Next Article in Journal
Low-Level Tetracycline Resistance Gene tet(O)_3 in Campylobacter jejuni
Next Article in Special Issue
Improving Antimicrobial Use to Protect the Environment: What Is the Role of Infection Specialists?
Previous Article in Journal
Establishing an Antimicrobial Stewardship Program in Sierra Leone: A Report of the Experience of a Low-Income Country in West Africa
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Relevance of Pharmacogenomics to the Safe Use of Antimicrobials

Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
Antibiotics 2023, 12(3), 425; https://doi.org/10.3390/antibiotics12030425
Submission received: 27 January 2023 / Revised: 11 February 2023 / Accepted: 20 February 2023 / Published: 21 February 2023

Abstract

:
There has been widespread implementation of pharmacogenomic testing to inform drug prescribing in medical specialties such as oncology and cardiology. Progress in using pharmacogenomic tests when prescribing antimicrobials has been more limited, though a relatively large number of pharmacogenomic studies on aspects such as idiosyncratic adverse drug reactions have now been performed for this drug class. Currently, there are recommendations in place from either National Regulatory Agencies and/or specialist Pharmacogenomics Advisory Groups concerning genotyping for specific variants in MT-RNR1 and CYP2C19 before prescribing aminoglycosides and voriconazole, respectively. Numerous additional pharmacogenomic associations have been reported concerning antimicrobial-related idiosyncratic adverse drug reactions, particularly involving specific HLA alleles, but, to date, the cost-effectiveness of genotyping prior to prescription has not been confirmed. Polygenic risk score determination has been investigated to a more limited extent but currently suffers from important limitations. Despite limited progress to date, the future widespread adoption of preemptive genotyping and genome sequencing may provide pharmacogenomic data to prescribers that can be used to inform prescribing and increase the safe use of antimicrobials.

1. Introduction

Pharmacogenomic testing either preemptively or immediately prior to the prescription of a new drug is an increasingly available option worldwide [1]. However, the implementation of testing remains limited, though it is increasingly being applied in certain medical specialties, especially oncology, cardiology and psychiatry. Antimicrobials are among the most widely prescribed drugs but, at present, the implementation of pharmacogenomic testing in conjunction with their prescription is very limited. The current perspective considers the potential for increasing the use of pharmacogenomic testing when prescribing drugs for the treatment of bacterial and fungal infections. Testing is already used fairly widely when using certain anti-viral drugs [2], but a detailed discussion of this area is outside the scope of the current article.
While many antimicrobials have wide therapeutic windows, which are helpful in terms of safe use, and also show good efficacy when prescribed appropriately, there are particular issues with idiosyncratic adverse drug reactions with some widely used members of this drug class [3]. This aspect and current progress in understanding the underlying basis for susceptibility will be considered in detail, together with examples of recommendations for pharmacogenomic testing in relation to the use of certain antimicrobials.

2. Adverse Drug Reactions Involving HLA Gene Associations

Idiosyncratic reactions, particularly involving the skin or the liver (and occasionally both organs), are the most common type of adverse reaction seen for antimicrobials [4]. As reviewed in detail elsewhere [5], there is evidence that many of these reactions are immune-mediated. There are also data pointing to genetic susceptibility to the reactions [6]. The most polymorphic immune-related genes in humans are the human leukocyte antigen (HLA) genes, which contribute to genetic susceptibility to a range of diseases where there is a genetic component. The possibility that a HLA genotype contributes to the risk of idiosyncratic adverse drug reactions has therefore been investigated in detail and a number of specific HLA gene associations involving antimicrobial agents have been reported (Table 1) [6]. Other immune-related genes may also contribute to this risk (see Section 5 below), but, to date, the strongest associations reported have been for the HLA genes.
The reports of strong associations between specific HLA genotypes and susceptibility to adverse drug reactions with antimicrobials summarised in Table 1 have been generally well replicated. This has prompted further studies on both the mechanism for the reaction and the feasibility of genotyping for the risk alleles before prescribing the relevant drug. Good progress has been made in understanding the underlying mechanism, with clear data showing that it likely involves the covalent binding of the drug to cellular proteins followed by an inappropriate T-cell response [20]. Despite the strong and mostly well-replicated associations seen, clinical implementation of genotyping before drug prescription for the associations summarised in Table 1 has not been seen. Even in the case of the strongest association listed (flucloxacillin-induced liver injury and HLA-B*57:01), detailed analysis suggests that genotyping is not cost-effective [21]. It is possible that this may change as the preemptive sequencing of whole genomes with the inclusion of data in electronic medical records becomes more common and data on HLA genotypes are immediately available to prescribers.

3. Other Common Adverse Drug Reactions That Appear Independent of HLA Genotype

3.1. Anti-TB Drug Treatment and Liver Toxicity

Patients suffering from tuberculosis (TB) are most commonly treated with a combination of the drugs isoniazid, rifampicin, pyrazinamide and ethambutol [3,22]. Mild hepatotoxicity characterised by transient elevation of liver transaminase enzyme levels is common during treatment and affects up to 20% of those treated, as discussed in detail elsewhere [23,24]. However, it is estimated that approximately 1% of those treated with this drug combination suffer significant drug-induced liver injury (DILI) and have more prolonged and severe transaminase elevation combined with other symptoms of hepatitis [25]. These patients usually recover when the drugs are withdrawn but a subgroup may develop liver failure [24]. Most cases of significant liver injury with the anti-TB drugs appear to be due to isoniazid [24,26]. However, rifampicin may contribute to the reaction severity [25], and some cases of DILI due to pyrazinamide alone have also been reported [26]. Patients undergoing treatment with isoniazid alone for latent tuberculosis may also develop DILI [24]. A number of risk factors for the development of DILI due to isoniazid have been identified, including age and some environmental factors, such as alcohol consumption, but genetic risk factors also contribute [24].
The most extensively investigated genetic risk factor for the development of isoniazid-related liver injury has been the NAT2 gene, which encodes N-acetyltransferase 2 (NAT2), the major metabolising enzyme for isoniazid (Figure 1). Depending on ethnicity, between 10 and 60% of individuals lack this enzyme activity due to homozygosity for variants causing amino acid changes (for review, see [27]). There is a large body of literature suggesting that NAT2 deficiency is associated with an increased risk of developing DILI, though not all reports agree that this is the case (for review, see [28]). Two recent studies involving genome-wide association approaches suggest that there is a genuine NAT2 association with isoniazid-related DILI [29,30], but one of these [30] found that only those homozygous for variants associated with strongly impaired enzyme activity [31] were at risk. This difference in overall conclusions may be due to ethnic differences in NAT2 allele frequencies and reflect the fact that one study was performed in Thailand and the second in Europe and India. As indicated in Figure 1, the mechanism for toxicity may involve the formation of high levels of hydrazine in those classified as “ultraslow” for NAT2 activity. One study in animals suggested that hydrazine is highly hepatotoxic compared with other metabolites of isoniazid [32]. Though further work is needed in this area, especially on the underlying mechanism for toxicity, results from a clinical trial on assigning an isoniazid dose based on NAT2 genotype are reasonably encouraging [33].

3.2. Aminoglycosides and Deafness

Aminoglycoside antibiotics have been known to cause ototoxicity in some patients for many years. There are several different risk factors for this serious toxicity (for review, see [34]), but genetic predisposition is one of these. Genetic susceptibility involves the mitochondrial gene RNR1 (MT-RNR1), which encodes the 12S ribosomal RNA subunit of the mitochondrial ribosome. The basis for this association is now clear, with certain variants showing increased binding of aminoglycoside molecules, which may lead directly to this toxicity within auditory cells [35]. Carriage of one of three variants in MT-RNR1 (1095C > T, 1494C > T and 1555A > G) is considered to be associated with a high risk of aminoglycoside-induced ototoxicity, though there are also a number of other variants where an increased risk is also considered to be possible [36]. Unlike the other gene examples considered in this perspective, the variants relevant to aminoglycoside ototoxicity are very rare. Although the association between RNR1 genotype and susceptibility has been known for some time, recommendations from both National Drug Regulators and a Specialist Pharmacogenomics Advisory Group that testing should be performed in patients prior to aminoglycoside prescription and efforts made to prescribe a suitable alternative antimicrobial to those positive have only recently appeared [37,38]. A recent study has shown the utility of a point-of-care genotyping test prior to gentamycin administration in neonates [36]. This approach can also be applied to adult patients who are also at risk of toxicity if they carry an “at risk” mitochondrial variant [38]. Not all cases of aminoglycoside ototoxicity are due to genetic risk factors, so other risk factors need to be also considered when prescribing these drugs (for review, see [39]).

3.3. Fluoroquinolone Toxicity

The fluoroquinolones as a class have been associated with a number of adverse reactions, including skin and liver toxicity, but also a range of other effects, including aortic and tendon ruptures and Clostridium difficile infections [40,41]. To date, no associations with HLA genes or indeed any other gene have been reported for any of these reactions. The absence of reports on genetic associations to date may be due to the heterogeneity of the reported reactions and, despite some surveys suggesting adverse reactions to fluoroquinolones are relatively common [40,41], only small numbers of cases are available for detailed analysis. Restrictions on the prescribing of these agents have been recommended since 2018 by the European Medicines Agency (https://www.ema.europa.eu/en/news/fluoroquinolone-quinolone-antibiotics-prac-recommends-new-restrictions-use-following-review, accessed on 27 January 2023), but in view of their broad specificity, understanding better the basis for adverse reactions would be helpful, especially if individuals at increased risk of these reactions can be identified by genetic testing.

4. Voriconazole and CYP2C19

Voriconazole is a triazole antifungal agent that is useful in the treatment of invasive fungal infections, particularly aspergillosis [42]. The metabolism of this agent involves a key role for the cytochrome P450 CYP2C19. This gene is subject to common genetic polymorphisms, which can result in either unusually high (ultrarapid metaboliser) or low (poor metaboliser) phenotypes [43], and, in line with this, it is well established that individual plasma concentrations of voriconazole vary considerably [44]. This variability led to a recommendation to perform therapeutic drug monitoring to ensure that adequate levels of the drug are achieved [44]. More recently, it has been proposed by the Clinical Pharmacogenetics Implementation Consortium (CPIC) (a USA-based Pharmacogenomics Advisory Group) that genotyping for CYP2C19 be performed prior to the start of treatment, with the use of alternative antifungals recommended in both ultrarapid metabolisers who have two copies of the CYP2C19*17 allele (approximately 4% of Europeans), and in poor metabolisers who are typically homozygous for CYP2C19*2 (approximately 2% of Europeans) [45]. The ultrarapid metabolisers are unlikely to achieve adequate plasma levels, whereas the poor metabolisers are at risk of a range of adverse drug reactions, including hepatoxicity and visual disturbances. Recent Dutch Pharmacogenomics guidelines (https://www.pharmgkb.org/chemical/PA10233/guidelineAnnotation/PA166104990, accessed on 27 January 2023) suggest that voriconazole dose adjustment based on CYP2C19 genotype may be an alternative approach to the use of other drugs. Voriconazole is the best example of an antimicrobial where a common pharmacogenetic polymorphism in a cytochrome P450 gene affects plasma levels directly and where genotyping for this polymorphism is of considerable value. Access to CYP2C19 genotyping is not always available, even in well-developed healthcare systems, but, as discussed above, therapeutic drug monitoring may be a helpful alternative.

5. Polygenic Risk Scores

Many complex traits, including both responses to drugs and susceptibility to adverse drug reactions, are likely to involve contributions from a number of different genetic variants [46,47]. As a result of the considerable progress made in studies involving genome-wide association studies and genome sequencing, a number of polygenic risk scores based on a number of genetic variants have been developed, but very few of these relate to pharmacogenomics [47]. Though a very promising approach, implementation in routine clinical practice to date is limited. At least two studies involving the development of polygenic risk scores to predict susceptibility to drug-induced liver injury due to antimicrobials have now appeared [11,48]. The first of these involved combining data from a genome-wide association study with data from in vitro studies on liver gene expression to select appropriate variants for inclusion in the score [48]. This resulted in the development of a score involving genotypes for almost 28,000 different variants. The genome-wide association study data used to develop this score did not include data from patients with either flucloxacillin- or amoxicillin-clavulanate-induced DILI, and the score was then assessed as a predictor of DILI due to either of these drugs in a new patient cohort. It was found that the score was a statistically significant predictor of both forms of DILI, with higher average risk scores seen in cases of DILI compared with healthy controls. The complexity of the score makes clinical implementation unlikely, but the finding is still interesting because it suggests that there are common susceptibility factors for DILI due to different drugs, unlike the findings with HLA genes discussed in Section 1 above, where different drugs causing DILI tend to show separate HLA associations. It seems likely that the drug non-specific risk factors involve variants, which each contribute a very small increase in risk.
A more recent study on amoxicillin-clavulanate DILI only has led to the development of a less complex polygenic risk score that is more specific for this drug only [11]. This builds on the three HLA associations for DILI due to this drug listed in Table 1. In addition to these HLA associations, two other genetic risk factors for this form of DILI have recently been detected. These are in the immune-related genes PTPN22 and ERAP2 [11,49]. While the effect sizes for the variants in these two genes are lower than those seen with HLA alleles, the two associations have been replicated in an independent cohort [11]. A polygenic risk score for amoxicillin-clavulanate DILI has been developed to include the three HLA risk factors listed in Table 1 (HLA-A*02:01, HLA-B*15:18 and HLA-DRB1*15:01) together with the two additional variants. In a typical population of European ethnicity, 99% of individuals will show a score of 4.5 or lower, with approximately 44% scoring below 0.5. Individuals with a score of 1.58 or higher are at increased risk of developing this form of DILI, with a specificity of 79% and sensitivity of 56%. In the study reporting this finding [11], 56% of the amoxicillin-clavulanate DILI cases had a score of >1.58, compared with 21% of controls. Whether this level of risk makes determining the risk score in advance of prescribing useful is doubtful, but it is possible that the risk score could be used as a diagnostic tool when assessing which drug is responsible for DILI in patients who have been exposed to several different drugs.

6. Conclusions

Use of pharmacogenomic tests before prescribing antimicrobials remains limited but, over the last 5 to 10 years, some progress has been made in very specific areas, including the prevention of aminoglycoside ototoxicity and more effective prescription of voriconazole. Both these examples are most relevant to specialist hospital settings, and no pharmacogenomic tests that are suitable for implementation in the primary care prescription of antimicrobials have yet been developed. Although these current examples of implementation are most relevant to specialist hospital settings, the technology required to perform the tests is quite cheap and can be used at the point of care [36,50]. CYP2C19, which is relevant to voriconazole use, is also part of a “direct to the consumer” genotyping panel (https://www.23andme.com/en-gb/test-info/pharmacogenetics/, accessed on 7 February 2023).
When antimicrobials used more widely in primary care and outpatient settings are considered, adverse reactions to these drugs have been well studied at the pharmacogenomic level and interesting associations with HLA and a few other genes have emerged. This has led to a better understanding of the biological basis of these reactions. However, it is currently not cost-effective to implement testing for the risk genotypes described. As both preemptive genotyping and genome sequencing become more widespread [51], this situation may change, with further cost-effectiveness analysis likely to be useful. Refinement and improvement of approaches involving the use of polygenic risk scores is also likely.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. van der Wouden, C.H.; Guchelaar, H.J.; Swen, J.J. Precision Medicine Using Pharmacogenomic Panel-Testing: Current Status and Future Perspectives. Clin. Lab. Med. 2022, 42, 587–602. [Google Scholar] [CrossRef]
  2. Cusato, J.; Allegra, S.; Nicolò, A.; Calcagno, A.; D’Avolio, A. Precision medicine for HIV: Where are we? Pharmacogenomics 2018, 19, 145–165. [Google Scholar] [CrossRef]
  3. Brunton, L.; Knollmann, B. Goodman and Gilman’s the Pharmacological Basis of Therapeutics, 14th ed.; McGraw-Hill Education: New York, NY, USA, 2022. [Google Scholar]
  4. Blumenthal, K.G.; Peter, J.G.; Trubiano, J.A.; Phillips, E.J. Antibiotic allergy. Lancet 2019, 393, 183–198. [Google Scholar] [CrossRef]
  5. Uetrecht, J.; Naisbitt, D.J. Idiosyncratic adverse drug reactions: Current concepts. Pharmacol. Rev. 2013, 65, 779–808. [Google Scholar] [CrossRef] [Green Version]
  6. Daly, A.K. Genetics of drug-induced liver injury: Current knowledge and future prospects. Clin. Transl. Sci. 2022, 16, 37–42. [Google Scholar] [CrossRef]
  7. Lucena, M.I.; Molokhia, M.; Shen, Y.; Urban, T.J.; Aithal, G.P.; Andrade, R.J.; Day, C.P.; Ruiz-Cabello, F.; Donaldson, P.T.; Stephens, C.; et al. Susceptibility to Amoxicillin-Clavulanate-Induced Liver Injury Is Influenced by Multiple HLA Class I and II Alleles. Gastroenterology 2011, 141, 338–347. [Google Scholar] [CrossRef]
  8. Nicoletti, P.; Aithal, G.P.; Bjornsson, E.S.; Andrade, R.J.; Sawle, A.; Arrese, M.; Barnhart, H.X.; Bondon-Guitton, E.; Hayashi, P.H.; Bessone, F.; et al. Association of Liver Injury From Specific Drugs, or Groups of Drugs, With Polymorphisms in HLA and Other Genes in a Genome-Wide Association Study. Gastroenterology 2017, 152, 1078–1089. [Google Scholar] [CrossRef] [Green Version]
  9. Zhang, F.R.; Liu, H.; Irwanto, A.; Fu, X.A.; Li, Y.; Yu, G.Q.; Yu, Y.X.; Chen, M.F.; Low, H.Q.; Li, J.H.; et al. HLA-B*13:01 and the dapsone hypersensitivity syndrome. N. Engl. J. Med. 2013, 369, 1620–1628. [Google Scholar] [CrossRef] [Green Version]
