Evaluation of the effect of UGT1A1 polymorphisms on the pharmacokinetics of oral and long-acting injectable cabotegravir

Parul Patel, Zhengyu Xue, Karen S King, Laura Parham, Susan Ford, Yu Lou, Kalpana K Bakshi, Kenneth Sutton, David Margolis, Arlene R Hughes, William R Spreen, Parul Patel, Zhengyu Xue, Karen S King, Laura Parham, Susan Ford, Yu Lou, Kalpana K Bakshi, Kenneth Sutton, David Margolis, Arlene R Hughes, William R Spreen

Abstract

Background: Cabotegravir is an HIV integrase inhibitor in clinical development with both oral and long-acting (LA) injectable formulations. Cabotegravir is primarily metabolized by uridine 5'-diphospho-glucuronosyltransferase (UGT) 1A1, a known polymorphic enzyme with functional variants that can affect drug metabolism and exposure.

Objectives: To investigate the pharmacogenetic effects of the reduced-function alleles UGT1A1*6, UGT1A1*28 and/or UGT1A1*37 on steady-state pharmacokinetics (PK) and safety of oral cabotegravir (30 mg/day) and intramuscular cabotegravir LA (400 mg every 4 weeks or 600 mg every 8 weeks).

Methods: Plasma cabotegravir PK was assessed in 346 UGT-genotyped participants with and without UGT1A1 functional variants across six studies (four Phase I and two Phase II) of oral cabotegravir, including 215 HIV-infected participants who received oral cabotegravir followed by cabotegravir LA. Changes from baseline in total bilirubin and ALT were assessed in one study (LATTE; NCT01641809).

Results: Statistically significant (P < 0.05) associations were observed between UGT1A1 genotype and plasma cabotegravir PK parameters, with 28%-50% increases following oral cabotegravir [plasma cabotegravir concentration at the end of the dosing interval (Ctau), 1.50-fold; AUCtau, 1.41-fold; and Cmax, 1.28-fold] and 16%-24% increases following cabotegravir LA administration (48 week Ctau, 1.24-fold; AUCtau, 1.16-fold; and Cmax, 1.18-fold) among those with low-versus-normal genetically predicted UGT1A1 activity. A statistically significant (P < 10-5) association between predicted UGT1A1 activity and maximum change in total bilirubin was also observed (2.45-fold asymptomatic increase for low versus normal) without a corresponding change in ALT.

Conclusions: This modest increase in oral and parenteral cabotegravir exposure associated with a reduced function of UGT1A1 is not considered clinically relevant based on accumulated safety data; no dose adjustment is required.

© The Author(s) 2020. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy.

Figures

Figure 1.
Figure 1.
Box and whiskers plot showing the effects of predicted UGT1A1 activity on (a) steady-state cabotegravir trough concentrations in pooled data from subjects receiving oral cabotegravir 30 mg/day; and (b) maximum on-treatment Tbili from subjects receiving oral cabotegravir 10, 30 or 60 mg/day in the LATTE study (NCT01641809). Boxes represent the range between the 25th and 75th percentiles, the horizontal lines within the boxes show medians and whiskers show the 5th and 95th percentiles. Panel (a) pale closed circles, UGT1A1*1 homozygotes; dark closed circles, UGT1A1*36 carriers; plus symbols, UGT1A1*6 carriers; asterisks, UGT1A1*37 carriers.
Figure 2.
Figure 2.
Mean (±95% CI) plasma cabotegravir concentrations by visit in the LATTE-2 study among subjects receiving parenteral cabotegravir LA plus rilpivirine LA maintenance (a) every 4 weeks (400 mg cabotegravir) or (b) every 8 weeks (600 mg cabotegravir) after suppressive treatment on daily oral cabotegravir and nucleoside analogues, according to normal, reduced or low predicted UGT1A1 activity. CAB, cabotegravir.
Figure 3.
Figure 3.
Trough cabotegravir concentrations for oral versus IM administration according to predicted UGT1A1 activity in subjects from LATTE-2 (NCT02120352) for (a) IM administration every 4 weeks and (b) IM administration every 8 weeks. L, low predicted UGT1A1 activity; N, normal predicted UGT1A1 activity; R, reduced predicted UGT1A1 activity. On Day 1, oral cabotegravir trough was assessed 20–28 h after the last oral cabotegravir 30 mg dose taken at home. Boxes represent the range between the 25th and 75th percentiles, the horizontal lines within the boxes show medians and whiskers show the 5th and 95th percentiles.

References

    1. Bari H. A prolonged release parenteral drug delivery system: an overview. Int J Pharm Sci Rev Res 2010; 3: 1–11.
    1. Trezza C, Ford SL, Spreen W. et al. Formulation and pharmacology of long-acting cabotegravir. Curr Opin HIV AIDS 2015; 10: 239–45.
    1. Markowitz M, Frank I, Grant RM. et al. Safety and tolerability of long-acting cabotegravir injections in HIV-uninfected men (ECLAIR): a multicentre, double-blind, randomised, placebo-controlled, phase 2a trial. Lancet HIV 2017; 4: e331–40.
    1. Bowers GD, Culp A, Reese MJ. et al. Disposition and metabolism of cabotegravir: a comparison of biotransformation and excretion between different species and routes of administration in humans. Xenobiotica 2016; 46: 147–62.
    1. Reese MJ, Bowers GD, Humphreys JE. et al. Drug interaction profile of the HIV integrase inhibitor cabotegravir: assessment from in vitro studies and a clinical investigation with midazolam. Xenobiotica 2016; 46: 445–56.
    1. Court MH, Zhang X, Ding X. et al. Quantitative distribution of mRNAs encoding the 19 human UDP-glucuronosyltransferase enzymes in 26 adult and 3 fetal tissues. Xenobiotica 2012; 42: 266–77.
    1. Barbarino JM, Haidar CE, Klein TE. et al. PharmGKB summary: very important pharmacogene information for UGT1A1. Pharmacogenet Genomics 2014; 24: 177–83.
    1. Fujita K, Sparreboom A.. Pharmacogenetics of irinotecan disposition and toxicity: a review. Curr Clin Pharmacol 2010; 5: 209–17.
    1. Stingl JC, Bartels H, Viviani R. et al. Relevance of UDP-glucuronosyltransferase polymorphisms for drug dosing: a quantitative systematic review. Pharmacol Ther 2014; 141: 92–116.
    1. Strassburg CP. Pharmacogenetics of Gilbert’s syndrome. Pharmacogenomics 2008; 9: 703–15.
    1. Sneitz N, Bakker CT, de Knegt RJ. et al. Crigler-Najjar syndrome in The Netherlands: identification of four novel UGT1A1 alleles, genotype-phenotype correlation, and functional analysis of 10 missense mutants. Hum Mutat 2010; 31: 52–9.
    1. Seppen J, Bosma PJ, Goldhoorn BG. et al. Discrimination between Crigler-Najjar type I and II by expression of mutant bilirubin uridine diphosphate-glucuronosyltransferase. J Clin Invest 1994; 94: 2385–91.
    1. Kadakol A, Ghosh SS, Sappal BS. et al. Genetic lesions of bilirubin uridine-diphosphoglucuronate glucuronosyltransferase (UGT1A1) causing Crigler-Najjar and Gilbert syndromes: correlation of genotype to phenotype. Hum Mutat 2000; 16: 297–306.
    1. Bosma PJ, Chowdhury JR, Bakker C. et al. The genetic basis of the reduced expression of bilirubin UDP-glucuronosyltransferase 1 in Gilbert’s syndrome. N Engl J Med 1995; 333: 1171–5.
    1. Yamamoto K, Sato H, Fujiyama Y. et al. Contribution of two missense mutations (G71R and Y486D) of the bilirubin UDP glycosyltransferase (UGT1A1) gene to phenotypes of Gilbert’s syndrome and Crigler-Najjar syndrome type II. Biochim Biophys Acta 1998; 1406: 267–73.
    1. Boyd MA, Srasuebkul P, Ruxrungtham K. et al. Relationship between hyperbilirubinaemia and UDP-glucuronosyltransferase 1A1 (UGT1A1) polymorphism in adult HIV-infected Thai patients treated with indinavir. Pharmacogenet Genomics 2006; 16: 321–9.
    1. Akaba K, Kimura T, Sasaki A. et al. Neonatal hyperbilirubinemia and mutation of the bilirubin uridine diphosphate-glucuronosyltransferase gene: a common missense mutation among Japanese, Koreans and Chinese. Biochem Mol Biol Int 1998; 46: 21–6.
    1. Beutler E, Gelbart T, Demina A.. Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism? Proc Natl Acad Sci USA 1998; 95: 8170–4.
    1. Hall D, Ybazeta G, Destro-Bisol G. et al. Variability at the uridine diphosphate glucuronosyltransferase 1A1 promoter in human populations and primates. Pharmacogenetics 1999; 9: 591–9.
    1. Haverfield EV, McKenzie CA, Forrester T. et al. UGT1A1 variation and gallstone formation in sickle cell disease. Blood 2005; 105: 968–72.
    1. Hu ZY, Yu Q, Pei Q. et al. Dose-dependent association between UGT1A1*28 genotype and irinotecan-induced neutropenia: low doses also increase risk. Clin Cancer Res 2010; 16: 3832–42.
    1. Onoue M, Terada T, Kobayashi M. et al. UGT1A1*6 polymorphism is most predictive of severe neutropenia induced by irinotecan in Japanese cancer patients. Int J Clin Oncol 2009; 14: 136–42.
