ST, Gallmeier E, Kolligs FT, Göke B, et al: JNK inhibition sensitises hepatocellular carcinoma cells but not normal hepatocytes to the TNF-related apoptosis-inducing ligand. Gut. 58:688–698. 2009. View Article : Google Scholar

20 

Lee HH, Jeong JW, Lee JH, Kim GY, Cheong J, Jeong YK, Yoo YH and Choi YH: Cordycepin increases sensitivity of Hep3B human hepatocellular carcinoma cells to TRAIL-mediated apoptosis by inactivating the JNK signaling pathway. Oncol Rep. 30:1257–1264. 2013.PubMed/NCBI

21 

Liu J, Qu X, Xu L, Zhang Y, Qu J, Hou K and Liu Y: Phosphoinositide 3-kinase/Akt and nuclear factor κB pathways are involved in tumor necrosis factor-related apoptosis-inducing ligand resistance in human gastric cancer cells. Mol Med Rep. 3:491–496. 2010. View Article : Google Scholar

22 

Wan Z, Pan H, Liu S, Zhu J, Qi W, Fu K, Zhao T and Liang J: Downregulation of SNAIL sensitizes hepatocellular carcinoma cells to TRAIL-induced apoptosis by regulating the NF-κB pathway. Oncol Rep. 33:1560–1566. 2015.PubMed/NCBI

23 

Nazim UM, Jeong JK, Seol JW, Hur J, Eo SK, Lee JH and Park SY: Inhibition of the autophagy flux by gingerol enhances TRAIL-induced tumor cell death. Oncol Rep. 33:2331–2336. 2015.PubMed/NCBI

24 

Liu YJ, Lin YC, Lee JC, Kuo SC, Ho CT, Huang LJ, Kuo DH and Way TD: CCT327 enhances TRAIL-induced apoptosis through the induction of death receptors and downregulation of cell survival proteins in TRAIL-resistant human leukemia cells. Oncol Rep. 32:1257–1264. 2014.PubMed/NCBI

25 

Pasquier E, Carré M, Pourroy B, Camoin L, Rebaï O, Briand C and Braguer D: Antiangiogenic activity of paclitaxel is associated with its cytostatic effect, mediated by the initiation but not completion of a mitochondrial apoptotic signaling pathway. Mol Cancer Ther. 3:1301–1310. 2004.PubMed/NCBI

26 

Gong J, Yang D, Kohanim S, Humphreys R, Broemeling L and Kurzrock R: Novel in vivo imaging shows up-regulation of death receptors by paclitaxel and correlates with enhanced antitumor effects of receptor agonist antibodies. Mol Cancer Ther. 5:2991–3000. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Hunter TB, Manimala NJ, Luddy KA, Catlin T and Antonia SJ: Paclitaxel and TRAIL synergize to kill paclitaxel-resistant small cell lung cancer cells through a caspase-independent mechanism mediated through AIF. Anticancer Res. 31:3193–3204. 2011.PubMed/NCBI

28 

Nimmanapalli R, Perkins CL, Orlando M, O'Bryan E, Nguyen D and Bhalla KN: Pretreatment with paclitaxel enhances apo-2 ligand/tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis of prostate cancer cells by inducing death receptors 4 and 5 protein levels. Cancer Res. 61:759–763. 2001.PubMed/NCBI

29 

Lee MJ, Ye AS, Gardino AK, Heijink AM, Sorger PK, MacBeath G and Yaffe MB: Sequential application of anticancer drugs enhances cell death by rewiring apoptotic signaling networks. Cell. 149:780–794. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Tsuburaya A, Yoshida K, Kobayashi M, Yoshino S, Takahashi M, Takiguchi N, Tanabe K, Takahashi N, Imamura H, Tatsumoto N, et al: Sequential paclitaxel followed by tegafur and uracil (UFT) or S-1 versus UFT or S-1 monotherapy as adjuvant chemotherapy for T4a/b gastric cancer (SAMIT): A phase 3 factorial randomised controlled trial. Lancet Oncol. 15:886–893. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Trivedi R, Maurya R and Mishra DP: Medicarpin, a legume phytoalexin sensitizes myeloid leukemia cells to TRAIL-induced apoptosis through the induction of DR5 and activation of the ROS-JNK-CHOP pathway. Cell Death Dis. 5:e14652014. View Article : Google Scholar : PubMed/NCBI

