Population pharmacokinetics of letermovir following oral and intravenous administration in healthy participants and allogeneic hematopoietic cell transplantation recipients

Marita Prohn, Anders Viberg, Da Zhang, Kevin Dykstra, Casey Davis, Sreeraj Macha, Philip Sabato, Dinesh de Alwis, Marian Iwamoto, Craig Fancourt, Carolyn R Cho, Marita Prohn, Anders Viberg, Da Zhang, Kevin Dykstra, Casey Davis, Sreeraj Macha, Philip Sabato, Dinesh de Alwis, Marian Iwamoto, Craig Fancourt, Carolyn R Cho

Abstract

Letermovir is indicated for prophylaxis of cytomegalovirus infection and disease in allogeneic hematopoietic stem cell transplant (HSCT) recipients. Two-stage population pharmacokinetic (PK) modeling of letermovir was conducted to support dose rationale and evaluate the impact of intrinsic/extrinsic factors. Data from healthy phase I study participants over a wide dose range were modeled to evaluate the effects of selected intrinsic factors, including pharmacogenomics; next, phase III HSCT-recipient data at steady-state following clinical doses were modeled. The model in HSCT recipients adequately described letermovir PK following both oral or i.v. administration, and was consistent with the healthy participant model at steady-state clinical doses. Intrinsic factor effects were not clinically meaningful. These staged analyses indicate that letermovir PK in HSCT recipients and healthy participants differ only with respect to bioavailability and absorption rate. The HSCT recipient model was suitable for predicting exposure for exposure-response analysis supporting final dose selection.

Trial registration: ClinicalTrials.gov NCT02137772.

Conflict of interest statement

Marita Prohn is an employee of qPharmetra and a former employee of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA. Anders Viberg and Kevin Dykstra are former employees of qPharmetra. Da Zhang, Casey Davis, Sreeraj Macha, Philip Sabato, Dinesh de Alwis, Marian Iwamoto, Craig Fancourt, and Carlyn R. Cho are current or former employees of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, and may own stock and/or stock options in Merck & Co., Inc., Kenilworth, NJ, USA.

© 2021 Merck Sharp & Dohme Corp. CPT: Pharmacometrics & Systems Pharmacology published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Graphical representation of the (a) healthy participant (phase I model), and (b) HSCT recipient (phase III model) popPK models. Atr, drug amount in a transit compartment; CL, clearance; CLmax, maximal clearance rate; CLmbase, CLmax for a typical 65.8 kg subject; CLMwt, exponent describing the weight effect on CLmax; CP, letermovir plasma concentration; EAI, induction compartment amount; HSCT, hematopoietic stem cell transplant; IMAG, scalar of the induction effect; kin, production rate induction compartment; KMCL, Michaelis‐Menten constant for clearance; KMQ, Michaelis‐Menten constant for intercompartmental clearance; kout, elimination rate induction compartment; Ktr, transit rate constant; MTT, mean transit time; MTTdose, dose effect on MTT; NTR, number of transit compartments; popPK, population pharmacokinetics; Q1, intercompartment clearance to the first peripheral compartment; Q1max, maximal intercompartment clearance to the first peripheral compartment; Q2, intercompartment clearance to the second peripheral compartment; Q3, intercompartment clearance to the third peripheral compartment; TVMTT, MTT for a typical dose of 240 mg; V1, central volume of distribution; V2, first peripheral volume of distribution; V3, second peripheral volume of distribution; V4, third peripheral volume of distribution; V1–4base, V1–4 for a typical 65.8 kg subject; Vd, volume of distribution (sum of V1, V2, V3, and V4); Vdbase, Vd for a typical 65.8 kg subject; Vdjpn, Asian effect on Vdbase; Vdwt, weight effect on Vd; WT, wild type
Figure 2
Figure 2
Simulated letermovir exposure using the healthy participant (phase I) popPK model after single‐dose or multiple‐dose: (a) oral administration; (b) i.v. administration. Box and whisker plot: the dot is the sample median, the boxes define the interquartile range, whiskers extend to 1.5 times the interquartile range. AUC, area under the concentration‐time curve; Cmax, maximum concentration (multiple‐dose Cmax at steady‐state); dn, dose normalized; popPK, population pharmacokinetics
Figure 3
Figure 3
HSCT recipient (phase III) popPK model predictions of letermovir exposure in HSCT recipients: (a) histograms of individual predicted letermovir AUCss and Ctrough; (b) box‐whisker plots of simulated AUCss and Ctrough following different letermovir dosing regimens. AUCss, area under the concentration‐time curve from 0 to 24 h postdose at steady‐state; CSA, cyclosporine A; Ctrough, minimum concentration; HSCT, hematopoietic stem cell transplant; popPK, population pharmacokinetics

References

    1. Ljungman P, Hakki M, Boeckh M. Cytomegalovirus in hematopoietic stem cell transplant recipients. Infect Dis Clin North Am. 2010;24:319‐337.
    1. Jacobsen T, Sifontis N. Drug interactions and toxicities associated with the antiviral management of cytomegalovirus infection. Am J Health Syst Pharm. 2010;67:1417‐1425.
    1. Salzberger B, Bowden RA, Hackman RC, Davis C, Boeckh M. Neutropenia in allogeneic marrow transplant recipients receiving ganciclovir for prevention of cytomegalovirus disease: Risk factors and outcome. Blood. 1997;90:2502‐2508.
    1. Le Page AK, Jager MM, Iwasenko JM, Scott GM, Alain S, Rawlinson WD. Clinical aspects of cytomegalovirus antiviral resistance in solid organ transplant recipients. Clin Infect Dis. 2013;56:1018‐1029.
    1. Kotton CN, Kumar D, Caliendo AM, et al. The third international consensus guidelines on the management of cytomegalovirus in solid‐organ transplantation. Transplantation. 2018;102:900‐931.
    1. Goldner T, Hewlett G, Ettischer N, Ruebsamen‐Schaeff H, Zimmermann H, Lischka P. The novel anticytomegalovirus compound AIC246 (Letermovir) inhibits human cytomegalovirus replication through a specific antiviral mechanism that involves the viral terminase. J Virol. 2011;85:10884‐10893.
    1. Zimmermann H, Lischka P, Ruebsamen‐Schaeff H. Letermovir (AIC246) a novel drug under development for prevention and treatment of cytomegalovirus infections acting via a novel mechanism of action. Eur Infect Dis. 2011;5:112‐114.
    1. Chemaly RF, Ullmann AJ, Stoelben S, et al. Letermovir for cytomegalovirus prophylaxis in hematopoietic‐cell transplantation. N Engl J Med. 2014;370:1781‐1789.
    1. Merck & Co. Inc. Prevymis™ prescribing information. 2019. . Accessed February 12, 2020.
    1. Merck Sharp & Dohme Ltd., London, UK . Prevymis summary of product characteristics. 2017. . Accessed February 12, 2020.
    1. Marty FM, Ljungman P, Chemaly RF, et al. Letermovir prophylaxis for cytomegalovirus in hematopoietic‐cell transplantation. N Engl J Med. 2017;377:2433‐2444.
    1. Kropeit D, Scheuenpflug J, Erb‐Zohar K, et al. Pharmacokinetics and safety of letermovir, a novel anti‐human cytomegalovirus drug, in patients with renal impairment. Br J Clin Pharmacol. 2017;83:1944‐1953.
    1. Kropeit D, von Richter O, Stobernack HP, Rübsamen‐Schaeff H, Zimmermann H. Pharmacokinetics and safety of letermovir coadministered with cyclosporine A or tacrolimus in healthy subjects. Clin Pharmacol Drug Dev. 2018;7:9‐21.
    1. Stoelben S, Arns W, Renders L, et al. Preemptive treatment of cytomegalovirus infection in kidney transplant recipients with letermovir: Results of a phase 2a study. Transpl Int. 2014;27:77‐86.
    1. Erb‐Zohar K, Kropeit D, Scheuenpflug J, et al. Intravenous hydroxypropyl β‐cyclodextrin formulation of letermovir: A phase I, randomized, single‐ascending, and multiple‐dose trial. Clin Trans Sci. 2017;10:487‐495.
    1. McCrea JB, Macha S, Adedoyin A, et al. Pharmacokinetic drug‐drug interactions between letermovir and the immunosuppressants cyclosporine, tacrolimus, sirolimus, and mycophenolate mofetil. J Clin Pharmacol. 2019;59:1331‐1339.
    1. Bergstrand M, Hooker AC, Wallin JE, Karlsson MO. Prediction‐corrected visual predictive checks for diagnosing nonlinear mixed‐effects models. AAPS J. 2011;13:143‐151.
    1. Lindbom L, Pihlgren P, Jonsson EN. PsN‐Toolkit–a collection of computer intensive statistical methods for non‐linear mixed effect modeling using NONMEM. Comput Methods Programs Biomed. 2005;79:241‐257.
    1. Matis JH, Wehrly TE. Generalized stochastic compartmental models with Erlang transit times. J Pharmacokinet Biopharm. 1990;18:589‐607.
    1. Savic RM, Jonker DM, Kerbusch T, Karlsson MO. Implementation of a transit compartment model for describing drug absorption in pharmacokinetic studies. J Pharmacokinet Pharmacodyn. 2007;34:711‐726.
    1. Jacobson PA, Ng J, Green KG, Rogosheske J, Brundage R. Posttransplant day significantly influences pharmacokinetics of cyclosporine after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2003;9:304‐311.
    1. Zhang D, Chow DS. Clinical pharmacokinetics of mycophenolic acid in hematopoietic stem cell transplantation recipients. Eur J Drug Metab Pharmacokinet. 2017;42:183‐189.
    1. Vanstraelen K, Prattes J, Maertens J, et al. Posaconazole plasma exposure correlated to intestinal mucositis in allogeneic stem cell transplant patients. Eur J Clin Pharmacol. 2016;72:953‐963.
    1. Chen D, Hartmann G, Menzel K, Cho C. Development of a physiologically‐based pharmacokinetic model to understand the nonlinear pharmacokinetics of letermovir and exposure differences between populations. Drug Metab Pharmacokinet. 2018;33:S36.
    1. Fancourt C, Cho C, Viberg A, Prohn M, Dykstra K, Macha S. Comparison of two population pharmacokinetic models characterizing letermovir pharmacokinetics separately in healthy subjects and patients. Abstracts for the Ninth American Conference on Pharmacometrics (ACoP9). W‐091 ed; 2018.

Source: PubMed

3
Abonnere