    1. Kishi S, Yang W, Boureau B. et al. Effects of prednisone and genetic polymorphisms on etoposide disposition in children with acute lymphoblastic leukemia. Blood 2004; 103: 67–72.
    1. Trontelj J, Marc J, Zavratnik A. et al. Effects of UGT1A1*28 polymorphism on raloxifene pharmacokinetics and pharmacodynamics. Br J Clin Pharmacol 2009; 67: 437–44.
    1. Lankisch TO, Moebius U, Wehmeier M. et al. Gilbert’s disease and atazanavir: from phenotype to UDP-glucuronosyltransferase haplotype. Hepatology 2006; 44: 1324–32.
    1. Motzer RJ, Johnson T, Choueiri TK. et al. Hyperbilirubinemia in pazopanib- or sunitinib-treated patients in COMPARZ is associated with UGT1A1 polymorphisms. Ann Oncol 2013; 24: 2927–8.
    1. Danoff TM, Campbell DA, McCarthy LC. et al. A Gilbert’s syndrome UGT1A1 variant confers susceptibility to tranilast-induced hyperbilirubinemia. Pharmacogenomics J 2004; 4: 49–53.
    1. Xu CF, Reck BH, Xue Z. et al. Pazopanib-induced hyperbilirubinemia is associated with Gilbert’s syndrome UGT1A1 polymorphism. Br J Cancer 2010; 102: 1371–7.
    1. Yamanaka H, Nakajima M, Katoh M. et al. A novel polymorphism in the promoter region of human UGT1A9 gene (UGT1A9*22) and its effects on the transcriptional activity. Pharmacogenetics 2004; 14: 329–32.
    1. Cui C, Shu C, Cao D. et al. UGT1A1*6, UGT1A7*3 and UGT1A9*1b polymorphisms are predictive markers for severe toxicity in patients with metastatic gastrointestinal cancer treated with irinotecan-based regimens. Oncol Lett 2016; 12: 4231–7.
    1. Linakis MW, Cook SF, Kumar SS. et al. Polymorphic expression of UGT1A9 is associated with variable acetaminophen glucuronidation in neonates: a population pharmacokinetic and pharmacogenetic study. Clin Pharmacokinet 2018; 57: 1325–36.
    1. Yang J, Cai L, Huang H. et al. Genetic variations and haplotype diversity of the UGT1 gene cluster in the Chinese population. PLoS One 2012; 7: e33988..
    1. Ford SL, Lou Y, Lewis N. et al. Effect of rifabutin on the pharmacokinetics of oral cabotegravir in healthy subjects. Antivir Ther 2019; 24: 301–8.
    1. Ford SL, Gould E, Chen S. et al. Lack of pharmacokinetic interaction between rilpivirine and integrase inhibitors dolutegravir and GSK1265744. Antimicrob Agents Chemother 2013; 57: 5472–7.
    1. Spreen W, Williams P, Margolis D. et al. Pharmacokinetics, safety, and tolerability with repeat doses of GSK1265744 and rilpivirine (TMC278) long-acting nanosuspensions in healthy adults. J Acquir Immune Defic Syndr 2014; 67: 487–92.
    1. Margolis DA, Brinson CC, Smith GHR. et al. Cabotegravir plus rilpivirine, once a day, after induction with cabotegravir plus nucleoside reverse transcriptase inhibitors in antiretroviral-naive adults with HIV-1 infection (LATTE): a randomised, phase 2b, dose-ranging trial. Lancet Infect Dis 2015; 15: 1145–55.
    1. Margolis DA, Gonzalez-Garcia J, Stellbrink HJ. et al. Long-acting intramuscular cabotegravir and rilpivirine in adults with HIV-1 infection (LATTE-2): 96-week results of a randomised, open-label, phase 2b, non-inferiority trial. Lancet 2017; 390: 1499–510.
    1. Reese MJ, Savina PM, Generaux GT. et al. In vitro investigations into the roles of drug transporters and metabolizing enzymes in the disposition and drug interactions of dolutegravir, a HIV integrase inhibitor. Drug Metab Dispos 2013; 41: 353–61.
    1. Chen S, St Jean P, Borland J. et al. Evaluation of the effect of UGT1A1 polymorphisms on dolutegravir pharmacokinetics. Pharmacogenomics 2014; 15: 9–16.
    1. Speed B, Bu HZ, Pool WF. et al. Pharmacokinetics, distribution, and metabolism of [14C]sunitinib in rats, monkeys, and humans. Drug Metab Dispos 2012; 40: 539–55.
    1. Fujita K, Sugiyama M, Akiyama Y. et al. The small-molecule tyrosine kinase inhibitor nilotinib is a potent noncompetitive inhibitor of the SN-38 glucuronidation by human UGT1A1. Cancer Chemother Pharmacol 2011; 67: 237–41.
    1. Spraggs CF, Xu CF, Hunt CM.. Genetic characterization to improve interpretation and clinical management of hepatotoxicity caused by tyrosine kinase inhibitors. Pharmacogenomics 2013; 14: 541–54.

Source: PubMed

3
订阅