Skip to main content
Erschienen in: Cancer Cell International 1/2005

Open Access 01.12.2005 | Review

Preclinical evaluation of the proteasome inhibitor bortezomib in cancer therapy

verfasst von: Mario Boccadoro, Gareth Morgan, Jamie Cavenagh

Erschienen in: Cancer Cell International | Ausgabe 1/2005

Abstract

Bortezomib is a highly selective, reversible inhibitor of the 26S proteasome that is indicated for single-agent use in the treatment of patients with multiple myeloma who have received at least 2 prior therapies and are progressing on their most recent therapy. Clinical investigations have been completed or are under way to evaluate the safety and efficacy of bortezomib alone or in combination with chemotherapy in multiple myeloma, both at relapse and presentation, as well as in other cancer types. The antiproliferative, proapoptotic, antiangiogenic, and antitumor activities of bortezomib result from proteasome inhibition and depend on the altered degradation of a host of regulatory proteins. Exposure to bortezomib has been shown to stabilize p21, p27, and p53, as well as the proapoptotic Bid and Bax proteins, caveolin-1, and inhibitor κB-α, which prevents activation of nuclear factor κB-induced cell survival pathways. Bortezomib also promoted the activation of the proapoptotic c-Jun-NH2 terminal kinase, as well as the endoplasmic reticulum stress response. The anticancer effects of bortezomib as a single agent have been demonstrated in xenograft models of multiple myeloma, adult T-cell leukemia, lung, breast, prostate, pancreatic, head and neck, and colon cancer, and in melanoma. In these preclinical in vivo studies, bortezomib treatment resulted in decreased tumor growth, angiogenesis, and metastasis, as well as increased survival and tumor apoptosis. In several in vitro and/or in vivo cancer models, bortezomib has also been shown to enhance the antitumor properties of several antineoplastic treatments. Importantly, bortezomib was generally well tolerated and did not appear to produce additive toxicities when combined with other therapies in the dosing regimens used in these preclinical in vivo investigations. These findings provide a rationale for further clinical trials using bortezomib alone or in combination regimens with chemotherapy, radiation therapy, immunotherapy, or novel agents in patients with hematologic malignancies or solid tumors.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1475-2867-5-18) contains supplementary material, which is available to authorized users.

Competing interests

M.B. has received consulting and lecture fees from Millennium Pharmaceuticals, Inc. G.M. declares that he has no competing interests; he is supported by the Leukaemia Research Fund and the International Myeloma Foundation. J.C. has received advisory board and speakers' bureau fees from Millennium Pharmaceuticals, Inc. and Ortho Biotech.

Authors' contributions

M.B. reviewed the literature and drafted the manuscript. G.M. and J.C. reviewed and revised the manuscript. All authors read and approved the final version.
Abkürzungen
NF-κB
nuclear factor-κB
IκBα
inhibitor κB-α
VEGF
vascular endothelial growth factor
ER
endoplasmic reticulum.

Introduction

Bortezomib (VELCADE®, formerly PS-341) was approved for the treatment of patients with relapsed or refractory multiple myeloma in May 2003 by the US Food and Drug Administration [1] and in April 2004 by the Committee for Proprietary Medicinal Products of the European Union. A number of clinical studies evaluating the activity and safety of bortezomib in multiple myeloma, as well as in other types of cancer, have been conducted [210] or are ongoing [11]. Therefore, a review of key preclinical studies that have explored the mechanisms of action and provided the rationale for clinical investigation of this novel agent in multiple myeloma and other cancer types is warranted.

Mechanism of action

Bortezomib, a boronic acid dipeptide (Figure 1A) [12], is a highly selective, reversible inhibitor of the 26S proteasome that was first shown to exhibit antitumor properties in a panel of 60 cancer cell lines from the US National Cancer Institute [13]. The proteasome is an enzyme complex that primarily functions in the degradation of misfolded proteins and is essential for the regulation of the cell cycle. Proteasomes are localized in the nucleus and cytosol, where they are largely associated with centrosomes, the cytoskeleton, and the outer endoplasmic reticulum [14]. Damaged intracellular proteins are targeted for elimination by the proteasome through ubiquitination (Figure 1B). Many of the substrates that have been identified are proteins that function in the regulation of transcriptional activation, signal transduction, cell cycle proliferation, and apoptosis (Figure 2, Table 1) [1529].
Table 1
Intracellular targets of bortezomib.
Protein
Function
Effect of bortezomib
References
IκBα
Regulates the activity of the transcription factor, NF-κB
Stabilization
Hideshima et al [15], 2001
Russo et al [16], 2001
Sunwoo et al [17], 2001
Hideshima et al [18], 2002
Tan and Waldmann [19], 2002
Ma et al [20], 2003
JNK
Phosphorylates and activates the transcription factor c-Jun
Activation
Hideshima et al [21], 2003
Chauhan et al [22], 2004
Yang et al [23], 2004
p21, p27
CDK inhibitors
Stabilization
Shah et al [24], 2001
Hideshima et al [15], 2001
Yang et al [23], 2004
p53
Transcription factor and Tumor suppressor
Stabilization
Williams and McConkey [25], 2003
Hideshima et al [21], 2003
Yang et al [23], 2004
Bid
Proapoptotic protein
Stabilization
Breitschopf et al [26], 2000
Bax
Proapoptotic protein
Stabilization
Li and Dou [27], 2000
Caveolin-1
Promotes cell migration
Inhibition of activation
Podar et al [28], 2004
gp130
Cytokine signaling receptor
Downregulation
Hideshima et al [29], 2003
DNA-PKcs
DNA repair
Cleavage
Hideshima et al [21], 2003
ATM
DNA repair
Cleavage
Hideshima et al [21], 2003
IκBα = inhibitor κB-α; JNK = c-Jun-NH2 terminal kinase; CDK = cyclin-dependent kinase; DNA-PKcs = DNA protein kinase catalytic subunit; ATM = ataxia telangiectasia, mutated.
Several independent investigators have found that bortezomib inhibits activation of the transcription factor nuclear factor κB (NF-κB) [1520, 30]. NF-κB is important for cell survival and is activated in response to cell stress, including that induced by cytotoxic agents, radiation, or DNA damage. NF-κB is overexpressed in several tumors and regulates the expression of genes involved in apoptosis (including Bcl-2 and Bcl-xL), cell cycle progression, inflammation, and angiogenesis (including interleukin [IL]-6, IL-8, and vascular endothelial growth factor [VEGF]) [3133]. NF-κB is normally bound in the cytosol to inhibitor κB-α (IκBα). Phosphorylation, ubiquitination, and degradation of IκBα are required for NF-κB to translocate to the nucleus and activate the transcription of target genes. Bortezomib blocks the activation of NF-κB by preventing proteasomal degradation of IκBα. Through inhibition of NF-κB, bortezomib not only promotes apoptosis of cancer cells but also sensitizes these cells to chemotherapy [15, 20, 30], radiation [16], or immunotherapy [19]. However, because specific NF-κB inhibition alone via PS-1145 only partially inhibits proliferation of tumor cells [18], the cytotoxic activity of bortezomib must also depend on altered regulation of other signal transduction pathway targets [18].
The intracellular levels of a number of other proteins that regulate gene transcription, apoptosis, and proliferation are significantly affected by bortezomib (Table 1). c-Jun-NH2 terminal kinase (JNK) is a protein that promotes cell death in response to stress and increased levels of misfolded proteins [34]. Bortezomib treatment leads to activation of JNK in multiple myeloma [21, 22] and non-small cell lung cancer cells [23]. These studies further showed that specific inhibition of JNK activation, either genetic or pharmacologic, prevented mitochondrial release of cytochrome c and Smac, activation of caspase-8, -9, and -3, and apoptosis.
Proteasome inhibition has also been shown to stabilize the cyclin-dependent kinase inhibitors p21 and p27, the tumor suppressor p53, and the proapoptotic proteins Bid and Bax [15, 21, 2327]. The increased levels of activated p21, p27, p53, Bid, and Bax result in inhibition of cell cycle progression and/or promotion of apoptosis in response to bortezomib. Interestingly, sensitivity to proteasome inhibition was partially dependent on the p53 status of breast [35] and lung cancer in vitro [36], but bortezomib-induced apoptosis and/or chemosensitization were p53 independent in prostate [13], multiple myeloma [15], and colon cancer cells [30]. Therefore, the degree of variability in the sensitivity to bortezomib with respect to p53 status appears cell-type dependent.
A recently published study found that bortezomib prevented activation of caveolin-1 in multiple myeloma cells [28]. Activation of caveolin-1, a protein that functions in cell motility or migration in a number of tissues, requires phosphorylation. In this report, bortezomib was shown to prevent phosphorylation of caveolin-1 by VEGF, a proangiogenic cytokine and transcriptional target of NF-κB [37]. Bortezomib also inhibited VEGF secretion by the bone marrow. Together, these findings demonstrate important mechanisms by which bortezomib may inhibit migration of cancer cells as well as tumor angiogenesis.
The specific proteins mentioned have all been shown to be at least partially responsible in various models for the antiproliferative, proapoptotic, antiangiogenic, and antitumor effects of bortezomib. However, recent studies have found that bortezomib results in cytotoxic activity through activation of the endoplasmic reticulum stress response [3841]. The mechanism appears to involve blockade of retrograde transportation and degradation of damaged endoplasmic reticulum proteins by proteasome inhibition [42]. Further studies are necessary to link these new findings with the specific intracellular signals that have been previously implicated in the anticancer activities of bortezomib.

