Androgen ablation and chemotherapy provide effective palliation for most patients with advanced prostate cancer, but eventually progressing androgen-independent prostate cancer threatens the lives of patients usually within a few years, mandating improvement in therapy. Proteasome inhibition has been proposed as a therapy target for the treatment of solid and hematological malignancies. The proteasome is a ubiquitous enzyme complex that is a hub for the regulation of many intracellular regulatory pathways; because of its essential function, this enzyme has become a new target for cancer treatment. Studies with bortezomib (VELCADE, formerly known as PS-341) and other proteasome inhibitors indicate that cancer cells are especially dependent on the proteasome for survival, and several mechanisms used by prostate cancer cells require proteasome function. Bortezomib has been studied extensively in vitro and in vivo, and anticancer activity has been seen in cell and animal models for several solid tumor types, including prostate cancer. A Phase I trial to determine the maximum tolerated dose of once-weekly bortezomib has been completed. This trial included a large fraction of patients with androgen-independent prostate cancer. The maximum tolerated dose was reached at 1.6 mg/m2. A correlation was seen among bortezomib dose, proteasome inhibition, and positive modulation of serum prostate-specific antigen. There was also evidence of down-regulation of serum interleukin 6, a downstream nuclear factor κB effector. This Phase I trial and preclinical studies support additional testing of bortezomib in combination with radiation or chemotherapy for androgen-independent prostate cancer.

Androgens are necessary for normal prostate cell growth (1), and in animal models, androgen ablation causes cell atrophy and death of prostate epithelial cells (2). Likewise, in the early stages of prostate cancer, the growth of cancerous prostatic epithelial cells is often androgen dependent (3). Androgen ablation remains the main initial treatment of advanced prostate cancer and provides palliation of symptoms and survival benefit. However, androgen-independent cells are eventually selected during androgen ablation therapy (4), and progression to an androgen-independent state remains the primary cause of mortality in these patients within an average of 1.5 years.

Treatment strategies currently available for treating androgen-independent prostate cancer (AIPC; i.e., radiation or chemotherapy) provide temporary palliation, but eventually prostate cancer cells become resistant to chemotherapy and radiation, with ensuing failure to control tumor growth. New treatments and modulation of established treatment regimens (i.e., restoring sensitivity to chemotherapy) are therefore being sought.

Several mechanisms have been proposed to account for the development of androgen independence in prostate cancer cells (4). Intrinsic activation of receptors or activation of androgen receptor (AR) coregulatory molecules (e.g., ARA55, ARA54, ARA70, BRCA1, and heat-shock proteins) may allow cells to become independent of androgens (intrinsic activation). Induction of growth factors [interleukin 6 (IL-6), insulin-like growth factor-1, and vascular endothelial growth factor/platelet-derived growth factor) may indirectly activate the AR and/or its downstream signaling pathways (5, 6, 7). In addition, loss of tumor suppressor activity or proapoptotic factors can contribute to androgen-independent growth (8) and has been reviewed (9). However, it is likely that no single pathway exists for the loss of androgen dependence, and all of these pathways (and possibly others) may work together to bring about androgen independence and disease progression.

In this review we discuss current knowledge about the structure and function of the proteasome and preclinical effects of proteasome inhibition, as well as initial clinical experience with bortezomib (VELCADE), the first proteasome inhibitor, in patients with androgen-independent prostate carcinoma.

The Proteasome: A New Cellular Target for Prostate Cancer Treatment.

Bortezomib is a proteasome inhibitor. In preclinical experiments, bortezomib induces growth arrest and apoptosis in many tumor types (10), including androgen-dependent (LNCaP) and androgen-independent prostate cancer cell lines (PC-3 and DU145; Refs. 11, 12, 13, 14, 15). As demonstrated in these experiments, proteasome inhibition could be a novel method for treating androgen-dependent and androgen-independent prostate cancer.

The proteasome is a highly conserved and essential mechanism for degrading the majority of intracellular proteins in the eukaryotic cell. Cellular homeostasis and intracellular signaling pathways depend on the proteasome for regulation of key proteins. Encouraging results from Phase I studies of the proteasome inhibitor bortezomib in hematological malignancies (16), AIPC (17, 18), and other solid tumors (19) provide ample justification for additional study of this drug in human malignancies.

The Proteasome and the Ubiquitin-Proteasome Pathway.

The proteasome is a large multiprotein complex present in all cells, both in the cytoplasm and nucleus, that degrades ubiquitinated proteins (20, 21). Degradation by the proteasome is a highly regulated process that controls the expression of a wide variety of cellular targets including cyclins (i.e., cyclin B), cyclin-dependent kinase inhibitors (i.e., p21 and p27; Ref. 22), pro- and antiapoptosis factors such as Bax and Bcl-2 (23, 24), early immediate transcription factors such as c-Myc (25), tumor suppressors such as p53 (26), and transcriptional inhibitors such as Id (27) and inhibitor of nuclear factor-κB (IκB)α (28). PEST (proline-glutamate-serine-threonine) sequences and/or PEST-like sequences are often involved in the cellular targeting of proteins for degradation (29). Phosphorylation of PEST sequences in response to cellular signals may mediate the initiation of the ubiquitination and the degradative process (30). Phosphorylated PEST sequences are recognized by ubiquitin ligases, which catalyze the attachment of a single ubiquitin molecule (31, 32). Ubiquitin molecules are subsequently added in a characteristic formation to create a polyubiquitin chain on the protein, and it is these ubiquitin chains that the proteasome uses to recognize proteins targeted for degradation (33). This mechanism, in part, allows tight regulation of protein degradation by the proteasome.

The 26S proteasome is a multiprotein complex composed of a 20S proteolytic core particle flanked at each end by the 19S regulatory complexes (34, 35). The 20S core is composed of four subunits that include two β and two α rings. Within each β ring, there are three active sites: a chymotrypsin-like site, a trypsin-like site, and a postacidic or caspase-like site (also sometimes called peptidyl glutamyl peptide hydrolase-like site; Ref. 36). Of the three catalytic activities in the 20S core structure, the chymotrypsin-like site appears to have the most prevalent proteolytic activity (Fig. 1; Refs. 37, 38). The 19S complexes recognize the polyubiquitin chain on target proteins, recycle the polyubiquitin chain, and denature the target protein so that it can be moved into the catalytic chamber in the 20S core.

Bortezomib is a reversible but potent and selective inhibitor (Ki = 0.6 nm) of the chymotryptic-like activity of the proteasome (39). Whereas the other proteolytic activities are not inhibited by bortezomib, inhibition of the chymotrypsin-like activity alone results in significant blockade of proteasomal protein degradation within the cell (40).

Androgen-Dependent and Androgen-Independent Prostate Cancer Growth.

Androgens act through several pathways to support prostate cancer cell growth, and deprivation of androgens can induce cell death (1, 2). Androgens block Fas- or tumor necrosis factor α-induced apoptosis in androgen-dependent prostate cancer (LNCaP) cells by obstructing caspase activation in a manner that is both independent of the phosphatidylinositol 3′-kinase-PTEN-Akt pathway and without nuclear factor κB (NFκB) activation (41, 42). Androgen-independent growth can develop through several mechanisms.

AR mutations have been found in AIPC metastatic tumors and cell lines (43); these mutations (44, 45) can alter AR ligand specificity and lead to inappropriate AR activation (43). In the androgen-independent cell line MDAPa, mutations in the AR gene allowed binding to nonandrogen molecules such as cortisol, cortisone, and synthetic glucocorticoids (46).

