Skip to main content
Top
Published in: BMC Cancer 1/2015

Open Access 01-12-2015 | Research article

Identification of markers that functionally define a quiescent multiple myeloma cell sub-population surviving bortezomib treatment

Authors: Alfred Adomako, Veronica Calvo, Noa Biran, Keren Osman, Ajai Chari, James C Paton, Adrienne W Paton, Kateri Moore, Denis M Schewe, Julio A Aguirre-Ghiso

Published in: BMC Cancer | Issue 1/2015

Login to get access

Abstract

Background

The mechanisms allowing residual multiple myeloma (MM) cells to persist after bortezomib (Bz) treatment remain unclear. We hypothesized that studying the biology of bortezomib-surviving cells may reveal markers to identify these cells and survival signals to target and kill residual MM cells.

Methods

We used H2B-GFP label retention, biochemical tools and in vitro and in vivo experiments to characterize growth arrest and the unfolded protein responses in quiescent Bz-surviving cells. We also tested the effect of a demethylating agent, 5-Azacytidine, on Bz-induced quiescence and whether inhibiting the chaperone GRP78/BiP (henceforth GRP78) with a specific toxin induced apoptosis in Bz-surviving cells. Finally, we used MM patient samples to test whether GRP78 levels might associate with disease progression. Statistical analysis employed t-test and Mann-Whitney tests at a 95% confidence.

Results

We report that Bz-surviving MM cells in vitro and in vivo enter quiescence characterized by p21CIP1 upregulation. Bz-surviving MM cells also downregulated CDK6, Ki67 and P-Rb. H2B-GFP label retention showed that Bz-surviving MM cells are either slow-cycling or deeply quiescent. The Bz-induced quiescence was stabilized by low dose (500nM) of 5-azacytidine (Aza) pre-treatment, which also potentiated the initial Bz-induced apoptosis. We also found that expression of GRP78, an unfolded protein response (UPR) survival factor, persisted in MM quiescent cells. Importantly, GRP78 downregulation using a specific SubAB bacterial toxin killed Bz-surviving MM cells. Finally, quantification of Grp78high/CD138+ MM cells from patients suggested that high levels correlated with progressive disease.

Conclusions

We conclude that Bz-surviving MM cells display a GRP78HIGH/p21HIGH/CDK6LOW/P-RbLOW profile, and these markers may identify quiescent MM cells capable of fueling recurrences. We further conclude that Aza + Bz treatment of MM may represent a novel strategy to delay recurrences by enhancing Bz-induced apoptosis and quiescence stability.
Appendix
Available only for authorised users
Literature
1.
go back to reference Chauhan D, Singh A, Brahmandam M, Podar K, Hideshima T, Richardson P, et al. Combination of proteasome inhibitors bortezomib and NPI-0052 trigger in vivo synergistic cytotoxicity in multiple myeloma. Blood. 2008;111(3):1654–64.CrossRefPubMedPubMedCentral Chauhan D, Singh A, Brahmandam M, Podar K, Hideshima T, Richardson P, et al. Combination of proteasome inhibitors bortezomib and NPI-0052 trigger in vivo synergistic cytotoxicity in multiple myeloma. Blood. 2008;111(3):1654–64.CrossRefPubMedPubMedCentral
2.
go back to reference Laubach JP, Mahindra A, Mitsiades CS, Schlossman RL, Munshi NC, Ghobrial IM, et al. The use of novel agents in the treatment of relapsed and refractory multiple myeloma. Leukemia. 2009;23(12):2222–32.CrossRefPubMedPubMedCentral Laubach JP, Mahindra A, Mitsiades CS, Schlossman RL, Munshi NC, Ghobrial IM, et al. The use of novel agents in the treatment of relapsed and refractory multiple myeloma. Leukemia. 2009;23(12):2222–32.CrossRefPubMedPubMedCentral
3.
go back to reference Lee HC, Shah JJ, Orlowski RZ. Novel approaches to treatment of double-refractory multiple myeloma. Am Soc Clin Oncol Educ Book. 2013;2013:302–6.CrossRefPubMedCentral Lee HC, Shah JJ, Orlowski RZ. Novel approaches to treatment of double-refractory multiple myeloma. Am Soc Clin Oncol Educ Book. 2013;2013:302–6.CrossRefPubMedCentral
4.
go back to reference McMillin DW, Jacobs HM, Delmore JE, Buon L, Hunter ZR, Monrose V, et al. Molecular and cellular effects of NEDD8-activating enzyme inhibition in myeloma. Mol Cancer Ther. 2012;11(4):942–51.CrossRefPubMedPubMedCentral McMillin DW, Jacobs HM, Delmore JE, Buon L, Hunter ZR, Monrose V, et al. Molecular and cellular effects of NEDD8-activating enzyme inhibition in myeloma. Mol Cancer Ther. 2012;11(4):942–51.CrossRefPubMedPubMedCentral
5.
go back to reference Badros AZ. The role of maintenance therapy in the treatment of multiple myeloma. J Natl Compr Canc Netw. 2010;8 Suppl 1:S21–27.PubMed Badros AZ. The role of maintenance therapy in the treatment of multiple myeloma. J Natl Compr Canc Netw. 2010;8 Suppl 1:S21–27.PubMed
6.
go back to reference Schewe DM, Aguirre-Ghiso JA. Inhibition of eIF2alpha dephosphorylation maximizes bortezomib efficiency and eliminates quiescent multiple myeloma cells surviving proteasome inhibitor therapy. Cancer Res. 2009;69(4):1545–52.CrossRefPubMedPubMedCentral Schewe DM, Aguirre-Ghiso JA. Inhibition of eIF2alpha dephosphorylation maximizes bortezomib efficiency and eliminates quiescent multiple myeloma cells surviving proteasome inhibitor therapy. Cancer Res. 2009;69(4):1545–52.CrossRefPubMedPubMedCentral
7.
go back to reference Flanders A, Stetler-Stevenson M, Landgren O. Minimal residual disease testing in multiple myeloma by flow cytometry: major heterogeneity. Blood. 2013;122(6):1088–9.CrossRefPubMedPubMedCentral Flanders A, Stetler-Stevenson M, Landgren O. Minimal residual disease testing in multiple myeloma by flow cytometry: major heterogeneity. Blood. 2013;122(6):1088–9.CrossRefPubMedPubMedCentral
8.
go back to reference Rajkumar SV, Richardson PG, Hideshima T, Anderson KC. Proteasome inhibition as a novel therapeutic target in human cancer. J Clin Oncol. 2005;23(3):630–9.CrossRefPubMed Rajkumar SV, Richardson PG, Hideshima T, Anderson KC. Proteasome inhibition as a novel therapeutic target in human cancer. J Clin Oncol. 2005;23(3):630–9.CrossRefPubMed
9.
go back to reference Massey AJ, Williamson DS, Browne H, Murray JB, Dokurno P, Shaw T, et al. A novel, small molecule inhibitor of Hsc70/Hsp70 potentiates Hsp90 inhibitor induced apoptosis in HCT116 colon carcinoma cells. Cancer Chemother Pharmacol. 2010;66(3):535–45.CrossRefPubMed Massey AJ, Williamson DS, Browne H, Murray JB, Dokurno P, Shaw T, et al. A novel, small molecule inhibitor of Hsc70/Hsp70 potentiates Hsp90 inhibitor induced apoptosis in HCT116 colon carcinoma cells. Cancer Chemother Pharmacol. 2010;66(3):535–45.CrossRefPubMed
10.
go back to reference Aguirre Ghiso JA, Kovalski K, Ossowski L. Tumor dormancy induced by downregulation of urokinase receptor in human carcinoma involves integrin and MAPK signaling. J Cell Biol. 1999;147(1):89–104.CrossRefPubMed Aguirre Ghiso JA, Kovalski K, Ossowski L. Tumor dormancy induced by downregulation of urokinase receptor in human carcinoma involves integrin and MAPK signaling. J Cell Biol. 1999;147(1):89–104.CrossRefPubMed
