Molecular mechanisms of bortezomib resistant adenocarcinoma cells

Erika Suzuki, Susan Demo, Edgar Deu, Jonathan Keats, Shirin Arastu-Kapur, P Leif Bergsagel, Mark K Bennett, Christopher J Kirk, Erika Suzuki, Susan Demo, Edgar Deu, Jonathan Keats, Shirin Arastu-Kapur, P Leif Bergsagel, Mark K Bennett, Christopher J Kirk

Abstract

Bortezomib (Velcade™) is a reversible proteasome inhibitor that is approved for the treatment of multiple myeloma (MM). Despite its demonstrated clinical success, some patients are deprived of treatment due to primary refractoriness or development of resistance during therapy. To investigate the role of the duration of proteasome inhibition in the anti-tumor response of bortezomib, we established clonal isolates of HT-29 adenocarcinoma cells adapted to continuous exposure of bortezomib. These cells were ~30-fold resistant to bortezomib. Two novel and distinct mutations in the β5 subunit, Cys63Phe, located distal to the binding site in a helix critical for drug binding, and Arg24Cys, found in the propeptide region were found in all resistant clones. The latter mutation is a natural variant found to be elevated in frequency in patients with MM. Proteasome activity and levels of both the constitutive and immunoproteasome were increased in resistant cells, which correlated to an increase in subunit gene expression. These changes correlated with a more rapid recovery of proteasome activity following brief exposure to bortezomib. Increased recovery rate was not due to increased proteasome turnover as similar findings were seen in cells co-treated with cycloheximide. When we exposed resistant cells to the irreversible proteasome inhibitor carfilzomib we noted a slower rate of recovery of proteasome activity as compared to bortezomib in both parental and resistant cells. Importantly, carfilzomib maintained its cytotoxic potential in the bortezomib resistant cell lines. Therefore, resistance to bortezomib, can be overcome with irreversible inhibitors, suggesting prolonged proteasome inhibition induces a more potent anti-tumor response.

Conflict of interest statement

Competing Interests: Christopher J. Kirk, Erika Suzuki and Shirin Arastu-Kapur are current employees of Onyx Pharmaceuticals. There are no patents to declare, however this study is in support of the mechanism of action of Carfilzomib, a product currently in development from Onyx. This does not alter the authors' adherence to all the PLoS ONE policies on sharing data and materials.

© 2011 Suzuki et al.

Figures

Figure 1. Altered proteasome expression in bortezomib-resistant…
Figure 1. Altered proteasome expression in bortezomib-resistant HT-29 cells.
(A) Parental (▪,□), BR100 (▴, Δ), and BR200 cells (•,○) were cultured for 3 or 40 days in the absence of drug prior to exposure to varying concentrations (1 nM–1 µM) of bortezomib (closed symbols) or carfilzomib (open symbols) for 72 hrs. Viability was normalized to DMSO controls and data are presented as mean viability of triplicate cultures (± S.E.M). Data is from 1 of 3 replicate experiments with similar results. (B) Western blot analysis of constitutive and immunoproteasome active site subunits in parental (1), BR100 (2), and BR200 (3) cells cultured in the absence of drug for 14 days. β-actin or GAPDH was used as an to verify equal loading. (C) Constitutive and immunoproteasome subunit levels (ng subunit/µg total protein) in parental, BR100 and BR200 cell lines cultured in the absence of drug for 14 or 40 days were measured using ProCISE. Data at day 14 are presented as mean ± S.D. of 3 independent experiments. *** = P<0.001 by one-way ANOVA followed by Bonferroni's Multiple Comparison Test. Data at day 40 is representative of one experiment.
Figure 2. Increased proteasome turnover in bortezomib-resistant…
Figure 2. Increased proteasome turnover in bortezomib-resistant cells.
(A) Parental (▪,□) and BR200 cells (•,○) were cultured for 3 days in the absence of drug prior to exposure to varying concentrations (1 nM–1 µM) of bortezomib (closed symbols) or carfilzomib (open symbols) for 1 hr. Proteasome chymotrypsin-like activity was measured using LLVY-AMC as substrate and specific activity values were normalized to DMSO controls. Data are presented as the mean relative activity (± S.E.M.) and is representative of 2 replicate experiments. (B) Parental (▪,□) and BR200 cells (•,○) were exposed to 100 nM bortezomib (closed symbols) or carfilzomib (open symbols) for 1 hr, washed and cultured in drug free media with or without cycloheximide for 1, 2, 4, 6, 8, 12 , and 24 hr prior to measurement of chymotrypsin-like activity. Parental cells (Δ) and BR200 (◊) cells treated with CHX alone in the absence of drug are included as additional controls. Data are presented as the mean relative activity (± S.E.M.) and is representative of 2 replicate experiments. (C) Relative chymotrypsin-like activity in parental and BR200 cells at 4 or 8 hr after a 1 hr pulse exposure to 100 nM bortezomib or carfilzomib in the presence or absence of cycloheximide. ** = P<0.01; *** = P<0.001 by one-way ANOVA followed by Newman-Keuls post-hoc comparisons.
Figure 3. Cys63Phe mutation is a critical…
Figure 3. Cys63Phe mutation is a critical structural mutation.
(A) Cys63Phe mutation is part of a critical helix at the α5 (cyan)/β5 (gray) subunit interface subunit that directly points into the active site, with Ala49/50 making direct contacts with proteasome inhibitors. (B) Mutant PRE unbound (purple) overlaid with mutant PRE bound to bortezomib (olive) and epoxomicin (midnight blue). (C) Active site view of bortezomib and epoxomicin bound to the wild-type (blue) and mutant (red) PRE2 structures.

References

    1. Ciechanover A. Proteolysis: from the lysosome to ubiquitin and the proteasome. Nat Rev Mol Cell Biol. 2005;6:79–87.
    1. Murata S, Yashiroda H, Tanaka K. Molecular mechanisms of proteasome assembly. Nat Rev Mol Cell Biol. 2009;10:104–115.
    1. McConkey DJ, Zhu K. Mechanisms of proteasome inhibitor action and resistance in cancer. Drug Resist Updat. 2008;11:164–179.
    1. Adams J. Development of the proteasome inhibitor PS-341. Oncologist. 2002;7:9–16.
    1. Bennett MK, Kirk CJ. Development of proteasome inhibitors in oncology and autoimmune diseases. Curr Opin Drug Discov Devel. 2008;11:616–625.
    1. Dick LR, Fleming PE. Building on bortezomib: second-generation proteasome inhibitors as anti-cancer therapy. Drug Discov Today. 2010;15:243–249.
    1. Kumar S, Rajkumar SV. Many facets of bortezomib resistance/susceptibility. Blood. 2008;112:2177–2178.
    1. Shah JJ, Orlowski RZ. Proteasome inhibitors in the treatment of multiple myeloma. Leukemia. 2009;23:1964–1979.
    1. Mulligan G, Mitsiades C, Bryant B, Zhan F, Chng WJ, et al. Gene expression profiling and correlation with outcome in clinical trials of the proteasome inhibitor bortezomib. Blood. 2007;109:3177–3188.
    1. Kraus M, Ruckrich T, Reich M, Gogel J, Beck A, et al. Activity patterns of proteasome subunits reflect bortezomib sensitivity of hematologic malignancies and are variable in primary human leukemia cells. Leukemia. 2007;21:84–92.
    1. Lu S, Chen Z, Yang J, Chen L, Gong S, et al. Overexpression of the PSMB5 gene contributes to bortezomib resistance in T-lymphoblastic lymphoma/leukemia cells derived from Jurkat line. Exp Hematol. 2008;36:1278–1284.
    1. Lu S, Yang J, Song X, Gong S, Zhou H, et al. Point mutation of the proteasome beta5 subunit gene is an important mechanism of bortezomib resistance in bortezomib-selected variants of Jurkat T cell lymphoblastic lymphoma/leukemia line. J Pharmacol Exp Ther. 2008;326:423–431.
    1. Lu S, Yang J, Chen Z, Gong S, Zhou H, et al. Different mutants of PSMB5 confer varying bortezomib resistance in T lymphoblastic lymphoma/leukemia cells derived from the Jurkat cell line. Exp Hematol. 2009;37:831–837.
    1. Oerlemans R, Franke NE, Assaraf YG, Cloos J, van ZI, et al. Molecular basis of bortezomib resistance: proteasome subunit beta5 (PSMB5) gene mutation and overexpression of PSMB5 protein. Blood. 2008;112:2489–2499.
    1. Ruckrich T, Kraus M, Gogel J, Beck A, Ovaa H, et al. Characterization of the ubiquitin-proteasome system in bortezomib-adapted cells. Leukemia. 2009;23:1098–1105.
    1. Parlati F, Lee SJ, Aujay M, Suzuki E, Levitsky K, et al. Carfilzomib can induce tumor cell death through selective inhibition of the chymotrypsin-like activity of the proteasome. Blood. 2009;114:3439–3447.
    1. Oerlemans R, Franke NE, Assaraf YG, Cloos J, van ZI, et al. Molecular basis of bortezomib resistance: proteasome subunit beta5 (PSMB5) gene mutation and overexpression of PSMB5 protein. Blood. 2008;112:2489–2499.
    1. Bianchi G, Oliva L, Cascio P, Pengo N, Fontana F, et al. The proteasome load versus capacity balance determines apoptotic sensitivity of multiple myeloma cells to proteasome inhibition. Blood. 2009;113:3040–3049.
    1. Ri M, Iida S, Nakashima T, Miyazaki H, Mori F, et al. Bortezomib-resistant myeloma cell lines: a role for mutated PSMB5 in preventing the accumulation of unfolded proteins and fatal ER stress. Leukemia. 2010;24:1506–1512.
    1. Politou M, Karadimitris A, Terpos E, Kotsianidis I, Apperley JF, et al. No evidence of mutations of the PSMB5 (beta-5 subunit of proteasome) in a case of myeloma with clinical resistance to Bortezomib. Leuk Res. 2006;30:240–241.
    1. Demo SD, Kirk CJ, Aujay MA, Buchholz TJ, Dajee M, et al. Antitumor activity of PR-171, a novel irreversible inhibitor of the proteasome. Cancer Res. 2007;67:6383–6391.
    1. Kraus M, Ruckrich T, Reich M, Gogel J, Beck A, et al. Activity patterns of proteasome subunits reflect bortezomib sensitivity of hematologic malignancies and are variable in primary human leukemia cells. Leukemia. 2007;21:84–92.
    1. Groll M, Berkers CR, Ploegh HL, Ovaa H. Crystal structure of the boronic acid-based proteasome inhibitor bortezomib in complex with the yeast 20S proteasome. Structure. 2006;14:451–456.
    1. Arastu-Kapur S, Anderl JL, Kraus M, Parlati F, Shenk KD, et al. Non-proteasomal targets of the proteasome inhibitors bortezomib and carfilzomib: a link to clinical adverse events. Clin Cancer Res 2011
    1. Doherty MK, Hammond DE, Clague MJ, Gaskell SJ, Beynon RJ. Turnover of the human proteome: determination of protein intracellular stability by dynamic SILAC. J Proteome Res. 2009;8:104–112.
    1. Klare N, Seeger M, Janek K, Jungblut PR, Dahlmann B. Intermediate-type 20 S proteasomes in HeLa cells: “asymmetric” subunit composition, diversity and adaptation. J Mol Biol. 2007;373:1–10.
    1. Busse A, Kraus M, Na IK, Rietz A, Scheibenbogen C, et al. Sensitivity of tumor cells to proteasome inhibitors is associated with expression levels and composition of proteasome subunits. Cancer. 2008;112:659–670.
    1. Kisselev AF, Garcia-Calvo M, Overkleeft HS, Peterson E, Pennington MW, et al. The caspase-like sites of proteasomes, their substrate specificity, new inhibitors and substrates, and allosteric interactions with the trypsin-like sites. J Biol Chem. 2003;278:35869–35877.
    1. Arendt CS, Hochstrasser M. Eukaryotic 20S proteasome catalytic subunit propeptides prevent active site inactivation by N-terminal acetylation and promote particle assembly. EMBO J. 1999;18:3575–3585.
    1. Jager S, Groll M, Huber R, Wolf DH, Heinemeyer W. Proteasome beta-type subunits: unequal roles of propeptides in core particle maturation and a hierarchy of active site function. J Mol Biol. 1999;291:997–1013.
    1. Wang L, Kumar S, Fridley BL, Kalari KR, Moon I, et al. Proteasome beta subunit pharmacogenomics: gene resequencing and functional genomics. Clin Cancer Res. 2008;14:3503–3513.
    1. Shringarpure R, Catley L, Bhole D, Burger R, Podar K, et al. Gene expression analysis of B-lymphoma cells resistant and sensitive to bortezomib. Br J Haematol. 2006;134:145–156.
    1. Chauhan D, Li G, Shringarpure R, Podar K, Ohtake Y, et al. Blockade of Hsp27 overcomes Bortezomib/proteasome inhibitor PS-341 resistance in lymphoma cells. Cancer Res. 2003;63:6174–6177.
    1. Siegel DS, Martin T, Wang M, Vij R, Jakubowiak AJ, et al. 2011. Results of PX-171-003-A1, An Open-Label, Single-Arm, Phase 2 (Ph 2) Study of Carfilzomib (CFZ) In Patients (pts) with Relapsed and Refractory Multiple Myeloma (MM). ASH 2010.
    1. Dasmahapatra G, Lembersky D, Kramer L, Fisher RI, Friedberg J, et al. The pan-HDAC inhibitor vorinostat potentiates the activity of the proteasome inhibitor carfilzomib in human DLBCL cells in vitro and in vivo. Blood. 2010;115:4478–4487.
    1. Kuhn DJ, Chen Q, Voorhees PM, Strader JS, Shenk KD, et al. Potent activity of carfilzomib, a novel, irreversible inhibitor of the ubiquitin-proteasome pathway, against preclinical models of multiple myeloma. Blood. 2007;110:3281–3290.
    1. Groll M, Kim KB, Kairies N, Huber R, Crews CM. Crystal Structure of Epoxomicin:20S Proteasome Reveals a Molecular Basis for Selectivity of a′ b′ - Epoxyketone Proteasome Inhibitors. J Am Chem Soc. 2000;122:1237–1238.

Source: PubMed

3
Abonnieren