32 

McDaid HM and Horwitz SB: Selective potentiation of paclitaxel (taxol)-induced cell death by mitogen-activated protein kinase kinase inhibition in human cancer cell lines. Mol Pharmacol. 60:290–301. 2001.PubMed/NCBI

33 

von Haefen C, Wieder T, Essmann F, Schulze-Osthoff K, Dörken B and Daniel PT: Paclitaxel-induced apoptosis in BJAB cells proceeds via a death receptor-independent, caspases-3/-8-driven mitochondrial amplification loop. Oncogene. 22:2236–2247. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Do MT, Na M, Kim HG, Khanal T, Choi JH, Jin SW, Oh SH, Hwang IH, Chung YC, Kim HS, et al: Ilimaquinone induces death receptor expression and sensitizes human colon cancer cells to TRAIL-induced apoptosis through activation of ROS-ERK/p38 MAPK-CHOP signaling pathways. Food Chem Toxicol. 71:51–59. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Paterson AL, Shannon NB, Lao-Sirieix P, Ong CA, Peters CJ, O'Donovan M and Fitzgerald RC: A systematic approach to therapeutic target selection in oesophago-gastric cancer. Gut. 62:1415–1424. 2013. View Article : Google Scholar

36 

Söderström TS, Poukkula M, Holmström TH, Heiskanen KM and Eriksson JE: Mitogen-activated protein kinase/extracellular signal-regulated kinase signaling in activated T cells abrogates TRAIL-induced apoptosis upstream of the mitochondrial amplification loop and caspase-8. J Immunol. 169:2851–2860. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Mandal R, Raab M, Matthess Y, Becker S, Knecht R and Strebhardt K: pERK 1/2 inhibit caspase-8 induced apoptosis in cancer cells by phosphorylating it in a cell cycle specific manner. Mol Oncol. 8:232–249. 2014. View Article : Google Scholar

38 

Yerbes R, López-Rivas A, Reginato MJ and Palacios C: Control of FLIP(L) expression and TRAIL resistance by the extracellular signal-regulated kinase1/2 pathway in breast epithelial cells. Cell Death Differ. 19:1908–1916. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Kim EY, Ryu JH and Kim AK: CAPE promotes TRAIL-induced apoptosis through the upregulation of TRAIL receptors via activation of p38 and suppression of JNK in SK-Hep1 hepatocellular carcinoma cells. Int J Oncol. 43:1291–1300. 2013.PubMed/NCBI

Related Articles

Journal Cover

May-2016
Volume 35 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li L, Wen X, Bu Z, Cheng X, Xing X, Wang X, Zhang L, Guo T, Du H, Hu Y, Hu Y, et al: Paclitaxel enhances tumoricidal potential of TRAIL via inhibition of MAPK in resistant gastric cancer cells. Oncol Rep 35: 3009-3017, 2016
APA
Li, L., Wen, X., Bu, Z., Cheng, X., Xing, X., Wang, X. ... Ji, J. (2016). Paclitaxel enhances tumoricidal potential of TRAIL via inhibition of MAPK in resistant gastric cancer cells. Oncology Reports, 35, 3009-3017. https://doi.org/10.3892/or.2016.4666
MLA
Li, L., Wen, X., Bu, Z., Cheng, X., Xing, X., Wang, X., Zhang, L., Guo, T., Du, H., Hu, Y., Fan, B., Ji, J."Paclitaxel enhances tumoricidal potential of TRAIL via inhibition of MAPK in resistant gastric cancer cells". Oncology Reports 35.5 (2016): 3009-3017.
Chicago
Li, L., Wen, X., Bu, Z., Cheng, X., Xing, X., Wang, X., Zhang, L., Guo, T., Du, H., Hu, Y., Fan, B., Ji, J."Paclitaxel enhances tumoricidal potential of TRAIL via inhibition of MAPK in resistant gastric cancer cells". Oncology Reports 35, no. 5 (2016): 3009-3017. https://doi.org/10.3892/or.2016.4666