We have recently shown that proteasome inhibitor PS-341 induces apoptosis in drug-resistant multiple myeloma (MM) cells, inhibits binding of MM cells in the bone marrow microenvironment, and inhibits cytokines mediating MM cell growth, survival, drug resistance, and migration in vitro. PS-341 also inhibits human MM cell growth and prolongs survival in a SCID mouse model. Importantly, PS-341 has achieved remarkable clinical responses in patients with refractory relapsed MM. We here demonstrate molecular mechanisms whereby PS-341 mediates anti-MM activity by inducing p53 and MDM2 protein expression; inducing the phosphorylation (Ser15) of p53 protein; activating c-Jun NH2-terminal kinase (JNK), caspase-8, and caspase-3; and cleaving the DNA protein kinase catalytic subunit, ATM, and MDM2. Inhibition of JNK activity abrogates PS-341–induced MM cell death. These studies identify molecular targets of PS-341 and provide the rationale for the development of second-generation, more targeted therapies.

The ubiquitin-proteasome pathway (UPP) is a proteolytic system in the cytosol and nucleus that regulates cyclins and cyclin-dependent kinase inhibitor cell-cycle regulatory proteins and thereby regulates cell-cycle progression.1 UPP also has a critical role in the selective removal of mutant, damaged, and misfolded proteins. Proteasome inhibitors have recently demonstrated promise as potential novel anticancer therapies2 because they induce the apoptosis of tumor cells in vitro, despite the accumulation of p21 and p273,4 and irrespective of the p53 wild-type or mutant status5,6 in tumor cells. Specifically, PS-341 (pyrazylcarbonyl-Phe-Leu-boronate) is representative of a class of peptide boronate proteasome inhibitors that inhibit 26S proteasome activity.1 This agent induces marked in vivo antitumor activity against human prostate cancer,2,7 Burkitt lymphoma in a murine model,8 and adult T-cell leukemia9; produces—with 5-fluorouracil, cisplatin, paclitaxel (Taxol), and doxorubicin hydrochloride (Adriamycin)—additive growth delays against Lewis lung carcinoma10; and demonstrates antiangiogenic activity in an orthotopic pancreatic cancer model.11 

Multiple myeloma (MM) is a hematologic malignancy that affected 14 000 new patients in the United States in 2000,12 and it remains incurable with conventional therapies. Novel biologically based therapies are, therefore, urgently needed. We have previously demonstrated that PS-341 directly induces apoptosis by the activation of caspase-3 and without the associated alteration of Bax or Bcl-2 protein expression, even in MM cell lines and patient MM cells that are resistant to conventional therapies. PS-341 also overcomes the antiapoptotic effects of interleukin-6 (IL-6) or adherence to bone marrow stromal cells (BMSCs).4 In vivo PS-341 inhibits human MM cell growth and associated angiogenesis and prolongs survival in a murine SCID mouse model. Most important, PS-341 has achieved responses, even complete responses, in a phase 2 clinical trial treating patients with relapsed MM refractory to conventional therapies, and it has an acceptable toxicity profile.13This drug, therefore, represents a new treatment paradigm targeting not only the tumor cell but also the MM cell–host interaction and the bone marrow (BM) milieu to overcome drug resistance and improve patient outcome.

Both proapoptotic and antiapoptotic proteins are substrates of UPP,1 and the molecular mechanisms whereby PS-341 mediates its anti-MM activity are not yet defined. In this study, we demonstrate that PS-341 induces p53 and MDM2 protein expression; induces phosphorylation (Ser15) of p53 protein; and activates c-Jun NH2-terminal kinase (JNK), which in turn activates caspase-8 and caspase-3. Activated caspase-3, in turn, cleaves DNA protein kinase catalytic subunit (DNA-PKcs), ATM, and MDM2; conversely, caspase inhibitors Z-VAD-FMK and Z-IETD-FMK abrogate these effects. Furthermore, the inhibition of JNK activity by SP600125 reduces PS-341–induced MM cell death. Our results demonstrate that PS-341 induces caspase activation, inhibits DNA repair, and activates p53 by phosphorylation and degradation of MDM2. Given the early clinical promise and favorable toxicity profile of PS-341 in patients with relapsed refractory MM, these studies provide the framework for further clinical evaluation of PS-341, alone and coupled with conventional or other novel therapies, to improve patient outcome in MM.

MM-derived cell lines and patient MM cells

Dr Steven Rosen (Northwestern University, Chicago, IL) kindly provided MM.1S (wild-type p53)14 human MM cell line. RPMI8226 and U266 human MM cells were obtained from American Type Culture Collection (Rockville, MD). All MM cell lines were cultured in RPMI 1640 containing 10% fetal bovine serum (FBS; Sigma Chemical, St Louis, MO), 2 μM L-glutamine, 100 U/mL penicillin, and 100 μg/mL streptomycin (Gibco, Grand Island, NY). Patient MM cells were purified from patient BM aspirates by negative selection using anti-CD2, -CD14, -CD33, -CD41, -CD45RA, and -CD66b antibodies (RosetteSep separation system; StemCell Technologies, Vancouver, BC, Canada). The purity of MM cells is greater than 95%, as confirmed by flow cytometric analysis using anti-CD138 antibody (BD PharMingen, San Diego, CA).