Growth factor signaling pathways can activate AR independently of androgen via phosphorylation at specific AR sites (47). This point is particularly important, because the up-regulated paracrine and autocrine expression of growth factors and their cognate receptors coincides with tumor metastasis and androgen-independent growth. IL-6 serum levels are elevated in AIPC with advanced disease (48), and IL-6 transcription is inhibited by dihydrotestosterone through IκBα in prostate cells (49), which suggests an inverse relationship between IL-6 and androgen in prostate cancer. The recent finding that IL-6 stimulation of androgen-responsive LNCaP cells results in phosphorylation of the AR and transcription of AR-driven promoters suggests a mechanism of cross-talk and redundancy between the IL-6 and AR growth-promoting pathways (50). Independent of their effects on the AR, other autocrine and paracrine factors are associated with progression of prostate cancer. IL-8 overexpression is correlated with angiogenesis and metastasis, as well as expression of matrix metalloproteases and vascular endothelial growth factor, in an animal androgen-independent tumor model (51, 52). Vascular endothelial growth factor is also expressed in neuroendocrine-positive tumor cells of human prostate carcinoma specimens, and its expression is strongly associated with higher microvessel density, increasing tumor stage, and a less differentiated cellular morphology (53).

Constitutive activation of the transcription factor NFκB may also be associated with more aggressive prostate cancers. Bortezomib-induced inhibition of IκBα degradation by the proteasome would result in cytoplasmic NFκB inhibition (through IκBα binding) and inability of NFκB to translocate to the nucleus and bind to the promoters of multiple genes. These NFκB-targeted genes include proinflammatory cytokines (i.e., IL-1, IL-6, and tumor necrosis factor-α), cell adhesion molecules (i.e., vascular cell adhesion molecule, intercellular cell-adhesion molecule, and E-selectin), stress response enzymes (i.e., cyclooxygenase 2, nitric-oxide synthase, and 5-lipoxygenase; Ref. 54), and antiapoptotic proteins (i.e., inhibitor of apoptosis protein and the BCL-2 family; Refs. 55, 56, 57, 58; Fig. 2). Many of these pathways are involved in tumor growth, angiogenesis, metastasis, and resistance to chemotherapy in both solid and hematological malignancies (59, 60, 61, 62, 63).

Blockade of NFκB activity via the expression of a dominant-negative IκBα construct resulted in the suppression of angiogenesis and prostate cancer cell invasion and metastasis (64). The in vivo expression of vascular endothelial growth factor, IL-8, and matrix metalloproteinase 9 was decreased in nude mice bearing xenografted tumors of human prostate cells transfected with the mutated IκBα constructs (64). Additionally, NFκB-dependent transcriptional activity was ∼10 times higher in invasive PC-3 clones compared with less invasive lines, suggesting that NFκB activity is associated with a highly metastatic phenotype (65). Constitutive NFκB activity is also associated with resistance to the apoptosis-inducing effects of tumor necrosis factor α in AIPC cells (66). Muenchen et al. (67) found that both androgen-dependent (LNCaP) and androgen-independent (PC-3) prostate cancer cell lines could be made tumor necrosis factor α sensitive by transfecting them with a dominant-negative IκB construct, resulting in reduction of IL-6 mRNA and protein expression. Although constitutive NFκB activity alone is not sufficient for androgen independence and other cellular factors must be involved in mediating androgen independence in prostate cancer cells (68), it appears that NFκB inhibition through proteasome inhibition or other methods remains a potential cellular target for cancer treatment (69, 70).

Mutations to tumor suppressor genes and oncogenes are also evident in established human androgen-independent and androgen-dependent prostate cancer cell lines, as well as primary prostate cancers and metastases (71, 72, 73). The human prostate cancer cell lines LNCaP, DU145, and PC-3 vary in their expression of important tumor suppressor genes such as p53, retinoblastoma (Rb), and phosphatase/tensin homologue deleted on chromosome 10 (PTEN), but no specific genotype has been assigned to cancer cells with androgen independence or dependence (Table 1). Inactivating mutations within the PTEN tumor suppressor gene have been identified in primary prostate tumors, the prostate cell line DU145, and breast and brain tumors (71, 72). PTEN is a dual-specific phosphatidyl phosphatase that modulates cell-cycle progression and negatively regulates the phosphatidylinositol 3′-kinase/Akt (protein kinase B) pathway (74). PTEN loss is associated with up-regulation of Bcl-2 transcription and expression (64), loss of p27 expression, and tumor progression (75). The cyclin-dependent kinase inhibitor p27 has been identified as a prognostic factor in prostate cancer, and loss of p27 expression is correlated with advanced AIPC (76). PTEN inactivation also results in the constitutive activation of the phosphatidylinositol 3′-kinase/Akt pathway in both androgen-dependent and androgen-independent prostate tumor cells (75, 77, 78, 79). In turn, the activation of the Akt serine/threonine kinase modulates the expression of the AR in normal and prostate tumor cells (80).

Interestingly, the overexpression of the epidermal growth factor-like receptor tyrosine kinase Her-2/neu has been seen in prostate cancer (72) and in the primary tumors of patients who have undergone androgen ablation therapy (81). Her-2/neu overexpression was identified in primary tumors, and continued overexpression occurred in androgen-independent tumors that developed after androgen ablation therapy (82, 83). Like PTEN mutations, Her-2/neu overexpression activates Akt (84) and may trigger the AR transcriptional activity and promote growth in an androgen-independent manner (85). Her-2/neu-mediated Akt activation has also been associated with the adaptation to androgen independence by LNCaP cells (84), possibly through an NFκB-mediated pathway (86). Akt phosphorylates IκBα, prompting degradation of IκBα by the proteasome (28) and activating NFκB (87), thereby protecting both androgen-dependent and androgen-independent cells from apoptosis. Thus, the transition to androgen-independent growth probably involves several different mechanisms related to the perturbation of normal cell growth and tumor suppression in the cell, which may involve, in part, activation of AR, NFκB, Akt, and other growth-signaling pathways (Fig. 3).

Promotion of Apoptosis by Bortezomib in Prostate Cancer Models.

The proteasome is a hub for the regulation of many cellular signaling pathways, and therefore proteasome inhibition appears to induce apoptosis through a number of mechanisms (39). In prostate cancer, initial evidence indicates that sensitivity is independent of the requirements of the tumor for androgens. Experiments with bortezomib show that this agent induces apoptosis in androgen-dependent and androgen-independent prostate cancer cell lines (11, 12, 13). Deregulation of proteasomal function by inhibition of proteolysis induces apoptosis in tumor cells, including those without functional p53, PTEN, or p21 (13); cells overexpressing the antiapoptosis factor Bcl-2 (13, 88) or those that are resistant to conventional chemotherapies (89) are also sensitive. Additional studies examining the effects of proteasome inhibitors in animal tumor models support the proapoptotic activities demonstrated in cell culture systems (Table 2; Refs. 10, 11, 14, 15, 90, 91, 92). Frankel et al. (93) found that when androgen-independent DU-145 prostate cancer cells are grown in spheroid cultures, a system that mimics the low growth fractions of prostate cancer in vivo(94), bortezomib treatment caused cell death in a greater proportion than when the same prostate cancer cells were grown in monolayers. In mouse xenograft tumor models, athymic mice bearing PC-3 tumors were treated with four weekly i.v. doses of bortezomib, and dose-dependent tumor size reductions were observed. When the drug was injected into the tumor on 4 consecutive days, 70% of the animals had no detectable tumor at the end of the study (11).

Proteasome activity may also overcome resistance mechanisms in response to chemotherapy and radiation and may augment antitumor activity of these agents. Proteasome inhibition may increase the effectiveness of these therapeutic modalities by either restoring sensitivity to radiation or chemotherapy or by showing additive or synergistic effect, allowing for decreased dosing and potentially reduced toxicity. Cooperative antitumor activity has been seen with bortezomib and the topoisomerase I inhibitor CPT-11 (irinotecan; Refs. 14, 15). One known topoisomerase inhibitor resistance mechanism in cancer cells is the up-regulation of topoisomerase I/topoisomerase II proteasomal degradation (95). However, proteasome inhibition can block solid tumor resistance to topoisomerase II inhibitors (96). In addition, bortezomib pretreatment blocked CPT-11-mediated NFκB DNA binding activity in colon cancer cells (14). This presumably occurs through the bortezomib-mediated stabilization of the proteasomal substrate IκBα. These experiments suggest that bortezomib may contribute to increase chemosensitivity of tumors to topoisomerase I inhibitors and may be a promising second-line or adjuvant treatment for CPT-11, VP16, or other topoisomerase inhibitors.

