Pharmacogenomics of hypertension in chronic kidney disease: the CKD-PGX study ============================================================================= * Michael T. Eadon * Judith Maddatu * Sharon M. Moe * Arjun D. Sinha * Ricardo Melo Ferreira * Brent W. Miller * S. Jawad Sher * Jing Su * Victoria M. Pratt * Arlene B. Chapman * Todd S. Skaar * Ranjani N. Moorthi * for the CKD-PGX investigators ## ABSTRACT **Background** Patients with chronic kidney disease (CKD) often have uncontrolled hypertension despite polypharmacy. Pharmacogenomic drug-gene interactions (DGIs) may impact the metabolism or efficacy of antihypertensive agents. We hypothesized that providing a panel of 11 pharmacogenomic predictors of antihypertensive response would improve hypertension control. **Methods** A prospective cohort with CKD and hypertension was followed to assess the effect of pharmacogenomic testing on blood pressure control. The analysis population included 382 hypertensive subjects genotyped for cross-sectional assessment of drug-gene interactions and 335 subjects followed for 1 year to assess systolic (SBP) and diastolic blood pressure (DBP). **Results** Most participants (58.2%) with uncontrolled hypertension had a DGI reducing the efficacy of one or more antihypertensive agents. Subjects with a DGI had 1.88-fold (95% CI 1.2-2.8) higher odds of uncontrolled hypertension as compared to those without a DGI, adjusted for race and CKD grade. *CYP2C9* reduced metabolism genotypes were associated with losartan response and uncontrolled hypertension (Odds Ratio 5.2, CI 1.9 -14.7). *CYP2D6* intermediate or poor metabolizers had less frequent uncontrolled hypertension compared to normal metabolizers taking metoprolol or carvedilol (OR 0.55, CI 0.3-0.95). In 335 subjects completing 1 year follow-up, SBP (−4.0 mmHg, CI 1.6-6.5) and DBP (−3.3 mmHg, CI 2.0-4.6) were improved. The magnitude of reductions in SBP (−14.8 mmHg, CI 10.3-19.3) and DBP (−8.4 mmHg, CI 5.9-10.9) were greatest in the 90 individuals with uncontrolled hypertension and an actionable genotype. **Conclusions** There is a potential role for the addition of pharmacogenomic testing to optimize antihypertensive regimens in patients with CKD. Keywords * pharmacogenomics * blood pressure * genotype * CYP2D6 * CYP2C9 ## INTRODUCTION Hypertension and chronic kidney disease (CKD) are common intersecting diseases with enormous economic burden, morbidity, and mortality. The Center for Disease Control (CDC) reports that 45% of the US population has hypertension, with approximately half of those individuals inadequately controlled1. The prevalence of hypertension increases with the severity of CKD as 36% of grade 1, 48% of grade 2, 60% of grade 3, and 84% of grade 4/5 CKD patients have concomitant hypertension2. Impaired sodium excretion, extracellular volume expansion, activation of the renin-angiotensin system, and numerous vasoconstrictive effects all conspire to impair blood pressure control in CKD patients3. International guidelines emphasize control of hypertension to reduce cardiovascular events in patients with CKD4. Many antihypertensive agents are subject to different forms of pharmacokinetic or pharmacodynamic drug-gene interactions, each of which impact efficacy. For example, metoprolol is metabolized by the enzyme cytochrome P450 2D6 (CYP2D6), wherein poor metabolizers possess higher circulating concentrations of the drug at a given dose5. The beta-blocker class may be pharmacodynamically affected by beta-1 adrenergic receptor (ADRB1) polymorphisms6. The angiotensin receptor blocker (ARB) losartan potassium is a pro-drug metabolized by cytochrome P450 2C9 (CYP2C9)7. Poor metabolizers of CYP2C9 have lower concentrations of its active metabolite8. Hydralazine hydrochloride undergoes phase 2 metabolism by N-acetyltransferase 2 (NAT2)9. Fast and intermediate acetylators will have lower concentrations and reduced efficacy of hydralazine at a given dose. The evidence supporting these (and other) pharmacogenomic drug-gene pairs has been generated through prior clinical studies and this evidence has been previously summarized10, 11. Previous studies have identified individual drug-gene pairs relevant to antihypertensives in the general population and those with CKD. Over 80% of individuals with CKD and hypertension take 2 or more antihypertensives and 32% take four or more agents12. Despite polypharmacy, 10.3 million Americans with apparent treatment resistant hypertension remain uncontrolled13. We hypothesized that embedding a panel of pharmacogenomic predictors of antihypertensive response in routine clinical practice would aid patients and practitioners in arriving at an efficacious blood pressure regimen, either by identifying less efficacious medications in an individual’s current regimen or selecting an efficacious drug as the “next” antihypertensive agent. To facilitate this, a clinical genotyping assay was developed and implemented across multiple health systems, with results and recommendations recorded in the electronic health records (EHR) for 40 variants and 11 drug-gene pairs relevant to hypertension control14. We present the results of a prospective cohort study entitled CKD-PGX, that enrolled and genotyped 382 adult hypertensive CKD patients from Indiana University Health Physicians nephrology clinics in three settings: a university health system, a county safety-net health system, and outlying suburban clinics near Indianapolis. The goal was to assess provider utilization, patient attitude, prevalence of actionable drug-gene interactions (DGIs), and blood pressure control after pharmacogenomic panel testing. ## MATERIALS AND METHODS ### Study Design This was a prospective, observational cohort study. Subjects were recruited and provided informed consent during a nephrology clinic visit in the IU Health or Eskenazi Health systems between 2017 and 2019. Blood pressure (BP) was assessed upon enrollment and at 1 year follow-up. This study was approved by the Institutional Review Board of Indiana University (IRB # 1705413046). ### Study Population Subjects were eligible for inclusion if of age ≥ 18 years old with the ability to provide consent and a genotyping sample. Subjects were required to have at least one of the following: systolic BP ≥ 140 mm Hg on any two readings in the 12 preceding months, estimated glomerular filtration rate (eGFR) less than 60 mL/min/1.73 m2, daily proteinuria > 0.2 g by 24-hour urine collection or > 0.2 g/g urine protein to creatinine ratio15. Our analysis population was comprised of N = 425 for the baseline subject survey, N = 382 for the cross-sectional analysis between genotype and hypertension control, and N = 335 for the longitudinal 1-year follow-up blood pressure outcomes. The prevalence of hypertension in those with CKD presenting to our clinics during the study period determined the sample size. ### Study Procedures BP was obtained at baseline immediately following a nephrology clinic appointment using a standard sphygmomanometer while seated at rest and again from the clinic visit closest to 1 year after enrollment (± 6 months). Three BP measurements were acquired, each separated by 5 minutes. Systolic and diastolic BP were each averaged separately for the three measurements. Participants provided a whole blood or saliva sample and were genotyped for 40 variants in 11 genes related to antihypertensive response (Table 1, Supplemental Table S1). Genotyping was performed on a custom Taqman™ OpenArray™ (FisherScientific, Waltham, MA) as previously described14. Genetic data were deposited in the EHR approximately two weeks after testing along with interpretations on drug efficacy. Providers received encrypted email alerts when the genetic information was available in the EHR. View this table: [Table 1:](http://medrxiv.org/content/early/2021/04/06/2021.03.30.21254665/T1) Table 1: Nephrology providers (N = 39) gave assent to enroll their patients and were trained on the interpretation of pharmacogenomic drug-gene interactions. The principal investigators did not alter or suggest changes to subjects’ prescriptions; all clinical care was at the behest of the primary nephrology provider. Three surveys were administered: 1) each subject’s attitude toward genetic testing was evaluated in a 15 question survey at baseline (Supplemental Document S1), 2) each provider (N = 76) completed a baseline survey regarding their attitude toward the testing16, and 3) each provider completed a return of results survey for every one of their enrolled patients to query whether they believed testing affected their clinical management. ### Variables Variables including demographic characteristics, biochemical parameters, CKD status, co-morbidities, and medication lists were obtained from the EHR at baseline and at 1 year follow-up. ### Outcomes Study outcomes included: 1) prevalence of uncontrolled hypertension associated with actionable DGIs at baseline, and 2) change in systolic blood pressure (SBP) and diastolic blood pressure (DBP) at 1 year follow-up. A DGI or “actionable” genotype was defined as the presence of at least one variant predicting reduced efficacy for an antihypertensive agent a subject was taking at the time of enrollment. Secondary outcomes included patient attitudes toward genetic testing and provider utilization as defined by the return of results survey. ### Statistical Analysis Baseline data and survey responses were analyzed descriptively and provided as percentages for categorical variables, mean ± standard deviation (SD) for normally distributed variables, and median (25th and 75th percentile) for non-normally distributed variables. Comparisons of categorical variables were expressed as an odds ratio (OR) with 95% confidence interval (CI). For our primary outcome analyses, subjects were considered to have a relevant DGI if one or more of their genetic variants predicted reduced efficacy of their prescribed antihypertensives. DGIs were coded as a binary variable (present or absent). Evaluation of the relationship between DGIs and hypertension control was performed by χ2 test and adjusted for significant covariates using logistic regression. Sub-group analyses were performed for each individual drug-gene(s) pair. Variants predicting increased efficacy of antihypertensives were assessed in sub-group analyses. Change in blood pressure within each individual at 1 year was assessed by Paired Student’s t-test. ## RESULTS ### Participants A total of 472 adult subjects were recruited and consented from outpatient nephrology clinics within the Indiana University Health system, the Eskenazi Health safety-net system, and associated outlying suburban clinics (Figure 1). Thirty-seven subjects withdrew from the study, most frequently due to personal reasons, an inability to follow-up during the coronavirus pandemic, or a genotyping failure for which they would not provide a repeat sample. The remaining 435 subjects were genotyped and completed baseline surveys. The characteristics of the overall population are given in Table 2. There were approximately equal numbers of female and male subjects and the average age was 58.1 ± 14.9 years old. The average body mass index (BMI) of 33.7 ± 8.4 kg/m2. The majority had baseline CKD stage 3 and 92.2% had an ICD10 diagnosis of hypertension (N = 401); however, only 382 subjects (87.8%) were treated with antihypertensive agents. The average number of antihypertensive agents prescribed was 2.7 ± 1.6 per subject. Angiotensin converting enzyme inhibitors (ACEIs) and ARBs (57.6%) or beta blockers (55.6%) were commonly prescribed. Common comorbidities included diabetes, heart disease, and sleep apnea. The overall mean SBP was 139.9 ± 22.1 mmHg and the mean DBP was 80.7 ± 12.0 mmHg. Of the 382 subjects on antihypertensives, 335 subjects completed 1 year follow-up. View this table: [Table 2:](http://medrxiv.org/content/early/2021/04/06/2021.03.30.21254665/T2) Table 2: Baseline Characteristics of Subjects included in Overall Genotype-Analysis ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/04/06/2021.03.30.21254665/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2021/04/06/2021.03.30.21254665/F1) Figure 1: Enrollment and inclusion in the analyses. Survey data was available in 435 adult participants who received the pharmacogenomic genotyping panel. There were 382 subjects who had a hypertension diagnosis and were prescribed 1 or more antihypertensives. These 382 were included in the cross-sectional analysis. In the longitudinal analysis, there were 335 subjects included who completed a 1 year follow-up with subsequent blood pressure assessment. ### Association of drug-gene interactions and blood pressure control Of the 382 individuals prescribed antihypertensive agents at baseline, 189 had uncontrolled hypertension (uHTN), defined as a SBP ≥ 140 mm Hg or DBP ≥ 90 (Table 2). The mean SBP in subjects with uHTN was 158.0 ± 17.6 mmHg. The mean SBP in individuals with controlled hypertension (cHTN, N = 193) was significantly lower at 123.3 ± 9.7 (P < 0.001). Similarly, the average DBP in uHTN subjects was higher (P < 0.001). Those with cHTN and uHTN were similar in age, sex distribution, BMI, and frequency of comorbidities. CKD was more prevalent in the uHTN group (P = 0.038). The distribution of race was significantly different between groups as 64.8% of cHTN subjects were white while only 48.7% of uHTN subjects were white (P = 0.004). In the 382 subjects at baseline, relevant DGIs predicted to reduce efficacy of a currently prescribed antihypertensive agent were assessed. The genotype distribution is summarized in Supplemental Table S2. The majority of participants with uHTN had an actionable genotype (58.2%). Subjects with relevant DGIs were more likely to have uncontrolled hypertension (P = 0.0008). Subjects with an actionable genotype had 2-fold (95% CI 1.3-3.0) higher odds of uHTN as compared to those without an actionable genotype (Table 3). When adjusted for race and presence of CKD stage 3 or greater, subjects with a relevant DGI had 1.88-fold (95% CI 1.2-2.8) increased odds of uHTN. In summary, individuals who had uHTN were more likely to have a genetic variant that predicted reduced efficacy of an anti-hypertensive agent that they were prescribed at baseline. View this table: [Table 3:](http://medrxiv.org/content/early/2021/04/06/2021.03.30.21254665/T3) Table 3: Association of antihypertensive drug-gene interactions with blood pressure ### Individual drug-gene analyses As exploratory analyses, we examined the association between relevant DGIs and baseline uHTN for each individual drug-gene interaction (Supplemental Table 3). Significant associations between uHTN and DGIs were found for participants prescribed losartan, metoprolol, and carvedilol. Variants in *CYP2C9* that predicted reduced efficacy of losartan were associated with uHTN in participants taking the drug (OR 5.2, 95% CI 1.9 to 14.7). Intermediate or poor CYP2D6 metabolizers have higher circulating concentrations of metoprolol or carvedilol. These individuals were less likely to have uncontrolled hypertension than normal metabolizers taking either agent (OR 0.55, 95% CI 0.3-0.95). No other significant DGIs were identified in this relatively small sample size. ### Longitudinal blood pressure control Overall, the 335 subjects who completed a 1 year follow-up in the Nephrology clinic had a significant decrease in blood pressures, both systolic (−4.0 (95% CI 1.6,6.5) mmHg) and diastolic (−3.3 (95% CI 2.0,4.6) mmHg). Amongst the 160 individuals with uHTN, 71 were “controlled” by 1 year follow-up with a SBP < 140 and a DBP < 90 mmHg. Table 4 shows the within group comparisons of baseline and 1 year follow-up blood pressures in the overall cohort, in those with a DGI, in those with uHTN at baseline and with uHTN and a concurrent actionable genotype. All comparisons were significant (p<0.001). The magnitude of reductions in both SBP at 14.8 (95%CI 10.3, 19.3) mmHg and DBP at 8.4 (95%CI 5.9,10.9) mmHg were largest in the 90 individuals with uHTN and an actionable genotype. Taken together, the inference is that the detection of an actionable genotype provided an opportunity for control of blood pressure over one year in those uncontrolled at baseline. View this table: [Table 4:](http://medrxiv.org/content/early/2021/04/06/2021.03.30.21254665/T4) Table 4: Longitudinal Blood Pressure assessment ### Provider utilization Action driven item surveys for each recruited subject were completed by their primary nephrology provider. The surveys queried the utility of genotype results in each subjects’ anti-hypertensive drug management. The response rate was 53.7% (180 of 335). Physicians reported that the genetic testing altered their diagnosis or management in 36% of cases. In 85% of survey responses, physicians stated they had or would discuss results with their patients. ### Patient reported attitudes to genotyping Among the full cohort of 435 subjects with hypertension, proteinuria, or eGFR < 60 mL/min/1.73 m2, 425 subjects completed a baseline survey. Supplemental Figure 1 illustrates the response from selected questions regarding subjects’ attitude toward genetic testing. The surveys revealed that few subjects (5.4%) were familiar with the terms “pharmacogenomics, genetic testing or personalized medicine”. More than 96% reported that knowledge of their genetic code would prompt them to invest more to control their blood pressure as well as enable their providers to deliver enhanced antihypertensive care. ## DISCUSSION Antihypertensive medication response is frequently unpredictable and varies among individuals. In CKD, blood pressure control at recommended targets is clearly important to prevent cardiovascular disease and end organ dysfunction. However, 24.9 to 33.4% of individuals with grade 3 or greater CKD have uncontrolled BP despite therapy with three or more medications17. Society guidelines often provide initial and secondary agent recommendations extrapolated from the general population18, 19, but may fail to provide specific direction for those with CKD and apparent treatment resistant (uncontrolled) hypertension. In this study, relevant drug-gene interactions were found in 58.2 % of subjects. We identified a significant odds ratio of 1.8 between reduced efficacy drug-gene interactions and uncontrolled hypertension, even after adjusting for presence of CKD and race. When providers were armed with and trained on the genotype information, they reported adjusting the antihypertensive regimen in 36% of subjects. Study subjects were receiving ongoing care in nephrology clinics including frequent follow-up, ambulatory BP monitoring when required, secondary HTN work-up, and dietary counseling. The only addition to standard care was the provision of the genotype report in the electronic health record and notification to the subject’s physician. Medication changes, if any, were made by the primary nephrologists who indicated they discussed the genetic results with 85% of subjects. Patient engagement is a necessary component of hypertension management. Indeed, surveyed subjects agreed that understanding their pharmacogenomic report would facilitate greater action on their part to control BP. The adjustments made by the patients and providers resulted in a decrease of 4 mm Hg in SBP within individuals across the entire cohort. A remarkable reduction in SBP of 14.8 mm Hg was observed in those with baseline uHTN and an actionable genotype. Our pharmacogenomics panel-based approach included a wide array of variants. A summary of the available evidence is beyond the scope of this discussion; however, antihypertensive variants were selected based on the strength of evidence, minor allele frequency, FDA label annotations, and guidelines. The FDA drug labels of hydralazine20, losartan8, and metoprolol21 each reference metabolic enzymes which affect concentrations of these drugs. Where major society guidelines were available, such as for metoprolol and carvedilol from the Dutch Pharmacogenomic Working Group (DPWG)22, they were used to guide recommendations. As additional guidelines from DPWG and the Clinical Pharmacogenomics Implementation Consortium (CPIC) are made available, these will need to be incorporated into future genotype-guided dosing recommendations. We included 40 variants associated with efficacy in a breadth of agents to maximize actionability and impact for enrolled subjects. In some cases, pharmacokinetic and pharmacodynamic variants predicted opposite effects for the same drug. For example, the CYP2D6 poor metabolizer status predicts increased circulating concentrations of metoprolol, yet a subject with a reduced function variant of *ADRB1* would have the opposite predicted efficacy. There is insufficient evidence to reconcile these two effects; thus, all variants were reported separately and the prescribing clinician decided how they would use the information. Consideration was given to evidence in multiple populations. The Pharmacogenomics Evaluation of Antihypertensive Response (PEAR) group identified different predictors of thiazide efficacy in African Americans and Caucasians. As such, *YEATS4* was used as a predictor in individuals with African ancestry23 while a different three gene model was used in Caucasians24. In this study, the outcomes of utilization and blood pressure control incorporate a level of decision making on the part of the provider. The direction of effect for drug-gene prediction, the identification of race by the provider and subject, and the choice of medications to treat apparent treatment resistant hypertension are inherent aspects of the decision-making process for all providers. A significant strength of our study is generalizability. Our study population was 57% white, 40% black and recruited from three diverse environments: a university hospital, a safety net health system, and several suburban clinics. The primary limitation of this study is that it was a prospective cohort, not a randomized controlled trial. Thus, observation bias in BP control (the Hawthorne effect) may contribute to the outcomes measured. This bias is inherent in all prospective cohort studies. An additional weakness, although also more generalizable, is that we did not mandate specific changes to the BP regimen. Multiple prior studies have11. We were underpowered to detect the significance of specific prescribing behavior. In practice, this may hold less relevance as CKD patients are on multiple antihypertensives, frequently with multiple actionable genotypes. Some providers would elect to change dose, select an alternate medication, or add a medication. We relied upon a binary variable of provider self-report of utility in each subject. A pharmacogenomic panel-based approach was utilized, and our sample size was insufficient to detect the effect of any single variant. Significant loss to follow-up (8.6%) did occur, in part due to the advent of the coronavirus pandemic which shifted follow-up to telehealth and impacted assessment of BP. Finally, we did not track medication compliance. These limitations are counterbalanced by our clinically translatable outcomes such as the association of drug-gene interactions with BP, longitudinal BP control, and provider utilization. This study outlines the implementation of pharmacogenomic panel testing in an outpatient nephrology setting. In our cohort, we showed that nephrology providers will use information regarding drug-gene interactions to effect change in blood pressures in their patients. Currently, individual patient demographics such as obesity, gender, and race are important factors in the selection process for an adequate anti-hypertensive regimen25. This study illustrates that there is a convincing role for the addition of pharmacogenomic data to make choices in antihypertensive regimens. We expect that in the future, whole genome or exome sequencing will be integrated into the clinical setting. At that time, genotype information will be readily available to providers and will usher in an exciting era in the care of patients with hypertension and chronic kidney disease. ## Supporting information Supplemental Document [[supplements/254665_file03.pdf]](pending:yes) ## Data Availability There is no publicly available data source ## Author contributions Conception, study design: MTE, RNM, ABC, TCS, SMM Analysis, interpretation of data: MTE, RNM, JM, SMM, RMF, JS. Conduct of study: VMP, MTE Drafting / revising the article: MTE, RNM, JM. Providing intellectual content: ADS, BWM, SJS. Final approval of the version to be published: All authors. ## Funding Support for this work was provided by the NIH/NIDDK K08DK107864 (M.T.E.) and the Indiana University Grand Challenge Precision Health Initiative. SMM was funded by R01DK11087103 and P30AR072581-01. RNM was supported by K23DK102824 and R01AR077273. **Collaborators:** “for the CKD-PGX investigators” Allon N. Friedman MD1, Kimberly S. Collins PhD1, Nehal A. Sheth BA1, Katherine M. Spiech BS1, Asif A. Sharfuddin MBBS1, Nupur Gupta MBBS1, Ayman Hallab MD1, Simit Doshi MBBS MPH1, Matthew D. Dollins MD1, Emma M. Tillman PharmD PhD1, Elizabeth Rowe PhD1, Tyler Shugg PharmD PhD1, Chad A. Zarse MD1, Jonathan W. Bazeley MD MS1, Jay B. Wish MD1, David S. Hains MD2, Myda Khalid MBBS2, Tae-Hwi Schwantes-An PhD3, Elizabeth B. Medeiros MSFS3 1Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA 2Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA 3Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA ## CONFLICT OF INTEREST DISCLOSURES ADS reports consulting for George Clinical and Johnson & Johnson outside of the submitted work and contracted research for Bayer outside of the submitted work. ## Figure Legends **Supplemental Figure 1: Subject reported survey results**. Of the 435 subjects who underwent genotyping, 425 completed a baseline survey regarding their beliefs and attitudes toward genetic testing. All responses were given according to a Likert scale. A) Subjects were asked about their familiarity with pharmacogenomics. B) Subjects were asked whether genetic testing would increase their own efforts to control their blood pressure. C-D) Subjects were asked whether the genetic testing would help their providers select medications and control blood pressure. ## ACKNOWLEDGEMENTS * Received March 30, 2021. * Revision received March 30, 2021. * Accepted April 6, 2021. * © 2021, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. 1.1. SERVICES Usdohah Ostchega YF, C.D.; Nwankwo, T.; Nguyen, D.T. : Hypertension Prevalence Among Adults Aged 18 and Over: United States, 2017–2018. In: SERVICES Usdohah (Ed.), National Center for Health Statistics, 2020 pp 1–8 2. 2.Tedla FM, Brar A, Browne R, Brown C: Hypertension in chronic kidney disease: navigating the evidence. Int J Hypertens, 2011: 132405, 2011 doi:10.4061/2011/132405 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.4061/2011/132405&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21747971&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 3. 3.Ku E, Lee BJ, Wei J, Weir MR: Hypertension in CKD: Core Curriculum 2019. Am J Kidney Dis, 74: 120-131, 2019 doi:10.1053/j.ajkd.2018.12.044 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1053/j.ajkd.2018.12.044&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30898362&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 4. 4.Cheung AK, Chang TI, Cushman WC, Furth SL, Hou FF, Ix JH, et al: Executive summary of the KDIGO 2021 Clinical Practice Guideline for the Management of Blood Pressure in Chronic Kidney Disease. Kidney Int, 99: 559–569, 2021 doi:10.1016/j.kint.2020.10.026 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.kint.2020.10.026&link_type=DOI) 5. 5.Thomas CD, Mosley SA, Kim S, Lingineni K, El Rouby N, Langaee TY, et al: Examination of Metoprolol Pharmacokinetics and Pharmacodynamics Across CYP2D6 Genotype-Derived Activity Scores. CPT Pharmacometrics Syst Pharmacol, 9: 678–685, 2020 doi:10.1002/psp4.12563 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/psp4.12563&link_type=DOI) 6. 6.Terra SG, Pauly DF, Lee CR, Patterson JH, Adams KF, Schofield RS, et al: beta-Adrenergic receptor polymorphisms and responses during titration of metoprolol controlled release/extended release in heart failure. Clin Pharmacol Ther, 77: 127–137, 2005 doi:10.1016/j.clpt.2004.10.006 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.clpt.2004.10.006&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15735607&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000227261700003&link_type=ISI) 7. 7.Sekino K, Kubota T, Okada Y, Yamada Y, Yamamoto K, Horiuchi R, et al: Effect of the single CYP2C9*3 allele on pharmacokinetics and pharmacodynamics of losartan in healthy Japanese subjects. Eur J Clin Pharmacol, 59: 589–592, 2003 doi:10.1007/s00228-003-0664-5 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00228-003-0664-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=14504849&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000187147100004&link_type=ISI) 8. 8.Merck & Co. I: Cozaar prescribing information [package inset]. Food and Drug Administration, 1998 9. 9.Collins KS, Raviele ALJ, Elchynski AL, Woodcock AM, Zhao Y, Cooper-DeHoff RM, et al: Genotype-Guided Hydralazine Therapy. Am J Nephrol, 51: 764–776, 2020 doi:10.1159/000510433 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1159/000510433&link_type=DOI) 10. 10.Eadon MT, Chapman AB: A Physiologic Approach to the Pharmacogenomics of Hypertension. Advances in chronic kidney disease, 23: 91–105, 2016 doi:10.1053/j.ackd.2016.02.003 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1053/j.ackd.2016.02.003&link_type=DOI) 11. 11.Eadon MT, Kanuri SH, Chapman AB: Pharmacogenomic studies of hypertension: paving the way for personalized antihypertensive treatment. Expert Rev Precis Med Drug Dev, 3: 33–47, 2018 doi:10.1080/23808993.2018.1420419 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/23808993.2018.1420419&link_type=DOI) 12. 12.Muntner P, Anderson A, Charleston J, Chen Z, Ford V, Makos G, et al: Hypertension awareness, treatment, and control in adults with CKD: results from the Chronic Renal Insufficiency Cohort (CRIC) Study. Am J Kidney Dis, 55: 441–451, 2010 doi:10.1053/j.ajkd.2009.09.014 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1053/j.ajkd.2009.09.014&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19962808&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000275109000009&link_type=ISI) 13. 13.Carey RM, Sakhuja S, Calhoun DA, Whelton PK, Muntner P: Prevalence of Apparent Treatment-Resistant Hypertension in the United States. Hypertension, 73: 424–431, 2019 doi:10.1161/HYPERTENSIONAHA.118.12191 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1161/HYPERTENSIONAHA.118.12191&link_type=DOI) 14. 14.Collins KS, Pratt VM, Stansberry WM, Medeiros EB, Kannegolla K, Swart M, et al: Analytical validity of a genotyping assay for use with personalized antihypertensive and chronic kidney disease therapy. Pharmacogenet Genomics, 29: 18–22, 2019 doi:10.1097/FPC.0000000000000361 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/FPC.0000000000000361&link_type=DOI) 15. 15.Levey AS, Stevens LA, Schmid CH, Zhang YL, Castro AF, 3rd, Feldman HI, et al: A new equation to estimate glomerular filtration rate. Ann Intern Med, 150: 604–612, 2009 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.7326/0003-4819-150-9-200905050-00006&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19414839&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000265903800004&link_type=ISI) 16. 16.Spiech KM, Tripathy PR, Woodcock AM, Sheth NA, Collins KS, Kannegolla K, et al: Implementation of a Renal Precision Medicine Program: Clinician Attitudes and Acceptance. Life (Basel), 10, 2020 doi:10.3390/life10040032 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/life10040032&link_type=DOI) 17. 17.Tanner RM, Calhoun DA, Bell EK, Bowling CB, Gutierrez OM, Irvin MR, et al: Prevalence of apparent treatment-resistant hypertension among individuals with CKD. Clin J Am Soc Nephrol, 8: 1583–1590, 2013 doi:10.2215/CJN.00550113 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiY2xpbmphc24iO3M6NToicmVzaWQiO3M6ODoiOC85LzE1ODMiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8wNC8wNi8yMDIxLjAzLjMwLjIxMjU0NjY1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 18. 18.Arnett DK, Blumenthal RS, Albert MA, Buroker AB, Goldberger ZD, Hahn EJ, et al: 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol, 74: 1376–1414, 2019 doi:10.1016/j.jacc.2019.03.009 [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czo0OiJhY2NqIjtzOjU6InJlc2lkIjtzOjEwOiI3NC8xMC8xMzc2IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDQvMDYvMjAyMS4wMy4zMC4yMTI1NDY2NS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 19. 19.James PA, Oparil S, Carter BL, Cushman WC, Dennison-Himmelfarb C, Handler J, et al: 2014 evidence-based guideline for the management of high blood pressure in adults: report from the panel members appointed to the Eighth Joint National Committee (JNC 8). Jama, 311: 507–520, 2014 doi:10.1001/jama.2013.284427 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2013.284427&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24352797&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 20. 20.LLC AP: BiDil prescribing information [package insert], Available at: [https://www.accessdata.fda.gov/drugsatfda\_docs/label/2019/020727s010lbl.pdf](https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/020727s010lbl.pdf). Accessed June 8 21. 21.Corporation NP: Lopressor Prescribing Information [Package Insert]. United States Food and Drug Administration, 2008 22. 22.Swen JJ, Nijenhuis M, de Boer A, Grandia L, Maitland-van der Zee AH, Mulder H, et al: Pharmacogenetics: from bench to byte--an update of guidelines. Clin Pharmacol Ther, 89: 662–673, 2011 doi:10.1038/clpt.2011.34 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/clpt.2011.34&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21412232&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000289833000014&link_type=ISI) 23. 23.Turner ST, Bailey KR, Fridley BL, Chapman AB, Schwartz GL, Chai HS, et al: Genomic association analysis suggests chromosome 12 locus influencing antihypertensive response to thiazide diuretic. Hypertension, 52: 359–365, 2008 doi:10.1161/HYPERTENSIONAHA.107.104273 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1161/HYPERTENSIONAHA.107.104273&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18591461&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 24. 24.Gong Y, McDonough CW, Wang Z, Hou W, Cooper-DeHoff RM, Langaee TY, et al: Hypertension susceptibility loci and blood pressure response to antihypertensives: results from the pharmacogenomic evaluation of antihypertensive responses study. Circ Cardiovasc Genet, 5: 686–691, 2012 doi:10.1161/CIRCGENETICS.112.964080 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiY2lyY2N2ZyI7czo1OiJyZXNpZCI7czo3OiI1LzYvNjg2IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDQvMDYvMjAyMS4wMy4zMC4yMTI1NDY2NS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 25. 25.Carey RM, Calhoun DA, Bakris GL, Brook RD, Daugherty SL, Dennison-Himmelfarb CR, et al: Resistant Hypertension: Detection, Evaluation, and Management: A Scientific Statement From the American Heart Association. Hypertension, 72: e53–e90, 2018 doi:10.1161/HYP.0000000000000084 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1161/HYP.0000000000000084&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30354828&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 26. 26.Siu YA, Lai WG: Impact of Probe Substrate Selection on Cytochrome P450 Reaction Phenotyping Using the Relative Activity Factor. Drug Metab Dispos, 45: 183–189, 2017 doi:10.1124/dmd.116.073510 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiZG1kIjtzOjU6InJlc2lkIjtzOjg6IjQ1LzIvMTgzIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDQvMDYvMjAyMS4wMy4zMC4yMTI1NDY2NS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 27. 27.Sica DA, Gehr TW, Ghosh S: Clinical pharmacokinetics of losartan. Clin Pharmacokinet, 44: 797–814, 2005 doi:10.2165/00003088-200544080-00003 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2165/00003088-200544080-00003&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16029066&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000231190000003&link_type=ISI) 28. 28.Huang HX, Wu H, Zhao Y, Zhou T, Ai X, Dong Y, et al: Effect of CYP2C9 genetic polymorphism and breviscapine on losartan pharmacokinetics in healthy subjects. Xenobiotica: 1–16, 2021 doi:10.1080/00498254.2021.1880670 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/00498254.2021.1880670&link_type=DOI) 29. 29.Cabaleiro T, Roman M, Ochoa D, Talegon M, Prieto-Perez R, Wojnicz A, et al: Evaluation of the relationship between sex, polymorphisms in CYP2C8 and CYP2C9, and pharmacokinetics of angiotensin receptor blockers. Drug Metab Dispos, 41: 224–229, 2013 doi:10.1124/dmd.112.046292 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiZG1kIjtzOjU6InJlc2lkIjtzOjg6IjQxLzEvMjI0IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDQvMDYvMjAyMS4wMy4zMC4yMTI1NDY2NS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 30. 30.Bae JW, Choi CI, Lee HI, Lee YJ, Jang CG, Lee SY: Effects of CYP2C9\*1/\*3 and \*1/\*13 on the pharmacokinetics of losartan and its active metabolite E-3174. Int J Clin Pharmacol Ther, 50: 683–689, 2012 doi:10.5414/CP201467 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.5414/CP201467&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22735459&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 31. 31.Bae JW, Choi CI, Kim MJ, Oh DH, Keum SK, Park JI, et al: Frequency of CYP2C9 alleles in Koreans and their effects on losartan pharmacokinetics. Acta Pharmacol Sin, 32: 1303–1308, 2011 doi:10.1038/aps.2011.100 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/aps.2011.100&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21841812&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 32. 32.Joy MS, Dornbrook-Lavender K, Blaisdell J, Hilliard T, Boyette T, Hu Y, et al: CYP2C9 genotype and pharmacodynamic responses to losartan in patients with primary and secondary kidney diseases. Eur J Clin Pharmacol, 65: 947–953, 2009 doi:10.1007/s00228-009-0707-7 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00228-009-0707-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19669737&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 33. 33.Babaoglu MO, Yasar U, Sandberg M, Eliasson E, Dahl ML, Kayaalp SO, et al: CYP2C9 genetic variants and losartan oxidation in a Turkish population. Eur J Clin Pharmacol, 60: 337–342, 2004 doi:10.1007/s00228-004-0785-5 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00228-004-0785-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15197523&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 34. 34.Yasar U, Forslund-Bergengren C, Tybring G, Dorado P, Llerena A, Sjoqvist F, et al: Pharmacokinetics of losartan and its metabolite E-3174 in relation to the CYP2C9 genotype. Clin Pharmacol Ther, 71: 89–98, 2002 doi:10.1067/mcp.2002.121216 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1067/mcp.2002.121216&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11823761&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000173770500011&link_type=ISI) 35. 35.Hamadeh IS, Langaee TY, Dwivedi R, Garcia S, Burkley BM, Skaar TC, et al: Impact of CYP2D6 polymorphisms on clinical efficacy and tolerability of metoprolol tartrate. Clin Pharmacol Ther, 96: 175–181, 2014 doi:10.1038/clpt.2014.62 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/clpt.2014.62&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24637943&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 36. 36.Do AN, Lynch AI, Claas SA, Boerwinkle E, Davis BR, Ford CE, et al: The effects of genes implicated in cardiovascular disease on blood pressure response to treatment among treatment-naive hypertensive African Americans in the GenHAT study. J Hum Hypertens, 30: 549–554, 2016 doi:10.1038/jhh.2015.121 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/jhh.2015.121&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26791477&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 37. 37.Pacanowski MA, Gong Y, Cooper-Dehoff RM, Schork NJ, Shriver MD, Langaee TY, et al: beta-adrenergic receptor gene polymorphisms and beta-blocker treatment outcomes in hypertension. Clin Pharmacol Ther, 84: 715–721, 2008 doi:10.1038/clpt.2008.139 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/clpt.2008.139&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18615004&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000261235600018&link_type=ISI) 38. 38.Baudhuin LM, Miller WL, Train L, Bryant S, Hartman KA, Phelps M, et al: Relation of ADRB1, CYP2D6, and UGT1A1 polymorphisms with dose of, and response to, carvedilol or metoprolol therapy in patients with chronic heart failure. Am J Cardiol, 106: 402–408, 2010 doi:10.1016/j.amjcard.2010.03.041 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.amjcard.2010.03.041&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20643254&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 39. 39.Magvanjav O, McDonough CW, Gong Y, McClure LA, Talbert RL, Horenstein RB, et al: Pharmacogenetic Associations of beta1-Adrenergic Receptor Polymorphisms With Cardiovascular Outcomes in the SPS3 Trial (Secondary Prevention of Small Subcortical Strokes). Stroke, 48: 1337–1343, 2017 doi:10.1161/STROKEAHA.116.015936 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToic3Ryb2tlYWhhIjtzOjU6InJlc2lkIjtzOjk6IjQ4LzUvMTMzNyI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzA0LzA2LzIwMjEuMDMuMzAuMjEyNTQ2NjUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 40. 40.Johnson JA, Zineh I, Puckett BJ, McGorray SP, Yarandi HN, Pauly DF: Beta 1- adrenergic receptor polymorphisms and antihypertensive response to metoprolol. Clin Pharmacol Ther, 74: 44–52, 2003 doi:10.1016/S0009-9236(03)00068-7 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0009-9236(03)00068-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12844134&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000184023300006&link_type=ISI) 41. 41.Vandell AG, Lobmeyer MT, Gawronski BE, Langaee TY, Gong Y, Gums JG, et al: G protein receptor kinase 4 polymorphisms: beta-blocker pharmacogenetics and treatment-related outcomes in hypertension. Hypertension, 60: 957–964, 2012 doi:10.1161/HYPERTENSIONAHA.112.198721 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTU6Imh5cGVydGVuc2lvbmFoYSI7czo1OiJyZXNpZCI7czo4OiI2MC80Lzk1NyI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzA0LzA2LzIwMjEuMDMuMzAuMjEyNTQ2NjUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 42. 42.Bhatnagar V, O’Connor DT, Brophy VH, Schork NJ, Richard E, Salem RM, et al: G-protein-coupled receptor kinase 4 polymorphisms and blood pressure response to metoprolol among African Americans: sex-specificity and interactions. Am J Hypertens, 22: 332–338, 2009 doi:10.1038/ajh.2008.341 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ajh.2008.341&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19119263&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 43. 43.McDonough CW, Burbage SE, Duarte JD, Gong Y, Langaee TY, Turner ST, et al: Association of variants in NEDD4L with blood pressure response and adverse cardiovascular outcomes in hypertensive patients treated with thiazide diuretics. J Hypertens, 31: 698–704, 2013 doi:10.1097/HJH.0b013e32835e2a71 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/HJH.0b013e32835e2a71&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23353631&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000315952100010&link_type=ISI) 44. 44.Svensson-Farbom P, Wahlstrand B, Almgren P, Dahlberg J, Fava C, Kjeldsen S, et al: A functional variant of the NEDD4L gene is associated with beneficial treatment response with beta-blockers and diuretics in hypertensive patients. J Hypertens, 29: 388–395, 2011 doi:10.1097/HJH.0b013e3283410390 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/HJH.0b013e3283410390&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21052022&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F06%2F2021.03.30.21254665.atom) 45. 45.Rimpela JM, Kontula KK, Fyhrquist F, Donner KM, Tuiskula AM, Sarin AP, et al: Replicated evidence for aminoacylase 3 and nephrin gene variations to predict antihypertensive drug responses. Pharmacogenomics, 18: 445–458, 2017 doi:10.2217/pgs-2016-0204 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2217/pgs-2016-0204&link_type=DOI) 46. 46.Hiltunen TP, Donner KM, Sarin AP, Saarela J, Ripatti S, Chapman AB, et al: Pharmacogenomics of hypertension: a genome-wide, placebo-controlled cross-over study, using four classes of antihypertensive drugs. J Am Heart Assoc, 4: e001521, 2015 doi:10.1161/JAHA.115.001778 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NToiYWhhb2EiO3M6NToicmVzaWQiO3M6MTE6IjQvMS9lMDAxNTIxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDQvMDYvMjAyMS4wMy4zMC4yMTI1NDY2NS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 47. 47.Shahin MH, Sa AC, Webb A, Gong Y, Langaee T, McDonough CW, et al: Genome-Wide Prioritization and Transcriptomics Reveal Novel Signatures Associated With Thiazide Diuretics Blood Pressure Response. Circ Cardiovasc Genet, 10, 2017 doi:10.1161/CIRCGENETICS.116.001404 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiY2lyY2N2ZyI7czo1OiJyZXNpZCI7czoxMjoiMTAvMS9lMDAxNDA0IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDQvMDYvMjAyMS4wMy4zMC4yMTI1NDY2NS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 48. 48.Duarte JD, Turner ST, Tran B, Chapman AB, Bailey KR, Gong Y, et al: Association of chromosome 12 locus with antihypertensive response to hydrochlorothiazide may involve differential YEATS4 expression. Pharmacogenomics J, 13: 257–263, 2013 doi:10.1038/tpj.2012.4 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/tpj.2012.4&link_type=DOI)