Skip to main content
Log in

The histone deacetylase inhibitor valproic acid sensitizes human and canine osteosarcoma to doxorubicin

  • Original Article
  • Published:
Cancer Chemotherapy and Pharmacology Aims and scope Submit manuscript

Abstract

Purpose

Osteosarcoma (OS) remains an incurable and ultimately fatal disease in many patients, and novel forms of therapy are needed. Improved models of OS that more closely mimic human disease would provide more robust information regarding the utility of novel therapies. Spontaneous OS in dogs may provide such a model. Pharmacologic inhibition of histone deacetylase (HDAC) enzymes has a variety of anti-tumor effects but may demonstrate the most utility when utilized in combination with standard cytotoxic therapies. We sought to determine the in vitro and in vivo effects of the HDAC inhibitor valproic acid (VPA) on doxorubicin (DOX) sensitivity in canine and human OS.

Methods

We evaluated the in vitro anti-proliferative and apoptotic effects of VPA/DOX combination treatment, alterations in histone acetylation and nuclear DOX accumulation resulting from VPA treatment, and the in vivo efficacy of combination therapy in a xenograft model.

Results

Treatment of canine and human OS cell lines with clinically achievable VPA concentrations resulted in increased histone acetylation but modest anti-proliferative effects. Pre-incubation with VPA followed by doxorubicin (DOX) resulted in significant growth inhibition and potentiation of apoptosis, associated with a dose-dependent increase in nuclear DOX accumulation. The combination of VPA and DOX was superior to either monotherapy in a canine OS xenograft model.

Conclusion

These results demonstrate a rationale for the addition of HDAC inhibitors to current protocols for the treatment of OS and illustrate the similarities in response to HDAC inhibitors between human and canine OS, lending further credibility to the canine OS model.

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.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Baylin SB, Ohm JE (2006) Epigenetic gene silencing in cancer–a mechanism for early oncogenic pathway addiction? Nat Rev Cancer 6:107–116

    Article  CAS  PubMed  Google Scholar 

  2. Marks P, Rifkind RA, Richon VM, Breslow R, Miller T, Kelly WK (2001) Histone deacetylases and cancer: causes and therapies. Nat Rev Cancer 1:194–202

    Article  CAS  PubMed  Google Scholar 

  3. Mai A, Massa S, Rotili D, Cerbara I, Valente S, Pezzi R, Simeoni S, Ragno R (2005) Histone deacetylation in epigenetics: an attractive target for anticancer therapy. Med Res Rev 25:261–309

    Article  CAS  PubMed  Google Scholar 

  4. Ouaissi M, Sielezneff I, Silvestre R, Sastre B, Bernard JP, Lafontaine JS, Payan MJ, Dahan L, Pirro N, Seitz JF, Mas E, Lombardo D, Ouaissi A (2008) High histone deacetylase 7 (HDAC7) expression is significantly associated with adenocarcinomas of the pancreas. Ann Surg Oncol 15:2318–2328

    Article  PubMed  Google Scholar 

  5. Zhu P, Martin E, Mengwasser J, Schlag P, Janssen KP, Gottlicher M (2004) Induction of HDAC2 expression upon loss of APC in colorectal tumorigenesis. Cancer Cell 5:455–463

    Article  CAS  PubMed  Google Scholar 

  6. Ropero S, Ballestar E, Alaminos M, Arango D, Schwartz S Jr, Esteller M (2008) Transforming pathways unleashed by a HDAC2 mutation in human cancer. Oncogene 27:4008–4012

    Article  CAS  PubMed  Google Scholar 

  7. Weichert W, Roske A, Gekeler V, Beckers T, Stephan C, Jung K, Fritzsche FR, Niesporek S, Denkert C, Dietel M, Kristiansen G (2008) Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy. Br J Cancer 98:604–610

    Article  CAS  PubMed  Google Scholar 

  8. Weichert W, Roske A, Gekeler V, Beckers T, Ebert MP, Pross M, Dietel M, Denkert C, Rocken C (2008) Association of patterns of class I histone deacetylase expression with patient prognosis in gastric cancer: a retrospective analysis. Lancet Oncol 9:139–148

    Article  CAS  PubMed  Google Scholar 

  9. Carraway HE, Gore SD (2007) Addition of histone deacetylase inhibitors in combination therapy. J Clin Oncol 25:1955–1956

    Article  PubMed  Google Scholar 

  10. McIntyre J, Angels-Moral M, Bozzo J (2007) Combination therapy with valproic acid in cancer: initial clinical approach. Drugs Future 32(1):45

    Google Scholar 

  11. Marchion DC, Bicaku E, Daud AI, Sullivan DM, Munster PN (2005) In vivo synergy between topoisomerase II and histone deacetylase inhibitors: predictive correlates. Mol Cancer Ther 4:1993–2000

    Article  CAS  PubMed  Google Scholar 

  12. Kim MS, Blake M, Baek JH, Kohlhagen G, Pommier Y, Carrier F (2003) Inhibition of histone deacetylase increases cytotoxicity to anticancer drugs targeting DNA. Cancer Res 63:7291–7300

    CAS  PubMed  Google Scholar 

  13. Catalano MG, Fortunati N, Pugliese M, Poli R, Bosco O, Mastrocola R, Aragno M, Boccuzzi G (2006) Valproic acid, a histone deacetylase inhibitor, enhances sensitivity to doxorubicin in anaplastic thyroid cancer cells. J Endocrinol 191:465–472

    Article  CAS  PubMed  Google Scholar 

  14. Marchion DC, Bicaku E, Daud AI, Sullivan DM, Munster PN (2005) Valproic acid alters chromatin structure by regulation of chromatin modulation proteins. Cancer Res 65:3815–3822

    Article  CAS  PubMed  Google Scholar 

  15. Kostrouchova M, Kostrouch Z, Kostrouchova M (2007) Valproic acid, a molecular lead to multiple regulatory pathways. Folia Biol (Praha) 53:37–49

    CAS  Google Scholar 

  16. Munster P, Marchion D, Bicaku E, Schmitt M, Lee JH, DeConti R, Simon G, Fishman M, Minton S, Garrett C, Chiappori A, Lush R, Sullivan D, Daud A (2007) Phase I trial of histone deacetylase inhibition by valproic acid followed by the topoisomerase II inhibitor epirubicin in advanced solid tumors: a clinical and translational study. J Clin Oncol 25:1979–1985

    Article  CAS  PubMed  Google Scholar 

  17. Zhu WG, Otterson GA (2003) The interaction of histone deacetylase inhibitors and DNA methyltransferase inhibitors in the treatment of human cancer cells. Curr Med Chem Anticancer Agents 3:187–199

    Article  CAS  PubMed  Google Scholar 

  18. Yamanegi K, Yamane J, Hata M, Ohyama H, Yamada N, Kato-Kogoe N, Futani H, Nakasho K, Okamura H, Terada N (2009) Sodium valproate, a histone deacetylase inhibitor, decreases the secretion of soluble Fas by human osteosarcoma cells and increases their sensitivity to Fas-mediated cell death. J Cancer Res Clin Oncol 135:879–889

    Article  CAS  PubMed  Google Scholar 

  19. Watanabe K, Okamoto K, Yonehara S (2005) Sensitization of osteosarcoma cells to death receptor-mediated apoptosis by HDAC inhibitors through downregulation of cellular FLIP. Cell Death Differ 12:10–18

    Article  CAS  PubMed  Google Scholar 

  20. Imai T, Adachi S, Nishijo K, Ohgushi M, Okada M, Yasumi T, Watanabe K, Nishikomori R, Nakayama T, Yonehara S, Toguchida J, Nakahata T (2003) FR901228 induces tumor regression associated with induction of Fas ligand and activation of Fas signaling in human osteosarcoma cells. Oncogene 22:9231–9242

    Article  CAS  PubMed  Google Scholar 

  21. Roh MS, Kim CW, Park BS, Kim GC, Jeong JH, Kwon HC, Suh DJ, Cho KH, Yee SB, Yoo YH (2004) Mechanism of histone deacetylase inhibitor Trichostatin A induced apoptosis in human osteosarcoma cells. Apoptosis 9:583–589

    Article  CAS  PubMed  Google Scholar 

  22. Jaboin J, Wild J, Hamidi H, Khanna C, Kim CJ, Robey R, Bates SE, Thiele CJ (2002) MS-27–275, an inhibitor of histone deacetylase, has marked in vitro and in vivo antitumor activity against pediatric solid tumors. Cancer Res 62:6108–6115

    CAS  PubMed  Google Scholar 

  23. Hamada K, Tomita Y, Inoue A, Fujimoto T, Hashimoto N, Myoui A, Yoshikawa H, Hatazawa J (2009) Evaluation of chemotherapy response in osteosarcoma with FDG-PET. Ann Nucl Med 23:89–95

    Article  CAS  PubMed  Google Scholar 

  24. Felx M, Guyot M-C, Isler M, Turcotte RE, Doyon J, Khatib A-M, Leclerc S, Moreau A, Moldovan F (2006) Endothelin-1 (ET-1) promotes MMP-2 and MMP-9 induction involving the transcription factor NF-κB in human osteosarcoma. Clin Sci 110:645–654

    Article  CAS  PubMed  Google Scholar 

  25. Hansen K, Khanna C (2004) Spontaneous and genetically engineered animal models; use in preclinical cancer drug development. Eur J Cancer 40:858–880

    Article  CAS  PubMed  Google Scholar 

  26. Kimmelman J, Nalbantoglu J (2007) Faithful companions: a proposal for neurooncology trials in pet dogs. Cancer Res 67:4541–4544

    Article  CAS  PubMed  Google Scholar 

  27. Khanna C (2008) Novel targets with potential therapeutic applications in osteosarcoma. Curr Oncol Rep 10:330–358

    Article  Google Scholar 

  28. Federman N, Bernthal N, Eilber FC, Tap WD (2009) The multidisciplinary management of osteosarcoma. Curr Treat Options Oncol 10:82–93

    Article  PubMed  Google Scholar 

  29. Marchion DC, Bicaku E, Daud AI, Richon V, Sullivan DM, Munster PN (2004) Sequence-specific potentiation of topoisomerase II inhibitors by the histone deacetylase inhibitor suberoylanilide hydroxamic acid. J Cell Biochem 92:223–237

    Article  CAS  PubMed  Google Scholar 

  30. Berg J, Weinstein MJ, Springfield DS, Rand WM (1995) Results of surgery and doxorubicin chemotherapy in dogs with osteosarcoma. J Am Vet Med Assoc 206:1555–1560

    CAS  PubMed  Google Scholar 

  31. Mueller F, Fuchs B, Kaser-Hotz B (2007) Comparative biology of human and canine osteosarcoma. Anticancer Res 27:155–164

    CAS  PubMed  Google Scholar 

  32. Vail D, Thamm D (2004) Spontaneously occurring tumors in companion animals as models for drug development. In: Teicher B, Andrews P (eds) Cancer drug discovery and development guide: preclinical screening, clinical trials, and approval. Humana Press Inc, Totowa, pp 259–284

    Google Scholar 

  33. Khanna C, Wan X, Bose S, Cassaday R, Olomu O, Mendoza A, Yeung C, Gorlick R, Hewitt SM, Helman LJ (2004) The membrane-cytoskeleton linker ezrin is necessary for osteosarcoma metastasis. Nat Med 10:182–186

    Article  CAS  PubMed  Google Scholar 

  34. MacEwen EG, Kurzman ID, Rosenthal RC, Smith BW, Manley PA, Roush JK, Howard PE (1989) Therapy for osteosarcoma in dogs with intravenous injection of liposome-encapsulated muramyl tripeptide. J Natl Cancer Inst 81:935–938

    Article  CAS  PubMed  Google Scholar 

  35. Kleinerman ES, Gano JB, Johnston DA, Benjamin RS, Jaffe N (1995) Efficacy of liposomal muramyl tripeptide (CGP 19835A) in the treatment of relapsed osteosarcoma. Am J Clin Oncol 18:93–99

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

Morris Animal Foundation (CSU Fund #5354500), National Institutes of Health NCRR T32-RR-007072-06, American Cancer Society RSG-04-219-01.

Conflict of interest statement

None.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Luke A. Wittenburg.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wittenburg, L.A., Bisson, L., Rose, B.J. et al. The histone deacetylase inhibitor valproic acid sensitizes human and canine osteosarcoma to doxorubicin. Cancer Chemother Pharmacol 67, 83–92 (2011). https://doi.org/10.1007/s00280-010-1287-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00280-010-1287-z

Keywords

Navigation