Bortezomib alone

Bortezomib has shown promising antitumor activity in a number of preclinical cancer murine models in vivo (Table 2) [13, 17, 19, 24, 30, 4349]. In a xenograft model of multiple myeloma, bortezomib treatment resulted in significant inhibition of tumor growth, an increase in overall survival, and a decrease in tumor angiogenesis [43]. The proteasome inhibitor was well tolerated up to 0.5 mg/kg intravenously (IV) twice weekly for 4 weeks, with dose-limiting toxicities, including weight loss, at 1 mg/kg. Two recent reports evaluating the efficacy of bortezomib in murine xenograft models of adult T-cell leukemia have reached contradictory conclusions. Tan and Waldman found that bortezomib treatment alone, 0.06 mg/kg intraperitoneally (IP), daily for 3 weeks, did not produce significant antitumor effects [19]. However, a second group reported that bortezomib 1.0 mg/kg IP twice weekly for 2 weeks resulted in antitumor activity [44]. Whereas Tan and Waldman reported lethality at a more aggressive dosing regimen of bortezomib 0.1 mg/kg IP twice daily for 2 weeks, slight, temporary weight loss was the only adverse effect described by the second group of investigators [44]. It seems plausible that the discrepancy in activity and toxicity may be due to the differences in the doses and dosing regimens. Based on the finding that proteasome inhibition by bortezomib lasts for up to 72 hours [8], more recent preclinical studies have used twice-weekly, rather than daily or twice-daily dosing, and have shown overall greater activity with less toxicity.
Table 2
Activity of bortezomib in tumor models in vivo.
Cancer
Activity
MTD
References
Multiple myeloma
Decreased tumor growth and angiogenesis; increased survival
0.5 mg/kg IV twice weekly for 4 weeks
LeBlanc et al [43], 2002
Adult T-cell leukemia
Decreased or no effect on tumor growth
1.0 mg/kg IP twice weekly for 2 weeks
Tan and Waldmann [19], 2002
Satou et al [44], 2004
Lung
Tumor growth delay and decreased lung metastases
1.0 mg/kg PO once daily for 18 days
Teicher et al [45], 1999
Breast
Decreased surviving fraction of tumor cells
5.0 mg/kg IP once
Teicher et al [45], 1999
Prostate
Decreased tumor growth; decrease or no effect on angiogenesis
1.0 mg/kg IV weekly for 4 weeks, or q 72 hrs for 15 days
Adams et al [13], 1999
Williams et al [46], 2003
Pancreatic
Decreased or no effect on tumor growth and angiogenesis; increased or no effect on tumor apoptosis
1.0 mg/kg IV biweekly for 2 to 3 weeks, or weekly for 4 weeks or 0.25 mg/kg IP biweekly for 4 weeks
Shah et al [24], 2001
Nawrocki et al [47], 2002
Bold et al [48], 2001
Head and neck
Decreased tumor growth and angiogenesis
1.5 mg/kg IP 3 times per week for 3 weeks
Sunwoo et al [17], 2001
Colon
Decreased tumor growth and increased tumor apoptosis
1.0 mg/kg IV twice weekly
Cusack et al [30], 2001
Melanoma
Decreased tumor growth and angiogenesis; increased tumor apoptosis
1.25 mg/kg SC twice weekly for 5 weeks
Amiri et al [49], 2004
MTD = maximum tolerated dose; IV = intravenous; IP = intraperitoneal; PO = by mouth; SC = subcutaneous.
An evaluation of the effects of bortezomib in murine xenograft models of both lung and breast cancer was also conducted [45]. Treatment with oral bortezomib 1.0 mg/kg daily for 18 days caused tumor growth delays, as well as a decrease in the number of metastases in the Lewis lung cancer model. Furthermore, in a murine model, bortezomib at a single IP dose of up to 5 mg/kg significantly decreased the surviving fraction of breast tumor cells. A decrease in the level of colony-forming-unit granulocyte macrophages was the only toxicity noted in these experiments.
Two groups of investigators have evaluated the efficacy of bortezomib in murine xenograft models of prostate cancer. The first study concluded that bortezomib 1.0 mg/kg IV weekly for 4 weeks reduced tumor growth by 60% [13]. The second study, in which bortezomib 1.0 mg/kg IV every 72 hours for 15 days was administered, produced similar results, with 50% and 80% inhibition in tumor growth in two xenograft models [46]. This report further showed that bortezomib significantly inhibited tumor angiogenesis in one of the models, as measured by a decrease in the number of CD31+ vessels in tumor sections. No toxicities were detected in either of these studies.
In a study of pancreatic cancer murine xenografts, treatment with bortezomib 1.0 mg/kg IV or IP weekly for 4 weeks resulted in a 72% or 84% reduction in tumor growth, as well as an increase in tumor cell apoptosis, with no evidence of toxicity [24]. Another group found that bortezomib 1.0 mg/kg IV biweekly for 2 to 3 weeks significantly inhibited tumor growth and angiogenesis and promoted apoptosis in 1 of 2 pancreatic cancer xenograft murine models [47]. These investigators also reported adverse events, including decreased body weight, diarrhea, and gastrointestinal inflammation at doses above 1.0 mg/kg and lethality at doses above 1.5 mg/kg. Finally, bortezomib produced significant antitumor, proapoptotic, and/or antiangiogenic effects in murine xenograft models of head and neck [17] and colon cancer [30], as well as melanoma [49]. Adverse events, including dehydration, lethargy, weight loss, and death, were noted at doses of 2.0 mg/kg IP 3 times weekly for 3 weeks in one of these studies [17], whereas the other studies did not report any toxicities at lower doses (Table 2).
These preclinical investigations have collectively demonstrated the antitumor activity of bortezomib as a single agent at tolerable levels in a variety of murine cancer models. However, because the standard approach to antineoplastic therapy generally involves the administration of more than one agent or modality in an effort to prevent the development of chemoresistance and increase tumor cell kill, the effects of bortezomib in combination with chemotherapy, radiation, immunotherapy, or novel agents have been investigated in vitro and/or in vivo.

Combination therapy

A number of preclinical studies have evaluated the activity of bortezomib in combination with other therapies (Table 3) [15, 16, 19, 20, 24, 30, 39, 40, 45, 4861]. The finding that several cytotoxic agents as well as radiation [62, 63] activate NF-κB is a major rationale for combining proteasome inhibitors with these therapies in the treatment of cancer. Activated NF-κB is free to translocate to the nucleus and induce the expression of proinflammatory and antiapoptotic genes, such as Bcl-2 and Bcl-xL, which promote tumor cell survival [3133]. Furthermore, inhibition of NF-κB has been implicated as an important mechanism by which bortezomib sensitizes tumor cells to various drugs or radiation [15, 16, 20, 24, 30, 4850, 55, 57, 59, 60].
Table 3
Evaluation of bortezomib in combination with other therapies.
Type of therapy
Drug or agent
Cancer
References
Chemotherapy
5-Fluorouracil, cisplatin, paclitaxel, doxorubicin, cyclophosphamide
Breast, lung
Teicher et al [45], 1999
 
Melphalan, doxorubicin, dexamethasone
Multiple myeloma
Hideshima et al [15], 2001
Mitsiades et al [50], 2003
Ma et al [20], 2003
 
Topoisomerase inhibitor: irinotecan
Colon Pancreatic
Cusack et al [30], 2001
Shah et al [24], 2001
 
Gemcitabine
Pancreatic Bladder
Bold et al [48], 2001
Kamat et al [51], 2004
 
Pegylated liposomal doxorubicin
Breast
Small et al [52], 2004
 
Docetaxel
Pancreatic
Nawrocki et al [53], 2004
 
Temozolomide
Melanoma
Amiri et al [49], 2004
Radiation therapy
 
Breast Colon Prostate
Teicher et al [45], 1999
Russo et al [16], 2001
Pervan et al [54], 2001
Immunotherapy
Daclizumab
Adult T-cell leukemia
Tan and Waldmann [19], 2002
Novel agents
TRAIL/Apo2L
Multiple myeloma, myeloid leukemia, renal
Mitsiades et al [55], 2001
Sayers et al [56], 2003
 
HSP90 inhibitor: 17-AAG
Breast
Mimnaugh et al [39], 2004
 
HDAC inhibitors: SAHA, sodium butyrate
CML, multiple myeloma, lung
Denlinger et al [40], 2004
Yu et al [57], 2003
Mitsiades et al [58], 2004
Denlinger et al [59], 2004
Pei et al [60], 2004
Transplantation
Allogeneic BMT
Leukemia
Sun et al [61], 2004
HSP = heat shock protein; HDAC = histone deacetylase; CML = chronic myelogenous leukemia; BMT = bone marrow transplantation.

Bortezomib with chemotherapy

In an investigation of lung cancer, bortezomib in combination with chemotherapeutic agents, including 5-fluorouracil, cisplatin, paclitaxel, or doxorubicin, produced additive antitumor and antimetastatic effects [45]. In this same study, bortezomib also increased tumor-cell killing by cyclophosphamide and cisplatin, as well as tumor-cell killing by radiation, in an in vitro-in vivo model of breast cancer. Although toxicity is difficult to assess in preclinical models, no added toxicities were observed when bortezomib was added to the other therapies, and bortezomib dose modifications were not required.
Several studies evaluating the effects of bortezomib in combination with other therapies have been conducted in multiple myeloma. These in vitro experiments collectively confirmed that bortezomib enhanced the antiproliferative and proapoptotic effects of conventional antimyeloma agents, including melphalan, doxorubicin, and dexamethasone [15, 20, 50]. These investigators also reported that multiple myeloma cell lines that were previously resistant to melphalan, doxorubicin, dexamethasone, or mitoxantrone were sensitized up to 1,000,000-fold by prior exposure to subtoxic concentrations of bortezomib. Finally, the researchers showed that bortezomib was not only directly cytotoxic to the multiple myeloma cells but that it also altered the microenvironment through inhibition of IL-6 to prevent the growth of tumor cells in proximity to the bone marrow [15].
Two groups of researchers examined the effects of bortezomib in combination with the topoisomerase inhibitor irinotecan in murine xenograft models of colon [30] and pancreatic [24] cancer. Both these studies concluded that combined inhibition of the proteasome and topoisomerase resulted in enhanced antiproliferative and proapoptotic effects. The combination of bortezomib and irinotecan therapy further resulted in a 94% [30] or 89% [24] decrease in tumor size compared with controls. These tumors were also significantly smaller than those of the mice receiving either bortezomib (26% or 65% decrease in tumor size relative to controls) or irinotecan (48% or 43% decrease in tumor size relative to controls) as single agents.
Xenograft models of pancreatic cancer were also used to evaluate the activity of bortezomib in combination with other standard chemotherapies [48, 53]. Inhibition of the proteasome increased the sensitivity of tumors to both gemcitabine [48] and docetaxel [53], because bortezomib in combination with these agents resulted in significant enhancement of antiproliferative, proapoptotic, antitumor, and/or antiangiogenic activities.
The combination of bortezomib with temozolomide was studied in models of malignant melanoma [49]. Bortezomib enhanced the antiproliferative and cytotoxic effects of temozolomide in melanoma cells in vitro, and this combined therapy produced complete remission of tumors lasting more than 200 days in murine xenografts in vivo. In contrast, tumors eventually progressed in mice receiving either drug alone. Although specific toxicity evaluations were not performed, no toxicities were observed in any of these investigations.

Bortezomib with radiation, immunotherapy, or novel agents

A number of studies have reported on the radiosensitizing properties of bortezomib. In addition to the previously mentioned study in breast cancer [45], other laboratories have demonstrated that bortezomib sensitizes colon [16] and prostate [54] cancers to radiation-induced cytotoxicity. In both clonogenic cell survival assays and murine xenograft tumor models, pretreatment with bortezomib enhanced the anticancer effects of irradiation with no observed toxicity.
Another murine xenograft model was used to investigate the effects of bortezomib in combination with daclizumab, a humanized anti-IL-2Rα antibody in adult T-cell leukemia [19]. Although either agent alone resulted in partial or no responses, bortezomib plus daclizumab resulted in prolongation of the survival of tumor-bearing mice. The only adverse effect of the combined therapy was a slight temporary weight loss during the treatment regimen.
Bortezomib has also demonstrated enhanced in vitro and/or in vivo anticancer effects when combined with novel agents such as TRAIL/Apo2L, a cell death-inducing ligand [55, 56], 17-N-allylamino-17-demethoxygeldanamycin (17-AAG), an inhibitor of heat shock protein 90 [39], and suberoylanilide hydroxamic acid (SAHA) or sodium butyrate, both histone deactylase inhibitors [40, 57, 59, 60]. Pretreatment with bortezomib sensitized multiple myeloma, myeloid leukemia, and renal cancer cells but not normal B lymphocytes to TRAIL/Apo2L-induced apoptosis [55, 56]. In an in vivo experiment, bone marrow and renal cancer cell mixtures, with or without bortezomib and/or TRAIL/Apo2L, were transplanted into the bone marrow of mice. Whereas all the mice receiving cells treated with TRAIL/Apo2L died of leukemia within 35 days, 50% of those receiving cells treated with bortezomib and 90% of those receiving cells treated with both TRAIL/Apo2L and bortezomib survived more than 100 days [56]. A mild transient thrombocytopenia was the only toxicity observed in this study. Finally, while the combination of bortezomib and 17-AAG, SAHA, or sodium butyrate resulted in synergistic antiproliferative and proapoptotic effects in vitro [39, 40, 5760], these combinations have yet to be evaluated in tumor xenograft models in vivo.

Sequence of administration

One of the areas of controversy has been the appropriate timing of therapy with bortezomib in relation to other antineoplastic agents. A brief discussion of some of the conflicting results is warranted as these preclinical studies may ultimately provide the rationale for clinical trials. In vitro experiments conducted by Mitsiades and colleagues revealed that optimal antimyeloma activity was achieved when bortezomib was administered 24 hours after doxorubicin [50]. The sequence of chemotherapy (gemcitabine) followed by bortezomib was also found to be the most effective in a model of pancreatic cancer [64]. In contrast, Pei et al showed that bortezomib followed by a second antineoplastic agent (SAHA) yielded the highest level of apoptosis in a model of myeloma [60], and Mimnaugh and colleagues found that maximal antiproliferative effects were observed upon simultaneous administration of bortezomib and a heat shock protein 90 inhibitor in a model of breast cancer [39]. It is important to note that these studies utilized bortezomib in combination with different antineoplastic agents in unique cancer models. Further studies may elucidate the reasons for the inconsistent findings.

Conclusion

The proteasome inhibitor bortezomib exhibits antiproliferative, proapoptotic, antiangiogenic, and antitumor activities in several cancer models. The mechanism of action of bortezomib involves stabilization of NF-κB, p21, p27, p53, Bid, and Bax, inhibition of caveolin-1 activation, and activation of JNK as well as the endoplasmic reticulum stress response. These preclinical evaluations have found that bortezomib is well tolerated at doses that demonstrated antitumor activity in xenograft models of multiple myeloma, adult T-cell leukemia, cancer of the lung, breast, prostate, pancreas, head and neck, and colon, as well as melanoma. Bortezomib also enhances the anticancer effects of chemotherapy, radiation therapy, immunotherapy, or novel agents, without added toxicities requiring dose modifications. The studies provide a rationale for clinical trials of bortezomib alone or in combination with other therapies in patients with solid tumors or hematologic malignancies.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

M.B. has received consulting and lecture fees from Millennium Pharmaceuticals, Inc. G.M. declares that he has no competing interests; he is supported by the Leukaemia Research Fund and the International Myeloma Foundation. J.C. has received advisory board and speakers' bureau fees from Millennium Pharmaceuticals, Inc. and Ortho Biotech.