12.
go back to reference Wilson A, Laurenti E, Oser G, van der Wath RC, Blanco-Bose W, Jaworski M, et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell. 2008;135(6):1118–29.CrossRefPubMed Wilson A, Laurenti E, Oser G, van der Wath RC, Blanco-Bose W, Jaworski M, et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell. 2008;135(6):1118–29.CrossRefPubMed
13.
go back to reference Sharma A, Heuck CJ, Fazzari MJ, Mehta J, Singhal S, Greally JM, et al. DNA methylation alterations in multiple myeloma as a model for epigenetic changes in cancer. Wiley Interdiscip Rev Syst Biol Med. 2010;2(6):654–69.CrossRefPubMed Sharma A, Heuck CJ, Fazzari MJ, Mehta J, Singhal S, Greally JM, et al. DNA methylation alterations in multiple myeloma as a model for epigenetic changes in cancer. Wiley Interdiscip Rev Syst Biol Med. 2010;2(6):654–69.CrossRefPubMed
14.
go back to reference Silverman LR, Fenaux P, Mufti GJ, Santini V, Hellstrom-Lindberg E, Gattermann N, et al. Continued azacitidine therapy beyond time of first response improves quality of response in patients with higher-risk myelodysplastic syndromes. Cancer. 2011;117(12):2697–702.CrossRefPubMedPubMedCentral Silverman LR, Fenaux P, Mufti GJ, Santini V, Hellstrom-Lindberg E, Gattermann N, et al. Continued azacitidine therapy beyond time of first response improves quality of response in patients with higher-risk myelodysplastic syndromes. Cancer. 2011;117(12):2697–702.CrossRefPubMedPubMedCentral
15.
go back to reference Kiziltepe T, Hideshima T, Catley L, Raje N, Yasui H, Shiraishi N, et al. 5-Azacytidine, a DNA methyltransferase inhibitor, induces ATR-mediated DNA double-strand break responses, apoptosis, and synergistic cytotoxicity with doxorubicin and bortezomib against multiple myeloma cells. Mol Cancer Ther. 2007;6(6):1718–27.CrossRefPubMed Kiziltepe T, Hideshima T, Catley L, Raje N, Yasui H, Shiraishi N, et al. 5-Azacytidine, a DNA methyltransferase inhibitor, induces ATR-mediated DNA double-strand break responses, apoptosis, and synergistic cytotoxicity with doxorubicin and bortezomib against multiple myeloma cells. Mol Cancer Ther. 2007;6(6):1718–27.CrossRefPubMed
16.
go back to reference Yang Q, Sarnow P. Location of the internal ribosome entry site in the 5′ non-coding region of the immunoglobulin heavy-chain binding protein (BiP) mRNA: evidence for specific RNA-protein interactions. Nucleic Acids Res. 1997;25(14):2800–7.CrossRefPubMedPubMedCentral Yang Q, Sarnow P. Location of the internal ribosome entry site in the 5′ non-coding region of the immunoglobulin heavy-chain binding protein (BiP) mRNA: evidence for specific RNA-protein interactions. Nucleic Acids Res. 1997;25(14):2800–7.CrossRefPubMedPubMedCentral
17.
go back to reference Ranganathan AC, Zhang L, Adam AP, Aguirre-Ghiso JA. Functional coupling of p38-induced up-regulation of BiP and activation of RNA-dependent protein kinase-like endoplasmic reticulum kinase to drug resistance of dormant carcinoma cells. Cancer Res. 2006;66(3):1702–11.CrossRefPubMedPubMedCentral Ranganathan AC, Zhang L, Adam AP, Aguirre-Ghiso JA. Functional coupling of p38-induced up-regulation of BiP and activation of RNA-dependent protein kinase-like endoplasmic reticulum kinase to drug resistance of dormant carcinoma cells. Cancer Res. 2006;66(3):1702–11.CrossRefPubMedPubMedCentral
18.
go back to reference Dong D, Dubeau L, Bading J, Nguyen K, Luna M, Yu H, et al. Spontaneous and controllable activation of suicide gene expression driven by the stress-inducible grp78 promoter resulting in eradication of sizable human tumors. Hum Gene Ther. 2004;15(6):553–61.CrossRefPubMed Dong D, Dubeau L, Bading J, Nguyen K, Luna M, Yu H, et al. Spontaneous and controllable activation of suicide gene expression driven by the stress-inducible grp78 promoter resulting in eradication of sizable human tumors. Hum Gene Ther. 2004;15(6):553–61.CrossRefPubMed
19.
go back to reference Eslick R, Talaulikar D. Multiple myeloma: from diagnosis to treatment. Aust Fam Physician. 2013;42(10):684–8.PubMed Eslick R, Talaulikar D. Multiple myeloma: from diagnosis to treatment. Aust Fam Physician. 2013;42(10):684–8.PubMed
20.
go back to reference Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, Irwin D, et al. A phase 2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med. 2003;348(26):2609–17.CrossRefPubMed Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, Irwin D, et al. A phase 2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med. 2003;348(26):2609–17.CrossRefPubMed
21.
go back to reference Bross PF, Kane R, Farrell AT, Abraham S, Benson K, Brower ME, et al. Approval summary for bortezomib for injection in the treatment of multiple myeloma. Clin Canc Res. 2004;10(12 Pt 1):3954–64.CrossRef Bross PF, Kane R, Farrell AT, Abraham S, Benson K, Brower ME, et al. Approval summary for bortezomib for injection in the treatment of multiple myeloma. Clin Canc Res. 2004;10(12 Pt 1):3954–64.CrossRef
22.
go back to reference Herndon TM, Deisseroth A, Kaminskas E, Kane RC, Koti KM, Rothmann MD, et al. U.s. Food and Drug Administration approval: carfilzomib for the treatment of multiple myeloma. Clin Canc Res. 2013;19(17):4559–63.CrossRef Herndon TM, Deisseroth A, Kaminskas E, Kane RC, Koti KM, Rothmann MD, et al. U.s. Food and Drug Administration approval: carfilzomib for the treatment of multiple myeloma. Clin Canc Res. 2013;19(17):4559–63.CrossRef
23.
go back to reference Aguirre-Ghiso JA, Estrada Y, Liu D, Ossowski L. ERK(MAPK) activity as a determinant of tumor growth and dormancy; regulation by p38(SAPK). Cancer Res. 2003;63(7):1684–95.PubMed Aguirre-Ghiso JA, Estrada Y, Liu D, Ossowski L. ERK(MAPK) activity as a determinant of tumor growth and dormancy; regulation by p38(SAPK). Cancer Res. 2003;63(7):1684–95.PubMed
24.
go back to reference Schewe DM, Aguirre-Ghiso JA. ATF6alpha-Rheb-mTOR signaling promotes survival of dormant tumor cells in vivo. Proc Natl Acad Sci U S A. 2008;105(30):10519–24.CrossRefPubMedPubMedCentral Schewe DM, Aguirre-Ghiso JA. ATF6alpha-Rheb-mTOR signaling promotes survival of dormant tumor cells in vivo. Proc Natl Acad Sci U S A. 2008;105(30):10519–24.CrossRefPubMedPubMedCentral
25.
26.
go back to reference Seidl S, Ackermann J, Kaufmann H, Keck A, Nosslinger T, Zielinski CC, et al. DNA-methylation analysis identifies the E-cadherin gene as a potential marker of disease progression in patients with monoclonal gammopathies. Cancer. 2004;100(12):2598–606.CrossRefPubMed Seidl S, Ackermann J, Kaufmann H, Keck A, Nosslinger T, Zielinski CC, et al. DNA-methylation analysis identifies the E-cadherin gene as a potential marker of disease progression in patients with monoclonal gammopathies. Cancer. 2004;100(12):2598–606.CrossRefPubMed
27.
go back to reference Chim CS, Fung TK, Liang R. Disruption of INK4/CDK/Rb cell cycle pathway by gene hypermethylation in multiple myeloma and MGUS. Leukemia. 2003;17(12):2533–5.CrossRefPubMed Chim CS, Fung TK, Liang R. Disruption of INK4/CDK/Rb cell cycle pathway by gene hypermethylation in multiple myeloma and MGUS. Leukemia. 2003;17(12):2533–5.CrossRefPubMed
28.
go back to reference Hendershot LM. The ER function BiP is a master regulator of ER function. Mount Sinai J Med New York. 2004;71(5):289–97. Hendershot LM. The ER function BiP is a master regulator of ER function. Mount Sinai J Med New York. 2004;71(5):289–97.
29.
go back to reference Zhuang L, Scolyer RA, Lee CS, McCarthy SW, Cooper WA, Zhang XD, et al. Expression of glucose-regulated stress protein GRP78 is related to progression of melanoma. Histopathology. 2009;54(4):462–70.CrossRefPubMed Zhuang L, Scolyer RA, Lee CS, McCarthy SW, Cooper WA, Zhang XD, et al. Expression of glucose-regulated stress protein GRP78 is related to progression of melanoma. Histopathology. 2009;54(4):462–70.CrossRefPubMed
30.
go back to reference Kuroda K, Horiguchi A, Asano T, Ito K, Asakuma J, Sato A, et al. Glucose-regulated protein 78 positivity as a predictor of poor survival in patients with renal cell carcinoma. Urol Int. 2011;87(4):450–6.CrossRefPubMed Kuroda K, Horiguchi A, Asano T, Ito K, Asakuma J, Sato A, et al. Glucose-regulated protein 78 positivity as a predictor of poor survival in patients with renal cell carcinoma. Urol Int. 2011;87(4):450–6.CrossRefPubMed
31.
go back to reference Thornton M, Aslam MA, Tweedle EM, Ang C, Campbell F, Jackson R, et al. The unfolded protein response regulator GRP78 is a novel predictive biomarker in colorectal cancer. Int J Canc. 2013;133(6):1408–18.CrossRef Thornton M, Aslam MA, Tweedle EM, Ang C, Campbell F, Jackson R, et al. The unfolded protein response regulator GRP78 is a novel predictive biomarker in colorectal cancer. Int J Canc. 2013;133(6):1408–18.CrossRef
Metadata
Title
Identification of markers that functionally define a quiescent multiple myeloma cell sub-population surviving bortezomib treatment
Authors
Alfred Adomako
Veronica Calvo
Noa Biran
Keren Osman
Ajai Chari
James C Paton
Adrienne W Paton
Kateri Moore
Denis M Schewe
Julio A Aguirre-Ghiso
Publication date
01-12-2015
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2015
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-015-1460-1

Other articles of this Issue 1/2015

BMC Cancer 1/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine