TRAIL and agonistic death receptor-specific antibodies can induce apoptosis in cancer cells with little cytotoxicity to normal cells. To improve TRAIL-induced antitumor effects, we tested its effectiveness in combination with pifithrin (PFT)-μ, which has the potential to inhibit HSP70 function and autophagy, both of which participate in TRAIL resistance in cancer cells. Among the four human pancreatic cancer cell lines tested, MiaPaca-2, Panc-1, and BxPC-3 cells showed varying sensitivities to TRAIL. In MiaPaca-2 and Panc-1 cells, knockdown of HSP70 or beclin-1, the latter an autophagy-related molecule, by RNA interference augmented TRAIL-induced antitumor effects, decreasing cell viability, and increasing apoptosis. On the basis of these findings, we next determined whether the TRAIL-induced antitumor effects could be augmented by its combination with PFT-μ. The combination of TRAIL plus PFT-μ significantly decreased the viability and colony-forming ability of MiaPaca-2 and Panc-1 cells compared with cells treated with either agent alone. When applied alone, PFT-μ increased Annexin V+ cells in both caspase-dependent and -independent manners. It also promoted TRAIL-induced apoptosis and arrested cancer cell growth. Furthermore, PFT-μ antagonized TRAIL-associated NF-κB activation in cancer cells. In a xenograft mouse model, combination therapy significantly inhibited MiaPaca-2 tumor growth compared with treatment with either agent alone. The results of this study suggest protective roles for HSP70 and autophagy in TRAIL resistance in pancreatic cancer cells and suggest that PFT-μ is a promising agent for use in therapies intended to enhance the antitumor effects of TRAIL. Mol Cancer Ther; 12(4); 341–51. ©2013 AACR.

Members of the TNF cytokine family, such as TNF-α and the Fas ligand, play important roles in inflammation and immunity (1). However, their use in anticancer therapy is limited because they cause severe cytotoxicity to normal cells. In contrast, TRAIL, a member of the TNF superfamily, can induce apoptosis in cancer cells while causing almost no cytotoxicity to normal cells (2). Therefore, TRAIL has been applied for several clinical trials (3, 4). TRAIL binds to death receptors (DR) and activates caspases, leading to apoptosis. However, some cancer cells acquire TRAIL resistance (5). The binding of TRAIL to death receptors activates NF-κB, PKC, mitogen-activated protein kinase (MAPK), and AKT signaling (6, 7), thereby promoting cell proliferation and apoptosis resistance. Notably, activation of NF-κB signaling is an important TRAIL resistance mechanism (6, 8). Indeed, combination therapy with TRAIL plus NF-κB inhibitors has been reported to be a promising treatment strategy (9–11).

Cancer cells acquire therapy resistance through a variety of mechanisms. One is increased expression of HSP70 (12). In contrast to the very low level in unstressed normal cells, HSP70 expression is rapidly increased in response to various stresses (13, 14). Importantly, increased expression of HSP70 in cancer cells has been reported to be associated with malignant features and poor prognosis in patients with cancer (15). Autophagy, meanwhile, has received much attention as a mechanism of therapy resistance in cancer cells. Its fundamental role in cells is cytoprotection under starvation and stress conditions (16), but this protective function can render cancer cells therapy-resistant (17). Indeed, many reports suggest that inhibition of autophagy in cancer cells can restore their susceptibility to anticancer therapy (18–20). Recently, we reported that autophagy inhibits apoptosis of human prostate and breast cancer cells treated with an innate adjuvant receptor ligand (21, 22). These lines of evidence suggest that HSP70 and autophagy are promising targets for therapies intended to enhance TRAIL-induced antitumor effects, something that has not yet been fully investigated.

Pifithrin (PFT)-μ (2-phenylethynesulfonamide) was initially identified as a small-molecule inhibitor of p53 binding to mitochondria (23). Recently, it was revealed to interact selectively with HSP70 and to inhibit its functions (24). In addition, it induces altered autophagy, leading to inhibition of the later autophagic pathway. Furthermore, PFT-μ inhibits the degradation of IκBα, resulting in inhibition of the NF-κB pathway. This information led us to test the possibility that PFT-μ enhances TRAIL-induced antitumor effects in human pancreatic cancer. In this study, after confirming that HSP70 and autophagy play protective roles in TRAIL-induced antitumor effects, we showed that the combination of TRAIL and PFT-μ decreased the viability and colony-forming ability of pancreatic cancer cells and increased their cell death. Using a xenograft mouse model, we also showed that the combination therapy significantly decreased pancreatic tumor growth as compared with treatment with either agent alone. These results suggest that PFT-μ is a promising enhancer of TRAIL-induced antitumor effects on human pancreatic cancer.

Cell lines

Four human pancreatic cancer cell lines (MiaPaca-2, Panc-1, AsPC-1, and BxPC-3), which were kindly provided by Dr. K. Takenaga (Shimane University Faculty of Medicine, Shimane, Japan), were maintained in Dulbecco's Modified Eagle's Medium (Sigma-Aldrich) supplemented with 10% fetal calf serum (FCS; Invitrogen) and 20 μg/mL gentamicin (Sigma-Aldrich) at 37°C in a humidified atmosphere containing 5% CO2. No authentication was done by the authors.

Cell viability assay

Cell viability was evaluated using the 2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2, 4-disulfophenyl)-2H-tetrazolium monosodium salt (WST-8) assay (Nacalai Tesque). Briefly, cells were seeded in flat-bottomed 96-well plates. The next day, TRAIL (PeproTech Inc.) and/or PFT-μ (Santa Cruz Biotechnology, or Cayman Chemical) were added. Two days later, WST-8 was added to each well and the plates were read at a wavelength of 450 nm after 3 hours. For inhibition assays, z-VAD-fmk (R&D Systems) was added and dimethyl sulfoxide (DMSO) was used as a vehicle control.

Flow cytometry

Cell death was measured using the Annexin V–FITC Apoptosis Detection Kit (BioVision) and propidium iodide (PI). To examine the cell cycle and proliferation of cancer cells, a bromodeoxyuridine (BrdUrd)/7-amino-actinomycin D (7AAD) Proliferation Kit (Becton Dickinson) was used according to the manufacturer's instructions. Analysis was conducted using a FACSCalibur flow cytometer (Becton Dickinson).

Immunoblot

Cells were lysed with a mammalian protein extraction reagent (M-PER; Thermo Scientific) containing a protease inhibitor cocktail (Nacalai Tesque). Equal amounts of protein were resolved on 4% to 12% gradient or 12% SDS-PAGE gels and then transferred to polyvinylidene fluoride membranes. The membranes were blocked and the blots then incubated with the following primary antibodies: anti-LC3 (MBL), anti-cyclin D1 (Cell Signaling Technology), anti-c-Myc (Epitomics), anti-beclin-1 (Cell Signaling Technology), anti-cathepsin L (Santa Cruz Biotechnology), anti-IκBα (Cell Signaling Technology), anti-β-actin (BioLegend), and anti-α-tubulin (Santa Cruz Biotechnology). Goat anti-rabbit and goat anti-mouse alkaline phosphatase–conjugated secondary antibodies (Invitrogen) were used to detect the primary antibodies.

Transfection of siRNA

Transfection of siRNA was conducted using Lipofectamine RNAiMAX (Invitrogen), according to the manufacturer's instructions. HSP70 siRNA (sc-29357) and beclin-1 siRNA (sc-29797) were purchased from Santa Cruz Biotechnology. Control siRNA (#6568) was purchased from Cell Signaling Technology. Three days after siRNA transfection, cancer cells were used for subsequent experiments.

Confocal imaging

LC3B (NM_022818) was amplified by PCR and inserted into the pcDNA3.1/NT-GFP-TOPO vector (Invitrogen) in frame with the GFP sequence. Transfection of plasmids was conducted using Lipofectamine 2000 (Invitrogen), according to the manufacturer's instructions. Cells were cultured on round microscope cover glasses in 24-well plates with the indicated reagents for 2 days. After incubation with Hoechst 33342 (5 μg/mL) for 30 minutes, cells were fixed with 3% formalin and placed on slide glasses with 4 μL of mounting medium for fluorescence (Vectashield; Vector Laboratories, Inc.). To examine NF-κB translocation to the nucleus, cells were cultured on round microscope cover glasses in 24-well plates for 1 day. After incubation with PFT-μ (20 μmol/L) for 5 hours, cells were additionally cultured in the presence of TRAIL and Hoechst 33342 (5 μg/mL) for 30 minutes. After fixation and permeabilization with 3% formalin and 1% Triton X, respectively, cells were stainined with anti-NF-κB p65 antibody (Cell Signaling Technology), followed by Alexa Fluor 488–conjugated anti-rabbit immunoglobulin G (IgG) F(ab′)2 fragment (Cell Signaling Technology). Confocal imaging was conducted using an Olympus FV1000-D laser scanning microscope (Olympus).

Colony-forming assay

Cancer cells were seeded in 6-well plates in the presence or absence of TRAIL and/or PFT-μ. Two days later, the medium was replaced with medium that contained no reagent and the culture was continued for an additional 10 days. Thereafter, colonies were counted after fixation with methanol and staining with 0.05% crystal violet.

In vivo xenograft model

BALB nu/nu female mice, purchased from CLEA Japan Inc., were maintained under specific pathogen-free conditions. Experiments were carried out according to the ethical guidelines for animal experimentation of the Shimane University Faculty of Medicine (approval number: IZ24-5). Mice were inoculated in the right flank with 4 × 106 MiaPaca-2 cells with Matrigel (Japan BD Biosciences) at a volume ratio of 1:1. On day 20, the mice were pooled and divided into 4 groups. On days 1, 2, and 3 after grouping, the mice were injected intraperitoneally with PFT-μ (100 μL). As a vehicle control, 100 μL of DMSO was injected. On day 2 after grouping, the mice were injected once intratumorally with TRAIL (50 μL). As a vehicle control, 50 μL of culture medium was injected. Thereafter, tumor size was measured twice weekly.

Statistical analyses

Data were evaluated statistically using an unpaired two-tailed Student t test or ANOVA with Scheffe post hoc test. A P value of less than 0.05 was considered to indicate statistical significance.

Protective role of HSP70 in TRAIL-induced antitumor effects on pancreatic cancer cells

First, we examined the expression of DR4 and DR5 on 4 pancreatic cancer cell lines. Although the expression of DR4 was relatively low on 3 cell lines and was negative on Panc-1, all cell lines were strongly positive for DR5 (Supplementary Fig. S1). We next determined the TRAIL susceptibility of these cell lines (Fig. 1A). MiaPaca-2, BxPC-3, and Panc-1 cells were highly, moderately, and lowly sensitive to TRAIL, respectively, and their susceptibility was dose-dependent. In contrast, AsPC-1 cells were entirely resistant to TRAIL. In subsequent experiments, we focused on MiaPaca-2 and Panc-1 cells.

Figure 1.

Protective role of HSP70 in TRAIL-induced antitumor effects. A, 4 cell lines were cultured with PFT-μ. After 48 hours, cell viability (%) was determined by WST-8 assay. The data are the means of 3 wells. B, after transfection of HSP70 siRNA or control siRNA, HSP70 expression was evaluated by immunoblotting. α-Tubulin was used as the control. C and D, MiaPaca-2 and Panc-1 cells that had been transfected with HSP70 siRNA or control siRNA 3 days previously were cultured with TRAIL. After 48 hours, cell viability (%) and the percentage of Annexin V+ cells were determined by WST-8 assay and flow cytometry, respectively. The data are the means of 3 wells. E, representative flow cytometry result of Panc-1. The numbers represent the percentage of each subset.

Figure 1.

Protective role of HSP70 in TRAIL-induced antitumor effects. A, 4 cell lines were cultured with PFT-μ. After 48 hours, cell viability (%) was determined by WST-8 assay. The data are the means of 3 wells. B, after transfection of HSP70 siRNA or control siRNA, HSP70 expression was evaluated by immunoblotting. α-Tubulin was used as the control. C and D, MiaPaca-2 and Panc-1 cells that had been transfected with HSP70 siRNA or control siRNA 3 days previously were cultured with TRAIL. After 48 hours, cell viability (%) and the percentage of Annexin V+ cells were determined by WST-8 assay and flow cytometry, respectively. The data are the means of 3 wells. E, representative flow cytometry result of Panc-1. The numbers represent the percentage of each subset.

Close modal

To elucidate the roles of HSP70 in TRAIL-induced antitumor effects on human pancreatic cancer cells, we compared the sensitivities of MiaPaca-2 and Panc-1 cells in which HSP70 expression was blocked by RNA interference (Fig. 1B). Knockdown of HSP70 decreased the viability of MiaPaca-2 and Panc-1 cells treated with TRAIL (Fig. 1C). Knockdown of HSP70 markedly increased the percentage of Annexin V+ Panc-1, but not MiaPaca-2, cells upon TRAIL treatment (Fig. 1D). As shown in Fig. 1E, knockdown of HSP70 increased Annexin V+/PI early apoptotic and Annexin V+/PI+ late apoptotic and/or necrotic Panc-1 cells upon TRAIL treatment. A notable finding is that Panc-1 cells, which were less sensitive to TRAIL, became sensitive to low-dose TRAIL after knockdown of HSP70. Overall, these results indicate that HSP70 plays a protective role in TRAIL-induced antitumor effects on human pancreatic cells.

Protective role of autophagy in TRAIL-induced antitumor effects on pancreatic cancer cells

We next tested the possibility that autophagy plays a protective role in the TRAIL resistance of pancreatic cancer cells. LC3 exists in 2 forms: LC3-type I, which is cytosolic, and its proteolytic derivative, LC3-type II, which localizes to the autophagosomal membrane (25). As shown in Fig. 2A, TRAIL treatment increased the expression of LC3-type II in MiaPaca-2 cells, whereas LC3-type II was expressed in Panc-1 cells without TRAIL treatment. We also assessed autophagy by confocal imaging of LC3 foci in GFP-LC3 fusion protein-expressing cancer cells (Fig. 2B). These 2 cell lines were transiently transfected with a plasmid-encoding GFP-LC3, and GFP-LC3 foci were subsequently examined. No GFP-LC3 foci were detected in control GFP/NT–transfected MiaPaca-2 and Panc-1 cells. Although GFP-LC3 foci were detected in GFP-LC3–transfected MiaPaca-2 and Panc-1 cells, even without TRAIL treatment, GFP-LC3 foci seemed to increase in both number and size after TRAIL treatment. To elucidate the role of autophagy in TRAIL-induced antitumor effects, the expression of beclin-1, a molecule required for autophagy, was knocked down by transfection of beclin-1 siRNA (Fig. 2C). Knockdown of beclin-1 decreased the viabilities and increased the percentages of Annexin V+ MiaPaca-2 and Panc-1 cells upon TRAIL treatment (Fig. 2D and E). Collectively, these results indicate that, similar to HSP70, autophagy plays a protective role in TRAIL-induced antitumor effects on pancreatic cancer cells.

Figure 2.

Protective role of autophagy in the antitumor effect of TRAIL on human pancreatic cancer cell lines. A, MiaPaca-2 and Panc-1 cells were cultured with TRAIL at 25 and 100 ng/mL, respectively, for 24 hours, and LC3 expression was evaluated by immunoblotting. β-Actin was used as the control. B, MiaPaca-2 and Panc-1 cells that had been pretransfected with a plasmid-encoding GFP-LC3 or GFP/NT were cultured with the indicated TRAIL concentration. After 18 hours, the expression of LC3 (green) and nuclear staining with Hoechst 33342 (blue) were visualized by confocal microscopy. Scale bar, 10 μm. C, MiaPaca-2 and Panc-1 cells that had been transfected with beclin-1 siRNA or control siRNA 3 days previously were evaluated by immunoblotting. β-Actin was used as the control, Bec-1, beclin-1. D, MiaPaca-2 and Panc-1 cells that had been transfected with beclin-1 siRNA or control siRNA 3 days previously were cultured with TRAIL and their viabilities were assessed by WST-8 assay. The data are the means of 3 wells. E, flow cytometry analysis was conducted after staining with fluorescein isothiocyanate (FITC)–conjugated Annexin V and PI. The data are the means of 3 wells.

Figure 2.

Protective role of autophagy in the antitumor effect of TRAIL on human pancreatic cancer cell lines. A, MiaPaca-2 and Panc-1 cells were cultured with TRAIL at 25 and 100 ng/mL, respectively, for 24 hours, and LC3 expression was evaluated by immunoblotting. β-Actin was used as the control. B, MiaPaca-2 and Panc-1 cells that had been pretransfected with a plasmid-encoding GFP-LC3 or GFP/NT were cultured with the indicated TRAIL concentration. After 18 hours, the expression of LC3 (green) and nuclear staining with Hoechst 33342 (blue) were visualized by confocal microscopy. Scale bar, 10 μm. C, MiaPaca-2 and Panc-1 cells that had been transfected with beclin-1 siRNA or control siRNA 3 days previously were evaluated by immunoblotting. β-Actin was used as the control, Bec-1, beclin-1. D, MiaPaca-2 and Panc-1 cells that had been transfected with beclin-1 siRNA or control siRNA 3 days previously were cultured with TRAIL and their viabilities were assessed by WST-8 assay. The data are the means of 3 wells. E, flow cytometry analysis was conducted after staining with fluorescein isothiocyanate (FITC)–conjugated Annexin V and PI. The data are the means of 3 wells.

Close modal

Antitumor effects on pancreatic cancer cells of the combination of TRAIL and PFT-μ

Because both HSP70 and autophagy were suggested to function protectively upon TRAIL treatment of pancreatic cancer cells, we next determined whether TRAIL-induced antitumor effects were enhanced by PFT-μ, which inhibits both the functions of HSP70 and autophagy (24), as described in the Introduction. Figure 3A shows the structure of PFT-μ. As shown in Fig. 3B, when administered alone, PFT-μ dose-dependently decreased the viability of all 4 pancreatic cancer cell lines. When suboptimal doses of TRAIL and PFT-μ were combined, additive effects were evident in 3 cell lines other than AsPC-1 (Fig. 3C). Combined treatment with suboptimal doses of TRAIL and PFT-μ significantly decreased the colony-forming ability of MiaPaca-2 and Panc-1 cells compared with those treated with either agent alone (Fig. 3D). Because the continuous presence of PFT-μ for 12 days drastically inhibited colony formation, we cultured cancer cells for 2 days with TRAIL and/or PFT-μ and subsequently for 10 days without reagents.

Figure 3.

PFT-μ enhances the antitumor effects of TRAIL on pancreatic cancer cell lines. A, the structure of PFT-μ. B, 4 pancreatic cancer cell lines were cultured with the indicated PFT-μ concentrations, and cell viability (%) was determined by the WST-8 assay after 48 hours. Results are mean ± SD of 3 wells. C, 4 cancer cell lines were cultured with TRAIL and/or PFT-μ. After 48 hours, cell viability (%) was determined by WST-8 assay. *, P < 0.05; **, P < 0.01 compared with the other 3 groups. D, MiaPaca-2 and Panc-1 cells were cultured with TRAIL and/or PFT-μ for 2 days and without TRAIL and PFT-μ for an additional 10 days. The results are mean ± SD of 3 wells. *, P < 0.05; **, P < 0.01.

Figure 3.

PFT-μ enhances the antitumor effects of TRAIL on pancreatic cancer cell lines. A, the structure of PFT-μ. B, 4 pancreatic cancer cell lines were cultured with the indicated PFT-μ concentrations, and cell viability (%) was determined by the WST-8 assay after 48 hours. Results are mean ± SD of 3 wells. C, 4 cancer cell lines were cultured with TRAIL and/or PFT-μ. After 48 hours, cell viability (%) was determined by WST-8 assay. *, P < 0.05; **, P < 0.01 compared with the other 3 groups. D, MiaPaca-2 and Panc-1 cells were cultured with TRAIL and/or PFT-μ for 2 days and without TRAIL and PFT-μ for an additional 10 days. The results are mean ± SD of 3 wells. *, P < 0.05; **, P < 0.01.

Close modal

PFT-μ induces cell death and growth arrest of pancreatic cancer cells

In the studies described earlier, we examined the antitumor effects on pancreatic cancer cells by measuring viability 2 days after the administration of TRAIL and/or PFT-μ. However, such effects on viability may reflect alterations in cell death and/or growth. Therefore, we next examined the underlying mechanism of action of PFT-μ in detail. Although suboptimal doses of TRAIL slightly increased Annexin V+ MiaPaca-2 and Panc-1 cells, PFT-μ moderately increased the percentages of Annexin V+ cells (Fig. 4A). Combining PFT-μ with TRAIL further increased the number of Annexin V+ cells. Because PFT-μ was reported to induce caspase-independent apoptosis (24), we attempted to confirm the result in our experimental systems. TRAIL-induced increase of Annexin V+ MiaPaca-2 and Panc-1 cells was completely inhibited by the pan-caspase inhibitor z-VAD, whereas z-VAD rescued in part PFT-μ–induced cell death (Fig. 4B). This result indicates that PFT-μ increased Annexin V+ cells in both caspase-dependent and -independent manners. We next determined whether growth arrest was involved in the antitumor effects of PFT-μ by evaluating BrdUrd uptake and 7AAD staining. PFT-μ significantly decreased the percentages of BrdUrd+ S-phase MiaPaca-2 and Panc-1 cells (Fig. 4C and D) and increased the apoptotic sub-G1 fraction in MiaPaca-2 cells. We also examined the expression of proliferation-related proteins in MiaPca-2 and Panc-1 cells and found that PFT-μ decreased cyclin D1 expression in MiaPaca-2 cells but not Panc-1 cells. Collectively, these results indicate that PFT-μ induces 2 types of antitumor responses in pancreatic cancer cells: cell death, both caspase-dependent and -independent, and cell-growth arrest.

Figure 4.

Induction of cell death and growth arrest in pancreatic cancer cells by PFT-μ. A, MiaPaca-2 and Panc-1 cells were cultured with TRAIL (20 and 100 ng/mL, respectively) and PFT-μ (15 μmol/L). After 48 hours, flow cytometry analysis was conducted after staining with FITC-conjugated Annexin V and PI. A representative result is shown. The numbers represent the percentages of each subset. B, MiaPaca-2 and Panc-1 cells were cultured with TRAIL (20 and 100 ng/mL, respectively) and PFT-μ (15 μmol/L) in the presence of z-VAD or DMSO. After 48 hours, flow cytometry analysis was conducted after staining with FITC-conjugated Annexin V and PI. The results are shown as the mean ± SD of 3 wells. *, P < 0.05; **, P < 0.01. C, MiaPaca-2 and Panc-1 cells were cultured with PFT-μ (10 μmol/L) for 2 days. During the last 90 minutes for MiaPaca-2 cells and 5 hours for Panc-1 cells, cells were cultured with BrdUrd (10 μmol/L). Harvested cells were stained with 7AAD, and flow cytometry analysis was conducted. Numbers represent the percentages of each subset. D, the results are shown as the mean ± SD of 3 wells. *, P < 0.05; **, P < 0.01. E, MiaPaca-2 and Panc-1 cells were cultured with PFT-μ (10 μmol/L) for 2 days, and c-Myc and cyclin D1 protein levels were determined by immunoblotting. β-Actin was used as the control.

Figure 4.

Induction of cell death and growth arrest in pancreatic cancer cells by PFT-μ. A, MiaPaca-2 and Panc-1 cells were cultured with TRAIL (20 and 100 ng/mL, respectively) and PFT-μ (15 μmol/L). After 48 hours, flow cytometry analysis was conducted after staining with FITC-conjugated Annexin V and PI. A representative result is shown. The numbers represent the percentages of each subset. B, MiaPaca-2 and Panc-1 cells were cultured with TRAIL (20 and 100 ng/mL, respectively) and PFT-μ (15 μmol/L) in the presence of z-VAD or DMSO. After 48 hours, flow cytometry analysis was conducted after staining with FITC-conjugated Annexin V and PI. The results are shown as the mean ± SD of 3 wells. *, P < 0.05; **, P < 0.01. C, MiaPaca-2 and Panc-1 cells were cultured with PFT-μ (10 μmol/L) for 2 days. During the last 90 minutes for MiaPaca-2 cells and 5 hours for Panc-1 cells, cells were cultured with BrdUrd (10 μmol/L). Harvested cells were stained with 7AAD, and flow cytometry analysis was conducted. Numbers represent the percentages of each subset. D, the results are shown as the mean ± SD of 3 wells. *, P < 0.05; **, P < 0.01. E, MiaPaca-2 and Panc-1 cells were cultured with PFT-μ (10 μmol/L) for 2 days, and c-Myc and cyclin D1 protein levels were determined by immunoblotting. β-Actin was used as the control.

Close modal

PFT-μ inhibits the autophagy-lysosomal system and TRAIL-associated NF-κB activation in pancreatic cancer cells

We next attempted to elucidate the mechanisms by which PFT-μ enhanced TRAIL-induced antitumor effects. To evaluate the influence of PFT-μ on the autophagy system, we determined its effect on the degradation of pro-cathepsin L (Fig. 5A). PFT-μ increased the expression of LC3 type II. However, this increase was not the result of enhanced autophagy but rather inhibition of the autophagy-lysosomal system, because degradation of pro-cathepsin L to cathepsin L was clearly inhibited, as reported previously (24). We also examined the effect of PFT-μ on NF-κB signaling in TRAIL-treated cancer cells, because activation of NF-κB is a major mechanism of TRAIL resistance of cancer cells (6, 8). As expected, TRAIL significantly decreased the level of IκBα, indicating activation of the NF-κB pathway. Although the IκBα level was decreased in cells treated with PFT-μ alone, PFT-μ restored IκBα expression in cells treated with TRAIL (Fig. 5B). TRAIL and PFT-μ had no marked effect on HSP70 expression in cancer cells. We next directly confirmed that the combination treatment inhibited the NF-κB pathway using confocal imaging. Although the treatment with either of TRAIL or PFT-μ induced NF-κB translocation to the nucleus, the combination treatment decreased the expression of NF-κB in the nucleus (Fig. 5C). These results suggest that PFT-μ can inhibit the autophagy–lysosomal system and antagonize TRAIL-associated NF-κB activation in pancreatic cancer cells.

Figure 5.

PFT-μ inhibits autophagy and TRAIL-associated NF-κB activation. A, MiaPaca-2 and Panc-1 cells were cultured with PFT-μ (20 μmol/L), and LC3, beclin-1, pro-cathepsin L, and cathepsin L protein levels were determined by immunoblotting. β-Actin was used as the control. B, after culture with PFT-μ (20 μmol/L) for 5 hours, MiaPaca-2 and Panc-1 cells were additionally cultured with TRAIL (25 ng and 100 ng/mL, respectively) for 30 minutes. Thereafter, IκBα and HSP70 expressions were determined by immunoblotting. β-Actin and α-tubulin were used as controls. C, after culture with PFT-μ (20 μmol/L) for 5 hours, MiaPaca-2 and Panc-1 cells were additionally cultured with TRAIL (50 ng and 200 ng/mL, respectively) for 30 minutes. The expression of NF-κB p65 (green) and nuclear staining with Hoechst 33342 (blue) were visualized by confocal microscopy. Scale bar, 20 μm.

Figure 5.

PFT-μ inhibits autophagy and TRAIL-associated NF-κB activation. A, MiaPaca-2 and Panc-1 cells were cultured with PFT-μ (20 μmol/L), and LC3, beclin-1, pro-cathepsin L, and cathepsin L protein levels were determined by immunoblotting. β-Actin was used as the control. B, after culture with PFT-μ (20 μmol/L) for 5 hours, MiaPaca-2 and Panc-1 cells were additionally cultured with TRAIL (25 ng and 100 ng/mL, respectively) for 30 minutes. Thereafter, IκBα and HSP70 expressions were determined by immunoblotting. β-Actin and α-tubulin were used as controls. C, after culture with PFT-μ (20 μmol/L) for 5 hours, MiaPaca-2 and Panc-1 cells were additionally cultured with TRAIL (50 ng and 200 ng/mL, respectively) for 30 minutes. The expression of NF-κB p65 (green) and nuclear staining with Hoechst 33342 (blue) were visualized by confocal microscopy. Scale bar, 20 μm.

Close modal

In vivo antitumor effect of TRAIL plus PFT-μ combination therapy in a xenograft mouse model

Finally, we evaluated whether combination therapy with TRAIL plus PFT-μ exerted an antitumor effect against established human pancreatic cancer in a xenograft mouse model. Nude mice were inoculated with MiaPaca-2 cells and were grouped when tumor diameters reached 8 to 9 mm. After grouping, PFT-μ (25 mg/kg) was injected intraperitoneally on days 1, 2, and 3 and TRAIL was injected locally on day 2. No change in body weight resulted, suggesting no severe adverse event (data not shown). Although the systemic administration of PFT-μ had no antitumor effect and local injections of TRAIL decreased tumor growth moderately, but not significantly, combination therapy with TRAIL plus PFT-μ significantly suppressed tumor growth (Fig. 6A and B).

Figure 6.

In vivo antitumor effects of TRAIL and PFT-μ in a xenograft mouse model. A, BALB nu/nu mice were subcutaneously inoculated in the right flank with MiaPaca-2 cells (4 × 106) with Matrigel (total volume, 200 μL). On day 20, the mice were pooled and divided into 4 groups. On days 1, 2, and 3 after grouping, the mice were intraperitoneally injected with PFT-μ (25 mg/kg; 100 μL). As a vehicle control, an identical volume of DMSO was injected. On day 2 after grouping, the mice were injected once intratumorally with 1.5 μg of TRAIL (50 μL). As a vehicle control, an identical volume of complete medium was injected. Thereafter, tumor size was measured twice weekly. Arrowheads (PFT-μ) and arrows (TRAIL) represent the day of treatment. Each group contained 7 or 8 mice. B, tumor size on day 13 after grouping. The results are shown as the mean ± SD of 7 or 8 mice. *, P < 0.05 (ANOVA with Scheffe post hoc test). n.s., not significant.

Figure 6.

In vivo antitumor effects of TRAIL and PFT-μ in a xenograft mouse model. A, BALB nu/nu mice were subcutaneously inoculated in the right flank with MiaPaca-2 cells (4 × 106) with Matrigel (total volume, 200 μL). On day 20, the mice were pooled and divided into 4 groups. On days 1, 2, and 3 after grouping, the mice were intraperitoneally injected with PFT-μ (25 mg/kg; 100 μL). As a vehicle control, an identical volume of DMSO was injected. On day 2 after grouping, the mice were injected once intratumorally with 1.5 μg of TRAIL (50 μL). As a vehicle control, an identical volume of complete medium was injected. Thereafter, tumor size was measured twice weekly. Arrowheads (PFT-μ) and arrows (TRAIL) represent the day of treatment. Each group contained 7 or 8 mice. B, tumor size on day 13 after grouping. The results are shown as the mean ± SD of 7 or 8 mice. *, P < 0.05 (ANOVA with Scheffe post hoc test). n.s., not significant.

Close modal

Because pancreatic cancer is highly resistant to conventional anticancer therapies and is associated with a very poor prognosis (26), new treatment modalities to enhance the efficacy of current treatments are required. In this study, we investigated the possibility that PFT-μ, a small-molecule HSP70 inhibitor that has the ability to alter autophagy (24), could enhance TRAIL-induced antitumor effects on human pancreatic cancer cells. We found that both HSP70 and autophagy are, at least in part, responsible for the TRAIL resistance of cancer cells and that PFT-μ enhances TRAIL-induced antitumor effects on human pancreatic cancer cells.

HSP70 is a potent heat-inducible survival protein that confers cytoprotection against various death-inducing stimuli and increases tumorigenicity (27–29). It has been suggested to be a promising target in cancer treatment (30). In this study, knockdown of HSP70 significantly decreased cell viability and increased the percentage of Annexin V+ pancreatic cancer cells after TRAIL treatment (Fig. 1). The increase of Annexin V+ cells in response to TRAIL was stronger in the less TRAIL-sensitive Panc-1 cells than in highly TRAIL-sensitive MiaPaca-2 cells. Although only one report suggests that TRAIL-induced apoptosis in cancer cells is enhanced by HSP70 (31), many reports have shown that HSP70 inhibits TNF-α- and Fas-mediated apoptosis in cancer cells (32–34). In this study, we have shown that siRNA knockdown of HSP70 increased Annexin V+ Panc-1 cells after TRAIL treatment (Fig. 1). To our knowledge, this is the first report that HSP70 contributes to the TRAIL resistance of human pancreatic cancer cells. Another intriguing finding is that autophagy is protective in TRAIL-treated pancreatic cancer cells. This result is compatible with other reports (20). Autophagy has received much attention in various cell biology fields (16, 17). Despite the reports of autophagic cell death (18, 35, 36) and autophagy-dependent antitumor immune response (37), the fundamental role of autophagy is thought to be cytoprotection under starvation and stress conditions (16). Many reports suggest that autophagy functions cytoprotectively in cancer cells (19, 20, 36, 38). In line with this, we previously reported that autophagy protects against apoptosis in human prostate and breast cancer cells after treatment with an innate adjuvant receptor ligand (21, 22). Therefore, in this study, we investigated the participation of autophagy in the TRAIL resistance of pancreatic cancer cells. Autophagy was constitutively induced in MiaPaca-2 and Panc-1 cells, and TRAIL treatment apparently augmented autophagy in MiaPaca-2 cells. We also showed that autophagy inhibition by knockdown of beclin-1 increased the susceptibility of pancreatic cancer cells to TRAIL (Fig. 2). Our findings about the protective roles of HSP70 and autophagy in TRAIL treatment encouraged us to use the HSP70 and autophagy inhibitor PFT-μ to enhance TRAIL-induced antitumor effects on pancreatic cancer cells.

Death receptor–mediated TRAIL signaling is involved in activation of the NF-κB pathway, leading to TRAIL resistance (6, 8). Indeed, the proteasome inhibitor bortezomib enhances TRAIL-induced antitumor effects by inhibiting NF-κB (9–11). Interestingly, PFT-μ decreases IκBα degradation by inhibiting the autophagy–lysosomal system (24). PFT-μ also inhibits the proteasome system (39). We then determined whether PFT-μ inhibited TRAIL-induced activation of the NF-κB pathway. As expected, TRAIL decreased IκBα expression in cancer cells and cotreatment with PFT-μ inhibited IκBα degradation. In addition, confocal imaging revealed that the combination treatment inhibited NF-κB translocation to the nucleus. This may be the third mechanism by which PFT-μ augmented TRAIL-induced antitumor effects on pancreatic cancer cells.

How does HSP70 inhibit TRAIL-induced cell death? HSP70 interacts with Apaf-1 to prevent caspase activation (40, 41). Interestingly, HSP70 has been reported to localize to the membranes of lysosomes, promote cancer cell viability, and inhibit TNF-induced cell death by inhibiting lysosomal membrane permeabilization (42). Thus, HSP70 enhances survival by stabilizing the lysosomes in cancer cells. Because PFT-μ binds HSP70, inhibiting the autophagy–lysosomal system, it is possible that PFT-μ inhibits HSP70-induced stabilization of lysosomal membrane permeabilization, resulting in increased cell death. We plan to investigate this possibility.

HSP70 can bind to apoptosis-inducing factor (AIF), which induces caspase-independent apoptosis by translocation into the nucleus (43). As PFT-μ induces cell death in both caspase-dependent and -independent manners in cancer cells (Fig. 4B), we evaluated the role of AIF in PFT-μ–induced caspase-independent cell death. However, RNA interference of AIF had no effect on PFT-μ–induced cell death of pancreatic cancer cells (data not shown); thus, AIF likely does not participate in PFT-μ–induced cell death.

There are several possible explanations about how autophagy protects cancer cells from TRAIL-induced cell death. First, autophagy degrades caspases. Indeed, autophagy can degrade active caspase-8 (44), which is required for extrinsic signal-mediated caspase-dependent apoptosis. Second, although we cannot exclude the possibility of participation of the proteasome system, the autophagy–lysosomal system degrades IκBα, resulting in activation of the NF-κB pathway. Several reports have indicated that activation of this pathway can cause cancer cells to become resistant to TRAIL (6, 8). Interestingly, a recent report suggests that the mitochondrial tumor suppressor ARF interacts with HSP70 and that PFT-μ can reduce the ability of ARF to induce autophagy by selectively interacting with HSP70 and blocking ARF trafficking to mitochondria (45). ARF thus seems to play a crucial role in the altered autophagy induced by PFT-μ. We plan to elucidate this mechanism in more detail.

Besides cell death, PFT-μ induced growth arrest of pancreatic cancer cells. PFT-μ treatment significantly decreased the S-phase fraction in 2 cancer cell lines (Fig. 4C). The decreased cyclin D1 expression might, at least in part, account for PFT-μ–induced cell growth arrest. We conducted a long-term (12-day) colony-formation assay. A suboptimal dose of PFT-μ significantly decreased the colony-forming ability of MiaPaca-2 cells. These data support the hypothesis that PFT-μ arrests cell growth.

Among the 4 pancreatic cancer cell lines, only AsPC-1 was resistant to TRAIL and PFT-μ showed no sensitizing effects on AsPC-1. Thus, some TRAIL susceptibility seems to be necessary for the sensitizing effects of PFT-μ. In addition, only AsPC-1 was entirely TRAIL-resistant even though there was no difference in DR4 and DR5 expression among the 4 cell lines (Supplementary Fig. S1). Although we have not elucidated the underlying mechanisms, AsPC-1 may have been preferentially positive for nonfunctional decoy receptors or antiapoptotic molecules including bcl-2, bcl-xL, and c-FLIP, as reported previously (46, 47).

In conclusion, we show here that PFT-μ has the potential to induce 2 types of antitumor response; that is, cell death and cell growth arrest, in human pancreatic cancer cells and that it enhances TRAIL-induced antitumor effects both in vitro and in vivo. These results suggest that PFT-μ shows promise for use as a therapy intended to enhance the antitumor effects of TRAIL or agonistic antibodies against death receptors.

No potential conflicts of interest were disclosed.

Conception and design: H. Monma, M. Harada

Development of methodology: S. Okano

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): H. Monma, N. Harashima, M. Harada

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): H. Monma, M. Harada

Writing, review, and/or revision of the manuscript: H. Monma, Y. Tajima, M. Harada

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): N. Harashima, T. Inao

Study supervision: M. Harada

The authors thank Ms. Tamami Moritani for her technical assistance.

This study was supported in part by Grants-in aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology, Japan (no. 18591449 to M. Harada and no. 23701074 to N. Harashima), and from Shimane University “S-TAKUMI Medical Nanotechnology” Project (M. Harada).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ashkenazi
A
,
Dixit
VM
. 
Apoptosis control by death and decoy receptors
.
Curr Opin Cell Biol
1999
;
11
:
255
60
.
2.
Almasan
A
,
Ashkenazi
A
. 
Apo2L/TRAIL: apoptosis signaling, biology, and potential for cancer therapy
.
Cytokine Growth Factor Rev
2003
;
14
:
337
48
.
3.
Herbst
RS
,
Eckhardt
G
,
Kurzrock
R
,
Ebbinghaus
S
,
O'Dwyer
PJ
,
Gordon
MS
, et al
Phase I dose-escalation study of recombinant human Apo2L/TRAIL, a dual proapoptotic receptor agonist, in patients with advanced cance
r.
J Clin Oncol
2010
;
28
:
2839
46
.
4.
Soria
JC
,
Márk
Z
,
Zatloukal
P
,
Szima
B
,
Albert
I
,
Juhász
E
, et al
Randomized phase II study of dulanermin in comination with paclitaxel, carboplatin, and bevacizuan in advanced non–small-cell lung cancer
.
J Clin Oncol
2011
;
29
:
4442
51
.
5.
Newsom-Davis
T
,
Prieske
S
,
Walczak
H
. 
Is TRAIL the holy grail of cancer therapy?
Apoptosis
2009
;
14
:
607
23
.
6.
Trauzold
A
,
Wermann
H
,
Arlt
A
,
Schutze
S
,
Schafer
H
,
Oestern
S
, et al
CD95 and TRAIL receptor-mediated activation of protein kinase C and NF–kappaB contributes to apoptosis resistance in ductal pancreatic adenocarcinoma cells
.
Oncogene
2001
;
20
:
4258
69
.
7.
Siegmund
D
,
Klose
S
,
Zhou
D
,
Baumann
B
,
Roder
C
,
Kalthoff
H
, et al
Role of caspases in CD95L- and TRAIL-induced non-apoptotic signalling in pancreatic tumour cells
.
Cell Signal
2007
;
19
:
1172
84
.
8.
Khanbolooki
S
,
Nawrocki
ST
,
Arumugam
T
,
Andtbacka
R
,
Pino
MS
,
Kurzrock
R
, et al
Nuclear factor-KB maintains TRAIL resistance in human pancreatic cancer cells
.
Mol Cancer Ther
2006
;
5
:
2251
60
.
9.
Brooks
AD
,
Jacobsen
KM
,
Li
W
,
Shanker
A
,
Sayers
TJ
. 
Bortezomib sensitizes human renal cell carcinoma to TRAIL apoptosis through increased activation of caspase-8 in the death-inducing signaling complex
.
Mol Cancer Res
2010
;
8
:
729
38
.
10.
Seki
N
,
Toh
U
,
Sayers
TJ
,
Fuji
T
,
Miyagi
M
,
Akagi
Y
, et al
Bortzomib sensitizes human esophageal squamous cell carcinoma cells to TRAIL-mediated apoptosis via activation of both extrinsic and intrinsic apoptosis pathways
.
Mol Cancer Ther
2010
;
9
:
1842
51
.
11.
Johnson
T
,
Stone
K
,
Nikrad
M
,
Yeh
T
,
Zong
WX
,
Thompson
CB
, et al
The proteasome inhibitor PS-341 overcomes TRAIL resistance in Bax and caspase 9-negative or Bcl-xL overexpressing cells
.
Oncogene
2003
;
22
:
4953
63
.
12.
Samali
A
,
Cotter
TG
. 
Heat shock proteins increase resistance to apoptosis
.
Exp Cell Res
1996
;
223
:
163
70
.
13.
Mayer
MP
,
Bukau
B
. 
Hsp70 chaperones: cellular functions and molecular mechanism
.
Cell Mol Life Sci
2005
;
62
:
670
84
.
14.
Daugaard
M
,
Rohde
M
,
Jäättelä
M
. 
The heat shock protein 70 family: highly homologous proteins with overlapping and distinct functions
.
FEBS Lett
2007
;
581
:
3702
10
.
15.
Gress
TM
,
Muller-Pillasch
F
,
Weber
C
,
Lerch
MM
,
Friess
H
,
Buchler
M
, et al
Differential expression of heat shock proteins in pancreatic carcinoma
.
Cancer Res
1994
;
54
:
547
51
.
16.
Levine
B
,
Kroemer
G
. 
Autophagy in the pathogenesis of diseases
.
Cell
2008
;
132
:
27
42
.
17.
Gewirtz
DA
. 
Autophagy, senescence and tumor dormancy in cancer therapy
.
Authophagy
2009
;
5
:
1232
4
.
18.
Mujumdar
N
,
Mackenzie
TN
,
Dudeja
V
,
Chugh
R
,
Antonoff
MB
,
Borja-Cacho
D
, et al
Triptolide induces cell death in pancreatic cancer cells by apoptotic and autophagic pathways
.
Gastroenterology
2010
;
139
:
598
608
.
19.
Amaravadi
RK
,
Lippincott-Schwartz
J
,
Yin
XM
,
Weiss
WA
,
Takebe
N
,
Timmer
W
, et al
Principles and current strategies for targeting autophagy for cancer treatment
.
Clin Cancer Res
2011
;
17
:
654
66
.
20.
Han
J
,
Hou
W
,
Goldstein
LA
,
Lu
C
,
Stolz
DB
,
Yin
XM
, et al
Involvement of protective autophagy in TRAIL resistance of apoptosis-defective tumor cells
.
J Biol Chem
2008
;
283
:
19665
77
.
21.
Harashima
N
,
Inao
T
,
Imamura
R
,
Okano
S
,
Suda
T
,
Harada
M
. 
Roles of the PI3K/Akt pathway and autophagy in TLR3 signaling-induced apoptosis and growth arrest of human prostate cancer cells
.
Cancer Immunol Immunother
2012
;
61
:
667
76
.
22.
Inao
T
,
Harashima
N
,
Monma
H
,
Okano
S
,
Itakura
M
,
Tanaka
T
, et al
Antitumor effects of cytoplasmic delivery of an innate adjuvant receptor ligand, poly(I:C), on human breast cancer
.
Breast Cancer Res Treat
2012
;
134
:
89
100
.
23.
Strom
E
,
Sathe
S
,
Komarov
PG
,
Chernova
OB
,
Pavlovska
I
,
Shyshynova
I
, et al
Small-moleculae inhibitor of p53 binding to mitochondria protects mice from gamma radiation
.
Nat Chem Biol
2006
;
2
:
474
9
.
24.
Leu
JI
,
Pimkina
J
,
Frank
A
,
Murphy
ME
,
George
DL
. 
A small molecule inhibitor of inducible heat shock protein 70
.
Mol Cell
2009
;
36
:
15
27
.
25.
Kabeya
Y
,
Mizushima
N
,
Ueno
T
,
Yamamoto
A
,
Kirisako
T
,
Noda
T
, et al
LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing
.
EMBO J
2000
;
19
:
5720
8
.
26.
Jemal
A
,
Siegel
R
,
Ward
E
,
Hao
Y
,
Xu
J
,
Thun
MJ
. 
Cancer statistics 2009
.
CA Cancer J Clin
2009
;
59
:
225
49
.
27.
Rohde
M
,
Daugaard
M
,
Jensen
MH
,
Helin
K
,
Nylandsted
J
,
Jäättelä
M
. 
Members of the heat-shock protein 70 family promote cancer cell growth by distinct mechanisms
.
Genes Dev
2005
;
19
:
570
82
.
28.
Aghdassi
A
,
Phillips
P
,
Dudeja
V
,
Dhaulakhandi
D
,
Sharif
R
,
Dawra
R
, et al
Heat shock protein 70 increases tumorigenicity and inhibits apoptosis in pancreatic adenocarcinoma
.
Cancer Res
2007
;
67
:
616
25
.
29.
Dudeja
V
,
Mujumdar
N
,
Phillips
P
,
Chugh
R
,
Borja-Cacho
D
,
Dawara
RK
, et al
Heat shock protein 70 inhibits apoptosis in cancer cells through simultaneous and independent mechanisms
.
Gastroenterology
2009
;
136
:
1772
82
.
30.
Galluzzi
L
,
Giordanetto
F
,
Kroemer
G
. 
Targeting HSP70 for cancer therapy
.
Mol Cell
2009
;
36
:
176
7
.
31.
Clemons
NJ
,
Anderson
RL
. 
TRAIL-induced apoptosis is enhaced by heat shock protein 70 expression
.
Cell Stress Chaperones
2006
;
11
:
343
55
.
32.
Buzzard
KA
,
Giaccia
AJ
,
Killender
M
,
Anderson
RL
. 
Heat showck protein 72 modulates pathways of stress-induced apoptosis
.
J Biol Chem
1998
;
273
:
17147
53
.
33.
Clemons
NJ
,
Buzzard
K
,
Steel
R
,
Anderson
RL
. 
Hsp72 inhibits Fas-mediated apoptosis upstream of the mitochondria in type II cells
.
J Biol Chem
2005
;
280
:
9005
12
.
34.
Jäättelä
M
,
Wissing
D
,
Bauer
PA
,
Li
GC
. 
Major heat shock protein hsp70 protects tumor cells from tumor necrosis factor cytotoxicity
.
EMBO J
1992
;
11
:
3507
12
.
35.
Bonapace
L
,
Bornhauser
BC
,
Schmitz
M
,
Cario
G
,
Ziegler
U
,
Niggli
FK
, et al
Induction of autophagy-dependent necrosis is required for childhood acute lymphoblastic leukemia cells to overcome glucocorticoid resistance
.
J Clin Invest
2010
;
120
:
1310
23
.
36.
Jing
K
,
Song
KS
,
Shin
S
,
Kim
N
,
Jeong
S
,
Oh
HR
, et al
Docosahexaenoic acid induces autophagy through p53/AMPK/mTOR signaling and promotes apoptosis in human cancer cells harboring wild-type p53
.
Autophagy
2001
;
7
:
1348
58
.
37.
Michaud
M
,
Martins
I
,
Sukkurwala
AQ
,
Adjemian
S
,
Ma
Y
,
Pellegatti
P
, et al
Autophagy-dependent anticancer immune response induced by chemotherapeutic agents in mice
.
Science
2011
;
334
:
1573
7
.
38.
Buchser
WJ
,
Laskow
TC
,
Pavlik
PJ
,
Lin
HM
,
Lotze
MT
. 
Cell-mediated autophagy promotes cancer cell survival
.
Cancer Res
2012
;
72
:
2970
9
.
39.
Leu
JI
,
Pimkina
J
,
Pandey
P
,
Murphy
ME
,
George
DL
. 
HSP70 inhibition by the small-molecule 2-phenylethynesulfonamide impairs protein clearance pathways in tumor cells
.
Mol Cancer Res
2011
;
9
:
936
47
.
40.
Saleh
A
,
Srinivasula
SM
,
Balkir
L
,
Robbins
PD
,
Alnemri
ES
. 
Negative regulation of the Apaf-1 apoptosome by Hsp70
.
Nat Cell Biol
2000
;
2
:
476
83
.
41.
Beer
HM
,
Wolf
BB
,
Cain
K
,
Mosser
DD
,
Mahboubi
A
,
Kuwana
T
, et al
Heat-shock protein 70 inhibits apoptosis by preventing recruitment of procaspase-9 to the Apaf-1 apoptosome
.
Nat Cell Biol
2000
;
2
:
469
75
.
42.
Nylandsted
J
,
Gyrd-Hansen
M
,
Danielewicz
A
,
Fehrenbacher
N
,
Lademann
U
,
Høyer-Hansen
M
, et al
Heat showck protein 70 promotes cell survival by inhibiting lysosomal membrane permeabilization
.
J Exp Med
2004
;
200
:
425
35
.
43.
Cabde
C
,
Vahsen
N
,
arrido
C
,
Kroemer
G
. 
Apoptosis-inducing factor (AIF): caspase-independent after all
.
Cell Death Differ
2004
;
11
:
591
5
.
44.
Hou
W
,
Han
J
,
Lu
C
,
Goldstein
LA
,
Rabinowich
H
. 
Autophagic degradation of active caspase-8: a crosstalk mechanism between autophagy and apoptosis
.
Autophagy
2010
;
6
:
891
900
.
45.
Pimkina
JS
,
Murphy
ME
. 
Interaction of the ARF tumor suppressor with cytosolic HSP70 contributes to its autophagy function
.
Cancer Biol Ther
2011
;
12
:
503
9
.
46.
Ibrahim
SM
,
Ringel
J
,
Schmidt
C
,
Ringel
B
,
Müller
P
,
Koczan
D
, et al
Pancreatic adenocarcinoma cell lines show variable susceptibility to TRAIL-mediated cell death
.
Pancreas
2001
;
23
:
72
9
.
47.
Wang
P
,
Zhang
J
,
Bellail
A
,
Jiang
W
,
Hugh
J
,
Kneteman
NM
, et al
Inhibition of RIP and c-FLIP enhances TRAIL-induced apoptosis in pancreatic cancer cells
.
Cell Signal
2007
;
19
:
2237
46
.