Open Access

Overcoming resistance to TRAIL-induced apoptosis in solid tumor cells by simultaneously targeting death receptors, c-FLIP and IAPs

  • Authors:
    • Ying Huang
    • Xiang Yang
    • Tianrui Xu
    • Qinghong Kong
    • Yaping Zhang
    • Yuehai Shen
    • Yunlin Wei
    • Guanlin Wang
    • Kwen-Jen Chang
  • View Affiliations

  • Published online on: May 16, 2016     https://doi.org/10.3892/ijo.2016.3525
  • Pages: 153-163
  • Copyright: © Huang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The discovery of the TRAIL protein and its death receptors DR4/5 changed the horizon of cancer research because TRAIL specifically kills cancer cells. However, the validity of TRAIL-based cancer therapies has yet to be established, as most cancer cells are TRAIL-resistant. In this report, we demonstrate that TRAIL-resistance of many cancer cell lines can be overcome after siRNA- or rocaglamide-mediated downregulation of c-FLIP expression and simultaneous inhibition of IAPs activity using AT406, a pan-antagonist of IAPs. Combined triple actions of the TRAIL, the IAPs inhibitor, AT406, and the c-FLIP expression inhibitor, rocaglamide (ART), markedly improve TRAIL-induced apoptotic effects in most solid cancer cell lines through the activation of an extrinsic apoptosis pathway. Furthermore, this ART combination does not harm normal cells. Among the 18 TRAIL-resistant cancer cell lines used, 15 cell lines become sensitive or highly sensitive to ART, and two out of three glioma cell lines exhibit high resistance to ART treatment due to very low levels of procaspase-8. This study provides a rationale for the development of TRAIL-induced apoptosis-based cancer therapies.

Introduction

Traditional chemotherapy and radiotherapy of cancer treatments suffer from severe side effects, development of drug resistance and cross-resistance, cancer migration and recurrence (1). In light of recent breakthroughs in molecular oncology, targeted therapies using monoclonal antibodies to mutated cell surface receptors and small molecule agents inhibiting tyrosine kinases, serine/threonine kinases, small GTP-binding proteins and other oncogenic proteins in the proliferation-driving signaling pathways have become standard in the current treatment of cancer (2). However, due to the genetic heterogeneous nature of cancer cells, particularly for solid tumors, resistance to these targeted agents can develop rapidly, and thus limit the overall efficacy (3). Novel cancer therapies are needed. One potential immuno-surveillance mechanism for therapy is apoptosis induced by cytokines produced by immune cells such as T and natural killer (NK) cells.

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), discovered two decades ago by Wiley et al (4) and Pitti et al (5), attracted enthusiastic attention worldwide as a potential cancer therapy because of its capacity to specifically induce cancer cell death, but not the death of normal and healthy cells (6). TRAIL produced from immune NK cells (7), can induce apoptosis of cancer cells upon binding to the cell surface death receptors (DR, TRAIL receptor), DR4 (or TRAIL R1) and/or DR5 (or TRAIL R2). In addition, TRAIL recruits the adaptor Fas-associated death domain (FADD) and procaspase-8 to form death-inducing signaling complexes (DISC), which results in the activation of the initiator caspase-8, leading to the activation of extrinsic and intrinsic apoptotic signaling downstream of caspase-3 (4,8). Recently, several phase 2 clinical studies based on the use of recombinant human TRAIL or agonistic monoclonal antibodies against DR4/5 have failed to show clinical efficacy, even when combined with traditional chemotherapy (9,10). Thus, enthusiasm has greatly dampened for cancer therapies based on TRAIL-induced apoptosis. Moreover, in the past decade, studies have demonstrated that only a small portion of cancer cells are sensitive to TRAIL, while most tumors were TRAIL-resistant (11,12). This property limits the potential of TRAIL-based cancer therapy.

Currently, inhibitors of the apoptosis proteins, cellular FLICE-like inhibitory protein (c-FLIP) and inhibitors of apoptosis protein (IAPs, including XIAP) are considered to be responsible for cellular TRAIL resistance. The utility of TRAIL-based therapy is dependent on mitigating this TRAIL resistance. IAPs bind to downstream executor caspases-3/6/7/9 to inhibit their activities and prevent the execution of apoptosis (13,14). To overcome this obstacle, IAPs antagonists with excellent activity in vivo have been developed, and several of these antagonist (e.g., AT406) are currently under clinical investigation (1518). These IAP antagonists are second mitochondria-derived activator of caspase (Smac) mimetics. c-FLIP, a procaspase-8 homologue, can compete with procaspase-8 to bind to the death effective domain (DED) of FADD and block the apoptotic signal from upstream of the apoptosis pathway (19). In vitro studies with some cytotoxic anticancer agents revealed that the downregulation of c-FLIP induced by these agents was partly responsible for their pro-apoptotic effects (20).

However, there is no specific antagonist available for c-FLIP (21). Downregulating the expression of c-FLIP through specific siRNA sensitized resistant melanoma cells to TRAIL-induced apoptosis (22). Rocaglamide, a natural product isolated from Aglaia species, is a translational inhibitor of de novo c-FLIP synthesis (23,24). Previous studies showed that a c-FLIP inhibitor and a XIAP inhibitor cooperatively sensitized TRAIL-mediated apoptosis in Hodgkin's lymphoma cells (25). However, no studies have shown that a triple combination can be effective in other solid tumors. Recent genetic analysis for various tumor cells revealed the extremely heterogeneous nature of cancers (1). The results in a single cancer cell line cannot be generalized to other types of cancer cells without empirical evidence. Furthermore, there is no safety testing on normal cells for this combination treatment. In our investigation, a combination of AT406 (A) a pan-antagonist of IAPs, rocaglamide (R) or c-FLIP-siRNA and TRAIL (T) (ART triple combination) was used to evaluate its possible broad spectrum activities on selected 17 solid cancer cell lines (from different tissues or organs), three glioma cell lines and two normal cells (pulp cells and MRC5). In addition, various combination effects were assessed. Our study showed that the ART-triple combination may be applied as a broad-spectrum antitumor therapeutic approach for cancer treatment. We also confirmed that our triple combination treatment had no harmful effects on normal cells tested, similar to TRAIL-only treatment. These features provide a theoretical and experimental basis for the TRAIL-induced apoptosis pathway as a potential target for cancer treatment.

Materials and methods

Cell lines and culture conditions

The cancer cell lines U87, SW480, U251 and U373 were purchased from the Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China). HCT116, HT29, LOVO, H460, SK-OV-3, MDA-MB-231, A549, MCF7, SK-BR-3, T-47D, BT474, U2OS, HeLa, HepG2, MDA-MB-468, Vcap, and MRC5 were purchased from ATCC (MD, USA). HCT116, HT29, LOVO, H460, SK-OV-3, MDA-MB-231, A549, U87, MCF7, SK-BR-3, T-47D, BT474 and SW480 were maintained in RPMI-1640 (Hyclone, USA). U2OS, HeLa, HepG2, MDA-MB-468, Vcap, U251 and U373 were cultured in Dulbecco's modified minimal essential medium (DMEM) growth medium (Hyclone). MRC5 cells (human embryonic lung cells) were maintained in MEM growth medium (Hyclone). All culture media were supplemented with 10% fetal bovine serum (Hyclone). All cancer cells were maintained in a humidified incubator at 37°C with 5% CO2, and passaged with 0.25% trypsin-EDTA when ~80% confluence was reached. The pulp cells were isolated and cultured according to a previously described method (26).

Antibodies and chemicals

The antibodies for immunoblotting were from the following sources: mouse anti-caspase-8 p55/53/43/41/18 (#9746), rabbit anti-PARP p118/89 (#9532) and mouse anti-caspase-3 p35/19/17 (#9668) were from Cell Signaling Technology (Beverly, MA, USA); mouse anti-FADD (#F8053)was from Sigma (St. Louis, MO, USA);rabbit anti-DR5 was from Abcam (Cambridge, MA, USA); mouse anti-c-FLIP (clone 7F10, #ALX-804-961-0100) was from Enzo Life (New York, NY, USA) and mouse anti-GAPDH (sc-365062) was from Santa Cruz Biotechnology (Dallas, TX, USA). Recombinant hTRAIL (#310-04) was from R&D Systems (Minneapolis, MN, USA), rocaglamide (#350-121-C100) was purchased from Enzo Life, AT406 (#S2754) was from Selleckchem (Houston, TX, USA) and valproic acid was from J&K Scientific.

Cell viability assay

Cells were seeded in 96-well cell culture plates and treated the next day with the given agents for the indicated times. The cells were then incubated with 100 μg/well of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) at 37°C for 4 h. Finally, the medium was discarded carefully and 150 μl of dimethyl sulfoxide (DMSO) was added to solubilize the formazan crystals. The absorbance was measured using a Microplate Reader (Perkin-Elmer 2030) at a wavelength of 490 nm. The experiments were performed in triplicate.

Caspase activity

Cells were seeded in 96-well cell culture plates (white/black walled) and treated the next day with the given agents for the indicated times, then manipulated according to the technical bulletin of Caspase-Glo 3/7 assay (Promega).

Western blotting

For each sample, 1×106 cells were lysed using a solubilizing solution [20 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1% NP-40, 1 mM PMSF, 0.02% NaN3, protease inhibitor cocktail tablet; Roche, Mannheim, Germany]. Protein concentration was determined using a Bio-Rad Protein assay kit (Hercules, CA, USA). An equal quantity (10–30 μg) of proteins was separated by 10–15% SDS-PAGE and transferred onto a PVDF membrane (Millipore Corp., Billerica, MA, USA). The membrane was blocked in 10% skim milk (in TBS, pH 7.2, containing 0.1% Tween-20) overnight at 4°C, then incubated with primary antibodies followed by peroxidase-conjugated anti-mouse or anti-rabbit IgG (Thermo Fisher, Inc., Rockford, IL, USA). The epitope was detected using an ECL western blot detection kit (Millipore Corp.). GADPH was used as an internal control for all western blots.

Data analysis

Data were expressed as the means ± SD values for western blots or mean ± SEM for cell viability MTT assays. Statistical analysis was performed by using Graphpad Prism 5 software. The IC50 values were evaluated by SPSS version 19. The one-way ANOVA followed by a post hoc multiple comparison test was used to compare control (positive control) and treated groups. A p-value <0.05 was considered statistically significant. The cell viability and blot experiments were performed in triplicate. Densitometry analysis was performed by using ImageJ software.

Results

Inhibition of c-FLIPS/L and IAPs can increase the sensitivity of U2OS to TRAIL-induced apoptosis

By evaluating TRAIL promotion of tumor cell apoptosis, we found that U2OS is resistant to TRAIL-induced apoptosis. Because c-FLIP is a crucial apoptotic resistance factor, we used the c-FLIP-siRNA overexpression plasmid which was reported in our previous study to inhibit the expression of c-FLIP (Fig. 1A) (27). Furthermore, we determined the effective concentrations of AT406 (SM406), a synthetic Smac mimetic that is a pan-antagonist of IAPs, including XIAP (Fig. 2A, left). The results of an MTT assay showed that in combination with TRAIL with or without c-FLIP-siRNA, AT406 could increase the cell death rate of U2OS (Fig. 1A).

Results of the Caspase-Glo 3/7 assay (Fig. 1B) confirmed that c-FLIP-siRNA in the presence of AT406 activated caspase proteins in the TRAIL-activated apoptotic pathway. That is, apoptosis initiator protein caspase-8, apoptosis executor protein caspase-3 and apoptosis substrate poly(ADP-ribose) polymerase (PARP) were all in their cleaved states to various degrees (Fig. 1C and D). All of these results demonstrated that c-FLIP-siRNA and AT406 together activated the extrinsic apoptotic pathway in the presence of TRAIL to induce apoptosis.

Triple combination of TRAIL/AT406/rocaglamide functions via an extrinsic apoptosis pathway

The c-FLIP-siRNA, mentioned above, cannot be transfected successfully to most cell lines. Rocaglamide is a natural product isolated from Aglaia species and has been previously shown to downregulate the expression of c-FLIP in leukemic T cells (23) and Hodgkin's lymphoma cells (25). Therefore, we tested whether rocaglamide could replace the siRNA. After determining the scope of drug safety of rocaglamide (Fig. 2A), we found that rocaglamide could also inhibit the expression of c-FLIP effectively (Fig. 2C and D). Because AT406 did not interfere with rocaglamide (Fig. 2E), we combined AT406 and rocaglamide with TRAIL. This combination could also increase the cell death rate of U2OS cells (Fig. 2B) and similarly activated caspase-8, caspase-3 and PARP by increasing the cleaved and active forms of these caspases (Fig. 2F and G). This result showed that, rocaglamide could inhibit c-FLIP as well as c-FLIP-siRNA, and further activated the extrinsic apoptotic pathway in combination with AT406 in the presence of TRAIL.

The actions of the triple combination overcame the resistance of most solid tumor cell lines to TRAIL-induced apoptosis

We collected 20 tumor cell lines that originated from colorectal cancers (HCT116, HT29, LOVO, SW480), lung cancers (H460, A549), ovarian cancer (SK-OV-3), osteosarcoma (U2OS), breast cancers (MDA-MB-231, SK-BR-3, T-47D, BT474, MDA-MB-468, MCF7), cervical cancer (HeLa), liver cancer (HepG2), prostate cancer (Vcap) and gliomas (U87, U251, U373) to test for apoptotic effects induced by TRAIL alone (Table I). Among these twenty tumor cell lines, only two cell lines, HCT116 and LOVO, responded to TRAIL; the remaining 18 tumor cell lines were highly resistant.

Table I

The sensitivity to TRAIL-induced apoptosis of 20 cancer cell lines with or without pre-treatment of AT406 (A) or rocaglamide (R) or A + R combination.

Table I

The sensitivity to TRAIL-induced apoptosis of 20 cancer cell lines with or without pre-treatment of AT406 (A) or rocaglamide (R) or A + R combination.

IC50 (ng/ml)Imax (%)


EntryCell lineTATRTARTTART-24 hART-72 h
1LOVO4.2±1.40.3±0.97.4±7.90.5±0.322±733±86±0.3
2HCT11654.2±1912±2.95.4±5.12.7±3.322±817±62±0.1
3HT29>20048.7±21106.8±563.4±1.191±314±37±0.5
4H460>200158±40.79±2.94.3±0.951±734±66±0.6
5BT474>200>20089.3±2626.2±7.897±320±13±0,7
6SK-BR-3>200>200122.3±11921.9±2.197±629±15±0.2
7U2OS>200>200>20011.3±1291±824±23±0.5
8T-47D>200>200>20035.8±13.2102±132±37±0.3
9MDA-MB-231>200>200>20017.8±7.381±633±27±0.4
10SK-OV-3>200>200>20056.6±32.883±741±213±1.8
11Vcap>200>200>20052.5±20100±243±315±1.2
12SW480>200>200>200>20088±1062±814±5.3
13MDA-MB-468>200>200>200>20093±471±27±2.4
14HeLa>200>200>200>200101±357±427±0.4
15Hep G2>200>200>200>20070±676±717±7.9
16A549>200>200>200>20081±868±1232±10.9
17U87>200>200>200>20085±973±836±2.7
18MCF7>200>200>200>200104±993±748±13.2
19U251>200>200>200>200108±7101±1281±3.4
20U373>200>200>200>20094±4102±493±4.4

[i] Twenty different kinds of cancer cell lines were treated with different concentrations of TRAIL for 24 or 72 h, or pretreated with A for 1 h and/or R for 4 h. The cell viability was determined by an MTT assay. The IC50 and Imax (%) were obtained from Fig. 3 as well as the concentration-cell viability inhibition curves (not shown) for the AT and RT conditions. The concentrations which cause 50% reduction of cell viability (IC50 values) were calculated using GraphPad Prism5 software. Imax (%) represents the percentage (%) of cells that remain alive at a maximum dose of 200 ng/ml of TRAIL. The results are shown as the means ± SEM of three independent experiments.

The addition of AT406 to HCT116 (a TRAIL sensitive cell line) or HT29 (a TRAIL-resistant cell line) to antagonize IAPs significantly improved the apoptotic effects of TRAIL (Fig. 3A and B). Pre-treating these two cell lines with rocaglamide to downregulate the expression of c-FLIP showed similar effects (Fig. 3A and B). Furthermore, after a combined pre-treatment of rocaglamide and AT406, TRAIL-resistant HT29 cells became highly sensitive to TRAIL (Fig. 3B).

Next, we treated the 20 cancer cell lines with a combination of AT406 (1 μg/ml), rocaglamide (40 nM) and TRAIL at various concentrations (ART combination) (Table I) for 24 h of treatment. Our results showed that these 20 tumor cell lines could be divided into the following three groups: group 1, highly sensitive (<50% relative cell viability), 11 cell lines (e.g., HCT116) (Fig. 3A); group 2, sensitive (50–80% relative cell viability), 6 cell lines (e.g., HT29) (Fig. 3B); and group 3, resistant (>80% relative cell viability), 3 cell lines (e.g., MCF7) (Fig. 3C). This ART-induced cell death can be further improved over time by extending the treatment to a 72-h incubation. Only three cancer cell lines were resistant to ART even after 72-h treatment (Table I), i.e., the breast cancer cell line, MCF7, and brain cancer cell lines, U373 and U251.

Inhibition of IAPs or c-FLIP alone is not sufficient to achieve the remarkable cell death induced with an ART triple combination

To confirm that an ART triple combination is necessary for inducing profound cell death, we determined the cell viability inhibition concentration (IC50 values) of TRAIL on cancer cells with or without AT406 or rocaglamide pre-treatment (Table I). When cells were treated with TRAIL alone, IC50 values could be obtained only on TRAIL-sensitive LOVO and HCT116 cells. After combination with either AT406 or rocaglamide pre-treatment (AT or RT combination), the potency of TRAIL increased >4-fold (lower IC50 values) in these two cell lines (entries 1 and 2), and IC50 values could be obtained on other cancer cells (8 cell lines for AT, 11 cell lines for RT). ART triple combination further enhanced the apoptosis-inducing effects of TRAIL. For the 11 highly sensitive cancer cell lines, >50% apoptosis was observed after 24-h treatment (entries 1–11). IC50 values using TRAIL on these cells were markedly lower than those values using AT or RT combinations, and 7 of them were <10 ng/ml. When ART exposure was extended to 72 h, the relative cell viability (Imax values) further decreased to <10% for 10 cell lines, and 10–40% for 7 cell lines (column of ART-72 h). Only three cells remained resistant to ART (entries 18–20).

On a protein level, cleaved caspases and PARP were barely detected when U2OS cells were treated with TRAIL alone. However, ART completely activated the extrinsic apoptotic pathway and resulted in cleavage of all related proteins (Fig. 2F and G).

Triple combination is safe for normal cells

Mounting evidence has demonstrated that TRAIL does not hurt normal cells. To assess the safety of ART, cell viability assays were carried out using pulp and MRC5 cells. Our results showed that both ART and its equivalent AT406/c-FLIP-siRNA/TRAIL combination could not induce cell death in these normal cells (Fig. 4A–C). Both of these normal cells were resistant to ART even after a 72-h treatment (Fig. 4D). Next, we evaluated the variation of protein expression levels during TRAIL-induced extrinsic apoptosis. Pulp expression of procaspase-3 and procaspase-8 were much lower than expression in the ovarian cancer cell line SK-OV-3. After stimulation by A/R/T in different combination patterns, we did not observe cleavage of the apoptotic proteins procaspase-3/8, but rather, only a slight upregulation of these two apoptotic proteins (Fig. 4E and F).

Figure 4

Normal cell pulp and MRC5 are resistant to ART combination treatment. (A) The pulp cells were treated with 40 nM rocaglamide for different durations or with/without c-FLIP-siRNA transfection for 24 h. The c-FLIPL/S expression was determined using western blots. (B) Left, the normal cell pulp was treated with different concentrations of TRAIL for 24 h in combination with c-FLIP-siRNA (si-FLIP, transient transfection with c-FLIP-siRNA for 24 h), AT406 (A) or empty vector (EV) as indicated in the graph. (B) Right, pulp cells were treated with various concentrations of TRAIL for 24 h with or without the combination of A and R as indicated in the graph. (C) MRC5 cells were treated with various concentrations of TRAIL for 24 h with or without the combination of A and R as indicated in the graph. In experiments of B and C, cells were pretreated by R at 40 nM for 4 h, and A at 1 μg/ml for 1-h pretreatment. The cell viability was determined by an MTT assay. (D) The time course of cell viability of pulp and MRC5 cells after treatment with the ART combination. In cell viability assays, results are shown as the means ± SEM of three independent experiments. (E) The ART triple combination did not activate the extrinsic apoptosis pathway. The normal cell pulp was treated with TRAIL at 100 ng/ml for 5 h, with or without the pretreatment of A at 1 μg/ml for 1 h and R at 40 nM for 4 h as indicated in the graph. The ovarian cancer cell SK-OV-3 is shown as a positive control after ART combination treatments. (F) Statistical analysis of western blots in (E). Means ± SD are shown in all bar graphs of western blots on the right side. The results are representative of three independent experiments.

ART-resistance may attribute to the low expression of procaspases

To investigate the molecular mechanism of TRAIL- and ART-resistance in normal and cancer cells, western blot analysis was carried out for the key proteins involved in the TRAIL apoptotic pathway, i.e., DR5, FADD, procaspase-3/-8 and c-FLIPL/S (Fig. 5A and B). DR5 was expressed in all of the cell lines, but levels in TRAIL-resistant cells were lower. FADD was expressed in all cells at levels unrelated to TRAIL- or ART-resistance. The expression of c-FLIPL was found in all ten cell lines, although it was relatively higher in U87, U251 and pulp cells. c-FLIPS could be clearly observed in HT29, U87 and pulp cells, and it was difficult to detect in other cells. Notably, procaspase-8 was expressed at relatively high levels in ART-sensitive cells (LOVO, HCT116, HT29, U2OS and SK-OV-3 cells) and at significantly lower levels in ART-resistant cells (U251, U373 and pulp cells). In contrast, procaspase-3 in ART-resistant MCF7 cells was nearly invisible. Valproic acid (VA) is an antiepileptic drug with histone deacetylase inhibitory activity (28). We used VA at an appropriate concentration to increase procaspase-8 expression (Fig. 6). The improvement of procaspase-8 expression resulted in the enhancement of apoptotic rate (Fig. 6B–D). Overall, levels of procaspase-3/-8 correlated with ART-sensitivity.

Discussion

Recombinant human TRAIL and several agonistic monoclonal antibodies of DR4/5 have been developed and used in clinical trials (2830). However, their development as a cancer therapy is hampered by the resistance observed in most cancer cells (31,32). Although synergy has been described for combinations of TRAIL with a variety of cytotoxic agents, including etoposide, 5-FU, oxamflatin, sorbitol, staurosporine, MG132, bortezomib, doxorubicin, azacitidine and sorafenibin, these synergistic effects were apparent in only TRAIL-sensitive tumor cells (33). However, in resistant tumor cells, a combination of TRAIL or DR4/5 agonist with chemotherapy showed no profound effect on cell death.

In this study, we hypothesized that TRAIL resistance could be overcome by simultaneously attacking three apoptosis inhibiting factors. First, we demonstrated that a higher concentration of TRAIL (200 ng/ml) would be necessary for competitive binding of DcR1/2 (34). Second, we showed that the apoptosis-inhibiting function of IAPs can be blocked by adding AT406, a small molecule IAPs inhibitor in clinical trials. Third, we showed that the cytosolic apoptotic inhibitor c-FLIPL/S can be downregulated by transient transfection of a plasmid containing c-FLIP-siRNA or by adding rocaglamide, a known c-FLIP expression suppressor (Fig. 5C). We firmly believe that only with the elimination of the main inhibitory factors of an extrinsic apoptotic pathway, can a drug combination based on TRAIL achieve the strongest and broadest therapeutic effects. This may also be the reason for failure of clinical trials of TRAIL or agonist monoclonal antibodies of death receptors and cytotoxic agent combinations on different cancer types. Additionally, in vitro culture systems are limited in making in vivo predictions because TRAIL is quickly degraded in vivo (35,36).

A double combination of either AT or RT only slightly improved the TRAIL-induced apoptosis on cancer cells. In contrast, powerful combined effects were observed for ART on most of the resistant cancer cells. Among the 18 TRAIL-resistant cancer cells, after a 72-h triple combination treatment, 8 became highly sensitive (<10% relative cell viability), and 7 became sensitive (10–50% relative cell viability). The normal pulp and MRC5 cells remained highly resistant to an ART triple combination treatment (Fig. 3 and Table I). These results clearly demonstrate that the ART triple combination generates effects that specifically activates an extrinsic apoptotic pathway in solid tumor cells. The profound apoptotic effects of ART triple combination on cell lines originally from solid tumors also suggests that c-FLIPs and IAPs contribute to the high resistance of these cancer cells to TRAIL-induced apoptosis. Therefore, a combination of DR 4/5 agonist, IAPs antagonist and a c-FLIP antagonist, i.e., the ART combination and its equivalents, is likely a better cancer therapy than TRAIL or agonistic monoclonal antibodies of the death receptors alone. These results also suggest that simultaneous activation of death receptors, inhibition of c-FLIP and antagonization of IAPs are minimally required for induction of profound apoptosis of solid tumor cells.

It is worth noting that ART triple combination does not impair normal cells. Previous research suggests that TRAIL-resistant cancer cells are resistant due to the expression of only one anti-apoptotic protein and that these cells have lost the redundancy in resistance mechanisms observed in non-transformed cells (37). In contrast, our study suggests that most of these highly resistant cancer cells rely on multiple tolerance mechanisms rather than only one resistance mechanism. We suggest that TRAIL-resistance mechanisms of normal cells are far more complicated than cancer cells and at the same time, confirm that ART triple combination is safe for normal primary culture cells from adults or normal passage cells from the human embryo.

Among the 20 cancer cell lines used in this study, the breast cancer cell line MCF7 and two brain cancer cell lines remained highly resistant to ART triple combination, even after a 72-h exposure (Table I and Fig. 3). Western blot analysis revealed that c-FLIP is expressed in these cell lines; however, the levels of procaspase-8 in these cell lines are lower than in the other cell lines. In addition, MCF7 showed extremely low, nearly undetectable, levels of procaspase-3 (Fig. 5). These results suggest that low expression or loss of procaspase-8 and -3 is a likely mechanism for resistance to the ART combination. MCF-7 is deficient of caspase-3 and is relatively insensitive to many traditional chemotherapeutic agents (3840). Because of the indispensable role of the extrinsic apoptotic pathway, deficiency of caspase-3 in MCF-7 results in high TRAIL-resistance during exposure to the ART combination.

Eggert et al (41) reported that only one out of 18 neuroblastoma cell lines showed sensitivity to TRAIL-induced apoptosis. This number of TRAIL sensitive cell lines increased to five by adding cycloheximide (CHX, a protein synthesis inhibitor) to cultured cells to inhibit the synthesis of c-FLIP. The remaining 13 TRAIL-resistant neuroblastoma cell lines (70%) showed a loss of procaspase-8 expression, correlating with resistance to TRAIL-induced apoptosis. These results suggest that in addition to c-FLIP, the loss of procaspase-8 plays a major role in TRAIL resistance in brain cancer cells that originated from a neuroblastoma. In our present study, similarly, all three glioma cell lines and normal pulp cells showed high resistance to TRAIL, as well as ART triple combination treatment (Fig. 3 and Table I). Two (U373 and U251) of these glioma cell lines and pulp cells also expressed at very low levels of procaspase-8 and variable levels of c-FLIPL/S, suggesting that the resistance of these cells to TRAIL and ART are the results of low level expression of procaspase-8 and the presence of anti-apoptosis factors, such as c-FLIP and/or IAPs. The underlying mechanisms of TRAIL resistance caused by decreased expression of the caspase-8 initiator have been attributed to epigenetic silencing, such as DNA methylation, histone acetylation modification (4245). Indeed, when histone deacetylase inhibitor was used on three glioma cell lines, the procaspase-8 expression increased. There is more cell apoptosis under ART treatment (Fig. 6). Decreased caspase-8 activities in some cancer cells are related to procaspase-8 gene mutation (41,4648), decrease in stability, and incomplete activation (4246,49,50) (Fig. 5C). The loss of procaspase-8 expression is particularly prevalent in both neuroblastoma (39) and glioma brain cancer cells. These results may explain the poor prognosis of patients suffering from malignant brain cancers regardless of the therapy.

Recently reported unsuccessful clinical phase 2 studies of rhTRAIL or agonistic monoclonal antibodies to death receptors using a randomly unselected patient population, with and without traditional chemotherapy (10,11), suggest that future clinical studies should consider the aforementioned mechanisms of TRAIL resistance in tumors. For peripheral solid tumors, it appears that a majority of tumor cells are sensitive to ART triple combination therapy; however, a small number of these tumors may have mutations in the procaspase-8 gene rendering caspase-8 inactive (50). Future clinical protocol designs and prognosis analysis based on death receptors should exclude patients with abnormal expression of procaspase-8 (51).

Indeed, the TRAIL resistance of cancer cell lines was successfully reversed with ART in the majority of peripheral solid tumor cells with the exception of brain cancer cells. While being used alone, AT406 has only a limited effect on cancer cell death (Fig. 2A). For rocaglamide, certain degrees of cytotoxicity were observed on both cancer cell lines and normal cells (Fig. 2A). The fact that rocaglamide is cytotoxic to normal cells could raise concerns for the future application of ART combination therapy. To study the cause of this cytotoxicity, we carried out additional experiments in which rocaglamide was replaced with c-FLIP-siRNA. However, similar results were obtained using c-FLIP-siRNA alone or in combination with TRAIL and AT406, confirming that this cytotoxicity is the result of c-FLIP downregulation and the suppression of the anti-apoptotic functions of c-FLIP (52,53). Therefore, it is highly desirable to develop a specific disruptor or antagonist of c-FLIP-FADD interactions to avoid the cyto-toxicity caused by changes in the cellular levels of c-FLIP in normal cells.

Acknowledgements

This study was supported by Chinese National Natural Science Foundation, grant no. 81360162, 81260351 and U1302225.

References

1 

Tannock IF: Cancer: Resistance through repopulation. Nature. 517:152–153. 2015. View Article : Google Scholar

2 

Santarpia L, Lippman SM and El-Naggar AK: Targeting the MAPK-RAS-RAF signaling pathway in cancer therapy. Expert Opin Ther Targets. 16:103–119. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Sriraman SK, Aryasomayajula B and Torchilin VP: Barriers to drug delivery in solid tumors. Tissue Barriers. 2:e295282014. View Article : Google Scholar : PubMed/NCBI

4 

Wiley SR, Schooley K, Smolak PJ, Din WS, Huang CP, Nicholl JK, Sutherland GR, Smith TD, Rauch C, Smith CA, et al: Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity. 3:673–682. 1995. View Article : Google Scholar : PubMed/NCBI

5 

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

6 

Ashkenazi A, Pai RC, Fong S, Leung S, Lawrence DA, Marsters SA, Blackie C, Chang L, McMurtrey AE, Hebert A, et al: Safety and antitumor activity of recombinant soluble Apo2 ligand. J Clin Invest. 104:155–162. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Takeda K, Hayakawa Y, Smyth MJ, Kayagaki N, Yamaguchi N, Kakuta S, Iwakura Y, Yagita H and Okumura K: Involvement of tumor necrosis factor-related apoptosis-inducing ligand in surveillance of tumor metastasis by liver natural killer cells. Nat Med. 7:94–100. 2001. View Article : Google Scholar : PubMed/NCBI

8 

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

9 

Lemke J, von Karstedt S, Zinngrebe J and Walczak H: Getting TRAIL back on track for cancer therapy. Cell Death Differ. 21:1350–1364. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Holland PM: Death receptor agonist therapies for cancer, which is the right TRAIL? Cytokine Growth Factor Rev. 25:185–193. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Dyer MJ, MacFarlane M and Cohen GM: Barriers to effective TRAIL-targeted therapy of malignancy. J Clin Oncol. 25:4505–4506. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Koschny R, Walczak H and Ganten TM: The promise of TRAIL - potential and risks of a novel anticancer therapy. J Mol Med Berl. 85:923–935. 2007. View Article : Google Scholar

13 

Salvesen GS and Duckett CS: IAP proteins: Blocking the road to death's door. Nat Rev Mol Cell Biol. 3:401–410. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Fulda S: Inhibitor of apoptosis proteins as targets for anticancer therapy. Expert Rev Anticancer Ther. 7:1255–1264. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Cai Q, Sun H, Peng Y, Lu J, Nikolovska-Coleska Z, McEachern D, Liu L, Qiu S, Yang CY, Miller R, et al: A potent and orally active antagonist (SM-406/AT-406) of multiple inhibitor of apoptosis proteins (IAPs) in clinical development for cancer treatment. J Med Chem. 54:2714–2726. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Fulda S, Wick W, Weller M and Debatin KM: Smac agonists sensitize for Apo2L/TRAIL- or anticancer drug-induced apoptosis and induce regression of malignant glioma in vivo. Nat Med. 8:808–815. 2002.PubMed/NCBI

17 

Fakler M, Loeder S, Vogler M, Schneider K, Jeremias I, Debatin KM and Fulda S: Small molecule XIAP inhibitors cooperate with TRAIL to induce apoptosis in childhood acute leukemia cells and overcome Bcl-2-mediated resistance. Blood. 113:1710–1722. 2009. View Article : Google Scholar

18 

Vogler M, Walczak H, Stadel D, Haas TL, Genze F, Jovanovic M, Bhanot U, Hasel C, Möller P, Gschwend JE, et al: Small molecule XIAP inhibitors enhance TRAIL-induced apoptosis and antitumor activity in preclinical models of pancreatic carcinoma. Cancer Res. 69:2425–2434. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Irmler M, Thome M, Hahne M, Schneider P, Hofmann K, Steiner V, Bodmer JL, Schröter M, Burns K, Mattmann C, et al: Inhibition of death receptor signals by cellular FLIP. Nature. 388:190–195. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Aoudjit F and Vuori K: Integrin signaling in cancer cell survival and chemoresistance. Chemother Res Pract. 2012:2831812012.PubMed/NCBI

21 

Safa AR and Pollok KE: Targeting the anti-apoptotic protein c-FLIP for cancer therapy. Cancers (Basel). 3. pp. 1639–1671. 2011, View Article : Google Scholar

22 

Chawla-Sarkar M, Bae SI, Reu FJ, Jacobs BS, Lindner DJ and Borden EC: Downregulation of Bcl-2, FLIP or IAPs (XIAP and survivin) by siRNAs sensitizes resistant melanoma cells to Apo2L/TRAIL-induced apoptosis. Cell Death Differ. 11:915–923. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Bleumink M, Köhler R, Giaisi M, Proksch P, Krammer PH and Li-Weber M: Rocaglamide breaks TRAIL resistance in HTLV-1-associated adult T-cell leukemia/lymphoma by translational suppression of c-FLIP expression. Cell Death Differ. 18:362–370. 2011. View Article : Google Scholar :

24 

Zhu JY, Giaisi M, Köhler R, Müller WW, Mühleisen A, Proksch P, Krammer PH and Li-Weber M: Rocaglamide sensitizes leukemic T cells to activation-induced cell death by differential regulation of CD95L and c-FLIP expression. Cell Death Differ. 16:1289–1299. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Giaisi M, Kohler R, Fulda S, Krammer PH and Li-Weber M: Rocaglamide and a XIAP inhibitor cooperatively sensitize TRAIL-mediated apoptosis in Hodgkin's lymphomas. Int J Cancer. 131:1003–1008. 2012. View Article : Google Scholar

26 

Gronthos S, Mankani M, Brahim J, Robey PG and Shi S: Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci USA. 97:13625–13630. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Zhang YP, Kong QH, Huang Y, Wang GL and Chang KJ: Inhibition of c-FLIP by RNAi enhances sensitivity of the human osteogenic sarcoma cell line U2OS to TRAIL-induced apoptosis. Asian Pac J Cancer Prev. 16:2251–2256. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Ziauddin MF, Yeow WS, Maxhimer JB, Baras A, Chua A, Reddy RM, Tsai W, Cole GW Jr, Schrump DS and Nguyen DM: Valproic acid, an antiepileptic drug with histone deacetylase inhibitory activity, potentiates the cytotoxic effect of Apo2L/TRAIL on cultured thoracic cancer cells through mitochondria-dependent caspase activation. Neoplasia. 8:446–457. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Micheau O, Shirley S and Dufour F: Death receptors as targets in cancer. Br J Pharmacol. 169:1723–1744. 2013. View Article : Google Scholar : PubMed/NCBI

30 

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

31 

Graves JD, Kordich JJ, Huang TH, Piasecki J, Bush TL, Sullivan T, Foltz IN, Chang W, Douangpanya H, Dang T, et al: Apo2L/TRAIL and the death receptor 5 agonist antibody AMG 655 cooperate to promote receptor clustering and antitumor activity. Cancer Cell. 26:177–189. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Fulda S: Safety and tolerability of TRAIL receptor agonists in cancer treatment. Eur J Clin Pharmacol. 71:525–527. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Sayers TJ: Targeting the extrinsic apoptosis signaling pathway for cancer therapy. Cancer Immunol Immunother. 60:1173–1180. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Menke C, Bin L, Thorburn J, Behbakht K, Ford HL and Thorburn A: Distinct TRAIL resistance mechanisms can be overcome by proteasome inhibition but not generally by synergizing agents. Cancer Res. 71:1883–1892. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Daniel PT, Wieder T, Sturm I and Schulze-Osthoff K: The kiss of death: Promises and failures of death receptors and ligands in cancer therapy. Leukemia. 15:1022–1032. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Schneider P, Olson D, Tardivel A, Browning B, Lugovskoy A, Gong D, Dobles M, Hertig S, Hofmann K, Van Vlijmen H, et al: Identification of a new murine tumor necrosis factor receptor locus that contains two novel murine receptors for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). J Biol Chem. 278:5444–5454. 2003. View Article : Google Scholar

37 

Wu GS, Burns TF, Zhan Y, Alnemri ES and El-Deiry WS: Molecular cloning and functional analysis of the mouse homologue of the KILLER/DR5 tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptor. Cancer Res. 59:2770–2775. 1999.PubMed/NCBI

38 

van Dijk M, Halpin-McCormick A, Sessler T, Samali A and Szegezdi E: Resistance to TRAIL in non-transformed cells is due to multiple redundant pathways. Cell Death Dis. 4:e7022013. View Article : Google Scholar : PubMed/NCBI

39 

Jänicke RU, Sprengart ML, Wati MR and Porter AG: Caspase-3 is required for DNA fragmentation and morphological changes associated with apoptosis. J Biol Chem. 273:9357–9360. 1998. View Article : Google Scholar : PubMed/NCBI

