Open Access

Proteasome inhibitors in glioblastoma (Review)

Retraction in: /10.3892/ol.2017.6921

  • Authors:
    • Wen-Juan Huang
    • Wei-Wei Chen
    • Xia Zhang
  • View Affiliations

  • Published online on: January 11, 2017     https://doi.org/10.3892/ol.2017.5585
  • Pages: 1058-1062
  • Copyright: © Huang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Glioblastomas (GBM) are the tumors originating from the star shaped supportive cells in brain known as astrocytes. These tumors are highly cancerous as they have the ability to proliferate very quickly. New therapeutic strategies are being developed worldwide to fight against deadly GBM, which has median survival time of just 14 months. Proteasome inhibition is an upcoming strategy for GBM. Proteasome inhibition has shown promising results in cancers such as myeloma. However, in the recent past this form of therapy has also shown positive results in brain tumors in the form of elevated apoptosis. We searched the electronic database PubMed for pre-clinical as well as clinical controlled trials reporting importance of proteasome inhibitors during GBM. It was observed clearly that this approach is evolving and has been observed to be promising therapeutic avenue against GBM. Thus, the present review aims to enlighten the present views on use of proteasome inhibition strategy in the case of GBM.

Introduction

The prominent tumor at brain site with malignant nature is glioblastoma (GBM) with an incidence of 3 cases/100,000 person years (1,2). Earlier GBM was treated by surgical resection and radiation. However, in a recent past increased survival has been observed in GBM patients treated with radiation plus temozolomide, an alkylating agent that causes DNA damage-induced cancer cell death (3). Even with this improvement in treatment, however, the median survival of patients only increased incrementally to 14.2 months (3,4). The continued poor prognosis of GBM patients highlights the need for novel therapeutic strategies to enhance survival.

The extensive sequence information has allowed classification of GBM into 4 main molecular subtypes: the proneural subtype, which is characterized by aberrations in platelet-derived growth factor receptor-α (PDGFRA), isocitrate dehydrogenase-1 (IDH1), and tumor protein p53 (TP53); the classical subtype, which is characterized by aberrant epidermal growth factor receptor (EGFR) expression; the mesenchymal subtype, with defects in neurofibromin 1 (NF1) expression; and the neural subtype, which is associated with gene signatures closer to more differentiated cells such as astrocytes and neurons (5). Another effort to identify a large amount of data from sequencing of brain tumors is the Repository of Molecular Brain Neoplasia Data (REMBRANDT), which includes data for gene expression and clinical endpoints for over 800 samples from different kinds of brain tumors (6). Though these efforts have greatly contributed to our knowledge base on GBM, more recent studies have indicated that these large scale sequencing efforts can greatly underestimate the remarkable heterogeneity that occurs within individual GBM tumors. Single cell studies identified subpopulations of cells within individual tumors that had a single clonal origin, but had acquired expression of receptor tyrosine kinases from multiple tumor subtypes, particularly of EGFR, MET, and PDGFRA (7,8). Expression of these receptors was often mutually exclusive in the different subpopulations within the tumor. Additionally, another study performed single cell RNA sequencing in multiple cells for 5 different primary GBM samples and found notable heterogeneity within single tumors (9). Notably, they identified cells that represented different subtypes (mesenchymal, classical, proneural, and neural) within the same tumor.

This heterogeneity has important implications for therapeutic strategies, as it indicates that targeting just one or even two of these abnormalities may not be effective. The present body of knowledge emphasizes the complexity of GBM and highlights a need for therapeutic strategies that could target broad mechanisms at work in cancer cells that may have a variety of genetic mutations. One potential approach to this problem is targeting the ubiquitin-proteasome system for GBM therapy. In the present review we discuss important current aspects of the strategy and the promising inhibitors for GBM therapy.

