Cancer Letters

Cancer Letters

Volume 293, Issue 1, 1 July 2010, Pages 15-22
Cancer Letters

Mini-review
Proteasome inhibition: A new therapeutic strategy to cancer treatment

https://doi.org/10.1016/j.canlet.2009.12.002Get rights and content

Abstract

The ubiquitin–proteasome system is a major pathway for protein degradation. Targeting this pathway using proteasome inhibitors represents a novel approach for the treatment of cancer. Proteasome inhibitors lower cell proliferation and induce apoptosis in solid and hematologic malignancies through multiple mechanisms, including stabilization of cell cycle regulators and pro-apoptotic factors, stimulation of bone morphogenetic protein signaling, inhibition of protein translation, and sensitization to ligand-induced apoptosis. In this connection, proteasome inhibition activates macroautophagy, a compensatory protein degradation system, as well as other pro-survival signaling pathways. Inhibition of these auto-protective responses sensitizes cancer cells to the anti-proliferative effects of proteasome inhibitors.

Introduction

The ubiquitin–proteasome system is the major catabolic pathway for degradation of short-lived and misfolded proteins [1]. Proteins to be degraded through this pathway first undergo polyubiquitination followed by recognition and proteolysis by proteasome, a barrel-shaped multimeric protein complex. The process of ubiquitination involves several enzymes: E1 (ubiquitin-activating), E2 (ubiquitin-conjugating) and E3 (ubiquitin-ligase) enzymes (Fig. 1). The E1 enzyme hydrolyses ATP and forms a thioester linkage with the ubiquitin molecule. The activated ubiquitin is then transferred to the E2 enzyme. However, it is the E3 enzyme, which determines the substrate specificity, transfers the ubiquitin to the lysine residue on the target protein. The ubiquitin then serves as the substrate for another round of ubiquitination, forming a polyubiquitin chain on the target protein. Chain of four or more ubiquitin molecules allows the target protein to be shuttled to proteasome for degradation [2]. The 26S proteasome consists of one 20S core structure and two 19S regulatory caps. The 20S core structure is composed of four stacked rings made of different α (structural) and β (catalytic) subunits while the 19S regulatory cap consists of 19 subunits divided into a 10-protein α ring and a 9-protein ubiquitin-binding lid [3], [4]. In mammalian cells, β1, β2, and β5 subunits of the 20S core structure account for the caspase-like, trypsin-like and chymotrypsin-like activities of proteasome, respectively [4].

The increasing interest in the role of the ubiquitin–proteasome system in carcinogenesis has led to the discovery that many proteins involved in the regulation of cell proliferation and apoptosis are degraded through this pathway [5]. It therefore comes as no surprise that inhibition of the ubiquitin–proteasome system substantially alters cancer growth. In this respect, the ubiquitin–proteasome system can be targeted at different points, such as the E1 enzyme or specific E3 enzyme [6], [7]. Nevertheless, prevention of proteasomal degradation of ubiquitinated proteins by proteasome inhibitors remains the most common approach. Therapeutically, proteasome inhibitors display a broad-spectrum anti-proliferative or pro-apoptotic activity in vitro against different types of hematological and solid malignancies (Table 1) [8], [9], [10], [11], [12], [13], [14], [15], [16], [17], [18], [19], [20], [21], [22], [23], [24], [25], [26], [27], [28], [29], [30], [31], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45]. Proteasome inhibitors also sensitize cancer cells to the antitumor effects of conventional chemotherapeutics [46] and novel targeted cancer therapy [47], [48] as well as irradiation [49]. Clinically, the proteasome inhibitor bortezomib has been FDA-approved for the treatment of multiple myeloma and mantle cell lymphoma [50]. Clinical trials evaluating the efficacies of proteasome inhibitors for the treatment of solid tumors and other hematological malignancies are in progress [5], [51], [52]. However, it is worthwhile to notice that solid tumors in general, with the exception of prostate cancer and non-small cell lung carcinoma, do not respond too well to bortezomib and further studies are needed. The most common adverse drug reactions associated with proteasome inhibitors include peripheral neuropathy and myelosuppression, which are usually self-limiting and mild relative to treatment options available to advanced cancer patients [52].

Section snippets

Principle of actions of proteasome inhibitors

The proposed mechanisms by which proteasome inhibition mediates its antitumor effects are listed in Table 2 [11], [35]. Because of the enormity of the possible mechanisms, it is unwise to discuss each of them in detail in this review. Instead, several pathways, which are of biologic significance or novelty, will be discussed.

Induction of macroautophagy

Macroautophagy is a catabolic process by which the cell degrades its intracellular content and damaged organelles through the lysosomal system. In this capacity, macroautophagy serves complementarily with the ubiquitin–proteasome system as the two major protein degradation systems in mammalian cells [2]. The role of macroautophagy in carcinogenesis is paradoxical. As a tumor-suppressing mechanism, overactivation of macroautophagy induces cell death [110] and mutations of macroautophagy-related

Conclusive remarks

Proteasome has emerged as a novel target in cancer therapy. The use of proteasome inhibitor has yielded some success in the management of some forms of hematological malignancies and may be effective against solid tumors. It triggers a mixed repertoire of tumor-suppressing and pro-survival pathways in cancer cells. Their relative importance in the antitumor activity of and resistance to proteasome inhibitors remains to be elucidated. Combinational therapy targeting the auto-regulatory

Conflicts of interest

None declared.

Acknowledgements

This work was supported by research Grant from National Basic Research Program of China (973 Program, 2010CB529305).

References (119)

  • N. Mitsiades et al.

    The proteasome inhibitor PS-341 potentiates sensitivity of multiple myeloma cells to conventional chemotherapeutic agents: therapeutic applications

    Blood

    (2003)
  • C.P. Miller et al.

    NPI-0052, a novel proteasome inhibitor,induces caspase-8 and ROS-dependent apoptosis alone and in combination with HDAC inhibitors in leukemia cells

    Blood

    (2007)
  • A. Goel et al.

    PS-341-mediated selective targeting of multiple myeloma cells by synergistic increase in ionizing radiation-induced apoptosis

    Exp. Hematol.

    (2005)
  • S. Cresta et al.

    Phase I study of bortezomib with weekly paclitaxel in patients with advanced solid tumours

    Eur. J. Cancer

    (2008)
  • I. Nickeleit et al.

    Argyrin a reveals a critical role for the tumor suppressor protein p27(kip1) in mediating antitumor activities in response to proteasome inhibition

    Cancer Cell

    (2008)
  • C.P. Miller et al.

    NPI-0052, a novel proteasome inhibitor, induces caspase-8 and ROS-dependent apoptosis alone and in combination with HDAC inhibitors in leukemia cells

    Blood

    (2007)
  • W.K. Wu et al.

    Repression of protein translation and mTOR signaling by proteasome inhibitor in colon cancer cells

    Biochem. Biophys. Res. Commun.

    (2009)
  • K.F. Chen et al.

    Bortezomib overcomes tumor necrosis factor-related apoptosis-inducing ligand resistance in hepatocellular carcinoma cells in part through the inhibition of the phosphatidylinositol 3-kinase/Akt pathway

    J. Biol. Chem.

    (2009)
  • C.P. Miller et al.

    Caspase-8 dependent histone acetylation by a novel proteasome inhibitor, NPI-0052: a mechanism for synergy in leukemia cells

    Blood

    (2009)
  • J.H. Lim et al.

    Transcriptional repression of E2F gene by proteasome inhibitors in human osteosarcoma cells

    Biochem. Biophys. Res. Commun.

    (2004)
  • T. Hideshima et al.

    Bortezomib induces canonical nuclear factor-kappaB activation in multiple myeloma cells

    Blood

    (2009)
  • M.V. Blagosklonny et al.

    Proteasome-dependent regulation of p21WAF1/CIP1 expression

    Biochem. Biophys. Res. Commun.

    (1996)
  • P. Aza-Blanc et al.

    Proteolysis that is inhibited by hedgehog targets Cubitus interruptus protein to the nucleus and converts it to a repressor

    Cell

    (1997)
  • C.H. Chen et al.

    Nuclear trafficking of cubitus interruptus in the transcriptional regulation of hedgehog target gene expression

    Cell

    (1999)
  • W.K. Wu et al.

    Proteasome inhibitor MG-132 lowers gastric adenocarcinoma TMK1 cell proliferation via bone morphogenetic protein signaling

    Biochem. Biophys. Res. Commun.

    (2008)
  • A.L. Schwartz et al.

    The ubiquitin–proteasome pathway and pathogenesis of human diseases

    Annu. Rev. Med.

    (1999)
  • D.C. Rubinsztein

    The roles of intracellular protein-degradation pathways in neurodegeneration

    Nature

    (2006)
  • A.J. Marques et al.

    Catalytic mechanism and assembly of the proteasome

    Chem. Rev.

    (2009)
  • P.G. Richardson et al.

    Bortezomib: proteasome inhibition as an effective anticancer therapy

    Annu. Rev. Med.

    (2006)
  • Y. Yang et al.

    Inhibitors of ubiquitin-activating enzyme (E1), a new class of potential cancer therapeutics

    Cancer Res.

    (2007)
  • X. Liu et al.

    The proteasome inhibitor PS-341 (bortezomib) up-regulates DR5 expression leading to induction of apoptosis and enhancement of TRAIL-induced apoptosis despite up-regulation of c-FLIP and survivin expression in human NSCLC cells

    Cancer Res.

    (2007)
  • J. Sun et al.

    CEP1612, a dipeptidyl proteasome, inhibitor, induces p21WAF1 and p27KIP1 expression and apoptosis and inhibits the growth of the human lung adenocarcinoma A-549 in nude mice

    Cancer Res.

    (2001)
  • A. Fribley et al.

    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)
  • W.K. Wu et al.

    Bone morphogenetic protein signalling is required for the anti-mitogenic effect of the proteasome inhibitor MG-132 on colon cancer cells

    Br. J. Pharmacol.

    (2008)
  • X.M. Fan et al.

    Inhibition of proteasome function induced apoptosis in gastric cancer

    Int. J. Cancer

    (2001)
  • K.F. Chen et al.

    Down-regulation of phospho-Akt is a major molecular determinant of bortezomib-induced apoptosis in hepatocellular carcinoma cells

    Cancer Res.

    (2008)
  • Y. Ustundag et al.

    Proteasome inhibition-induces endoplasmic reticulum dysfunction and cell death of human cholangiocarcinoma cells

    World J. Gastroenterol.

    (2007)
  • A. Märten et al.

    Bortezomib is ineffective in an orthotopic mouse model of pancreatic adenocarcinoma

    Mol. Cancer Ther.

    (2008)
  • C.S. Mitsiades et al.

    Antitumor effects of the proteasome inhibitor bortezomib in medullary and anaplastic thyroid carcinoma cells in vitro

    J. Clin. Endocrinol. Metab.

    (2006)
  • J. An et al.

    Epidermal growth factor receptor inhibition sensitizes renal cell carcinoma cells to the cytotoxic effects of bortezomib

    Mol. Cancer Ther.

    (2007)
  • A.M. Kamat et al.

    The proteasome inhibitor bortezomib synergizes with gemcitabine to block the growth of human 253JB-V bladder tumors in vivo

    Mol. Cancer Ther.

    (2004)
  • R.B. Bazzaro et al.

    Ubiquitin proteasome system stress underlies synergistic killing of ovarian cancer cells by bortezomib and a novel HDAC6 inhibitor

    Clin. Cancer Res.

    (2008)
  • F. Cardoso et al.

    Bortezomib (PS-341, Velcade) increases the efficacy of trastuzumab (Herceptin) in HER-2-positive breast cancer cells in a synergistic manner

    Mol. Cancer Ther.

    (2006)
  • D.C. Birle et al.

    Suppression of the hypoxia-inducible factor-1 response in cervical carcinoma xenografts by proteasome inhibitors

    Cancer Res.

    (2007)
  • T. Ikezoe et al.

    Proteasome inhibitor PS-341 down-regulates prostate-specific antigen (PSA) and induces growth arrest and apoptosis of androgen-dependent human prostate cancer LNCaP cells

    Cancer Sci.

    (2004)
  • C. Brignole et al.

    Effect of bortezomib on human neuroblastoma cell growth, apoptosis, and angiogenesis

    J. Natl. Cancer Inst.

    (2006)
  • D. Yin et al.

    Proteasome inhibitor PS-341 causes cell growth arrest and apoptosis in human glioblastoma multiforme (GBM)

    Oncogene

    (2005)
  • E. Taniguchi et al.

    Bortezomib reverses a post-translational mechanism of tumorigenesis for patched1 haplo insufficiency in medulloblastoma

    Pediatr. Blood Cancer

    (2009)
  • V. Poulaki et al.

    The proteasome inhibitor bortezomib induces apoptosis in human retinoblastoma cell lines in vitro

    Invest. Ophthalmol. Vis. Sci.

    (2007)
  • J. Størling et al.

    Antitumorigenic effect of proteasome inhibitors on insulinoma cells

    Endocrinology

    (2005)
  • Cited by (91)

    • Best of most possible worlds: Hybrid gene therapy vectors based on parvoviruses and heterologous viruses

      2021, Molecular Therapy
      Citation Excerpt :

      Specifically, the two teams consistently reported that, akin to AAV and other parvoviruses,192 also AAVP is susceptible to proteasomal degradation and that proteasome inhibition enhances targeted tumor cell killing by AAVP in vitro and in vivo. Investigation of this particular pathway made sense considering that proteasome activity is frequently elevated in tumor cells,193 implying that it may pose a critical barrier for AAVP in these cells. Indeed, transduction was improved in the presence of two well-known and widely used proteasome inhibitors, MG132 and LLnL (peptide aldehyde inhibitors that block the 26S proteasome subunit).191

    • Ligand-directed tumor targeting with hybrid viral phage nanoparticles

      2018, Drug Targeting and Stimuli Sensitive Drug Delivery Systems
    View all citing articles on Scopus
    View full text