Publication Cover
Xenobiotica
the fate of foreign compounds in biological systems
Volume 47, 2017 - Issue 4
604
Views
21
CrossRef citations to date
0
Altmetric
Topics in Xenobiochemistry

Clinical pharmacokinetics of panobinostat, a novel histone deacetylase (HDAC) inhibitor: review and perspectives

Pages 354-368 | Received 03 Apr 2016, Accepted 26 Apr 2016, Published online: 25 May 2016

References

  • Abbas A, Gupta S. (2008). The role of histone deacetylases in prostate cancer. Epigenetics 3:300–9.
  • Bali P, Pranpat M, Bradner J, et al. (2005). Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors. J Biol Chem 280:26729–34.
  • Clive S, Woo MM, Nydam T, et al. (2012). Characterizing the disposition, metabolism, and excretion of an orally active pan-deacetylase inhibitor, panobinostat, via trace radiolabeled 14C material in advanced cancer patients. Cancer Chemother Pharmacol 70:513–22.
  • Coiffier B, Pro B, Prince HM, et al. (2012). Results from a pivotal, open-label, phase II study of romidepsin in relapsed or refractory peripheral T-cell lymphoma after prior systemic therapy. J Clin Oncol 30:631–6.
  • DeAngelo DJ, Spencer A, Bhalla KN, et al. (2013). Phase Ia/II, two-arm, open-label, dose-escalation study of oral panobinostat administered via two dosing schedules in patients with advanced hematologic malignancies. Leukemia 27:1628–36.
  • Drummond DC, Noble CO, Kirpotin DB, et al. (2005). Clinical development of histone deacetylase inhibitors as anticancer agents. Annu Rev Pharmacol Toxicol 45:495–528.
  • Ellis L, Pan Y, Smyth GK, et al. (2008). Histone deacetylase inhibitor panobinostat induces clinical responses with associated alterations in gene expression profiles in cutaneous T-cell lymphoma. Clin Cancer Res 14:4500–10.
  • FARYDAK® (panobinostat) capsules, for oral use. Prescribing information, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey 07936, February 2015. Available from: http://www.pharma.us.novartis.com/product/pi/pdf/farydak.pdf [last accessed 6 Feb 2016].
  • Feld R, Woo MM, Leighl N, et al. (2013). A clinical investigation of inhibitory effect of panobinostat on CYP2D6 substrate in patients with advanced cancer. Cancer Chemother Pharmacol 72:747–55.
  • Fukutomi A, Hatake K, Matsui K, et al. (2012). A phase I study of oral panobinostat (LBH589) in Japanese patients with advanced solid tumors. Invest New Drugs 30:1096–106.
  • Giles F, Fischer T, Cortes J, et al. (2006). A phase I study of intravenous LBH589, a novel cinnamic hydroxamic acid analogue histone deacetylase inhibitor, in patients with refractory hematologic malignancies. Clin Cancer Res 12:4628–35.
  • Glaser KB. (2007). HDAC inhibitors: clinical update and mechanism-based potential. Biochem Pharmacol 74:659–71.
  • Glozak MA, Sengupta N, Zhang X, Seto E. (2005). Acetylation and deacetylation of non-histone proteins. Gene 363:15–23.
  • Hamberg P, Woo MM, Chen LC, et al. (2011). Effect of ketoconazole-mediated CYP3A4 inhibition on clinical pharmacokinetics of panobinostat (LBH589), an orally active histone deacetylase inhibitor. Cancer Chemother Pharmacol 68:805–13.
  • Hanigan CL, Van Engeland M, De Bruine AP, et al. (2008). An inactivating mutation in HDAC2 leads to dysregulation of apoptosis mediated by APAF1. Gastroenterology 135:e2.
  • Hubbert C, Guardiola A, Shao R, et al. (2002). HDAC6 is a microtubule-associated deacetylase. Nature 417:455–8.
  • Kotlinska-Lemieszek A, Paulsen O, Kaasa S, Klepstad P. (2014). Polypharmacy in patients with advanced cancer and pain: a European cross-sectional study of 2282 patients. J Pain Symptom Manage 48:1145–59.
  • Laubach JP, Moreau P, San-Miguel JF, Richardson PG. (2015). Panobinostat for the treatment of multiple myeloma. Clin Cancer Res 21:4767–73.
  • Marks P, Rifkind RA, Richon VM, et al. (2001). Histone deacetylases and cancer: causes and therapies. Nat Rev Cancer 1:194–202.
  • Marquard L, Gjerdrum LM, Christensen IJ, et al. (2008). Prognostic significance of the therapeutic targets histone deacetylase 1, 2, 6 and acetylated histone H4 in cutaneous T-cell lymphoma. Histopathology 53:267–77.
  • Morita S, Oizumi S, Minami H, et al. (2012). Phase I dose-escalating study of panobinostat (LBH589) administered intravenously to Japanese patients with advanced solid tumors. Invest New Drugs 30:1950–7.
  • Mottamal M, Zheng S, Huang TL, Wang G. (2015). Histone deacetylase inhibitors in clinical studies as templates for new anticancer agents. Molecules 20:3898–941.
  • Mu S, Kuroda Y, Shibayama H, et al. (2016). Panobinostat PK/PD profile in combination with bortezomib and dexamethasone in patients with relapsed and relapsed/refractory multiple myeloma. Eur J Clin Pharmacol 72:153–61.
  • Neri P, Bahlis NJ, Lonial S. (2012). Panobinostat for the treatment of multiple myeloma. Expert Opin Investig Drugs 21:733–47.
  • Oki Y, Copeland A, Younes A. (2011). Clinical development of panobinostat in classical Hodgkin’s lymphoma. Expert Rev Hematol 4:245–52.
  • Pan DS, Yang QJ, Fu X, et al. (2014). Discovery of an orally active subtype-selective HDAC inhibitor, chidamide, as an epigenetic modulator for cancer treatment. Med Chem Commun 5:1789–96.
  • Piekarz RL, Frye R, Turner M, et al. (2009). Phase ii multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma. J Clin Oncol 27:5410–7.
  • Poole RM. (2014). Belinostat: first global approval. Drugs 74:1543–54.
  • Portanova P, Russo T, Pellerito O, et al. (2008). The role of oxidative stress in apoptosis induced by the histone deacetylase inhibitor suberoylanilide hydroxamic acid in human colon adenocarcinoma HT-29 cells. Int J Oncol 33:325–31.
  • Prince HM, Bishton MJ, Johnstone RW. (2009). Panobinostat (LBH589): a potent pan-deacetylase inhibitor with promising activity against hematologic and solid tumors. Future Oncol 5:601–12.
  • Prystowsky M, Feeney K, Kawachi N, et al. (2013). Inhibition of Plk1 and Cyclin B1 expression results in panobinostat-induced G2 delay and mitotic defects. Sci Rep 3:2640.
  • Rasheed W, Bishton M, Johnstone RW, Prince HM. (2008). Histone deacetylase inhibitors in lymphoma and solid malignancies. Expert Rev Anticancer Ther 8:413–32.
  • Rathkopf D, Wong BY, Ross RW, et al. (2010). A phase I study of oral panobinostat alone and in combination with docetaxel in patients with castration-resistant prostate cancer. Cancer Chemother Pharmacol 66:181–9.
  • Richon VM, Garcia-Vargas J, Hardwick JS. (2009). Development of vorinostat: current applications and future perspectives for cancer therapy. Cancer Lett 280:201–10.
  • Saijo K, Imamura J, Narita K, et al. (2015). Biochemical, biological and structural properties of romidepsin (FK228) and its analogs as novel HDAC/PI3K dual inhibitors. Cancer Sci 106:208–15.
  • San-Miguel JF1, Richardson PG, Günther A, et al. (2013). Phase Ib study of panobinostat and bortezomib in relapsed or relapsed and refractory multiple myeloma. J Clin Oncol 31:3696–703.
  • Savelieva M, Woo MM, Schran H, et al. (2015). Population pharmacokinetics of intravenous and oral panobinostat in patients with hematologic and solid tumors. Eur J Clin Pharmacol 71:663–72.
  • Shapiro GI, Frank R, Dandamudi UB, et al. (2012). The effect of food on the bioavailability of panobinostat, an orally active pan-histone deacetylase inhibitor, in patients with advanced cancer. Cancer Chemother Pharmacol 69:555–62.
  • Sharma S, Beck J, Mita M, et al. (2013). A phase I dose-escalation study of intravenous panobinostat in patients with lymphoma and solid tumors. Invest New Drugs 31:974–85.
  • Sharma S, Witteveen PO, Lolkema MP, et al. (2015). A phase I, open-label, multicenter study to evaluate the pharmacokinetics and safety of oral panobinostat in patients with advanced solid tumors and varying degrees of renal function. Cancer Chemother Pharmacol 75:87–95.
  • Slingerland M, Hess D, Clive S, et al. (2014). A phase I, open-label, multicenter study to evaluate the pharmacokinetics and safety of oral panobinostat in patients with advanced solid tumors and various degrees of hepatic function. Cancer Chemother Pharmacol 74:1089–98.
  • Struhl K. (1998). Histone acetylation and transcriptional regulatory mechanisms. Genes Dev 12:599–606.
  • Suzuki J, Chen YY, Scott GK, et al. (2009). Protein acetylation and histone deacetylase expression associated with malignant breast cancer progression. Clin Cancer Res 15:3163–71.
  • Turner JP, Shakib S, Singhal N, et al. (2014). Prevalence and factors associated with polypharmacy in older people with cancer. Support Care Cancer 22:1727–34.
  • Whittaker SJ, Demierre MF, Kim EJ, et al. (2010). Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma. J Clin Oncol 28:4485–91.
  • Wilson AJ, Byun DS, Nasser S, et al. (2008). HDAC4 promotes growth of colon cancer cells via repression of p21. Mol Biol Cell 19:4062–75.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.