Proteasome inhibitors

Inhibitors in use for proteasome inhibition therapy could be classified into 5 main classes. Reversible inhibitors are classified as either peptide aldehydes (e.g., MG132) or peptide boronates [e.g., bortezomib (BTZ)]. Irreversible inhibitors can be divided into β-lactones [e.g., marizomib (MRZ)], peptide vinyl sulfones, or peptide epoxyketones (e.g., carfilzomib) (10). The most well-established, clinically utilized proteasome inhibitor is the dipeptide boronic acid BTZ. Determination of the crystal structure of BTZ in complex with yeast 20S proteasomes revealed that the boronic acid moiety of BTZ interacts with amino acids surrounding the 20S proteasome active site threonine, forming a tetrahedral boronate adduct. BTZ also forms a hydrogen bridge between a hydroxyl group from the boronate group and the proteasome active site threonine itself (11). Previous studies of the binding mechanism have revealed that BTZ inhibits the proteasome in a manner that is slowly reversible. Crystal structure experiments as well as a study that utilized a probe for the active sites of the proteasome revealed that, while BTZ has the strongest affinity for binding and inhibiting β5, it can also inhibit β1 and β1i (12).

Studies of BTZ have identified its anticancer potential in a variety of tumors including colorectal cancer (13), pancreatic cancer (14,15), and lung cancer (16). BTZ has been particularly successful in myeloma, as it induces death in multiple myeloma cells at doses that are non-toxic to normal peripheral blood mononuclear cells, establishing it as a potential therapeutic agent for this disease (17). Subsequent clinical trials demonstrated therapeutic activity of BTZ (18). In the phase III Assessment of Proteasome Inhibition for Extending Remissions (APEX) trial, BTZ increased median survival from 23.7 months in patients receiving dexamethasone to 29.8 months (19). Additionally, BTZ treatment was associated with a 43% overall response rate and 9% complete response rate in the APEX trial.

The success of BTZ fueled development of other inhibitors in this class, including MRZ (formerly NPI-0052) (20). MRZ is a nonpeptide proteasome inhibitor that was isolated from the marine actinomycete Salinispora tropica and structurally resembles the natural proteasome inhibitor omuralide (21). MRZ contains a β-lactone ring with a chloride leaving group that is important for the irreversible inhibitory nature of MRZ; the leaving group allows formation of a cyclic ether with the active site threonine in the proteasome (22). In addition to causing more sustained proteasome inhibition compared to BTZ, MRZ also inhibits the β5 and β2 proteasome subunits at lower doses than BTZ (23). Differences in the mode of proteasome binding (irreversible for MRZ vs. reversible for BTZ) and target specificity (β5 and β2 for MRZ vs. β5 and β1 for BTZ) have led to some key differences in how these drugs induce death in cancer cells. While BTZ and MRZ both target the ‘standard’ 20S proteasome, specific inhibitors of the immunoproteasome have also been developed. IPSI-001, a specific inhibitor of β1i, induced death in myeloma cells and overcame resistance to BTZ (24). ONX-0914 (also known as PR-957) is another immunoproteasome-specific inhibitor. It specifically targets LMP7 (β5i), and most present studies have focused on the ability to attenuate immune-related diseases such as arthritis and colitis (25). Future investigations targeting alternative proteasome components in cancer are required.

Induction of apoptosis and autophagy by proteasome inhibitors in cancer

Proteasome inhibitors have been shown to induce caspase-dependent apoptosis in a variety of cancer types, including leukemia and myeloma (26). A family of cysteine proteases known as caspases carries out apoptosis. Caspases-2, 8 and 9 are initiator caspases; these proteases exist as inactive pro-caspases that are activated in response to signals that induce their recruitment to activation platforms and proteolytic processing of these caspases to their active forms (27). Specific apoptotic studies in other cancer models have found that BTZ and MRZ rely on different caspases; MRZ induced caspase-8-dependent apoptosis in leukemia and myeloma, while BTZ was more equally dependent on caspases-8 and 9 (26). Previous findings showed that BTZ can induce death in GBM cells as measured by MTT assay (28). BTZ also induces cell cycle arrest and cleavage of poly ADP ribose polymerase in GBM cells, suggesting that death may occur due to caspase activation and apoptosis but without clarifying the role of specific caspases (29). Preliminary evidence also suggested that MRZ induced death in glioma cells, but a more specific examination of the death mechanism is lacking (30). The dependence of proteasome inhibitors on activation of specific initiator caspases has not yet been reported in GBM.