Bortezomib also enhanced xenograft radiosensitivity in a mouse colon tumor model and blocked radiation-induced NFκB DNA binding (90). Terminal transferase-mediated dUTP nick-end labeling analysis of the xenografts for apoptosis revealed that 4% of tumor cells were undergoing apoptosis in control mice, compared with 18% in mice treated with 6 Gy of radiation, 26% in mice treated with 1 mg/kg of bortezomib, and 72% in mice treated with both therapies (90). Pervan et al. (12) also showed enhanced radiosensitivity by proteasome inhibition in the TRAMP-C1 prostate cancer tumor model. Mice with TRAMP-C1 tumors received either bortezomib or placebo before radiation treatment. Animals receiving bortezomib treatment showed enhanced radiosensitivity and delayed tumor growth compared with controls. Considered together, these data suggest that bortezomib sensitizes cells to the effects of radiation and increases tumor cell apoptosis in mice treated with both therapies.

Other studies suggest that bortezomib may enhance the effectiveness of other classes of chemotherapy agents. Mice with Mia-PaCa-2 xenografts treated with gemcitabine had an average of 59% tumor size reduction, whereas mice receiving the combined bortezomib-gemcitabine therapy saw an average tumor volume loss of 75%. Relatively high doses of bortezomib (1 μm) induced apoptosis in the cells in vitro by reducing Bcl-2 transcription without an effect on Bax or Bak protein levels. However, low doses of the drug (10 nm) appeared to enhance gemcitabine-induced apoptosis. Schedule-dependent modulation has also been seen with bortezomib and taxanes (97, 98, 99). Cells treated with bortezomib before taxane administration had lower apoptotic indices (98), whereas taxane treatment preceding proteasome inhibition appeared to preserve the proapoptotic effects of taxanes in some cancer cells (97, 98, 99). Synergy between the two agents may not be present, because paclitaxel has been shown to induce phosphorylation and subsequent proteasomal degradation of Bcl-2 in NIH-OVCAR-3 cells (24). However, it is likely that this proposed abrogation of Bcl-2 phosphorylation by proteasome inhibitors is only a partial explanation for schedule-dependent synergy between bortezomib and taxanes, because Bcl-2 overexpression does not block bortezomib-induced apoptosis in prostate cells (13). We showed that treatment with either simultaneous bortezomib and paclitaxel or bortezomib followed by paclitaxel did not change the proapoptotic rate of PC-3 cells compared with bortezomib alone and was associated with moderate down-regulation of microtubule-associated protein 4 and marked down-regulation of β-tubulin with concomitant increase of unbound paclitaxel. By contrast, pretreatment of PC-3 cells with paclitaxel followed by bortezomib decreased their apoptotic rate by 53%, while preserving the up-regulated expression of both microtubule-associated protein 4 and β-tubulin, suggesting a cytoprotective effect of this treatment sequence (100).

Both prednisone (101) and dexamethasone (102, 103) have shown some antitumor activity in patients with AIPC, but the mechanisms responsible for their effects are not fully known. Dexamethasone treatment, however, is known to affect IκBα and IL-6 regulation in multiple myeloma, a cancer that is highly sensitive to bortezomib (104), and both these mechanisms may be important in prostate cancer (48). In addition, bortezomib induced apoptosis in myeloma cells from patients with dexamethasone-resistant disease (89). In AIPC lines, dexamethasone inhibited proliferation, and this coincided with an increase of IκBα protein, cytosolic accumulation of NFκB, and decreased secretion of IL-6 (105).

Differential Sensitivity to Proteasome Inhibition.

An important observation that appears to be consistent across many tumor types and proteasome inhibitors is that tumor cells seem to be more sensitive to proteasome inhibitors than normal cells (25). Myeloma cells isolated from patients are ∼1000 times more sensitive to bortezomib-induced apoptosis than normal plasma cells of patients (89). Human leukemia cells that have been induced to differentiate are significantly less sensitive to the apoptotic effects of proteasome inhibitors than their rapidly proliferating precursors (106). Another line of evidence suggests that the induction of growth arrest of cancer cells in culture does not make cells less susceptible to bortezomib-induced apoptosis (107). Thus, it is unlikely that hypersensitivity of cancer cells to proteasome inhibition can be explained solely by their accelerated rate of division, because proteasome inhibition promotes apoptosis in slowly dividing prostate cancer cells grown in monolayer cultures and spheroids (93).

Early Clinical Experience with Bortezomib in AIPC.

Seminal Phase I clinical trials have suggested that bortezomib has biological activity in advanced solid tumors and hematological malignancies with manageable toxicities (19). A Phase I study conducted at the M. D. Anderson Cancer Center at the University of Texas examined bortezomib in 53 patients with advanced solid tumors (17). Forty-eight of these patients were elderly men with AIPC. The median age of the patients was 66 years; 39 had received prior chemotherapy. Patients were treated with i.v. bortezomib from 0.13 to 2.0 mg/m2, once weekly for 4 weeks every 5 weeks. Thirty-one of 53 patients enrolled completed at least two cycles of bortezomib; 39 patients completed at least one cycle of treatment. In addition to monitoring toxicity, measurable disease response, serum IL-6, and prostate-specific antigen levels, 20S proteasome inhibition was measured in patients pre- and postdosing to examine possible correlations among bortezomib dose, proteasome inhibition, toxicity, and tumor response. The most frequent adverse events on this schedule included diarrhea, fatigue/weakness, constipation, nausea, vomiting, other gastrointestinal complaints, hypotension, dizziness, hypertension, and neuropathy. The dose-limiting toxicities included grade 3 diarrhea and grade 3 postural hypotension at the 2.0 mg/m2 dose level (Table 3). In studies using a twice-weekly treatment regimen, peripheral sensory neuropathy was frequent; however, neuropathy was less common on the once-weekly schedule, occurring in 8% of patients (2% grade 3), although many patients had received potentially neurotoxic drugs previously.

Dose-related inhibition of whole-blood proteasome activity was observed, with most toxicity (diarrhea, hypotension, and fatigue) seen at doses (≥1.45 mg/m2) that yielded ≥70% proteasome inhibition 1 h after treatment. The maximum tolerated dose in this schedule was 1.6 mg/m2. Antitumor responses (measurable disease and prostate-specific antigen responses) were seen in 2 and 3 patients, respectively, among the 48 patients with AIPC. One of the 2 objective partial responses and all 3 of the prostate-specific antigen responses occurred at doses resulting in average 1-h postbortezomib whole-blood 20S proteasome inhibition of ≥70%, which also is associated with dose-limiting toxicity, suggesting a narrow therapeutic index with monotherapy in prostate carcinoma in this study. Proteasome inhibition was associated with decline in serum IL-6 levels, suggesting that bortezomib may be affecting NFκB signaling and autocrine/paracrine mechanisms in vivo. Additional studies of bortezomib in patients with prostate cancer are warranted, and we are focusing on combination treatments with cytotoxic agents that may reverse resistance to chemotherapy.

Two additional single-agent Phase I trials of bortezomib, one in solid tumors (19) and another in hematological malignancies (16), have been published that support additional studies of this agent for the treatment of cancer. Whereas these studies were not designed to assess efficacy, clinically significant responses were observed. Bortezomib has demonstrated significant biological activity in a Phase II study of relapsed and refractory myeloma patients (108) and has received United States Food and Drug Administration approval for patients with relapsed or refractory multiple myeloma who have received at least two prior therapies and have progressed on their last therapy (109). Additional trials studying bortezomib as monotherapy and in combination with other antineoplastic agents are ongoing in multiple myeloma and in other types of malignancy. Most are Phase I exploratory studies, and promising results are beginning to emerge (Table 4; Refs. 108, 110, 111, 112, 113, 114, 115). A Phase III randomized trial of the treatment of multiple myeloma with bortezomib monotherapy versus dexamethasone was halted recently at the interim analysis because of significantly greater efficacy in the bortezomib arm. Study analyses are ongoing.