Authors' contributions

M.B. reviewed the literature and drafted the manuscript. G.M. and J.C. reviewed and revised the manuscript. All authors read and approved the final version.
Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Kane RC, Bross PF, Farrell AT, Pazdur R: Velcade: U.S. FDA approval for the treatment of multiple myeloma progressing on prior therapy. Oncologist. 2003, 8: 508-513. 10.1634/theoncologist.8-6-508.CrossRefPubMed Kane RC, Bross PF, Farrell AT, Pazdur R: Velcade: U.S. FDA approval for the treatment of multiple myeloma progressing on prior therapy. Oncologist. 2003, 8: 508-513. 10.1634/theoncologist.8-6-508.CrossRefPubMed
2.
Zurück zum Zitat Aghajanian C, Soignet S, Dizon DS, Pien CS, Adams J, Elliott PJ, Sabbatini P, Miller V, Hensley ML, Pezzulli S, Canales C, Daud A, Spriggs DR: A phase I trial of the novel proteasome inhibitor PS341 in advanced solid tumor malignancies. Clin Cancer Res. 2002, 8: 2505-2511.PubMed Aghajanian C, Soignet S, Dizon DS, Pien CS, Adams J, Elliott PJ, Sabbatini P, Miller V, Hensley ML, Pezzulli S, Canales C, Daud A, Spriggs DR: A phase I trial of the novel proteasome inhibitor PS341 in advanced solid tumor malignancies. Clin Cancer Res. 2002, 8: 2505-2511.PubMed
3.
Zurück zum Zitat Orlowski RZ, Stinchcombe TE, Mitchell BS, Shea TC, Baldwin AS, Stahl S, Adams J, Esseltine DL, Elliott PJ, Pien CS, Guerciolini R, Anderson JK, Depcik-Smith ND, Bhagat R, Lehman MJ, Novick SC, O'Connor OA, Soignet SL: Phase I trial of the proteasome inhibitor PS-341 in patients with refractory hematologic malignancies. J Clin Oncol. 2002, 20: 4420-4427. 10.1200/JCO.2002.01.133.CrossRefPubMed Orlowski RZ, Stinchcombe TE, Mitchell BS, Shea TC, Baldwin AS, Stahl S, Adams J, Esseltine DL, Elliott PJ, Pien CS, Guerciolini R, Anderson JK, Depcik-Smith ND, Bhagat R, Lehman MJ, Novick SC, O'Connor OA, Soignet SL: Phase I trial of the proteasome inhibitor PS-341 in patients with refractory hematologic malignancies. J Clin Oncol. 2002, 20: 4420-4427. 10.1200/JCO.2002.01.133.CrossRefPubMed
4.
Zurück zum Zitat Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, Irwin D, Rajkumar SV, Srkalovic G, Alsina M, Alexanian R, Siegel D, Orlowski RZ, Kuter D, Limentani SA, Lee S, Hideshima T, Esseltine DL, Kauffman M, Adams J, Schenkein DP, Anderson KC: A phase 2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med. 2003, 348: 2609-2617. 10.1056/NEJMoa030288.CrossRefPubMed Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, Irwin D, Rajkumar SV, Srkalovic G, Alsina M, Alexanian R, Siegel D, Orlowski RZ, Kuter D, Limentani SA, Lee S, Hideshima T, Esseltine DL, Kauffman M, Adams J, Schenkein DP, Anderson KC: A phase 2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med. 2003, 348: 2609-2617. 10.1056/NEJMoa030288.CrossRefPubMed
5.
Zurück zum Zitat Davis NB, Taber DA, Ansari RH, Ryan CW, George C, Vokes EE, Vogelzang NJ, Stadler WM: Phase II trial of PS-341 in patients with renal cell cancer: a University of Chicago phase II consortium study. J Clin Oncol. 2004, 22: 115-119. 10.1200/JCO.2004.07.165.CrossRefPubMed Davis NB, Taber DA, Ansari RH, Ryan CW, George C, Vokes EE, Vogelzang NJ, Stadler WM: Phase II trial of PS-341 in patients with renal cell cancer: a University of Chicago phase II consortium study. J Clin Oncol. 2004, 22: 115-119. 10.1200/JCO.2004.07.165.CrossRefPubMed
6.
Zurück zum Zitat Papandreou CN, Daliani DD, Nix D, Yang H, Madden T, Wang X, Pien CS, Millikan RE, Tu SM, Pagliaro L, Kim J, Adams J, Elliott P, Esseltine D, Petrusich A, Dieringer P, Perez C, Logothetis CJ: Phase I trial of the proteasome inhibitor bortezomib in patients with advanced solid tumors with observations in androgen-independent prostate cancer. J Clin Oncol. 2004, 22: 2108-2121. 10.1200/JCO.2004.02.106.CrossRefPubMed Papandreou CN, Daliani DD, Nix D, Yang H, Madden T, Wang X, Pien CS, Millikan RE, Tu SM, Pagliaro L, Kim J, Adams J, Elliott P, Esseltine D, Petrusich A, Dieringer P, Perez C, Logothetis CJ: Phase I trial of the proteasome inhibitor bortezomib in patients with advanced solid tumors with observations in androgen-independent prostate cancer. J Clin Oncol. 2004, 22: 2108-2121. 10.1200/JCO.2004.02.106.CrossRefPubMed
7.
Zurück zum Zitat Kondagunta GV, Drucker B, Schwartz L, Bacik J, Marion S, Russo P, Mazumdar M, Motzer RJ: Phase II trial of bortezomib for patients with advanced renal cell carcinoma. J Clin Oncol. 2004, 22: 3720-3725. 10.1200/JCO.2004.10.155.CrossRefPubMed Kondagunta GV, Drucker B, Schwartz L, Bacik J, Marion S, Russo P, Mazumdar M, Motzer RJ: Phase II trial of bortezomib for patients with advanced renal cell carcinoma. J Clin Oncol. 2004, 22: 3720-3725. 10.1200/JCO.2004.10.155.CrossRefPubMed
8.
Zurück zum Zitat Cortes J, Thomas D, Koller C, Giles F, Estey E, Faderl S, Garcia-Manero G, McConkey D, Patel G, Guerciolini R, Wright J, Kantarjian H: Phase I study of bortezomib in refractory or relapsed acute leukemias. Clin Cancer Res. 2004, 10: 3371-3376.CrossRefPubMed Cortes J, Thomas D, Koller C, Giles F, Estey E, Faderl S, Garcia-Manero G, McConkey D, Patel G, Guerciolini R, Wright J, Kantarjian H: Phase I study of bortezomib in refractory or relapsed acute leukemias. Clin Cancer Res. 2004, 10: 3371-3376.CrossRefPubMed
9.
Zurück zum Zitat Jagannath S, Barlogie B, Berenson J, Siegel D, Irwin D, Richardson PG, Niesvizky R, Alexanian R, Limentani SA, Alsina M, Adams J, Kauffman M, Esseltine DL, Schenkein DP, Anderson KC: A phase 2 study of two doses of bortezomib in relapsed or refractory myeloma. Br J Haematol. 2004, 127: 165-172. 10.1111/j.1365-2141.2004.05188.x.CrossRefPubMed Jagannath S, Barlogie B, Berenson J, Siegel D, Irwin D, Richardson PG, Niesvizky R, Alexanian R, Limentani SA, Alsina M, Adams J, Kauffman M, Esseltine DL, Schenkein DP, Anderson KC: A phase 2 study of two doses of bortezomib in relapsed or refractory myeloma. Br J Haematol. 2004, 127: 165-172. 10.1111/j.1365-2141.2004.05188.x.CrossRefPubMed
10.
Zurück zum Zitat Shah MH, Young D, Kindler HL, Webb I, Kleiber B, Wright J, Grever M: Phase II study of the proteasome inhibitor bortezomib (PS-341) in patients with metastatic neuroendocrine tumors. Clin Cancer Res. 2004, 10: 6111-6118.CrossRefPubMed Shah MH, Young D, Kindler HL, Webb I, Kleiber B, Wright J, Grever M: Phase II study of the proteasome inhibitor bortezomib (PS-341) in patients with metastatic neuroendocrine tumors. Clin Cancer Res. 2004, 10: 6111-6118.CrossRefPubMed
11.
Zurück zum Zitat Bruno B, Rotta M, Giaccone L, Massaia M, Bertola A, Palumbo A, Boccadoro M: New drugs for treatment of multiple myeloma. Lancet Oncol. 2004, 5: 430-442. 10.1016/S1470-2045(04)01511-6.CrossRefPubMed Bruno B, Rotta M, Giaccone L, Massaia M, Bertola A, Palumbo A, Boccadoro M: New drugs for treatment of multiple myeloma. Lancet Oncol. 2004, 5: 430-442. 10.1016/S1470-2045(04)01511-6.CrossRefPubMed
12.
