Effects of Temsavir, Active Moiety of Antiretroviral Agent Fostemsavir, on QT Interval: Results From a Phase I Study and an Exposure-Response Analysis

Chakradhar Lagishetty, Katy Moore, Peter Ackerman, Cyril Llamoso, Mindy Magee, Chakradhar Lagishetty, Katy Moore, Peter Ackerman, Cyril Llamoso, Mindy Magee

Abstract

Fostemsavir, a prodrug of human immunodeficiency virus attachment inhibitor temsavir (TMR), is in phase III development in combination with other antiretroviral agents for the treatment of human immunodeficiency virus type I (HIV-1) infection in heavily treatment-experienced adults with multidrug-resistant HIV-1 infection for whom it is otherwise not possible to construct a suppressive antiviral regimen due to resistance, intolerance, or safety considerations. The proarrhythmic potential of fostemsavir was studied in a thorough QT study and exposure-response modeling was performed at therapeutic and supratherapeutic concentrations of TMR. Fostemsavir 1,200 mg b.i.d. did not result in a clinically meaningful change from placebo in baseline-adjusted Fridericia-corrected QTc (ddQTcF); however, at a supratherapeutic dose of 2,400 mg b.i.d., the upper bound of the two-sided 90% confidence interval (CI) of ddQTcF was 13.2 msec, exceeding the clinically important 10 msec threshold. A linear model of ddQTcF as a function of TMR plasma concentrations described these observations. Based on simulations with this model, TMR concentrations up to 7,500 ng/mL are expected to have an upper 90% CI bound for QTcF ≤ 10 msec. This concentration is 4.2-fold higher than the geometric mean TMR peak plasma concentration (Cmax ) of 1,770 ng/mL in heavily treatment-experienced HIV-1 infected patients administered fostemsavir 600 mg b.i.d. in the phase III BRIGHTE study (NCT02362503).

Conflict of interest statement

All authors are employees of ViiV Healthcare (K.M., P.A., and C.L.) or GlaxoSmithKline (C.L. and M.M.).

© 2020 ViiV Healthcare/GlaxoSmithKline. Clinical and Translational Science published by Wiley Periodicals, Inc. on behalf of the American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
The ddQTcF over time for fostemsavir 1,200 mg q.d., fostemsavir 2,400 mg b.i.d., and moxifloxacin. CI, confidence interval; ddQTcF, difference vs. placebo for change from baseline in Fridericia‐corrected QT interval; FTR, fostemsavir; MOX, moxifloxacin.
Figure 2
Figure 2
Scatterplot of ddQTcF vs. temsavir plasma concentrations for all measurements in subjects receiving fostemsavir 1,200 mg q.d. or 2,400 mg b.i.d. ddQTcF, difference vs. placebo for change from baseline in Fridericia‐corrected QT interval; TMR, temsavir.
Figure 3
Figure 3
Visual predictive check of final model. Circles are observed data; lines are median, 5th, and 95th percentiles of simulated data; shaded area is the 90% CI of simulated data. CI, confidence interval; ddQTcF, difference vs. placebo for change from baseline in Fridericia‐corrected QT interval; TMR, temsavir.
Figure 4
Figure 4
Model‐simulated effect on ddQTcF at increasing temsavir plasma concentrations. Solid line and shaded region represent model‐predicted mean ddQTcF and 90% CI as a function of plasma concentration. Horizontal line with tick marks shows range of concentrations divided into deciles. Circles and vertical bars denote observed means and 90% CIs for ddQTcF with each concentration decile. CI, confidence interval; ddQTcF, difference vs. placebo for change from baseline in Fridericia‐corrected QT interval; TMR, temsavir.

References

    1. Meanwell, N.A. et al Inhibitors of HIV‐1 attachment: the discovery and development of temsavir and its prodrug fostemsavir. J. Med. Chem. 61, 62–80 (2018).
    1. Langley, D.R. et al Homology models of the HIV‐1 attachment inhibitor BMS‐626529 bound to gp120 suggest a unique mechanism of action. Proteins 83, 331–350 (2015).
    1. Brown, J. et al Compartmental absorption modelling and site of absorption studies to determine feasibility of an extended‐release formulation of an HIV‐1 attachment inhibitor phosphate ester prodrug. J. Pharm. Sci. 102, 1742–1751 (2013).
    1. Gorycki, P. et al Pharmacokinetics, metabolism and excretion of radiolabelled fostemsavir administered with or without ritonavir in healthy male subjects. 19th International Workshop on Clinical Pharmacology of Antiviral Therapy; Baltimore, MD, USA; May 22–24, Abstract 42 (2018).
    1. Nowicka‐Sans, B. et al In vitro antiviral characteristics of HIV‐1 attachment inhibitor BMS‐626529, the active component of the prodrug BMS‐663068. Antimicrob. Agents Chemother. 56, 3498–3507 (2012).
    1. Li, Z. et al Activity of the HIV‐1 attachment inhibitor BMS‐626529, the active component of the prodrug BMS‐663068, against CD4‐independent viruses and HIV‐1 envelopes resistant to other entry inhibitors. Antimicrob. Agents Chemother. 57, 4172–4180 (2013).
    1. Zhou, N. et al Genotypic correlates of susceptibility to HIV‐1 attachment inhibitor BMS‐626529, the active agent of the prodrug BMS‐663068. J. Antimicrob. Chemother. 69, 573–581 (2014).
    1. Lalezari, J.P. et al Safety and efficacy of the HIV‐1 attachment inhibitor prodrug BMS‐663068 in treatment‐experienced individuals: 24 week results of AI438011, a phase 2b, randomised controlled trial. Lancet HIV 2, e427–e437 (2015).
    1. Landry, I. et al Model‐based phase 3 dose selection for HIV‐1 attachment inhibitor prodrug BMS‐663068 in HIV‐1‐infected patients: population pharmacokinetics/pharmacodynamics of the active moiety, BMS‐626529. Antimicrob. Agents Chemother. 60, 2782–2789 (2016).
    1. Nettles, R. et al Single and multiple dose pharmacokinetics and safety in non‐HIV‐infected healthy subjects dosed with BMS‐663068, an oral HIV attachment inhibitor. 12th International Workshop on Clinical Pharmacology of HIV Therapy; Miami, FL, USA; April 13–15, Abstract O_04 (2011).
    1. Janssen Pharmaceutical Companies . Edurant®; (rilpivirine) [package insert]. U.S. Food and Drug Administration (Drugs@FDA.com). U.S. Food and Drug Administration Website. <>. Accessed August 28, 2019.
    1. Abbvie Inc. KALETRA® (lopinavir and ritonavir) [package insert]. U.S. Food and Drug Administration (Drugs@FDA.com). U.S. Food and Drug Administration Website. <>. Accessed August 28, 2019.
    1. International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use. ICH Harmonised Tripartite Guideline E14: The Clinical Evaluation of QT/QTc Interval Prolongation and Proarrhythmic Potential for Non‐Antiarrhythmic Drugs. <> (May 2005). Accessed March 11, 2019.
    1. Williams, E.J. Experimental designs balanced for the estimation of residual effects of treatments. Aus. J. Sci. Res. 2, 149–168 (1949).
    1. Fridericia, L.S. Die systolendauer im elektrokardiogramm bei normalen menschen und bei herzkranken. Acta. Med. Scand. 53, 469–486 (1920).
    1. Bazett, H.C. An analysis of the time‐relations of the electrocardiograms. Heart 7, 353–370 (1920).
    1. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Statist. Soc. B 57, 289–300 (1995).
    1. Zhu, L. et al Pharmacokinetic interactions between BMS‐626529, the active moiety of the HIV‐1 attachment inhibitor prodrug BMS‐663068, and ritonavir or ritonavir‐boosted atazanavir in healthy subjects. Antimicrob. Agents Chemother. 59, 3816–3822 (2015).
    1. Hosmane, B. & Locke, C. A simulation study of power in thorough QT/QTc studies and a normal approximation for planning purposes. Ther. Innov. Regul. Sci. 39, 447–455 (2005).
    1. Natekar, M. et al Effect of number of replicate electrocardiograms recorded at each time point in a thorough QT study on sample size and study cost. J. Clin. Pharmacol. 51, 908–914 (2011).
    1. Vakkalagadda, B. et al HIV‐1 attachment inhibitor prodrug BMS‐663068: interactions with cobicistat and darunavir+cobicistat. ICAAC/ICC 2015; San Diego, CA, USA; September 17–21, Abstract 1676 (2015).
    1. Moore, K. et al Fostemsavir drug‐drug interaction profile, an attachment inhibitor and oral prodrug of temsavir, for heavily treatment‐experienced HIV‐1 infected patients. ID Week 2019; Washington, DC, USA; October 2–6, Abstract 2500.
    1. Lataillade, M. et al Week 96 safety and efficacy of the novel HIV‐1 attachment inhibitor prodrug fostemsavir in heavily treatment‐experienced participants infected with multi‐drug resistant HIV‐1 (BRIGHTE Study). International AIDS Conference on HIV Science (IAS) 2019; Mexico City, Mexico; July 21–24.
    1. Moore, K. et al Impact of mild, moderate, and severe renal impairment and hemodialysis on temsavir pharmacokinetics following oral administration of fostemsavir, an attachment inhibitor for heavily treatment experienced HIV‐1 infected patients. HIV Drug Therapy 2018; Glasgow, UK; October 28–31. Poster 261.
    1. Sevinsky, H. et al Pharmacokinetics of temsavir, the active moiety of the prodrug fostemsavir, in subjects with hepatic impairment. Open Forum Infect. Dis. 4 (suppl. 1), S430 (2017).

Source: PubMed

3
Prenumerera