Increases in reactive oxygen species (ROS)

One stimulus for proteasome inhibitor-induced death is increased ROS levels after proteasome inhibition. Proteasome inhibitors have been shown to increase ROS in numerous types of cancer, including multiple myeloma, mantle cell lymphoma, colon cancer, and lung cancer (31). Inhibition of mitochondrial electron transport chain components prevented BTZ-induced ROS increases, suggesting that ROS is likely produced by mitochondrial dysfunction. The importance of ROS was solidified by the fact that proteasome inhibitor-induced death in mantle cell lymphoma was blocked by treatment with the antioxidants glutathione ethyl ester (GSHEE) and N-acetyl cysteine, which acts as an antioxidant by increasing levels of the antioxidant glutathione (32). Some studies have presented conflicting findings. One study showed that lung cancer cells did not have increased ROS after BTZ treatment, and also were not protected from BTZ-induced death by NAC (33), while a conflicting study reported that induction of ROS and mitochondrial dysfunction were important events in BTZ-mediated apoptosis in lung cancer cells (16). These results suggest that effects on ROS and the protective effect of antioxidants may be cell-specific, possibly due to factors such as differences in basal antioxidant capacity in different cell types.

Stimulation of autophagy

Another pathway of cellular protein breakdown is autophagy, a process in which proteins and organelles are first engulfed via autophagosomes, leading to mortification. The role of autophagy in cancer and therapeutics is complex: Autophagy can aid survival by clearing cells of damaged proteins and aggregates, but may also promote death when stimulated at high levels (34). Proteasome inhibitors have been found to induce autophagy in a variety of cell types including prostate cancer (35) and melanoma (36). Induction of autophagy is generally thought to be a protective mechanism in cancer cells treated with proteasome inhibitors, and dual inhibition of the proteasome and autophagy has been shown to increase cell death (37). Increased death after dual inhibition of the proteasome and autophagy appears to be specific to transformed cells, possibly indicating an increased reliance of cancer cells on these cellular processes (38). Though it is possible that autophagy may also be responsible for cell death in some cases (39), careful studies are necessary to delineate whether this process is acting as a pro-death or pro-survival mechanism.

Clinical challenges in treatment of GBM with proteasome inhibitors

The blood-brain barrier (BBB)

The BBB forms a barrier that serves many functions, including restriction of the substances that are able to exit the capillaries in the brain and is formed by association among the endothelial cells (40). The BBB is, therefore, thought to prevent some therapeutic agents from being effectively delivered to lesions in the brain (41). One study in an orthotopic xenograft model of GBM investigated BTZ in combination with neural stem cells (NSCs) expressing TRAIL (42). NSCs have been shown to migrate to intracranial tumors, and expression of TRAIL allows them to trigger death receptors on cancer cells. This study found that BTZ increased 100-day survival by 20% in mice also receiving NSC-TRAIL. However, strong in vitro data showing the efficacy of the combination of BTZ and the histone deacetylase inhibitor (HDACi) vorinostat (43) did not translate in a clinical trial; a phase II study of BTZ and the HDACi vorinostat in relapsed GBM patients was closed at the interim analysis due to failure to prevent disease progression (44). BTZ increased the efficacy of NSC-TRAIL therapy in orthotopic brain tumors, but specific markers of proteasome inhibition were not reported in this study, so there was no direct measurement of proteasome inhibition (42). For MRZ, a previous study examined proteasome activity in mice from 10 min to 24 h following intravenous injection of 0.15 mg/kg MRZ and reported that MRZ did not decrease proteasome activity in the brain; however, this study was performed in mice without brain tumors, and therefore with intact BBB (45). Therefore, the present body of evidence concerning the extent of proteasome inhibition achieved by BTZ and MRZ in relevant orthotopic brain tumor models is incomplete.

