997
Views
0
CrossRef citations to date
0
Altmetric
Review

Next-generation sequencing in pharmacogenomics – fit for clinical decision support?

ORCID Icon & ORCID Icon
Pages 213-223 | Received 16 Oct 2023, Accepted 16 Jan 2024, Published online: 23 Jan 2024

ABSTRACT

Introduction

The technological advances of sequencing methods during the past 20 years have fuelled the generation of large amounts of sequencing data that comprise common variations, as well as millions of rare and personal variants that would not be identified by conventional genotyping. While comprehensive sequencing is technically feasible, its clinical utility for guiding personalized treatment decisions remains controversial.

Areas covered

We discuss the opportunities and challenges of comprehensive sequencing compared to targeted genotyping for pharmacogenomic applications. Current pharmacogenomic sequencing panels are heterogeneous and clinical actionability of the included genes is not a major focus. We provide a current overview and critical discussion of how current studies utilize sequencing data either retrospectively from biobanks, databases or repurposed diagnostic sequencing, or prospectively using pharmacogenomic sequencing.

Expert opinion

While sequencing-based pharmacogenomics has provided important insights into genetic variations underlying the safety and efficacy of a multitude pharmacological treatments, important hurdles for the clinical implementation of pharmacogenomic sequencing remain. We identify gaps in the interpretation of pharmacogenetic variants, technical challenges pertaining to complex loci and variant phasing, as well as unclear cost-effectiveness and incomplete reimbursement. It is critical to address these challenges in order to realize the promising prospects of pharmacogenomic sequencing.

1. Introduction

Inter-individual variability in drug response and toxicity constitutes a common phenomenon in clinical care that poses a significant burden on the healthcare system. A multitude of factors contribute to unintended treatment outcomes including drug–drug interactions, physiological and pathophysiological factors, environmental cues, adherence, and genetic differences, the latter of which are estimated to account for an estimated 20–30% of this variability [Citation1]. The majority of genetic variations that affect drug response or the risk of adverse drug reactions (ADRs) localize in loci involved in drug absorption, distribution, metabolism, and excretion (ADME) or in genes that encode drug targets. Genetic variants in these genes that robustly associate with drug efficacy or safety can provide biomarkers for the individualized guidance of pharmacotherapy and further identification of such pharmacogenomic associations thus constitutes a key aim of precision medicine [Citation2–4]. To date, around 180 drug labels by the US Food and Drug Administration (FDA) and European Medicines Agency (EMA) have actionable pharmacogenomic labels [Citation5]. Importantly, >90% of all individuals carry at least one actionable genotype [Citation6–8] and 58% of all adult patients are prescribed at least one medicine with available pharmacogenomic guidance, which increases to 89% for patients over 70 years of age [Citation9,Citation10]. Nevertheless, the use of pharmacogenomic biomarkers in clinical practice is limited and only few associations are routinely used in clinical care outside of targeted oncology.

From twin studies, it is clear that candidate variants that are typically evaluated in association studies only explain a part of the overall hereditary variability in pharmacogenetic traits [Citation11]. While consideration of common variations can improve the outcomes of pharmacotherapy, considerable variability up to 100-fold can remain even within genotype-stratified groups [Citation12,Citation13]. These findings strongly support the hypothesis that a considerable fraction of heritable drug response variations remains unexplained. This missing heritability can be due to rare or personal variants not typically considered in genotyping-based analyses, epigenetic mechanisms or caused by epistatic phenomena in cis or trans [Citation14].

Currently, pharmacogenomic testing is mainly conducted via the genotyping of pre-selected variants in a panel of pharmacogenes. The advancement of sequencing technologies during the past 20 years has, however, revealed that there are tens of thousands of different variations across the human pharmacogenome. Specifically, population-scale sequencing projects revealed that > 90% of all identified pharmacogenomic variants are rare with minor allele frequencies (MAFs) below 1% and each individual carries multiple rare functional variants across ADME and drug target genes [Citation15–17]. Overall, rare coding and non-coding variations are estimated to account for approximately 30% and 20% of the genetically encoded functional variability in pharmacogenes, respectively [Citation18,Citation19]. These estimates suggest that consideration of comprehensive sequencing data that includes such rare variations could considerably improve individualized predictions of pharmacophenotypes.

In this review, we discuss the advantages and limitations of Next-Generation Sequencing (NGS) and genotyping in clinical pharmacogenomics and compare the existing panels designed for pharmacogenomic sequencing in either research or clinical settings. Subsequently, we provide a current overview of sequencing studies that either repurpose clinical sequencing data or conducted targeted pharmacogenomic sequencing to predict drug response or explain extreme phenotypes. We conclude that NGS provides a powerful and versatile tool for precision medicine whose implementation into routine clinical care, however, requires further developments at multiple fronts.

2. Genotyping versus sequencing

Pharmacogenomic testing via genotyping has been widely adopted in many clinical institutions and pharmacogenomic programs [Citation20–22]. Genotyped variants usually have strong evidence for their association with drug efficacy or toxicity. Therefore, the test results are easier to interpret and, when respective guidelines are available, can be translated into dose adjustments or alternative drug selection.

In contrast to genotyping, which can only cover a limited number of candidate variants, sequencing provides comprehensive genetic information about the interrogated loci. Three different sequencing strategies can be distinguished: whole-genome sequencing (WGS), whole-exome sequencing (WES) and targeted sequencing of selected candidate loci. From a technical standpoint, pharmacogenomic variants can by now be accurately called from sequencing data and results show high concordance with targeted genotyping results [Citation23–25]. While genotyping of candidate polymorphisms detects most clinically significant pharmacogenomic variants, up to 13% of variants per pharmacogene are not covered in conventional genotyping panels and could only be detected by sequencing-based approaches [Citation26]. It is furthermore important to consider that conventional short-read sequencing also has technical limitations, particularly with regard to variant detection in complex pharmacogenetic regions that contain repetitive sequences, copy number variations (CNVs) or nearby homologous pseudogenes, such as CYP2D6, CYP2B6 and the human HLA locus [Citation27]. These issues can be addressed by using NGS with longer reads. For instance, while five out of nine CYP2B6 exons contained inaccessible regions when using 100bp single-end reads, all exons could be fully interrogated when 200bp reads were used instead [Citation28]. Alternatively, long-read sequencing is increasingly utilized, which produces continuous reads of 20-100kb, thus allowing to produce phased information even in highly complex loci. While not yet in routine clinical use, current developments in software and workflows have lowered the barriers for implementation [Citation29,Citation30].

Importantly, while some clinical testing facilities already use sequencing as a modality for pharmacogenomic testing, they only extract known variations with available guidelines and do not interpret results pertaining to pharmcogenomic variants of unknown significance. The main reason is that ACMG/AMP guidelines are designed for the interpretation of variants encountered through clinical sequencing for hereditary disorders [Citation31]. Specifically, they recommend the functional classification of variants based on population data, computational variant effect predictions, as well as functional and segregation data. While these categories are useful for the classification of variant pathogenicity, they are not applicable to pharmacogenomic variants that are often not disease associated. No similar guidance documents for the interpretation of pharmacogenetic sequence variants are presently available. Furthermore, if variants of unknown significant coincide with variants or haplotypes for which pharmacogenomic guidance documents are available, it is currently unclear how such information should be reported. Thus, while the added technical capabilities of pharmacogenomic sequencing are evident, its main added value pertaining to the detection of rare or novel variations is currently not leveraged. Investigations into whether and how this additional information can be clinically utilized are thus an important frontier of pharmacogenomic research.

3. Pharmacogene selection

Pharmacogenomic variability can be examined i) by analyzing preexisting genetic data that was generated for other purposes, such as diagnostic sequencing or ii) by actual sequencing of pharmacogenes. Analysis of preexisting data, often from WGS or WES, is commonly conducted in a clinical setting. This approach requires only a reinterpretation of already available data and, as such, virtually any number of genes that are of interest in the analytical context can be assessed. In contrast, sequencing for pharmacogenomic applications is predominantly conducted in a research setting. In this context, targeted panel-based approaches are typically preferred over WGS or WES due their favorable balance between cost, throughput, and coverage.

Multiple panels for such sequencing-based interrogations of pharmacogenetic variation in large cohorts have been presented, including PGRN-Seq [Citation32], ClinPharmSeq [Citation33], PGxSeq [Citation34] and PKSeq [Citation35]. These panels differ considerably in scope, ranging from 59 to 340 included genes, as well as in gene composition (, , Supplementary Table S1). Only 18 genes are included in all of the pharmacogenomic sequencing panels, whereas 261 and 91 were only included in one and two panels, respectively (). Genes encoding drug metabolizing enzymes were more extensively covered, whereas drug target genes were underrepresented. Of the 26 actionable pharmacogenes, i.e. genes encoded in the nuclear genome with guidelines from the Clinical Pharmacogenetics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Group (DPWG), 12 were covered by all sequencing panels (). These mostly included polymorphic CYPs and other phase I or phase II enzymes, as well as the drug transporter gene SLCO1B1 and VKORC1, encoding vitamin K epoxide reductase, the target enzyme of warfarin. Furthermore, NAT2, POR, SLC15A2, SLC22A2, SLCO2B1, and UGT1A4 were also evaluated by all panels, despite a lack of pharmacogenetic guidelines. In contrast, multiple loci associated with adverse events, such as HLA-A, HLA-B and CACNA1S, or treatment efficacy (IFNL3) were only covered in one or two of the sequencing panels. Combined, these results suggest that clinical actionability per se is not a major focus of most current pharmacogenomic sequencing panels, despite the ambition that panel-based approaches might eventually transition from research tools to clinical use.

Figure 1. Analysis of pharmacogenomic targeted sequencing panels. (a) The concordance of genes included in pharmacogenomic sequencing panels. Highest concordance is indicated in red and lowest in blue. (b) Column plot showing the overlap of pharmacogenes across the six analyzed panels. Note that pharmacogenes included in 4–6 panels are almost exclusively encoding factors involved in the absorption, distribution, metabolism and excretion (ADME) of drugs, whereas most drug target genes are only included in 1–3 panels. (c) Column plot showing the panel coverage of pharmacogenes with established actionable guidelines by CPIC or DPWG. While 12 out of 26 actionable genes are included in all panels, a total of nine genes are included in less than half of all panels.

Figure 1. Analysis of pharmacogenomic targeted sequencing panels. (a) The concordance of genes included in pharmacogenomic sequencing panels. Highest concordance is indicated in red and lowest in blue. (b) Column plot showing the overlap of pharmacogenes across the six analyzed panels. Note that pharmacogenes included in 4–6 panels are almost exclusively encoding factors involved in the absorption, distribution, metabolism and excretion (ADME) of drugs, whereas most drug target genes are only included in 1–3 panels. (c) Column plot showing the panel coverage of pharmacogenes with established actionable guidelines by CPIC or DPWG. While 12 out of 26 actionable genes are included in all panels, a total of nine genes are included in less than half of all panels.

Table 1. Number of pharmacogenes included in NGS-based pharmacogenomic studies.

4. Repurposing of diagnostic and population-scale sequencing data

Due to the plummeting cost and increasing accessibility of NGS, large amounts of human sequencing data have accumulated over the past decades. Where accessible, these datasets can be tapped into to conduct retrospective pharmacogenomic analyses. To this end, sequencing data is mostly interpreted as targeted genotyping tests, i.e. only a few common variants with known clinical significance are extracted and functionally interpreted to infer metabolic phenotypes, whereas rare and novel variants without actionable guidance are not considered. In this section, we first discuss the potential and limitations of pharmacogenomic repurposing of such clinical sequencing data generated for diagnostic applications. Subsequently, we highlight the importance of biobanks and large population-scale sequencing projects for pharmacogenomic analysis.

WES and WGS are powerful tools for the molecular diagnostics of congenital and rare diseases [Citation38,Citation39]. Repurposing of such clinical sequencing data for pharmacogenomic analyses can provide population frequencies as well as useful information for personal drug response predictions (). Analysis of WES and targeted exome sequencing data from 5,001 Spanish or Latin American individuals identified 280 alleles in 11 pharmacogenes that can be used to inform pharmacogenomic phenotypes impacting over 95% of the studied cohort [Citation40]. In addition, 197 novel variants were discovered, of which 26 were considered loss-of-function. Similarly, pharmacogenomic profiling of WES data from 1,116 Hong Kong Chinese revealed that almost all individuals carried at least one actionable pharmacogenomic variant across 19 pharmacogenes and, when taking prescription data into account, 13.4% of the population would benefit from pharmacogenetically guided drug prescriptions [Citation42]. Furthermore, above 93% of the individuals carry at least one putatively deleterious rare variant across 108 pharmacogenes. While repurposing of WES data resulted in meaningful pharmacogenomic profile, it suffers from difficulties in inferring functionality of UGT1A1, CYP3A5, CYP2C19 and CYP2D6, due to the lack of coverage of important non-exonic variations and problems pertaining to copy number variant calling. Consequently, functional interpretation of these loci requires additional validation before these data can provide guidance for clinical use [Citation41]. Such issues can be overcome by using WGS and multiple studies have successfully translated WGS data into pharmacogenomic phenotypes, mostly in pediatric cohorts [Citation46,Citation47].

Table 2. Studies repurposing clinical sequencing data for pharmacogenomic analysis. WES = whole-exome sequencing; WGS = whole-genome sequencing; TS = targeted sequencing.

NGS is furthermore recommended for a variety of solid tumors and hematological malignancies and has become standard in clinical oncology. While recommendations regarding the sequencing approach and scope can differ between tumor type and stage [Citation48], even comprehensive WGS has been shown to be feasible in clinical practice in a comprehensive cancer center setting [Citation49]. The majority of cancer patients receive NGS only on tumor samples to identify potential therapeutic vulnerabilities. However, whether somatic sequences can be reliably used to infer germline genotypes of drug response variants remains controversial [Citation50–52]. In recent years, there is an increase in paired somatic-germline sequencing. This approach allow offers multiple advantages, such as better guidance for treatment selection and the opportunity to reduce risk in family members [Citation53,Citation54]. Furthermore, due to the resulting availability of genomic germline data, such information can be repurposed for pharmacogenomic applications for instance, to make evidence-based treatment recommendations for common chemotherapeutics [Citation55].

While the aforementioned evidence indicates the feasibility and utility of repurposing diagnostic sequencing data for pharmacogenomic purposes, the seamless integration of this information into electronic health records (EHRs) remains challenging. A 2015 proof-of-concept study identified two key challenges – the accurate interpretation of incidental findings in agreement with pharmacogenomic guidelines and identification of those findings that are clinically meaningful [Citation44,Citation56]. For well-characterized variations, the first challenge has been largely addressed by pharmacogenomic variant calling and interpretation tools, such as Stargazer [Citation57], Aldy [Citation58] and PharmCAT [Citation59], whereas the interpretation of rare variants with unknown functional significance remains an open issue. Furthermore, a dedicated pharmacogenomic reporting system, named lmPGX, was recently presented that fully automates the translation of clinical short-read sequencing data into succinct recommendations for 11 clinically actionable pharmacogenes that can be readily interpreted by clinicians [Citation60]. The second challenge, however, remains largely unsolved since the functional interpretation of an individual’s pharmacogenomic profile remains, with few exceptions, imprecise.

In recent years, biobank evolved from collections of biological samples to complex multi-dimensional and often decentralized repositories in which diverse samples are stored and annotated with clinical information and, often, complex omics data [Citation61]. Many national biobanks were established that contain in-depth genetic and health information for population-specific research purpose. For example, the UK Biobank currently contains genotype data from 500,000 individuals, WES data from 470,000 individuals and WGS data from 200,000 individuals that are all linked to EHRs [Citation62]. Similarly, the Estonian Biobank includes genetic data from 200,000 individuals, WES data from 2,500 individuals and WGS data from 3,000 individuals accompanied by lifestyle and genealogical information [Citation63]. These biobanks serve as valuable resources for large-scale pharmacogenomic studies and have been used to generate comprehensive pharmacogenomic variation profiles. For example, analysis of the genetic variability across 14 pharmacogenes with CPIC guidelines based on the UK Biobank indicated that 99.5% of population are at risk of an atypical response to at least one drug [Citation8]. Similarly, preexisting genotype data of over 44,000 participants from the Estonian biobank, in comparison with WES and WGS data, showed that 99.8% of studied individuals carry at least one actionable variant and pharmacogenomic dosing adjustments will affect at least 50 daily drug doses per 1000 inhabitants [Citation7].

In addition, consolidated and harmonized international sequencing databases, such as the 1000 Genomes Project and The Genome Aggregation Database (gnomAD) have emerged as powerful tools for pharmacogenomic research. These resources not only allow analysis of pharmacogenomic germline variations across different ethnogeographic groups [Citation64–66] but also provide opportunities to estimate functional consequence of naturally occurring variants at the population-scale by integrating frequency information with functional data from other sources [Citation67]. Moreover, germline variation data from sequencing data stored in cancer databases, such as The Cancer Genome Atlas (TCGA), can be utilized to predict therapeutic effect of different cancer drugs [Citation68]. Even in the absence of EHR annotations, these resources constitute important repositories to assess global pharmacogenomic variability and, combined with established gene–drug relationships allow to estimate drug response phenotypes at the population scale.

5. Pharmacogenomic sequencing

In addition to retrospective pharmacogenomic analysis of preexisting sequencing data, NGS is increasingly applied prospectively (). More than 10 years ago, the RIGHT Study (The Right Drug, Right Dose, Right Time) was initiated that aimed to develop individualized treatment protocols using genomic data from cohort samples stored in the Mayo Clinic Biobank [Citation82]. A RIGHT pilot study was conducted in which 1,013 subjects were sequenced using the 84-gene PGRN-seq panel, followed by targeted analysis of SLCO1B1, CYP2C9, CYP2C19 and VKORC1. In addition, CYP2D6 was separately genotyped using a customized genotyping method. The result revealed that 99% of the subjects carried an actionable pharmacogenomic variant in as least one of the five genes [Citation69]. Functional consequences of novel variants identified in CYP2C9 and CYP2C19 were interrogated by in vitro and in silico methods in a follow-up study and the authors conclude that if novel variant interpretation can be generalized using high-throughput functional assays and highly accurate predictive algorithms, DNA sequencing will ultimately be preferable to genotyping for the clinical implementation of pharmacogenomic variants [Citation70]. Recently, the complete Mayo-Baylor RIGHT 10k Study was published, presenting the largest preemptive DNA sequence-based pharmacogenomic testing to date [Citation71]. In the frame of this study, targeted sequencing of 77 pharmacogenes was performed for 10,077 individuals. The results revealed that, in addition to clinically actionable variations, each individual carried on average 127 additional SNVs and indels, which would have been missed by genotyping. Importantly, 3.3 variants per individual were interpreted as putatively deleterious and more than 30 individuals carried rare functionally deleterious variants in CYP2C9 and CYP2C19 that were confirmed by deep mutational scanning. To facilitate clinical implementation, electronic consultations were sent to primary care providers of risk variant carriers to suggest dose adjustment or alternative therapy. These adjustments resulted in full remission or alleviation of ADRs in some cases. While clearly promising, stringent statistical evaluations of the results are required to demonstrate clinical utility.

Table 3. Pharmacogenomic sequencing studies. AUC = area under the curve; NGS = next-generation sequencing; WES = whole-exome sequencing; WGS = whole-genome sequencing; TS = targeted sequencing.

In a different study, a similar workflow from sequencing to pharmacogenomic result reporting was explored [Citation72]. Specifically, WES data from 100 individuals were analyzed for both star alleles and rare variants. The former was translated into individualized medication safety cards while the latter underwent complex computational assessments. Notably, the authors observed drastic discordance between different haplotype calling algorithms and proposed that, in the absence of more accurate and precise prediction methods, results from multiple tools should be integrated to obtain more confident results.

To harness the potential advantage of sequencing over genotyping, functional interpretation of rare and novel variants is of central importance. Resequencing of 137 DNA samples from the Genetic Testing Reference Material (GeT-RM) identified novel variants in CYP2C8, CYP2C9 and CYP2C19 that would have been missed in targeted genotyping assays, highlighting that many novel variants are yet to be discovered even in extensively studied pharmacogenes [Citation73]. To further address the issue of variant interpretation, an analytical workflow was developed in which 13 variants in CYP2D6 and CYP2C19 identified by targeted sequencing of 304 individuals were first assessed using in silico predictors and subsequently expressed in 293 FT cells for experimental analyses [Citation74]. The results demonstrated a high concordance between computational predictions and in vitro data, and emphasized the functional impact of novel variants for genotype–phenotype translations.

Besides prediction of drug response, pharmacogenomic data can also be utilized in forward genetics to identify genetic variations that explain ADRs or other pharmacophenotypes. For instance, WGS identified a novel rare missense variant in DPYD, p.R235Q, in a patient who developed life-threatening toxicity after capecitabine treatment after commonly tested risk variant could not explain the observed phenotype [Citation75]. This variant was predicted to be deleterious by the DPYD-specific prediction algorithm DPYD-Varifier [Citation83] and the prediction was validated in vitro. Combined, these mechanistic insights provide strong evidence for the conclusion that the identified variant is responsible for the observed phenotype despite the fact that the low frequency of the variant did not allow for epidemiological association studies. In another study, sequencing of DPYD coding and untranslated regions was conducted for 120 patients with grade 3–5 fluoropyrimidine toxicity and 104 matched controls [Citation76]. The authors found that rare variants were significantly enriched in the ADR group and that carrying at least one very rare DPYD variant increased the risk of developing acute fluoropyrimidine toxicity by fourfold (p = 0.017). In a separate example, a machine-learning algorithm trained on CYP2D6 long-read sequences from 561 breast cancer patients quantitatively predicted alterations of CYP2D6 enzyme activity [Citation78]. The model showed a 25% increase in the fraction of explained CYP2D6 variability compared to conventional approaches using star alleles, exemplifying the added value of using comprehensive sequencing for phenotype and drug response predictions.

Importantly, WGS also allows untargeted searches for gene-phenotype associations. In a study with 1,441 children with asthma who exhibited extreme albuterol bronchodilator drug response (BDR), rare variants in loci associated with lung capacity, immunity, and β-adrenergic signaling were shown to significantly contribute to genetically encoded variations in BDR [Citation77]. In addition to explaining outlier phenotypes, novel variants identified by targeted gene sequencing were also demonstrated to have important roles in predicting pharmacokinetic parameters, such as drug plasma concentration and clearance, and, as a result, can inform optimized treatment strategies [Citation79–81].

6. Expert opinion

Over the last decades, large-scale genetic association studies have clearly shown that common variants in ‘classical’ pharmacogenes can only explain a fraction of the genetically encoded inter-individual variability in drug response observed in the clinics. Sequencing technologies thus offer exciting opportunities to increase analytical breadth and detect associations with novel variants as well as additional loci that would not be included in classical genotyping arrays. Particularly pharmacogenomic sequencing panels allow to capture the full spectrum of pharmacogenomic variations and are suitable for large-scale profiling. Current panels differ in scope and composition and there is limited consensus as to which pharmacokinetic and pharmacodynamic genes are to be included in such panels. For instance, important pharmacogenes, such as HLA-A and HLA-B are not consistently covered in these panels, at least in part due to technical difficulties in analyzing these loci. The incorporation of sequencing data into consolidated databases as well as the increasing availability of biobanks can further fuel profiling of the human pharmacovariome at population-scale.

Despite the impressive developments in the research domain and promising prospects for clinical application of sequencing-based pharmacogenomics, significant hurdles remain to be overcome [Citation84–87]:

  1. Sequencing-based solutions require appropriate processes and workflows to handle variants of unknown significance if the added value of these technologies is to be leveraged. These could include computational pipelines for the functional interpretation of identified variants, which can, as one piece of evidence, assist clinical decision-making, for instance, by flagging the respective variant carriers for increased surveillance.

  2. Specific attention needs to be paid for genes that are challenging to evaluate, such as CYP2B6, CYP2D6 or HLA genes. This includes both the use of appropriate sequencing methods with sufficiently long read-lengths and the use of specific analytical software packages that allow for the accurate calling of genotypes in these genes [Citation88,Citation89].

  3. When one or more heterozygous variants are detected, the correct phasing of variants can be important for the inference of functional phenotypes [Citation90]. While imputation can increase the likelihood of correct phasing, only long-read sequencing can fully overcome these issues.

  4. The inclusion of sequencing needs to be feasible in clinical routine, i.e. the necessary infrastructure and expertise must be available, and workflows must be established to generate results with an acceptable turnaround time.

  5. The cost-effectiveness of sequencing must be demonstrated. While the majority of studies report that genotype-guided dosing might be cost-effective or even dominant in different disease areas [Citation91–93], most studies utilized genotyping and the cost-effectiveness of sequencing remains unclear. With declining sequencing costs, other factors pertaining to data analytics and storage are becoming increasingly more relevant [Citation94]. In parallel to declining sequencing costs, emerging cloud storage solutions have resulted in a rapid reduction of storage costs from hundreds of USD to ten-year costs of 14 USD per genome and 0.71 USD per exome [Citation95]. However, analytical costs, including bioinformatics, interpretation and results reporting, remain relatively constant and can be as high as one-third of the sequencing costs [Citation96].

  6. The reimbursement of pharmacogenomic tests remains incomplete and variable. A recent study from a tertiary academic medical center in the US indicated that panel tests are being reimbursed at a significantly higher rate than single-gene tests [Citation97]. However, there are considerable differences over time as well as between indications, genes tested, and the specific payer, likely limiting the generalizability of these results. Considering the most important genomic tests as medical devices has been suggested to stimulate test reimbursement [Citation98]. While not specific to NGS-based pharmacogenomics, the higher out-of-pocket payments for sequencing magnifies the importance of this aspect compared to genotype-based tests. However, such decisions are complex and need to consider how the costs of the approval process propagate to the diagnostic market. Thus, whether genomic tests should be considered as medical devices requires robust health economic evaluations and pricing systems in order to avoid an eventual increase in test costs and decrease in test availability.

While each of the aforementioned points is complex and their in-depth discussion would go beyond the scope of this article, they can nevertheless illustrate the multitude of challenges that remain to be overcome to implement clinical sequencing-based pharmacogenomics. As a consequence, the use of NGS for prospective pharmacogenomic applications remains at present mostly limited to research settings.

To unlock its full potential, parallel progress has to be made in several directions. We specifically highlight the need for efficient and accessible incorporation of sequencing data into electronic health records so that the responsible physicians can make maximal use of the available data when needed. Furthermore, more reliable computational tools are needed that support the interpretation of pharmacogenetic variants with unknown significance to extend clinical actionability to the entire personal genomic information. Finally, appropriate ethical frameworks need to be in place at the respective study site, which ensure that utilization of sequencing data balances patient privacy with anticipated healthcare benefit. Only such integrated developments promise to help a highly efficient translation of sequencing data into personalized health recommendations in pharmacogenomics and beyond. Judging from recent developments, we anticipate that the aforementioned technical challenges can be largely overcome within the next five to 10 years. We are thus optimistic that, with time, pharmacogenomic utilization of sequencing data will become increasingly common and be well positioned to facilitate more accurate translation of an individual’s genetic variability into pharmacophenotypes and personalized treatments.

Article highlights

  • Sequencing shows benefits over genotyping regarding genomic information provided and utility over time. Lack of guidance with regard to the interpretation of rare and novel variants as well as unclear cost-effectiveness remain the main challenges for its broader clinical use.

  • Current pharmacogenomic targeted sequencing panels are used primarily for research applications and differ drastically in scope and composition.

  • Actionability is not major design criterion for the inclusion of genes in most pharmacogenomic sequencing panels.

  • NGS data from clinical projects, biobanks and sequencing databases provide ample resources for large-scale pharmacogenomic variation profiling and rare variant analysis.

  • Sequencing instead of genotyping is becoming increasingly prevalent for the investigation of extreme phenotypes and drug response predictions.

Declaration of interest

VM Lauschke is co-founder, CEO and shareholder of HepaPredict AB. The authors have no other 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 apart from those disclosed.

Reviewer disclosures

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

Additional information

Funding

The laboratory received funding from the Swedish Research Council [grant numbers 2019-01837 and 2021-02801], the Knut and Alice Wallenberg Foundation [Grant VC-2021-0026], Cancerfonden [Grant 23-0763PT] and the Robert Bosch Foundation, Stuttgart, Germany.

References

  • Sim SC, Kacevska M, Ingelman-Sundberg M. Pharmacogenomics of drug-metabolizing enzymes: a recent update on clinical implications and endogenous effects. Pharmacogenomics J. 2013;13(1):1–11. doi: 10.1038/tpj.2012.45
  • Weinshilboum RM, Wang L. Pharmacogenomics: precision medicine and drug response. Mayo Clin Proc. 2017;92(11):1711–1722. doi: 10.1016/j.mayocp.2017.09.001
  • Lauschke VM, Zhou Y, Ingelman-Sundberg M. Novel genetic and epigenetic factors of importance for inter-individual differences in drug disposition, response and toxicity. Pharmacol Ther. 2019;197:122–152. doi: 10.1016/j.pharmthera.2019.01.002
  • Pirmohamed M. Pharmacogenomics: current status and future perspectives. Nat Rev Genet. 2023;24(6):350–362. doi: 10.1038/s41576-022-00572-8
  • Shekhani R, Steinacher L, Swen JJ, et al. Evaluation of current regulation and guidelines of pharmacogenomic drug labels: opportunities for improvements. Clin Pharmacol Ther. 2020;107(5):1240–1255. doi: 10.1002/cpt.1720
  • Driest SLV, Shi Y, Bowton EA, et al. Clinically actionable genotypes among 10,000 patients with preemptive pharmacogenomic testing. Clin Pharmacol Ther. 2014;95(4):423–431. doi: 10.1038/clpt.2013.229
  • Reisberg S, Krebs K, Lepamets M, et al. Translating genotype data of 44,000 biobank participants into clinical pharmacogenetic recommendations: challenges and solutions. Genet Med. 2019;21(6):1345–1354. doi: 10.1038/s41436-018-0337-5
  • McInnes G, Lavertu A, Sangkuhl K, et al. Pharmacogenetics at scale: an analysis of the UK biobank. Clin Pharmacol Ther. 2021;109(6):1528–1537. doi: 10.1002/cpt.2122
  • Kimpton JE, Carey IM, Threapleton CJD, et al. Longitudinal exposure of English primary care patients to pharmacogenomic drugs: an analysis to inform design of pre‐emptive pharmacogenomic testing. Br J Clin Pharmacol. 2019;85(12):2734–2746. doi: 10.1111/bcp.14100
  • Hayward J, McDermott J, Qureshi N, et al. Pharmacogenomic testing to support prescribing in primary care: a structured review of implementation models. Pharmacogenomics. 2021;22(12):761–776. doi: 10.2217/pgs-2021-0032
  • Lauschke VM, Ingelman-Sundberg M. Prediction of drug response and adverse drug reactions: from twin studies to next generation sequencing. Eur J Pharm Sci. 2019;130:65–77. doi: 10.1016/j.ejps.2019.01.024
  • Jukic MM, Smith RL, Haslemo T, et al. Effect of CYP2D6 genotype on exposure and efficacy of risperidone and aripiprazole: a retrospective, cohort study. Lancet Psychiatry. 2019;6(5):418–426. doi: 10.1016/S2215-0366(19)30088-4
  • Bråten LS, Haslemo T, Jukic MM, et al. Impact of CYP2C19 genotype on sertraline exposure in 1200 scandinavian patients. Neuropsychopharmacol. 2020;45(3):570–576. doi: 10.1038/s41386-019-0554-x
  • Lauschke VM, Zhou Y, Ingelman-Sundberg M. Pharmacogenomics beyond single common genetic variants: the way forward. Annu Rev Pharmacol Toxicol. 2023;64. doi: 10.1146/annurev-pharmtox-051921-091209. Online ahead of print.
  • Ingelman-Sundberg M, Mkrtchian S, Zhou Y, et al. Integrating rare genetic variants into pharmacogenetic drug response predictions. Hum Genomics. 2018;12(1):26. doi: 10.1186/s40246-018-0157-3
  • Wright GEB, Carleton B, Hayden MR, et al. The global spectrum of protein-coding pharmacogenomic diversity. Pharmacogenomics J. 2018;18(1):187–195. doi: 10.1038/tpj.2016.77
  • Schärfe CPI, Tremmel R, Schwab M, et al. Genetic variation in human drug-related genes. Genome Med. 2017;9(1):117. doi: 10.1186/s13073-017-0502-5
  • 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(1):20–29. doi: 10.1038/gim.2016.33
  • Tremmel R, Zhou Y, Schwab M, et al. Structural variation of the coding and non-coding human pharmacogenome. NPJ Genom Med. 2023;8(1):24. doi: 10.1038/s41525-023-00371-y
  • Dunnenberger HM, Crews KR, Hoffman JM, et al. Preemptive clinical pharmacogenetics implementation: Current programs in five US medical centers. Annu Rev Pharmacol Toxicol. 2015;55(1):89–106. doi: 10.1146/annurev-pharmtox-010814-124835
  • Krebs K, Milani L. Translating pharmacogenomics into clinical decisions: do not let the perfect be the enemy of the good. Hum Genomics. 2019;13(1):39. doi: 10.1186/s40246-019-0229-z
  • Duarte JD, Dalton R, Elchynski AL, et al. Multisite investigation of strategies for the clinical implementation of pre-emptive pharmacogenetic testing. Genet Med. 2021;23(12):2335–2341. doi: 10.1038/s41436-021-01269-9
  • Yang W, Wu G, Broeckel U, et al. Comparison of genome sequencing and clinical genotyping for pharmacogenes. Clin Pharmacol Ther. 2016;100(4):380–388. doi: 10.1002/cpt.411
  • Chua EW, Cree SL, Ton KNT, et al. Cross-Comparison of exome analysis, next-generation sequencing of amplicons, and the iPLEX ADME PGx panel for pharmacogenomic profiling. Front Pharmacol. 2016;7:1. doi: 10.3389/fphar.2016.00001
  • Rasmussen-Torvik LJ, Almoguera B, Doheny KF, et al. Concordance between research sequencing and clinical pharmacogenetic genotyping in the eMERGE-PGx study. J Mol Diagn. 2017;19(4):561–566. doi: 10.1016/j.jmoldx.2017.04.002
  • Lopes JL, Harris K, Karow MB, et al. Targeted genotyping in clinical pharmacogenomics what is missing? J Mol Diagn. 2022;24(3):253–261. doi: 10.1016/j.jmoldx.2021.11.008
  • Lauschke VM, Milani L, Ingelman-Sundberg M. Pharmacogenomic biomarkers for improved drug therapy—recent progress and future developments. AAPS J. 2017;20(1):4. doi: 10.1208/s12248-017-0161-x
  • Desta Z, El‐Boraie A, Gong L, et al. PharmVar GeneFocus: CYP2B6. Clin Pharmacol Ther. 2021;110(1):82–97. doi: 10.1002/cpt.2166
  • Schaal W, Ameur A, Olsson-Strömberg U, et al. Migrating to long-read sequencing for clinical routine BCR-ABL1 TKI resistance mutation screening. Cancer Inform. 2022;21:11769351221110872. doi: 10.1177/11769351221110872
  • Kobayashi ES, Batalov S, Wenger AM, et al. Approaches to long-read sequencing in a clinical setting to improve diagnostic rate. Sci Rep. 2022;12(1):16945. doi: 10.1038/s41598-022-20113-x
  • Richards S, Aziz N, Bale S, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American college of medical genetics and genomics and the association for molecular pathology. Genet Med. 2015;17(5):405–424. doi: 10.1038/gim.2015.30
  • Gordon AS, Fulton RS, Qin X, et al. Pgrnseq: a targeted capture sequencing panel for pharmacogenetic research and implementation. Pharmacogenet Genom. 2016;26(4):161–168. doi: 10.1097/FPC.0000000000000202
  • Lee S, Shin J-Y, Kwon N-J, et al. ClinPharmSeq: A targeted sequencing panel for clinical pharmacogenetics implementation. PloS One. 2022;17(7):e0272129. doi: 10.1371/journal.pone.0272129
  • Gulilat M, Lamb T, Teft WA, et al. Targeted next generation sequencing as a tool for precision medicine. BMC Med Genomics. 2019;12(1):81. doi: 10.1186/s12920-019-0527-2
  • Fukunaga K, Momozawa Y, Mushiroda T. Update on next generation sequencing of pharmacokinetics-related genes: development of the PKseq panel, a platform for amplicon sequencing of drug-metabolizing enzyme and drug transporter genes. Drug Metab Pharmacokinet. 2021;37:100370. doi: 10.1016/j.dmpk.2020.11.005
  • Han SM, Park J, Lee JH, et al. Targeted next-generation sequencing for comprehensive genetic profiling of pharmacogenes. Clin Pharmacol Ther. 2017;101(3):396–405. doi: 10.1002/cpt.532
  • Klein K, Tremmel R, Winter S, et al. A new panel-based next-generation sequencing method for ADME genes reveals novel associations of common and rare variants with expression in a human liver cohort. Front Genet. 2019;10:7. doi: 10.3389/fgene.2019.00007
  • Yang Y, Muzny DM, Reid JG, et al. Clinical whole-exome sequencing for the diagnosis of mendelian disorders. N Engl J Med. 2013;369:1502–1511. doi: 10.1056/NEJMoa1306555
  • Stavropoulos DJ, Merico D, Jobling R, et al. Whole-genome sequencing expands diagnostic utility and improves clinical management in paediatric medicine. NPJ Genom Med. 2016;1(1):15012. doi: 10.1038/npjgenmed.2015.12
  • Lanillos J, Carcajona M, Maietta P, et al. Clinical pharmacogenetic analysis in 5,001 individuals with diagnostic exome sequencing data. NPJ Genom Med. 2022;7(1):12. doi: 10.1038/s41525-022-00283-3
  • van der Lee M, Allard WG, Bollen S, et al. Repurposing of diagnostic whole exome sequencing data of 1,583 individuals for clinical pharmacogenetics. Clin Pharmacol Ther. 2020;107(3):617–627. doi: 10.1002/cpt.1665
  • Yu MHC, Chan MCY, Chung CCY, et al. Actionable pharmacogenetic variants in Hong Kong Chinese exome sequencing data and projected prescription impact in the Hong Kong population. PloS Genet. 2021;17:e1009323. doi: 10.1371/journal.pgen.1009323
  • Cousin MA, Matey ET, Blackburn PR, et al. Pharmacogenomic findings from clinical whole exome sequencing of diagnostic odyssey patients. Mol Genet Genomic Med. 2017;5(3):269–279. doi: 10.1002/mgg3.283
  • Nishimura AA, Shirts BH, Dorschner MO, et al. Development of clinical decision support alerts for pharmacogenomic incidental findings from exome sequencing. Genet Med. 2015;17(11):939–942. doi: 10.1038/gim.2015.5
  • Caspar SM, Schneider T, Meienberg J, et al. Added value of clinical sequencing: WGS-Based profiling of pharmacogenes. Int J Mol Sci. 2020;21(7):2308. doi: 10.3390/ijms21072308
  • Cohn I, Paton TA, Marshall CR, et al. Genome sequencing as a platform for pharmacogenetic genotyping: a pediatric cohort study. NPJ Genom Med. 2017;2(1):19. doi: 10.1038/s41525-017-0021-8
  • Pan A, Scodellaro S, Khan T, et al. Pharmacogenetic profiling via genome sequencing in children with medical complexity. Pediatr Res. 2023;93(4):905–910. doi: 10.1038/s41390-022-02313-3
  • Mosele F, Remon J, Mateo J, et al. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO precision medicine working group. Ann Oncol. 2020;31(11):1491–1505. doi: 10.1016/j.annonc.2020.07.014
  • Samsom K, Monkhorst K, Luuk J, et al. Feasibility of whole-genome sequencing in routine clinical practice. J Clin Oncol. 2021;39(15_suppl):3013. doi: 10.1200/JCO.2021.39.15_suppl.3013
  • Weiss JR, Baer MR, Ambrosone CB, et al. Concordance of pharmacogenetic polymorphisms in tumor and germ line DNA in adult patients with acute myeloid leukemia. Cancer Epidemiol Biomarkers Prev. 2007;16(5):1038–1041. doi: 10.1158/1055-9965.EPI-06-0964
  • Hertz DL, Kidwell KM, Thibert JN, et al. Genotyping concordance in DNA extracted from formalin‐fixed paraffin embedded (FFPE) breast tumor and whole blood for pharmacogenetic analyses. Mol Oncol. 2015;9(9):1868–1876. doi: 10.1016/j.molonc.2015.07.002
  • Gillis N, Etheridge AS, Patil SA, et al. Sequencing of genes of drug response in tumor DNA and implications for precision medicine in cancer patients. Pharmacogenomics J. 2023;23(4):73–81. doi: 10.1038/s41397-023-00299-7
  • Mandelker D, Ceyhan-Birsoy O. Evolving significance of tumor-normal sequencing in cancer care. Trends Cancer. 2020;6(1):31–39. doi: 10.1016/j.trecan.2019.11.006
  • Henson JW. Paired tumor-germline testing as a driver in better cancer care. JAMA Netw Open. 2022;5(5):e2213077. doi: 10.1001/jamanetworkopen.2022.13077
  • Hertz DL, Glatz A, Pasternak AL, et al. Integration of germline pharmacogenetics into a tumor sequencing program. JCO Precis Oncol. 2018;2(2):1–15. doi: 10.1200/PO.18.00011
  • Janssens ACJW, Evans JP. Returning pharmacogenetic secondary findings from genome sequencing: let’s not put the cart before the horse. Genet Med. 2015;17(11):854–856. doi: 10.1038/gim.2015.59
  • Lee S, Wheeler MM, Patterson K, et al. Stargazer: a software tool for calling star alleles from next-generation sequencing data using CYP2D6 as a model. Genet Med. 2019;21(2):361–372. doi: 10.1038/s41436-018-0054-0
  • Hari A, Zhou Q, Gonzaludo N, et al. An efficient genotyper and star-allele caller for pharmacogenomics. Genome Res. 2023;33(1):61–70. doi: 10.1101/gr.277075.122
  • Sangkuhl K, Whirl-Carrillo M, Whaley RM, et al. Pharmacogenomics clinical annotation tool (PharmCAT). Clin Pharmacol Ther. 2020;107(1):203–210. doi: 10.1002/cpt.1568
  • Klanderman BJ, Koch C, Machini K, et al. Automated pharmacogenomic reports for clinical genome sequencing. J Mol Diagn. 2022;24(3):205–218. doi: 10.1016/j.jmoldx.2021.12.001
  • Coppola L, Cianflone A, Grimaldi AM, et al. Biobanking in health care: evolution and future directions. J Transl Med. 2019;17(1):172. doi: 10.1186/s12967-019-1922-3
  • Halldorsson BV, Eggertsson HP, Moore KHS, et al. The sequences of 150,119 genomes in the UK biobank. Nature. 2022;607(7920):732–740. doi: 10.1038/s41586-022-04965-x
  • Leitsalu L, Alavere H, Tammesoo M-L, et al. Linking a population biobank with national health registries—the Estonian experience. J Pers Med. 2015;5(2):96–106. doi: 10.3390/jpm5020096
  • Zhou Y, Ingelman‐Sundberg M, Lauschke V. Worldwide distribution of cytochrome P450 alleles: a meta‐analysis of population‐scale sequencing projects. Clin Pharmacol Ther. 2017;102(4):688–700. doi: 10.1002/cpt.690
  • Santos M, Niemi M, Hiratsuka M, et al. Novel copy-number variations in pharmacogenes contribute to interindividual differences in drug pharmacokinetics. Genet Med. 2018;20(6):622–629. doi: 10.1038/gim.2017.156
  • Zhou Y, Lauschke VM. The genetic landscape of major drug metabolizing cytochrome P450 genes—an updated analysis of population-scale sequencing data. Pharmacogenomics J. 2022;22(5–6):284–293. doi: 10.1038/s41397-022-00288-2
  • Hauser AS, Chavali S, Masuho I, et al. Pharmacogenomics of GPCR drug targets. Cell. 2018;172(1–2):41–54. doi: 10.1016/j.cell.2017.11.033
  • Xiao Q, Zhou Y, Winter S, et al. Germline variant burden in multidrug resistance transporters is a therapy‐specific predictor of survival in breast cancer patients. Int J Cancer. 2020;146(9):2475–2487. doi: 10.1002/ijc.32898
  • Ji Y, Skierka JM, Blommel JH, et al. Preemptive pharmacogenomic testing for precision medicine a comprehensive analysis of five actionable pharmacogenomic genes using next-generation DNA sequencing and a customized CYP2D6 genotyping cascade. J Mol Diagn. 2016;18(3):438–445. doi: 10.1016/j.jmoldx.2016.01.003
  • Devarajan S, Moon I, Ho M, et al. Pharmacogenomic next-generation DNA sequencing: lessons from the identification and functional characterization of variants of unknown significance in CYP2C9 and CYP2C19. Drug Metab Dispos. 2019;47(4):425–435. doi: 10.1124/dmd.118.084269
  • Wang L, Scherer SE, Bielinski SJ, et al. Implementation of preemptive DNA sequence–based pharmacogenomics testing across a large academic medical center: the mayo-baylor RIGHT 10K study. Genet Med. 2022;24(5):1062–1072. doi: 10.1016/j.gim.2022.01.022
  • Tafazoli A, van der Lee M, Swen JJ, et al. Development of an extensive workflow for comprehensive clinical pharmacogenomic profiling: lessons from a pilot study on 100 whole exome sequencing data. Pharmacogenomics J. 2022;22(5–6):276–283. doi: 10.1038/s41397-022-00286-4
  • Gaedigk A, Boone EC, Scherer SE, et al. CYP2C8, CYP2C9, and CYP2C19 characterization using next-generation sequencing and haplotype analysis a GeT-RM collaborative project. J Mol Diagn. 2022;24(4):337–350. doi: 10.1016/j.jmoldx.2021.12.011
  • Siamoglou S, Koromina M, Hishinuma E, et al. Identification and functional validation of novel pharmacogenomic variants using a next-generation sequencing-based approach for clinical pharmacogenomics. Pharmacol Res. 2022;176:106087. doi: 10.1016/j.phrs.2022.106087
  • Ly RC, Schmidt RE, Kiel PJ, et al. Severe capecitabine toxicity associated with a rare DPYD variant identified through whole-genome sequencing. JCO Precis Oncol. 2020;4(4):632–638. doi: 10.1200/PO.20.00067
  • DeMattia E, Silvestri M, Polesel J, et al. Rare genetic variant burden in DPYD predicts severe fluoropyrimidine-related toxicity risk. Biomed Pharmacother. 2022;154:113644. doi: 10.1016/j.biopha.2022.113644
  • Mak ACY, White MJ, Eckalbar WL, et al. Whole-genome sequencing of pharmacogenetic drug response in racially diverse children with asthma. Am J Respir Crit Care Med. 2018;197(12):1552–1564. doi: 10.1164/rccm.201712-2529OC
  • van der Lee M, Allard WG, Vossen RHAM, et al. Toward predicting CYP2D6-mediated variable drug response from CYP2D6 gene sequencing data. Sci Transl Med. 2021;13(603):eabf3637. doi: 10.1126/scitranslmed.abf3637
  • Cruz-Correa OF, Len-Cachn RBR, Barrera-Saldaa HA, et al. Prediction of atorvastatin plasmatic concentrations in healthy volunteers using integrated pharmacogenetics sequencing. Pharmacogenomics. 2017;18(2):121–131. doi: 10.2217/pgs-2016-0072
  • Ramsey LB, Bruun GH, Yang W, et al. Rare versus common variants in pharmacogenetics: SLCO1B1 variation and methotrexate disposition. Genome Res. 2012;22(1):1–8. doi: 10.1101/gr.129668.111
  • Mizzi C, Peters B, Mitropoulou C, et al. Personalized pharmacogenomics profiling using whole-genome sequencing. Pharmacogenomics. 2014;15(9):1223–1234. doi: 10.2217/pgs.14.102
  • Bielinski SJ, Sauver JLS, Olson JE, et al. Cohort profile: the right drug, right dose, right time: using genomic data to individualize treatment protocol (RIGHT protocol). Int J Epidemiol. 2019;49(1):23–24k. doi: 10.1093/ije/dyz123
  • Shrestha S, Zhang C, Jerde CR, et al. Gene-specific variant classifier (DPYD-Varifier) to identify deleterious alleles of dihydropyrimidine dehydrogenase. Clin Pharmacol Ther. 2018;104(4):709–718. doi: 10.1002/cpt.1020
  • Lam YWF. Scientific challenges and implementation barriers to translation of pharmacogenomics in clinical practice. ISRN Pharmacol. 2013;2013:641089. doi: 10.1155/2013/641089
  • Manolio TA, Chisholm RL, Ozenberger B, et al. Implementing genomic medicine in the clinic: the future is here. Genet Med. 2013;15(4):258–267. doi: 10.1038/gim.2012.157
  • Qureshi S, Latif A, Condon L, et al. Understanding the barriers and enablers of pharmacogenomic testing in primary care: a qualitative systematic review with meta-aggregation synthesis. Pharmacogenomics. 2021;23(2):135–154. doi: 10.2217/pgs-2021-0131
  • Russell LE, Zhou Y, Almousa AA, et al. Pharmacogenomics in the era of next generation sequencing – from byte to bedside. Drug Metab Rev. 2021;53(2):253–278. doi: 10.1080/03602532.2021.1909613
  • Chen J, Madireddi S, Nagarkar D, et al. In silico tools for accurate HLA and KIR inference from clinical sequencing data empower immunogenetics on individual-patient and population scales. Brief Bioinform. 2021;22(3):bbaa223. doi: 10.1093/bib/bbaa223
  • Chen X, Shen F, Gonzaludo N, et al. Cyrius: accurate CYP2D6 genotyping using whole-genome sequencing data. Pharmacogenomics J. 2021;21(2):251–261. doi: 10.1038/s41397-020-00205-5
  • van der Lee M, Kriek M, Guchelaar H-J, et al. Technologies for pharmacogenomics: a review. Genes. 2020;11:1456. doi: 10.3390/genes11121456
  • Verbelen M, Weale ME, Lewis CM. Cost-effectiveness of pharmacogenetic-guided treatment: are we there yet? Pharmacogenomics J. 2017;17(5):395–402. doi: 10.1038/tpj.2017.21
  • Zhu Y, Swanson KM, Rojas RL, et al. Systematic review of the evidence on the cost-effectiveness of pharmacogenomics-guided treatment for cardiovascular diseases. Genet Med. 2020;22(3):475–486. doi: 10.1038/s41436-019-0667-y
  • Ghanbarian S, Wong GWK, Bunka M, et al. Cost-effectiveness of pharmacogenomic-guided treatment for major depression. CMAJ. 2023;195(44):E1499–E1508. doi: 10.1503/cmaj.221785
  • Muir P, Li S, Lou S, et al. The real cost of sequencing: scaling computation to keep pace with data generation. Genome Biol. 2016;17(1):53. doi: 10.1186/s13059-016-0917-0
  • Krumm N, Hoffman N. Practical estimation of cloud storage costs for clinical genomic data. Pract Lab Med. 2020;21:e00168. doi: 10.1016/j.plabm.2020.e00168
  • Weymann D, Laskin J, Roscoe R, et al. The cost and cost trajectory of whole‐genome analysis guiding treatment of patients with advanced cancers. Mol Genet Genomic Med. 2017;5(3):251–260. doi: 10.1002/mgg3.281
  • Lemke LK, Alam B, Williams R, et al. Reimbursement of pharmacogenetic tests at a tertiary academic medical center in the United States. Front Pharmacol. 2023;14:1179364. doi: 10.3389/fphar.2023.1179364
  • Vozikis A, Cooper DN, Mitropoulou C, et al. Test pricing and reimbursement in genomic medicine: towards a general strategy. Public Health Genomics. 2016;19(6):352–363. doi: 10.1159/000449152