Reagents

Proteasome inhibitor PS-341 (Millennium Pharmaceuticals, Cambridge, MA) was dissolved in dimethyl sulfoxide (DMSO), stored at −20°C, and used at 5 to 80 nM in the presence of 5% FBS. JNK inhibitor 60021515,16 (Calbiochem, San Diego, CA) was stored at −20°C and used at 5 to 40 μM. Pan-caspase inhibitor Z-VAD-FMK and caspase-8 inhibitor Z-IETD-FMK (Calbiochem) were also stored at −20°C and were used at 100 μM and 25 μM, respectively. For inhibition of caspase cleavage, cells were incubated with caspase inhibitors for 1 hour before incubation with PS-341 (20 nM) for 8 hours. Caffeine (Sigma) (0.5 and 1 mM) was used as an inhibitor of ATM/ATR.

Growth inhibition assay

MM.1S cells were cultured for 24 hours with 6-25 nM PS-341 in the presence (5-10 μM) or absence of SP600125. The inhibitory effect of PS-341 on MM cell growth in the presence or absence of SP600125 was assessed by measuring 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrasodium bromide (MTT; Chemicon International, Temecula, CA) dye absorbance, as previously described.14,17 

Preparation of biotinylated probes and hybridization on microarrays

MM.1S cells were incubated with 20 nM PS-341 for 0.5, 1, 3, and 6 hours in the presence of 5% FBS. mRNA expression profiling was analyzed using Affymetrix huGene FL arrays (Santa Clara, CA) containing 12 626 genes, as previously described.18 

Immunoblotting

MM cell lines and patient MM cells cultured with PS-341 were lysed, and protein lysates (50 μg) were subjected to Western blotting, as in prior studies.11,17 Nuclear extracts were prepared using the Nuclear Extract Kit (Activemotif, Carlsbad, CA), according to the manufacturer's instructions. Antibodies used for immunoblotting included anti–phospho-p53 (Ser6, Ser9, Ser15, Ser20, Ser37, Ser46, Ser392), p53, phospho-SEK1, phospho-SAPK, phospho-c-Jun, phospho-ATF-2, phospho-Bcl-2, and caspase-8 (Cell Signaling, Beverly, MA); anti-MDM2, -JNK1, -DNA-PKcs, -Bcl-2, -Bcl-XL, -α-tubulin, and -nucleolin (Santa Cruz Biotechnology, Santa Cruz, CA); anti-ATM (Oncogene Research Products, Boston, MA); and anti–caspase-3 (BD PharMingen). For immunoprecipitation, whole-cell lysates were incubated with anti-p53 and MDM2 overnight at 4°C and then incubated with protein A/G PLUS-Agarose (Santa Cruz Biotechnology) for 4 hours at 4°C as in our prior study.14,19 

Statistical analysis

Statistical significance of differences observed in drug-treated versus control cultures was determined using the Wilcoxon signed-rank test. The minimal level of significance was P < .05.

PS-341 up-regulates expression of p53 and MDM2 proteins and induces phosphorylation (Ser15) of p53

Because p53 is a substrate of UPP, we first examined the effect of PS-341 on p53 protein expression in MM.1S cells. MM.1S cells were treated with PS-341 (20 nM) for up to 16 hours in the presence of 5% FBS. As expected, p53 protein expression was transiently up-regulated after PS-341 treatment in MM.1S cells, with peak expression at 8 hours (Figure 1A). PS-341 also induces transient p53 phosphorylation on Ser15 without the phosphorylation of Ser6, Ser9, Ser20, Ser37, Ser46, or Ser392 residues (data not shown), in the same pattern as p53 protein expression. MDM2 protein is also transiently up-regulated after PS-341 treatment, with peak protein expression observed at 6 hours. We next examined the dose-dependent effect of PS-341 on phosphorylation and protein expression of these proteins. Phosphorylation (Ser15) and protein expression of p53 are up-regulated in a dose-dependent fashion; in contrast, MDM2 expression was decreased only after high-dose PS-341 treatment (Figure 1B). To assess the effect of PS-341 in patient MM cells, we next purified the MM cells from patient bone marrow aspirates. As can be seen in Figure1C, PS-341 treatment (20 nM, 8 hours) induces the phosphorylation of p53 (Ser15) and up-regulates p53 and MDM2 protein expression in patient MM cells. Because DNA damage induces p53 phosphorylation,20,21 these results suggest that PS-341 induces DNA damage in MM cell lines and patient MM cells. We further examined the distribution and interaction of p-p53, p53, and MDM2 proteins. As expected, most p-p53 is present in the nucleus, and PS-341 triggered increased protein expression associated with the nuclear translocation of p-p53. In contrast, cytoplasmic p53 is in its nonphosphorylated form (Figure 1D). We also examined the association of p53 and MDM2 protein using coimmunoprecipitation with anti-p53 and MDM2 antibodies, and we demonstrated that PS-341 induces coimmunoprecipitation of p53 with MDM2 protein in a time-dependent fashion (Figure 1E). PS-341–induced interaction of p53 and MDM2 occurs earlier than the induction of p53 protein.

Fig. 1.

Induction of phosphorylation and protein expression of p53 and MDM2 by PS-341.

(A) PS-341 induces p53 phosphorylation (Ser15) and increased protein expression of p53 and MDM2 in MM.1S cells in a time-dependent fashion. Anti–α-tubulin antibody is used to confirm equal loading of proteins. (B) PS-341 induces p53 phosphorylation (Ser15) and increased protein expression of p53 and MDM2 in MM.1S cells in a dose-dependent fashion. Anti–α-tubulin antibody is used to confirm equal loading of proteins. (C) PS-341 induces p53 phosphorylation (Ser15) and increased protein expression of p53 and MDM2, in purified patient MM cells. Pat indicates patient. (D) Localization of p-p53, p53, and MDM2 proteins in cytoplasmic and nuclear fractions of MM.1S cells treated with PS-341. Anti–α-tubulin and anti–nucleolin antibodies are used to confirm equal loading of cytoplasmic and nuclear proteins, respectively. (E) Coimmunoprecipitation of p53 and MDM2 in MM.1S cells treated with PS-341.

Fig. 1.

Induction of phosphorylation and protein expression of p53 and MDM2 by PS-341.

(A) PS-341 induces p53 phosphorylation (Ser15) and increased protein expression of p53 and MDM2 in MM.1S cells in a time-dependent fashion. Anti–α-tubulin antibody is used to confirm equal loading of proteins. (B) PS-341 induces p53 phosphorylation (Ser15) and increased protein expression of p53 and MDM2 in MM.1S cells in a dose-dependent fashion. Anti–α-tubulin antibody is used to confirm equal loading of proteins. (C) PS-341 induces p53 phosphorylation (Ser15) and increased protein expression of p53 and MDM2, in purified patient MM cells. Pat indicates patient. (D) Localization of p-p53, p53, and MDM2 proteins in cytoplasmic and nuclear fractions of MM.1S cells treated with PS-341. Anti–α-tubulin and anti–nucleolin antibodies are used to confirm equal loading of cytoplasmic and nuclear proteins, respectively. (E) Coimmunoprecipitation of p53 and MDM2 in MM.1S cells treated with PS-341.

Close modal

PS-341 induces c-Jun NH2-terminal kinase activation

We next examined whether PS-341 induces a stress response in MM.1S cells. PS-341 induces the phosphorylation of JNK, and upstream (SEK-1) and downstream (c-Jun and ATF-2) proteins, in a time- (Figure2A) and dose- (Figure 2B) dependent fashion. PS-341 similarly induces the phosphorylation of JNK in U266 and RPMI 8226 MM cell lines (Figure 2C). As we have done in dexamethasone-treated MM.1S cells,18 we next performed gene microarray profile analysis in MM.1S cells before and after PS-341 treatment. As seen in Figure 2D, PS-341 induces a significant (2.5- to 72-fold) increase in heat shock protein (HSP) transcription in a time-dependent fashion. These results indicate that PS-341 induces a stress response in MM cell lines.

Fig. 2.

PS-341 induces JNK activation and HSP gene expression.

(A) PS-341 induces the phosphorylation of JNK, upstream molecule SEK1, and downstream targets c-Jun and ATF-2, in MM.1S cells in a time-dependent fashion. Immunoblotting with anti–α-tubulin antibody confirms equal protein loading. (B) PS-341 induces the phosphorylation of JNK and c-Jun in dose-dependent fashion. (C) PS-341 induces the phosphorylation of JNK in MM.1S, U266, and RPMI 8226 MM cell lines. (D) Gene microarray analysis demonstrates the up-regulation of HSP27 (♦), HSP40 (▪), HSP70 (▴), HSP90 (●), and HSP110 (○) mRNA in MM.1S cells treated with PS-341.

Fig. 2.

PS-341 induces JNK activation and HSP gene expression.

(A) PS-341 induces the phosphorylation of JNK, upstream molecule SEK1, and downstream targets c-Jun and ATF-2, in MM.1S cells in a time-dependent fashion. Immunoblotting with anti–α-tubulin antibody confirms equal protein loading. (B) PS-341 induces the phosphorylation of JNK and c-Jun in dose-dependent fashion. (C) PS-341 induces the phosphorylation of JNK in MM.1S, U266, and RPMI 8226 MM cell lines. (D) Gene microarray analysis demonstrates the up-regulation of HSP27 (♦), HSP40 (▪), HSP70 (▴), HSP90 (●), and HSP110 (○) mRNA in MM.1S cells treated with PS-341.

Close modal

Inhibition of JNK activity reduces PS-341–induced cell death by inhibition of caspase-3 activation

To examine whether the activation of JNK plays a role in mediating PS-341–induced apoptosis, we used JNK-specific inhibitor SP600125.15,16 In the presence of SP600125, the phosphorylation of JNK triggered by PS-341 is completely abrogated in a dose-dependent fashion (Figure 3A). Phosphorylation of c-Jun or ATF-2 is also blocked by SP600125 (data not shown). SP600125 does not block the phosphorylation of p53 or the induction of MDM2 by PS-341. We further examined whether the inhibition of JNK activity also inhibits caspase-3 cleavage. As seen in Figure3B, SP600125 inhibits PS-341–induced caspase-3 cleavage, consistent with its inhibition of JNK phosphorylation. SP600125 in a dose-dependent fashion also significantly (P < .01) inhibits PS-341–induced cytotoxicity in MM.1S cells, assessed by MTT assay (Figure 3C). These results suggest that JNK plays a critical role in mediating PS-341–induced apoptosis through the activation of caspase-3.

Fig. 3.

JNK inhibitor blocks caspase-3 cleavage and abrogates PS-341–induced apoptosis in MM.1S cells.

(A) Inhibition of PS-341–induced JNK phosphorylation, but not of MDM2 expression or p53 phosphorylation, by JNK inhibitor SP600125. (B) Caspase-3 cleavage triggered by PS-341 is dependent on JNK activity. SP600125 inhibits the phosphorylation of JNK and caspase-3 cleavage triggered by PS-341. (C) Cell death triggered by PS-341 is partially dependent on JNK activation. MM.1S cell death triggered by PS-341 was measured by MTT assay, in the absence (■) or presence of 5 μM (░) and 10 μM (▪) SP600125. *P < .01 (versus 0 μM).

Fig. 3.

JNK inhibitor blocks caspase-3 cleavage and abrogates PS-341–induced apoptosis in MM.1S cells.

(A) Inhibition of PS-341–induced JNK phosphorylation, but not of MDM2 expression or p53 phosphorylation, by JNK inhibitor SP600125. (B) Caspase-3 cleavage triggered by PS-341 is dependent on JNK activity. SP600125 inhibits the phosphorylation of JNK and caspase-3 cleavage triggered by PS-341. (C) Cell death triggered by PS-341 is partially dependent on JNK activation. MM.1S cell death triggered by PS-341 was measured by MTT assay, in the absence (■) or presence of 5 μM (░) and 10 μM (▪) SP600125. *P < .01 (versus 0 μM).

Close modal

PS-341 induces DNA-PKcs and ATM cleavage in MM

Others have demonstrated that DNA-PKcs is a target for IL-1–converting enzyme (ICE)–like22 and CPP32-like apoptotic protease.23 We have previously reported that PS-341 induces caspase-3 cleavage in MM cells,4 so we next examined whether DNA-PKcs, ATM, or both are cleaved by caspase activation triggered by PS-341. As expected, PS-341 induces cleavage of DNA-PKcs (180-kDa protein) in a time- (Figure 4A, upper panel), and dose- (Figure 4A, lower panel) dependent fashion in MM.1S cells. PS-341–induced DNA-PKcs cleavage also occurs in RPMI 8226 (Figure 4B, upper panel) and U266 (Figure 4B, lower panel) MM cell lines. Although a constitutive DNA-PKcs cleaved form is present in patient MM cells, PS-341 also induces DNA-PKcs cleavage in these cells (Figure 4C). PS-341 similarly cleaves ATM in a time- (Figure 4D, upper panel), and dose- (Figure 4D, lower panel) dependent fashion in MM.1S cells. These results suggest that PS-341 may inhibit DNA repair by cleavage of DNA-PKcs, ATM, or both in MM cell lines and patient MM cells.

Fig. 4.

Cleavage of DNA-PKcs and ATM induced by PS-341.

(A) PS-341-induces time- (upper panel) and dose- (lower panel) dependent DNA-PKcs cleavage in MM.1S cells. (B) Time-dependent DNA-PKcs cleavage is induced by PS-341 treatment in RPMI 8226 (upper panel) and U266 (lower panel) MM cell lines. (C) DNA-PKcs cleavage is also triggered by PS-341 treatment in purified patient MM cells. (D) PS-341 induces time- (upper panel) and dose- (lower panel) dependent ATM cleavage in MM.1S cells.

Fig. 4.

Cleavage of DNA-PKcs and ATM induced by PS-341.

(A) PS-341-induces time- (upper panel) and dose- (lower panel) dependent DNA-PKcs cleavage in MM.1S cells. (B) Time-dependent DNA-PKcs cleavage is induced by PS-341 treatment in RPMI 8226 (upper panel) and U266 (lower panel) MM cell lines. (C) DNA-PKcs cleavage is also triggered by PS-341 treatment in purified patient MM cells. (D) PS-341 induces time- (upper panel) and dose- (lower panel) dependent ATM cleavage in MM.1S cells.

Close modal

Mechanism by which PS-341 cleaves DNA-PKcs and ATM

Having shown that PS-341 induces DNA-PKcs and ATM cleavage, we next examined the mechanism whereby PS-341 triggers the cleavage of these protein kinases. As in our prior study,4 PS-341 induces caspase-3 cleavage (Figure 5A, upper panel) and caspase-8 cleavage (Figure 5A, lower panel) in a time-dependent fashion. In contrast, there are no caspase-9 cleavage (data not shown) and no changes in phosphorylation of Bcl-2 or in protein expression of Bcl-2 and Bcl-XL (Figure 5B). To determine whether caspase-3 activation mediates DNA-PKcs or ATM cleavage induced by PS-341, we examined the effect of caspase inhibitors on DNA-PK and ATM cleavage (Figure 5C). As expected, pan-caspase inhibitor Z-VAD-FMK (4th lane) and caspase-8 inhibitor Z-IETD-FMK (3rd lane) completely abrogate DNA-PKcs and ATM cleavage induced by PS-341. These caspase inhibitors also abrogate PS-341–induced phosphorylation, but not protein expression, of p53 (Figure 5C). Because MDM2 is cleaved during apoptosis,24 we next examined whether PS-341 also cleaves MDM2. As seen in Figure 5D, MDM2 is completely cleaved after treatment with PS-341 for 10 hours. To define the role of ATM or ATR in the phosphorylation of p53 induced by PS-341 treatment, we used caffeine as an inhibitor of ATM25 and ATR.26 Caffeine inhibits PS-341–induced p53 phosphorylation, but not p53 protein expression, in MM.1S cells in a time- (Figure 5E, upper panel) and dose- (Figure 5E, lower panel) dependent fashion. These results suggest that PS-341 induces DNA-PKcs and ATM cleavage, phosphorylation of p53 and MDM2 cleavage through activation of caspase-3 by caspase-8.

Fig. 5.

Caspase inhibitors block PS-341–induced DNA-PKcs or ATM cleavage.

(A) PS-341 induces time-dependent caspase-3 and caspase-8 cleavage in MM.1S cells. (B) PS-341 does not affect the phosphorylation of Bcl-2 or the protein expression of Bcl-2 and Bcl-xL. Immunoblotting with anti–α-tubulin antibody confirms equal protein loading. (C) Caspase-8 inhibitor Z-IETD-FMK and pan-caspase inhibitor Z-VDD-FMK protect against the PS-341–induced cleavage of DNA-PKcs and ATM and the phosphorylation of p53 through the inhibition of caspase-3 cleavage. (D) PS-341 induces time-dependent MDM2 protein cleavage in MM.1S cells. (E) Caffeine inhibits PS-341–triggered phosphorylation of p53 in a time- (upper panel) and dose- (lower panel) dependent fashion in MM.1S cells.

Fig. 5.

Caspase inhibitors block PS-341–induced DNA-PKcs or ATM cleavage.

(A) PS-341 induces time-dependent caspase-3 and caspase-8 cleavage in MM.1S cells. (B) PS-341 does not affect the phosphorylation of Bcl-2 or the protein expression of Bcl-2 and Bcl-xL. Immunoblotting with anti–α-tubulin antibody confirms equal protein loading. (C) Caspase-8 inhibitor Z-IETD-FMK and pan-caspase inhibitor Z-VDD-FMK protect against the PS-341–induced cleavage of DNA-PKcs and ATM and the phosphorylation of p53 through the inhibition of caspase-3 cleavage. (D) PS-341 induces time-dependent MDM2 protein cleavage in MM.1S cells. (E) Caffeine inhibits PS-341–triggered phosphorylation of p53 in a time- (upper panel) and dose- (lower panel) dependent fashion in MM.1S cells.

Close modal

UPP is a major proteolytic system that selectively removes mutant, damaged, or misfolded proteins. It also regulates the expression of proteins mediating cell-cycle progression (p21Cip,1 p27Kip,1cyclins), oncogenesis (p53, IκB), and apoptosis (Bcl, cIAP, XIAP, Bax).1 Proteasome inhibitors block UPP and induce caspase activation despite the expression of antiapoptotic proteins (Bcl, cIAP, XIAP) and independent of p53 status. The proteasome inhibitor PS-341 demonstrates remarkable anti-MM activity in vitro11 and in murine MM models,27 and most recently it has achieved stabilization or decrease in paraprotein in 77% of patients with refractory relapsed MM.13 The in vivo targets whereby PS-341 mediates anti-MM activity and, conversely, mechanisms of PS-341 resistance are at present undefined. In this study we define the molecular mechanisms of anti-MM activity of this agent to provide the framework for its optimal clinical use and to form the basis for the development of second-generation, more targeted, potent, and less toxic proteasome inhibitors.

Our previous study demonstrates that PS-341 induces apoptosis in MM cells with wild-type and mutant p53,4 consistent with previous reports that proteasome inhibitor-induced apoptosis occurs dependently5 or independently28 of p53 status. In this study, we hypothesized that DNA damage triggered by PS-341 treatment in MM cells is associated with the activation of DNA-PKcs, ATM/ATR, or both, and the activation of p53. We first report that PS-341 specifically induces the phosphorylation of p53 (Ser15). This induction of p53 phosphorylation is associated with increased p53 protein expression, as previously reported.29 PS-341 induces MDM2 protein and the association of p53 and MDM2 earlier than the phosphorylation of p53. Previous studies have demonstrated that the phosphorylation of p53 (Ser15) dissociates p53 from the p53/MDM2 complex; however, our results demonstrate p53 is still associated with MDM2 even after phosphorylation. Previous reports demonstrated that DNA damage induced the phosphorylation of p53 through the activation of DNA-PKcs20,30; therefore, our results strongly suggest that PS-341 induces DNA damage, activates DNA-PKcs, ATM/ATR, or both, and phosphorylates p53 (Ser15) in MM cell lines and primary patient MM cells.

We have studied mechanisms whereby conventional and novel therapies trigger MM cell apoptosis. For example, Dex triggers caspase-9–mediated MM cell death,19 whereas immunomodulatory derivatives of thalidomide31 and TRAIL32 induce caspase 8-mediated apoptosis. Our recent gene microarray data of MM.1S cells treated with PS-341 demonstrates transcriptional triggering of apoptotic cascades, down-regulation of growth/survival kinases, up-regulation of UPP, and induction of stress kinases, including heat shock proteins (HSPs). JNK, one of these stress-response proteins, mediates apoptosis triggered by unfolded proteins.33 JNK inhibitor SP600125 blocks PS-341–induced cell death by the abrogation of caspase-3 cleavage but does not affect the phosphorylation of p53. These data suggest that the activation of JNK modulates PS-341–induced caspase activation and apoptosis. Previous reports demonstrate that JNK increases the phosphorylation of antiapoptotic proteins Bcl-2 and Bcl-x, thereby reducing their antiapoptic activity34,35; however, our data demonstrate that PS-341 does not alter the protein expression of Bcl-2 and Bcl-XL or the phosphorylation of Bcl-2. This result is consistent with our data that PS-341 does not induce the activation of caspase-9 but rather induces caspase-8 apoptotic signaling.

DNA-PKcs is a phosphatidylinositol (PI) kinase that has a crucial role in the repair of damaged DNA and the phosphorylation of selective serine residues (Ser15) on p53,20,30 and it is a possible target for an ICE-like protease22 or a CPP32-like apoptotic protease.23 Therefore, we sought to determine whether the activation of caspases by PS-341 induces cleavage of DNA-PKcs, ATM, or both. We demonstrated that PS-341 cleaves DNA-PKcs and ATM in MM cell lines and primary patient MM cells. PS-341 activates caspase-3 through caspase-8 activation, whereas pan-caspase and caspase-8 inhibitors completely abrogate PS-341–induced caspase-8/caspase-3 activation and DNA-PKcs and ATM cleavage. These data suggest that the cleavage of DNA-PKcs and ATM triggered by PS-341 is dependent on caspase-8/caspase-3 signaling. Our results further demonstrate that the inhibition of caspase activation also inhibits p53 phosphorylation, but not protein expression of p53, suggesting that the phosphorylation of p53 is a secondary event following DNA damage induced by caspase-3. Taken together, our findings indicate that PS-341 activates caspase-3 through caspase-8 activation, impairs DNA repair by the cleavage of DNA-PKcs or ATM, and activates p53 through the phosphorylation of p53 and the degradation of MDM2 (Figure6). These effects all occur with PS-341 (20 nM) serum levels that are achieved in clinical trials; moreover, PS-341 irreversibly binds to proteasomes and accumulates in target cells. Further delineation of the molecular mechanisms mediating antitumor activity of these agents will provide the framework for their use, alone or coupled with other novel agents, to improve outcome. These studies will also establish the molecular basis for the development of more targeted, potent, and less toxic second-generation proteasome inhibitors.

Fig. 6.

Mechanism of action of PS-341–induced apoptosis.

PS-341 induces the up-regulation of p53 by the inhibition of UPP. Up-regulated p53 in turn induces MDM2 protein, which binds to p53 as a negative regulator of p53. JNK activates caspase-8 and caspase-3. Activation of caspase-3 induces DNA damage, which induces p53 (Ser15) phosphorylation. Phosphorylation of p53 by DNA-PKcs, ATM/ATR, or both and degradation of MDM2 by caspase-3 trigger the dissociation of MDM2/p53 binding. Furthermore, the activation of caspase-3 cleaves DNA-PKcs and ATM/ATR proteins, resulting in impaired DNA repair.

Fig. 6.

Mechanism of action of PS-341–induced apoptosis.

PS-341 induces the up-regulation of p53 by the inhibition of UPP. Up-regulated p53 in turn induces MDM2 protein, which binds to p53 as a negative regulator of p53. JNK activates caspase-8 and caspase-3. Activation of caspase-3 induces DNA damage, which induces p53 (Ser15) phosphorylation. Phosphorylation of p53 by DNA-PKcs, ATM/ATR, or both and degradation of MDM2 by caspase-3 trigger the dissociation of MDM2/p53 binding. Furthermore, the activation of caspase-3 cleaves DNA-PKcs and ATM/ATR proteins, resulting in impaired DNA repair.

Close modal

Prepublished online as Blood First Edition Paper, September 26, 2002; DOI 10.1182/blood-2002-08-2543.

Supported by National Institutes of Health grants PO-1 78378 and RO-1 CA 50947; the Doris Duke Distinguished Clinical Research Scientist Award (K.C.A.); the Multiple Myeloma Research Foundation (T.H., C.M., T.H., D.C., and N.M.); and the Myeloma Research Fund (K.C.A.).

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Kisselev
AF
Goldberg
AL
Proteasome inhibitors: from research tools to drug candidates.
Chem Biol.
8
2001
739
758
2
Adams
J
Palombella
VJ
Sausville
EA
et al
Proteasome inhibitors: a novel class of potent and effective antitumor agents.
Cancer Res.
59
1999
2615
2622
3
Sun
J
Nam
S
Lee
CS
et al
CEP1612, a dipeptidyl proteasome inhibitor, induces p21WAF1 and p27KIP1 expression and apoptosis and inhibits the growth of the human lung adenocarcinoma A-579 nude mice.
Cancer Res.
61
2001
1280
1284
4
Hideshima
T
Richardson
P
Chauhan
D
et al
The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells.
Cancer Res.
61
2001
3071
3076
5
Lopes
UG
Erhardt
P
Yao
R
Cooper
GM
p53-dependent induction of apoptosis by proteasome inhibitors.
J Biol Chem.
272
1997
12893
12896
6
Herrmann
JL
Briones
FJ
Brisbay
S
Logothetis
CJ
McDonnell
TJ
Prostate carcinoma cell death resulting from inhibition of proteasome activity is independent of functional Bcl-2 and p53.
Oncogene.
17
1998
2889
2899
7
Frankel
A
Man
S
Elliott
P
Adams
J
Kerbel
RS
Lack of multicellular drug resistance observed in human ovarian and prostate carcinoma treated with the proteasome inhibitor PS-341.
Clin Cancer Res.
6
2000
3719
3728
8
Orkowski
RZ
Eswara
JR
Lafond-Walker
A
Grever
MR
Orlowski
M
Dang
CV
Tumor growth inhibition induced in a murine model of human Burkitt's lymphoma by a proteasome inhibitor.
Cancer Res.
58
1998
4342
4348
9
Tan
C
Waldmann
TA
Proteasome inhibitor PS-341, a potential therapeutic agent for adult T-cell leukemia.
Cancer Res.
62
2002
1083
1086
10
Teicher
BA
Ara
G
Herbst
R
Palombella
VJ
Adams
J
The proteasome inhibitor PS-341 in cancer therapy.
Clin Cancer Res.
5
1999
2638
2645
11
Harbison
MT
Bruns
CJ
Bold
RJ
et al
Proteasome inhibitor PS-341 is effective as an antiangiogenic agent in the treatment of human pancreatic carcinoma via the inhibition of NF-κB and subsequent inhibition of vascular endothelial growth factor production [abstract].
Proc Am Assoc Cancer Res.
41
2000
71
12
Greenlee
RT
Murray
T
Bolden
S
Wingo
PA
Cancer Statistics, 2000.
CA Cancer J Clin.
50
2000
7
33
13
Richardson
PG
Berenson
J
Irwin
D
et al
Phase II trial of PS-341, a novel proteasome inhibitor, alone or in combination with dexamethasone, in patients with multiple myeloma who have relapsed following front-line therapy and are refractory to their most recent therapy [abstract].
Blood.
98
2001
774
14
Hideshima
T
Chauhan
D
Shima
Y
et al
Thalidomide and its analogues overcome drug resistance of human multiple myeloma cells to conventional therapy.
Blood.
96
2000
2943
2950
15
Han
Z
Boyle
DL
Chang
L
et al
c-Jun N-terminal kinase is required for metalloproteinase expression and joint destruction in inflammatory arthritis.
J Clin Invest.
108
2001
73
81
16
Bennett
BL
Sasaki
DT
Murray
BW
et al
SP600125, an anthrapyrazolone inhibitor of Jun N-terminal kinase.
Proc Natl Acad Sci U S A.
98
2001
13681
13686
17
Hideshima
T
Chauhan
D
Richardson
P
et al
NF-κB as a therapeutic target in multiple myeloma.
J Biol Chem.
277
2002
6639
6647
18
Chauhan
D
Auclair
D
Robinson
EK
et al
Identification of genes regulated by dexamethasone in multiple myeloma cells using oligonucleotide arrays.
Oncogene.
21
2002
1346
1358
19
Hideshima
T
Nakamura
N
Chauhan
D
Anderson
KC
Biologic sequelae of interleukin-6 induced PI3-K/Akt signaling in multiple myeloma.
Oncogene.
20
2001
5991
6000
20
Tibbetts
RS
Brumbaugh
KM
Williams
JM
et al
A role for ATR in the DNA damaged-induced phosphorylation of p53.
Genes Dev.
13
1999
152
157
21
Buschmann
T
Potapova
O
Bar-Shira
A
et al
Jun NH2-terminal kinase phosphorylation of p53 on Thr-81 is important for p53 stabilization and transcriptional activities in response to stress.
Mol Cell Biol.
21
2001
2743
2754
22
Song
Q
Lees-Miller
SP
Kumar
S
et al
DNA-dependent protein kinase catalytic subunit: a target for an ICE-like protease in apoptosis.
EMBO J.
15
1996
3238
3246
23
Han
Z
Malik
N
Carter
T
Reeves
WH
Wyche
JH
Hendrickson
EA
DNA-dependent protein kinase is a target for a CPP32-like apoptotic protease.
J Biol Chem.
271
1996
25035
25040
24
Chen
L
Marechal
V
Moreau
J
Levine
AJ
Chen
J
Proteolytic cleavage of the mdm2 oncoprotein during apoptosis.
J Biol Chem.
272
1997
22966
22973
25
Sarkaria
JN
Busby
EC
Tibbetts
RS
et al
Inhibition of ATM and ATR kinase activity by the radiosensitizing agent, caffeine.
Cancer Res.
59
1999
4375
4382
26
Hall-Jackson
CA
Cross
DA
Morrice
N
Smythe
C
ATR is a caffeine-sensitive, DNA-activated protein kinase with a substrate specificity distinct from DNA-PK.
Oncogene.
18
1999
6707
6713
27
LeBlanc
R
Catley
LP
Hideshima
T
et al
Proteasome inhibitor PS-341 inhibits human myeloma cell growth in vivo and prolongs survival in a murine model.
Cancer Res.
62
2002
4996
5000
28
An
WG
Hwang
SG
Trepel
JB
Blagosklonny
MV
Proteasome inhibitor-induced apoptosis: accumulation of wt p53, p21WAF1/CIP1, and induction of apoptosis are independent markers of proteasome inhibition.
Leukemia.
14
2000
1276
1283
29
Klibanov
SA
O'Hagan
HM
Ljungman
M
Accumulation of soluble and nucleolar-associated p53 proteins following cellular stress.
J Cell Sci.
114
2001
1867
1973
30
Shieh
SY
Ikeda
M
Taya
Y
Prives
C
DNA damaged-induced phosphorylation of p53 alleviates inhibition by MDM2.
Cell.
91
1997
325
334
31
Mitsiades
N
Mitsiades
CS
Poulaki
V
et al
Apoptotic signaling induced by immunomodulatory thalidomide analogs in human multiple myeloma cells: therapeutic implications.
Blood.
99
2002
4525
4530
32
Mitsiades
CS
Treon
SP
Mitsiades
N
et al
TRAIL/Apo2L ligand selectively induces apoptosis and overcomes drug resistance in multiple myeloma: therapeutic applications.
Blood.
98
2001
795
804
33
Gabai
VL
Meriin
AB
Yaglom
JA
Volloch
VZ
Sherman
MY
Role of Hsp70 in regulation of stress-kinase JNK: implications in apoptosis and aging.
FEBS Lett.
438
1998
1
4
34
Srivastava
RK
Sollott
SJ
Khan
L
Hansford
R
Lakatta
EG
Longo
DL
Bcl-2 and Bcl-X(L) block thapsigargin-induced nitric oxide generation, c-Jun NH(2)-terminal kinase activity, and apoptosis.
Mol Cell Biol.
19
1999
5659
5674
35
Kharbanda
S
Saxena
S
Yoshida
K
et al
Translocation of SAPK/JNK to mitochondria and interaction with Bcl-x(L) in response to DNA damage.
J Biol Chem.
275
1999
322
327

Author notes

Kenneth C. Anderson, Dana-Farber Cancer Institute, M557, 44 Binney St, Boston, MA 02115; e-mail:kenneth_anderson@dfci.harvard.edu.

Sign in via your Institution