Zurück zum Zitat Adams J: The development of proteasome inhibitors as anticancer drugs. Cancer Cell. 2004, 5: 417-421. 10.1016/S1535-6108(04)00120-5.CrossRefPubMed Adams J: The development of proteasome inhibitors as anticancer drugs. Cancer Cell. 2004, 5: 417-421. 10.1016/S1535-6108(04)00120-5.CrossRefPubMed
13.
Zurück zum Zitat Adams J, Palombella VJ, Sausville EA, Johnson J, Destree A, Lazarus DD, Maas J, Pien CS, Prakash S, Elliott PJ: Proteasome inhibitors: a novel class of potent and effective antitumor agents. Cancer Res. 1999, 59: 2615-2622.PubMed Adams J, Palombella VJ, Sausville EA, Johnson J, Destree A, Lazarus DD, Maas J, Pien CS, Prakash S, Elliott PJ: Proteasome inhibitors: a novel class of potent and effective antitumor agents. Cancer Res. 1999, 59: 2615-2622.PubMed
14.
Zurück zum Zitat Wojcik C, DeMartino GN: Intracellular localization of proteasomes. Int J Biochem Cell Biol. 2003, 35: 579-589. 10.1016/S1357-2725(02)00380-1.CrossRefPubMed Wojcik C, DeMartino GN: Intracellular localization of proteasomes. Int J Biochem Cell Biol. 2003, 35: 579-589. 10.1016/S1357-2725(02)00380-1.CrossRefPubMed
15.
Zurück zum Zitat Hideshima T, Richardson P, Chauhan D, Palombella VJ, Elliott PJ, Adams J, Anderson KC: The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells. Cancer Res. 2001, 61: 3071-3076.PubMed Hideshima T, Richardson P, Chauhan D, Palombella VJ, Elliott PJ, Adams J, Anderson KC: The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells. Cancer Res. 2001, 61: 3071-3076.PubMed
16.
Zurück zum Zitat Russo SM, Tepper JE, Baldwin ASJ, Liu R, Adams J, Elliott P, Cusack JCJ: Enhancement of radiosensitivity by proteasome inhibition: implications for a role of NF-kappaB. Int J Radiat Oncol Biol Phys. 2001, 50: 183-193. 10.1016/S0360-3016(01)01446-8.CrossRefPubMed Russo SM, Tepper JE, Baldwin ASJ, Liu R, Adams J, Elliott P, Cusack JCJ: Enhancement of radiosensitivity by proteasome inhibition: implications for a role of NF-kappaB. Int J Radiat Oncol Biol Phys. 2001, 50: 183-193. 10.1016/S0360-3016(01)01446-8.CrossRefPubMed
17.
Zurück zum Zitat Sunwoo JB, Chen Z, Dong G, Yeh N, Crowl BC, Sausville E, Adams J, Elliott P, Van Waes C: Novel proteasome inhibitor PS-341 inhibits activation of nuclear factor-kappa B, cell survival, tumor growth, and angiogenesis in squamous cell carcinoma. Clin Cancer Res. 2001, 7: 1419-1428.PubMed Sunwoo JB, Chen Z, Dong G, Yeh N, Crowl BC, Sausville E, Adams J, Elliott P, Van Waes C: Novel proteasome inhibitor PS-341 inhibits activation of nuclear factor-kappa B, cell survival, tumor growth, and angiogenesis in squamous cell carcinoma. Clin Cancer Res. 2001, 7: 1419-1428.PubMed
18.
Zurück zum Zitat Hideshima T, Chauhan D, Richardson P, Mitsiades C, Mitsiades N, Hayashi T, Munshi N, Dang L, Castro A, Palombella V, Adams J, Anderson KC: NF-kappa B as a therapeutic target in multiple myeloma. J Biol Chem. 2002, 277: 16639-16647. 10.1074/jbc.M200360200.CrossRefPubMed Hideshima T, Chauhan D, Richardson P, Mitsiades C, Mitsiades N, Hayashi T, Munshi N, Dang L, Castro A, Palombella V, Adams J, Anderson KC: NF-kappa B as a therapeutic target in multiple myeloma. J Biol Chem. 2002, 277: 16639-16647. 10.1074/jbc.M200360200.CrossRefPubMed
19.
Zurück zum Zitat Tan C, Waldmann TA: Proteasome inhibitor PS-341, a potential therapeutic agent for adult T-cell leukemia. Cancer Res. 2002, 62: 1083-1086.PubMed Tan C, Waldmann TA: Proteasome inhibitor PS-341, a potential therapeutic agent for adult T-cell leukemia. Cancer Res. 2002, 62: 1083-1086.PubMed
20.
Zurück zum Zitat Ma MH, Yang HH, Parker K, Manyak S, Friedman JM, Altamirano C, Wu ZQ, Borad MJ, Frantzen M, Roussos E, Neeser J, Mikail A, Adams J, Sjak-Shie N, Vescio RA, Berenson JR: The proteasome inhibitor PS-341 markedly enhances sensitivity of multiple myeloma tumor cells to chemotherapeutic agents. Clin Cancer Res. 2003, 9: 1136-1144.PubMed Ma MH, Yang HH, Parker K, Manyak S, Friedman JM, Altamirano C, Wu ZQ, Borad MJ, Frantzen M, Roussos E, Neeser J, Mikail A, Adams J, Sjak-Shie N, Vescio RA, Berenson JR: The proteasome inhibitor PS-341 markedly enhances sensitivity of multiple myeloma tumor cells to chemotherapeutic agents. Clin Cancer Res. 2003, 9: 1136-1144.PubMed
21.
Zurück zum Zitat Hideshima T, Mitsiades C, Akiyama M, Hayashi T, Chauhan D, Richardson P, Schlossman R, Podar K, Munshi NC, Mitsiades N, Anderson KC: Molecular mechanisms mediating antimyeloma activity of proteasome inhibitor PS-341. Blood. 2003, 101: 1530-1534. 10.1182/blood-2002-08-2543.CrossRefPubMed Hideshima T, Mitsiades C, Akiyama M, Hayashi T, Chauhan D, Richardson P, Schlossman R, Podar K, Munshi NC, Mitsiades N, Anderson KC: Molecular mechanisms mediating antimyeloma activity of proteasome inhibitor PS-341. Blood. 2003, 101: 1530-1534. 10.1182/blood-2002-08-2543.CrossRefPubMed
22.
Zurück zum Zitat Chauhan D, Li G, Podar K, Hideshima T, Mitsiades C, Schlossman R, Munshi N, Richardson P, Cotter FE, Anderson KC: Targeting mitochondria to overcome conventional and bortezomib/proteasome inhibitor PS-341 resistance in multiple myeloma (MM) cells. Blood. 2004, 104: 2458-2466. 10.1182/blood-2004-02-0547.CrossRefPubMed Chauhan D, Li G, Podar K, Hideshima T, Mitsiades C, Schlossman R, Munshi N, Richardson P, Cotter FE, Anderson KC: Targeting mitochondria to overcome conventional and bortezomib/proteasome inhibitor PS-341 resistance in multiple myeloma (MM) cells. Blood. 2004, 104: 2458-2466. 10.1182/blood-2004-02-0547.CrossRefPubMed
23.
Zurück zum Zitat Yang Y, Ikezoe T, Saito T, Kobayashi M, Koeffler HP, Taguchi H: Proteasome inhibitor PS-341 induces growth arrest and apoptosis of non-small cell lung cancer cells via the JNK/c-Jun/AP-1 signaling. Cancer Sci. 2004, 95: 176-180.CrossRefPubMed Yang Y, Ikezoe T, Saito T, Kobayashi M, Koeffler HP, Taguchi H: Proteasome inhibitor PS-341 induces growth arrest and apoptosis of non-small cell lung cancer cells via the JNK/c-Jun/AP-1 signaling. Cancer Sci. 2004, 95: 176-180.CrossRefPubMed
24.
Zurück zum Zitat Shah SA, Potter MW, McDade TP, Ricciardi R, Perugini RA, Elliott PJ, Adams J, Callery MP: 26S proteasome inhibition induces apoptosis and limits growth of human pancreatic cancer. J Cell Biochem. 2001, 82: 110-122. 10.1002/jcb.1150.CrossRefPubMed Shah SA, Potter MW, McDade TP, Ricciardi R, Perugini RA, Elliott PJ, Adams J, Callery MP: 26S proteasome inhibition induces apoptosis and limits growth of human pancreatic cancer. J Cell Biochem. 2001, 82: 110-122. 10.1002/jcb.1150.CrossRefPubMed
25.
Zurück zum Zitat Williams SA, McConkey DJ: The proteasome inhibitor bortezomib stabilizes a novel active form of p53 in human LNCaP-Pro5 prostate cancer cells. Cancer Res. 2003, 63: 7338-7344.PubMed Williams SA, McConkey DJ: The proteasome inhibitor bortezomib stabilizes a novel active form of p53 in human LNCaP-Pro5 prostate cancer cells. Cancer Res. 2003, 63: 7338-7344.PubMed
26.
Zurück zum Zitat Breitschopf K, Zeiher AM, Dimmeler S: Ubiquitin-mediated degradation of the proapoptotic active form of bid. A functional consequence on apoptosis induction. J Biol Chem. 2000, 275: 21648-21652. 10.1074/jbc.M001083200.CrossRefPubMed Breitschopf K, Zeiher AM, Dimmeler S: Ubiquitin-mediated degradation of the proapoptotic active form of bid. A functional consequence on apoptosis induction. J Biol Chem. 2000, 275: 21648-21652. 10.1074/jbc.M001083200.CrossRefPubMed
27.
Zurück zum Zitat Li B, Dou QP: Bax degradation by the ubiquitin/proteasome-dependent pathway: involvement in tumor survival and progression. Proc Natl Acad Sci U S A. 2000, 97: 3850-3855. 10.1073/pnas.070047997.PubMedCentralCrossRefPubMed Li B, Dou QP: Bax degradation by the ubiquitin/proteasome-dependent pathway: involvement in tumor survival and progression. Proc Natl Acad Sci U S A. 2000, 97: 3850-3855. 10.1073/pnas.070047997.PubMedCentralCrossRefPubMed
28.
Zurück zum Zitat Podar K, Shringarpure R, Tai YT, Simoncini M, Sattler M, Ishitsuka K, Richardson PG, Hideshima T, Chauhan D, Anderson KC: Caveolin-1 is required for vascular endothelial growth factor-triggered multiple myeloma cell migration and is targeted by bortezomib. Cancer Res. 2004, 64: 7500-7506.CrossRefPubMed Podar K, Shringarpure R, Tai YT, Simoncini M, Sattler M, Ishitsuka K, Richardson PG, Hideshima T, Chauhan D, Anderson KC: Caveolin-1 is required for vascular endothelial growth factor-triggered multiple myeloma cell migration and is targeted by bortezomib. Cancer Res. 2004, 64: 7500-7506.CrossRefPubMed
29.
Zurück zum Zitat Hideshima T, Chauhan D, Hayashi T, Akiyama M, Mitsiades N, Mitsiades C, Podar K, Munshi NC, Richardson PG, Anderson KC: Proteasome inhibitor PS-341 abrogates IL-6 triggered signaling cascades via caspase-dependent downregulation of gp130 in multiple myeloma. Oncogene. 2003, 22: 8386-8393. 10.1038/sj.onc.1207170.CrossRefPubMed Hideshima T, Chauhan D, Hayashi T, Akiyama M, Mitsiades N, Mitsiades C, Podar K, Munshi NC, Richardson PG, Anderson KC: Proteasome inhibitor PS-341 abrogates IL-6 triggered signaling cascades via caspase-dependent downregulation of gp130 in multiple myeloma. Oncogene. 2003, 22: 8386-8393. 10.1038/sj.onc.1207170.CrossRefPubMed
30.
Zurück zum Zitat Cusack JCJ, Liu R, Houston M, Abendroth K, Elliott PJ, Adams J, Baldwin ASJ: Enhanced chemosensitivity to CPT-11 with proteasome inhibitor PS-341: implications for systemic nuclear factor-kappaB inhibition. Cancer Res. 2001, 61: 3535-3540.PubMed Cusack JCJ, Liu R, Houston M, Abendroth K, Elliott PJ, Adams J, Baldwin ASJ: Enhanced chemosensitivity to CPT-11 with proteasome inhibitor PS-341: implications for systemic nuclear factor-kappaB inhibition. Cancer Res. 2001, 61: 3535-3540.PubMed
31.
Zurück zum Zitat 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-kappaB that blocks TNFalpha-induced apoptosis. Genes Dev. 1999, 13: 382-387.PubMedCentralCrossRefPubMed 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-kappaB that blocks TNFalpha-induced apoptosis. Genes Dev. 1999, 13: 382-387.PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Dong QG, Sclabas GM, Fujioka S, Schmidt C, Peng B, Wu T, Tsao MS, Evans DB, Abbruzzese JL, McDonnell TJ, Chiao PJ: The function of multiple IkappaB : NF-kappaB complexes in the resistance of cancer cells to Taxol-induced apoptosis. Oncogene. 2002, 21: 6510-6519. 10.1038/sj.onc.1205848.CrossRefPubMed Dong QG, Sclabas GM, Fujioka S, Schmidt C, Peng B, Wu T, Tsao MS, Evans DB, Abbruzzese JL, McDonnell TJ, Chiao PJ: The function of multiple IkappaB : NF-kappaB complexes in the resistance of cancer cells to Taxol-induced apoptosis. Oncogene. 2002, 21: 6510-6519. 10.1038/sj.onc.1205848.CrossRefPubMed
33.
Zurück zum Zitat Loercher A, Lee TL, Ricker JL, Howard A, Geoghegen J, Chen Z, Sunwoo JB, Sitcheran R, Chuang EY, Mitchell JB, Baldwin ASJ, Van Waes C: Nuclear factor-kappaB is an important modulator of the altered gene expression profile and malignant phenotype in squamous cell carcinoma. Cancer Res. 2004, 64: 6511-6523.CrossRefPubMed Loercher A, Lee TL, Ricker JL, Howard A, Geoghegen J, Chen Z, Sunwoo JB, Sitcheran R, Chuang EY, Mitchell JB, Baldwin ASJ, Van Waes C: Nuclear factor-kappaB is an important modulator of the altered gene expression profile and malignant phenotype in squamous cell carcinoma. Cancer Res. 2004, 64: 6511-6523.CrossRefPubMed
34.
Zurück zum Zitat 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. 1998, 438: 1-4. 10.1016/S0014-5793(98)01242-3.CrossRefPubMed 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. 1998, 438: 1-4. 10.1016/S0014-5793(98)01242-3.CrossRefPubMed
35.
Zurück zum Zitat MacLaren AP, Chapman RS, Wyllie AH, Watson CJ: p53-dependent apoptosis induced by proteasome inhibition in mammary epithelial cells. Cell Death Differ. 2001, 8: 210-218. 10.1038/sj.cdd.4400801.CrossRefPubMed MacLaren AP, Chapman RS, Wyllie AH, Watson CJ: p53-dependent apoptosis induced by proteasome inhibition in mammary epithelial cells. Cell Death Differ. 2001, 8: 210-218. 10.1038/sj.cdd.4400801.CrossRefPubMed
36.
Zurück zum Zitat Ling YH, Liebes L, Jiang JD, Holland JF, Elliott PJ, Adams J, Muggia FM, Perez-Soler R: Mechanisms of proteasome inhibitor PS-341-induced G(2)-M-phase arrest and apoptosis in human non-small cell lung cancer cell lines. Clin Cancer Res. 2003, 9: 1145-1154.PubMed Ling YH, Liebes L, Jiang JD, Holland JF, Elliott PJ, Adams J, Muggia FM, Perez-Soler R: Mechanisms of proteasome inhibitor PS-341-induced G(2)-M-phase arrest and apoptosis in human non-small cell lung cancer cell lines. Clin Cancer Res. 2003, 9: 1145-1154.PubMed
37.
Zurück zum Zitat Bancroft CC, Chen Z, Dong G, Sunwoo JB, Yeh N, Park C, Van Waes C: Coexpression of proangiogenic factors IL-8 and VEGF by human head and neck squamous cell carcinoma involves coactivation by MEK-MAPK and IKK-NF-kappaB signal pathways. Clin Cancer Res. 2001, 7: 435-442.PubMed Bancroft CC, Chen Z, Dong G, Sunwoo JB, Yeh N, Park C, Van Waes C: Coexpression of proangiogenic factors IL-8 and VEGF by human head and neck squamous cell carcinoma involves coactivation by MEK-MAPK and IKK-NF-kappaB signal pathways. Clin Cancer Res. 2001, 7: 435-442.PubMed
38.
Zurück zum Zitat Lee AH, Iwakoshi NN, Anderson KC, Glimcher LH: Proteasome inhibitors disrupt the unfolded protein response in myeloma cells. Proc Natl Acad Sci U S A. 2003, 100: 9946-9951. 10.1073/pnas.1334037100.PubMedCentralCrossRefPubMed Lee AH, Iwakoshi NN, Anderson KC, Glimcher LH: Proteasome inhibitors disrupt the unfolded protein response in myeloma cells. Proc Natl Acad Sci U S A. 2003, 100: 9946-9951. 10.1073/pnas.1334037100.PubMedCentralCrossRefPubMed
39.
Zurück zum Zitat Mimnaugh EG, Xu W, Vos M, Yuan X, Isaacs JS, Bisht KS, Gius D, Neckers L: Simultaneous inhibition of hsp 90 and the proteasome promotes protein ubiquitination, causes endoplasmic reticulum-derived cytosolic vacuolization, and enhances antitumor activity. Mol Cancer Ther. 2004, 3: 551-566.CrossRefPubMed Mimnaugh EG, Xu W, Vos M, Yuan X, Isaacs JS, Bisht KS, Gius D, Neckers L: Simultaneous inhibition of hsp 90 and the proteasome promotes protein ubiquitination, causes endoplasmic reticulum-derived cytosolic vacuolization, and enhances antitumor activity. Mol Cancer Ther. 2004, 3: 551-566.CrossRefPubMed
