Skip to main content

Advertisement

Log in

Caffeic acid phenethyl ester exerts apoptotic and oxidative stress on human multiple myeloma cells

  • PRECLINICAL STUDIES
  • Published:
Investigational New Drugs Aims and scope Submit manuscript

Summary

Caffeic acid phenethyl ester (CAPE) is a phenolic compound initially identified in bee glue. CAPE is reported to exhibit antitumor activity in many cancer models. However, the effect of CAPE on multiple myeloma (MM) is not well studied. We investigated the anti-myeloma effect of CAPE, and the data showed that CAPE inhibited the growth of human MM cells in a dose (1 ~ 30 μM) and time (24 ~72 h) dependent manner without altering the viability of normal human peripheral blood B cells. Stress and toxicity pathway analysis demonstrated that CAPE, in a dose- and time-related fashion, induced the expression of apoptotic and oxidative stress-response genes including growth arrest and DNA-damage inducible, alpha and gamma (GADD45A and GADD45G) and heme oxygenase-1. Apoptosis of MM cells by CAPE was further confirmed through flow cytometric analysis with up to 50% apoptotic cells induced by 50 μM CAPE within 24 h. Western blot analysis revealed the CAPE-induced activation of apoptosis executioner enzyme caspase-3, and corresponding cleavage of its downstream target poly(ADP-ribose)polymerase (PARP). The oxidative stress caused by CAPE cytotoxicity in MM cells was evaluated through measurement of reactive oxygen species (ROS) level, antioxidant intervention and glutathione depletion. The intracellular ROS level was not elevated by CAPE, but the pretreatment of antioxidant (N-acetyl cysteine) and glutathione synthesis inhibitor (buthionine sulfoximine) suggested that CAPE may cause oxidative stress by decrease of intracellular antioxidant level rather than over production of ROS. These data suggest that CAPE promotes apoptosis through oxidative stress in human multiple myeloma cells.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Palumbo A, Anderson K (2011) Multiple myeloma. N Engl J Med 364(11):1046–1060. https://doi.org/10.1056/NEJMra1011442

    Article  CAS  PubMed  Google Scholar 

  2. Fairfax KA, Kallies A, Nutt SL, Tarlinton DM (2008) Plasma cell development: from B-cell subsets to long-term survival niches. Semin Immunol 20(1):49–58. https://doi.org/10.1016/j.smim.2007.12.002

    Article  CAS  PubMed  Google Scholar 

  3. Bakkus MH, Heirman C, Van Riet I, Van Camp B, Thielemans K (1992) Evidence that multiple myeloma Ig heavy chain VDJ genes contain somatic mutations but show no intraclonal variation. Blood 80(9):2326–2335

    Article  CAS  PubMed  Google Scholar 

  4. Fonseca R, Barlogie B, Bataille R, Bastard C, Bergsagel PL, Chesi M, Davies FE, Drach J, Greipp PR, Kirsch IR, Kuehl WM, Hernandez JM, Minvielle S, Pilarski LM, Shaughnessy JD Jr, Stewart AK, Avet-Loiseau H (2004) Genetics and cytogenetics of multiple myeloma: a workshop report. Cancer Res 64(4):1546–1558

    Article  CAS  PubMed  Google Scholar 

  5. Chauhan D, Uchiyama H, Urashima M, Yamamoto K, Anderson KC (1995) Regulation of interleukin 6 in multiple myeloma and bone marrow stromal cells. Stem Cells 13(Suppl 2):35–39

    CAS  PubMed  Google Scholar 

  6. Podar K, Tai YT, Davies FE, Lentzsch S, Sattler M, Hideshima T, Lin BK, Gupta D, Shima Y, Chauhan D, Mitsiades C, Raje N, Richardson P, Anderson KC (2001) Vascular endothelial growth factor triggers signaling cascades mediating multiple myeloma cell growth and migration. Blood 98(2):428–435

    Article  CAS  PubMed  Google Scholar 

  7. Mitsiades CS, Mitsiades NS, Munshi NC, Richardson PG, Anderson KC (2006) The role of the bone microenvironment in the pathophysiology and therapeutic management of multiple myeloma: interplay of growth factors, their receptors and stromal interactions. Eur J Cancer 42(11):1564–1573. https://doi.org/10.1016/j.ejca.2005.12.025

    Article  CAS  PubMed  Google Scholar 

  8. Hideshima T, Mitsiades C, Tonon G, Richardson PG, Anderson KC (2007) Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer 7(8):585–598. https://doi.org/10.1038/nrc2189

    Article  CAS  PubMed  Google Scholar 

  9. Hazlehurst LA, Dalton WS (2001) Mechanisms associated with cell adhesion mediated drug resistance (CAM-DR) in hematopoietic malignancies. Cancer Metastasis Rev 20(1–2):43–50

    Article  CAS  PubMed  Google Scholar 

  10. Dalton WS (2003) The tumor microenvironment: focus on myeloma. Cancer Treat Rev 29(Suppl 1):11–19

    Article  CAS  PubMed  Google Scholar 

  11. Younes H, Leleu X, Hatjiharissi E, Moreau AS, Hideshima T, Richardson P, Anderson KC, Ghobrial IM (2007) Targeting the phosphatidylinositol 3-kinase pathway in multiple myeloma. Clin Cancer Res 13(13):3771–3775. https://doi.org/10.1158/1078-0432.CCR-06-2921

    Article  CAS  PubMed  Google Scholar 

  12. Ishikawa H, Tsuyama N, Abroun S, Liu S, Li FJ, Otsuyama K, Zheng X, Kawano MM (2003) Interleukin-6, CD45 and the src-kinases in myeloma cell proliferation. Leuk Lymphoma 44(9):1477–1481. https://doi.org/10.3109/10428190309178767

    Article  CAS  PubMed  Google Scholar 

  13. Li ZW, Chen H, Campbell RA, Bonavida B, Berenson JR (2008) NF-kappaB in the pathogenesis and treatment of multiple myeloma. Curr Opin Hematol 15(4):391–399. https://doi.org/10.1097/MOH.0b013e328302c7f4

    Article  CAS  PubMed  Google Scholar 

  14. Hideshima T, Bergsagel PL, Kuehl WM, Anderson KC (2004) Advances in biology of multiple myeloma: clinical applications. Blood 104(3):607–618. https://doi.org/10.1182/blood-2004-01-0037

    Article  CAS  PubMed  Google Scholar 

  15. Gentile M, Recchia AG, Mazzone C, Lucia E, Vigna E, Morabito F (2013) Perspectives in the treatment of multiple myeloma. Expert Opin Biol Ther 13(Suppl 1):S1–S22. https://doi.org/10.1517/14712598.2013.799132

    Article  CAS  PubMed  Google Scholar 

  16. Gullett NP, Ruhul Amin AR, Bayraktar S, Pezzuto JM, Shin DM, Khuri FR, Aggarwal BB, Surh YJ, Kucuk O (2010) Cancer prevention with natural compounds. Semin Oncol 37(3):258–281. https://doi.org/10.1053/j.seminoncol.2010.06.014

    Article  CAS  PubMed  Google Scholar 

  17. Patel S (2016) Emerging adjuvant therapy for Cancer: Propolis and its constituents. J Diet Suppl 13(3):245–268. https://doi.org/10.3109/19390211.2015.1008614

    Article  CAS  PubMed  Google Scholar 

  18. Son S, Lewis BA (2002) Free radical scavenging and antioxidative activity of caffeic acid amide and ester analogues: structure-activity relationship. J Agric Food Chem 50(3):468–472

    Article  CAS  PubMed  Google Scholar 

  19. Toyoda T, Tsukamoto T, Takasu S, Shi L, Hirano N, Ban H, Kumagai T, Tatematsu M (2009) Anti-inflammatory effects of caffeic acid phenethyl ester (CAPE), a nuclear factor-kappaB inhibitor, on helicobacter pylori-induced gastritis in Mongolian gerbils. Int J Cancer 125(8):1786–1795. https://doi.org/10.1002/ijc.24586

    Article  CAS  PubMed  Google Scholar 

  20. Tseng TH, Lee YJ (2006) Evaluation of natural and synthetic compounds from east Asiatic folk medicinal plants on the mediation of cancer. Anti Cancer Agents Med Chem 6(4):347–365

    Article  CAS  Google Scholar 

  21. Natarajan K, Singh S, Burke TR Jr, Grunberger D, Aggarwal BB (1996) Caffeic acid phenethyl ester is a potent and specific inhibitor of activation of nuclear transcription factor NF-kappa B. Proc Natl Acad Sci U S A 93(17):9090–9095

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB (2010) Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med 49(11):1603–1616. https://doi.org/10.1016/j.freeradbiomed.2010.09.006

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wu J, Omene C, Karkoszka J, Bosland M, Eckard J, Klein CB, Frenkel K (2011) Caffeic acid phenethyl ester (CAPE), derived from a honeybee product propolis, exhibits a diversity of anti-tumor effects in pre-clinical models of human breast cancer. Cancer Lett 308(1):43–53. https://doi.org/10.1016/j.canlet.2011.04.012

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lin HP, Jiang SS, Chuu CP (2012) Caffeic acid phenethyl ester causes p21 induction, Akt signaling reduction, and growth inhibition in PC-3 human prostate cancer cells. PLoS One 7(2):e31286. https://doi.org/10.1371/journal.pone.0031286

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Akyol S, Ozturk G, Ginis Z, Armutcu F, Yigitoglu MR, Akyol O (2013) In vivo and in vitro antineoplastic actions of caffeic acid phenethyl ester (CAPE): therapeutic perspectives. Nutr Cancer 65(4):515–526. https://doi.org/10.1080/01635581.2013.776693

    Article  CAS  PubMed  Google Scholar 

  26. Wang X, Stavchansky S, Zhao B, Bynum JA, Kerwin SM, Bowman PD (2008) Cytoprotection of human endothelial cells from menadione cytotoxicity by caffeic acid phenethyl ester: the role of heme oxygenase-1. Eur J Pharmacol 591(1–3):28–35. https://doi.org/10.1016/j.ejphar.2008.06.017

    Article  CAS  PubMed  Google Scholar 

  27. Zaman S, Wang R, Gandhi V (2014) Targeting the apoptosis pathway in hematologic malignancies. Leuk Lymphoma 55(9):1980–1992. https://doi.org/10.3109/10428194.2013.855307

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Walker RE, Lawson MA, Buckle CH, Snowden JA, Chantry AD (2014) Myeloma bone disease: pathogenesis, current treatments and future targets. Br Med Bull 111(1):117–138. https://doi.org/10.1093/bmb/ldu016

    Article  CAS  PubMed  Google Scholar 

  29. Bynum JA, Wang X, Stavchansky SA, Bowman PD (2017) Time course expression analysis of 1[2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]imidazole induction of Cytoprotection in human endothelial cells. Gene Regul Syst Bio 11: https://doi.org/10.1177/1177625017701106

  30. Nicholson JK, Connelly J, Lindon JC, Holmes E (2002) Metabonomics: a platform for studying drug toxicity and gene function. Nat Rev Drug Discov 1(2):153–161. https://doi.org/10.1038/nrd728

    Article  CAS  PubMed  Google Scholar 

  31. Orrenius S, Nicotera P, Zhivotovsky B (2011) Cell death mechanisms and their implications in toxicology. Toxicol Sci 119(1):3–19. https://doi.org/10.1093/toxsci/kfq268

    Article  CAS  PubMed  Google Scholar 

  32. Beauregard AP, Harquail J, Lassalle-Claux G, Belbraouet M, Jean-Francois J, Touaibia M, Robichaud GA (2015) CAPE analogs induce growth arrest and apoptosis in breast Cancer cells. Molecules 20(7):12576–12589. https://doi.org/10.3390/molecules200712576

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ozturk G, Ginis Z, Akyol S, Erden G, Gurel A, Akyol O (2012) The anticancer mechanism of caffeic acid phenethyl ester (CAPE): review of melanomas, lung and prostate cancers. Eur Rev Med Pharmacol Sci 16(15):2064–2068

    CAS  PubMed  Google Scholar 

  34. Nowsheen S, Yang ES (2012) The intersection between DNA damage response and cell death pathways. Exp Oncol 34(3):243–254

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Ozben T (2007) Oxidative stress and apoptosis: impact on cancer therapy. J Pharm Sci 96(9):2181–2196. https://doi.org/10.1002/jps.20874

    Article  CAS  PubMed  Google Scholar 

  36. Kirshner JR, He S, Balasubramanyam V, Kepros J, Yang CY, Zhang M, Du Z, Barsoum J, Bertin J (2008) Elesclomol induces cancer cell apoptosis through oxidative stress. Mol Cancer Ther 7(8):2319–2327. https://doi.org/10.1158/1535-7163.MCT-08-0298

    Article  CAS  PubMed  Google Scholar 

  37. Leon-Gonzalez AJ, Auger C, Schini-Kerth VB (2015) Pro-oxidant activity of polyphenols and its implication on cancer chemoprevention and chemotherapy. Biochem Pharmacol 98(3):371–380. https://doi.org/10.1016/j.bcp.2015.07.017

    Article  CAS  PubMed  Google Scholar 

  38. Rahal A, Kumar A, Singh V, Yadav B, Tiwari R, Chakraborty S, Dhama K (2014) Oxidative stress, prooxidants, and antioxidants: the interplay. Biomed Res Int 2014:761264–761219. https://doi.org/10.1155/2014/761264

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work was supported by the American Association of Colleges of Pharmacy (AACP) New Investigator Award and The Center for Chronic Disorders of Aging (CCDA) funding from Philadelphia College of Osteopathic Medicine to XW. It was partly supported by AHA (11SDG5710004) and NIAID (AI128254-01A1) to RS.

Author information

Authors and Affiliations

Authors

Contributions

Conceived and designed the experiments: XW. Performed the experiments: EM, HP, ML.YB, and MK. Analyzed the data: EM, HP, ML, RS, and XW. Critically reviewed the paper: RS. Wrote the paper: XW.

Corresponding author

Correspondence to Xinyu Wang.

Ethics declarations

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Ethical approval

This article does not contain any studies with human participants or animals performed by any of the authors.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Marin, E.H., Paek, H., Li, M. et al. Caffeic acid phenethyl ester exerts apoptotic and oxidative stress on human multiple myeloma cells. Invest New Drugs 37, 837–848 (2019). https://doi.org/10.1007/s10637-018-0701-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10637-018-0701-y

Keywords

Navigation