The proteasome plays a central role in regulation of the cell cycle, proliferation, cell death, angiogenesis, metastasis, and resistance to chemotherapy and radiation therapy. Results from cell culture systems, animal tumor models, and early clinical studies suggest that proteasome inhibition represents a new therapy target for human cancers, including AIPC. Bortezomib is the first agent of this novel class to enter clinical trials, and antitumor activity has been reported from preclinical investigations in a variety of tumor models. In some diseases, like multiple myeloma, antitumor activity is significant even as a single agent, and bortezomib has been approved for the treatment of patients with multiple myeloma who have received at least two prior therapies and have demonstrated disease progression on their last therapy. In AIPC, bortezomib seems to have weak single agent antitumor activity. Preclinical models suggest synergy with some conventional chemotherapeutic agents with activity against AIPC. Additional studies are under way to additionally examine the role of bortezomib in combination with chemotherapy in AIPC as well as other cancers in which the proteasome has an active role in transformation, chemoresistance, and disease progression.

Grant support: CaPCURE and Millennium Inc. (trial support).

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.

Requests for reprints: Christos N. Papandreou, Genitourinary Medical Oncology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030. Phone: (713) 792-2830; Fax: (713) 745-1625; E-mail: cpapandr@notes.mdacc.tmc.edu

Fig. 1.

Cross-sectional view of the 20S proteasome. Cross-section of the proteasome β ring, which contains the three active sites. Bortezomib binds and inhibits the chymotryptic site. The function of the other four β subunits is unclear.

Fig. 1.

Cross-sectional view of the 20S proteasome. Cross-section of the proteasome β ring, which contains the three active sites. Bortezomib binds and inhibits the chymotryptic site. The function of the other four β subunits is unclear.

Close modal
Fig. 2.

Nuclear factor κB (NF-κB) activation pathway. Viruses, growth factors, antigens, radiation, or chemotherapeutic drugs activate signaling pathways that lead to the degradation of inhibitor of nuclear factor-κB (IκB) by the proteasome. Once released from IκB inhibition, NF-κB translocates to the nucleus to activate transcription of genes that protect the cell from apoptosis and promote cell growth and differentiation. The synthesis of growth factors, cell-adhesion molecules, angiogenesis factors, and antiapoptotic factors is increased.

Fig. 2.

Nuclear factor κB (NF-κB) activation pathway. Viruses, growth factors, antigens, radiation, or chemotherapeutic drugs activate signaling pathways that lead to the degradation of inhibitor of nuclear factor-κB (IκB) by the proteasome. Once released from IκB inhibition, NF-κB translocates to the nucleus to activate transcription of genes that protect the cell from apoptosis and promote cell growth and differentiation. The synthesis of growth factors, cell-adhesion molecules, angiogenesis factors, and antiapoptotic factors is increased.

Close modal
Fig. 3.

Important cellular mediators of prostate cancer growth. PTEN and HER-2/neu regulate the phosphatidylinositol 3′-kinase (PI3-Kinase)/Akt signaling pathway, which, in turn, regulates the activity of important mediators of proliferation, survival, and apoptosis. NF-κB, nuclear factor κB.

Fig. 3.

Important cellular mediators of prostate cancer growth. PTEN and HER-2/neu regulate the phosphatidylinositol 3′-kinase (PI3-Kinase)/Akt signaling pathway, which, in turn, regulates the activity of important mediators of proliferation, survival, and apoptosis. NF-κB, nuclear factor κB.

Close modal
Table 1

Genotypes for common human prostate cancer cell lines

Cell lineTissue of originAndrogen- dependentNFκBa constitutive activationGenotype
Rbp53PTEN
LNCaP Lymph node Yes Yes +/+ +/+ −/− 
PC-3 Bone marrow No Yes −/− −/− −/− 
DU145 Brain tumor No Yes +/+ −/− +/+ 
Cell lineTissue of originAndrogen- dependentNFκBa constitutive activationGenotype
Rbp53PTEN
LNCaP Lymph node Yes Yes +/+ +/+ −/− 
PC-3 Bone marrow No Yes −/− −/− −/− 
DU145 Brain tumor No Yes +/+ −/− +/+ 
a

NFκB, nuclear factor κB.

Table 2

Antitumor activities of bortezomib in solid tumor models

CellsAnimal model and dosing scheduleAntitumor activity
Human prostate cancer, PC-3 (11) Athymic nude mice, weekly ×4 60% decrease in tumor volume 
Human MIA-PACa-2 pancreatic cancer (10) Athymic nude mice, twice weekly ×3.5 75% growth inhibition with combination with bortezomib and gemcitabine versus 59% growth inhibition with gemcitabine alone 
Human pancreatic cancer, BxPc-3 (15) Athymic nude mice, weekly ×4 89% growth inhibition with combination bortezomib and CPT-11 versus 43% with CPT-11 alone 
Human LOVO colon cancer (14) Athymic nude mice, twice weekly 94% decrease in tumor size with combination bortezomib and CPT-11 versus 48% with CPT-11 alone 
Human LOVO colon cancer (90) Athymic nude mice, 6 h before irradiation 7–41% increase in radiosensitivity with bortezomib leading to an 84% reduction in tumor volume 
Squamous cell carcinoma: murine PAM-LY2 (91) Balb/C SCID mice, three times weekly 50–80% dose-dependent decrease in tumor volume 
Murine lung cancer and human breast cancer (92) Lewis lung model, EMT-6 mouse model; daily treatment Lewis lung model: growth delay and decreased number of lung metastases alone and in combination (5-fluorouracil, cisplatin, paclitaxel, or docetaxel); EMT-6: dose-dependent cytotoxicity alone and potentiated tumor cell killing with cyclophosphamide, allowing dose reduction 
CellsAnimal model and dosing scheduleAntitumor activity
Human prostate cancer, PC-3 (11) Athymic nude mice, weekly ×4 60% decrease in tumor volume 
Human MIA-PACa-2 pancreatic cancer (10) Athymic nude mice, twice weekly ×3.5 75% growth inhibition with combination with bortezomib and gemcitabine versus 59% growth inhibition with gemcitabine alone 
Human pancreatic cancer, BxPc-3 (15) Athymic nude mice, weekly ×4 89% growth inhibition with combination bortezomib and CPT-11 versus 43% with CPT-11 alone 
Human LOVO colon cancer (14) Athymic nude mice, twice weekly 94% decrease in tumor size with combination bortezomib and CPT-11 versus 48% with CPT-11 alone 
Human LOVO colon cancer (90) Athymic nude mice, 6 h before irradiation 7–41% increase in radiosensitivity with bortezomib leading to an 84% reduction in tumor volume 
Squamous cell carcinoma: murine PAM-LY2 (91) Balb/C SCID mice, three times weekly 50–80% dose-dependent decrease in tumor volume 
Murine lung cancer and human breast cancer (92) Lewis lung model, EMT-6 mouse model; daily treatment Lewis lung model: growth delay and decreased number of lung metastases alone and in combination (5-fluorouracil, cisplatin, paclitaxel, or docetaxel); EMT-6: dose-dependent cytotoxicity alone and potentiated tumor cell killing with cyclophosphamide, allowing dose reduction 
Table 3

Principal toxic effects during bortezomib treatment in androgen-independent prostate cancer (17) 

Any grade toxicity All cycles Patients (%)Grade 3 toxicitya Cycle 1 Patients (%)
Parameter n = 53 n = 53 
Likely related to bortezomib   
Diarrhea 28 (53) 3 (6) 
Fatigue 27 (51) 1 (2) 
Constipation 25 (47) 1 (2) 
Nausea 22 (42) 
Vomiting 18 (34) 
Weakness 14 (26) 2 (4) 
Abdominal pain 13 (25) 
Appetite decreased 12 (23) 
Hypotension 10 (19) 3 (6) 
Dizziness 8 (15) 
Abdominal distension 7 (13) 
Hypertension 6 (11) 1 (2) 
Flatulence 6 (11) 
Neuropathy 4 (8) 1 (2) 
Likely unrelated to bortezomib   
Back pain 15 (28) 
Catheter related complication 15 (28) 
Bone pain 13 (25) 
Pyrexia 13 (25) 
Arthralgia 11 (21) 
Headache 11 (21) 
Urinary tract infection 11 (21) 
Dyspnea 9 (17) 
Anemia 8 (15) 
Cardiac murmur 8 (15) 
Hematuria 8 (15) 
Hypomagnesemia 7 (13) 
Edema lower limb 7 (13) 1 (2) (due to DVT) 
Pain in limb 7 (13) 
Nocturia 6 (11)  
Rigors 6 (11)  
Urinary frequency 6 (11)  
Any grade toxicity All cycles Patients (%)Grade 3 toxicitya Cycle 1 Patients (%)
Parameter n = 53 n = 53 
Likely related to bortezomib   
Diarrhea 28 (53) 3 (6) 
Fatigue 27 (51) 1 (2) 
Constipation 25 (47) 1 (2) 
Nausea 22 (42) 
Vomiting 18 (34) 
Weakness 14 (26) 2 (4) 
Abdominal pain 13 (25) 
Appetite decreased 12 (23) 
Hypotension 10 (19) 3 (6) 
Dizziness 8 (15) 
Abdominal distension 7 (13) 
Hypertension 6 (11) 1 (2) 
Flatulence 6 (11) 
Neuropathy 4 (8) 1 (2) 
Likely unrelated to bortezomib   
Back pain 15 (28) 
Catheter related complication 15 (28) 
Bone pain 13 (25) 
Pyrexia 13 (25) 
Arthralgia 11 (21) 
Headache 11 (21) 
Urinary tract infection 11 (21) 
Dyspnea 9 (17) 
Anemia 8 (15) 
Cardiac murmur 8 (15) 
Hematuria 8 (15) 
Hypomagnesemia 7 (13) 
Edema lower limb 7 (13) 1 (2) (due to DVT) 
Pain in limb 7 (13) 
Nocturia 6 (11)  
Rigors 6 (11)  
Urinary frequency 6 (11)  
a

No grade 4 toxicity was observed during the study.

DVT, Deep Vein Thrombosis.

Table 4

Clinical activity of single-agent bortezomib

Tumor typeDose schedule (intravenous bolus)Activity
Relapsed, refractory multiple myeloma (108) 1.3 mg/m2 twice weekly every 3 weeks for up to 8 cycles 35% overall response rate (complete response + partial response + minimal response) by European Blood and Marrow Transplantation criteria (110); 18% clinical remission rate by Southwest Oncology Group criteria 
Malignant lymphoma (111, 112) 1.5 mg/m2 twice weekly every 3 weeks Promising activity, particularly in mantle cell lymphoma, in patients with relapsed disease (studies are ongoing) 
Metastatic renal cell carcinoma 1.3–1.5 mg/m2 twice weekly every 3 weeks 3 durable partial responses among 32 treated patients in an ongoing trial (113); 1 partial response in 21 evaluable patients (114) 
Non-small cell lung carcinoma 1.5 mg/m2 twice weekly every 3 weeks (115) 1 partial response and 2 patients with stable disease among 8 evaluable patients (115); 1 partial response in 8 patients enrolled in a dose-escalation study (19) 
Metastatic androgen-independent prostate cancer 0.13–2.0 mg/m2 weekly ×4 wks q 5 wks 2 radiographic partial responses in lymph nodes and 3 PSA partial responses among 48 treated patients (17). All responses occurred at doses resulting in average 1-h post-bortezomib whole-blood 20S proteasome inhibition ≥70%, which was also associated with a decline in serum interleukin 6 levels 
Tumor typeDose schedule (intravenous bolus)Activity
Relapsed, refractory multiple myeloma (108) 1.3 mg/m2 twice weekly every 3 weeks for up to 8 cycles 35% overall response rate (complete response + partial response + minimal response) by European Blood and Marrow Transplantation criteria (110); 18% clinical remission rate by Southwest Oncology Group criteria 
Malignant lymphoma (111, 112) 1.5 mg/m2 twice weekly every 3 weeks Promising activity, particularly in mantle cell lymphoma, in patients with relapsed disease (studies are ongoing) 
Metastatic renal cell carcinoma 1.3–1.5 mg/m2 twice weekly every 3 weeks 3 durable partial responses among 32 treated patients in an ongoing trial (113); 1 partial response in 21 evaluable patients (114) 
Non-small cell lung carcinoma 1.5 mg/m2 twice weekly every 3 weeks (115) 1 partial response and 2 patients with stable disease among 8 evaluable patients (115); 1 partial response in 8 patients enrolled in a dose-escalation study (19) 
Metastatic androgen-independent prostate cancer 0.13–2.0 mg/m2 weekly ×4 wks q 5 wks 2 radiographic partial responses in lymph nodes and 3 PSA partial responses among 48 treated patients (17). All responses occurred at doses resulting in average 1-h post-bortezomib whole-blood 20S proteasome inhibition ≥70%, which was also associated with a decline in serum interleukin 6 levels 
1
Denmeade SR, Lin XS, Isaacs JT. Role of programmed (apoptotic) cell death during the progression and therapy for prostate cancer.
Prostate
,
28
:
251
-65,  
1996
.
2
Waltregny D, Leav I, Signoretti S, et al Androgen-driven prostate epithelial cell proliferation and differentiation in vivo involve the regulation of p27.
Mol Endocrinol
,
15
:
765
-82,  
2001
.
3
Cunha GR, Donjacour A. Stromal-epithelial interactions in normal and abnormal prostatic development.
Prog Clin Biol Res
,
239
:
251
-72,  
1987
.
4
Grossmann ME, Huang H, Tindall DJ. Androgen receptor signaling in androgen-refractory prostate cancer.
J Natl Cancer Inst
,
93
:
1687
-97,  
2001
.
5
Culig Z, Bartsch G, Hobisch A. Interleukin-6 regulates androgen receptor activity and prostate cancer cell growth.
Mol Cell Endocrinol
,
197
:
231
-8,  
2002
.
6
Hobisch A, Eder IE, Putz T, et al Interleukin-6 regulates prostate-specific protein expression in prostate carcinoma cells by activation of the androgen receptor.
Cancer Res
,
58
:
4640
-5,  
1998
.
7
Orio F, Terouanne B, Georget V, et al Potential action of IGF-1 and EGF on androgen receptor nuclear transfer and transactivation in normal and cancer human prostate cell lines.
Mol Cell Endocrinol
,
198
:
105
-14,  
2002
.
8
Fribourg AF, Knudsen KE, Strobeck MW, Lindhorst CM, Knudsen ES. Differential requirements for ras and the retinoblastoma tumor suppressor protein in the androgen dependence of prostatic adenocarcinoma cells.
Cell Growth Differ
,
11
:
361
-72,  
2000
.
9
Navarro D, Luzardo OP, Fernandez L, Chesa N, Diaz-Chico BN. Transition to androgen-independence in prostate cancer.
J Steroid Biochem Mol Biol
,
81
:
191
-201,  
2002
.
10
Bold RJ, Virudachalam S, McConkey DJ. Chemosensitization of pancreatic cancer by inhibition of the 26S proteasome.
J Surg Res
,
100
:
11
-7,  
2001
.
11
Adams J, Palombella VJ, Sausville EA, et al Proteasome inhibitors: a novel class of potent and effective antitumor agents.
Cancer Res
,
59
:
2615
-22,  
1999
.
12
Pervan M, Pajonk F, Sun JR, Withers HR, McBride WH. Molecular pathways that modify tumor radiation response.
Am J Clin Oncol
,
24
:
481
-5,  
2001
.
13
Herrmann JL, Briones F, Jr, 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
:
2889
-99,  
1998
.
14
Cusack JC, Jr, Liu R, Houston M, et al Enhanced chemosensitivity to CPT-11 with proteasome inhibitor PS-341: implications for systemic nuclear factor-κB inhibition.
Cancer Res
,
61
:
3535
-40,  
2001
.
15
Shah SA, Potter MW, McDade TP, et al 26S proteasome inhibition induces apoptosis and limits growth of human pancreatic cancer.
J Cell Biochem
,
82
:
110
-22,  
2001
.
16
Orlowski RZ, Stinchcombe TE, Mitchell BS, et al Phase I trial of the proteasome inhibitor PS-341 in patients with refractory hematologic malignancies.
J Clin Oncol
,
20
:
4420
-7,  
2002
.
17
Papandreou C, Daliani D, Millikan RE, et al Phase I trial of the proteasome inhibitor bortezomib (PS-341) in patients with advanced solid tumors with observations in androgen-independent prostate cancer.
J Clin Oncol
,
22
:
2108
-21,  
2004
.
18
Logothetis CJ, Yang H, Daliani D, et al Dose-dependent inhibition of 20S proteasome results in serum IL-6 and PSA decline in patients (pts) with androgen-independent prostate cancer (AI PCa) treated with the proteasome inhibitor PS-341.
Proc Am Soc Clin Oncol
,
20
:
186a
2001
.
19
Aghajanian C, Soignet S, Dizon DS, et al A phase I trial of the novel proteasome inhibitor PS341 in advanced solid tumor malignancies.
Clin Cancer Res
,
8
:
2505
-11,  
2002
.
20
Arrigo AP, Tanaka K, Goldberg AL, Welch WJ. Identity of the 19S ‘prosome’ particle with the large multifunctional protease complex of mammalian cells (the proteasome).
Nature
,
331
:
192
-4,  
1988
.
21
Dahlmann B, Kopp F, Kuehn L, et al The multicatalytic proteinase (prosome) is ubiquitous from eukaryotes to archaebacteria.
FEBS Lett
,
251
:
125
-31,  
1989
.
22
Pagano M. Cell cycle regulation by the ubiquitin pathway.
FASEB J
,
11
:
1067
-75,  
1997
.
23
Li B, Dou QP. Bax degradation by the ubiquitin/proteasome-dependent pathway: involvement in tumor survival and progression.
Proc Natl Acad Sci USA
,
97
:
3850
-5,  
2000
.
24
Chadebech P, Brichese L, Baldin V, Vidal S, Valette A. Phosphorylation and proteasome-dependent degradation of Bcl-2 in mitotic-arrested cells after microtubule damage.
Biochem Biophys Res Commun
,
262
:
823
-7,  
1999
.
25
Orlowski 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
:
4342
-8,  
1998
.
26
Maki CG, Huibregtse JM, Howley PM. In vivo ubiquitination and proteasome-mediated degradation of p53(1).
Cancer Res
,
56
:
2649
-54,  
1996
.
27
Bounpheng MA, Dimas JJ, Dodds SG, Christy BA. Degradation of Id proteins by the ubiquitin-proteasome pathway.
FASEB J
,
13
:
2257
-64,  
1999
.
28
Palombella VJ, Rando OJ, Goldberg AL, Maniatis T. The ubiquitin-proteasome pathway is required for processing the NF-κB1 precursor protein and the activation of NF-κ B.
Cell
,
78
:
773
-85,  
1994
.
29
Roth AF, Davis NG. Ubiquitination of the PEST-like endocytosis signal of the yeast a-factor receptor.
J Biol Chem
,
275
:
8143
-53,  
2000
.
30
Marchal C, Haguenauer-Tsapis R, Urban-Grimal D. A PEST-like sequence mediates phosphorylation and efficient ubiquitination of yeast uracil permease.