40 

Yang XH, Sladek TL, Liu X, Butler BR, Froelich CJ and Thor AD: Reconstitution of caspase 3 sensitizes MCF-7 breast cancer cells to doxorubicin- and etoposide-induced apoptosis. Cancer Res. 61:348–354. 2001.PubMed/NCBI

41 

Eggert A, Grotzer MA, Zuzak TJ, Wiewrodt BR, Ho R, Ikegaki N and Brodeur GM: Resistance to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in neuroblastoma cells correlates with a loss of caspase-8 expression. Cancer Res. 61:1314–1319. 2001.PubMed/NCBI

42 

Hopkins-Donaldson S, Ziegler A, Kurtz S, Bigosch C, Kandioler D, Ludwig C, Zangemeister-Wittke U and Stahel R: Silencing of death receptor and caspase-8 expression in small cell lung carcinoma cell lines and tumors by DNA methylation. Cell Death Differ. 10:356–364. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Duiker EW, Meijer A, van der Bilt ARM, Meersma GJ, Kooi N, van der Zee AG, de Vries EG and de Jong S: Drug-induced caspase 8 upregulation sensitises cisplatin-resistant ovarian carcinoma cells to rhTRAIL-induced apoptosis. Br J Cancer. 104:1278–1287. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Jia SH, Parodo J, Kapus A, Rotstein OD and Marshall JC: Dynamic regulation of neutrophil survival through tyrosine phosphorylation or dephosphorylation of caspase-8. J Biol Chem. 283:5402–5413. 2008. View Article : Google Scholar

45 

Jin Z, Li Y, Pitti R, Lawrence D, Pham VC, Lill JR and Ashkenazi A: Cullin3-based polyubiquitination and p62-dependent aggregation of caspase-8 mediate extrinsic apoptosis signaling. Cell. 137:721–735. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Bellail AC, Olson JJ, Yang X, Chen ZJ and Hao C: A20 ubiquitin ligase-mediated polyubiquitination of RIP1 inhibits caspase-8 cleavage and TRAIL-induced apoptosis in glioblastoma. Cancer Discov. 2:140–155. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Fulda S, Küfer MU, Meyer E, van Valen F, Dockhorn-Dworniczak B and Debatin KM: Sensitization for death receptor- or drug-induced apoptosis by re-expression of caspase-8 through demethylation or gene transfer. Oncogene. 20:5865–5877. 2001. View Article : Google Scholar : PubMed/NCBI

48 

Casciano I, De Ambrosis A, Croce M, Pagnan G, Di Vinci A, Allemanni G, Banelli B, Ponzoni M, Romani M and Ferrini S: Expression of the caspase-8 gene in neuroblastoma cells is regulated through an essential interferon-sensitive response element (ISRE). Cell Death Differ. 11:131–134. 2004. View Article : Google Scholar

49 

Fulda S and Debatin KM: IFNgamma sensitizes for apoptosis by upregulating caspase-8 expression through the Stat1 pathway. Oncogene. 21:2295–2308. 2002. View Article : Google Scholar : PubMed/NCBI

50 

Crowder RN and El-Deiry WS: Caspase-8 regulation of TRAIL-mediated cell death. Exp Oncol. 34:160–164. 2012.PubMed/NCBI

51 

Soung YH, Lee JW, Kim SY, Sung YJ, Park WS, Nam SW, Kim SH, Lee JY, Yoo NJ and Lee SH: Caspase-8 gene is frequently inactivated by the frameshift somatic mutation 1225_1226delTG in hepatocellular carcinomas. Oncogene. 24:141–147. 2005. View Article : Google Scholar

52 

Shirley S and Micheau O: Targeting c-FLIP in cancer. Cancer Lett. 332:141–150. 2013. View Article : Google Scholar

53 

Wajant H, Haas E, Schwenzer R, Muhlenbeck F, Kreuz S, Schubert G, Grell M, Smith C and Scheurich P: Inhibition of death receptor-mediated gene induction by a cycloheximide-sensitive factor occurs at the level of or upstream of Fas-associated death domain protein (FADD). J Biol Chem. 275:24357–24366. 2000. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2016
Volume 49 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang Y, Yang X, Xu T, Kong Q, Zhang Y, Shen Y, Wei Y, Wang G and Chang K: Overcoming resistance to TRAIL-induced apoptosis in solid tumor cells by simultaneously targeting death receptors, c-FLIP and IAPs. Int J Oncol 49: 153-163, 2016
APA
Huang, Y., Yang, X., Xu, T., Kong, Q., Zhang, Y., Shen, Y. ... Chang, K. (2016). Overcoming resistance to TRAIL-induced apoptosis in solid tumor cells by simultaneously targeting death receptors, c-FLIP and IAPs. International Journal of Oncology, 49, 153-163. https://doi.org/10.3892/ijo.2016.3525
MLA
Huang, Y., Yang, X., Xu, T., Kong, Q., Zhang, Y., Shen, Y., Wei, Y., Wang, G., Chang, K."Overcoming resistance to TRAIL-induced apoptosis in solid tumor cells by simultaneously targeting death receptors, c-FLIP and IAPs". International Journal of Oncology 49.1 (2016): 153-163.
Chicago
Huang, Y., Yang, X., Xu, T., Kong, Q., Zhang, Y., Shen, Y., Wei, Y., Wang, G., Chang, K."Overcoming resistance to TRAIL-induced apoptosis in solid tumor cells by simultaneously targeting death receptors, c-FLIP and IAPs". International Journal of Oncology 49, no. 1 (2016): 153-163. https://doi.org/10.3892/ijo.2016.3525