40.
Zurück zum Zitat Denlinger CE, Rundall BK, Jones DR: Proteasome inhibition sensitizes non-small cell lung cancer to histone deacetylase inhibitor-induced apoptosis through the generation of reactive oxygen species. J Thorac Cardiovasc Surg. 2004, 128: 740-748. 10.1016/j.jtcvs.2004.07.010.CrossRefPubMed Denlinger CE, Rundall BK, Jones DR: Proteasome inhibition sensitizes non-small cell lung cancer to histone deacetylase inhibitor-induced apoptosis through the generation of reactive oxygen species. J Thorac Cardiovasc Surg. 2004, 128: 740-748. 10.1016/j.jtcvs.2004.07.010.CrossRefPubMed
41.
Zurück zum Zitat Fribley A, Zeng Q, Wang CY: Proteasome inhibitor PS-341 induces apoptosis through induction of endoplasmic reticulum stress-reactive oxygen species in head and neck squamous cell carcinoma cells. Mol Cell Biol. 2004, 24: 9695-9704. 10.1128/MCB.24.22.9695-9704.2004.PubMedCentralCrossRefPubMed Fribley A, Zeng Q, Wang CY: Proteasome inhibitor PS-341 induces apoptosis through induction of endoplasmic reticulum stress-reactive oxygen species in head and neck squamous cell carcinoma cells. Mol Cell Biol. 2004, 24: 9695-9704. 10.1128/MCB.24.22.9695-9704.2004.PubMedCentralCrossRefPubMed
42.
Zurück zum Zitat Kostova Z, Wolf DH: For whom the bell tolls: protein quality control of the endoplasmic reticulum and the ubiquitin-proteasome connection. EMBO J. 2003, 22: 2309-2317. 10.1093/emboj/cdg227.PubMedCentralCrossRefPubMed Kostova Z, Wolf DH: For whom the bell tolls: protein quality control of the endoplasmic reticulum and the ubiquitin-proteasome connection. EMBO J. 2003, 22: 2309-2317. 10.1093/emboj/cdg227.PubMedCentralCrossRefPubMed
43.
Zurück zum Zitat LeBlanc R, Catley LP, Hideshima T, Lentzsch S, Mitsiades CS, Mitsiades N, Neuberg D, Goloubeva O, Pien CS, Adams J, Gupta D, Richardson PG, Munshi NC, Anderson KC: Proteasome inhibitor PS-341 inhibits human myeloma cell growth in vivo and prolongs survival in a murine model. Cancer Res. 2002, 62: 4996-5000.PubMed LeBlanc R, Catley LP, Hideshima T, Lentzsch S, Mitsiades CS, Mitsiades N, Neuberg D, Goloubeva O, Pien CS, Adams J, Gupta D, Richardson PG, Munshi NC, Anderson KC: Proteasome inhibitor PS-341 inhibits human myeloma cell growth in vivo and prolongs survival in a murine model. Cancer Res. 2002, 62: 4996-5000.PubMed
44.
Zurück zum Zitat Satou Y, Nosaka K, Koya Y, Yasunaga JI, Toyokuni S, Matsuoka M: Proteasome inhibitor, bortezomib, potently inhibits the growth of adult T-cell leukemia cells both in vivo and in vitro. Leukemia. 2004, 18: 1357-1363. 10.1038/sj.leu.2403400.CrossRefPubMed Satou Y, Nosaka K, Koya Y, Yasunaga JI, Toyokuni S, Matsuoka M: Proteasome inhibitor, bortezomib, potently inhibits the growth of adult T-cell leukemia cells both in vivo and in vitro. Leukemia. 2004, 18: 1357-1363. 10.1038/sj.leu.2403400.CrossRefPubMed
45.
Zurück zum Zitat Teicher BA, Ara G, Herbst R, Palombella VJ, Adams J: The proteasome inhibitor PS-341 in cancer therapy. Clin Cancer Res. 1999, 5: 2638-2645.PubMed Teicher BA, Ara G, Herbst R, Palombella VJ, Adams J: The proteasome inhibitor PS-341 in cancer therapy. Clin Cancer Res. 1999, 5: 2638-2645.PubMed
46.
Zurück zum Zitat Williams S, Pettaway C, Song R, Papandreou C, Logothetis C, McConkey DJ: Differential effects of the proteasome inhibitor bortezomib on apoptosis and angiogenesis in human prostate tumor xenografts. Mol Cancer Ther. 2003, 2: 835-843.PubMed Williams S, Pettaway C, Song R, Papandreou C, Logothetis C, McConkey DJ: Differential effects of the proteasome inhibitor bortezomib on apoptosis and angiogenesis in human prostate tumor xenografts. Mol Cancer Ther. 2003, 2: 835-843.PubMed
47.
Zurück zum Zitat Nawrocki ST, Bruns CJ, Harbison MT, Bold RJ, Gotsch BS, Abbruzzese JL, Elliott P, Adams J, McConkey DJ: Effects of the proteasome inhibitor PS-341 on apoptosis and angiogenesis in orthotopic human pancreatic tumor xenografts. Mol Cancer Ther. 2002, 1: 1243-1253.PubMed Nawrocki ST, Bruns CJ, Harbison MT, Bold RJ, Gotsch BS, Abbruzzese JL, Elliott P, Adams J, McConkey DJ: Effects of the proteasome inhibitor PS-341 on apoptosis and angiogenesis in orthotopic human pancreatic tumor xenografts. Mol Cancer Ther. 2002, 1: 1243-1253.PubMed
48.
Zurück zum Zitat Bold RJ, Virudachalam S, McConkey DJ: Chemosensitization of pancreatic cancer by inhibition of the 26S proteasome. J Surg Res. 2001, 100: 11-17. 10.1006/jsre.2001.6194.CrossRefPubMed Bold RJ, Virudachalam S, McConkey DJ: Chemosensitization of pancreatic cancer by inhibition of the 26S proteasome. J Surg Res. 2001, 100: 11-17. 10.1006/jsre.2001.6194.CrossRefPubMed
49.
Zurück zum Zitat Amiri KI, Horton LW, LaFleur BJ, Sosman JA, Richmond A: Augmenting chemosensitivity of malignant melanoma tumors via proteasome inhibition: implication for bortezomib (VELCADE, PS-341) as a therapeutic agent for malignant melanoma. Cancer Res. 2004, 64: 4912-4918.CrossRefPubMed Amiri KI, Horton LW, LaFleur BJ, Sosman JA, Richmond A: Augmenting chemosensitivity of malignant melanoma tumors via proteasome inhibition: implication for bortezomib (VELCADE, PS-341) as a therapeutic agent for malignant melanoma. Cancer Res. 2004, 64: 4912-4918.CrossRefPubMed
50.
Zurück zum Zitat Mitsiades N, Mitsiades CS, Richardson PG, Poulaki V, Tai YT, Chauhan D, Fanourakis G, Gu X, Bailey C, Joseph M, Libermann TA, Schlossman R, Munshi NC, Hideshima T, Anderson KC: The proteasome inhibitor PS-341 potentiates sensitivity of multiple myeloma cells to conventional chemotherapeutic agents: therapeutic applications. Blood. 2003, 101: 2377-2380. 10.1182/blood-2002-06-1768.CrossRefPubMed Mitsiades N, Mitsiades CS, Richardson PG, Poulaki V, Tai YT, Chauhan D, Fanourakis G, Gu X, Bailey C, Joseph M, Libermann TA, Schlossman R, Munshi NC, Hideshima T, Anderson KC: The proteasome inhibitor PS-341 potentiates sensitivity of multiple myeloma cells to conventional chemotherapeutic agents: therapeutic applications. Blood. 2003, 101: 2377-2380. 10.1182/blood-2002-06-1768.CrossRefPubMed
51.
Zurück zum Zitat Kamat AM, Karashima T, Davis DW, Lashinger L, Bar-Eli M, Millikan R, Shen Y, Dinney CP, McConkey DJ: The proteasome inhibitor bortezomib synergizes with gemcitabine to block the growth of human 253JB-V bladder tumors in vivo. Mol Cancer Ther. 2004, 3: 279-290.PubMed Kamat AM, Karashima T, Davis DW, Lashinger L, Bar-Eli M, Millikan R, Shen Y, Dinney CP, McConkey DJ: The proteasome inhibitor bortezomib synergizes with gemcitabine to block the growth of human 253JB-V bladder tumors in vivo. Mol Cancer Ther. 2004, 3: 279-290.PubMed
52.
Zurück zum Zitat Small GW, Shi YY, Edmund NA, Somasundaram S, Moore DT, Orlowski RZ: Evidence that mitogen-activated protein kinase phosphatase-1 induction by proteasome inhibitors plays an antiapoptotic role. Mol Pharmacol. 2004, 66: 1478-1490. 10.1124/mol.104.003400.CrossRefPubMed Small GW, Shi YY, Edmund NA, Somasundaram S, Moore DT, Orlowski RZ: Evidence that mitogen-activated protein kinase phosphatase-1 induction by proteasome inhibitors plays an antiapoptotic role. Mol Pharmacol. 2004, 66: 1478-1490. 10.1124/mol.104.003400.CrossRefPubMed
53.
Zurück zum Zitat Nawrocki ST, Sweeney-Gotsch B, Takamori R, McConkey DJ: The proteasome inhibitor bortezomib enhances the activity of docetaxel in orthotopic human pancreatic tumor xenografts. Mol Cancer Ther. 2004, 3: 59-70.PubMed Nawrocki ST, Sweeney-Gotsch B, Takamori R, McConkey DJ: The proteasome inhibitor bortezomib enhances the activity of docetaxel in orthotopic human pancreatic tumor xenografts. Mol Cancer Ther. 2004, 3: 59-70.PubMed
54.
Zurück zum Zitat Pervan M, Pajonk F, Sun JR, Withers HR, McBride WH: Molecular pathways that modify tumor radiation response. Am J Clin Oncol. 2001, 24: 481-485. 10.1097/00000421-200110000-00013.CrossRefPubMed Pervan M, Pajonk F, Sun JR, Withers HR, McBride WH: Molecular pathways that modify tumor radiation response. Am J Clin Oncol. 2001, 24: 481-485. 10.1097/00000421-200110000-00013.CrossRefPubMed
55.
Zurück zum Zitat Mitsiades CS, Treon SP, Mitsiades N, Shima Y, Richardson P, Schlossman R, Hideshima T, Anderson KC: TRAIL/Apo2L ligand selectively induces apoptosis and overcomes drug resistance in multiple myeloma: therapeutic applications. Blood. 2001, 98: 795-804. 10.1182/blood.V98.3.795.CrossRefPubMed Mitsiades CS, Treon SP, Mitsiades N, Shima Y, Richardson P, Schlossman R, Hideshima T, Anderson KC: TRAIL/Apo2L ligand selectively induces apoptosis and overcomes drug resistance in multiple myeloma: therapeutic applications. Blood. 2001, 98: 795-804. 10.1182/blood.V98.3.795.CrossRefPubMed
56.
Zurück zum Zitat Sayers TJ, Brooks AD, Koh CY, Ma W, Seki N, Raziuddin A, Blazar BR, Zhang X, Elliott PJ, Murphy WJ: The proteasome inhibitor PS-341 sensitizes neoplastic cells to TRAIL-mediated apoptosis by reducing levels of c-FLIP. Blood. 2003, 102: 303-310. 10.1182/blood-2002-09-2975.CrossRefPubMed Sayers TJ, Brooks AD, Koh CY, Ma W, Seki N, Raziuddin A, Blazar BR, Zhang X, Elliott PJ, Murphy WJ: The proteasome inhibitor PS-341 sensitizes neoplastic cells to TRAIL-mediated apoptosis by reducing levels of c-FLIP. Blood. 2003, 102: 303-310. 10.1182/blood-2002-09-2975.CrossRefPubMed
57.
Zurück zum Zitat Yu C, Rahmani M, Conrad D, Subler M, Dent P, Grant S: The proteasome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitors to induce apoptosis in Bcr/Abl+ cells sensitive and resistant to STI571. Blood. 2003, 102: 3765-3774. 10.1182/blood-2003-03-0737.CrossRefPubMed Yu C, Rahmani M, Conrad D, Subler M, Dent P, Grant S: The proteasome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitors to induce apoptosis in Bcr/Abl+ cells sensitive and resistant to STI571. Blood. 2003, 102: 3765-3774. 10.1182/blood-2003-03-0737.CrossRefPubMed
58.
Zurück zum Zitat Mitsiades CS, Mitsiades NS, McMullan CJ, Poulaki V, Shringarpure R, Hideshima T, Akiyama M, Chauhan D, Munshi N, Gu X, Bailey C, Joseph M, Libermann TA, Richon VM, Marks PA, Anderson KC: Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications. Proc Natl Acad Sci U S A. 2004, 101: 540-545. 10.1073/pnas.2536759100.PubMedCentralCrossRefPubMed Mitsiades CS, Mitsiades NS, McMullan CJ, Poulaki V, Shringarpure R, Hideshima T, Akiyama M, Chauhan D, Munshi N, Gu X, Bailey C, Joseph M, Libermann TA, Richon VM, Marks PA, Anderson KC: Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications. Proc Natl Acad Sci U S A. 2004, 101: 540-545. 10.1073/pnas.2536759100.PubMedCentralCrossRefPubMed
59.
Zurück zum Zitat Denlinger CE, Keller MD, Mayo MW, Broad RM, Jones DR: Combined proteasome and histone deacetylase inhibition in non-small cell lung cancer. J Thorac Cardiovasc Surg. 2004, 127: 1078-1086. 10.1016/S0022-5223(03)01321-7.CrossRefPubMed Denlinger CE, Keller MD, Mayo MW, Broad RM, Jones DR: Combined proteasome and histone deacetylase inhibition in non-small cell lung cancer. J Thorac Cardiovasc Surg. 2004, 127: 1078-1086. 10.1016/S0022-5223(03)01321-7.CrossRefPubMed
60.
Zurück zum Zitat Pei XY, Dai Y, Grant S: Synergistic induction of oxidative injury and apoptosis in human multiple myeloma cells by the proteasome inhibitor bortezomib and histone deacetylase inhibitors. Clin Cancer Res. 2004, 10: 3839-3852.CrossRefPubMed Pei XY, Dai Y, Grant S: Synergistic induction of oxidative injury and apoptosis in human multiple myeloma cells by the proteasome inhibitor bortezomib and histone deacetylase inhibitors. Clin Cancer Res. 2004, 10: 3839-3852.CrossRefPubMed
61.
Zurück zum Zitat Sun K, Welniak LA, Panoskaltsis-Mortari A, O'Shaughnessy MJ, Liu H, Barao I, Riordan W, Sitcheran R, Wysocki C, Serody JS, Blazar BR, Sayers TJ, Murphy WJ: Inhibition of acute graft-versus-host disease with retention of graft-versus-tumor effects by the proteasome inhibitor bortezomib. Proc Natl Acad Sci U S A. 2004, 101: 8120-8125. 10.1073/pnas.0401563101.PubMedCentralCrossRefPubMed Sun K, Welniak LA, Panoskaltsis-Mortari A, O'Shaughnessy MJ, Liu H, Barao I, Riordan W, Sitcheran R, Wysocki C, Serody JS, Blazar BR, Sayers TJ, Murphy WJ: Inhibition of acute graft-versus-host disease with retention of graft-versus-tumor effects by the proteasome inhibitor bortezomib. Proc Natl Acad Sci U S A. 2004, 101: 8120-8125. 10.1073/pnas.0401563101.PubMedCentralCrossRefPubMed
62.
Zurück zum Zitat Wang CY, Cusack JCJ, Liu R, Baldwin ASJ: Control of inducible chemoresistance: enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-kappaB. Nat Med. 1999, 5: 412-417. 10.1038/10577.CrossRefPubMed Wang CY, Cusack JCJ, Liu R, Baldwin ASJ: Control of inducible chemoresistance: enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-kappaB. Nat Med. 1999, 5: 412-417. 10.1038/10577.CrossRefPubMed
63.
Zurück zum Zitat Mayo MW, Baldwin AS: The transcription factor NF-kappaB: control of oncogenesis and cancer therapy resistance. Biochim Biophys Acta. 2000, 1470: M55-M62.PubMed Mayo MW, Baldwin AS: The transcription factor NF-kappaB: control of oncogenesis and cancer therapy resistance. Biochim Biophys Acta. 2000, 1470: M55-M62.PubMed
64.
Zurück zum Zitat Fahy BN, Schlieman MG, Virudachalam S, Bold RJ: Schedule-dependent molecular effects of the proteasome inhibitor bortezomib and gemcitabine in pancreatic cancer. J Surg Res. 2003, 113: 88-95. 10.1016/S0022-4804(03)00201-4.CrossRefPubMed Fahy BN, Schlieman MG, Virudachalam S, Bold RJ: Schedule-dependent molecular effects of the proteasome inhibitor bortezomib and gemcitabine in pancreatic cancer. J Surg Res. 2003, 113: 88-95. 10.1016/S0022-4804(03)00201-4.CrossRefPubMed
Metadaten
Titel
Preclinical evaluation of the proteasome inhibitor bortezomib in cancer therapy
verfasst von
Mario Boccadoro
Gareth Morgan
Jamie Cavenagh
Publikationsdatum
01.12.2005
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2005
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/1475-2867-5-18

Weitere Artikel der Ausgabe 1/2005

Cancer Cell International 1/2005 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.