Effect of AGTR1 and BDKRB2 gene polymorphisms on atorvastatin metabolism in a Mexican population

  • Authors:
    • Sarahí Herrera‑González
    • Denisse Aideé Martínez‑Treviño
    • Marcelino Aguirre‑Garza
    • Magdalena Gómez‑Silva
    • Hugo Alberto Barrera‑Saldaña
    • Rafael Baltazar Reyes León‑Cachón
  • View Affiliations

  • Published online on: October 25, 2017     https://doi.org/10.3892/br.2017.1009
  • Pages: 579-584
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Discrepancies in the response to drugs are partially due to polymorphisms in genes involved in drug metabolism and transport. The frequency, pattern and impact of these polymorphisms vary among populations. In the present study, the pharmacokinetics and pharmacogenetics of atorvastatin (ATV) in a Mexican population were investigated. The study cohort exhibited differing ATV metabolizing phenotypes, and in subsequent allelic discrimination assays, single nucleotide polymorphisms in the angiotensinogen, angiotensin II type 1 receptor (AGTR1) and bradykinin B2 receptor (BDKRB2) genes were genotyped and their effects on the pharmacokinetic parameters of ATV were assessed. Additionally, association studies were performed to test for a correlation between metabolizing phenotypes and genetic variants. It was observed that carriers of the genotypes A/C and C/T in AGTR1 and BDKRB2 had higher area under the plasma concentration‑time curve values from time 0 to the time of the last measurement and from time 0 extrapolated to infinity, and lower values of clearance of the fraction dose absorbed compared with homozygous carriers (P<0.05). Only the C/C genotype of BDKRB2 was associated with the fast metabolizer phenotype. These data suggest that AGTR1 and BDKRB2 are involved in ATV pharmacokinetics; a novel finding that requires confirmation in further studies.

Introduction

The prevalence of chronic degenerative diseases has increased in the adult Mexican population (1,2). In Mexico, cardiovascular disease (CVD) was a leading cause of death in 2015 (3), while hypercholesterolemia, a major risk factor for CVD, was the most prevalent type of dyslipidemia in the Mexican population between 2003 and 2005 (4). Statins are cholesterol-lowering drugs, and in 2012, atorvastatin (ATV) was the most frequently prescribed statin in Mexico (5). Within liver cells, ATV disrupts cholesterol biosynthesis by blocking 3-hydroxy-3-methylglutaryl-coenzyme A reductase, which reduces the amount of cholesterol released into the blood. As a consequence, low-density lipoprotein cholesterol uptake by liver cells increases and blood cholesterol levels diminish (6). However, in clinical trials of ATV, pharmacokinetic parameters including maximum plasma concentration (Cmax), time to reach Cmax (Tmax), area under the plasma concentration-time curve (AUC) from time 0 to the time of last measurement (AUC0-t), AUC from time 0 extrapolated to infinity (AUC0-∞), apparent clearance of the fraction dose absorbed (Cl/F), elimination rate constant in the terminal drug phase (Ke) and the half-life in the terminal drug phase (T1/2) are variable (7). This reflects the underlying variability in the absorption, distribution, metabolism and excretion (ADME) processes of ATV, which may affect the pharmacological response (8). Although in general, genetic factors influence ~30% of variation in drug disposition and response (9,10), recent results have indicated that genetic variability may contribute to >90% of the variance in ATV plasma concentrations (11). These differences in the ADME characteristics of ATV have been attributed to polymorphisms in genes associated with drug pharmacokinetics, particularly those encoding enzymes and transporters (7,10,11).

The anti-inflammatory effect of statins has been investigated (12). The angiotensin II type 1 receptor (AGTR1) blocks the angiotensin II pathway and has been associated with the development of atherosclerosis (12). In addition, polymorphisms in AGTR1 have been associated with muscle toxicity in patients treated with statins (13). In addition to AGTR1, angiotensinogen (AGT) is part of the renin-angiotensin-aldosterone system (14). An improved response to diuretics has been observed in the presence of the AGTR1 A1166C and AGT G-6A polymorphisms in African-American women and a Chinese population (15,16).

The kallikrein-kinin system is also involved in multiple cardiovascular events; it modulates the renin-angiotensin-aldosterone system, promotes vasodilation, modulates neovascularization and stimulates the inflammatory response (17). Genetic variants in the bradykinin B2 receptor (BDKRB2) and endothelial nitric oxide synthase (eNOS) genes have been associated with CVD risk (18,19). Notably, the BDKRB2 C(−58)T polymorphism has been associated with hypertension in an Asian population; carriers of the C/C genotype had an increased risk, whereas carriers of the T/T genotype had a decreased risk. However, in Asian heterozygous carriers, Americans and Europeans, no association has been identified (20), though an improved response to enalapril for the treatment of hypertension has been observed in individuals with the C/C genotype (21).

Although polymorphisms in the AGTR1, AGT and BDKRB2 genes have been described, there is a lack of studies on their frequency and effect on ATV pharmacokinetics. Therefore, the present study aimed to: i) Identify novel polymorphic variants influencing the pharmacokinetic parameters of ATV; and ii) associate genotypes with metabolizing phenotypes.

Materials and methods

Design

A randomized clinical study was conducted in 60 healthy volunteers of Mexican origin to assess the bioequivalence of a single oral dose (80 mg) of ATV (coated tablets; Pfizer, Inc., New York, NY, USA) (7). The study was performed according to the guidelines of the Declaration of Helsinki (22), of Tokyo for Good Clinical Practice Standards (23), and to Mexican regulations for studies of bioavailability and bioequivalence (24). The clinical protocol was approved by the Research and Ethics Committee of the Clinical and Experimental Pharmacology Center, Ipharma S.A. (Monterrey, Mexico), and the pharmacogenetic procedure was approved by the Ethics, Research and Biosecurity Committees of the University of Monterrey (Monterrey, Mexico). The study was registered with the Federal Commission for Protection Against Health Risks under code Atorvastatina/A95-10Bis and in the Register of Clinical Trials of Australia and New Zealand (registration no. ACTRN12614000851662). Written informed consent was obtained from all subjects.

Study population

As described in our preliminary pilot study (7), a total of 60 healthy male volunteers of Mexican origin were included in the study from January 2011 to February 2011, with a mean age of 24.01±4.35 years. The inclusion criteria were as follows: Non-smoker; 18–45 years old; weight, ≥50 kg; body mass index, 20–26 kg/m2; availability to complete the study and normal health status (free from disease). Health status was assessed based on physical examination, medical history and clinical and biochemical tests. Insufficiency in any requirement (abnormal laboratory results, drug abuse, ingestion of alcohol 1 week prior to the study, prescription or over-the-counter medication prior to enrollment and reluctance to complete the study) was reason for exclusion from the study. Women were excluded as ATV is classified as a pregnancy category X drug (25). All subjects were informed of the aims of the study.

Sampling

ATV administration and blood sampling were performed as described in the pilot study (7). Briefly, peripheral blood (4 ml) was collected in K2EDTA-coated BD Vacutainers® (BD Diagnostics, Franklin Lakes, NJ, USA) at different time points: Prior to drug administration (time 0) and at 17 time points (0.25, 0.5, 0.75, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 12, 24, 36 and 48 h) after drug administration. The plasma was used for pharmacokinetic analysis and DNA was isolated from blood cells using an alkaline lysis method (26). Genomic DNA was quantified by UV absorbance using a Nanodrop 1000 Spectrophotometer (Thermo Fisher Scientific, Inc., Waltham, MA, USA). An absorbance 260/280 ratio between 1.8 and 2 was considered of adequate quality for subsequent use. The DNA concentration was adjusted to 10 ng/µl and stored at −20°C until analysis.

Pharmacokinetic analysis

ATV plasma concentrations were measured by high-performance liquid chromatography tandem mass spectrometry with an Agilent 1100 system (Agilent Technologies, Inc., Santa Clara, CA, USA) using a method validated by Ipharma S.A. (7,27,28). The Cmax and Tmax parameters were obtained from the concentration-time data of the plasma. Pharmacokinetic parameters including AUC0-t, AUC0-∞, Cl/F, Ke, and T1/2, were calculated with a non-compartmental method (29) using WinNonlin® software v5.3 (Pharsight Corp., Mountain View, CA, USA) as described in the pilot study (7).

Metabolic phenotype classification

The metabolizer phenotypes were determined according to the results of a multivariate analysis of the combined pharmacokinetic parameters Cmax and AUC0-t (7). First, Cmax and AUC0-t were standardized to minimize the effect of scale differences, and a distance matrix was made from the combined standardized Cmax and AUC0-t values. Subsequently, hierarchical cluster analysis (HCA) using the Ward linkage method (30) was performed on individual Cmax and AUC0-t values. Finally, the interindividual Manhattan distances were computed. Minitab 16 software (Minitab Inc., State College, PA, USA) was used for standardization and HCA.

Pharmacogenetic tests

DNA samples were genotyped for the polymorphisms AGT-rs699, AGTR1-rs5186 and BDKRB2-rs1799722 using real-time polymerase chain reaction and Taqman® probes (Applied Biosystems; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Three quality controls thresholds were applied: A genotype call rate equal to 1.0, a Hardy-Weinberg equilibrium (HWE) test with P>0.05, and a minor allele frequency of >0.01.

Statistical analysis

The HWE was determined by comparing the genotype frequencies with the expected values using the maximum likelihood method (31). All statistical analysis was performed with SPSS v20 software (IBM Corp., Armonk, NY, USA). To assess the effects of polymorphisms on the ATV pharmacokinetic parameters, comparisons between two and three groups were made. The Student's t-test and one-way analysis of variance were used for parametric distributions, while Mann-Whitney U and Kruskal-Wallis H tests were used for nonparametric distributions. To confirm the contribution of genetic factors to the variability of pharmacokinetic parameters, linear regression analysis was performed. Possible associations of genotypes or combinations of genotypes with phenotypes were evaluated using χ2 and Fisher's exact tests. Linear regression and associations were assessed under three different models (dominant, over-dominant and recessive) (32). The odds ratio (OR) was estimated with a 95% confidence interval (95% CI). All P-values were two-tailed. Corrected P-values (Pc) were obtained using the Bonferroni correction for exclusion of spurious associations. P<0.05 was considered to indicate statistical significance.

Results

Metabolic phenotype classification

As reported in our previous study (7), the classification of metabolizer phenotypes, based on the combination of the Cmax and AUC0-t parameters, identified three ATV metabolizer phenotypes: Slow metabolizers (30.00%), normal metabolizers (41.66%) and fast metabolizers (28.33%). The Cmax and AUC0-t parameters used for the classification were significantly different between the three phenotypes (7); the parameters were significantly higher for the slow phenotype compared with the normal and fast phenotypes, and significantly higher for the normal phenotype compared with the fast phenotype (P<0.05; Fig. 1). None of the subjects reported any side effects (7).

Pharmacogenetic tests

Allele frequencies of the genetic polymorphisms were consistent with HWE (P>0.05). All genetic polymorphisms satisfied the quality control tests.

Association between genotypes and ATV pharmacokinetics

There was no significant effect of the AGT-rs699 polymorphism on ATV pharmacokinetic parameters (Table I). Conversely, AUC0-t and AUC0-∞ values were significantly higher in individuals with the heterozygous genotype (A/C) of the AGTR1-rs5186 polymorphism when compared with the C/C genotype (P<0.05). In addition, the AUC0-∞ of the A/C genotype was increased compared with that observed for the combination of the homozygous wild-type and homozygous variant alleles (A/A+C/C; P<0.05). In heterozygous carriers, Cl/F values were significantly lower than those observed for homozygous variant allele carriers (C/C) and for the combination of homozygous alleles (A/A+C/C; P<0.05; Table I and Fig. 2A and B).

Table I.

Effect of polymorphisms on the pharmacokinetic parameters of atorvastatin.

Table I.

Effect of polymorphisms on the pharmacokinetic parameters of atorvastatin.

GenotypesNCmax (ng/ml)AUC0-t (ng/ml/h)AUC0-∞ (ng/ml/h)Cl/F (l/h)KeT1/2 (h)
AGT-rs699A/A4 45.72±11.03 109.72±38.89 121.44±43.81 729.06±266.90 0.05±0.02 14.97±9.68
A/G29 43.21±21.06 141.98±51.00 159.05±56.37 588.08±279.10 0.07±0.04 12.43±7.99
G/G27 51.20±26.65 189.40±11.65 200.34±110.61 516.63±246.52 0.08±0.02 9.22±3.25
A/A+A/G33 43.51±20.00 138.07±50.32 154.49±55.82 605.17±277.53 0.07±0.04 12.74±8.09
AGTR1-rs5186A/A34 44.14±22.86 155.21±83.44 166.63±81.73 585.92±269.36 0.08±0.03 9.78±5.53
A/C21 54.80±23.47 186.44±92.45a 204.55±94.76a 473.67±220.44a 0.06±0.03 13.49±8.12
C/C5 33.36±18.79 95.57±43.10 109.28±40.84 810.19±275.81 0.07±0.02 10.72±3.30
A/A+C/C39 42.76±22.46 147.56±81.54 159.28±79.71b 614.68±277.11b 0.08±0.03 9.90±5.27
BDKRB2-rs1799722C/C24 44.00±27.21 151.53±103.77 163.00±102.00 649.67±316.78 0.09±0.02 8.78±3.27
C/T27 51.80±20.93 180.66±78.48c 194.17±77.38d 469.55±168.36e 0.07±0.04 12.64±7.21
T/T9 40.40±17.73 128.38±42.26 150.31±66.35 627.71±287.80 0.06±0.02 13.05±9.53
C/C+T/T33 43.02±24.76 145.22±91.08 159.54±92.80 643.68±304.85 0.08±0.02 9.94±5.84

{ label (or @symbol) needed for fn[@id='tfn1-br-0-0-1009'] } Data are presented as the mean ± standard deviation.

a P =0.015 (A/C vs. C/C)

b P =0.040 (A/A+C/C vs. A/C)

c P =0.021 (C/C+T/T vs. C/T)

d P =0.023 (C/C+T/T vs. C/T)

e P =0.007 (C/C+T/T vs. C/T). AGT, AGTR1, angiotensin II type 1 receptor; BDKRB2, bradykinin B2 receptor; Cmax, maximum plasma concentration; AUC, area under the plasma concentration-time curve; AUC0-t, AUC from time 0 to the time of last measurement; AUC0-∞, AUC from time 0 extrapolated to infinity; Cl/F, apparent clearance of the fraction dose absorbed; Ke, elimination rate constant in the terminal drug phase; T1/2, half-life in the terminal drug phase.

For BDKRB2-rs1799722, carriers of the heterozygous genotype (C/T) were identified to have significantly higher values of AUC0-t (P=0.021) and AUC0-∞ (P=0.023) and lower values of Cl/F (P=0.007) compared with those obtained for the combination of homozygous alleles (C/C+T/T; Table I and Fig. 2C and D).

The linear regression analysis under the over-dominant genetic model indicated that the AGTR1-rs5186 polymorphism significantly affected the values of T1/2 (adjusted R2=0.053, P=0.043); however, on comparison of the means by genotype, no significant differences were observed. Similarly, the BDKRB2-rs1799722 polymorphism significantly affected the Cl/F values (adjusted R2=0.093, P=0.01; data not shown).

Association between genotypes and metabolizer phenotypes

Of the two polymorphisms with an effect on ATV pharmacokinetics, BDKRB2-rs1799722 was associated with fast metabolism when considering genetic models; association analysis using the dominant model identified that the C/C genotype of BDKRB2-rs1799722 was associated with the fast metabolizer phenotype (OR, 0.47; 95% CI, 0.26–0.83), whereas the C/T+T/T combination was associated with the normal and slow phenotypes (OR, 1.98; 95% CI, 1.01–3.88; P=0.014). When using the over-dominant model, the C/C+T/T combination was associated with the fast metabolizer phenotype (OR, 0.68; 95% CI, 0.40–0.92) and C/T was associated with the normal and slow phenotypes (OR, 2.27; 95% CI, 0.92–5.60; P=0.036). However, following Bonferroni's correction, only the associations under the dominant model remained statistically significant (Pc=0.03; Table II).

Table II.

Association between genotypes and metabolizer phenotypes.

Table II.

Association between genotypes and metabolizer phenotypes.

GenePolymorphismModelOR (95% CI)P-valuePc-value
BDKRB2rs1799722Dominant (C/C vs. C/T+T/T)C/C: Fast metabolizers0.0140.03
0.47 (0.26–0.83)
C/T+T/T: Normal/slow metabolizers
1.98 (1.01–3.88)
BDKRB2rs1799722Over-dominant (C/C+T/T vs. C/T)C/C+T/T: Fast metabolizers0.0360.07
0.68 (0.40–0.92)
C/T: Normal/slow metabolizers
2.27 (0.92–5.60)

[i] BDKRB2, bradykinin B2 receptor; OR, odds ratio; CI, confidence interval; Pc, Bonferroni-corrected P-values.

Discussion

The pharmacokinetic parameters of ATV, namely Cmax and AUC, may vary by >10-fold (7,33). Following analysis of pharmacokinetic discrepancies, three major metabolizer phenotypes of ATV have been identified in Chinese and Mexican populations (7,33). This variation has been associated with polymorphisms in genes encoding drug metabolizing enzymes and transporters (34). However, differences in allele frequencies and their effect on quantitative parameters including cholesterol levels, arterial pressure and pharmacokinetic parameters, and associations of genotypes with drug metabolism and response have been documented across various populations (7,33,35,36).

In the current study, the effect on ATV pharmacokinetics of three polymorphic variants in genes related to drug metabolism and response were evaluated. AGT-rs699 had no significant effect on pharmacokinetic parameters. However, to the best of our knowledge, the study is the first to identify an effect of AGTR1-rs5186 and BDKRB2-rs1799722 on ATV pharmacokinetics.

Heterozygous carriers of rs5186 (A/C) exhibited higher AUC0-t and AUC0-∞ values, while having lower Cl/F values, than homozygous carriers (C/C), which thus indicates a diminished clearance activity and longer permanence of ATV in heterozygous carriers. The effect of the AGTR1 polymorphism on T1/2 was consistent with this interpretation. This decreased clearance and longer exposure to ATV may lead to an improved response to drug therapy, or to an adverse effect. By contrast, carriers of homozygous genotypes (A/A or C/C) had increased clearance, and thus may have a poorer response to treatment. However, there was no association between the A/A or C/C genotypes with the fast metabolizer phenotype. To the best of our knowledge, this is the first report on the influence of rs5186 on ATV pharmacokinetics, though the rs5186 polymorphism has previously been associated with lipid levels (37), and ATV, as a statin, has lipid-lowering effects (38). Consistent with the present results, the C/C genotype has been related to higher levels of triglycerides in a healthy Malayan population (37). Additionally, in a case-control study conducted in a Northern Indian population, the C/C genotype was associated with essential hypertension and higher gene expression of AGTR1 (39). Regarding the anti-inflammatory effect of statins, ATV may affect activation of the angiotensin pathway through AGTR1 by attenuating the activity of angiotensin II (ANG II), as observed in rats, whereby ATV modulated ANG II-induced expression of inflammatory and fibrogenic genes in the liver (40); however there is a lack of data regarding the association of AGTR1-rs5186 with the anti-inflammatory response.

Regarding BDKRB2-rs1799722, significant differences in the AUC0-t, AUC0-∞ and Cl/F parameters were identified between heterozygous carriers (C/T) and homozygous carriers (C/C or T/T). The current results suggest that the BDKRB2-rs1799722 polymorphism affects ATV clearance activity. This effect was demonstrated by linear regression and association analyses under over-dominant and dominant models. Notably, it was indicated that the C allele promotes fast metabolism, while accumulation of the T allele leads to a shift towards slower metabolism. However, the pharmacokinetic parameters of the T/T homozygous carriers did not differ significantly compared with heterozygous carriers. To the best of our knowledge, BDKRB2-rs1799722 has not previously been associated with statin metabolism. However, a meta-analysis identified that the C allele of rs1799722 increased the risk of hypertension in Asian and African-American populations (20). A pharmacogenetic study conducted in a Brazilian population revealed that carriers of the C allele responded to Enalapril, an antihypertensive drug that serves as an inhibitor of angiotensin-converting enzyme (21). The statins are also established for their antihypertensive effects in hypercholesterolemic patients (41). Nonetheless, there is a lack of studies into the effect of BDKRB2-rs1799722 on the metabolism or response of patients to statins.

Although a number of polymorphisms have been suggested as candidate responsible for the pharmacokinetic variability of ATV, the present study is the first to indicate the involvement of AGTR1-rs5186 and BDKRB2-rs1799722. The inclusion of these biomarkers in future studies may improve the prediction of the pharmacokinetic variability of ATV or decrease the number of variants required for prediction (7,11). While the current study demonstrated the contribution of genetic polymorphisms to ATV pharmacokinetics, there were a number of limitations. Firstly, there was a lack of data, such as cholesterol levels at one month post-treatment, for complete analysis of polymorphism effect on response. Secondly, in some cases the number of subjects per genotype was small, and thus associations may have been lost following correction. To validate the results, further studies should be performed in a larger population. In addition, the influence of other genes may explain the lack of association with the slow metabolizer phenotype.

In conclusion, a significant effect of AGTR1-rs5186 and BDKRB2-rs1799722 on ATV pharmacokinetics was detected. The present findings suggest that the A/C genotype of AGTR1-rs5186 is associated with slow ATV metabolism, while the C/C or A/A+C/C genotypes are associated with fast metabolism. Additionally, the C/T genotype of BDKRB2-rs1799722 may be associated with the slow metabolizer phenotype, while the homozygous genotypes may be associated with the fast metabolizer phenotype. These novel findings increase the panel of potential genetic biomarkers associated with ATV metabolism, and should be verified in future pharmacogenetic studies in larger populations with different genetic backgrounds.

Acknowledgements

The authors would like to thank the University of Monterrey, Italy, for funding the current study (grant no. UIN15009) and Dr Irene Meester from the University of Monterrey for reviewing and improving the manuscript.

References

1 

Valdez Morales M, Medina Godoy S, Chacón López MA and Espinosa Alonso LG: Comprehensive approach of diet importance on health status of the Mexican population. Biotecnia. 18:102016. View Article : Google Scholar

2 

Kuri-Morales PA: La transición en salud y su impacto en la demanda de servicios. Gac Med Mex. 147:451–454. 2011.(In Spanish). PubMed/NCBI

3 

National Institute of Statistics and Geography (INEGI), . Mortality Statistics. INEGI, Mexico City. 2015.http://www.beta.inegi.org.mx/contenidos/proyectos/registros/vitales/mortalidad/doc/presentacion.pdfJune 15–2016(In Spanish).

4 

Escobedo-de la Peña J, de Jesús-Pérez R, Schargrodsky H and Champagne B: Prevalence of dyslipidemias in Mexico city and Its relation to other cardiovascular risk factors. Results from the CARMELA study. Gac Med Mex. 150:128–136. 2014.(In Spanish).

5 

Canalizo-Miranda E, Favela-Pérez EA, Salas-Anaya JA, Gómez-Díaz R, Jara-Espino R, Del Pilar Torres-Arreola L and Viniegra-Osorio A: Clinical practice guideline. Diagnosis and treatment of dyslipidemia. Rev Med Inst Mex Seguro Soc. 51:700–709. 2013.(In Spanish).

6 

McFarland AJ, Anoopkumar-Dukie S, Arora DS, Grant GD, McDermott CM, Perkins AV and Davey AK: Molecular mechanisms underlying the effects of statins in the central nervous system. Int J Mol Sci. 15:20607–20637. 2014. View Article : Google Scholar : PubMed/NCBI

7 

León-Cachón RBR, Ascacio-Martínez JA, Gamino-Peña ME, Cerda-Flores RM, Meester I, Gallardo-Blanco HL, Gómez-Silva M, Piñeyro-Garza E and Barrera-Saldaña HA: A pharmacogenetic pilot study reveals MTHFR, DRD3, and MDR1 polymorphisms as biomarker candidates for slow atorvastatin metabolizers. BMC Cancer. 16:742016. View Article : Google Scholar : PubMed/NCBI

8 

León-Cachón RBR, Ascacio-Martínez JAI, Gómez-Silva M, Piñeyro-Garza E, González-González JG, Pogue G, Simón-Buela L and Barrera-Saldaña HA: Application of genomic technologies in clinical pharmacology research. Rev Inves Clin. 67:212–218. 2015.

9 

US Food and Drug Administration, . Draft guidance on atorvastatin calcium and ezetimibe. US Department of Health and Human Services; Silver Spring, MD: 2014

10 

León-Cachón RB1, Ascacio-Martínez JA and Barrera-Saldaña HA: Individual response to drug therapy: Bases and study approaches. Rev Invest Clin. 64:364–376. 2012.PubMed/NCBI

11 

Cruz-Correa OF, León-Cachón RB, Barrera-Saldaña HA and Soberón X: Prediction of atorvastatin plasmatic concentrations in healthy volunteers using integrated pharmacogenetics sequencing. Pharmacogenomics. 18:121–131. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Ma Y, Chen Z, Zou Y and Ge J: Atorvastatin represses the angiotensin 2-induced oxidative stress and inflammatory response in dendritic cells via the PI3K/Akt/Nrf 2 pathway. Oxid Med Cell Longev. 2014:1487982014. View Article : Google Scholar : PubMed/NCBI

13 

Ruaño G, Thompson PD, Windemuth A, Smith A, Kocherla M, Holford TR, Seip R and Wu AH: Physiogenomic analysis links serum creatine kinase activities during statin therapy to vascular smooth muscle homeostasis. Pharmacogenomics. 6:865–872. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Peters BJ, Klungel OH, de Boer A, Ch Stricker BH and Maitland-van der Zee AH: Pharmacogenetics of cardiovascular drug therapy. Clin Cases Miner Bone Metab. 6:55–65. 2009.PubMed/NCBI

15 

Frazier L, Turner ST, Schwartz GL, Chapman AB and Boerwinkle E: Multilocus effects of the renin-angiotensin-aldosterone system genes on blood pressure response to a thiazide diuretic. Pharmacogenomics J. 4:17–23. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Jiang X, Sheng HH, Lin G, Li J, Lu XZ, Cheng YL, Huang J, Xiao HS and Zhan YY: Effect of renin-angiotensin-aldosterone system gene polymorphisms on blood pressure response to antihypertensive treatment. Chin Med J (Engl). 120:782–786. 2007.PubMed/NCBI

17 

Bryant JW and Shariat-Madar Z: Human plasma kallikrein-kinin system: Physiological and biochemical parameters. Cardiovasc Hematol Agents Med Chem. 7:234–250. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Bentley JP, Asselbergs FW, Coffey CS, Hebert PR, Moore JH, Hillege HL and van Gilst WH: Cardiovascular risk associated with interactions among polymorphisms in genes from the renin-angiotensin, bradykinin, and fibrinolytic systems. PLoS One. 5:e127572010. View Article : Google Scholar : PubMed/NCBI

19 

Pal GK, Adithan C, Umamaheswaran G, Pal P, Nanda N, Indumathy J and Syamsunder AN: Endothelial nitric oxide synthase gene polymorphisms are associated with cardiovascular risks in prehypertensives. J Am Soc Hypertens. 10:865–872. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Luo K, Kang W and Xu G: The risk of bradykinin B2 receptor-58T/C gene polymorphism on hypertension: A meta-analysis. Int J Clin Exp Med. 8:19917–19927. 2015.PubMed/NCBI

21 

Silva PS, Fontana V, Luizon MR, Lacchini R, Silva WA Jr, Biagi C and Tanus-Santos JE: eNOS and BDKRB2 genotypes affect the antihypertensive responses to enalapril. Eur J Clin Pharmacol. 69:167–177. 2013. View Article : Google Scholar : PubMed/NCBI

22 

World Medical Association, . World Medical Association Declaration of Helsinki: Ethical principles for medical research involving human subjects. JAMA. 310:2191–2194. 2013. View Article : Google Scholar : PubMed/NCBI

23 

World Medical Association, . WMA Declaration of Tokyo - Guidelines for physicians concerning torture and other cruel, inhuman or degrading treatment or punishment in relation to detention and imprisonment. 29th WMA General Assembly. Tokyo, Japan. 1975;

24 

Solorzano-Flores LI: Official Mexican Standard NOM-177-SSA1-1998, establishing tests and procedures to demonstrate that a drug is interchangeable. Requirements must be subject to third party authorized to perform the tests. Secretaria de Salud, Mexico. 1999.

25 

Briggs GG, Freeman RK, Towers CV and Forinash AB: Drugs in Pregnancy and Lactation. 11th. Williams & Wilkins; Philadelphia, PA: 2017

26 

Sambrook J and Russell DW: Preparation and analysis of eukaryotic genomic DNAMolecular Cloning: A Laboratory Manual. 3rd. Cold Spring Harbor Laboratory Press; New York, NY: 2001

27 

Ahmed T, Kollipara S, Gautam A, Gigras R, Kothari M, Saha N, Batra V and Paliwal J: Bioavailability and interaction potential of atorvastatin and losartan on co-administration in healthy human subjects. J Bioequiv Availab. 1:18–27. 2009.

28 

Stanisz B and Kania L: Validation of HPLC method for determination of atorvastatin in tablets and for monitoring stability in solid phase. Acta Pol Pharm. 63:471–476. 2006.PubMed/NCBI

29 

Rowland M and Tozer TN: Clinical Pharmacokinetics and Pharmacodynamics: Concepts and Applications. 4th. Williams & Willkins; Philadelphia, PA: 2017

30 

Ward JH Jr: Hierarchical grouping to optimize an objective function. J Am Stat Assoc. 58:236–244. 1963. View Article : Google Scholar

31 

Reed TE and Schull WJ: A general maximum likelihood estimation program. Am J Hum Genet. 20:579–580. 1968.PubMed/NCBI

32 

Horita N and Kaneko T: Genetic model selection for a case-control study and a meta-analysis. Meta Gene. 5:1–8. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Huang Q, Aa J, Jia H, Xin X, Tao C, Liu L, Zou B, Song Q, Shi J, Cao B, et al: A Pharmacometabonomic approach to predicting metabolic phenotypes and pharmacokinetic parameters of atorvastatin in healthy volunteers. J Proteome Res. 14:3970–3981. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Niemi M: Transporter pharmacogenetics and statin toxicity. Clin Pharmacol Ther. 87:130–133. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Kadam P, Ashavaid TF, Ponde CK and Rajani RM: Genetic determinants of lipid-lowering response to atorvastatin therapy in an Indian population. J Clin Pharm Ther. 41:329–333. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Prado Y, Zambrano T and Salazar LA: Transporter genes ABCG2 rs2231142 and ABCB1 rs1128503 polymorphisms and atorvastatin response in Chilean subjects. J Clin Pharm Ther. Aug 19–2017.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

37 

Yap RWK, Shidoji Y, Yap WS and Masaki M: Association and interaction effect of AGTR1 and AGTR2 gene polymorphisms with dietary pattern on metabolic risk factors of cardiovascular disease in Malaysian adults. Nutrients. 9:E8532017. View Article : Google Scholar : PubMed/NCBI

38 

Isley WL, Miles JM, Patterson BW and Harris WS: The effect of high-dose simvastatin on triglyceride-rich lipoprotein metabolism in patients with type 2 diabetes mellitus. J Lipid Res. 47:193–200. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Chandra S, Narang R, Sreenivas V, Bhatia J, Saluja D and Srivastava K: Association of angiotensin II type 1 receptor (A1166C) gene polymorphism and its increased expression in essential hypertension: A case-control study. PLoS One. 9:e1015022014. View Article : Google Scholar : PubMed/NCBI

40 

Moreno M, Ramalho LN, Sancho-Bru P, Ruiz-Ortega M, Ramalho F, Abraldes JG, Colmenero J, Dominguez M, Egido J, Arroyo V, et al: Atorvastatin attenuates angiotensin II-induced inflammatory actions in the liver. Am J Physiol Gastrointest Liver Physiol. 296:G147–G156. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Morgado M, Rolo S, Macedo AF and Castelo-Branco M: Association of statin therapy with blood pressure control in hypertensive hypercholesterolemic outpatients in clinical practice. J Cardiovasc Dis Res. 2:44–49. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2017
Volume 7 Issue 6

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Herrera‑González S, Martínez‑Treviño DA, Aguirre‑Garza M, Gómez‑Silva M, Barrera‑Saldaña HA and León‑Cachón RB: Effect of AGTR1 and BDKRB2 gene polymorphisms on atorvastatin metabolism in a Mexican population. Biomed Rep 7: 579-584, 2017
APA
Herrera‑González, S., Martínez‑Treviño, D.A., Aguirre‑Garza, M., Gómez‑Silva, M., Barrera‑Saldaña, H.A., & León‑Cachón, R.B. (2017). Effect of AGTR1 and BDKRB2 gene polymorphisms on atorvastatin metabolism in a Mexican population. Biomedical Reports, 7, 579-584. https://doi.org/10.3892/br.2017.1009
MLA
Herrera‑González, S., Martínez‑Treviño, D. A., Aguirre‑Garza, M., Gómez‑Silva, M., Barrera‑Saldaña, H. A., León‑Cachón, R. B."Effect of AGTR1 and BDKRB2 gene polymorphisms on atorvastatin metabolism in a Mexican population". Biomedical Reports 7.6 (2017): 579-584.
Chicago
Herrera‑González, S., Martínez‑Treviño, D. A., Aguirre‑Garza, M., Gómez‑Silva, M., Barrera‑Saldaña, H. A., León‑Cachón, R. B."Effect of AGTR1 and BDKRB2 gene polymorphisms on atorvastatin metabolism in a Mexican population". Biomedical Reports 7, no. 6 (2017): 579-584. https://doi.org/10.3892/br.2017.1009