1,528
Views
1
CrossRef citations to date
0
Altmetric
Editorial

Current approaches and future strategies for the implementation of pharmacogenomics in the clinical use of azole antifungal drugs

&
Pages 509-514 | Received 17 Nov 2020, Accepted 11 Feb 2021, Published online: 23 Feb 2021

1. Introduction

Azoles are a heterogeneous group of antifungal drugs. They can be classified into two groups, those with two nitrogens in the azole ring, called imidazoles (include clotrimazole, econazole, ketoconazole, miconazole, and tioconazole) and those with three nitrogens in the azole ring, called triazoles (fluconazole, itraconazole, posaconazole, voriconazole and isavuconazole).

Agents within the azole class work primarily by inhibiting the fungal cytochrome P450 enzyme lanosterol 14α-demethylase. This leads to disruption of ergosterol biosynthesis, a component of the fungal cell membrane, which alters the functionality and structure of the fungal cell membrane, resulting in cell lysis and death [Citation1].

Imidazoles are now usually confined to topical use due to its unfavorable safety profile, while triazoles, with superior pharmacokinetics and improved safety profiles, are widely used for prophylaxis and treatment of invasive fungal disease, such as invasive aspergillosis, for which voriconazole has become the standard of care [Citation1,Citation2].

Pharmacokinetic properties vary between drugs, but all classes of azoles have the potential to be both substrates and inhibitors of different CYP (cytochrome P450) enzymes, conjugative enzymes and transport proteins [Citation2]. Polymorphic expression of these enzymes and transporters has been widely recognized as an important source of variability in the disposition and response to these drugs, which makes them potential biomarkers to predict optimal doses and drug response. However, the implementation of these biomarkers in the clinical setting has been uneven [Citation3].

2. Body

The difficulties to validate and implement the use of these biomarkers are well described in the literature, including low institutional promotion, insufficiency of available evidence and cost-effectiveness, lack of clinical guidelines and protocols, and economic and institutional issues as well as ethical, legal, and social implications [Citation4,Citation5]. Implementation of pharmacogenetics into clinical practice needs of a sequential process. Once a gene-drug interaction is identified, evidence of statistically associations between the pharmacogene and clinical outcomes needs to be provided. The prevalence of the clinically actionable pharmacogenes in the target population is another key aspect to consider. In addition, to assist physicians to apply this information, pharmacogenomic guidelines with consistent recommendations have to be developed. Finally, demonstrating cost-effectiveness and impact on cost is essential to justify and facilitate the adoption of Pharmacogenetics as a tool able to save significant costs to healthcare systems [Citation5,Citation6].

Polymorphic expression of genes coding azole-metabolizing enzymes and transporters has been described, and some of these genetic variations have been associated to interindividual heterogeneity in disposition, response, or toxicity of these drugs. Still, for the majority of azoles there are no clinical guidelines or recommendations that take into account the pharmacogenetic information to achieve an accurate and optimized therapeutic plan.

In a review done in the Pharmacogenomics Knowledgebase website (PharmGKB; www.pharmgkb.org) no actionable pharmacogenetic information was found for imidazole drugs, no clinical or drug label annotations and no prescribing information were available. Only a few variant annotations (genetic variant–drug associations as reported in a single publication) were described for ketoconazole. Information found for most triazoles was in line with the previous group. There is no information about isavuconazole, some variant annotations are available for fluconazole, one for itraconazole, and there is one annotation of EMA Label for posaconazole and CYP3A4 but it is related to metabolism drug–drug interactions with CYP3A4 substrates. On the contrary, there is significant information for voriconazole: clinical annotations, drug label annotations with actionable pharmacogenetics information and prescribing information including clinical guidelines providing drug dosing information based on pharmacogenetic data [Citation7].

The lack of pharmacogenetics information available for azoles except voriconazole can be explained by diverse reasons. All triazoles are substrates of CYP3A4 to a different extent, and although interindividual variability in CYP3A4 activity exists, only a few common genetic variants have been described and so far there is scarce evidence supporting dose adjustment for the majority of drugs metabolized by CYP3A4 [Citation3,Citation8]. Only isavuconazole undergoes metabolism by CYP3A5, but to a lesser extent than by CYP3A4 [Citation9,Citation10]. Thus far, the documented influence of CYP3A4 and CYP3A5 genotype on azoles is mostly related to interactions with CYP3A4/5 substrates, as they may significantly increase its concentrations in individuals homozygous for CYP3A5*3 variant, which is associated with a lack of CYP3A5 activity [Citation3,Citation11].

The triazoles are also substrates and/or inhibitors of conjugative enzymes and transporter proteins such as uridine diphosphate glucuronosyltransferase (UGT) or P-Glycoprotein (P-gp), but further insights are needed to clarify its role in triazole disposition, efficacy, and drug interactions [Citation2,Citation3]. Additionally, polymorphisms in genes coding regulatory proteins that interact with CYP enzymes, such as POR or NR1I2, seem to have an additive effect on the pharmacokinetics of some CYP-metabolized drugs, but not much actionable information is available in relation to this [Citation12].

Two enzymes of the CYP2C subfamily are involved in fluconazole and voriconazole, metabolism, CYP2C19 and CYP2C9 [Citation3,Citation13]. The CYP2C9 gene is highly polymorphic, but the most prevalent allele is CYP2C9*1, associated with normal enzyme activity, being the variants with decreased function notably infrequent [Citation3,Citation14]. Furthermore, fluconazole is mostly excreted as unchanged drug, and its metabolic pathway is not well described.

Conversely, lot of information is available related to Voriconazole metabolism. It is estimated that around 70–75% of its total metabolism is mediated through CYP P450 enzymes, mostly CYP2C19, CYP3A4, and CYP2C9, and the remaining 25–30% is mediated by the FMO (flavin-containing monooxy- genase) family [Citation12]. Despite several SNPs located in the genes associated with voriconazole metabolism have been related to variability in its clearance, to date, dosing guidelines for voriconazole are monogenic and rely exclusively on CYP2C19 [Citation15]. Further, even when guidelines are available, ambiguity in how to apply pharmacogenomic biomarker information exists. The Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for voriconazole and CYP2C19, recommends to select other antifungal agents in those patients with ultrarapid and poor metabolizer genotypes, and to prescribe the usual dosing in other genotypes. In contrast, the Dutch Pharmacogenetics Working Group guideline propose a dose adjustments for ultrarapid and poor metabolizer genotypes. Also, Hicks et al. propose a dose adjustment based on CYP2C19 genotype in children with leukemia, which would be a very good option for this first line drug in invasive aspergillosis [Citation15–18].

summarizes for each azole the gene–drug interactions described in the literature and the level of evidence.

Table 1. Summary of gene–drug interaction described in the literature for each azole and level of evidence

3. Expert opinion

Pharmacogenetics has emerged in the last years as a key discipline in the progress toward personalized medicine. Despite the fact that data available to this subject are experiencing exponential growth, pharmacogenomic testing are rapidly evolving and its costs progressively dropping, the clinical utility and cost-effectiveness of pharmacogenetic testing is sometimes restricted to certain gene–drug combinations and/or specific populations.

Herein, we have summarized the current status of Pharmacogenetics in the clinical use of azole antifungal drugs. Although several gene gene–drug associations are described for different azoles, for the majority of combinations, current levels of evidence are insufficient to support recommendations for changing clinical practice.

Evidence to support clinical recommendations is only available for voriconazole, albeit recommendations are not concurrent and large-scale implementation is not established.

It is difficult to predict how implementation of pharmacogenomics in the clinical use of azole will be carried out in the future. High-level evidence on genetic markers’ efficacy, effectivity and efficiency, needs to be obtained along with consensus on recommendations. To this aim, large population-based studies associating these biomarkers to drug response should be undertaken.

We suggest that, the ideal strategy to implement into clinical practice an efficacious strategy for dose individualization would be the development of prediction algorithms based on those factors contributing to drug variability in efficacy and safety, including pharmacogenetic and other clinical and/or physiological information. We also find that a preemptive genotyping strategy in risk populations (those who are susceptible to receive a drug) for which the pharmacogenetics information would be available at the moment of prescription allowing a precise initial dosing, is essential for bringing pharmacogenetics and clinics together.

In the specific case of voriconazole, there is sufficient evidence supporting implementation of pharmacogenetics in the clinical use (plenty of association studies and clinical guidelines are available), but it is necessary to evaluate the effectiveness and efficiency of the implementation. In this regard, our group is developing a multicenter, randomized clinical trial to evaluate the effectiveness and efficiency of a preemptive genotyping strategy for voriconazole, including an economic evaluation from the perspective of the Spanish National Health System [Citation19]. In our view, the data available strongly support the hypothesis that voriconazole pharmacogenetics combined with close therapeutic drug monitoring of serum levels (which is already implemented in many hospitals), could improve the individualization of voriconazole dosing and we hope that the information obtained from this clinical trial will contribute to facilitate the implementation of pharmacogenetics into clinical practice [Citation20].

Recent studies suggest that as drug exposure can be influenced by inflammation-induced phenoconversion, in addition to pharmacogenetics, the inflammatory status should be also taken into account when selecting voriconazole dose, especially in individuals with invasive fungal infections [Citation21].

With respect to other azoles, further studies are needed to obtain evidence to support the influence of genomic biomarkers over dosing, safety, or efficacy.

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This paper was not funded.

References

  • Nocua-Báez LC, Uribe-Jerez P, Tarazona-Guaranga L, et al. Azoles de antes y ahora: una revisión [Azoles of then and now: a review]. Rev Chilena Infectol. 2020 Jun;37(3):219–230. Spanish. PMID: 32853312.
  • Ashbee HR, Gilleece MH. 2014. Chapter 38 - pharmacogenomics of antifungal agents. Handbook of Pharmacogenomics and Stratified Medicine, 879–900. 978-0-12-386882-4. DOI:10.1016/B978-0-12-386882-4.00038-4.
  • Amsden JR, Gubbins PO. Pharmacogenomics of triazole antifungal agents: implications for safety, tolerability and efficacy. Expert Opin Drug Metab Toxicol. 2017 Nov;13(11):1135–1146. Epub 2017 Oct 20. PMID: 29022838.
  • Borobia AM, Dapia I, Tong HY, et al. Clinical implementation of pharmacogenetic testing in a hospital of the Spanish national health system: strategy and experience over 3 years. Clin Transl Sci. 2018 Mar;11(2):189–199. Epub 2017 Nov 28. PMID: 29193749; PMCID: PMC5866958.
  • Hippman C, Nislow C. Pharmacogenomic testing: clinical evidence and implementation challenges. J Pers Med. 2019 Aug 7;9(3):40. PMID: 31394823; PMCID: PMC6789586.
  • Mizzi C, Dalabira E, Kumuthini J, et al. Spectrum of pharmacogenomic biomarkers: implications for clinical pharmacogenomics. PLoS One. 2016 Sep 16;11(9):e0162866. Erratum in: PLoS One. 2017 Feb 16;12 (2):e0172595. PMID: 27636550; PMCID: PMC5026342.
  • Whirl-Carrillo M, McDonagh EM, Hebert JM, et al. Pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther. 2012 Oct [Accesed 2020 Nov 13];92(4):414–417. PMID: 22992668; PMCID: PMC3660037. Available from: www.pharmgkb.org.
  • Saiz-Rodríguez M, Almenara S, Navares-Gómez M, et al. Effect of the most relevant CYP3A4 and CYP3A5 polymorphisms on the pharmacokinetic parameters of 10 CYP3A substrates. Biomedicines. 2020 Apr 22;8(4):94. PMID: 32331352; PMCID: PMC7235792.
  • Darnaud L, Lamoureux F, Godet C, et al. Isavuconazole kinetic exploration for clinical practice. Drugs R D. 2018 Dec;18(4):317–321. PMID: 30306415; PMCID: PMC6277320.
  • Townsend R, Dietz A, Hale C, et al. Pharmacokinetic evaluation of CYP3A4-mediated drug-drug interactions of isavuconazole with rifampin, ketoconazole, midazolam, and ethinyl estradiol/norethindrone in healthy adults. Clin Pharmacol Drug Dev. 2017 Jan;6(1):44–53. Epub 2016 Jul 25. PMID: 27273461; PMCID: PMC5298035.
  • Shirasaka Y, Chang SY, Grubb MF, et al. Effect of CYP3A5 expression on the inhibition of CYP3A-catalyzed drug metabolism: impact on modeling CYP3A-mediated drug-drug interactions. Drug Metab Dispos. 2013 Aug;41(8):1566–1574. Epub 2013 May 30. PMID: 23723360; PMCID: PMC3716306.
  • Dapía I, García I, Martinez JC, et al. Prediction models for voriconazole pharmacokinetics based on pharmacogenetics: AN exploratory study in a Spanish population. Int J Antimicrob Agents. 2019 Oct;54(4):463–470. Epub 2019 Jul 4. PMID: 31279853.
  • Niwa T, Shiraga T, Takagi A. Effect of antifungal drugs on cytochrome P450 (CYP) 2C9, CYP2C19, and CYP3A4 activities in human liver microsomes. Biol Pharm Bull. 2005 Sep;28(9):1805–1808. . PMID: 16141567.
  • Scordo MG, Aklillu E, Yasar U, et al. Genetic polymorphism of cytochrome P450 2C9 in a Caucasian and a black African population. Br J Clin Pharmacol. 2001 Oct;52(4):447–450. PMID: 11678789; PMCID: PMC2014581.
  • García IG, Carcas AJ, Borobia AM. Strategy to effectively and efficiently implement voriconazole pharmacogenetics in clinical practice. Pharmacogenomics. 2020 Jul;21(10):647–649. Epub 2020 Jun 3. PMID: 32486895.
  • Hicks JK, Crews KR, Flynn P, et al. Voriconazole plasma concentrations in immunocompromised pediatric patients vary by CYP2C19 diplotypes. Pharmacogenomics. 2014 Jun;15(8):1065–1078. PMID: 25084200; PMCID: PMC4155516.
  • Moriyama B, Obeng AO, Barbarino J, et al. Clinical pharmacogenetics implementation consortium (CPIC) guidelines for CYP2C19 and voriconazole therapy. Clin Pharmacol Ther. 2017 Jul;102(1):45–51. Epub 2017 Apr 18. Erratum in: Clin Pharmacol Ther. 2018 Feb;103(2):349. PMID: 27981572; PMCID: PMC5474211.
  • Dutch Pharmacogenetics Working Group. Pharmacogenetic recommendations; 2020. [cited 2020 Nov 1]. Available from: https://www.knmp.nl/@@search.
  • Monserrat Villatoro J, García García I, Bueno D, et al. Randomised multicentre clinical trial to evaluate voriconazole pre-emptive genotyping strategy in patients with risk of aspergillosis: vorigenipharm study protocol. BMJ Open. 2020 Oct 1;10(10):e037443. PMID: 33004392; PMCID: PMC7534724.
  • Miller MA, Lee YM. Applying pharmacogenomics to antifungal selection and dosing: are we there yet? Curr Fungal Infect Rep. 2020 Mar;14(1):63–75. Epub 2020 Jan 16. PMID: 32256938; PMCID: PMC7111498.
  • Gautier-Veyret E, Thiebaut-Bertrand A, Roustit M, et al. Optimization of voriconazole therapy for treatment of invasive aspergillosis: pharmacogenomics and inflammatory status need to be evaluated. Br J Clin Pharmacol. 2020 Nov 20. Epub ahead of print. PMID: 33217017. DOI:10.1111/bcp.14661

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.