Skip to main content
Experimental Biology and Medicine logoLink to Experimental Biology and Medicine
. 2017 Sep 26;243(3):291–299. doi: 10.1177/1535370217733425

Biomarkers of adverse drug reactions

Daniel F Carr 1,2, Munir Pirmohamed 1,2,
PMCID: PMC5813863  PMID: 28950720

Short abstract

Adverse drug reactions can be caused by a wide range of therapeutics. Adverse drug reactions affect many bodily organ systems and vary widely in severity. Milder adverse drug reactions often resolve quickly following withdrawal of the casual drug or sometimes after dose reduction. Some adverse drug reactions are severe and lead to significant organ/tissue injury which can be fatal. Adverse drug reactions also represent a financial burden to both healthcare providers and the pharmaceutical industry. Thus, a number of stakeholders would benefit from development of new, robust biomarkers for the prediction, diagnosis, and prognostication of adverse drug reactions. There has been significant recent progress in identifying predictive genomic biomarkers with the potential to be used in clinical settings to reduce the burden of adverse drug reactions. These have included biomarkers that can be used to alter drug dose (for example, Thiopurine methyltransferase (TPMT) and azathioprine dose) and drug choice. The latter have in particular included human leukocyte antigen (HLA) biomarkers which identify susceptibility to immune-mediated injuries to major organs such as skin, liver, and bone marrow from a variety of drugs. This review covers both the current state of the art with regard to genomic adverse drug reaction biomarkers. We also review circulating biomarkers that have the potential to be used for both diagnosis and prognosis, and have the added advantage of providing mechanistic information. In the future, we will not be relying on single biomarkers (genomic/non-genomic), but on multiple biomarker panels, integrated through the application of different omics technologies, which will provide information on predisposition, early diagnosis, prognosis, and mechanisms.

Impact statement

• Genetic and circulating biomarkers present significant opportunities to personalize patient therapy to minimize the risk of adverse drug reactions. ADRs are a significant heath issue and represent a significant burden to patients, healthcare providers, and the pharmaceutical industry.

• This review details the current state of the art in biomarkers of ADRs (both genetic and circulating). There is still significant variability in patient response which cannot be explained by current knowledge of genetic risk factors for ADRs; however, we discussed how specific advances in genomics have the potential to yield better and more predictive models.

• Many current clinically utilized circulating biomarkers of tissue injury are valid biomarkers for a number of ADRs. However, they often give little insight into the specific cell or tissue subtype which may be affected. Emerging circulating biomarkers with potential to provide greater information on the etiology/pathophysiology of ADRs are described.

Keywords: Adverse drug reactions, drug safety, biomarkers, pharmacogenomics

Introduction

An adverse drug reaction (ADR) is defined by the World Health Organization as “A response to a drug which is noxious and unintended, and which occurs at doses normally used in man for the prophylaxis, diagnosis, or therapy of disease, or for the modifications of physiological function”.1 The majority of ADRs fall into the two broad categories:

  • Type A – Reactions which are predictable from the drug’s known pharmacology and typically result from an augmented on-target pharmacological response when given at a usual therapeutic dose.

  • Type B – Reactions which are also termed idiosyncratic and are not predictable from the known pharmacological actions of the drug. These are typically rare and safety signals are often not detected prior to marketing.

As demonstrated in Table 1, ADRs can affect a significant number of organ systems in the body and can range in severity from mild reactions (e.g. skin rash or mild liver enzyme elevation) which resolve upon withdrawal of the causal drug to severe, life-threatening reactions including skin blistering reactions (Stevens-Johnson syndrome/ toxic epidermal necrolysis (SJS/TEN)) and fulminant liver failure.

Table 1.

Examples of immunogenetic biomarkers of ADRs by organ system.

System ADR Causal drug Indication Associated genetic variant References
Skin Hypersensitivity (SJS/TEN/DRESS/maculopapular exanthem) Carbamazepine Phenytoin Epilepsy HLA-B*15:02 (Han Chinese) HLA-A*31:01 (Caucasian/Japanese) HLA-B*15:02 (Han Chinese) 2,3,4,5
Allopurinol Gout HLA-B*58:01 6
Nevirapine HIV HLA-C*04:01HLA-B*35:05 (Thai)HLA-DRB1*01:01 7,7,7
Abacavir HIV HLA-B*57:01 8
Gastrointestinal Hepatotoxicity Flucloxacillin Gram +ve bacterial infection HLA-B*57:01 9
Co-amoxiclav Bacterial infection HLA-A*02:01, DRB1*15:01-DQB1*06:02 10
Nevirapine HIV HLA-DRB1:01:01 11
Minocycline Bacterial infection HLA-B*35:02 12
Lapatinib Breast cancer HLA-DQA1*02:01/HLA-DRB1*07:01 13,14
Pancreatitis Azathioprine Rheumatoid arthritis, Crohn’s disease HLA-DRB1, HLA-DQB1 15
Renal Nephrotoxicity 5-aminosalicylic acid Inflammatory bowel disease HLA-DRB1*03:01 16
Hematological Agranulocytosis Clozapine Schizophrenia HLA-B/HLA-DQB1 17
Sulfasalazine Inflammatory joint/bowel disease HLA-B*08:01, HLA-A*31:01 18
Antithyroid drugs Hyperthyroidism HLA-B*27:05 (Caucasian) HLA-B*38:02, HLA-DRB1*08:03 (Han Chinese) DRB1*08032 (Japanese) 19,20,21
Musculoskeletal Necrotizing autoimmune myopathy Statins Hypercholesterolemia HLA-DRB1*11:01 22

ADR: adverse drug reaction; DRESS: drug reaction with eosinophilia and systemic symptoms; SJS: Stevens-Johnson syndrome; TEN: toxic epidermal necrolysis.

ADRs account for between 6.5% (hospital admissions) and 25% (primary care) of attendances for medical treatment, representing a significant burden on healthcare services.23 This translates to a cost in excess of £1 billion every year in the UK (with equivalent figures in other countries).24 Indeed, in the US, the figure has been estimated to be as high as $30.1 billion.25 The financial burden on pharmaceutical research and development is also significant; between 1990 and 2013, 43 drugs were withdrawn from market due to severe ADRs.26

From both a patient and healthcare perspective, there are potential benefits in developing biomarkers for identifying individuals predisposed to ADRs prior to initiation of therapy. Biomarkers can also have a prognostic role in determining the likelihood of recovery postreaction and potentially in the development of severe sequelae (e.g. ophthalmic complications postreaction in SJS/TEN survivors) (Figure 1). This review covers the current knowledge base of biomarkers of ADRs (both genomic and non-genomic) and discusses potential advances and directions for the development and implementation of new biomarkers.

Figure 1.

Figure 1.

Schematic of a typical delayed onset idiosyncratic ADR and indicative points at which theoretical predictive, prognostic, and diagnostic biomarkers could be used for informing patient treatment decisions and care pathways. (A color version of this figure is available in the online journal.)

ADR: adverse drug reaction.

Genomic biomarkers

Pharmacodynamic/pharmacokinetic-related genetic biomarkers

Polymorphisms in genes encoding drug metabolizing enzymes or drug transporter proteins have been associated with a number of type A ADRs (Table 2). Indeed, clinical implementation guidelines exist for a number of drugs where pharmacokinetic genetic variation can be critical in determining the risk of an ADR. These include (but are not limited to) TPMT and azathioprine/mercaptopurine-induced bone marrow toxicity;27,28 CYP2D6 and codeine (morphine)-related respiratory depression;29 and SLCO1B1 and simvastatin-induced myotoxicity.30 An interesting recent study has shown that a non-synonymous variant in the SLCO2A1 gene, which encodes a prostaglandin transporter, is associated with thiazide-induced hyponatraemia.31

Table 2.

Examples of pharmacokinetic/pharmacodynamic genetic biomarkers of ADRs by organ system.

System ADR Causal drug Indication Associated genetic variation References
Skin SJS/TEN Phenytoin Epilepsy CYP2C9*2/*3 5
Gastrointestinal Hepatotoxicity Isoniazid Tuberculosis NAT2 slow acetylator 32
Hyperbilirubinemia Atazanavir HIV UGT1A1*28 33
CNS Respiratory depression Codeine (morphine) Analgesia CYP2D6 Ultrarapid metabolizer 29,34
Renal Nephrotoxicity Tacrolimus Immunosuppressant CYP3A5*3/*6/*7 35
Hematological Bone marrow suppression Azathioprine Rheumatoid arthritis, Crohn’s disease TPMT*2/*3A/*3C/*4 27,28
Neutropenia Irinotecan Colorectal cancer UGT1A1*28 36
Cardiovascular Bleeding Warfarin Anticoagulant CYP2C9*2/*3, VKORC1 37
Myocardial infarction, stroke, bleeding Clopidogrel Antiplatelet CYP2C19*2/*3/*17 38,39
Musculoskeletal Myopathy Simvastatin Hypercholesterolemia SLCO1B1*5 30

ADR: adverse drug reaction.

In addition to PK-related pharmacogenomic biomarkers, some type A ADRs are also associated with polymorphisms in genes encoding pharmacodynamic targets. Perhaps the most prominent example of this is the anticoagulant warfarin which is indicated in the treatment of atrial fibrillation, deep-vein thrombosis, and pulmonary embolism. The CYP2C9*2 and CYP2C9*3 polymorphisms in the gene-encoding P4502C9, which is responsible for the metabolism of the active S enantiomer of warfarin, are a key determinant of daily dose requirement in patients. Additionally, a promoter region polymorphism (c.1639A > G) which reduces the hepatic expression of vitamin K epoxide reductase (VKORC1), the pharmacological target of warfarin, is also strongly associated with warfarin dose requirement. The combination of genetic polymorphisms in the CYP2C9 and VKORC1 genes in fact accounts for almost 50% of the variation in daily dose requirement.40 The translation of these findings into clinical practice has been challenging,41 but a randomized controlled trial in Europe has shown that genotype-guided dosing was superior to the current standard of care in improving overall anticoagulation control (including time in therapeutic international normalized ratio (INR) range and reducing overshoot to an INR > 4).37

Immunogenetic biomarkers

Many type B (idiosyncratic) ADRs, including SJS/TEN and drug-induced liver injury (DILI), have an immune pathogenesis. This is consistent with the fact that very strong genetic associations between such reactions and individual HLA genetic loci within the major histocompatibility complex region on chromosome 6 have been reported (Table 2). Indeed, for two of these associations (HLA-B*57:01 and abacavir hypersensitivity; and HLA-B*15:02 and carbamazepine-induced SJS/TEN in some SE Asian populations), preprescription genotyping is recommended by most regulatory agencies including the FDA. This has been of clinical value since the incidence of these reactions has shown a marked decrease where the genetic test has been consistently implemented.42,43

An increasingly important issue to consider in the implementation of pharmacogenetic testing is the cost-effectiveness – this is vital to persuade the healthcare providers to pay for testing. There is now an increasing number of such studies being performed; this requires the collection of different types of data (including quality of life) which can then be incorporated into health economic models. Examples of tests which have been demonstrated to be cost-effective include HLA-B*57:01 for abacavir hypersensitivity,44 HLA-B*15:02 and HLA-A*31:01 for carbamazepine hypersensitivity,45,46 and TPMT for azathioprine.47 In other situations, even though a test result may be highly significant, the rarity of the ADR may make genetic testing cost-ineffective. For instance, the antibiotic flucloxacillin, which is widely used to treat Gram-positive bacterial infections, can lead to hepatotoxicity, which shows a strong association with the HLA-B*57:01 allelotype.9 However, the incidence of flucloxacillin-induced liver injury is approximately 8.5 cases per 100,000 individuals,48 and it has been estimated that in order to prevent one case of hepatotoxicity, a total of 13,513 individuals would need to be tested.

Barriers to the clinical implementation of genomic biomarkers of ADRs

Very few genomic markers have been translated into clinical practice as pre-emptive screening tools to identify individuals at risk of ADRs. Although there is significant evidence for many genetic associations with ADRs (Table 1), lack of replication remains a key factor in hampering translation of genomic biomarkers. Two key reasons for the failure of genetic associations to progress beyond discovery stage are:

  1. Heterogeneity of phenotype definition between independent studies: Many promising, biologically plausible genetic associations of ADRs fail replicate in part due to disparities in the clinical definition of the phenotype. In order to overcome this, in recent years a number of projects have been undertaken to standardize ADR phenotypes including cutaneous hypersensitivity,49 liver injury,50 Torsade de Pointes,51 and statin-induced myopathy.52 Such standardization will help not only for replication but also in undertaking meta-analyses of different studies.

  2. Statistically underpowered studies: Many severe ADRs are rare and by virtue of this, identification of patients and recruitment to pharmacogenomic studies is challenging, requiring international collaborative initiatives. Because of this, many ADR studies tend to be small and statistically underpowered, particularly for replication purposes, where the odds ratio in the original discovery set may be inflated. Despite the smaller numbers of patients available, it is important to note that pharmacogenetic association traits, on average, have significantly larger effect sizes than complex disease associations,53 and therefore may find significant associations despite relatively small (compared with complex disease studies) sample sizes.

It is perhaps also important to note that genomic biomarkers can also be used for purposes other than prediction. Our recent paper has outlined the case as to how genomic biomarkers can be used for diagnosis, selection of patients, pre-emptive genotyping, and for understanding mechanisms.41 Pre-emptive genotyping is now being tested in several countries, and data on clinical outcomes are keenly awaited.54 Indeed, as whole genome sequencing becomes more widespread, a wider perspective on how genetic tests can be used in practice will help to improve the benefit–risk ratio of medicine and in implementing precision medicine in its broadest sense.

Genomic biomarkers of ADRs: Opportunities

Common genetic variants represent the “low hanging fruit” as predictive risk factors for ADRs but it is clear that there is still a significant degree of interindividual variability in drug response that cannot be accounted for by our existing knowledge of genetic and non-genetic risk factors. Efforts in a number of research areas have the potential to shed light on this unexplained variability.

Rare variants

With the increased availability of sequencing technologies has come the ability to type patients for rare genetic variants (minor allele frequency <1%) and assess the role they may play in predicting ADRs. Warfarin is an example where pharmacogenetic (CYP2C9 and VKORC1) and non-genetic determinants account for approximately 60% of the dose requirement,55 with 40% of the variability unexplained. It is plausible that rare variants in both known and as yet unknown gene loci may play a role in warfarin response, particularly in individuals with extreme phenotypes, i.e. requirement of either very low or very high daily warfarin doses. Furthermore, twin studies of the pharmacokinetic variability of torsemide and metoprolol56 have shown that only around 40% of the genetic variability can be explained by known genetic polymorphisms. Recent work by Kozyra et al.57 has indicated that between 30% and 40% of the variability in pharmacogenes is due to rare variants.

Metagenomic risk factors for ADRs

There is growing interest in the role of the human microbiome in predicting drug response.58 More than 50 drugs are known to be metabolized by the microbiome (by hydrolysis or reduction in the majority of cases).59 Examples include soruvidine, lovastatin, and paracetamol. Theoretically, this could alter their disposition in the host and potentially lead to lack of efficacy or predisposition to adverse effects. Whether the microbiome, in the gut and other locations, has a significant role in explaining the missing variability with different drugs requires further study. For instance, with the antidiabetic agent metformin, there is evidence that modulation of the gut microbiome is responsible, at least partly, for its therapeutic effects.60 Whether disturbance in the gut microbiome is also responsible for its common adverse effect of GI intolerance requires further study.

Multiomics/systems biology approaches

Genomic biomarkers clearly still have much to offer for predicting ADRs. However, alternative but complementary omics technologies need to be considered both in insolation but also within an integrated multiomic/systems biology framework.61 This will allow the identification of complexed multifaceted traits which predispose to ADRs but also uncover novel mechanistic biological pathways with the potential to yield novel biomarkers of both a genetic and non-genetic nature.

Circulating protein and nucleic acid biomarkers

Biomarkers of generalized tissue injury applicable to ADRs

A plethora of “traditional” circulating protein biomarkers which correlate with specific tissue injury, regardless of etiology, can be used for diagnosis of ADRs, and in some cases, for determining prognosis (Table 3). Typical examples include plasma alanine transaminase (ALT) and aspartate transaminase (AST) for liver injury and serum creatinine for kidney injury. While these markers of tissue injury have been used for many years, they can have limitations in terms of sensitivity (they become elevated only when a significant proportion of the organ is damaged) and specificity (they can be produced by multiple organ or multiple toxic insults). Additionally, in the context of ADRs, they are also limited in informing as to the specific mechanism of toxicity or the affected cell type within an organ system. A typical example is serum creatine kinase (CK) where a level of >4×upper limit of normal (ULN) has been used for diagnosing muscle toxicity associated with the use of statins.52 CK elevation can occur due to a number of commonly occurring events including strenuous exercise62 and trauma.63 In addition, other unrelated comorbidities such as myocardial infarction can also cause CK elevation.64 Broadly speaking, CK elevation thus offers low specificity for diagnosing statin myopathy and gives little indication of the specific mechanisms of statin myopathy.

Table 3.

Clinically utilized and putative biomarkers of ADRs.

System ADR Biomarker Tissue/fluid Indicative of Currently utilized for diagnosis/prognosis References
Skin Hypersensitivity (SJS/TEN/DRESS/rash) C-reactive protein Serum Non-specific inflammation Y 65
Granulysin Blister fluid N 66
Interleukin-15 Serum Innate immune response N 67
HMGB1 Serum Sterile tissue injury/innate immune response N 68
miR-124 Serum/skin Unknown N 69
miR-18a-5p Serum/skin Unknown N 70
Gastrointestinal Hepatotoxicity Alanine transaminase (ALT)/aspartate transaminase (AST) Serum Hepatocellular injury Y 71
Alkaline phosphatase (ASP) Serum Hepatic bile canaliculi cell injury Y 71
Bilirubin Serum Hepatic injury/biliary obstruction but also extrahepatic disorder Y 72
HMGB1 Serum Sterile tissue injury/innate immune response N 73
miR-122 Serum Hepatocyte-specific cell injury N 24,74
Cytokeratin-18 Serum Apoptotic/necrotic hepatic cell death N 75
Lower GI toxicity C-reactive protein Serum Non-specific inflammation Y 76
Lactoferrin Fecal Inflammation Y 77
Calprotectin Fecal Inflammation Y 78
Renal Creatinine Serum Glomerular filtration rate and proximal tubular secretion Y 79
Cystatin-C Serum Glomerular filtration rate and proximal tubular secretion Y 80
NGAL Urine Proximal and distal tubule secretion N 81
KIM-1 Urine Proximal Tubule N 90
Musculoskeletal Myopathy/rhabdomyolysis Creatine kinase Serum Skeletal muscle injury but also cardiac muscle injury Y 52

GI: gastrointestinal; HMGB1: High Mobility Group Box-1; N: no; Y: yes.N-GAL: Neutrophil gelatinase-associated lipocalin; KIM-1: kidney injury molecule 1.

By contrast, recent advances in biomarkers for the detection of DILI serve as a paradigm for how novel markers can have significant advantages over traditional markers for diagnosing and understanding the etiology of an ADR. Of particular promise is serum miR-122, which has been shown to be a highly specific marker for acute hepatocyte injury in paracetamol overdose82 and more sensitive than traditional liver function tests for early toxicity detection. However, further work is required to determine the utility of miR-122 as a diagnostic/prognostic marker of late-onset idiosyncratic DILI. Evidence for other miR as diagnostic tools for ADRs is currently low, although a number of other putative miR biomarkers have been reported, including miR-12469 and miR-18a-5p70 for SJS/TEN. Understanding how and when miRs become elevated also provides an opportunity to gain insight into the specific cell source and pathogenesis of the injury. This may also help in the future in drug development during both preclinical toxicology testing and early phase human trials.

Mechanistic biomarkers

The field of kidney injury research is perhaps the best example of how a new generation of biomarkers has the potential to provide not only early sensitive detection of renal toxicity but also provide information as to the specific site of injury within the nephron.83 The additional information could have tremendous potential benefits to both drug development and healthcare professionals. This sort of mechanistic approach to biomarker discovery could certainly be applied to other tissues/organs commonly affected by ADRs where toxicity may be specific to particular cell types such as the liver and gastrointestinal (GI) tract.

High Mobility Group Box-1 (HMGB1) is a biomarker which has significant potential as a prognostic mechanistic biomarker for ADRs. HMGB1 is an example of a Damage Associated Molecular Pattern molecule, which is critical in linking cell death to inflammation and in the progression of disease. HMGB1 sits at the intersection between infectious and sterile inflammation. HMGB1 is actively released in an acetylated form from activated immune cells and passively released in the non-acetylated form during necrotic cell death.84 Furthermore, the HMGB1 molecule can exist in three redox states, each of which infers a different physiological function related to the innate immune response. Disulfide HMGB1 has been demonstrated to engage with MD2 as part of the toll-like receptor 4 (TLR4) complex on monocytes in order to elicit cytokine induction85 while the fully reduced isoform is thought to interact with CXCL12 to engage with CXCR4 to induce chemotaxis.86 Work undertaken in patients with liver damage who have overdosed on acetaminophen has shown that early elevation of HMGB1, in patients with normal ALTs, was able to predict more severe forms of liver injury later during the course of the overdose.82

Total serum HMGB1 is also elevated in a number of immune-mediated type B ADRs including DRESS,87 SJS/TEN,68 and in principle DILI.88 HMGB1 isoforms could theoretically be utilized for early distinction of hypersensitivity reactions leading to significant tissue injury (e.g. SJS/TEN) as opposed to milder phenotypes (maculopapular exanthem), but more work needs to be done on this.

Identifying and implementing diagnostic biomarkers which can predict the onset of ADRs (Figure 1) is clearly of benefit to both patients and healthcare professionals. However, in some examples, such as SJS/TEN, severe long-term sequelae can occur a significant time after the acute reaction and subsequent drug withdrawal.89 Although at present no examples exist, the development of prognostic biomarkers for prediction of these sequelae, which can include vision loss, has the potential to provide tools to improve treatment decisions and clinical care pathways.

Conclusions

While much progress has been made in identifying predictive genomic biomarkers of ADRs, only a small number have been translated to clinical practice. With the increasing use of sequencing technologies, greater focus is being placed on the role of rare variants in ADR predisposition. Additionally, other omics technologies are likely to yield significant biomarkers for ADRs in the future. A new generation of circulating biomarkers of ADRs, typified by miR-122 and HMGB1, have great potential as highly specific and sensitive diagnostic and/or prognostic markers. The example of renal toxicity highlights the potential of using panels of biomarkers as indicators of specific cellular or tissue sites of injury and provide greater mechanistic understanding of ADRs. Application of this mechanistic approach to other target organs of ADRs (liver, skin, GI tract) could yield substantial benefits in producing robust biomarkers for the benefit of patient care and future drug development pipelines. It is likely that in the future we will not be relying on single biomarkers (genomic/non-genomic), but on multiple biomarker panels, integrated through the application of different omics technologies, which will provide information on predisposition, early diagnosis, prognosis, and mechanisms. This is however likely to introduce huge complexity in terms of the evidence that will be required for regulation and clinical implementation, and in the interpretation of these complex tests for clinical care of patients. This will need to be aligned to studies of cost-effectiveness and inclusion in clinical guidelines, as well as education and training of new and existing healthcare professionals.

Authors’ contributions

DC and MP contributed equally to the synthesis, writing, and production of this review.

Declaration of conflicting interests

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding

We would like to thank the MRC Centre for Drug Safety Science (MR/L006758/1) for support. MP is NIHR Emeritus Senior Investigator.

References

  • 1. International drug monitoring: the role of national centres. Report of a WHO meeting. World Health Organization technical report series 1972;498:1–25. [PubMed]
  • 2.Chung WH, Hung SI, Hong HS, Hsih MS, Yang LC, Ho HC, Wu JY, Chen YT. Medical genetics: a marker for Stevens–Johnson syndrome. Nature 2004; 428:486. [DOI] [PubMed] [Google Scholar]
  • 3.McCormack M, Alfirevic A, Bourgeois S, Farrell JJ, Kasperaviciute D, Carrington M, Sills GJ, Marson T, Jia X, de Bakker PI, Chinthapalli K, Molokhia M, Johnson MR, O'Connor GD, Chaila E, Alhusaini S, Shianna KV, Radtke RA, Heinzen EL, Walley N, Pandolfo M, Pichler W, Park BK, Depondt C, Sisodiya SM, Goldstein DB, Deloukas P, Delanty N, Cavalleri GL, Pirmohamed M. HLA-A*3101 and carbamazepine-induced hypersensitivity reactions in Europeans. N Engl J Med 2011; 364:1134–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ozeki T, Mushiroda T, Yowang A, Takahashi A, Kubo M, Shirakata Y, Ikezawa Z, Iijima M, Shiohara T, Hashimoto K, Kamatani N, Nakamura Y. Genome-wide association study identifies HLA-A*3101 allele as a genetic risk factor for carbamazepine-induced cutaneous adverse drug reactions in Japanese population. Hum Mol Genet 2011; 20:1034–41 [DOI] [PubMed] [Google Scholar]
  • 5.Caudle KE, Rettie AE, Whirl-Carrillo M, Smith LH, Mintzer S, Lee MT, Klein TE, Callaghan JT; Clinical Pharmacogenetics Implementation C. Clinical Pharmacogenetics Implementation Consortium guidelines for CYP2C9 and HLA-B genotypes and phenytoin dosing. Clin Pharmacol Ther 2014; 96:542–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Saito Y, Stamp LK, Caudle KE, Hershfield MS, McDonagh EM, Callaghan JT, Tassaneeyakul W, Mushiroda T, Kamatani N, Goldspiel BR, Phillips EJ, Klein TE, Lee MT; Clinical Pharmacogenetics Implementation C. Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for human leukocyte antigen B (HLA-B) genotype and allopurinol dosing: 2015 update. Clin Pharmacol Ther 2016; 99:36–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Cornejo Castro EM, Carr DF, Jorgensen AL, Alfirevic A, Pirmohamed M. HLA-allelotype associations with nevirapine-induced hypersensitivity reactions and hepatotoxicity: a systematic review of the literature and meta-analysis. Pharmacogenet Genomics 2015; 25:186–98 [DOI] [PubMed] [Google Scholar]
  • 8.Martin MA, Hoffman JM, Freimuth RR, Klein TE, Dong BJ, Pirmohamed M, Hicks JK, Wilkinson MR, Haas DW, Kroetz DL; Clinical Pharmacogenetics Implementation C. Clinical Pharmacogenetics Implementation Consortium guidelines for HLA-B genotype and abacavir dosing: 2014 update. Clin Pharmacol Ther 2014; 95:499–500 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Daly AK, Donaldson PT, Bhatnagar P, Shen Y, Pe'er I, Floratos A, Daly MJ, Goldstein DB, John S, Nelson MR, Graham J, Park BK, Dillon JF, Bernal W, Cordell HJ, Pirmohamed M, Aithal GP, Day CP, Study D; International SAEC. HLA-B*5701 genotype is a major determinant of drug-induced liver injury due to flucloxacillin. Nat Genet 2009; 41:816–9 [DOI] [PubMed] [Google Scholar]
  • 10.Lucena MI, Molokhia M, Shen Y, Urban TJ, Aithal GP, Andrade RJ, Day CP, Ruiz-Cabello F, Donaldson PT, Stephens C, Pirmohamed M, Romero-Gomez M, Navarro JM, Fontana RJ, Miller M, Groome M, Bondon-Guitton E, Conforti A, Stricker BH, Carvajal A, Ibanez L, Yue QY, Eichelbaum M, Floratos A, Pe'er I, Daly MJ, Goldstein DB, Dillon JF, Nelson MR, Watkins PB, Daly AK. Susceptibility to amoxicillin-clavulanate-induced liver injury is influenced by multiple HLA class I and II alleles. Gastroenterology 2011; 141:338–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Martin AM, Nolan D, James I, Cameron P, Keller J, Moore C, Phillips E, Christiansen FT, Mallal S. Predisposition to nevirapine hypersensitivity associated with HLA-DRB1*0101 and abrogated by low CD4 T-cell counts. AIDS 2005; 19:97–9 [DOI] [PubMed] [Google Scholar]
  • 12.Urban TJ, Nicoletti P, Chalasani N, Serrano J, Stolz A, Daly AK, Aithal GP, Dillon J, Navarro V, Odin J, Barnhart H, Ostrov D, Long N, Cirulli ET, Watkins PB, Fontana RJ; Drug-Induced Liver Injury N, Pharmacogenetics of Drug-Induced Liver Injury G, International Serious Adverse Events C. Minocycline hepatotoxicity: clinical characterization and identification of HLA-B *35:02 as a risk factor. J Hepatol 2017; 67:137–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Parham LR, Briley LP, Li L, Shen J, Newcombe PJ, King KS, Slater AJ, Dilthey A, Iqbal Z, McVean G, Cox CJ, Nelson MR, Spraggs CF. Comprehensive genome-wide evaluation of lapatinib-induced liver injury yields a single genetic signal centered on known risk allele HLA-DRB1*07:01. Pharmacogenomics J 2016; 16:180–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Spraggs CF, Budde LR, Briley LP, Bing N, Cox CJ, King KS, Whittaker JC, Mooser VE, Preston AJ, Stein SH, Cardon LR. HLA-DQA1*02:01 is a major risk factor for lapatinib-induced hepatotoxicity in women with advanced breast cancer. J Clin Oncol 2011; 29:667–73 [DOI] [PubMed] [Google Scholar]
  • 15.Heap GA, Weedon MN, Bewshea CM, Singh A, Chen M, Satchwell JB, Vivian JP, So K, Dubois PC, Andrews JM, Annese V, Bampton P, Barnardo M, Bell S, Cole A, Connor SJ, Creed T, Cummings FR, D'Amato M, Daneshmend TK, Fedorak RN, Florin TH, Gaya DR, Greig E, Halfvarson J, Hart A, Irving PM, Jones G, Karban A, Lawrance IC, Lee JC, Lees C, Lev-Tzion R, Lindsay JO, Mansfield J, Mawdsley J, Mazhar Z, Parkes M, Parnell K, Orchard TR, Radford-Smith G, Russell RK, Reffitt D, Satsangi J, Silverberg MS, Sturniolo GC, Tremelling M, Tsianos EV, van Heel DA, Walsh A, Watermeyer G, Weersma RK, Zeissig S, Rossjohn J, Holden AL; International Serious Adverse Events C, Group IBDPS, Ahmad T. HLA-DQA1-HLA-DRB1 variants confer susceptibility to pancreatitis induced by thiopurine immunosuppressants. Nat Genet 2014; 46:1131–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Heap GA, So K, Weedon M, Edney N, Bewshea C, Singh A, Annese V, Beckly J, Buurman D, Chaudhary R. Clinical features and HLA association of 5-aminosalicylate (5-ASA)-induced nephrotoxicity in inflammatory bowel disease. J Crohns Colitis 2016; 10:149–58 [DOI] [PubMed] [Google Scholar]
  • 17.Goldstein JI, Jarskog LF, Hilliard C, Alfirevic A, Duncan L, Fourches D, Huang H, Lek M, Neale BM, Ripke S, Shianna K, Szatkiewicz JP, Tropsha A, van den Oord EJ, Cascorbi I, Dettling M, Gazit E, Goff DC, Holden AL, Kelly DL, Malhotra AK, Nielsen J, Pirmohamed M, Rujescu D, Werge T, Levy DL, Josiassen RC, Kennedy JL, Lieberman JA, Daly MJ, Sullivan PF. Clozapine-induced agranulocytosis is associated with rare HLA-DQB1 and HLA-B alleles. Nat Commun 2014; 5:4757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wadelius M, Eriksson N, Kreutz R, Bondon-Guitton E, Ibanez L, Carvajal A, Lucena MI, Sancho Ponce E, Molokhia M, Martin J, Axelsson T, Kohnke H, Yue QY, Magnusson PKE, Bengtsson M, Hallberg P, EuDac. Sulfasalazine-induced agranulocytosis is associated with the human leukocyte antigen locus. Clin Pharmacol Ther 2017. Epub ahead of print 1 August. doi: 10.1002/cpt.805. [DOI] [PMC free article] [PubMed]
  • 19.Hallberg P, Eriksson N, Ibanez L, Bondon-Guitton E, Kreutz R, Carvajal A, Lucena MI, Ponce ES, Molokhia M, Martin J, Axelsson T, Yue QY, Magnusson PK, Wadelius M; Eu DACc. Genetic variants associated with antithyroid drug-induced agranulocytosis: a genome-wide association study in a European population. Lancet Diabetes Endocrinol 2016; 4:507–16 [DOI] [PubMed] [Google Scholar]
  • 20.Chen PL, Shih SR, Wang PW, Lin YC, Chu CC, Lin JH, Chen SC, Chang CC, Huang TS, Tsai KS, Tseng FY, Wang CY, Lu JY, Chiu WY, Chang CC, Chen YH, Chen YT, Fann CS, Yang WS, Chang TC. Genetic determinants of antithyroid drug-induced agranulocytosis by human leukocyte antigen genotyping and genome-wide association study. Nat Commun 2015; 6:7633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Tamai H, Sudo T, Kimura A, Mukuta T, Matsubayashi S, Kuma K, Nagataki S, Sasazuki T. Association between the DRB1*08032 histocompatibility antigen and methimazole-induced agranulocytosis in Japanese patients with Graves disease. Ann Intern Med 1996; 124:490–4 [DOI] [PubMed] [Google Scholar]
  • 22.Mammen AL, Gaudet D, Brisson D, Christopher-Stine L, Lloyd TE, Leffell MS, Zachary AA. Increased frequency of DRB1*11:01 in anti-hydroxymethylglutaryl-coenzyme A reductase-associated autoimmune myopathy. Arthritis Care Res 2012; 64:1233–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Pillans PI. Clinical perspectives in drug safety and adverse drug reactions. Expert Rev Clin Pharmacol 2008; 1:695–705 [DOI] [PubMed] [Google Scholar]
  • 24.Pirmohamed M, James S, Meakin S, Green C, Scott AK, Walley TJ, Farrar K, Park BK, Breckenridge AM. Adverse drug reactions as cause of admission to hospital: prospective analysis of 18 820 patients. BMJ 2004; 329:15–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sultana J, Cutroneo P, Trifiro G. Clinical and economic burden of adverse drug reactions. J Pharmacol Pharmacother 2013; 4:S73–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wei CY, Lee MT, Chen YT. Pharmacogenomics of adverse drug reactions: implementing personalized medicine. Hum Mol Genet 2012; 21:R58–65 [DOI] [PubMed] [Google Scholar]
  • 27.Relling MV, Gardner EE, Sandborn WJ, Schmiegelow K, Pui CH, Yee SW, Stein CM, Carrillo M, Evans WE, Hicks JK, Schwab M, Klein TE; Clinical Pharmacogenetics Implementation C. Clinical Pharmacogenetics Implementation Consortium guidelines for thiopurine methyltransferase genotype and thiopurine dosing: 2013 update. Clin Pharmacol Ther 2013; 93:324–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Relling MV, Gardner EE, Sandborn WJ, Schmiegelow K, Pui CH, Yee SW, Stein CM, Carrillo M, Evans WE, Klein TE; Clinical Pharmacogenetics Implementation C. Clinical Pharmacogenetics Implementation Consortium guidelines for thiopurine methyltransferase genotype and thiopurine dosing. Clin Pharmacol Ther 2011; 89:387–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Crews KR, Gaedigk A, Dunnenberger HM, Klein TE, Shen DD, Callaghan JT, Kharasch ED, Skaar TC. Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for codeine therapy in the context of cytochrome P450 2D6 (CYP2D6) genotype. Clin Pharmacol Ther 2012; 91:321–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ramsey LB, Johnson SG, Caudle KE, Haidar CE, Voora D, Wilke RA, Maxwell WD, McLeod HL, Krauss RM, Roden DM, Feng Q, Cooper-DeHoff RM, Gong L, Klein TE, Wadelius M, Niemi M. The Clinical Pharmacogenetics Implementation Consortium guideline for SLCO1B1 and simvastatin-induced myopathy: 2014 update. Clin Pharmacol Ther 2014; 96:423–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ware JS, Wain LV, Channavajjhala SK, Jackson VE, Edwards E, Lu R, Siew K, Jia W, Shrine N, Kinnear S, Jalland M, Henry AP, Clayton J, O'Shaughnessy KM, Tobin MD, Schuster V, Cook S, Hall IP, Glover M. Phenotypic and pharmacogenetic evaluation of patients with thiazide-induced hyponatremia. J Clin Invest 2017; 127:3367–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Huang YS, Chern HD, Su WJ, Wu JC, Lai SL, Yang SY, Chang FY, Lee SD. Polymorphism of the N-acetyltransferase 2 gene as a susceptibility risk factor for antituberculosis drug-induced hepatitis. Hepatology 2002; 35:883–9 [DOI] [PubMed] [Google Scholar]
  • 33.Gammal RS, Court MH, Haidar CE, Iwuchukwu OF, Gaur AH, Alvarellos M, Guillemette C, Lennox JL, Whirl-Carrillo M, Brummel SS, Ratain MJ, Klein TE, Schackman BR, Caudle KE, Haas DW; Clinical Pharmacogenetics Implementation C. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for UGT1A1 and atazanavir prescribing. Clin Pharmacol Ther 2016; 99:363–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Gasche Y, Daali Y, Fathi M, Chiappe A, Cottini S, Dayer P, Desmeules J. Codeine intoxication associated with ultrarapid CYP2D6 metabolism. N Engl J Med 2004; 351:2827–31 [DOI] [PubMed] [Google Scholar]
  • 35.Birdwell KA, Decker B, Barbarino JM, Peterson JF, Stein CM, Sadee W, Wang D, Vinks AA, He Y, Swen JJ, Leeder JS, van Schaik R, Thummel KE, Klein TE, Caudle KE, MacPhee IA. Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for CYP3A5 genotype and tacrolimus dosing. Clin Pharmacol Ther 2015; 98:19–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Etienne-Grimaldi MC, Boyer JC, Thomas F, Quaranta S, Picard N, Loriot MA, Narjoz C, Poncet D, Gagnieu MC, Ged C, Broly F, Le Morvan V, Bouquie R, Gaub MP, Philibert L, Ghiringhelli F, Le Guellec C; Collective work by Groupe de Pharmacologie Clinique O, French Reseau National de Pharmacogenetique H. UGT1A1 genotype and irinotecan therapy: general review and implementation in routine practice. Fundam Clin Pharmacol 2015; 29:219–37 [DOI] [PubMed] [Google Scholar]
  • 37.Johnson JA, Caudle KE, Gong L, Whirl-Carrillo M, Stein CM, Scott SA, Lee MT, Gage BF, Kimmel SE, Perera MA, Anderson JL, Pirmohamed M, Klein TE, Limdi NA, Cavallari LH, Wadelius M. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for pharmacogenetics-guided warfarin dosing: 2017 Update. Clin Pharmacol Ther 2017; 102:397–404 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Scott SA, Sangkuhl K, Gardner EE, Stein CM, Hulot JS, Johnson JA, Roden DM, Klein TE, Shuldiner AR; Clinical Pharmacogenetics Implementation C. Clinical Pharmacogenetics Implementation Consortium guidelines for cytochrome P450-2C19 (CYP2C19) genotype and clopidogrel therapy. Clin Pharmacol Ther 2011; 90:328–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Scott SA, Sangkuhl K, Stein CM, Hulot JS, Mega JL, Roden DM, Klein TE, Sabatine MS, Johnson JA, Shuldiner AR; Clinical Pharmacogenetics Implementation C. Clinical Pharmacogenetics Implementation Consortium guidelines for CYP2C19 genotype and clopidogrel therapy: 2013 update. Clin Pharmacol Ther 2013; 94:317–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Bourgeois S, Jorgensen A, Zhang EJ, Hanson A, Gillman MS, Bumpstead S, Toh CH, Williamson P, Daly AK, Kamali F, Deloukas P, Pirmohamed M. A multi-factorial analysis of response to warfarin in a UK prospective cohort. Genome Med 2016; 8:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Alfirevic A, Pirmohamed M. Genomics of adverse drug reactions. Trends Pharmacol Sci 2017; 38:100–9 [DOI] [PubMed] [Google Scholar]
  • 42.Chen P, Lin JJ, Lu CS, Ong CT, Hsieh PF, Yang CC, Tai CT, Wu SL, Lu CH, Hsu YC, Yu HY, Ro LS, Lu CT, Chu CC, Tsai JJ, Su YH, Lan SH, Sung SF, Lin SY, Chuang HP, Huang LC, Chen YJ, Tsai PJ, Liao HT, Lin YH, Chen CH, Chung WH, Hung SI, Wu JY, Chang CF, Chen L, Chen YT, Shen CY, Taiwan SJSC. Carbamazepine-induced toxic effects and HLA-B*1502 screening in Taiwan. N Engl J Med 2011; 364:1126–33 [DOI] [PubMed] [Google Scholar]
  • 43.Mallal S, Phillips E, Carosi G, Molina JM, Workman C, Tomazic J, Jagel-Guedes E, Rugina S, Kozyrev O, Cid JF, Hay P, Nolan D, Hughes S, Hughes A, Ryan S, Fitch N, Thorborn D, Benbow A. HLA-B*5701 screening for hypersensitivity to abacavir. N Engl J Med 2008; 358:568–79 [DOI] [PubMed] [Google Scholar]
  • 44.Hughes DA, Vilar FJ, Ward CC, Alfirevic A, Park BK, Pirmohamed M. Cost-effectiveness analysis of HLA B*5701 genotyping in preventing abacavir hypersensitivity. Pharmacogenetics 2004; 14:335–42 [DOI] [PubMed] [Google Scholar]
  • 45.Plumpton CO, Yip VL, Alfirevic A, Marson AG, Pirmohamed M, Hughes DA. Cost-effectiveness of screening for HLA-A*31:01 prior to initiation of carbamazepine in epilepsy. Epilepsia 2015; 56:556–63 [DOI] [PubMed] [Google Scholar]
  • 46.Tiamkao S, Jitpimolmard J, Sawanyawisuth K, Jitpimolmard S. Cost minimization of HLA-B*1502 screening before prescribing carbamazepine in Thailand. Int J Clin Pharm 2013; 35:608–12 [DOI] [PubMed] [Google Scholar]
  • 47.Thompson AJ, Newman WG, Elliott RA, Roberts SA, Tricker K, Payne K. The cost-effectiveness of a pharmacogenetic test: a trial-based evaluation of TPMT genotyping for azathioprine. Value Health 2014; 17:22–33 [DOI] [PubMed] [Google Scholar]
  • 48.Sgro C, Clinard F, Ouazir K, Chanay H, Allard C, Guilleminet C, Lenoir C, Lemoine A, Hillon P. Incidence of drug-induced hepatic injuries: a French population-based study. Hepatology 2002; 36:451–5 [DOI] [PubMed] [Google Scholar]
  • 49.Pirmohamed M, Friedmann PS, Molokhia M, Loke YK, Smith C, Phillips E, La Grenade L, Carleton B, Papaluca-Amati M, Demoly P, Shear NH. Phenotype standardization for immune-mediated drug-induced skin injury. Clin Pharmacol Ther 2011; 89:896–901 [DOI] [PubMed] [Google Scholar]
  • 50.Aithal GP, Watkins PB, Andrade RJ, Larrey D, Molokhia M, Takikawa H, Hunt CM, Wilke RA, Avigan M, Kaplowitz N, Bjornsson E, Daly AK. Case definition and phenotype standardization in drug-induced liver injury. Clin Pharmacol Ther 2011; 89:806–15 [DOI] [PubMed] [Google Scholar]
  • 51.Behr ER, January C, Schulze-Bahr E, Grace AA, Kaab S, Fiszman M, Gathers S, Buckman S, Youssef A, Pirmohamed M, Roden D. The International Serious Adverse Events Consortium (iSAEC) phenotype standardization project for drug-induced torsades de pointes. Eur Heart J 2012; 34:1958–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Alfirevic A, Neely D, Armitage J, Chinoy H, Cooper RG, Laaksonen R, Carr DF, Bloch KM, Fahy J, Hanson A, Yue QY, Wadelius M, Maitland-van Der Zee AH, Voora D, Psaty BM, Palmer CN, Pirmohamed M. Phenotype standardization for statin-induced myotoxicity. Clin Pharmacol Ther 2014; 96:470–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Maranville JC, Cox NJ. Pharmacogenomic variants have larger effect sizes than genetic variants associated with other dichotomous complex traits. Pharmacogenomics J 2016; 16:388–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.van der Wouden CH, Cambon-Thomsen A, Cecchin E, Cheung KC, Dávila-Fajardo CL, Deneer VH, Dolžan V, Ingelman-Sundberg M, Jönsson S, Karlsson MO, Kriek M, Mitropoulou C, Patrinos GP, Pirmohamed M, Samwald M, Schaeffeler E, Schwab M, Steinberger D, Stingl J, Sunder-Plassmann G, Toffoli G, Turner RM, van Rhenen MH, Swen JJ, Guchelaar HJ; on behalf of the Ubiquitous Pharmacogenomics C. Implementing pharmacogenomics in Europe: design and implementation strategy of the ubiquitous Pharmacogenomics Consortium. Clin Pharmacol Ther 2017; 101:341–58 [DOI] [PubMed] [Google Scholar]
  • 55.Johnson JA, Gong L, Whirl-Carrillo M, Gage BF, Scott SA, Stein CM, Anderson JL, Kimmel SE, Lee MT, Pirmohamed M, Wadelius M, Klein TE, Altman RB. Clinical Pharmacogenetics Implementation Consortium guidelines for CYP2C9 and VKORC1 genotypes and warfarin dosing. Clin Pharmacol Ther 2011; 90:625–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Matthaei J, Brockmoller J, Tzvetkov MV, Sehrt D, Sachse-Seeboth C, Hjelmborg JB, Moller S, Halekoh U, Hofmann U, Schwab M, Kerb R. Heritability of metoprolol and torsemide pharmacokinetics. Clin Pharmacol Ther 2015; 98:611–21 [DOI] [PubMed] [Google Scholar]
  • 57.Kozyra M, Ingelman-Sundberg M, Lauschke VM. Rare genetic variants in cellular transporters, metabolic enzymes, and nuclear receptors can be important determinants of interindividual differences in drug response. Genet Med 2017; 19:20–9 [DOI] [PubMed] [Google Scholar]
  • 58.Klaassen CD, Cui JY. Review: mechanisms of how the intestinal microbiota alters the effects of drugs and bile acids. Drug Metab Dispos 2015; 43:1505–21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Spanogiannopoulos P, Bess EN, Carmody RN, Turnbaugh PJ. The microbial pharmacists within us: a metagenomic view of xenobiotic metabolism. Nat Rev Microbiol 2016; 14:273–87 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S, Prifti E, Vieira-Silva S, Gudmundsdottir V, Pedersen HK, Arumugam M, Kristiansen K, Voigt AY, Vestergaard H, Hercog R, Costea PI, Kultima JR, Li J, Jorgensen T, Levenez F, Dore J, Meta HITc, Nielsen HB, Brunak S, Raes J, Hansen T, Wang J, Ehrlich SD, Bork P, Pedersen O. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 2015; 528:262–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Boland MR, Jacunski A, Lorberbaum T, Romano JD, Moskovitch R, Tatonetti NP. Systems biology approaches for identifying adverse drug reactions and elucidating their underlying biological mechanisms. Wiley Interdiscip Rev Syst Biol Med 2016; 8:104–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kindermann W. Creatine kinase levels after exercise. Dtsc Arztebl Int 2016; 113:344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Assanangkornchai N, Akaraborworn O, Kongkamol C, Kaewsaengrueang K. Characteristics of trauma patients with creatine kinase elevation. Crit Care 2015; 19:P282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Duma RJ, Siegel AL. Serum creatinine phosphokinase in acute myocardial infarction: diagnostic value. Arch Int Med 1965; 115:443–51 [DOI] [PubMed] [Google Scholar]
  • 65.Watanabe R, Watanabe H, Sotozono C, Kokaze A, Iijima M. Critical factors differentiating erythema multiforme majus from Stevens–Johnson syndrome (SJS)/toxic epidermal necrolysis (TEN). Eur J Dermatol 2011; 21:889–94 [DOI] [PubMed] [Google Scholar]
  • 66.Chung WH, Hung SI, Yang JY, Su SC, Huang SP, Wei CY, Chin SW, Chiou CC, Chu SC, Ho HC, Yang CH, Lu CF, Wu JY, Liao YD, Chen YT. Granulysin is a key mediator for disseminated keratinocyte death in Stevens–Johnson syndrome and toxic epidermal necrolysis. Nat Med 2008; 14:1343–50 [DOI] [PubMed] [Google Scholar]
  • 67.Su SC, Mockenhaupt M, Wolkenstein P, Dunant A, Le Gouvello S, Chen CB, Chosidow O, Valeyrie-Allanore L, Bellon T, Sekula P, Wang CW, Schumacher M, Kardaun SH, Hung SI, Roujeau JC, Chung WH. Interleukin-15 is associated with severity and mortality in Stevens–Johnson syndrome/toxic epidermal necrolysis. J Invest Dermatol 2017; 137:1065–73 [DOI] [PubMed] [Google Scholar]
  • 68.Nakajima S, Watanabe H, Tohyama M, Sugita K, Iijima M, Hashimoto K, Tokura Y, Nishimura Y, Doi H, Tanioka M, Miyachi Y, Kabashima K. High-mobility group box 1 protein (HMGB1) as a novel diagnostic tool for toxic epidermal necrolysis and Stevens–Johnson syndrome. Arch Dermatol 2011; 147:1110–2 [DOI] [PubMed] [Google Scholar]
  • 69.Sato S, Ichihara A, Jinnin M, Izuno Y, Fukushima S, Ihn H. Serum miR-124 up-regulation as a disease marker of toxic epidermal necrolysis. Eur J Dermatol 2015; 25:457–62 [DOI] [PubMed] [Google Scholar]
  • 70.Ichihara A, Wang Z, Jinnin M, Izuno Y, Shimozono N, Yamane K, Fujisawa A, Moriya C, Fukushima S, Inoue Y, Ihn H. Upregulation of miR-18a-5p contributes to epidermal necrolysis in severe drug eruptions. J Allergy Clin Immunol 2013 [DOI] [PubMed]
  • 71.Hall P, Cash J. What is the real function of the liver 'function' tests? Ulster Med J 2012; 81:30–6 [PMC free article] [PubMed] [Google Scholar]
  • 72.Giannini EG, Testa R, Savarino V. Liver enzyme alteration: a guide for clinicians. CMAJ 2005; 172:367–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lea JD, Clarke JI, McGuire N, Antoine DJ. Redox-dependent HMGB1 isoforms as pivotal co-ordinators of drug-induced liver injury: mechanistic biomarkers and therapeutic targets. Antioxid Redox Signal 2016; 24:652–65 [DOI] [PubMed] [Google Scholar]
  • 74.Starkey Lewis PJ, Dear J, Platt V, Simpson KJ, Craig DG, Antoine DJ, French NS, Dhaun N, Webb DJ, Costello EM, Neoptolemos JP, Moggs J, Goldring CE, Park BK. Circulating microRNAs as potential markers of human drug-induced liver injury. Hepatology 2011; 54:1767–76 [DOI] [PubMed] [Google Scholar]
  • 75.Thulin P, Nordahl G, Gry M, Yimer G, Aklillu E, Makonnen E, Aderaye G, Lindquist L, Mattsson CM, Ekblom B, Antoine DJ, Park BK, Linder S, Harrill AH, Watkins PB, Glinghammar B, Schuppe-Koistinen I. Keratin-18 and microRNA-122 complement alanine aminotransferase as novel safety biomarkers for drug-induced liver injury in two human cohorts. Liver Int 2014; 34:367–78 [DOI] [PubMed] [Google Scholar]
  • 76.Ki Y, Kim W, Nam J, Kim D, Park D, Kim D. C-reactive protein levels and radiation-induced mucositis in patients with head-and-neck cancer. Int J Radiat Oncol Biol Phys 2009; 75:393–8 [DOI] [PubMed] [Google Scholar]
  • 77.Walker TR, Land ML, Kartashov A, Saslowsky TM, Lyerly DM, Boone JH, Rufo PA. Fecal lactoferrin is a sensitive and specific marker of disease activity in children and young adults with inflammatory bowel disease. J Pediatr Gastroenterol Nutr 2007; 44:414–22 [DOI] [PubMed] [Google Scholar]
  • 78.Burri E, Beglinger C. The use of fecal calprotectin as a biomarker in gastrointestinal disease. Expert Rev Gastroenterol Hepatol 2014; 8:197–210 [DOI] [PubMed] [Google Scholar]
  • 79.Rule AD, Larson TS, Bergstralh EJ, Slezak JM, Jacobsen SJ, Cosio FG. Using serum creatinine to estimate glomerular filtration rate: accuracy in good health and in chronic kidney disease. Ann Intern Med 2004; 141:929–37 [DOI] [PubMed] [Google Scholar]
  • 80.Kyhse-Andersen J, Schmidt C, Nordin G, Andersson B, Nilsson-Ehle P, Lindstrom V, Grubb A. Serum cystatin C, determined by a rapid, automated particle-enhanced turbidimetric method, is a better marker than serum creatinine for glomerular filtration rate. Clin Chem 1994; 40:1921–6 [PubMed] [Google Scholar]
  • 81.Devarajan P. Neutrophil gelatinase-associated lipocalin (NGAL): a new marker of kidney disease. Scand J Clin Lab Invest Suppl 2008; 241:89–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Antoine DJ, Dear JW, Lewis PS, Platt V, Coyle J, Masson M, Thanacoody RH, Gray AJ, Webb DJ, Moggs JG, Bateman DN, Goldring CE, Park BK. Mechanistic biomarkers provide early and sensitive detection of acetaminophen-induced acute liver injury at first presentation to hospital. Hepatology 2013; 58:777–87 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Bonventre JV, Vaidya VS, Schmouder R, Feig P, Dieterle F. Next-generation biomarkers for detecting kidney toxicity. Nat Biotechnol 2010; 28:436–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Andersson U, Antoine DJ, Tracey KJ. The functions of HMGB1 depend on molecular localization and post-translational modifications. J Intern Med 2014; 276:420–4 [DOI] [PubMed] [Google Scholar]
  • 85.Yang Y, Li F, Du J, Shen Y, Lin J, Zhu X, Luo X, Liang J, Xu J. Variable levels of apoptotic signal-associated cytokines in the disease course of patients with Stevens-Johnson syndrome and toxic epidermal necrolysis. Australas J Dermatol 2016 [DOI] [PubMed]
  • 86.Schiraldi M, Raucci A, Munoz LM, Livoti E, Celona B, Venereau E, Apuzzo T, De Marchis F, Pedotti M, Bachi A, Thelen M, Varani L, Mellado M, Proudfoot A, Bianchi ME, Uguccioni M. HMGB1 promotes recruitment of inflammatory cells to damaged tissues by forming a complex with CXCL12 and signaling via CXCR4. J Exp Med 2012; 209:551–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Fujita H, Matsukura S, Watanabe T, Komitsu N, Watanabe Y, Takahashi Y, Kambara T, Ikezawa Z, Aihara M. The serum level of HMGB1 (high mobility group box 1 protein) is preferentially high in drug-induced hypersensitivity syndrome/drug reaction with eosinophilia and systemic symptoms. Br J Dermatol 2014; 171:1585–8 [DOI] [PubMed] [Google Scholar]
  • 88.Ogese MO, Faulkner L, Jenkins RE, French NS, Copple IM, Antoine DJ, Elmasry M, Malik H, Goldring CE, Kevin Park B, Betts C, Naisbitt DJ. Characterisation of drug-specific signalling between primary human hepatocytes and immune cells. Toxicol Sci Epub ahead of print 21 April 2017. doi: 10.1093/toxsci/kfx069 [DOI] [PubMed]
  • 89.Lee HY, Walsh SA, Creamer D. Long term complications of Stevens–Johnson syndrome/toxic epidermal necrolysis: the spectrum of chronic problems in patients who survive an episode of SJS/TEN necessitates multi-disciplinary follow up. Br J Dermatol Epub ahead of print 1 February 2017. doi: 10.1111/bjd.15360 [DOI] [PubMed]
  • 90. Bonventre JV. Kidney Injury Molecule-1 (KIM-1): a specific and sensitive biomarker of kidney injury. Scand J Clin Lab Invest Suppl 2008;241:78–83. [DOI] [PubMed]

Articles from Experimental Biology and Medicine are provided here courtesy of Frontiers Media SA

RESOURCES