Mol Cell Biol
,
18
:
314
-21,  
1998
.
31
Conaway RC, Brower CS, Conaway JW. Emerging roles of ubiquitin in transcription regulation.
Science
,
296
:
1254
-8,  
2002
.
32
Lee PL, Midelfort CF, Murakami K, Hatcher VB. Multiple forms of ubiquitin-protein ligase. Binding of activated ubiquitin to protein substrates.
Biochemistry
,
25
:
3134
-8,  
1986
.
33
Hershko A, Ciechanover A. The ubiquitin system.
Annu Rev Biochem
,
67
:
425
-79,  
1998
.
34
Eytan E, Ganoth D, Armon T, Hershko A. ATP-dependent incorporation of 20S protease into the 26S complex that degrades proteins conjugated to ubiquitin.
Proc Natl Acad Sci USA
,
86
:
7751
-5,  
1989
.
35
Driscoll J, Goldberg AL. The proteasome (multicatalytic protease) is a component of the 1500-kDa proteolytic complex which degrades ubiquitin-conjugated proteins.
J Biol Chem
,
265
:
4789
-92,  
1990
.
36
Lupas A, Koster AJ, Baumeister W. Structural features of 26S and 20S proteasomes.
Enzyme Protein
,
47
:
252
-73,  
1993
.
37
Tokunaga F, Goto T, Koide T, et al ATP- and antizyme-dependent endoproteolysis of ornithine decarboxylase to oligopeptides by the 26 S proteasome.
J Biol Chem
,
269
:
17382
-5,  
1994
.
38
Kisselev AF, Goldberg AL. Proteasome inhibitors: from research tools to drug candidates.
Chem Biol
,
8
:
739
-58,  
2001
.
39
Adams J, Behnke M, Chen S, et al Potent and selective inhibitors of the proteasome: dipeptidyl boronic acids.
Bioorg Med Chem Lett
,
8
:
333
-8,  
1998
.
40
Nussbaum AK, Dick TP, Keilholz W, et al Cleavage motifs of the yeast 20S proteasome β subunits deduced from digests of enolase 1.
Proc Natl Acad Sci USA
,
95
:
12504
-9,  
1998
.
41
Kimura K, Bowen C, Spiegel S, Gelmann EP. Tumor necrosis factor-α sensitizes prostate cancer cells to γ-irradiation-induced apoptosis.
Cancer Res
,
59
:
1606
-14,  
1999
.
42
Kimura K, Markowski M, Bowen C, Gelmann EP. Androgen blocks apoptosis of hormone-dependent prostate cancer cells.
Cancer Res
,
61
:
5611
-8,  
2001
.
43
Buchanan G, Greenberg NM, Scher HI, Harris JM, Marshall VR, Tilley WD. Collocation of androgen receptor gene mutations in prostate cancer.
Clin Cancer Res
,
7
:
1273
-81,  
2001
.
44
Marcelli M, Ittmann M, Mariani S, et al Androgen receptor mutations in prostate cancer.
Cancer Res
,
60
:
944
-9,  
2000
.
45
Taplin ME, Bubley GJ, Shuster TD, et al Mutation of the androgen-receptor gene in metastatic androgen-independent prostate cancer.
N Engl J Med
,
332
:
1393
-8,  
1995
.
46
Krishnan AV, Zhao XY, Swami S, et al A glucocorticoid-responsive mutant androgen receptor exhibits unique ligand specificity: therapeutic implications for androgen-independent prostate cancer.
Endocrinology
,
143
:
1889
-900,  
2002
.
47
Gioeli D, Ficarro SB, Kwiek JJ, et al Androgen receptor phosphorylation. Regulation and identification of the phosphorylation sites.
J Biol Chem
,
277
:
29304
-14,  
2002
.
48
Smith PC, Hobisch A, Lin DL, Culig Z, Keller ET. Interleukin-6 and prostate cancer progression.
Cytokine Growth Factor Rev
,
12
:
33
-40,  
2001
.
49
Keller ET, Chang C, Ershler WB. Inhibition of NFκB activity through maintenance of IκBα levels contributes to dihydrotestosterone-mediated repression of the interleukin-6 promoter.
J Biol Chem
,
271
:
26267
-75,  
1996
.
50
Ueda T, Bruchovsky N, Sadar MD. Activation of the androgen receptor N-terminal domain by interleukin-6 via MAPK and STAT3 signal transduction pathways.
J Biol Chem
,
277
:
7076
-85,  
2002
.
51
Inoue K, Slaton JW, Eve BY, et al Interleukin 8 expression regulates tumorigenicity and metastases in androgen-independent prostate cancer.
Clin Cancer Res
,
6
:
2104
-19,  
2000
.
52
Kim SJ, Uehara H, Karashima T, Mccarty M, Shih N, Fidler IJ. Expression of interleukin-8 correlates with angiogenesis, tumorigenicity, and metastasis of human prostate cancer cells implanted orthotopically in nude mice.
Neoplasia
,
3
:
33
-42,  
2001
.
53
Borre M, Nerstrom B, Overgaard J. Association between immunohistochemical expression of vascular endothelial growth factor (VEGF), VEGF-expressing neuroendocrine-differentiated tumor cells, and outcome in prostate cancer patients subjected to watchful waiting.
Clin Cancer Res
,
6
:
1882
-90,  
2000
.
54
Chen C, Edelstein LC, Gelinas C. The Rel/NF-κB family directly activates expression of the apoptosis inhibitor Bcl-x(L).
Mol Cell Biol
,
20
:
2687
-3695,  
2000
.
55
Wang CY, Mayo MW, Korneluk RG, et al NF-κB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation.
Science
,
281
:
1680
-3,  
1998
.
56
Wang CY, Guttridge DC, Mayo MW, et al NF-κB induces expression of the Bcl-2 homologue A1/Bfl-1 to preferentially suppress chemotherapy-induced apoptosis.
Mol Cell Biol
,
19
:
5923
-9,  
1999
.
57
Zong WX, Edelstein LC, Chen C, Bash J, Gelinas C. The prosurvival Bcl-2 homolog Bfl-1/A1 is a direct transcriptional target of NF-κB that blocks TNFα-induced apoptosis.
Genes Dev
,
13
:
382
-7,  
1999
.
58
Kim JY, Lee S, Hwangho B. NF-κB activation is related to the resistance of lung cancer cells to TNF-α-induced apoptosis.
Biochem Biophys Res Commun
,
273
:
140
-6,  
2000
.
59
Huang Y, Johnson KR, Norris JS, Fan W. Nuclear factor-κB/IκB signaling pathway may contribute to the mediation of paclitaxel-induced apoptosis in solid tumor cells.
Cancer Res
,
60
:
4426
-32,  
2000
.
60
Andela VB, Schwarz EM, Puzas JE, O’Keefe RJ, Rosier RN. Tumor metastasis and the reciprocal regulation of prometastatic and antimetastatic factors by nuclear factor κB.
Cancer Res
,
60
:
6557
-62,  
2000
.
61
Damiano JS, Cress AE, Hazlehurst LA, Shtil AA, Dalton WS. Cell adhesion mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human myeloma cell lines.
Blood
,
93
:
1658
-67,  
1999
.
62
Hazlehurst LA, Damiano JS, Buyuksal I, Pledger WJ, Dalton WS. Adhesion to fibronectin via β1 integrins regulates p27kip1 levels and contributes to cell adhesion mediated drug resistance (CAM-DR).
Oncogene
,
19
:
4319
-27,  
2000
.
63
Borset M, Waage A, Brekke OL, Helseth E. TNF and IL-6 are potent growth factors for OH-2, a novel human myeloma cell line.
Eur J Haematol
,
53
:
31
-7,  
1994
.
64
Huang H, Cheville JC, Pan Y, Roche PC, Schmidt LJ, Tindall DJ. PTEN induces chemosensitivity in PTEN-mutated prostate cancer cells by suppression of Bcl-2 expression.
J Biol Chem
,
276
:
38830
-6,  
2001
.
65
Lindholm PF, Bub J, Kaul S, Shidham VB, Kajdacsy-Balla A. The role of constitutive NF-κB activity in PC-3 human prostate cancer cell invasive behavior.
Clin Exp Metastasis
,
18
:
471
-9,  
2001
.
66
Sumitomo M, Tachibana M, Nakashima J, et al An essential role for nuclear factor kappa B in preventing TNF-α-induced cell death in prostate cancer cells.
J Urol
,
161
:
674
-9,  
1999
.
67
Muenchen HJ, Lin DL, Walsh MA, Keller ET, Pienta KJ. Tumor necrosis factor-α-induced apoptosis in prostate cancer cells through inhibition of nuclear factor-κB by an IκBα “super-repressor.”.
Clin Cancer Res
,
6
:
1969
-77,  
2000
.
68
Palayoor ST, Youmell MY, Calderwood SK, Coleman CN, Price BD. Constitutive activation of IκB kinase α and NF-κB in prostate cancer cells is inhibited by ibuprofen.
Oncogene
,
18
:
7389
-94,  
1999
.
69
Cusack JC, Liu R, Baldwin AS. NF-κB and chemoresistance: potentiation of cancer drugs via inhibition of NF-κB.
Drug Resist Updat
,
2
:
271
-3,  
1999
.
70
Hideshima T, Chauhan D, Richardson P, et al NF-κB as a therapeutic target in multiple myeloma.
J Biol Chem
,
277
:
16639
-47,  
2002
.
71
Li J, Yen C, Liaw D, et al PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer.
Science
,
275
:
1943
-7,  
1997
.
72
Verma RS, Manikal M, Conte RA, Godec CJ. Chromosomal basis of adenocarcinoma of the prostate.
Cancer Investig
,
17
:
441
-7,  
1999
.
73
Al Maghrabi J, Vorobyova L, Chapman W, Jewett M, Zielenska M, Squire JA. p53 Alteration and chromosomal instability in prostatic high-grade intraepithelial neoplasia and concurrent carcinoma: analysis by immunohistochemistry, interphase in situ hybridization, and sequencing of laser-captured microdissected specimens.
Mod Pathol
,
14
:
1252
-62,  
2001
.
74
Myers MP, Stolarov JP, Eng C, et al P-TEN, the tumor suppressor from human chromosome 10q23, is a dual-specificity phosphatase.
Proc Natl Acad Sci USA
,
94
:
9052
-7,  
1997
.
75
Graff JR, Konicek BW, McNulty AM, et al Increased AKT activity contributes to prostate cancer progression by dramatically accelerating prostate tumor growth and diminishing p27Kip1 expression.
J Biol Chem
,
275
:
24500
-5,  
2000
.
76
Lloyd RV, Erickson LA, Jin L, et al p27kip1: a multifunctional cyclin-dependent kinase inhibitor with prognostic significance in human cancers.
Am J Pathol
,
154
:
313
-23,  
1999
.
77
Persad S, Attwell S, Gray V, et al Inhibition of integrin-linked kinase (ILK) suppresses activation of protein kinase B/Akt and induces cell cycle arrest and apoptosis of PTEN-mutant prostate cancer cells.
Proc Natl Acad Sci USA
,
97
:
3207
-12,  
2000
.
78
Li P, Nicosia SV, Bai W. Antagonism between PTEN/MMAC1/TEP-1 and androgen receptor in growth and apoptosis of prostatic cancer cells.
J Biol Chem
,
276
:
20444
-50,  
2001
.
79
Lin HK, Yeh S, Kang HY, Chang C. Akt suppresses androgen-induced apoptosis by phosphorylating and inhibiting androgen receptor.
Proc Natl Acad Sci USA
,
98
:
7200
-5,  
2001
.
80
Manin M, Baron S, Goossens K, et al Androgen receptor expression is regulated by the phosphoinositide 3- kinase/Akt pathway in normal and tumoral epithelial cells.
Biochem J
,
366
:
729
-36,  
2002
.
81
Signoretti S, Montironi R, Manola J, et al Her-2-neu expression and progression toward androgen independence in human prostate cancer.
J Natl Cancer Inst
,
92
:
1918
-25,  
2000
.
82
Shi Y, Brands FH, Chatterjee S, et al Her-2/neu expression in prostate cancer: high level of expression associated with exposure to hormone therapy and androgen independent disease.
J Urol
,
166
:
1514
-9,  
2001
.
83
Osman I, Scher HI, Drobnjak M, et al HER-2/neu (p185neu) protein expression in the natural or treated history of prostate cancer.
Clin Cancer Res
,
7
:
2643
-7,  
2001
.
84
Wen Y, Hu MC, Makino K, et al HER-2/neu promotes androgen-independent survival and growth of prostate cancer cells through the Akt pathway.
Cancer Res
,
60
:
6841
-5,  
2000
.
85
Craft N, Shostak Y, Carey M, Sawyers CL. A mechanism for hormone-independent prostate cancer through modulation of androgen receptor signaling by the HER-2/neu tyrosine kinase.
Nat Med
,
5
:
280
-5,  
1999
.
86
Pianetti S, Arsura M, Romieu-Mourez R, Coffey RJ, Sonenshein GE. Her-2/neu overexpression induces NF-κB via a PI3-kinase/Akt pathway involving calpain-mediated degradation of IκBα that can be inhibited by the tumor suppressor PTEN.
Oncogene
,
20
:
1287
-99,  
2001
.
87
Zhou BP, Hu MC, Miller SA, et al HER-2/neu blocks tumor necrosis factor-induced apoptosis via the Akt/NF-κB pathway.
J Biol Chem
,
275
:
8027
-31,  
2000
.
88
Feinman R, Aris VM, Vergano S, et al Transcriptional gene expression profiles in drug-sensitive and Bcl-2-resistant myeloma cells in response to dexamethasone, arsenic trioxide, and PS-341.
Blood
,
98
:
368a
2001
.
89
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
:
3071
-6,  
2001
.
90
Russo SM, Tepper JE, Baldwin AS, Jr, et al Enhancement of radiosensitivity by proteasome inhibition: implications for a role of NF-κB.
Int J Radiat Oncol Biol Phys
,
50
:
183
-93,  
2001
.
91
Sunwoo JB, Chen Z, Dong G, et al Novel proteasome inhibitor PS-341 inhibits activation of nuclear factor-κB, cell survival, tumor growth, and angiogenesis in squamous cell carcinoma.
Clin Cancer Res
,
7
:
1419
-28,  
2001
.
92
Teicher BA, Ara G, Herbst R, Palombella VJ, Adams J. The proteasome inhibitor PS-341 in cancer therapy.
Clin Cancer Res
,
5
:
2638
-45,  
1999
.
93
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
:
3719
-28,  
2000
.
94
Berges RR, Vukanovic J, Epstein JI, et al Implication of cell kinetic changes during the progression of human prostatic cancer.
Clin Cancer Res
,
1
:
473
-80,  
1995
.
95
Desai SD, Liu LF, Vazquez-Abad D, D’Arpa P. Ubiquitin-dependent destruction of topoisomerase I is stimulated by the antitumor drug camptothecin.
J Biol Chem
,
272
:
24159
-64,  
1997
.
96
Ogiso Y, Tomida A, Lei S, Omura S, Tsuruo T. Proteasome inhibition circumvents solid tumor resistance to topoisomerase II-directed drugs.
Cancer Res
,
60
:
2429
-34,  
2000
.
97
McConkey DJ, Williams SA, Logothetis C, Papandreou CN. Preclinical evaluation of proteasome inhibitor-based combination chemotherapy in prostate cancer.
Proc Am Assoc Cancer Res
,
42
:
229
2001
.
98
Williams SA, Papandreou C, McConkey D. Preclinical effects of proteasome inhibitor PS-341 in combination chemotherapy for prostate cancer.
Proc Am Soc Clin Oncol
,
20
:
169b
2001
.
99
Gumerlock PH, Moisan LP, Lau AH, Mack PC, Lara PN, Gandara DR. Docetaxel followed by PS-341 results in phosphorylation and stabilization of p27 and increases response in non-small cell lung carcinoma (NSCLC).
Clin Cancer Res
,
7
:
3809S
-10S,  
2001
.
100
Yang H, Assikis V, Daliani D, Logothetis CJ, Adams J, Papandreou CN. The proteasome inhibitor PS-341 alters the chemosensitivity of prostate cancer cells.
Proc Am Assoc Cancer Res
,
44
:
6526
2003
.
101
Tannock IF, Osoba D, Stockler MR, et al Chemotherapy with mitoxantrone plus prednisone or prednisone alone for symptomatic hormone-resistant prostate cancer: a Canadian randomized trial with palliative end points.
J Clin Oncol
,
14
:
1756
-64,  
1996
.
102
Morioka M, Kobayashi T, Furukawa Y, et al Prostate-specific antigen levels and prognosis in patients with hormone- refractory prostate cancer treated with low-dose dexamethasone.
Urol Int
,
68
:
10
-5,  
2002
.
103
Saika T, Kusaka N, Tsushima T, et al Treatment of androgen-independent prostate cancer with dexamethasone: a prospective study in stage D2 patients.
Int J Urol
,
8
:
290
-4,  
2001
.
104
Hideshima T, Richardson PG, Chauhan D, et al The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma (MM) cells.
Blood
,
96
:
461a
2000
.
105
Nishimura K, Nonomura N, Satoh E, et al Potential mechanism for the effects of dexamethasone on growth of androgen-independent prostate cancer.
J Natl Cancer Inst
,
93
:
1739
-46,  
2001
.
106
Drexler HC. Activation of the cell death program by inhibition of proteasome function.
Proc Natl Acad Sci USA
,
94
:
855
-60,  
1997
.
107
Neumeier H, Hoar K, Pink M, et al Hypoxia increases potency of the proteasome inhibitor VELCADE (TM) (bortezomib) for injection: Potential for a hypoxic cell cytotoxin in solid tumors.
Eur J Cancer
,
38
:
166
2002
.
108
Richardson PG, Barlogie B, Berenson J, et al A phase 2 study of bortezomib in relapsed, refractory myeloma.
N Engl J Med
,
348
:
2609
-17,  
2003
.
109
Oncology Tools: New Approvals [cited 2004 Jan 31]. Available from: http://www.fda.gov/cder/cancer/whatsnew.htm.
110
Blade J, Samson D, Reece D, for the Myeloma Subcommittee of the European Group for Blood and Marrow Transplantet al Criteria for evaluating disease response and progression in patients with multiple myeloma treated by high-dose therapy and haemopoietic stem cell transplantation.
Br J Haematol
,
102
:
1115
-23,  
1998
.
111
Goy A, Hart S, Pro B, et al Report of a phase II study of proteasome inhibitor bortezomib (VELCADE™) in patients with relapsed or refractory indolent or aggressive lymphomas.
Blood
,
102
:
180a
2003
.
112
O’Connor O, Wright J, Moskowitz CH, et al Promising activity of the proteasome inhibitor bortezomib (VELCADE™) in the treatment of indolent non-Hodgkins lymphoma and mantle cell lymphoma.
Blood
,
102
:
636a
2003
.
113
Drucker BJ, Schwartz L, Bacik J, Mazumdar M, Marion S, Motzer RJ. Phase II trial of PS-341 shows response in patients with advanced renal cell carcinoma.
Proc Am Soc Clin Oncol
,
22
:
386
2003
.
114
Davis NB, Taber DA, Ansari RH, et al Phase II trial of PS-341 in patients with renal cell cancer: a University of Chicago phase II consortium study.
J Clin Oncol
,
22
:
115
-9,  
2004
.
115
Stevenson J, Nho CW, Schick J, et al Phase II clinical/pharmacodynamic trial of the proteasome inhibitor PS-341 in advanced non-small cell lung cancer.
Proc Am Soc Clin Oncol
,
22
:
202
2003
.