Adverse effects

Between 33 and 66% of myeloma patients treated with BTZ experience peripheral neuropathy (46). However, its incidence is rarely reported in patients with solid tumors undergoing treatment with BTZ (47), it is still an important consideration for patients. Treatment with antioxidants such as vitamin E, NAC, and glutathione have produced promising results for decreasing peripheral neuropathy (48). However, supplementation with antioxidants must be implemented with caution. Previous findings have shown that vitamin C can directly inactivate BTZ, reducing its efficacy (33). Notably, a phase I trial in myeloma indicated that MRZ was not associated with peripheral neuropathy (49).

Drug resistance

The observed relapses in some patients on BTZ therapy are the result of drug resistance (33). Several mechanisms of BTZ resistance have been described. In some cases, cells induce changes to the proteasome itself; this may include upregulation of proteasome subunits, alterations in the composition of the proteasome subunit pool, or mutations that limit binding of BTZ (50). For these cases, resistance may be overcome by new-generation proteasome inhibitors such as MRZ, which targets the proteasome at lower concentrations and induces death by mechanisms different from BTZ, or immunoproteasome inhibitors, which target alternative catalytic subunits (51).

In other cases, resistance occurs due to differences in the cellular environment that attenuate the toxic functions of proteasome inhibition. For example, cells with a higher basal antioxidant capacity, such as elevated expression of the antioxidant transcription factor Nrf2, have been found to be more resistant to proteasome inhibitors (52). Also, activation of autophagy, an alternative cellular degradation pathway, was found to protect cells from proteasome inhibitors in prostate cancer (36). Together, these studies indicate that cells may have ways of escaping the toxic effects of proteasome inhibitors by modulating the cellular environment and inducing alternative pathways for protein disposal. Another route of resistance involves direct inhibition of apoptosis pathways. Overexpression of anti-apoptotic regulators of mitochondrial integrity such as Bcl-2 and Bcl-XL is seen in some types of cancer, including GBM, which could make these cells resistant to therapies that target the intrinsic pathway of apoptosis (53).

Need for combination treatment strategies

To maximize therapeutic efficacy and limit toxicity, many drugs used in the clinic are administered as part of rationally designed combination strategies. MRZ and BTZ are both good candidates for combination therapy strategies. An improved understanding of BTZ and MRZ and their cell death mechanisms in GBM will definitely aid in the design of rational combination strategies that potentiate the efficacy of these agents in the clinic.

One combination strategy that has been investigated in other cancer types is that of proteasome inhibitors with HDACi. Expression of HDACs has been shown to be altered in several types of cancer, including GBM (54). The first clinically relevant HDACi was the hydroxamic acid vorinostat (also called SAHA) (55). There are four classes of HDACs, and vorinostat inhibits classes I and II. Vorinostat leads to accumulation of acetylated histones, and alters expression of 2–10% of genes in transformed cells (56). Vorinostat induces death in a variety of cancer types, and it has FDA-approval for cutaneous T-cell lymphoma (57). Several other HDACi have been developed. Some HDACi have more narrow inhibitory profiles, such as the benzamide derivative entinostat, a specific inhibitor of class I HDACs (58). Panobinostat is currently in several clinical trials, including a phase I/II trial in combination with the angiogenesis inhibitor bevacizumab for malignant glioma (59).

Previous studies have indicated the efficacy of the combination of HDACi with proteasome inhibitors. The overlap between the mechanisms of HDACi and proteasome inhibitors may contribute toward their synergistic effects. Therefore, the combination of proteasome inhibitors and HDACi may be a potent combination in GBM.

Conclusion

The above citations have shown that proteasome inhibitory strategy especially in combination therapeutic mode is an upcoming efficient module for GBM management. However, further studies are required to establish it as a gold standard therapy against GBM.

References

1 

Gilbert AN, Shevin RS, Anderson JC, Langford CP, Eustace N, Gillespie GY, Singh R and Willey CD: Generation of microtumors using 3D human biogel culture system and patient-derived glioblastoma cells for kinomic profiling and drug response testing. J Vis Exp. Jun 9–2016.(Epub ahead of print). doi: 10.3791/54026. View Article : Google Scholar

2 

Hess KR, Broglio KR and Bondy ML: Adult glioma incidence trends in the United States, 1977–2000. Cancer. 101:2293–2299. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al: European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups; National Cancer Institute of Canada Clinical Trials Group: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Johnson DR and O'Neill BP: Glioblastoma survival in the United States before and during the temozolomide era. J Neurooncol. 107:359–364. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Verhaak RGW, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, Miller CR, Ding L, Golub T, Mesirov JP, et al: Cancer Genome Atlas Research Network: Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 17:98–110. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Madhavan S, Zenklusen JC, Kotliarov Y, Sahni H, Fine HA and Buetow K: Rembrandt: helping personalized medicine become a reality through integrative translational research. Mol Cancer Res. 7:157–167. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Snuderl M, Fazlollahi L, Le LP, Nitta M, Zhelyazkova BH, Davidson CJ, Akhavanfard S, Cahill DP, Aldape KD, Betensky RA, et al: Mosaic amplification of multiple receptor tyrosine kinase genes in glioblastoma. Cancer Cell. 20:810–817. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Szerlip NJ, Pedraza A, Chakravarty D, Azim M, McGuire J, Fang Y, Ozawa T, Holland EC, Huse JT, Jhanwar S, et al: Intratumoral heterogeneity of receptor tyrosine kinases EGFR and PDGFRA amplification in glioblastoma defines subpopulations with distinct growth factor response. Proc Natl Acad Sci USA. 109:3041–3046. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H, Cahill DP, Nahed BV, Curry WT, Martuza RL, et al: Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 344:1396–1401. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Kisselev AF and Goldberg AL: Proteasome inhibitors: from research tools to drug candidates. Chem Biol. 8:739–758. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Groll M, Berkers CR, Ploegh HL and Ovaa H: Crystal structure of the boronic acid-based proteasome inhibitor bortezomib in complex with the yeast 20S proteasome. Structure. 14:451–456. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Berkers CR, Verdoes M, Lichtman E, Fiebiger E, Kessler BM, Anderson KC, Ploegh HL, Ovaa H and Galardy PJ: Activity probe for in vivo profiling of the specificity of proteasome inhibitor bortezomib. Nat Methods. 2:357–362. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Cusack JC Jr, Liu R, Houston M, Abendroth K, Elliott PJ, Adams J and Baldwin AS Jr: Enhanced chemosensitivity to CPT-11 with proteasome inhibitor PS-341: implications for systemic nuclear factor-kappaB inhibition. Cancer Res. 61:3535–3540. 2001.PubMed/NCBI

14 

Nawrocki ST, Bruns CJ, Harbison MT, Bold RJ, Gotsch BS, Abbruzzese JL, Elliott P, Adams J and McConkey DJ: Effects of the proteasome inhibitor PS-341 on apoptosis and angiogenesis in orthotopic human pancreatic tumor xenografts. Mol Cancer Ther. 1:1243–1253. 2002.PubMed/NCBI

15 

Nawrocki ST, Sweeney-Gotsch B, Takamori R and McConkey DJ: The proteasome inhibitor bortezomib enhances the activity of docetaxel in orthotopic human pancreatic tumor xenografts. Mol Cancer Ther. 3:59–70. 2004.PubMed/NCBI

16 

Ling YH, Liebes L, Zou Y and Perez-Soler R: Reactive oxygen species generation and mitochondrial dysfunction in the apoptotic response to Bortezomib, a novel proteasome inhibitor, in human H460 non-small cell lung cancer cells. J Biol Chem. 278:33714–33723. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Hideshima T, Richardson P, Chauhan D, Palombella VJ, Elliott PJ, Adams J and Anderson KC: The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells. Cancer Res. 61:3071–3076. 2001.PubMed/NCBI

18 

Jagannath S, Barlogie B, Berenson J, Siegel D, Irwin D, Richardson PG, Niesvizky R, Alexanian R, Limentani SA, Alsina M, et al: A phase 2 study of two doses of bortezomib in relapsed or refractory myeloma. Br J Haematol. 127:165–172. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Richardson PG, Sonneveld P, Schuster M, Irwin D, Stadtmauer E, Facon T, Harousseau JL, Ben-Yehuda D, Lonial S, Goldschmidt H, et al: Extended follow-up of a phase 3 trial in relapsed multiple myeloma: final time-to-event results of the APEX trial. Blood. 110:3557–3560. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Potts BC, Albitar MX, Anderson KC, Baritaki S, Berkers C, Bonavida B, Chandra J, Chauhan D, Cusack JC Jr, Fenical W, et al: Marizomib, a proteasome inhibitor for all seasons: preclinical profile and a framework for clinical trials. Curr Cancer Drug Targets. 11:254–284. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Williams PG, Buchanan GO, Feling RH, Kauffman CA, Jensen PR and Fenical W: New cytotoxic salinosporamides from the marine Actinomycete Salinispora tropica. J Org Chem. 70:6196–6203. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Groll M, Huber R and Potts BCM: Crystal structures of Salinosporamide A (NPI-0052) and B (NPI-0047) in complex with the 20S proteasome reveal important consequences of beta-lactone ring opening and a mechanism for irreversible binding. J Am Chem Soc. 128:5136–5141. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Chauhan D, Catley L, Li G, Podar K, Hideshima T, Velankar M, Mitsiades C, Mitsiades N, Yasui H, Letai A, et al: A novel orally active proteasome inhibitor induces apoptosis in multiple myeloma cells with mechanisms distinct from Bortezomib. Cancer Cell. 8:407–419. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Kuhn DJ, Hunsucker SA, Chen Q, Voorhees PM, Orlowski M and Orlowski RZ: Targeted inhibition of the immunoproteasome is a potent strategy against models of multiple myeloma that overcomes resistance to conventional drugs and nonspecific proteasome inhibitors. Blood. 113:4667–4676. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Muchamuel T, Basler M, Aujay MA, Suzuki E, Kalim KW, Lauer C, Sylvain C, Ring ER, Shields J, Jiang J, et al: A selective inhibitor of the immunoproteasome subunit LMP7 blocks cytokine production and attenuates progression of experimental arthritis. Nat Med. 15:781–787. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Miller CP, Ban K, Dujka ME, McConkey DJ, Munsell M, Palladino M and Chandra J: NPI-0052, a novel proteasome inhibitor, induces caspase-8 and ROS-dependent apoptosis alone and in combination with HDAC inhibitors in leukemia cells. Blood. 110:267–277. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Boatright KM and Salvesen GS: Mechanisms of caspase activation. Curr Opin Cell Biol. 15:725–731. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Styczynski J, Olszewska-Slonina D, Kolodziej B, Napieraj M and Wysocki M: Activity of bortezomib in glioblastoma. Anticancer Res. 26:4499–4503. 2006.PubMed/NCBI

29 

Yin D, Zhou H, Kumagai T, Liu G, Ong JM, Black KL and Koeffler HP: Proteasome inhibitor PS-341 causes cell growth arrest and apoptosis in human glioblastoma multiforme (GBM). Oncogene. 24:344–354. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Vlashi E, Mattes M, Lagadec C, Donna LD, Phillips TM, Nikolay P, McBride WH and Pajonk F: Differential effects of the proteasome inhibitor NPI-0052 against glioma cells. Transl Oncol. 3:50–55. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Pei XY, Dai Y and 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. 10:3839–3852. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Pérez-Galán P, Roué G, Villamor N, Montserrat E, Campo E and Colomer D: The proteasome inhibitor bortezomib induces apoptosis in mantle-cell lymphoma through generation of ROS and Noxa activation independent of p53 status. Blood. 107:257–264. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Zou W, Yue P, Lin N, He M, Zhou Z, Lonial S, Khuri FR, Wang B and Sun SY: Vitamin C inactivates the proteasome inhibitor PS-341 in human cancer cells. Clin Cancer Res. 12:273–280. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Baehrecke EH: Autophagy: dual roles in life and death? Nat Rev Mol Cell Biol. 6:505–510. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Zhu K, Dunner K Jr and McConkey DJ: Proteasome inhibitors activate autophagy as a cytoprotective response in human prostate cancer cells. Oncogene. 29:451–462. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Selimovic D, Porzig BBOW, El-Khattouti A, Badura HE, Ahmad M, Ghanjati F, Santourlidis S, Haikel Y and Hassan M: Bortezomib/proteasome inhibitor triggers both apoptosis and autophagy-dependent pathways in melanoma cells. Cell Signal. 25:308–318. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Yao F, Wang G, Wei W, Tu Y, Tong H and Sun S: An autophagy inhibitor enhances the inhibition of cell proliferation induced by a proteasome inhibitor in MCF-7 cells. Mol Med Rep. 5:84–88. 2012.PubMed/NCBI

38 

Ding WX, Ni HM, Gao W, Chen X, Kang JH, Stolz DB, Liu J and Yin XM: Oncogenic transformation confers a selective susceptibility to the combined suppression of the proteasome and autophagy. Mol Cancer Ther. 8:2036–2045. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Belloni D, Veschini L, Foglieni C, Dell'Antonio G, Caligaris-Cappio F, Ferrarini M and Ferrero E: Bortezomib induces autophagic death in proliferating human endothelial cells. Exp Cell Res. 316:1010–1018. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Abbott NJ, Patabendige AAK, Dolman DEM, Yusof SR and Begley DJ: Structure and function of the blood-brain barrier. Neurobiol Dis. 37:13–25. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Zünkeler B, Carson RE, Olson J, Blasberg RG, DeVroom H, Lutz RJ, Saris SC, Wright DC, Kammerer W, Patronas NJ, et al: Quantification and pharmacokinetics of blood-brain barrier disruption in humans. J Neurosurg. 85:1056–1065. 1996. View Article : Google Scholar : PubMed/NCBI

42 

Balyasnikova IV, Ferguson SD, Han Y, Liu F and Lesniak MS: Therapeutic effect of neural stem cells expressing TRAIL and bortezomib in mice with glioma xenografts. Cancer Lett. 310:148–159. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Asklund T, Kvarnbrink S, Holmlund C, Wibom C, Bergenheim T, Henriksson R and Hedman H: Synergistic killing of glioblastoma stem-like cells by bortezomib and HDAC inhibitors. Anticancer Res. 32:2407–2413. 2012.PubMed/NCBI

44 

Friday BB, Anderson SK, Buckner J, Yu C, Giannini C, Geoffroy F, Schwerkoske J, Mazurczak M, Gross H, Pajon E, et al: Phase II trial of vorinostat in combination with bortezomib in recurrent glioblastoma: a north central cancer treatment group study. Neuro Oncol. 14:215–221. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Singh AV, Palladino MA, Lloyd GK, Potts BC, Chauhan D and Anderson KC: Pharmacodynamic and efficacy studies of the novel proteasome inhibitor NPI-0052 (marizomib) in a human plasmacytoma xenograft murine model. Br J Haematol. 149:550–559. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Berkowitz A and Walker S: Bortezomib-induced peripheral neuropathy in patients with multiple myeloma. Clin J Oncol Nurs. 16:86–89. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Argyriou AA, Iconomou G and Kalofonos HP: Bortezomib-induced peripheral neuropathy in multiple myeloma: a comprehensive review of the literature. Blood. 112:1593–1599. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Wolf S, Barton D, Kottschade L, Grothey A and Loprinzi C: Chemotherapy-induced peripheral neuropathy: prevention and treatment strategies. Eur J Cancer. 44:1507–1515. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Richardson PG, Spencer A, Cannell P, Harrison SJ, Catley L, Underhill C, Zimmerman TM, Hofmeister CC, Jakubowiak AJ, Laubach J, et al: Phase I clinical evaluation of twice-weekly marizomib (NPI-0052), a novel proteasome inhibitor, in patients with relapsed/refractory multiple myeloma (MM). Blood (ASH Annual Meeting abst). 118:3022011.

50 

Fuchs D, Berges C, Opelz G, Daniel V and Naujokat C: Increased expression and altered subunit composition of proteasomes induced by continuous proteasome inhibition establish apoptosis resistance and hyperproliferation of Burkitt lymphoma cells. J Cell Biochem. 103:270–283. 2008. View Article : Google Scholar : PubMed/NCBI

51 

Chauhan D, Hideshima T and Anderson KC: A novel proteasome inhibitor NPI-0052 as an anticancer therapy. Br J Cancer. 95:961–965. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Weniger MA, Rizzatti EG, Pérez-Galán P, Liu D, Wang Q, Munson PJ, Raghavachari N, White T, Tweito MM, Dunleavy K, et al: Treatment-induced oxidative stress and cellular antioxidant capacity determine response to bortezomib in mantle cell lymphoma. Clin Cancer Res. 17:5101–5112. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Liu G, Yuan X, Zeng Z, Tunici P, Ng H, Abdulkadir IR, Lu L, Irvin D, Black KL and Yu JS: Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer. 5:672006. View Article : Google Scholar : PubMed/NCBI

54 

Lucio-Eterovic AK, Cortez MA, Valera ET, Motta FJ, Queiroz RG, Machado HR, Carlotti CG Jr, Neder L, Scrideli CA and Tone LG: Differential expression of 12 histone deacetylase (HDAC) genes in astrocytomas and normal brain tissue: class II and IV are hypoexpressed in glioblastomas. BMC Cancer. 8:2432008. View Article : Google Scholar : PubMed/NCBI

55 

Richon VM, Webb Y, Merger R, Sheppard T, Jursic B, Ngo L, Civoli F, Breslow R, Rifkind RA and Marks PA: Second generation hybrid polar compounds are potent inducers of transformed cell differentiation. Proc Natl Acad Sci USA. 93:5705–5708. 1996. View Article : Google Scholar : PubMed/NCBI

56 

Marks PA and Breslow R: Dimethyl sulfoxide to vorinostat: development of this histone deacetylase inhibitor as an anticancer drug. Nat Biotechnol. 25:84–90. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Mann BS, Johnson JR, Cohen MH, Justice R and Pazdur R: FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist. 12:1247–1252. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Saito A, Yamashita T, Mariko Y, Nosaka Y, Tsuchiya K, Ando T, Suzuki T, Tsuruo T and Nakanishi O: A synthetic inhibitor of histone deacetylase, MS-27-275, with marked in vivo antitumor activity against human tumors. Proc Natl Acad Sci USA. 96:4592–4597. 1999. View Article : Google Scholar : PubMed/NCBI

59 

Lee EQ, Reardon DA, Schiff D, Drappatz J, Muzikansky A, Hammond S, Grimm SA, Norden AD, Beroukhim R, McCluskey CS, et al: Interim analysis of a phase I/II study of panobinostat in combination with bevacizumab for recurrent glioblastoma. J Clin Oncol. 31:ASCO abst. 2013.

Related Articles

Journal Cover

March-2017
Volume 13 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang W, Chen W and Zhang X: Proteasome inhibitors in glioblastoma (Review) Retraction in /10.3892/ol.2017.6921. Oncol Lett 13: 1058-1062, 2017
APA
Huang, W., Chen, W., & Zhang, X. (2017). Proteasome inhibitors in glioblastoma (Review) Retraction in /10.3892/ol.2017.6921. Oncology Letters, 13, 1058-1062. https://doi.org/10.3892/ol.2017.5585
MLA
Huang, W., Chen, W., Zhang, X."Proteasome inhibitors in glioblastoma (Review) Retraction in /10.3892/ol.2017.6921". Oncology Letters 13.3 (2017): 1058-1062.
Chicago
Huang, W., Chen, W., Zhang, X."Proteasome inhibitors in glioblastoma (Review) Retraction in /10.3892/ol.2017.6921". Oncology Letters 13, no. 3 (2017): 1058-1062. https://doi.org/10.3892/ol.2017.5585