  10. Li, Y.J.; Phillips, E.J.; Dellinger, A.; Nicoletti, P.; Schutte, R.; Li, D.; Ostrov, D.A.; Fontana, R.J.; Watkins, P.B.; Stolz, A.; et al. Human Leukocyte Antigen B*14:01 and B*35:01 Are Associated With Trimethoprim-Sulfamethoxazole Induced Liver Injury. Hepatology 2021, 73, 268–281. [Google Scholar] [CrossRef]
  11. Nicoletti, P.; Dellinger, A.; Li, Y.J.; Barnhart, H.X.; Chalasani, N.; Fontana, R.J.; Odin, J.A.; Serrano, J.; Stolz, A.; Etheridge, A.S.; et al. Identification of Reduced ERAP2 Expression and a Novel HLA Allele as Components of a Risk Score for Susceptibility to Liver Injury Due to Amoxicillin-Clavulanate. Gastroenterology 2022, in press. [Google Scholar] [CrossRef]
  12. Urban, T.J.; Nicoletti, P.; Chalasani, N.; Serrano, J.; Stolz, A.; Daly, A.K.; Aithal, G.P.; Dillon, J.; Navarro, V.; Odin, J.; et al. Minocycline hepatotoxicity: Clinical characterization and identification of HLA-B∗35:02 as a risk factor. J. Hepatol. 2017, 67, 137–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Krebs, K.; Bovijn, J.; Zheng, N.; Lepamets, M.; Censin, J.C.; Jürgenson, T.; Särg, D.; Abner, E.; Laisk, T.; Luo, Y.; et al. Genome-wide Study Identifies Association between HLA-B(∗)55:01 and Self-Reported Penicillin Allergy. Am. J. Hum. Genet. 2020, 107, 612–621. [Google Scholar] [CrossRef]
  14. Daly, A.K.; Donaldson, P.T.; Bhatnagar, P.; Shen, Y.; Pe’er, I.; Floratos, A.; Daly, M.J.; Goldstein, D.B.; John, S.; Nelson, M.R.; et al. HLA-B*5701 genotype is a major determinant of drug-induced liver injury due to flucloxacillin. Nat. Genet. 2009, 41, 816–819. [Google Scholar] [CrossRef] [PubMed]
  15. Petros, Z.; Kishikawa, J.; Makonnen, E.; Yimer, G.; Habtewold, A.; Aklillu, E. HLA-B*57 Allele Is Associated with Concomitant Anti-tuberculosis and Antiretroviral Drugs Induced Liver Toxicity in Ethiopians. Front. Pharmacol. 2017, 8, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Nicoletti, P.; Aithal, G.P.; Chamberlain, T.C.; Coulthard, S.; Alshabeeb, M.; Grove, J.I.; Andrade, R.J.; Bjornsson, E.; Dillon, J.F.; Hallberg, P.; et al. Drug-Induced Liver Injury due to Flucloxacillin: Relevance of Multiple Human Leukocyte Antigen Alleles. Clin. Pharmacol. Ther. 2019, 106, 245–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Nicoletti, P.; Carr, D.F.; Barrett, S.; McEvoy, L.; Friedmann, P.S.; Shear, N.H.; Nelson, M.R.; Chiriac, A.M.; Blanca-López, N.; Cornejo-García, J.A.; et al. Beta-lactam-induced immediate hypersensitivity reactions: A genome-wide association study of a deeply phenotyped cohort. J. Allergy Clin. Immunol. 2021, 147, 1830–1837.e1815. [Google Scholar] [CrossRef] [PubMed]
  18. Chalasani, N.; Li, Y.J.; Dellinger, A.; Navarro, V.; Bonkovsky, H.; Fontana, R.J.; Gu, J.; Barnhart, H.; Phillips, E.; Lammert, C.; et al. Clinical features, outcomes, and HLA risk factors associated with nitrofurantoin-induced liver injury. J. Hepatol. 2023, 78, 293–300. [Google Scholar] [CrossRef] [PubMed]
  19. Romano, A.; Oussalah, A.; Chery, C.; Guéant-Rodriguez, R.M.; Gaeta, F.; Cornejo-Garcia, J.A.; Rouyer, P.; Josse, T.; Mayorga, C.; Torres, M.J.; et al. Next-generation sequencing and genotype association studies reveal the association of HLA-DRB3*02:02 with delayed hypersensitivity to penicillins. Allergy 2022, 77, 1827–1834. [Google Scholar] [CrossRef]
  20. Jee, A.; Sernoskie, S.C.; Uetrecht, J. Idiosyncratic Drug-Induced Liver Injury: Mechanistic and Clinical Challenges. Int. J. Mol. Sci. 2021, 22, 2954. [Google Scholar] [CrossRef]
  21. Manson, L.E.N.; van den Hout, W.B.; Guchelaar, H.J. Genotyping for HLA Risk Alleles to Prevent Drug Hypersensitivity Reactions: Impact Analysis. Pharmaceuticals 2021, 15, 4. [Google Scholar] [CrossRef]
  22. Alffenaar, J.W.C.; Stocker, S.L.; Forsman, L.D.; Garcia-Prats, A.; Heysell, S.K.; Aarnoutse, R.E.; Akkerman, O.W.; Aleksa, A.; van Altena, R.; de Oñata, W.A.; et al. Clinical standards for the dosing and management of TB drugs. Int. J. Tuberc. Lung Dis. 2022, 26, 483–499. [Google Scholar] [CrossRef] [PubMed]
  23. Saukkonen, J.J.; Cohn, D.L.; Jasmer, R.M.; Schenker, S.; Jereb, J.A.; Nolan, C.M.; Peloquin, C.A.; Gordin, F.M.; Nunes, D.; Strader, D.B.; et al. An official ATS statement: Hepatotoxicity of antituberculosis therapy. Am. J. Respir. Crit. Care Med. 2006, 174, 935–952. [Google Scholar] [CrossRef] [Green Version]
  24. Ramappa, V.; Aithal, G.P. Hepatotoxicity Related to Anti-tuberculosis Drugs: Mechanisms and Management. J. Clin. Exp. Hepatol. 2013, 3, 37–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Garibaldi, R.A.; Drusin, R.E.; Ferebee, S.H.; Gregg, M.B. Isoniazid-associated hepatitis. Report of an outbreak. Am. Rev. Respir. disease 1972, 106, 357–365. [Google Scholar] [CrossRef] [PubMed]
  26. Senousy, B.E.; Belal, S.I.; Draganov, P.V. Hepatotoxic effects of therapies for tuberculosis. Nat. Rev. Gastroenterol. Hepatol. 2010, 7, 543–556. [Google Scholar] [CrossRef] [PubMed]
  27. Hein, D.W.; Millner, L.M. Arylamine N-acetyltransferase acetylation polymorphisms: Paradigm for pharmacogenomic-guided therapy- a focused review. Expert Opin. Drug Metab. Toxicol. 2021, 17, 9–21. [Google Scholar] [CrossRef]
  28. Richardson, M.; Kirkham, J.; Dwan, K.; Sloan, D.J.; Davies, G.; Jorgensen, A.L. NAT2 variants and toxicity related to anti-tuberculosis agents: A systematic review and meta-analysis. Int. J. Tuberc. Lung Dis. 2019, 23, 293–305. [Google Scholar] [CrossRef]
  29. Suvichapanich, S.; Wattanapokayakit, S.; Mushiroda, T.; Yanai, H.; Chuchottawon, C.; Kantima, T.; Nedsuwan, S.; Suwankesawong, W.; Sonsupap, C.; Pannarunothai, R.; et al. Genomewide Association Study Confirming the Association of NAT2 with Susceptibility to Antituberculosis Drug-Induced Liver Injury in Thai Patients. Antimicrob. Agents Chemother. 2019, 63, e02692-18. [Google Scholar] [CrossRef] [Green Version]
  30. Nicoletti, P.; Devarbhavi, H.; Goel, A.; Venkatesan, R.; Eapen, C.E.; Grove, J.I.; Zafer, S.; Bjornsson, E.; Lucena, M.I.; Andrade, R.J.; et al. Genetic Risk Factors in Drug-Induced Liver Injury Due to Isoniazid-Containing Antituberculosis Drug Regimens. Clin. Pharmacol. Ther. 2021, 109, 1125–1135. [Google Scholar] [CrossRef]
  31. Fukunaga, K.; Kato, K.; Okusaka, T.; Saito, T.; Ikeda, M.; Yoshida, T.; Zembutsu, H.; Iwata, N.; Mushiroda, T. Functional Characterization of the Effects of N-acetyltransferase 2 Alleles on N-acetylation of Eight Drugs and Worldwide Distribution of Substrate-Specific Diversity. Front. Genet. 2021, 12, 652704. [Google Scholar] [CrossRef]
  32. Richards, V.E.; Chau, B.; White, M.R.; McQueen, C.A. Hepatic gene expression and lipid homeostasis in C57BL/6 mice exposed to hydrazine or acetylhydrazine. Toxicol. Sci. 2004, 82, 318–332. [Google Scholar] [CrossRef] [Green Version]
  33. Azuma, J.; Ohno, M.; Kubota, R.; Yokota, S.; Nagai, T.; Tsuyuguchi, K.; Okuda, Y.; Takashima, T.; Kamimura, S.; Fujio, Y.; et al. NAT2 genotype guided regimen reduces isoniazid-induced liver injury and early treatment failure in the 6-month four-drug standard treatment of tuberculosis: A randomized controlled trial for pharmacogenetics-based therapy. Eur. J. Clin. Pharmacol. 2013, 69, 1091–1101. [Google Scholar] [CrossRef] [Green Version]
  34. Steyger, P.S. Mechanisms of Aminoglycoside- and Cisplatin-Induced Ototoxicity. Am. J. Audiol. 2021, 30, 887–900. [Google Scholar] [CrossRef]
  35. Hutchin, T.P.; Cortopassi, G.A. Mitochondrial defects and hearing loss. Cell Mol. Life Sci. 2000, 57, 1927–1937. [Google Scholar] [CrossRef]
  36. McDermott, J.H.; Mahaveer, A.; James, R.A.; Booth, N.; Turner, M.; Harvey, K.E.; Miele, G.; Beaman, G.M.; Stoddard, D.C.; Tricker, K.; et al. Rapid Point-of-Care Genotyping to Avoid Aminoglycoside-Induced Ototoxicity in Neonatal Intensive Care. JAMA Pediatr. 2022, 176, 486–492. [Google Scholar] [CrossRef]
  37. Medicines and Healthcare Products Regulatory Agency. Aminoglycosides (Gentamicin, Amikacin, Tobramycin, and Neomycin): Increased Risk of Deafness in Patients with Mitochondrial Mutations; Medicines and Healthcare Products Regulatory Agency: London, UK, 2021; pp. 19–21.
  38. McDermott, J.H.; Wolf, J.; Hoshitsuki, K.; Huddart, R.; Caudle, K.E.; Whirl-Carrillo, M.; Steyger, P.S.; Smith, R.J.H.; Cody, N.; Rodriguez-Antona, C.; et al. Clinical Pharmacogenetics Implementation Consortium Guideline for the Use of Aminoglycosides Based on MT-RNR1 Genotype. Clin. Pharmacol. Ther. 2022, 111, 366–372. [Google Scholar] [CrossRef]
  39. Kros, C.J.; Steyger, P.S. Aminoglycoside- and Cisplatin-Induced Ototoxicity: Mechanisms and Otoprotective Strategies. Cold Spring Harb. Perspect. Med. 2019, 9, a033548. [Google Scholar] [CrossRef]
  40. Kuula, L.S.M.; Backman, J.T.; Blom, M.L. Healthcare costs and mortality associated with serious fluoroquinolone-related adverse reactions. Pharmacol. Res. Perspect. 2022, 10, e00931. [Google Scholar] [CrossRef]
  41. Bove, C.; Baldock, R.A.; Champigneulle, O.; Martin, L.; Bennett, C.L. Fluoroquinolones: Old drugs, putative new toxicities. Expert Opin. Drug Saf. 2022, 21, 1365–1378. [Google Scholar] [CrossRef]
  42. Walsh, T.J.; Anaissie, E.J.; Denning, D.W.; Herbrecht, R.; Kontoyiannis, D.P.; Marr, K.A.; Morrison, V.A.; Segal, B.H.; Steinbach, W.J.; Stevens, D.A.; et al. Treatment of aspergillosis: Clinical practice guidelines of the Infectious Diseases Society of America. Clin. Infect. Dis. 2008, 46, 327–360. [Google Scholar] [CrossRef]
  43. Botton, M.R.; Whirl-Carrillo, M.; Del Tredici, A.L.; Sangkuhl, K.; Cavallari, L.H.; Agúndez, J.A.G.; Duconge, J.; Lee, M.T.M.; Woodahl, E.L.; Claudio-Campos, K.; et al. PharmVar GeneFocus: CYP2C19. Clin. Pharmacol. Ther. 2021, 109, 352–366. [Google Scholar] [CrossRef]
  44. Trifilio, S.; Pennick, G.; Pi, J.; Zook, J.; Golf, M.; Kaniecki, K.; Singhal, S.; Williams, S.; Winter, J.; Tallman, M.; et al. Monitoring plasma voriconazole levels may be necessary to avoid subtherapeutic levels in hematopoietic stem cell transplant recipients. Cancer 2007, 109, 1532–1535. [Google Scholar] [CrossRef]
  45. Moriyama, B.; Obeng, A.O.; Barbarino, J.; Penzak, S.R.; Henning, S.A.; Scott, S.A.; Agúndez, J.; Wingard, J.R.; McLeod, H.L.; Klein, T.E.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guidelines for CYP2C19 and Voriconazole Therapy. Clin. Pharmacol. Ther. 2017, 102, 45–51. [Google Scholar] [CrossRef] [Green Version]
  46. Evans, D.M.; Visscher, P.M.; Wray, N.R. Harnessing the information contained within genome-wide association studies to improve individual prediction of complex disease risk. Hum. Mol. Genet. 2009, 18, 3525–3531. [Google Scholar] [CrossRef] [Green Version]
  47. Cross, B.; Turner, R.; Pirmohamed, M. Polygenic risk scores: An overview from bench to bedside for personalised medicine. Front. Genet. 2022, 13, 1000667. [Google Scholar] [CrossRef]
  48. Koido, M.; Kawakami, E.; Fukumura, J.; Noguchi, Y.; Ohori, M.; Nio, Y.; Nicoletti, P.; Aithal, G.P.; Daly, A.K.; Watkins, P.B.; et al. Polygenic architecture informs potential vulnerability to drug-induced liver injury. Nat. Med. 2020, 26, 1541–1548. [Google Scholar] [CrossRef]
  49. Cirulli, E.T.; Nicoletti, P.; Abramson, K.; Andrade, R.J.; Bjornsson, E.S.; Chalasani, N.; Fontana, R.J.; Hallberg, P.; Li, Y.J.; Lucena, M.I.; et al. A Missense Variant in PTPN22 is a Risk Factor for Drug-induced Liver Injury. Gastroenterology 2019, 156, 1707–1716.e1702. [Google Scholar] [CrossRef] [Green Version]
  50. Baudhuin, L.M.; Train, L.J.; Goodman, S.G.; Lane, G.E.; Lennon, R.J.; Mathew, V.; Murthy, V.; Nazif, T.M.; So, D.Y.F.; Sweeney, J.P.; et al. Point of care CYP2C19 genotyping after percutaneous coronary intervention. Pharm. J. 2022, 22, 303–307. [Google Scholar] [CrossRef]
  51. Ji, X.; Ning, B.; Liu, J.; Roberts, R.; Lesko, L.; Tong, W.; Liu, Z.; Shi, T. Towards population-specific pharmacogenomics in the era of next-generation sequencing. Drug Discov. Today 2021, 26, 1776–1783. [Google Scholar] [CrossRef]
Figure 1. Metabolism of isoniazid by NAT2 and other enzymes. The main metabolites are shown, including hydrazine, which shows hepatotoxicity in animal models.
Figure 1. Metabolism of isoniazid by NAT2 and other enzymes. The main metabolites are shown, including hydrazine, which shows hepatotoxicity in animal models.
Antibiotics 12 00425 g001
Table 1. HLA allele risk factors for idiosyncratic adverse reactions related to antimicrobials that affect the skin or liver.
Table 1. HLA allele risk factors for idiosyncratic adverse reactions related to antimicrobials that affect the skin or liver.
HLA AlleleType of Idiosyncratic Adverse ReactionDrugOdds Ratio (Allelic)p-ValueReference
A*02:01Liver injuryAmoxicillin-clavulanate2.3 (1.8–2.9)1.8 × 10−10[7]
A*33:01Liver injuryTerbinafine40.5 (12.5–131.4)6.7 × 10−10[8]
B*13:01Hypersensitivity syndrome (particularly involving skin, with possible liver involvement)Dapsone21.7 (10.4–45.1)2.0 × 10−16[9]
B*14:01-C*08:02Liver injuryTrimethoprim-sulfamethoxazole8.7 (3.2–19.5)2.3 × 10−4[10]
B*15:18Liver injuryAmoxicillin-clavulanate3.1 (6.08–1.58)0.001[11,12]
B*35:02Liver injuryMinocycline29.6 (7.8–89.8)2.57 × 10−8[12]
B*55:01Early and delayed allergic reactions (mainly involving skin)Penicillins1.4 (1.3–1.5)2.0 × 10−31[13]
B*57:01Liver injuryFlucloxacillin36.6 (26.1–51.3)2.6 × 10−97[14]
B*57:02 and B*57:03Liver injuryAnti-HIV and anti-TB comb30.1 (3.4–263.1) 0.002[15]
B*57:03Liver injuryFlucloxacillin 19.8 (3.37–116.1)0.001[16]
DRB1*10:01Immediate hypersensitivity reactionPenicillins2.9 (2.0–4.4)5.4 × 10−7[17]
DRB1*11:04Liver injuryNitrofurantoin4.29 (2.18–7.66)1.2 × 10−4[18]
DRB1*15:01Liver injuryAmoxicillin-clavulanate2.8 (2.1–3.8)3.5 × 10−11[7]
DRB3*02:02Delayed hypersensitivity reactionPenicillins8.9 (3.4–23.3)p < 0.0001[19]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Daly, A.K. Relevance of Pharmacogenomics to the Safe Use of Antimicrobials. Antibiotics 2023, 12, 425. https://doi.org/10.3390/antibiotics12030425

AMA Style

Daly AK. Relevance of Pharmacogenomics to the Safe Use of Antimicrobials. Antibiotics. 2023; 12(3):425. https://doi.org/10.3390/antibiotics12030425

Chicago/Turabian Style

Daly, Ann K. 2023. "Relevance of Pharmacogenomics to the Safe Use of Antimicrobials" Antibiotics 12, no. 3: 425. https://doi.org/10.3390/antibiotics12030425

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop