Next Article in Journal
Genetic Distinctness and Diversity of American Aberdeen Cattle Compared to Common Beef Breeds in the United States
Next Article in Special Issue
G-Protein Signaling Modulator 2 as a Potential Biomarker in Colorectal Cancer: Integrative Analysis Using Genetic Profiling and Pan-Cancer Studies
Previous Article in Journal
Population Pharmacogenomics for Health Equity
Previous Article in Special Issue
Personal Genomes in Practice: Exploring Citizen and Healthcare Professionals’ Perspectives on Personalized Genomic Medicine and Personal Health Data Spaces Using a Mixed-Methods Design
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Implementation of Pharmacogenetics in First-Line Care: Evaluation of Its Use by General Practitioners

1
Department of Clinical Chemistry, Erasmus MC University Medical Center, 3015 GD Rotterdam, The Netherlands
2
Department of Hospital Pharmacy, Erasmus MC University Medical Center, 3015 CN Rotterdam, The Netherlands
3
Department of Clinical Chemistry, Result Laboratory, 3318 AT Dordrecht, The Netherlands
4
Department of General Practice, Erasmus MC University Medical Center, 3015 GD Rotterdam, The Netherlands
*
Author to whom correspondence should be addressed.
Genes 2023, 14(10), 1841; https://doi.org/10.3390/genes14101841
Submission received: 28 August 2023 / Revised: 16 September 2023 / Accepted: 20 September 2023 / Published: 22 September 2023
(This article belongs to the Special Issue Genetics and Pharmacogenetics in Primary Care)

Abstract

:
Pharmacogenetics (PGx) can explain/predict drug therapy outcomes. There is, however, unclarity about the use and usefulness of PGx in primary care. In this study, we investigated PGx tests ordered by general practitioners (GPs) in 2021 at Dept. Clinical Chemistry, Erasmus MC, and analyzed the gene tests ordered, drugs/drug groups, reasons for testing and single-gene versus panel testing. Additionally, a survey was sent to 90 GPs asking about their experiences and barriers to implementing PGx. In total, 1206 patients and 6300 PGx tests were requested by GPs. CYP2C19 was requested most frequently (17%), and clopidogrel was the most commonly indicated drug (23%). Regarding drug groups, antidepressants (51%) were the main driver for requesting PGx, followed by antihypertensives (26%). Side effects (79%) and non-response (27%) were the main indicators. Panel testing was preferred over single-gene testing. The survey revealed knowledge on when and how to use PGx as one of the main barriers. In conclusion, PGx is currently used by GPs in clinical practice in the Netherlands. Side effects are the main reason for testing, which mostly involves antidepressants. Lack of knowledge is indicated as a major barrier, indicating the need for more education on PGx for GPs.

1. Introduction

Pharmacogenetics is the field of analyzing genetic polymorphisms in order to explain or predict drug therapy outcomes. Mostly, this is focused on analyzing single-nucleotide polymorphisms (SNPs) in genes encoding drug-metabolizing enzymes and/or drug transporters. The rationale behind using DNA information to guide drug therapy lies in the fact that only 30–60% of patients respond properly to treatment with antidepressants, antipsychotics, β-blockers and statins [1], and that 5–7% of hospitalizations are due to adverse drug reactions (ADRs) [2,3,4]. Having DNA information available can help to reduce adverse drug reactions by 30%, as was recently demonstrated in the PREPARE study [5]. Guidelines on how to dose drugs based on genetic information are currently available for several gene–drug combinations (www.cpicpgx.org, accessed on 1 August 2023). In total, there are now 28 CPIC guidelines available, addressed in 43 publications: antidepressants/CYP2D6 and CYP2C19 [6,7]; SSRIs/CYP2D6 and CYP2C19 [8]; SSRIs/CYP2D6, CYP2C19, CYP2B6, SLC6A4 and HTR2A [9]; codeine/CYP2D6 [10,11]; opioids/CYP2D6, OPRM1 and COMT [12]; atomoxetine/CYP2D6 [13]; statins/SLCO1B1, ABCG2 and CYP2C9 [14,15,16]; warfarin/CYP2C9 and VKORC1 [17,18]; clopidogrel/CYP2C19 [19,20,21]; proton pump inhibitors (PPI)/CYP2C19 [22]; odansetron/CYP2D6 [23]; tamoxifen/CYP2D6 [24]; voriconazol/CYP2C19 [25]; non-steroidal anti-inflammatory drugs (NSAIDs)/CYP2C9 [26]; fluoropyrimidines/DPYD [27,28]; efavirenz/CYP2B6 [29]; phenytoin/CYP2C9 and HLA-B [30,31]; rasburicase/G6PD [32,33]; tacrolimus/CYP3A5 [34]; carbamazepine/HLA-B [35,36]; abacavir/HLA-B [37,38]; allopurinol/HLA-B [39,40]; PEG-interferon α/IFNL3(IL28B) [41]; aminoglycosides/MT-RNR1 [42]; volatile anesthetic agents and succinylcholine/RYR1 and CACNA1S [43]); thiopurines/TPMT (and NUDT15) [44,45,46]); atazanavir/UGT1A1 [47]; and ivacaftor/CFTR [48]. In addition, the FDA has published a table of pharmacogenetic associations (https://www.fda.gov/medical-devices/precision-medicine/table-pharmacogenetic-associations, accessed on 1 August 2023), divided into three sections: (1) associations for which the data support therapeutic management recommendations; (2) associations for which the data indicate a potential impact on safety or response; and (3) associations for which the data demonstrate a potential impact on pharmacokinetic properties only. The CPIC guidelines and the FDA association table do not, however, overlap completely [49]. The Dutch Pharmacogenetics Working Group (DPWG), a working group of the Royal Dutch Society for Advancing Pharmacotherapy (KNMP: Koninklijke Maatschappij ter bevordering der Pharmacie), saw the first publication of their guidelines in 2008 [50], with an update in 2011 [51]. Currently, the DPWG has 108 recommendations (www.pharmgkb.org, accessed on 1 August 2023), many of which were also published separately with the background information on how the advice was reached: SSRIs/CYP2D6 and CYP2C19 [52], antipsychotics/CYP2D6, CYP3A4 and CYP1A2 [53], irinotecan/UGT1A1 [54], fluoropyrimidines/DPYD [55], atomoxetine and methylphenidate/CYP2D6 and COMT [56], opioids/CYP2D6 [57], allopurinol/ABCG2 and HLA-B [58]), and folic acid and methotrexate/MTHFR [58]. In the Netherlands, these guidelines are in use and every pharmacist has access to genotype-based dosing recommendations. Based on the increased interest and availability of guidance documents, an increasing number of laboratories are nowadays offering pharmacogenetic testing.
As to the position of pharmacogenetic testing in the healthcare setting, there has been debate about its position in first-line care. Whereas pharmacogenetics undergoes adequate implementation now in second-line care for a small group of high-risk medications (clopidogrel/CYP2C19, abacavir/HLA-B*5701, and capecitabine/DPYD), the majority of medicines are being prescribed in primary care. In 2015, 27 out of the 80 medicines for which the DPWG had guidelines available at the time were regularly prescribed in first-line care [59]. Several publications on the potential of pharmacogenetic testing in first-line care have been published, also addressing pharmacist-initiated testing for GPs [60,61]. A recent study, estimating the potential impact of implementing pre-emptive pharmacogenetic testing in primary care in the UK, demonstrated that 19–21% of all new prescriptions for 56 drugs (5.2–5.7 million prescriptions/year in total) were, in fact, involving an actionable gene–drug interaction, according to CPIC and DPWG guidelines, showing the high potential of PGx testing in primary care [62]. A comparable study, performed in the Netherlands, reported 23.6% (n = 856,002) of all new prescriptions for 45 drugs given by GPs as actionable [63]. An initiative to identify potential barriers to implementing pharmacogenetics among primary care physicians in the Netherlands, however, was not successful, due to low response rates [64]. At Erasmus MC at the Dept. Clinical Chemistry, we have been offering clinical diagnostic testing for pharmacogenetics since 2005, being an expert center recognized by the International Federation of Clinical Chemistry (IFCC) since 2007. Starting in 2005, we have seen a growth in the number of test requests from 25 tests in 2005 up to 23,284 tests in 2021, representing 8500 patients in total.
The aim of this present study was to analyze the number of requests sent in by general practitioners (GPs), and we subsequently used our real-world clinical data to identify the genes, as well as the medications, of interest. Furthermore, we wanted to investigate the reasons for requesting a PGx test. Finally, we were interested in experiences and barriers so we distributed a questionnaire to GPs in our academic GP network associated with Erasmus MC (named “PrimEUR”), to ask them about their experiences and barriers for using PGx in primary care.

2. Materials and Methods

In the period January 1 to December 31, 2021, 8500 PGx request forms in total, representing 8500 patients, were received at Dept. Clinical Chemistry, Erasmus MC, representing 23,284 test requests. Erasmus MC offers 26 individual genes for testing, but also allows standardized panel testing, including DNA panel—basic; DNA panel—extended; a psychiatry panel; cardiac panel; pain panel and oncology panel (Table 1). The mean turnaround time was 7 working days, except for individual CYP2C19/clopidogrel testing requests, which had a turnaround time of 24 h. Clinicians could choose whether to send in blood samples, or have a buccal swab kit sent directly to their patients.
The GP survey was conducted among 90 GPs from the Erasmus MC Academic GP network PrimEUR, and sent out by email in February 2022, with a reminder after one week. The GPs were not selected based on whether or not they had experiences with PGx testing. The survey contained 13 questions and was divided into questions regarding characteristics of the participating GP, the application of PGx and the sources that were used to obtain information about PGx. The survey contained multiple choice questions, but also included an “other…” option. The survey closed 3 weeks after the first invite. Analyses were performed using IBM SPSS Statistics 25 for Windows. Descriptive statistics were used to summarize each question.

3. Results

3.1. Materials, Request Forms and Genes Requested

From the total of 8500 patients sent to Erasmus MC for PGx testing, 1281 patients (15%) were sent in by GPs, either directly (76%) or through specific GP laboratories (24%). For analysis, 75 test request forms were excluded: 8 forms were not available, 12 were duplicates and 55 forms were not sent in on behalf of a different physician, leaving 1206 patients (14%), representing 6301 out of 23,284 test requests (27%). From the 1206 request forms obtained from primary care, 65% were sent in as buccal swabs and 35% as blood samples. The total number of individual tests was 6301, resulting in an average of 5.2 tests/patient. The top two genes were CYP2C19 (17%) and CYP2D6 (16%) (Figure 1a). PGx panels were requested in 795/1206 patients (66%), representing 5257/6301 tests (83%). Among these, 43% were for the DNA panel—extended; 24% were for the psychiatry panel; and 15% were for the DNA panel—basic (15%) (Figure 1b). Individual gene testing (so outside of a panel) was highest for CYP2D6 (272/978; 27.9%) and CYP2C19 (206/895; 23.7%), and lowest for VKORC1 (2/493; 0.4%), OPRM1 (3/91; 3.3%), ABCB1 (2/64; 3.1%) and SLCO1B1 (16/510; 3.1%).

3.2. Drugs

The majority of GPs (706/1206; 59%) listed on the request form which drugs (groups) the PGx test was requested for: 146 (12%) listed both the drug and drug group, 505 (42%) listed only the drug and 55 (4.5%) listed only the drug group. In total, 237 different drugs were mentioned on the requested forms, representing 30 different drug groups. PGx testing was most often requested for clopidogrel, at 23% (147/639), followed by citalopram (8%), amitriptyline (6.8%) and metoprolol (6.8%). In total, 50 drugs were mentioned in at least 1% of requests (Table 2).
Regarding drug groups, antidepressants were most frequently mentioned (70% of forms; 850/1206), followed by antihypertensive (26%) and antiplatelet drugs (26%) (Figure 2).

3.3. Reasons for Requesting PGx Testing

Reasons for requesting PGx testing, in 70% of patients (850/1206), were as follows: (a) low drug plasma concentrations, (b) high drug plasma concentrations, (c) side effects, (d) non-response, (e) prior to therapy, or (f) other. On 195/1206 forms (16%), more than one indication was given. The most frequent reason for requesting a PGx test was “side effects”, as stated in 79% of request forms (669/846), followed by “non-response” (27%) and “prior to therapy” (13%). In addition to the standard choices, in 2.8% of request forms (24/857), “PGx-variant in family member” was mentioned (Figure 3).

3.4. Results of the Survey on PGx

The survey was returned by 29 out of 90 GPs, resulting in a response rate of 32%. Most responders (20/29; 69%) were working in a group practice/healthcare center. Regarding their work experience, 2/29 (7%) worked for 0–5 years as GPs, 14/29 (48%) worked for 5–20 years and 13/29 (45%) worked for more than 20 years. Self-reported familiarity with PGx ranged from “slightly familiar” (11/29; 38%) and “moderately familiar” (7/29; 24%) to “familiar” (10/29; 34%) and “very familiar” (1/29; 3%). None of the GPs reported being “unfamiliar” with PGx testing. As to how the GPs learned about PGx, most respondents reported a refresher course/magazine (20/29; 69%) and patient cases (13/29; 45%). In total, 16/29 respondents (55%) had requested a PGx test in the last year, with the majority (10/16; 63%) requesting a basic DNA passport, followed by requests for CYP2C19 (7/14; 50%) and CYP2D6 (6/14; 38%). On the question regarding the barriers experienced in using PGx, 59% of GPs reported “unclear when to test”, “cost for patient” (48%) and “lack of experience” (45%). Other barriers reflected unclarity in testing or test interpretation (Figure 4).
Regarding the sources consulted by GPs with regard to PGx, pharmacists were mentioned mostly as the main source (34%), followed by publications of the Dutch College of General Practitioners (NHG) (21%) and the website www.farmacogenetica.nl (accessed on 1 August 2023) (17%). The laboratory offering PGx testing, the pharmacology compendium ‘Farmacotherapeutisch Kompas’ and the GP genetics website (www.huisartsengenetica.nl, accessed on 1 August 2023) each scored 10% for consulted sources. GPs did not use information from the drug label, PharmGKB, CPIC, DPWG scientific literature or the KNMP PGx database.

4. Discussion

Our results indicate that PGx testing is being taken up by GPs and has a place in clinical practice, both through the total number of patients (1281) and the number of test requests (6301) in 2021. It is important to note that the GPs ordering these test were GPs from all over the Netherlands, and were not restricted to Erasmus MC or GPs from the Erasmus MC-associated PrimEUR Network. Buccal swab testing (75%) was clearly preferred above blood sampling (25%), compared to the percentage of 20% of buccal swabs when taking all PGx test received at our department in 2021 into account. There is, in addition, a preference by GPs for requesting multiple tests per patient, resulting in an average of 5.2 tests/patient. This is higher than the number of hospital-based test requests, which have an average of 2.3 tests/patient (Dept. Clinical Chemistry, PGx test request data 2021). Most tests ordered by GPs were requested inside a panel, ranging from 72% for CYP2D6 to 99.6% for VKORC1. Apparently, statin use itself was not regarded as a specific reason for requesting a PGx test, as seen in the relatively low number of requests for individual SLCO1B1 tests. It cannot be excluded, however, that side effects of statins triggered PGx panel testing. Interestingly, DNA panel—extended was the most highly requested panel test (43%). This, in our opinion, could reflect that, when considering a PGx test, a total overview is preferred over a smaller, perhaps more focused panel by patients and/or GPs. Alternatively, this could also reflect the limited experience of GPs in terms of which test to order.
Although from individual drugs, clopidogrel was most often mentioned (23%), on a group level, antidepressants represented the major reason (51%) for requesting a PGx test. The other drug groups in the top seven were antihypertensives, antiplatelet drugs, opioids, statins, β-blockers and proton pump inhibitors. In total, 30 different drug groups were indicated. From the individual drugs, it is interesting that of the drugs mentioned in >1% of request forms, almost half (27/50) do not have a direct indication for PGx testing based on DPWG guidelines. This may possibly reflect that communication with physicians on when PGx testing can be helpful, should be improved. Otherwise, it could reflect that physicians want to provide all relevant information on the medication a patient is on. When a non-guideline drug was the only reason for asking for PGx testing, no test was performed. In other instances, the gene was tested for which an appropriate test and corresponding dosing guideline was available. The medication mentioned on the form was, however, still included in our overview.
As for reasons for PGx testing, side effects was by far the most reported (79%). This reflects the Dutch GP guideline that indicates that PGx testing should be considered only after side effects are experienced, or ineffective therapy, and not prior to therapy. Yet, 13% of forms still indicated “prior to therapy” as the reason. Since we did find combinations where both “side effects” and “prior to therapy”, or “non-response” and “prior to therapy” boxes were checked, it seems that this question was multi-interpretable. Indeed, after side effects (or non-response), a new drug is to be prescribed, meaning that both options on the form could be indicated. This also caused the total percentage for this question to exceed 100%. High/low plasma drug concentrations were hardly mentioned as a reason for requesting PGx testing (2.8%).
Regarding the survey among GPs, the response rate of 30% (29/90) was above expectations, since an earlier study on Dutch GPs was based on only eight interviews, which was reported as a very low response rate [64]. The GPs that were approached by us were part of an academic network (PrimEUR), which is why the response rate may have been higher in our case. The distribution of working experience in the interviewed GPs of the PrimEUR group ranged from relatively new (7%) to more than 30 years of experience (14%), and was quite balanced over the different years of experience. Most GPs were to some extent, familiar with PGx, with only one reporting “very familiar”. Most respondents had asked for a PGx test in the last year, reflecting the answers on familiarity. Consulting a pharmacist, refresher courses, magazines, Dutch literature, patient cases and congresses were reported as main sources of knowledge on PGx, whereas international literature scored 0%. As to barriers experienced, lack of knowledge on when to test (59%), which test to request (41%), how to order a test (34%), or lack of knowledge on how to interpret test results (31%) stood out, clearly reflecting the need for more information for GPs on how to use PGx.
As a second argument, concerns with costs for the patient (48%), or unclarity in terms of reimbursement (10%), were mentioned as barriers. In the Netherlands, PGx testing is reimbursed by insurance companies when requested by a GP, provided there is a valid reason for requesting the test, this usually being the occurrence of side effects or non-response. However, each patient has a maximum amount that he/she needs to contribute each year out of pocket, which is usually around EUR 385, meaning that there may still be costs for the patient when this maximum amount is not yet reached. Because of this policy, Erasmus MC experiences a doubling of the number of PGx panel test requests in the last 3 months of the year, when most patients have reached this maximum through their own contribution, and PGx testing does not result in extra costs for the patient. It is important to notice that the GPs in the PrimEUR group, because of their affiliation with academia, are not representative of the average GP. The fact that in this selected group a lack of knowledge is mentioned as a barrier to implementing PGx, again stresses the importance of providing more tools and education for GPs on PGx.
In a recent study performed in the US, comparing the attitudes of healthcare providers to PGx testing, it was shown that primary care providers overwhelmingly perceived PGx to be useful [65], as is the case in our own experience with teaching GPs. In fact, primary care providers were more likely to rate PGx testing as useful compared to specialty care providers [65], indicating that PGx is a tool that fits the needs of GPs more than those of specialty care providers. In a recent survey among thirty-four clinicians in the US that had minimal experience with PGx and PGx resources, it was shown that the expectation among participants was that PGx indeed has the ability to improve medication-related outcomes, but that many lack the confidence to apply PGx results in their practice [66]. This again indicates that GP education on PGx is of high importance. In this regard, pharmacist-initiated PGx testing could be of value, as revealed by van der Wouden et al. [61] via an investigation on the feasibility and real-world impact of this approach. In their prospective study, they provided community pharmacists the opportunity to pre-emptively request a panel of eight pharmacogenes to guide drug dispensing with a clinical decision support system for 200 primary care patients. Pre-emptive testing would relieve the pressure on GPs contemplating which PGx test should be ordered, since results are already available. In a mean 2.5-year follow up in this study, it was shown that PGx panel reports were successfully recorded in 96% of pharmacists, and in 67% of GP electronical medical records. In 97% of cases, this enabled patients to (re)use PGx panel results at least once. In total, 24.2% of prescriptions had actionable drug–gene interactions. The authors concluded that pre-emptive panel-based PGx testing is feasible and that real-world impact is believed to be substantial in primary care. These findings confirmed the findings of an earlier study in the Netherlands on the estimated nationwide impact of implementing a pre-emptive PGx panel approach to guide drug prescribing in primary care, where it was shown that 23.6% of all new prescriptions were, in fact, drug–gene interactions and that dose adjustments or switching to another drug would follow in 5.4% of all new prescriptions [63].

5. Conclusions

PGx testing does have a role in GP practice, and is indeed also already applied in the Netherlands by this group of healthcare practitioners. There is a preference for buccal swabs as a testing material over blood sampling. The occurrence of side effects is most prominently mentioned as the reason for requesting PGx testing, and the drug group mostly involved is antidepressants. Lack of knowledge on PGx is still indicated as a major barrier to implementing PGx in primary care, stressing the need for developing additional tools and education for GPs on PGx.

Author Contributions

Conceptualization: M.M., M.S. and R.H.N.v.S.; formal analysis: D.v.d.D., M.M. and M.S.; methodology: M.M., M.S. and R.H.N.v.S.; investigation: D.v.d.D., G.B., M.M., M.S. and R.H.N.v.S.; data curation: D.v.d.D., M.S. and M.M.; writing—original draft: D.v.d.D.; writing—review and editing: M.S., M.M., B.C.P.K., G.B., P.B. and R.H.N.v.S.; supervision: M.M., B.C.P.K. and R.H.N.v.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Eichelbaum, M.; Ingelman-Sundberg, M.; Evans, W.E. Pharmacogenomics and individualized drug therapy. Annu. Rev. Med. 2006, 57, 119–137. [Google Scholar] [CrossRef] [PubMed]
  2. Lazarou, J.; Pomeranz, B.H.; Corey, P.N. Incidence of adverse drug reactions in hospitalized patients: A meta-analysis of prospective studies. JAMA 1998, 279, 1200–1205. [Google Scholar] [CrossRef]
  3. Green, C.F.; Mottram, D.R.; Rowe, P.H.; Pirmohamed, M. Adverse drug reactions as a cause of admission to an acute medical assessment unit: A pilot study. J. Clin. Pharm. Ther. 2000, 25, 355–361. [Google Scholar] [CrossRef] [PubMed]
  4. Pirmohamed, M.; James, S.; Meakin, S.; Green, C.; Scott, A.K.; Walley, T.J.; Farrar, K.; Park, B.K.; Breckenridge, A.M. Adverse drug reactions as cause of admission to hospital: Prospective analysis of 18 820 patients. BMJ 2004, 329, 15–19. [Google Scholar] [CrossRef] [PubMed]
  5. Swen, J.J.; van der Wouden, C.H.; Manson, L.E.; Abdullah-Koolmees, H.; Blagec, K.; Blagus, T.; Bohringer, S.; Cambon-Thomsen, A.; Cecchin, E.; Cheung, K.C.; et al. A 12-gene pharmacogenetic panel to prevent adverse drug reactions: An open-label, multicentre, controlled, cluster-randomised crossover implementation study. Lancet 2023, 401, 347–356. [Google Scholar] [CrossRef] [PubMed]
  6. Hicks, J.K.; Swen, J.J.; Thorn, C.F.; Sangkuhl, K.; Kharasch, E.D.; Ellingrod, V.L.; Skaar, T.C.; Muller, D.J.; Gaedigk, A.; Stingl, J.C.; et al. Clinical Pharmacogenetics Implementation Consortium guideline for CYP2D6 and CYP2C19 genotypes and dosing of tricyclic antidepressants. Clin. Pharmacol. Ther. 2013, 93, 402–408. [Google Scholar] [CrossRef]
  7. Hicks, J.K.; Sangkuhl, K.; Swen, J.J.; Ellingrod, V.L.; Muller, D.J.; Shimoda, K.; Bishop, J.R.; Kharasch, E.D.; Skaar, T.C.; Gaedigk, A.; et al. Clinical pharmacogenetics implementation consortium guideline (CPIC) for CYP2D6 and CYP2C19 genotypes and dosing of tricyclic antidepressants: 2016 update. Clin. Pharmacol. Ther. 2017, 102, 37–44. [Google Scholar] [CrossRef]
  8. Hicks, J.K.; Bishop, J.R.; Sangkuhl, K.; Muller, D.J.; Ji, Y.; Leckband, S.G.; Leeder, J.S.; Graham, R.L.; Chiulli, D.L.; Llerena, A.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2D6 and CYP2C19 Genotypes and Dosing of Selective Serotonin Reuptake Inhibitors. Clin. Pharmacol. Ther. 2015, 98, 127–134. [Google Scholar] [CrossRef]
  9. Bousman, C.A.; Stevenson, J.M.; Ramsey, L.B.; Sangkuhl, K.; Hicks, J.K.; Strawn, J.R.; Singh, A.B.; Ruano, G.; Mueller, D.J.; Tsermpini, E.E.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2D6, CYP2C19, CYP2B6, SLC6A4, and HTR2A Genotypes and Serotonin Reuptake Inhibitor Antidepressants. Clin. Pharmacol. Ther. 2023, 114, 51–68. [Google Scholar] [CrossRef]
  10. Crews, K.R.; Gaedigk, A.; Dunnenberger, H.M.; Klein, T.E.; Shen, D.D.; Callaghan, J.T.; Kharasch, E.D.; Skaar, T.C.; Clinical Pharmacogenetics Implementation Consortium. Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for codeine therapy in the context of cytochrome P450 2D6 (CYP2D6) genotype. Clin. Pharmacol. Ther. 2012, 91, 321–326. [Google Scholar] [CrossRef]
  11. Crews, K.R.; Gaedigk, A.; Dunnenberger, H.M.; Leeder, J.S.; Klein, T.E.; Caudle, K.E.; Haidar, C.E.; Shen, D.D.; Callaghan, J.T.; Sadhasivam, S.; et al. Clinical Pharmacogenetics Implementation Consortium guidelines for cytochrome P450 2D6 genotype and codeine therapy: 2014 update. Clin. Pharmacol. Ther. 2014, 95, 376–382. [Google Scholar] [CrossRef] [PubMed]
  12. Crews, K.R.; Monte, A.A.; Huddart, R.; Caudle, K.E.; Kharasch, E.D.; Gaedigk, A.; Dunnenberger, H.M.; Leeder, J.S.; Callaghan, J.T.; Samer, C.F.; et al. Clinical Pharmacogenetics Implementation Consortium Guideline for CYP2D6, OPRM1, and COMT Genotypes and Select Opioid Therapy. Clin. Pharmacol. Ther. 2021, 110, 888–896. [Google Scholar] [CrossRef] [PubMed]
  13. Brown, J.T.; Bishop, J.R.; Sangkuhl, K.; Nurmi, E.L.; Mueller, D.J.; Dinh, J.C.; Gaedigk, A.; Klein, T.E.; Caudle, K.E.; McCracken, J.T.; et al. Clinical Pharmacogenetics Implementation Consortium Guideline for Cytochrome P450 (CYP)2D6 Genotype and Atomoxetine Therapy. Clin. Pharmacol. Ther. 2019, 106, 94–102. [Google Scholar] [CrossRef] [PubMed]
  14. Wilke, R.A.; Ramsey, L.B.; Johnson, S.G.; Maxwell, W.D.; McLeod, H.L.; Voora, D.; Krauss, R.M.; Roden, D.M.; Feng, Q.; Cooper-Dehoff, R.M.; et al. The clinical pharmacogenomics implementation consortium: CPIC guideline for SLCO1B1 and simvastatin-induced myopathy. Clin. Pharmacol. Ther. 2012, 92, 112–117. [Google Scholar] [CrossRef]
  15. Ramsey, L.B.; Johnson, S.G.; Caudle, K.E.; Haidar, C.E.; Voora, D.; Wilke, R.A.; Maxwell, W.D.; McLeod, H.L.; Krauss, R.M.; Roden, D.M.; et al. The clinical pharmacogenetics implementation consortium guideline for SLCO1B1 and simvastatin-induced myopathy: 2014 update. Clin. Pharmacol. Ther. 2014, 96, 423–428. [Google Scholar] [CrossRef]
  16. Cooper-DeHoff, R.M.; Niemi, M.; Ramsey, L.B.; Luzum, J.A.; Tarkiainen, E.K.; Straka, R.J.; Gong, L.; Tuteja, S.; Wilke, R.A.; Wadelius, M.; et al. The Clinical Pharmacogenetics Implementation Consortium Guideline for SLCO1B1, ABCG2, and CYP2C9 genotypes and Statin-Associated Musculoskeletal Symptoms. Clin. Pharmacol. Ther. 2022, 111, 1007–1021. [Google Scholar] [CrossRef] [PubMed]
  17. Johnson, J.A.; Gong, L.; Whirl-Carrillo, M.; Gage, B.F.; Scott, S.A.; Stein, C.M.; Anderson, J.L.; Kimmel, S.E.; Lee, M.T.; Pirmohamed, M.; et al. Clinical Pharmacogenetics Implementation Consortium Guidelines for CYP2C9 and VKORC1 genotypes and warfarin dosing. Clin. Pharmacol. Ther. 2011, 90, 625–629. [Google Scholar] [CrossRef] [PubMed]
  18. Johnson, J.A.; Caudle, K.E.; Gong, L.; Whirl-Carrillo, M.; Stein, C.M.; Scott, S.A.; Lee, M.T.; Gage, B.F.; Kimmel, S.E.; Perera, M.A.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Pharmacogenetics-Guided Warfarin Dosing: 2017 Update. Clin. Pharmacol. Ther. 2017, 102, 397–404. [Google Scholar] [CrossRef] [PubMed]
  19. Scott, S.A.; Sangkuhl, K.; Gardner, E.E.; Stein, C.M.; Hulot, J.S.; Johnson, J.A.; Roden, D.M.; Klein, T.E.; Shuldiner, A.R.; Clinical Pharmacogenetics Implementation Consortium. Clinical Pharmacogenetics Implementation Consortium guidelines for cytochrome P450-2C19 (CYP2C19) genotype and clopidogrel therapy. Clin. Pharmacol. Ther. 2011, 90, 328–332. [Google Scholar] [CrossRef]
  20. Scott, S.A.; Sangkuhl, K.; Stein, C.M.; Hulot, J.S.; Mega, J.L.; Roden, D.M.; Klein, T.E.; Sabatine, M.S.; Johnson, J.A.; Shuldiner, A.R.; et al. Clinical Pharmacogenetics Implementation Consortium guidelines for CYP2C19 genotype and clopidogrel therapy: 2013 update. Clin. Pharmacol. Ther. 2013, 94, 317–323. [Google Scholar] [CrossRef]
  21. Lee, C.R.; Luzum, J.A.; Sangkuhl, K.; Gammal, R.S.; Sabatine, M.S.; Stein, C.M.; Kisor, D.F.; Limdi, N.A.; Lee, Y.M.; Scott, S.A.; et al. Clinical Pharmacogenetics Implementation Consortium Guideline for CYP2C19 Genotype and Clopidogrel Therapy: 2022 Update. Clin. Pharmacol. Ther. 2022, 112, 959–967. [Google Scholar] [CrossRef]
  22. Lima, J.J.; Thomas, C.D.; Barbarino, J.; Desta, Z.; Van Driest, S.L.; El Rouby, N.; Johnson, J.A.; Cavallari, L.H.; Shakhnovich, V.; Thacker, D.L.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2C19 and Proton Pump Inhibitor Dosing. Clin. Pharmacol. Ther. 2021, 109, 1417–1423. [Google Scholar] [CrossRef]
  23. Bell, G.C.; Caudle, K.E.; Whirl-Carrillo, M.; Gordon, R.J.; Hikino, K.; Prows, C.A.; Gaedigk, A.; Agundez, J.; Sadhasivam, S.; Klein, T.E.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for CYP2D6 genotype and use of ondansetron and tropisetron. Clin. Pharmacol. Ther. 2017, 102, 213–218. [Google Scholar] [CrossRef]
  24. Goetz, M.P.; Sangkuhl, K.; Guchelaar, H.J.; Schwab, M.; Province, M.; Whirl-Carrillo, M.; Symmans, W.F.; McLeod, H.L.; Ratain, M.J.; Zembutsu, H.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2D6 and Tamoxifen Therapy. Clin. Pharmacol. Ther. 2018, 103, 770–777. [Google Scholar] [CrossRef]
  25. Moriyama, B.; Obeng, A.O.; Barbarino, J.; Penzak, S.R.; Henning, S.A.; Scott, S.A.; Agundez, J.; Wingard, J.R.; McLeod, H.L.; Klein, T.E.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guidelines for CYP2C19 and Voriconazole Therapy. Clin. Pharmacol. Ther. 2017, 102, 45–51. [Google Scholar] [CrossRef]
  26. Theken, K.N.; Lee, C.R.; Gong, L.; Caudle, K.E.; Formea, C.M.; Gaedigk, A.; Klein, T.E.; Agundez, J.A.G.; Grosser, T. Clinical Pharmacogenetics Implementation Consortium Guideline (CPIC) for CYP2C9 and Nonsteroidal Anti-Inflammatory Drugs. Clin. Pharmacol. Ther. 2020, 108, 191–200. [Google Scholar] [CrossRef]
  27. Caudle, K.E.; Thorn, C.F.; Klein, T.E.; Swen, J.J.; McLeod, H.L.; Diasio, R.B.; Schwab, M. Clinical Pharmacogenetics Implementation Consortium guidelines for dihydropyrimidine dehydrogenase genotype and fluoropyrimidine dosing. Clin. Pharmacol. Ther. 2013, 94, 640–645. [Google Scholar] [CrossRef]
  28. Amstutz, U.; Henricks, L.M.; Offer, S.M.; Barbarino, J.; Schellens, J.H.M.; Swen, J.J.; Klein, T.E.; McLeod, H.L.; Caudle, K.E.; Diasio, R.B.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Dihydropyrimidine Dehydrogenase Genotype and Fluoropyrimidine Dosing: 2017 Update. Clin. Pharmacol. Ther. 2018, 103, 210–216. [Google Scholar] [CrossRef]
  29. Desta, Z.; Gammal, R.S.; Gong, L.; Whirl-Carrillo, M.; Gaur, A.H.; Sukasem, C.; Hockings, J.; Myers, A.; Swart, M.; Tyndale, R.F.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2B6 and Efavirenz-Containing Antiretroviral Therapy. Clin. Pharmacol. Ther. 2019, 106, 726–733. [Google Scholar] [CrossRef]
  30. Caudle, K.E.; Rettie, A.E.; Whirl-Carrillo, M.; Smith, L.H.; Mintzer, S.; Lee, M.T.; Klein, T.E.; Callaghan, J.T.; Clinical Pharmacogenetics Implementation Consortium. Clinical pharmacogenetics implementation consortium guidelines for CYP2C9 and HLA-B genotypes and phenytoin dosing. Clin. Pharmacol. Ther. 2014, 96, 542–548. [Google Scholar] [CrossRef]
  31. Karnes, J.H.; Rettie, A.E.; Somogyi, A.A.; Huddart, R.; Fohner, A.E.; Formea, C.M.; Ta Michael Lee, M.; Llerena, A.; Whirl-Carrillo, M.; Klein, T.E.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2C9 and HLA-B Genotypes and Phenytoin Dosing: 2020 Update. Clin. Pharmacol. Ther. 2021, 109, 302–309. [Google Scholar] [CrossRef] [PubMed]
  32. Relling, M.V.; McDonagh, E.M.; Chang, T.; Caudle, K.E.; McLeod, H.L.; Haidar, C.E.; Klein, T.; Luzzatto, L.; Clinical Pharmacogenetics Implementation Consortium. Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for rasburicase therapy in the context of G6PD deficiency genotype. Clin. Pharmacol. Ther. 2014, 96, 169–174. [Google Scholar] [CrossRef]
  33. Gammal, R.S.; Pirmohamed, M.; Somogyi, A.A.; Morris, S.A.; Formea, C.M.; Elchynski, A.L.; Oshikoya, K.A.; McLeod, H.L.; Haidar, C.E.; Whirl-Carrillo, M.; et al. Expanded Clinical Pharmacogenetics Implementation Consortium Guideline for Medication Use in the Context of G6PD Genotype. Clin. Pharmacol. Ther. 2023, 113, 973–985. [Google Scholar] [CrossRef] [PubMed]
  34. Birdwell, K.A.; Decker, B.; Barbarino, J.M.; Peterson, J.F.; Stein, C.M.; Sadee, W.; Wang, D.; Vinks, A.A.; He, Y.; Swen, J.J.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guidelines for CYP3A5 Genotype and Tacrolimus Dosing. Clin. Pharmacol. Ther. 2015, 98, 19–24. [Google Scholar] [CrossRef] [PubMed]
  35. Leckband, S.G.; Kelsoe, J.R.; Dunnenberger, H.M.; George, A.L., Jr.; Tran, E.; Berger, R.; Muller, D.J.; Whirl-Carrillo, M.; Caudle, K.E.; Pirmohamed, M.; et al. Clinical Pharmacogenetics Implementation Consortium guidelines for HLA-B genotype and carbamazepine dosing. Clin. Pharmacol. Ther. 2013, 94, 324–328. [Google Scholar] [CrossRef]
  36. Phillips, E.J.; Sukasem, C.; Whirl-Carrillo, M.; Muller, D.J.; Dunnenberger, H.M.; Chantratita, W.; Goldspiel, B.; Chen, Y.T.; Carleton, B.C.; George, A.L., Jr.; et al. Clinical Pharmacogenetics Implementation Consortium Guideline for HLA Genotype and Use of Carbamazepine and Oxcarbazepine: 2017 Update. Clin. Pharmacol. Ther. 2018, 103, 574–581. [Google Scholar] [CrossRef] [PubMed]
  37. Martin, M.A.; Klein, T.E.; Dong, B.J.; Pirmohamed, M.; Haas, D.W.; Kroetz, D.L.; Clinical Pharmacogenetics Implementation Consortium. Clinical pharmacogenetics implementation consortium guidelines for HLA-B genotype and abacavir dosing. Clin. Pharmacol. Ther. 2012, 91, 734–738. [Google Scholar] [CrossRef]
  38. Martin, M.A.; Hoffman, J.M.; Freimuth, R.R.; Klein, T.E.; Dong, B.J.; Pirmohamed, M.; Hicks, J.K.; Wilkinson, M.R.; Haas, D.W.; Kroetz, D.L.; et al. Clinical Pharmacogenetics Implementation Consortium Guidelines for HLA-B Genotype and Abacavir Dosing: 2014 update. Clin. Pharmacol. Ther. 2014, 95, 499–500. [Google Scholar] [CrossRef]
  39. Hershfield, M.S.; Callaghan, J.T.; Tassaneeyakul, W.; Mushiroda, T.; Thorn, C.F.; Klein, T.E.; Lee, M.T. Clinical Pharmacogenetics Implementation Consortium guidelines for human leukocyte antigen-B genotype and allopurinol dosing. Clin. Pharmacol. Ther. 2013, 93, 153–158. [Google Scholar] [CrossRef]
  40. Saito, Y.; Stamp, L.K.; Caudle, K.E.; Hershfield, M.S.; McDonagh, E.M.; Callaghan, J.T.; Tassaneeyakul, W.; Mushiroda, T.; Kamatani, N.; Goldspiel, B.R.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for human leukocyte antigen B (HLA-B) genotype and allopurinol dosing: 2015 update. Clin. Pharmacol. Ther. 2016, 99, 36–37. [Google Scholar] [CrossRef]
  41. Muir, A.J.; Gong, L.; Johnson, S.G.; Lee, M.T.; Williams, M.S.; Klein, T.E.; Caudle, K.E.; Nelson, D.R.; Clinical Pharmacogenetics Implementation Consortium. Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for IFNL3 (IL28B) genotype and PEG interferon-α-based regimens. Clin. Pharmacol. Ther. 2014, 95, 141–146. [Google Scholar] [CrossRef]
  42. McDermott, J.H.; Wolf, J.; Hoshitsuki, K.; Huddart, R.; Caudle, K.E.; Whirl-Carrillo, M.; Steyger, P.S.; Smith, R.J.H.; Cody, N.; Rodriguez-Antona, C.; et al. Clinical Pharmacogenetics Implementation Consortium Guideline for the Use of Aminoglycosides Based on MT-RNR1 Genotype. Clin. Pharmacol. Ther. 2022, 111, 366–372. [Google Scholar] [CrossRef]
  43. Gonsalves, S.G.; Dirksen, R.T.; Sangkuhl, K.; Pulk, R.; Alvarellos, M.; Vo, T.; Hikino, K.; Roden, D.; Klein, T.E.; Poler, S.M.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for the Use of Potent Volatile Anesthetic Agents and Succinylcholine in the Context of RYR1 or CACNA1S Genotypes. Clin. Pharmacol. Ther. 2019, 105, 1338–1344. [Google Scholar] [CrossRef] [PubMed]
  44. Relling, M.V.; Gardner, E.E.; Sandborn, W.J.; Schmiegelow, K.; Pui, C.H.; Yee, S.W.; Stein, C.M.; Carrillo, M.; Evans, W.E.; Klein, T.E.; et al. Clinical Pharmacogenetics Implementation Consortium guidelines for thiopurine methyltransferase genotype and thiopurine dosing. Clin. Pharmacol. Ther. 2011, 89, 387–391. [Google Scholar] [CrossRef] [PubMed]
  45. Relling, M.V.; Gardner, E.E.; Sandborn, W.J.; Schmiegelow, K.; Pui, C.H.; Yee, S.W.; Stein, C.M.; Carrillo, M.; Evans, W.E.; Hicks, J.K.; et al. Clinical pharmacogenetics implementation consortium guidelines for thiopurine methyltransferase genotype and thiopurine dosing: 2013 update. Clin. Pharmacol. Ther. 2013, 93, 324–325. [Google Scholar] [CrossRef] [PubMed]
  46. Relling, M.V.; Schwab, M.; Whirl-Carrillo, M.; Suarez-Kurtz, G.; Pui, C.H.; Stein, C.M.; Moyer, A.M.; Evans, W.E.; Klein, T.E.; Antillon-Klussmann, F.G.; et al. Clinical Pharmacogenetics Implementation Consortium Guideline for Thiopurine Dosing Based on TPMT and NUDT15 Genotypes: 2018 Update. Clin. Pharmacol. Ther. 2019, 105, 1095–1105. [Google Scholar] [CrossRef]
  47. Gammal, R.S.; Court, M.H.; Haidar, C.E.; Iwuchukwu, O.F.; Gaur, A.H.; Alvarellos, M.; Guillemette, C.; Lennox, J.L.; Whirl-Carrillo, M.; Brummel, S.S.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for UGT1A1 and Atazanavir Prescribing. Clin. Pharmacol. Ther. 2016, 99, 363–369. [Google Scholar] [CrossRef]
  48. Clancy, J.P.; Johnson, S.G.; Yee, S.W.; McDonagh, E.M.; Caudle, K.E.; Klein, T.E.; Cannavo, M.; Giacomini, K.M.; Clinical Pharmacogenetics Implementation Consortium. Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for ivacaftor therapy in the context of CFTR genotype. Clin. Pharmacol. Ther. 2014, 95, 592–597. [Google Scholar] [CrossRef]
  49. Shekhani, R.; Steinacher, L.; Swen, J.J.; Ingelman-Sundberg, M. Evaluation of Current Regulation and Guidelines of Pharmacogenomic Drug Labels: Opportunities for Improvements. Clin. Pharmacol. Ther. 2020, 107, 1240–1255. [Google Scholar] [CrossRef]
  50. Swen, J.J.; Wilting, I.; de Goede, A.L.; Grandia, L.; Mulder, H.; Touw, D.J.; de Boer, A.; Conemans, J.M.; Egberts, T.C.; Klungel, O.H.; et al. Pharmacogenetics: From bench to byte. Clin. Pharmacol. Ther. 2008, 83, 781–787. [Google Scholar] [CrossRef]
  51. Swen, J.J.; Nijenhuis, M.; de Boer, A.; Grandia, L.; Maitland-van der Zee, A.H.; Mulder, H.; Rongen, G.A.; van Schaik, R.H.; Schalekamp, T.; Touw, D.J.; et al. Pharmacogenetics: From bench to byte—An update of guidelines. Clin. Pharmacol. Ther. 2011, 89, 662–673. [Google Scholar] [CrossRef] [PubMed]
  52. Brouwer, J.; Nijenhuis, M.; Soree, B.; Guchelaar, H.J.; Swen, J.J.; van Schaik, R.H.N.; Weide, J.V.; Rongen, G.; Buunk, A.M.; de Boer-Veger, N.J.; et al. Dutch Pharmacogenetics Working Group (DPWG) guideline for the gene-drug interaction between CYP2C19 and CYP2D6 and SSRIs. Eur. J. Hum. Genet. 2022, 30, 1114–1120. [Google Scholar] [CrossRef] [PubMed]
  53. Beunk, L.; Nijenhuis, M.; Soree, B.; de Boer-Veger, N.J.; Buunk, A.M.; Guchelaar, H.J.; Houwink, E.J.F.; Risselada, A.; Rongen, G.; van Schaik, R.H.N.; et al. Dutch Pharmacogenetics Working Group (DPWG) guideline for the gene-drug interaction between CYP2D6, CYP3A4 and CYP1A2 and antipsychotics. Eur. J. Hum. Genet. 2023. ahead of print. [Google Scholar] [CrossRef] [PubMed]
  54. Hulshof, E.C.; Deenen, M.J.; Nijenhuis, M.; Soree, B.; de Boer-Veger, N.J.; Buunk, A.M.; Houwink, E.J.F.; Risselada, A.; Rongen, G.; van Schaik, R.H.N.; et al. Dutch pharmacogenetics working group (DPWG) guideline for the gene-drug interaction between UGT1A1 and irinotecan. Eur. J. Hum. Genet. 2022, 31, 982–987. [Google Scholar] [CrossRef]
  55. Lunenburg, C.; van der Wouden, C.H.; Nijenhuis, M.; Crommentuijn-van Rhenen, M.H.; de Boer-Veger, N.J.; Buunk, A.M.; Houwink, E.J.F.; Mulder, H.; Rongen, G.A.; van Schaik, R.H.N.; et al. Dutch Pharmacogenetics Working Group (DPWG) guideline for the gene-drug interaction of DPYD and fluoropyrimidines. Eur. J. Hum. Genet. 2020, 28, 508–517. [Google Scholar] [CrossRef] [PubMed]
  56. Nijenhuis, M.; Soree, B.; Jama, W.O.M.; de Boer-Veger, N.J.; Buunk, A.M.; Guchelaar, H.J.; Houwink, E.J.F.; Rongen, G.A.; van Schaik, R.H.N.; Swen, J.J.; et al. Dutch pharmacogenetics working group (DPWG) guideline for the gene-drug interaction of CYP2D6 and COMT with atomoxetine and methylphenidate. Eur. J. Hum. Genet. 2022. ahead of print. [Google Scholar] [CrossRef]
  57. Matic, M.; Nijenhuis, M.; Soree, B.; de Boer-Veger, N.J.; Buunk, A.M.; Houwink, E.J.F.; Mulder, H.; Rongen, G.; Weide, J.V.; Wilffert, B.; et al. Dutch Pharmacogenetics Working Group (DPWG) guideline for the gene-drug interaction between CYP2D6 and opioids (codeine, tramadol and oxycodone). Eur. J. Hum. Genet. 2022, 30, 1105–1113. [Google Scholar] [CrossRef]
  58. van der Pol, K.H.; Nijenhuis, M.; Soree, B.; de Boer-Veger, N.J.; Buunk, A.M.; Guchelaar, H.J.; Risselada, A.; van Schaik, R.H.N.; Swen, J.J.; Touw, D.; et al. Dutch pharmacogenetics working group guideline for the gene-drug interaction of ABCG2, HLA-B and Allopurinol, and MTHFR, folic acid and methotrexate. Eur. J. Hum. Genet. 2022. ahead of print. [Google Scholar] [CrossRef]
  59. Houwink, E.J.; Rigter, T.; Swen, J.J.; Cornel, M.C.; Kienhuis, A.; Rodenburg, W.; Weda, M. [Pharmacogenetics in primary health care: Implementation and future expectations] Farmacogenetica in de eerstelijnszorg: Toepassing en toekomstverwachtingen. Ned. Tijdschr. Geneeskd. 2015, 159, A9204. [Google Scholar]
  60. Bank, P.C.D.; Swen, J.J.; Schaap, R.D.; Klootwijk, D.B.; Baak-Pablo, R.; Guchelaar, H.J. A pilot study of the implementation of pharmacogenomic pharmacist initiated pre-emptive testing in primary care. Eur. J. Hum. Genet. 2019, 27, 1532–1541. [Google Scholar] [CrossRef]
  61. van der Wouden, C.H.; Bank, P.C.D.; Ozokcu, K.; Swen, J.J.; Guchelaar, H.J. Pharmacist-Initiated Pre-Emptive Pharmacogenetic Panel Testing with Clinical Decision Support in Primary Care: Record of PGx Results and Real-World Impact. Genes 2019, 10, 416. [Google Scholar] [CrossRef] [PubMed]
  62. Youssef, E.; Kirkdale, C.L.; Wright, D.J.; Guchelaar, H.J.; Thornley, T. Estimating the potential impact of implementing pre-emptive pharmacogenetic testing in primary care across the UK. Br. J. Clin. Pharmacol. 2021, 87, 2907–2925. [Google Scholar] [CrossRef] [PubMed]
  63. Bank, P.C.D.; Swen, J.J.; Guchelaar, H.J. Estimated nationwide impact of implementing a preemptive pharmacogenetic panel approach to guide drug prescribing in primary care in The Netherlands. BMC Med. 2019, 17, 110. [Google Scholar] [CrossRef] [PubMed]
  64. Rigter, T.; Jansen, M.E.; de Groot, J.M.; Janssen, S.W.J.; Rodenburg, W.; Cornel, M.C. Implementation of Pharmacogenetics in Primary Care: A Multi-Stakeholder Perspective. Front. Genet. 2020, 11, 10. [Google Scholar] [CrossRef] [PubMed]
  65. Preys, C.L.; Blout Zawatsky, C.L.; Massmann, A.; Heukelom, J.V.; Green, R.C.; Hajek, C.; Hickingbotham, M.R.; Zoltick, E.S.; Schultz, A.; Christensen, K.D. Attitudes about pharmacogenomic testing vary by healthcare specialty. Pharmacogenomics 2023, 24, 539–549. [Google Scholar] [CrossRef]
  66. Behr, M.P.; Gammal, R.S.; Matthews, M.L.; Wang, V.C. Healthcare professionals’ knowledge, confidence and perceptions of pharmacogenomics in primary care and pain management. Pharmacogenomics 2023, 24, 371–379. [Google Scholar] [CrossRef]
Figure 1. PGx tests requested by GPs. (a) Distribution per gene, including both separate and panel testing requests (n = 6301); displayed are the genes above a 1% threshold. PGx tests requested, but not reaching a 1% threshold, were BChE, CYP2C8, CYP3A7, DPYD, GADL, HLA-B*1502, HLA-A*3101, HLA-B*5701, MTHFR, NUDT15, TPMT, UGT1A1 and UGT1A9; (b) distribution on requested panel testing (n = 795).
Figure 1. PGx tests requested by GPs. (a) Distribution per gene, including both separate and panel testing requests (n = 6301); displayed are the genes above a 1% threshold. PGx tests requested, but not reaching a 1% threshold, were BChE, CYP2C8, CYP3A7, DPYD, GADL, HLA-B*1502, HLA-A*3101, HLA-B*5701, MTHFR, NUDT15, TPMT, UGT1A1 and UGT1A9; (b) distribution on requested panel testing (n = 795).
Genes 14 01841 g001
Figure 2. Top 10 drug groups for which PGx testing was requested, based on 706 forms. PPIs: proton pump inhibitors; NSAIDs: non-steroidal anti-inflammatory drugs.
Figure 2. Top 10 drug groups for which PGx testing was requested, based on 706 forms. PPIs: proton pump inhibitors; NSAIDs: non-steroidal anti-inflammatory drugs.
Genes 14 01841 g002
Figure 3. Main reasons for requesting PGx testing, based on 850 forms.
Figure 3. Main reasons for requesting PGx testing, based on 850 forms.
Genes 14 01841 g003
Figure 4. Barriers to applying PGx experienced by respondents (n = 29).
Figure 4. Barriers to applying PGx experienced by respondents (n = 29).
Genes 14 01841 g004
Table 1. Available PGx tests and panel tests at Erasmus MC in 2021.
Table 1. Available PGx tests and panel tests at Erasmus MC in 2021.
PGx Panel NamePGx Tests
DNA panel—basicCYP2C9, CYP2C19, CYP2D6, CYP3A4, VKORC1, SLCO1B1
DNA panel—extendedIs DNA panel—basic + CYP1A2, CYP2B6, CYP3A5
Psychiatry panelCYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4
Cardiac panelCYP2C9, CYP2C19, CYP2D6, VKORC1, SLCO1B1, ABCB1
Pain panelCYP2C9, CYP2D6, CYP3A4, OPRM1, COMT
Oncology panelCYP2D6, CYP3A4, CYP3A5, DPYD
Individual testsCYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP3A4, CYP3A5, CYP3A7, BChE, DPYD, TPMT, NUDT15, UGT1A1, UGT1A9, ABCB1, ABCC2, ABCG2, SLCO1B1, HLA-A*3101, HLA-B*1502, HLA-B*5701, HLA-B*5801, HLA-B*1511, GADL1, VKORC1
Table 2. Drugs mentioned by GPs on request forms (n = 706); threshold >1%. Drugs for which the DPWG have no guidelines or no adapted dosing recommendations available are indicated in italics.
Table 2. Drugs mentioned by GPs on request forms (n = 706); threshold >1%. Drugs for which the DPWG have no guidelines or no adapted dosing recommendations available are indicated in italics.
Drug%Drug%Drug%
Clopidogrel23.0%Lisinopril3.2%Clomipramine1.8%
Citalopram8.0%Rosuvastatin2.9%Metformin1.7%
Amitriptyline6.8%Atorvastatin2.9%Bisoprolol1.7%
Metoprolol6.8%Fluoxetine2.9%Verapamil1.5%
Tramadol6.3%Quetiapine2.8%Amoxicillin1.5%
Sertraline6.1%Morphine2.8%Bupropion1.5%
Pantoprazole5.7%Pregabalin2.6%Ac. Salic. Acid1.5%
Venlafaxine5.4%Risperidone2.5%Duloxetine1.4%
Amlodipine5.4%Diclofenac2.5%Valsartan1.4%
Simvastatin4.9%Ezetimibe2.2%Diazepam1.2%
Oxycodone4.6%Paracetamol2.2%Perindopril1.2%
Paroxetine4.5%Aripiprazole2.0%Naproxen1.1%
Omeprazole4.3%Nifedipine2.0%Ibuprofen1.1%
Nortriptyline4.1%Methylphenidate2.0%Enalapril1.1%
Escitalopram3.7%Lidocaine2.0%Fentanyl1.1%
H.chlorothiazide3.5%Codeine1.8%Oxazepam1.1%
Mirtazapine3.5%Losartan1.8%
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

van der Drift, D.; Simoons, M.; Koch, B.C.P.; Brufau, G.; Bindels, P.; Matic, M.; van Schaik, R.H.N. Implementation of Pharmacogenetics in First-Line Care: Evaluation of Its Use by General Practitioners. Genes 2023, 14, 1841. https://doi.org/10.3390/genes14101841

AMA Style

van der Drift D, Simoons M, Koch BCP, Brufau G, Bindels P, Matic M, van Schaik RHN. Implementation of Pharmacogenetics in First-Line Care: Evaluation of Its Use by General Practitioners. Genes. 2023; 14(10):1841. https://doi.org/10.3390/genes14101841

Chicago/Turabian Style

van der Drift, Denise, Mirjam Simoons, Birgit C. P. Koch, Gemma Brufau, Patrick Bindels, Maja Matic, and Ron H. N. van Schaik. 2023. "Implementation of Pharmacogenetics in First-Line Care: Evaluation of Its Use by General Practitioners" Genes 14, no. 10: 1841. https://doi.org/10.3390/genes14101841

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

Article Metrics

Back to TopTop