Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 3/2018

01-09-2018 | Short Communication

A population pharmacokinetic/toxicity model for the reduction of platelets during a 48-h continuous intravenous infusion of the histone deacetylase inhibitor belinostat

Authors: Cody J. Peer, Oliver M. Hall, Tristan M. Sissung, Richard Piekarz, Sanjeeve Balasubramaniam, Susan E. Bates, William D. Figg

Published in: Cancer Chemotherapy and Pharmacology | Issue 3/2018

Login to get access

Abstract

Purpose

Belinostat is a second-generation histone deacetylase inhibitor (HDI) predominantly metabolized by UGT1A1-mediated glucuronidation. Two common polymorphisms (UGT1A1*28 and UGT1A1*60) were previously associated with impaired drug clearance and thrombocytopenia risk, likely from increased drug exposure. This latter phenomenon has been observed with other HDIs such as abexinostat, panobinostat, romidepsin, and vorinostat. It was the intention of this brief report to expand a population pharmacokinetic (PPK) model to include a pharmacodynamic (PD) model describing the change in platelet levels in patients with cancer administered belinostat as a 48-h continuous intravenous infusion, along with cisplatin and etoposide.

Methods

The PPK/PD model developed here introduced an additional rate constant to a commonly used mechanistic myelosuppression model to better describe the maturation of megakaryocytes into platelets before degradation and a feedback mechanism. The model employed a proportional error model to describe the observed circulating platelet data.

Results

Several covariates were explored, including sex, body weight, UGT1A1 genotype status, liver, and kidney function, but none significantly improved the model. Platelet levels rebounded to baseline within 21 days, before the next cycle of therapy. Simulations predicted that higher belinostat drug exposure does cause lower thrombocyte nadirs compared to lower belinostat levels. However, platelet levels rebound by the start of the next belinostat cycle.

Conclusions

This model suggests a q3week schedule allows for sufficient platelet recovery before the next belinostat infusion is optimal.
Appendix
Available only for authorised users
Literature
1.
go back to reference Thomas A, Rajan A, Szabo E et al (2014) A phase I/II trial of belinostat in combination with cisplatin, doxorubicin, and cyclophosphamide in thymic epithelial tumors: a clinical and translational study. Clin Cancer Res 20:5392–5402CrossRefPubMedPubMedCentral Thomas A, Rajan A, Szabo E et al (2014) A phase I/II trial of belinostat in combination with cisplatin, doxorubicin, and cyclophosphamide in thymic epithelial tumors: a clinical and translational study. Clin Cancer Res 20:5392–5402CrossRefPubMedPubMedCentral
2.
go back to reference Luchenko VL, Salcido CD, Zhang Y et al (2011) Schedule-dependent synergy of histone deacetylase inhibitors with DNA damaging agents in small cell lung cancer. Cell Cycle 10:3119–3128CrossRefPubMedPubMedCentral Luchenko VL, Salcido CD, Zhang Y et al (2011) Schedule-dependent synergy of histone deacetylase inhibitors with DNA damaging agents in small cell lung cancer. Cell Cycle 10:3119–3128CrossRefPubMedPubMedCentral
3.
go back to reference Kim MS, Blake M, Baek JH et al (2003) Inhibition of histone deacetylase increases cytotoxicity to anticancer drugs targeting DNA. Cancer Res 63:7291–7300PubMed Kim MS, Blake M, Baek JH et al (2003) Inhibition of histone deacetylase increases cytotoxicity to anticancer drugs targeting DNA. Cancer Res 63:7291–7300PubMed
4.
go back to reference Balasubramaniam S, Redon CE, Peer CJ et al (2018) Phase I trial of belinostat with cisplatin and etoposide in advanced solid tumors, with a focus on neuroendocrine and small cell cancers of the lung. Anticancer Drugs 29:457–465CrossRefPubMed Balasubramaniam S, Redon CE, Peer CJ et al (2018) Phase I trial of belinostat with cisplatin and etoposide in advanced solid tumors, with a focus on neuroendocrine and small cell cancers of the lung. Anticancer Drugs 29:457–465CrossRefPubMed
5.
go back to reference Peer CJ, Goey AK, Sissung TM et al (2015) UGT1A1 genotype-dependent dose adjustment of belinostat in patients with advanced cancers using population pharmacokinetic modeling and simulation. J Clin Pharmacol 56:450CrossRefPubMed Peer CJ, Goey AK, Sissung TM et al (2015) UGT1A1 genotype-dependent dose adjustment of belinostat in patients with advanced cancers using population pharmacokinetic modeling and simulation. J Clin Pharmacol 56:450CrossRefPubMed
6.
go back to reference Goey AK, Sissung TM, Peer CJ et al. (2015) Effects of UGT1A1 genotype on the pharmacokinetics, pharmacodynamics and toxicities of belinostat administered by 48 h continuous infusion in patients with cancer. J Clin Pharmacol 56:461CrossRefPubMed Goey AK, Sissung TM, Peer CJ et al. (2015) Effects of UGT1A1 genotype on the pharmacokinetics, pharmacodynamics and toxicities of belinostat administered by 48 h continuous infusion in patients with cancer. J Clin Pharmacol 56:461CrossRefPubMed
7.
go back to reference Chalret du Rieu Q, Fouliard S, Jacquet-Bescond A et al (2013) Application of hematological toxicity modeling in clinical development of abexinostat (S-78454, PCI-24781), a new histone deacetylase inhibitor. Pharm Res 30:2640–2653CrossRefPubMed Chalret du Rieu Q, Fouliard S, Jacquet-Bescond A et al (2013) Application of hematological toxicity modeling in clinical development of abexinostat (S-78454, PCI-24781), a new histone deacetylase inhibitor. Pharm Res 30:2640–2653CrossRefPubMed
8.
go back to reference Fouliard S, Robert R, Jacquet-Bescond A et al (2013) Pharmacokinetic/pharmacodynamic modelling-based optimisation of administration schedule for the histone deacetylase inhibitor abexinostat (S78454/PCI-24781) in phase I. Eur J Cancer 49:2791–2797CrossRefPubMed Fouliard S, Robert R, Jacquet-Bescond A et al (2013) Pharmacokinetic/pharmacodynamic modelling-based optimisation of administration schedule for the histone deacetylase inhibitor abexinostat (S78454/PCI-24781) in phase I. Eur J Cancer 49:2791–2797CrossRefPubMed
9.
go back to reference Younes A, Sureda A, Ben-Yehuda D et al (2012) Panobinostat in patients with relapsed/refractory Hodgkin’s lymphoma after autologous stem-cell transplantation: results of a phase II study. J Clin Oncol 30:2197–2203CrossRefPubMed Younes A, Sureda A, Ben-Yehuda D et al (2012) Panobinostat in patients with relapsed/refractory Hodgkin’s lymphoma after autologous stem-cell transplantation: results of a phase II study. J Clin Oncol 30:2197–2203CrossRefPubMed
10.
go back to reference 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 27:5410–5417CrossRefPubMedPubMedCentral 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 27:5410–5417CrossRefPubMedPubMedCentral
11.
go back to reference Grant C, Rahman F, Piekarz R et al (2010) Romidepsin: a new therapy for cutaneous T-cell lymphoma and a potential therapy for solid tumors. Expert Rev Anticancer Ther 10:997–1008CrossRefPubMed Grant C, Rahman F, Piekarz R et al (2010) Romidepsin: a new therapy for cutaneous T-cell lymphoma and a potential therapy for solid tumors. Expert Rev Anticancer Ther 10:997–1008CrossRefPubMed
13.
go back to reference Duvic M, Talpur R, Ni X et al (2007) Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T-cell lymphoma (CTCL). Blood 109:31–39CrossRefPubMedPubMedCentral Duvic M, Talpur R, Ni X et al (2007) Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T-cell lymphoma (CTCL). Blood 109:31–39CrossRefPubMedPubMedCentral
14.
go back to reference Bishton MJ, Harrison SJ, Martin BP et al (2011) Deciphering the molecular and biologic processes that mediate histone deacetylase inhibitor-induced thrombocytopenia. Blood 117:3658–3668CrossRefPubMed Bishton MJ, Harrison SJ, Martin BP et al (2011) Deciphering the molecular and biologic processes that mediate histone deacetylase inhibitor-induced thrombocytopenia. Blood 117:3658–3668CrossRefPubMed
15.
go back to reference Sasaki T, Takane H, Ogawa K et al (2011) Population pharmacokinetic and pharmacodynamic analysis of linezolid and a hematologic side effect, thrombocytopenia, in Japanese patients. Antimicrob Agents Chemother 55:1867–1873CrossRefPubMedPubMedCentral Sasaki T, Takane H, Ogawa K et al (2011) Population pharmacokinetic and pharmacodynamic analysis of linezolid and a hematologic side effect, thrombocytopenia, in Japanese patients. Antimicrob Agents Chemother 55:1867–1873CrossRefPubMedPubMedCentral
16.
go back to reference Mikhailidis DP, Ganotakis ES (1996) Plasma albumin and platelet function: relevance to atherogenesis and thrombosis. Platelets 7:125–137CrossRefPubMed Mikhailidis DP, Ganotakis ES (1996) Plasma albumin and platelet function: relevance to atherogenesis and thrombosis. Platelets 7:125–137CrossRefPubMed
17.
go back to reference Bautista MJ, Ruiz-Villamor E, Salguero FJ et al (2002) Early platelet aggregation as a cause of thrombocytopenia in classical swine fever. Vet Pathol 39:84–91CrossRefPubMed Bautista MJ, Ruiz-Villamor E, Salguero FJ et al (2002) Early platelet aggregation as a cause of thrombocytopenia in classical swine fever. Vet Pathol 39:84–91CrossRefPubMed
Metadata
Title
A population pharmacokinetic/toxicity model for the reduction of platelets during a 48-h continuous intravenous infusion of the histone deacetylase inhibitor belinostat
Authors
Cody J. Peer
Oliver M. Hall
Tristan M. Sissung
Richard Piekarz
Sanjeeve Balasubramaniam
Susan E. Bates
William D. Figg
Publication date
01-09-2018
Publisher
Springer Berlin Heidelberg
Published in
Cancer Chemotherapy and Pharmacology / Issue 3/2018
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-018-3631-7

Other articles of this Issue 3/2018

Cancer Chemotherapy and Pharmacology 3/2018 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