Abstract
In developed countries cardiovascular disease is one of the leading causes of death. Cardiovascular drugs such as platelet aggregation inhibitors, oral anticoagulants, antihypertensives and cholesterol lowering drugs are abundantly prescribed to reduce risk of cardiovascular disease. Notable interindividual variation exists in the response to these pharmacotherapeutic interventions, which can be partially explained by factors such as gender, age, diet, concomitant drug use and environmental factors. Notwithstanding, a great part of this variability remains unknown. To a smaller or larger extent, genetic variability may contribute to the variability in response to these cardiovascular drugs. This review gives an overview of pharmacogenetic studies of genes that were reported to be associated with four commonly prescribed drugs/drug classes (platelet aggregation inhibitors, coumarins, antihypertensives and statins) and were studied at least 2 times with a similar outcome measure. In the field of cardiovascular drug therapy, polymorphisms in candidate genes such as the cycloxygenase-1, vitamin K reductase complex subunit 1, CYP2C9, alpha adducin and 3-hydroxy-3-methylglutaryl-CoA reductase have received a great amount of interest in the pharmacogenetics of aspirin, coumarins, antihypertensives and statins respectively. However, only variations in VKORC1 and CYP2C9 have consistently been associated with drug response (coumarins) and have clinical implications. Clinical trials should provide evidence for the effectiveness of genotyping before this procedure will be a part of every day anticoagulant therapy. In spite of the tremendous amount of publications in this field, there is no reason to advocate for genetic testing for any other drugs cardiovascular drug therapy yet. Current approaches in pharmacogenetic research do not seem to lead to results that meet our expectations of individualized medicine. Therefore, new approaches are needed addressing issues and challenges such as the number of SNPs studied, study power, study design and application of new statistical methods in (pharmaco-)genetic analysis.
Keywords: pharmacogenetics, antihypertensive drugs, statins, oral anticoagulants, aspirin.
Introduction
Cardiovascular disease is one of the leading causes of death, especially in developed countries (1). To reduce the risk on cardiovascular disease (CVD), cardiovascular drugs are abundantly prescribed all over the world for the treatment of conditions such as hypertension and hyperlipidemia. There is notable interindividual variation in response to these drugs, which is partially explained by factors such as gender, age, diet, concomitant drug use and environmental factors. Nevertheless, large part of this variability remains unknown. Genetic variability may contribute to the explanation of variability in response to these cardiovascular drugs (2). Pharmacogenetics focuses on genetic determinants of drug response and aims to optimize effectiveness and to minimize the risk of adverse drug reactions (ADR). Drug response can be modified by polymorphisms in genes that encode drug targets or components in the pathway through which a drug has its effect and are considered pharmacodynamic genes. Genes involved in the metabolism of a drug, such as cytochrome P450 (CYP) enzymes, are classified as pharmacokinetic genes. Finally, there are genes that are involved in the causal pathway of a disease and are able to modify the effect of a drug. In the past decade, the pharmacogenetics field has enjoyed a tremendous surge in activity due to advances in technology. Many association studies have been published. This review gives an overview of the pharmacogenetics of the most commonly prescribed drugs in the management of cardiovascular disease such as platelet aggregation inhibitors, coumarins, antihypertensives and statins.
Methodology
We chose the four most commonly used drug classes in the field of cardiovascular treatment, i.e. platelet aggregation inhibitors (aspirin, clopidogrel), anticoagulants (warfarin, acenocoumarol, phenprocoumon), antihypertensive drugs (diuretics, beta blockers, ACE inhibitors, angiotensin 2 type 1 antagonists, and cholesterol lowering drugs (statins). We restricted our search to those polymorphisms that were reported to be associated with modified response to these drugs and were studied at least 2 times with a similar clinical relevant outcome measure, because replication of genetic association studies are highly recommended to warrant a true association (3). We therefore, excluded studies that only reported pharmacokinetic parameters. Studies on genetic polymorphisms in strong linkage with other polymorphisms were also considered as replication studies.
In our literature search strategy, we searched MEDLINE for recently published reviews of the pharmacogenetics of each drug (class) to list the pharmacogenetic studies that have been conducted. An additional search for studies that were not yet addressed by recent reviews was conducted using MEDLINE. Tables presented in the article give the name of the author of the initial association with a brief description of the findings. The last two columns of each table give the references of studies that have reported either similar or dissimilar results.
Platelet aggregation inhibitors
Pharmacotherapy with platelet aggregation inhibitors such as aspirin and clopidogrel is an important intervention in cardiovascular risk management. In high risk patients, its role in the primary and particularly in the secondary prevention of cardiovascular death, myocardial infarction and stroke is well established (4). However, part of the patients suffer a (recurrent) thromboembolic vascular event despite platelet aggregation inhibitor therapy (5). Several studies have been conducted, investigating the possible genetic contribution to treatment failure. These studies mainly use platelet function tests, including thromboxane (TX) levels (urine or serum), arachidonic acid (AA) tests, adenosine-diphosphate (ADP) tests and others which are described elsewhere (6), either as their main outcome or to define ‘resistance’ to platelet aggregation therapy. Only few studies on clinically relevant outcomes such as MI and stroke have been conducted in relation to the pharmacogenetics of aspirin or clopidogrel. Table I gives an overview of the genes that were studied in vivo more than once.
Table I - .
Gene | Study | SNP | Findings | Similar | Dissimilar |
---|---|---|---|---|---|
Aspirin | |||||
COX1 | Maree et al. 2005 (7) | 5 SNPs** | 842G carriers less sensitive to aspirin treatment | (8, 9)* | (9, 10)* |
COX2 | Gonzalez-Conejero et al. 2005 (9) | G765C | 765C increased sensitivity to aspirin | (17) | |
ITGB3 | Undas et al. 1999 (12) | PlA1/PlA2 | PlA2 less sensitive to aspirin | (13, 14) | (15-18) |
Clopidogrel | |||||
ITGB3 | Angiolillo et al. 2004 (21) | PlA1/PlA2 | P1A2 carriers lower antiplatelet effect compared to A1A1 | (16, 22) | |
P2Y12 | Bura et al. 2006 (23) | H1/H2 | H2H2 carriers less responsive to clopidogrel | (16, 24, 122) | |
CYP2C19 | Hulot et al. 2006 (25) | *1/*2 | *1/*2 decreased platelet responsiveness | (26, 27) |
Result replicated in TXB2 assay, result not replicated in AA induced platelet aggregation assay.
A842G, C22T, G128A, C644A and C714A
Aspirin irreversibly inhibits the cycloxygenase-1 (COX-1) enzyme, ultimately resulting in a decreased amount of thromboxane A2 (TXA2). TXA2 is responsible for activation of platelet aggregation. Therefore, polymorphisms in the COX-1 gene may affect response to aspirin therapy.
In 2005, Maree et al. reported an association between a polymorphism in the COX-1 gene and platelet function in response to aspirin (7). Five common SNPs were genotyped in 144 patients with cardiovascular disease who were treated with aspirin for at least 2 weeks. Aspirin response, determined by serum TXB2 levels and AA-induced platelet aggregation, was associated with the A842G polymorphism. Patients carrying the -842G polymorphism were less sensitive to aspirin treatment (7). Lepantalo et al. reported similar results in 101 patients undergoing elective percutaneous coronary intervention (8). Gonzalez-Conejero et al. investigated the C50T polymorphism which was in complete linkage disequilibrium with the A842G polymorphism. Only the results of the TXB2 assay were similar to those reported in literature, whereas no drug gene interaction was shown using the AA-induced platelet aggregation (9). These results are generally consistent and show that the -842G allele (in linkage disequilibrium with -50T allele) is associated with reduced platelet sensitivity to aspirin. Only one small study including 38 healthy participants could not find any differences in the AA-induced platelet aggregation or TXB2 synthesis (10). The exact mechanism of the interaction between aspirin and the A842G and C50T polymorphisms has not been elucidated yet.
Another gene that has been investigated several times with regard to the pharmacogenetics of both aspirin and clopidogrel is the gene coding for the platelet glycoprotein IIIa (ITGB3) subunit, part of the glycoprotein IIb/IIIa receptor which is present on the platelet surface. Most research focused on the PlA1/A2 polymorphism, in which the P1A1 is the wild-type variant (11). Undas et al. were first to report on the effect of this polymorphism on platelet functioning after in vivo exposure to aspirin, showing that subjects carrying the PlA1/PlA2 genotype were less sensitive to aspirin than homozygous PlA1 carriers (12). These findings have been replicated in small studies (13, 14), whereas other larger studies could not find such an association (15-17) or even showed opposite (18) findings corroborating results from earlier in vitro studies (19, 20). Inconsistent results have been reported for the association between the ITGB3 P1A1/P1A2 polymorphism and variability in response to clopidogrel as well (16, 21, 22). The contribution of the PlA1/PlA2 polymorphism to the pharmacogenetics of platelet aggregation inhibitors has not been elucidated yet.
In addition to genetic variability in COX-1 and ITGB3, polymorphisms in COX-2 and P2Y12 have been associated with modified response to respectively aspirin (9) and clopidogrel (23), but were studied in a very small number of patients and larger studies did not replicate these results (16, 17, 24).
Clopidogrel is a prodrug and has to be activated by hepatic cytochrome P450 (CYP) isoenzymes in order to inhibit platelet aggregation. Among other CYP isoenzymes, clopidogrel is metabolized by CYP2C19 enzyme encoded by the CYP2C19 gene. CYP2C19*2 is a common genetic variant that encodes a deficient version of the enzyme. Hulot et al. showed in 28 healthy subjects that heterozygous carriers of the *2 allele had decreased platelet responsiveness to clopidogrel (25).
Two other studies including 74 (26) and 94 (27) healthy subjects also reported the CYP2C19*2 allele to be associated with decreased platelet responsiveness to clopidogrel. In conclusion, the CYP2C19*2 polymorphism seems to explain part of the interindividual response to clopidogrel, but larger studies are needed to elucidate the clinical implications of this drug gene interaction.
More comprehensive reviews that cover the pharmacogenetics of aspirin and clopidogrel can be found elsewhere (28, 29).
Anticoagulants
Coumarins – warfarin, phenprocoumon and acenocoumarol – are commonly used for the treatment and prevention of thrombotic diseases. Dosing of coumarins, however, is difficult as a result of a narrow therapeutic range and large interindividual differences in response to a certain dose (30).
Consequently, an overdose will increase the risk of haemorrhage and an insufficient dose may lead to failure of prevention of thromboembolism. Patients’ characteristics such as weight, diet, disease state and concomitant use of other medications have been shown to affect the response to coumarins (31). In addition, genetic variation seems to play a role in the explanation for the interindividual variation. To date, studies on the pharmacogenetics of coumarins have mainly focused on the CYP2C9 and VKORC1 polymorphisms and their modifying effect on mean daily dosage and the risk of bleeding, ultimately aiming to prevent bleedings and to predict dosage. Although the evidence for the efficacy of coumarins on secondary endpoints is well established (32), future pharmacogenetic research may also address the variation in the efficacy of coumarin in preventing thrombotic diseases.
The most extensively investigated variants of the CYP2C9 gene encoding the CYP2C9 enzyme are CYP2C9*2 (C430T) and CYP2C9*3 (A1075C), in which the *2 and/or *3 variants encode enzymes with a decreased activity compared to the wild type, CYP2C9*1. Sanderson et al. reported in a systematic meta-analysis that lower maintenance doses of warfarin are required in subjects carrying either variant allele. They showed that *2 and *3 carriers have an increased risk of bleeding as a result of higher plasma levels of warfarin due to slow metabolism of warfarin by the CYP2C9 enzyme (30, 33). Also for acenocoumarol, the CYP2C9*2 and CYP2C9*3 variant have been associated with low dose requirements (34-37).
Moreover, the CYP2C9*3 genotype has been associated with a decreased chance to achieve stable anticoagulation within 6 months, an increased risk for over anticoagulation, a higher initial INR after standard dose acenocoumarol (38) and an increased risk of major bleeding events (39).
Although research on phenprocoumon is sparse, the CYP2C9*2 and/or CYP2C9*3 allele have also been associated with lower dosage requirements, a decreased chance to achieve stability, an increased risk of over anticoagulation (40) and bleeding (41).
In the Rotterdam study, no such interactions between the CYP2C9 genotype and phenprocoumon treatment were found (37, 39), possibly due to power problems and use of different study design. Moreover, the contribution of the CYP2C9 genotype to the variation in response to phenprocoumon may be smaller as acenocoumarol and warfarin are more substantially metabolised by CYP2C9 enzyme than phenprocoumon.
Coumarins exert their anticoagulant effect by inhibiting the vitamin K epoxide reductase complex (VKORC), preventing VKOR from converting vitamin K epoxide to reduced vitamin K (42), which is essential for functioning of several clotting factors such as factor II, VII, IX and X.
The T allele of the C1173T polymorphism in the VKORC1 gene, coding for vitamin K epoxide reductase, has recently been associated with reduced dose needs of warfarin (30), acenocoumarol (43-45) and phenprocoumon (43, 44). In addition to this single SNP approach, five haplotype constructs of 10 SNPs in the VKORC1 gene have been allocated to low and high dose warfarin haplotype groups A and B explaining the interindividual variability in response to warfarin partly (46). Furthermore, it has been reported that being a carrier of the T allele is associated with an increased risk of severe over anticoagulation (INR > 6.0) (47) and with a higher risk for bleeding when using phenprocoumon (44).
Polymorphisms in genes encoding vitamin K-dependent proteins such as clotting factors II and VII have also been studied.
The T165M polymorphism of the factor II (F2) gene and the G-401T and G-402A polymorphisms of the factor VII (F7) gene have been part of pharmacogenetic research.
D’Ambrosio et al. reported that carriers of the M allele of the F2 T165M polymorphism require lower doses of warfarin (48).
Other studies did not confirm an association between the F2 T165M polymorphism and warfarin dose requirements (49, 50). Carriers of the A allele of the F7 G-402A polymorphism (50) and T allele carriers of the F7 G-401T (48) allele have been reported to require lower doses warfarin, but no other studies have confirmed these results yet (48, 50). To what extent these polymorphisms contribute to variability in response to warfarin remains to be determined.
Many articles present estimates of the contribution of various factors, including VKORC1 and CYP2C9 polymorphism, to the interindividual variation of coumarin response (up to 60%) and also propose algorithms for coumarin dosing (31). However, before we can implement this knowledge in the guidelines of anticoagulation therapy, a controlled clinical trial is needed to validate these dosing algorithms. A more comprehensive overview of the current status in the pharmacogenetics of coumarins can be found elsewhere (30, 31).
Antihypertensive drugs
The most prevalent indication in cardiovascular drug therapy is hypertension. Its high prevalence and the strong association with cardiovascular morbidity have given rise to the question of who to treat with which drug. In the past decades, the search for markers that can predict response to therapy has experienced a tremendous surge. The major drug classes available for the treatment of hypertension are diuretics, beta-blockers, ACE inhibitors and angiotensin 2 type 1 receptor antagonists. A complete review of the pharmacogenetics of antihypertensives can be found elsewhere (51).
Diuretics
Diuretics are considered the first line pharmacological intervention for most patients with hypertension (52). The long term beneficial effects of thiazides are thought to result from a reduction of peripheral vascular resistance, decreased blood vessel sensitivity to catecholamines and indirect activation of the renin-angiotensin-aldosterone system. To date, several pharmacogenetic studies have been published on the pharmacogenetics of diuretics. Polymorphisms in five genes were involved in more than 1 study, namely alpha adducin (ADD1) G460W, angiotensin converting enzyme (ACE) I/D, angiotensin (AGT) -6A, angiotensin receptor (AGTR1) A1166C and G-protein beta-3 subunit (GNB3) C825T.
The ADD1 G460W polymorphism has been shown to affect renal proximal tubule sodium reabsorption in hypertension with increased reabsorption in patients carrying the W allele (53), possibly influencing the responsiveness to diuretics. Cusi et al. first reported on the ADD1 G460W polymorphism and the association with altered response to hydrochlorothiazide (54). They found that heterozygous hypertensive patients experienced a greater fall in mean arterial pressure in response to 2 months’ treatment with hydrochlorothiazide than wild-type homozygous hypertensive patients (54). Following this study, these results were replicated in 2 other studies also showing that carriers of the ADD1 W allele respond better to hydrochlorothiazide (55, 56). Large scale studies with up to 36,000 patients (57) could not replicate these findings (57-60) and thereby rule out a major role for the ADD1 G460W polymorphism in predicting blood pressure response to diuretics.
Additionally, some studies have associated the ADD1 W allele with a better response to diuretics studying clinical outcomes like stroke or MI (61). Yet again, these results could not be replicated (57, 62).
Besides the ADD1 gene, researchers have taken a great interest in the I/D polymorphism of the ACE gene encoding the angiotensin converting enzyme which has a pivotal role in the reninangiotensin- aldosteron system (RAAS), converting angiotensin I into angiotensin II. ACE plays an important role in the regulation of the water sodium balance and in the maintenance of vascular tone. Judging from the studies listed in table II, nothing can be concluded about the effect of the ACE I/D polymorphism on blood pressure, as the direction of the effect of the genotype appears to go either way. In addition, no association has been found between the ACE I/D polymorphism and diuretic therapy on any clinical outcome (63). Other polymorphisms in the RAAS that have been studied are present in the AGT and AGTR1 gene. The AGT A(-6)G and M235T polymorphism and AGTR1 A1166C polymorphism have been associated with modified response to diuretics in African American women only (64). These results were not replicated in the Doetinchem Cohort Study (59). Contrary to earlier findings (64), a smaller study in forty-five Chinese subjects showed that G allele carriers of the AGT A(-6)G polymorphism experienced greater blood pressure response to diuretics (65). Clearly, larger studies are needed to elucidate the possible role of these polymorphisms in the response to diuretics. Furthermore, an association with the GNB3 C825T polymorphism has been reported and replicated, both studies showing that subjects carrying the TT genotype would benefit more from diuretics in lowering blood pressure (59, 66). The mechanistic pathway of this possible gene-treatment interaction is that the T allele of the GNB3 C825T polymorphism has been associated with low plasma renin and an elevated aldosterone-renin ratio (67) which could influence the response to diuretics. A study by Schelleman et al. could not corroborate previous findings concerning the association between the GNB3 C825T polymorphism and clinical events (62).
Table II - .
Gene | SNP | Outcome | Study | Findings | Similar | Dissimilar |
---|---|---|---|---|---|---|
Diuretics | ||||||
ADD1 | G460W | Blood pressure | Cusi et al. (54) | W allele carriers respond better | (55, 56) | (57-60) |
MI/stroke | Psaty et al. (61) | W allele carriers respond better | (57, 62) | |||
ACE | I/D | Blood pressure | Sciarrone et al. (56) | Better response in II carriers | (123)** | (123)** |
AGT | A(-6)G* | Blood pressure | Frazier et al. (64) | -6A greater response (in AfrA women) | (59, 65) | |
AGTR1 | A1166C | Blood pressure | Frazier et al. (64) | A allele better response (in AfrA women) | (59, 65) | |
GNB3 | C825T | Blood pressure | Turner et al. (66) | Better response TT compared to CC | (59) | |
Beta Blockers | ||||||
ADRB1 | R389G | Blood pressure | Sofowora et al. (71) | AA greater response than GG | (70, 72, 73) | (69, 74, 75) |
AGT | A(-6)G* | Blood pressure | Kurland et al. (76) | -6A allele greater response | (77, 78) | |
ACE inhibitors | ||||||
ACE | I/D | Blood pressure | Hingorani et al. (80) | No effect | (59, 63, 78, 79, 124- 133)*** | (133-137) (II > DD) (138-140) (DD>II) |
AGT | M235T | Blood pressure | Hingorani et al. (80) | MT and TT greater response than MM | (59, 60, | |
Clinical event | Bis et al. (143) | TT less risk of stroke (not MI) | (62) | |||
Angiotensin2 type 1 receptor antagonists | ||||||
ACE | I/D | Blood pressure | Kurland et al. (77) | II carriers greater response | (82, 83) | |
CYP11B2 | C-344T | Blood pressure | Kurland et al. (81) | TT carriers greater response | (83) | |
AGTR1 | A1166C | Blood pressure | Miller et al. (144) | C carriers greater response | (77, 82) |
In linkage disequilibrium with M235T polymorphism.
Similar result for women, men more benefit with DD genotype.
O’Toole et al. (133): Lisonopril no association, captopril associated. AfrA = African American descent.
Beta-blockers
The main target of β1-selective beta-blockers is the β1- adrenoreceptor encoded by ADRB1, in which the Arg389Gly polymorphism is a common variant located in the intracellular GTP binding domain. The Arg389 variant has been shown to possess a three- to four-fold higher adenylyl cyclase activity that in turn may cause a modified response to beta blockers (68). Although O’Shaughnessy did not report an association of the ADRB1 polymorphism and altered response to atenolol or bisoprolol (69), in the year 2003 three articles were published reporting an association of the Arg variant with better response to beta-blockers, either SBP (70-72), DBP (72, 73) or resting MBP (70, 72). This observation could not be confirmed by two other studies (74, 75).
Beta adrenergic receptors are G-protein coupled receptors. Therefore the GNB3 C825T polymorphism possibly modifies the blood pressure response to beta blockade. In females only, the CC genotype of the GNB3 C825T polymorphism has been associated with better blood pressure response to beta blockers (75), a finding that was not confirmed in the Doetinchem study (59). Also the AGT A(-6)G polymorphism has also been associated with the pharmacogenetics of beta blockers, but the association has only been reported once (76) whereas other studies failed to confirm this association (77, 78).
Larger scale studies should confirm these interactions because these studies are all underpowered.
ACE inhibitors
The ACE gene – coding for the drug target of ACE inhibitors – has been studied over 20 times.
Table II includes all studies that evaluated blood pressure response to ACE inhibitors according to ACE I/D genotype, including treatment duration ranging from single doses to several years, small scale studies as well as larger studies and studies in both healthy subject and patients. At first, smaller studies reported a more beneficial effect of ACE inhibitors on blood pressure in subjects carrying II. Large studies that have been published more recently (63, 79) could not confirm these results, suggesting that the ACE I/D polymorphism is not likely to be a strong modifier of blood pressure response to treatment with ACE inhibitors.
Another gene involved in the RAAS that has been associated with modified response to ACE inhibitors is the angiotensin (AGT) gene. Initially, a study in 125 subjects showed that the T allele of the M235T polymorphism was associated with better blood pressure lowering response to ACE inhibitors compared to homozygous M allele carriers (80). Several other studies, among which one included 1,447 patients, found no such association (Table II), concluding that there is no interaction between the AGT M235T polymorphism and the response to ACE inhibitors.
Angiotensin II antagonists
Another category of antihypertensives that act on the RAAS, are the angiotensin II type 1 receptor antagonists. Genes in the RAAS are considered candidate genes in the pharmacogenetics of angiotensin II type 1 receptor antagonists. Studies in the SILVHIA trial including almost 50 subjects taking irbesartan reported that the ACE I/D (77) and CYP11B2 C-344T polymorphism were associated with modified blood pressure response (77, 81). Subjects carrying the ACE II polymorphism (77) and subjects carrying the CYP11B2 TT variant (81) had a more pronounced blood pressure response to irbesartan. Redon et al. conducted a similar study including 206 subjects treated with telmisartan, but no significant associations were found in any of the RAAS gene polymorphisms (82). Contrary to the results in the SILVHIA trial, Ortlepp et al. reported the C allele of the CYP11B2 C-344T polymorphism to be associated with better response to candesartan (83). Currently, the role of polymorphisms in the RAAS in response to angiotensin II antagonists is unknown. Larger studies are needed to elucidate the pharmacogenetic contribution, as these studies are all underpowered.
Cholesterol lowering drugs
Statins primarily reduce the risk on coronary artery disease (CAD) by lowering blood cholesterol through inhibition of the HMG-CoA reductase enzyme. Although large clinical trials found a 27% average relative risk reduction of major coronary events (84), there is large variability in benefits from statin therapy. Many genes involved in the pharmacodynamic pathway of statins have been part of pharmacogenetic research in patients with primary hypercholesterolemia, with an emphasis on genes involved in the cholesterol pathway, although genes involved with possible pleiotropic effects of statins gain more and more interest.
The drug target of statins, encoded by the HMG-CoA reductase (HMGCR) gene was one of the candidate genes in the two studies addressing a considerable number of SNPs (85, 86). The PRINCE study found a pharmacogenetic association with two (SNP12 and SNP29) out of 33 SNPs investigated in the HMGCR gene on total cholesterol as well as on LDL cholesterol (85). SNP29 was also one of the four SNPs that was studied in the HMGCR gene in the ACCESS trial, showing no such association (86). Both SNP 12 and SNP 29 are located in a non-coding region and further research should determine whether there is a molecular explanation for the results found in the PRINCE study.
The cholesteryl ester transfer protein (CETP) enzyme plays a central role in transport of cholesterol from peripheral tissues back to the liver, reverse cholesterol transport (RCT). Taq1B is a common polymorphism in the CETP gene and is associated with variations in lipid transfer activity and high density lipoprotein (HDL) levels (87) and may therefore alter response to statins. CETP concentrations are believed to be highest in homozygous B1 carriers and lowest in CETP homozygous B2 carriers. The effect of Taq1B on statin therapy was first investigated by Kuivenhoven et al, showing that pravastatin therapy slowed the progression of coronary atherosclerosis in B1B1 (variant) carriers, whereas B2B2 carriers did not benefit from pravastatin therapy (88). Several other studies investigated the CETP polymorphism as well and could not find an association of the Taq1B polymorphism with altered efficacy of statins in preventing cardiovascular diseases (89-92) or found the opposite (93). A large meta-analysis including over 13,000 patients found no gene treatment interaction between statins and the Taq1B polymorphism of the CETP gene (94). Similar to CETP, lipoprotein lipase (LPL) and hepatic lipase (LIPC) are genes that code for enzymes that transfer lipids between lipoproteins and mediate lipolysis. These genes have been associated with altered lipid response to statin therapy but no conclusive results have been published (86, 95, 96).
Apolipoprotein E (ApoE) is a genetically polymorphic protein defined by three alleles – E2, E3 and E4 – that influences hepatic cholesterol content. Lipoproteins with the E4 isoform are taken up with greater affinity than those with the common E3 isoform, which in turn are taken up more efficiently than those with the E2 isoform. Ordovas et al. reported that carriers of the E2 genotype experience greatest LDL reduction in response to statin therapy in comparison to E3 and E4 carriers (97). With great interest, many similar studies were conducted trying to clarify the role of the ApoE polymorphism in the pharmacogenetics of statins (Table III). There is a reasonable body of evidence supporting the findings from Ordovas et al, among which are some large scale studies (86, 98, 99).
Table III - .
Gene | SNP | Outcome | Study | Findings | Similar | Dissimilar |
---|---|---|---|---|---|---|
ApoE | E2/E3/E4 | LDL reduction | Ordovas et al. (97) | Greatest LDLC reductions in E2 carriers | (86, 98, 99, 145-147) | (85, 148-153) |
HDL increase | Ballantyne et al. (154) | Enhanced HDL response E2 | (152, 153) | |||
Death | Gerdes et al. (100) | Attenuated mortality risk for E4 carriers | (101) | |||
LDLR | AvaII/PvuII | LDL reduction | Salazar et al. (155) | Attenuated LDL reduction A+ and P1 carriers | (85, 156) | |
ApoB | I/D | LDL reduction | Guzman et al. (157) | II carriers greatest LDL reduction | (85) | |
Xba1 | TC reduction | Ye et al. (145) | Greater TC reduction patients carrying X allele | (150) | ||
CETP | Taq1B | HDL increase | van Venrooy et al. (160) | B1 allele greater increase HDL-C | (85, 86, 161) | |
Changes MSD/CE | Kuivenhoven et al. (88) | Decreased atherosclerosis progression in B1B1 carriers | (90-93, 162) | |||
HMGCR | 33 SNPs | LDL reduction | Chasman et al. (85) | Reduced response heterozygous for SNP12, SNP29 | (86) | |
ACE | I/D | LDL reduction | Marian et al. (107) | Enhanced response in DD subjects | (86) | |
Angiogram/CE | Marian et al. (107) | Higher regression rate in DD subjects | (86, 108-110) | |||
CYP7A1 | A-204C | LDL reduction | Kajinami et al. (163) | C allele diminished LDL cholesterol response | (86, 147) | |
SREBP1 | G-36del | HDL response | Salek et al. (164) | Reduced ApoAI response in carriers del/del | (165) | |
SCAP | A2386G | TC/TG/LDL | Fiegenbaum et al. (165) | Better response in G allele carriers response | (164, 166) | |
LIPC | C-514T | HDL response | Lahoz et al. (96) | Greatest HDL response in T allele carriers | (86) | |
LPL | S447X | TC reduction | Thompson et al. (86) | Homozygous X greatest TC reduction | (95) | |
TLR4 | D299Q | CE reduction | Boekholdt et al. (111) | Carriers G allele greatest response | (112) | |
PON1 | R192Q | HDL increase | Malin et al. (103) | Carriers R allele greatest response | (104) | |
SLCO1B1 | T521C | TC reduction | Tachibana-Iimori et al. (113) | Better response in TT carriers | (86, 115, 116) | |
ABCB1 | C3435T | LDL reduction | Kajinami et al. 2004 (117) | CC carriers of C3435T polymorphism experience smaller LDL reduction. | (118, 167) | |
No effect of G2677T/A |
CE = clinical events, MSD = mean segment diameter, TC = total cholesterol, TG = triglyceride.
ApoE2 carriers seem to benefit most from statin therapy regarding lipid profile improvement, however a sub study of the Scandinavian Simvastatin Survival Study (S4) reported subjects carrying the ApoE4 genotype to have the largest risk reduction of mortality (100). These results were not replicated (101) and the effect of the possible gene treatment interaction on clinical outcomes remains inconclusive. Besides ApoE, genetic variations in the genes coding for apolipoprotein B (ApoB) and apolipoprotein A1 (APOA1) have been part of pharmacogenetic research with inconclusive results (Table III). PON1, encoding the paraoxonase enzyme, is closely related to ApoA1 and the transcriptional activity of the PON1 gene has been shown to be increased by simvastatin (102). One small study including 51 patients reported the R allele of the PON1 R192Q polymorphism to be associated with better HDL response than those carrying the QQ genotype (103). Another study, did not confirm these results (104) and further research is needed to elucidate the role of this candidate gene.
Some functional mutations in the low density lipoprotein receptor (LDLR) cause familial hypercholesterolemia (FH), whereas more common genetic variations in the LDLR have been shown to modify the cholesterol lowering response to statins. Sterol regulatory elements binding factor 1 (SREBF1) and SREBF chaperone (SCAP) have been shown to regulate LDLR expression (105) and other enzymes in the lipid metabolism (106) and have therefore also been investigated for genetic variation in relation with modified response. To date, replication of associations in pharmacogenetic studies of both the LDLR gene as well as the SREBF1 and SCAP genes lack (table 3). Future studies may explain the role of genetic variations in these important genes in the lipid metabolism.
In addition to genes involved in the lipid lowering response to statins, polymorphisms in the ACE and toll like receptor 4 (TLR4) genes have been studied. The ACE I/D polymorphism was first studied in the LCAS, showing cholesterol reduction and atherosclerosis progression was most beneficial in subject carrying the DD genotype (107). Large studies in GENHAT (108) and ACCESS (86) could not confirm the gene treatment interaction from LCAS and studies in CARE (109) and in the Rotterdam study (110) even reported contradicting results. If there is an interaction between the ACE I/D and the effectiveness of statins, the interaction is small and its direction and molecular basis is unknown.
The toll like receptor mediates innate and adaptive immunity. A functional polymorphism in the TLR4 gene D299G has been studied in REGRESS and SAS, both including approximately 650 patients. Both studies showed that carriers of the G allele treated with statins had a lower risk of cardiovascular events than non carriers treated with statins (111, 112), suggesting that statins interact with inflammatory factors like the toll like receptor.
Polymorphisms in pharmacokinetic genes encoding the solute carrier organic transporter (SCLO1B1) involved in the hepatic uptake of statins and the adenosine triphosphatebinding cassette (ABC) B1 transporter involved in the hepatobiliary excretion of statins may influence the pharmacokinetics of statins and thereby its lipid lowering response. The TT genotype of the SCLO1B1 T521C polymorphism has been associated with greater response to statins in lowering total cholesterol (113). A possible explanation for this finding is that the C allele is associated with impaired hepatic uptake of statins (114). The ACCESS study (86) and two small studies (115, 116) could not confirm the interaction between the SCLO1B1 T521C polymorphism and the effect on lowering plasma lipid levels. The C3435T polymorphism of the ABCB1 gene has been associated with modified response to statin therapy (117), which could not be replicated by Fiegenbaum et al. who found an association with the G2677T/A polymorphism of the ABCB1 gene (118). Currently there is no evidence for an interaction between these polymorphisms in the SCLO1B1 and ABCB1 gene and statins on pharmacodynamic endpoints.
An excellent review published by Mangravite et al. gives a very comprehensive overview of all studies conducted in the pharmacogenetics of statins (119, 120).
Clinical implications
By addressing some of the most extensively investigated pharmacogenetic associated genes in cardiovascular drug therapy, it is clear that the evidence for clinical implications of pharmacogenetics currently is very scarce. Although each initial association of a polymorphism with modified drug response seemed very promising, the reality is that most associations cannot be replicated. Furthermore, most effects that were found are very small. Therefore implementation of these interactions in clinical practice is still far away. Only for coumarins, there is a real opportunity for pharmacogenetics by genotyping the VKORC1 and CYP2C9 gene to optimize anticoagulant therapy. Even though the effect of the existing variations in these genes are quite clear, clinical trials should provide evidence for the effectiveness of genotyping regarding prevention of adverse drug events and cost-effectiveness, before this procedure will be a part of every day anticoagulant therapy. These studies are currently underway and will possibly advocate for a global implementation of VKORC1 and CYP2C9 genotyping into the anticoagulant therapy guidelines.
Nevertheless, in August 2007 the Food and Drug Administration updated the warfarin prescribing information and highlighted the opportunity to use genetic tests to improve their initial estimate of warfarin dose. For other cardiovascular drugs, future research may well elucidate the role of genetics in the response to these drugs.
Future directions
Current approaches in pharmacogenetic research do not seem to lead to results that meet our expectations of individualized medicine. Therefore, new approaches are needed addressing issues and challenges such as the number of SNPs studied, study power, study design and application of new statistical methods in (pharmaco-)genetic analysis.
Most studies have only examined one polymorphism in a candidate gene associated with modified response to a certain drug. However, drug response is likely to result from complex interactions among various biologic pathways. Hence, future studies should consider a set of candidate genes and/or a genome wide scan (GWS) rather than addressing a single or small number of SNPs. In recent years, the costs for a GWS have considerably decreased and will be increasingly available.
Examining multiple SNPs will require sufficient sample size, that many studies to date lack.
Moreover, analysis of large numbers of SNPs to identify a combination of SNPs that influence drug efficacy, will be a huge challenge due to statistical problems. Not only the issue of multiple testing of many SNP should be addressed with new tools in statistical analysis, also possible important effects of genegene interactions should be considered (121). Although a definitive statistical method for characterising statistical patterns of epistasis in not known yet, conventional statistical methods only will not be the appropriate tool to decipher the complexity of pharmacogenetics.
Finally, to elucidate mechanisms that lie behind the genetic associations, other fields of research including proteomics and transcriptomics should be integrated in the field of pharmacogenomics.
Conclusion
In conclusion, although pharmacogenetic testing is already part of everyday clinical practice in some areas (chemotherapy, psychiatry), for cardiovascular drugs currently only oral anticoagulant therapy seems to have a real opportunity to benefit from pharmacogenetic testing. In spite of the tremendous amount of publications in this field, there is no reason to advocate for genetic testing for any other cardiovascular drugs yet. Although future research will certainly benefit from emerging genetic technology as high throughput genome wide scans will be readily available, finding the genetic profile that will predict response to cardiovascular drugs will be a major challenge.
References
- 1.McGovern PG, et al. Trends in acute coronary heart disease mortality, morbidity, and medical care from 1985 through 1997: the Minnesota heart survey. Circulation. 2001;104(1):19–24. doi: 10.1161/01.cir.104.1.19. [DOI] [PubMed] [Google Scholar]
- 2.Weinshilboum R. Inheritance and drug response. N Engl J Med. 2003;348(6):529–37. doi: 10.1056/NEJMra020021. [DOI] [PubMed] [Google Scholar]
- 3.Freely associating. Nat Genet. 1999;22(1):1–2. doi: 10.1038/8702. [DOI] [PubMed] [Google Scholar]
- 4.Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, and stroke in high risk patients. BMJ. 2002;324(7329):71–86. doi: 10.1136/bmj.324.7329.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Grotemeyer KH, Scharafinski HW, Husstedt IW. Two-year follow-up of aspirin responder and aspirin non responder. A pilot-study including 180 post-stroke patients. Thromb Res. 1993;71(5):397–403. doi: 10.1016/0049-3848(93)90164-j. [DOI] [PubMed] [Google Scholar]
- 6. Michelson AD, Frelinger AL 3rd, Furman MI. Current options in platelet function testingCurrent options in platelet function testing. Am J Cardiol. 2006;98(10A):4N–10N. doi: 10.1016/j.amjcard.2006.09.008. [DOI] [PubMed] [Google Scholar]
- 7. Maree AO, et al. Cyclooxygenase-1 haplotype modulates platelet response to aspirinCyclooxygenase-1 haplotype modulates platelet response to aspirin. J Thromb Haemost. 2005;3(10):2340–5. doi: 10.1111/j.1538-7836.2005.01555.x. [DOI] [PubMed] [Google Scholar]
- 8.Lepantalo A, et al. Polymorphisms of COX-1 and GPVI associate with the antiplatelet effect of aspirin in coronary artery disease patients. Thromb Haemost. 2006;95(2):253–9. doi: 10.1160/TH05-07-0516. [DOI] [PubMed] [Google Scholar]
- 9. Gonzalez-Conejero R, et al. Biological assessment of aspirin efficacy on healthy individuals: heterogeneous response or aspirin failure?Biological assessment of aspirin efficacy on healthy individuals: heterogeneous response or aspirin failure? Stroke. 2005;36(2):276–80. doi: 10.1161/01.STR.0000151362.65339.f9. [DOI] [PubMed] [Google Scholar]
- 10. Halushka MK, Walker LP, Halushka PV. Genetic variation in cyclooxygenase 1: effects on response to aspirinGenetic variation in cyclooxygenase 1: effects on response to aspirin. Clin Pharmacol Ther. 2003;73(1):122–30. doi: 10.1067/mcp.2003.1. [DOI] [PubMed] [Google Scholar]
- 11. Nurden AT. Platelet glycoprotein IIIa polymorphism and coronary thrombosisPlatelet glycoprotein IIIa polymorphism and coronary thrombosis. Lancet. 1997;350(9086):1189–91. doi: 10.1016/s0140-6736(05)63447-2. [DOI] [PubMed] [Google Scholar]
- 12. Undas A, et al. Platelet glycoprotein IIIa polymorphism, aspirin, and thrombin generationPlatelet glycoprotein IIIa polymorphism, aspirin, and thrombin generation. Lancet. 1999;353(9157):982–3. doi: 10.1016/S0140-6736(98)05054-5. [DOI] [PubMed] [Google Scholar]
- 13. Szczeklik A, et al. Relationship between bleeding time, aspirin and the PlA1/A2 polymorphism of platelet glycoprotein IIIaRelationship between bleeding time, aspirin and the PlA1/A2 polymorphism of platelet glycoprotein IIIa. Br J Haematol. 2000;110(4):965–7. doi: 10.1046/j.1365-2141.2000.02267.x. [DOI] [PubMed] [Google Scholar]
- 14.Undas A, et al. Pl(A2) polymorphism of beta(3) integrins is associated with enhanced thrombin generation and impaired antithrombotic action of aspirin at the site of microvascular injury. Circulation. 2001;104(22):2666–72. doi: 10.1161/hc4701.099787. [DOI] [PubMed] [Google Scholar]
- 15. Pamukcu B, Oflaz H, Nisanci Y. The role of platelet glycoprotein IIIa polymorphism in the high prevalence of in vitro aspirin resistance in patients with intracoronary stent restenosisThe role of platelet glycoprotein IIIa polymorphism in the high prevalence of in vitro aspirin resistance in patients with intracoronary stent restenosis. Am Heart J. 2005;149(4):675–80. doi: 10.1016/j.ahj.2004.10.007. [DOI] [PubMed] [Google Scholar]
- 16.Lev EI, et al. Genetic polymorphisms of the platelet receptors P2Y(12), P2Y(1) and GP IIIa and response to aspirin and clopidogrel. Thromb Res. 2007;119(3):355–60. doi: 10.1016/j.thromres.2006.02.006. [DOI] [PubMed] [Google Scholar]
- 17.Jefferson BK, et al. Aspirin resistance and a single gene. Am J Cardiol. 2005;95(6):805–8. doi: 10.1016/j.amjcard.2004.11.045. [DOI] [PubMed] [Google Scholar]
- 18.Macchi L, et al. Resistance in vitro to low-dose aspirin is associated with platelet PlA1 (GP IIIa) polymorphism but not with C807T (GP Ia/IIa) and C-5T Kozak (GP Ibalpha) polymorphisms. J Am Coll Cardiol. 2003;42(6):1115–9. doi: 10.1016/s0735-1097(03)00921-5. [DOI] [PubMed] [Google Scholar]
- 19. Cooke GE, et al. PlA2 polymorphism and efficacy of aspirinPlA2 polymorphism and efficacy of aspirin. Lancet. 1998;351(9111):1253. doi: 10.1016/S0140-6736(05)79320-X. [DOI] [PubMed] [Google Scholar]
- 20.Michelson AD, et al. Platelet GP IIIa Pl(A) polymorphisms display different sensitivities to agonists. Circulation. 2000;101(9):1013–8. doi: 10.1161/01.cir.101.9.1013. [DOI] [PubMed] [Google Scholar]
- 21.Angiolillo DJ, et al. PlA polymorphism and platelet reactivity following clopidogrel loading dose in patients undergoing coronary stent implantation. Blood Coagul Fibrinolysis. 2004;15(1):89–93. doi: 10.1097/00001721-200401000-00014. [DOI] [PubMed] [Google Scholar]
- 22. Dropinski J, et al. Anti-thrombotic action of clopidogrel and P1(A1/A2) polymorphism of beta3 integrin in patients with coronary artery disease not being treated with aspirinAnti-thrombotic action of clopidogrel and P1(A1/A2) polymorphism of beta3 integrin in patients with coronary artery disease not being treated with aspirin. Thromb Haemost. 2005;94(6):1300–5. [PubMed] [Google Scholar]
- 23. Bura A, et al. Role of the P2Y12 gene polymorphism in platelet responsiveness to clopidogrel in healthy subjectsRole of the P2Y12 gene polymorphism in platelet responsiveness to clopidogrel in healthy subjects. J Thromb Haemost. 2006;4(9):2096–7. doi: 10.1111/j.1538-7836.2006.02113.x. [DOI] [PubMed] [Google Scholar]
- 24. von Beckerath N, et al. P2Y12 gene H2 haplotype is not associated with increased adenosine diphosphate-induced platelet aggregation after initiation of clopidogrel therapy with a high loading doseP2Y12 gene H2 haplotype is not associated with increased adenosine diphosphate-induced platelet aggregation after initiation of clopidogrel therapy with a high loading dose. Blood Coagul Fibrinolysis. 2005;16(3):199–204. doi: 10.1097/01.mbc.0000164429.21040.0a. [DOI] [PubMed] [Google Scholar]
- 25.Hulot JS, et al. Cytochrome P450 2C19 loss-of-function polymorphism is a major determinant of clopidogrel responsiveness in healthy subjects. Blood. 2006;108(7):2244–7. doi: 10.1182/blood-2006-04-013052. [DOI] [PubMed] [Google Scholar]
- 26. Brandt JT, et al. Common polymorphisms of CYP2C19 and CYP2C9 affect the pharmacokinetic and pharmacodynamic response to clopidogrel but not prasugrelCommon polymorphisms of CYP2C19 and CYP2C9 affect the pharmacokinetic and pharmacodynamic response to clopidogrel but not prasugrel. J Thromb Haemost. 2007;5(12):2429–36. doi: 10.1111/j.1538-7836.2007.02775.x. [DOI] [PubMed] [Google Scholar]
- 27.Fontana P, et al. Influence of CYP2C19 and CYP3A4 gene polymorphisms on clopidogrel responsiveness in healthy subjects. J Thromb Haemost. 2007;5(10):2153–5. doi: 10.1111/j.1538-7836.2007.02722.x. [DOI] [PubMed] [Google Scholar]
- 28.Maree AO, Fitzgerald DJ. . Variable platelet response to aspirin and clopidogrel in atherothrombotic disease. Circulation. 2007;115(16):2196–207. doi: 10.1161/CIRCULATIONAHA.106.675991. [DOI] [PubMed] [Google Scholar]
- 29. Goodman T, Sharma P, Ferro A. The genetics of aspirin resistanceThe genetics of aspirin resistance. Int J Clin Pract. 2007;61(5):826–34. doi: 10.1111/j.1742-1241.2007.01344.x. [DOI] [PubMed] [Google Scholar]
- 30. Wadelius M, Pirmohamed M. Pharmacogenetics of warfarin: current status and future challengesPharmacogenetics of warfarin: current status and future challenges. Pharmacogenomics J. 2007;7(2):99–111. doi: 10.1038/sj.tpj.6500417. [DOI] [PubMed] [Google Scholar]
- 31.Yin T, T Miyata. Warfarin dose and the pharmacogenomics of CYP2C9 and VKORC1 - rationale and perspectives. Thromb Res. 2007;120(1):1–10. doi: 10.1016/j.thromres.2006.10.021. [DOI] [PubMed] [Google Scholar]
- 32.Aguilar MI, Hart R. Oral anticoagulants for preventing stroke in patients with nonvalvular atrial fibrillation and no previous history of stroke or transient ischemic attacks. Cochrane Database Syst Rev. 2005 doi: 10.1002/14651858.CD001927.pub2. (3):CD001927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Sanderson S, Emery J, Higgins J. CYP2C9 gene variants, drug dose, and bleeding risk in warfarin-treated patients: a HuGEnet systematic review and meta-analysis. Genet Med. 2005;7(2):97–104. doi: 10.1097/01.gim.0000153664.65759.cf. [DOI] [PubMed] [Google Scholar]
- 34.Thijssen HH, Verkooijen IW, Frank HL. The possession of the CYP2C9*3 allele is associated with low dose requirement of acenocoumarol. Pharmacogenetics. 2000;10(8):757–60. doi: 10.1097/00008571-200011000-00010. [DOI] [PubMed] [Google Scholar]
- 35.Tassies D, et al. Pharmacogenetics of acenocoumarol: cytochrome P450 CYP2C9 polymorphisms influence dose requirements and stability of anticoagulation. Haematologica. 2002;87(11):1185–91. [PubMed] [Google Scholar]
- 36.Hermida J, et al. Differential effects of 2C9*3 and 2C9*2 variants of cytochrome P-450 CYP2C9 on sensitivity to acenocoumarol. Blood. 2002;99(11):4237–9. doi: 10.1182/blood.v99.11.4237. [DOI] [PubMed] [Google Scholar]
- 37.Visser LE, et al. The risk of overanticoagulation in patients with cytochrome P450 CYP2C9*2 or CYP2C9*3 alleles on acenocoumarol or phenprocoumon. Pharmacogenetics. 2004;14(1):27–33. doi: 10.1097/00008571-200401000-00003. [DOI] [PubMed] [Google Scholar]
- 38.Schalekamp T, et al. Acenocoumarol stabilization is delayed in CYP2C93 carriers. Clin Pharmacol Ther. 2004;75(5):394–402. doi: 10.1016/j.clpt.2003.12.017. [DOI] [PubMed] [Google Scholar]
- 39.Visser LE, et al. The risk of bleeding complications in patients with cytochrome P450 CYP2C9*2 or CYP2C9*3 alleles on acenocoumarol or phenprocoumon. Thromb Haemost. 2004;92(1):61–6. doi: 10.1160/TH03-12-0741. [DOI] [PubMed] [Google Scholar]
- 40.Schalekamp T, et al. Effects of cytochrome P450 2C9 polymorphisms on phenprocoumon anticoagulation status. Clin Pharmacol Ther. 2004;76(5):409–17. doi: 10.1016/j.clpt.2004.08.006. [DOI] [PubMed] [Google Scholar]
- 41.Hummers-Pradier E, et al. Determination of bleeding risk using genetic markers in patients taking phenprocoumon. Eur J Clin Pharmacol. 2003;59(3):213–9. doi: 10.1007/s00228-003-0580-8. [DOI] [PubMed] [Google Scholar]
- 42.Suttie JW. The biochemical basis of warfarin therapy. Adv Exp Med Biol. 1987;214:3–16. doi: 10.1007/978-1-4757-5985-3_2. [DOI] [PubMed] [Google Scholar]
- 43.Bodin L, et al. Cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKORC1) genotypes as determinants of acenocoumarol sensitivity. Blood. 2005;106(1):135–40. doi: 10.1182/blood-2005-01-0341. [DOI] [PubMed] [Google Scholar]
- 44.Reitsma PH, et al. A C1173T dimorphism in the VKORC1 gene determines coumarin sensitivity and bleeding risk. PLoS Med. 2005;2(10):e312. doi: 10.1371/journal.pmed.0020312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Schalekamp T, et al. VKORC1 and CYP2C9 genotypes and acenocoumarol anticoagulation status: interaction between both genotypes affects overanticoagulation. Clin Pharmacol Ther. 2006;80(1):13–22. doi: 10.1016/j.clpt.2006.04.006. [DOI] [PubMed] [Google Scholar]
- 46.Rieder MJ, et al. Effect of VKORC1 haplotypes on transcriptional regulation and warfarin dose. N Engl J Med. 2005;352(22):2285–93. doi: 10.1056/NEJMoa044503. [DOI] [PubMed] [Google Scholar]
- 47.Schalekamp T, et al. VKORC1 and CYP2C9 genotypes and phenprocoumon anticoagulation status: interaction between both genotypes affects dose requirement. Clin Pharmacol Ther. 2007;81(2):185–93. doi: 10.1038/sj.clpt.6100036. [DOI] [PubMed] [Google Scholar]
- 48.D’Ambrosio RL, et al. Polymorphisms in factor II and factor VII genes modulate oral anticoagulation with warfarin. Haematologica. 2004;89(12):1510–6. [PubMed] [Google Scholar]
- 49.Aquilante CL, et al. Influence of coagulation factor, vitamin K epoxide reductase complex subunit 1, and cytochrome P450 2C9 gene polymorphisms on warfarin dose requirements. Clin Pharmacol Ther. 2006;79(4):291–302. doi: 10.1016/j.clpt.2005.11.011. [DOI] [PubMed] [Google Scholar]
- 50.Shikata E, et al. Association of pharmacokinetic (CYP2C9) and pharmacodynamic (factors II, VII, IX, and X; proteins S and C; and gamma-glutamyl carboxylase) gene variants with warfarin sensitivity. Blood. 2004;103(7):2630–5. doi: 10.1182/blood-2003-09-3043. [DOI] [PubMed] [Google Scholar]
- 51.Arnett DK, Claas SA, Glasser SP. Pharmacogenetics of antihypertensive treatment. Vascul Pharmacol. 2006;44(2):107–18. doi: 10.1016/j.vph.2005.09.010. [DOI] [PubMed] [Google Scholar]
- 52.Psaty BM, et al. Health outcomes associated with various antihypertensive therapies used as first-line agents: a network meta-analysis. JAMA. 2003;289(19):2534–44. doi: 10.1001/jama.289.19.2534. [DOI] [PubMed] [Google Scholar]
- 53.Manunta P, et al. Adducin polymorphism affects renal proximal tubule reabsorption in hypertension. Hypertension. 1999;33(2):694–7. doi: 10.1161/01.hyp.33.2.694. [DOI] [PubMed] [Google Scholar]
- 54.Cusi D, et al. Polymorphisms of alpha-adducin and salt sensitivity in patients with essential hypertension. Lancet. 1997;349(9062):1353–7. doi: 10.1016/S0140-6736(97)01029-5. [DOI] [PubMed] [Google Scholar]
- 55.Glorioso N, et al. The role of alpha-adducin polymorphism in blood pressure and sodium handling regulation may not be excluded by a negative association study. Hypertension. 1999;34(4 Pt 1):649–54. doi: 10.1161/01.hyp.34.4.649. [DOI] [PubMed] [Google Scholar]
- 56.Sciarrone MT, et al. ACE and alpha-adducin polymorphism as markers of individual response to diuretic therapy. Hypertension. 2003;41(3):398–403. doi: 10.1161/01.HYP.0000057010.27011.2C. [DOI] [PubMed] [Google Scholar]
- 57.Davis BR, et al. Antihypertensive therapy, the alpha-adducin polymorphism, and cardiovascular disease in high-risk hypertensive persons: the Genetics of Hypertension-Associated Treatment Study. Pharmacogenomics J. 2007;7(2):112–22. doi: 10.1038/sj.tpj.6500395. [DOI] [PubMed] [Google Scholar]
- 58.Turner ST, et al. Effects of endothelial nitric oxide synthase, alpha-adducin, and other candidate gene polymorphisms on blood pressure response to hydrochlorothiazide. Am J Hypertens. 2003;16(10):834–9. doi: 10.1016/s0895-7061(03)01011-2. [DOI] [PubMed] [Google Scholar]
- 59.Schelleman H, et al. Interactions between five candidate genes and antihypertensive drug therapy on blood pressure. Pharmacogenomics J. 2006;6(1):22–6. doi: 10.1038/sj.tpj.6500339. [DOI] [PubMed] [Google Scholar]
- 60.Schelleman H, et al. The influence of the alpha-adducin G460W polymorphism and angiotensinogen M235T polymorphism on antihypertensive medication and blood pressure. Eur. J Hum Genet. 2006;14(7):860–6. doi: 10.1038/sj.ejhg.5201632. [DOI] [PubMed] [Google Scholar]
- 61.Psaty BM, et al. Diuretic therapy, the alpha-adducin gene variant, and the risk of myocardial infarction or stroke in persons with treated hypertension. JAMA. 2002;287(13):1680–9. doi: 10.1001/jama.287.13.1680. [DOI] [PubMed] [Google Scholar]
- 62.Schelleman H, et al. Diuretic-gene interaction and the risk of myocardial infarction and stroke. Pharmacogenomics J. 2007;7(5):346–52. doi: 10.1038/sj.tpj.6500428. [DOI] [PubMed] [Google Scholar]
- 63.Arnett DK, et al. Pharmacogenetic association of the angiotensin-converting enzyme insertion/deletion polymorphism on blood pressure and cardiovascular risk in relation to antihypertensive treatment: the Genetics of Hypertension-Associated Treatment (GenHAT) study. Circulation. 2005;111(25):3374–83. doi: 10.1161/CIRCULATIONAHA.104.504639. [DOI] [PubMed] [Google Scholar]
- 64.Frazier L, et al. Multilocus effects of the renin-angiotensin-aldosterone system genes on blood pressure response to a thiazide diuretic. Pharmacogenomics J. 2004;4(1):17–23. doi: 10.1038/sj.tpj.6500215. [DOI] [PubMed] [Google Scholar]
- 65.Jiang X, et al. Effect of renin-angiotensin-aldosterone system gene polymorphisms on blood pressure response to antihypertensive treatment. Chin Med J (Engl) 2007;120(9):782–6. [PubMed] [Google Scholar]
- 66.Turner ST, et al. C825T polymorphism of the G protein beta(3)-subunit and antihypertensive response to a thiazide diuretic. Hypertension. 2001;37(2 Part 2):739–43. doi: 10.1161/01.hyp.37.2.739. [DOI] [PubMed] [Google Scholar]
- 67.Schunkert H, et al. Association between a polymorphism in the G protein beta3 subunit gene and lower renin and elevated diastolic blood pressure levels. Hypertension. 1998;32(3):510–3. doi: 10.1161/01.hyp.32.3.510. [DOI] [PubMed] [Google Scholar]
- 68.Mason DA, et al. A gain-of-function polymorphism in a G-protein coupling domain of the human beta1-adrenergic receptor. J Biol Chem. 1999;274(18):12670–4. doi: 10.1074/jbc.274.18.12670. [DOI] [PubMed] [Google Scholar]
- 69.O’Shaughnessy KM, et al. The gain-of-function G389R variant of the beta1-adrenoceptor does not influence blood pressure or heart rate response to beta-blockade in hypertensive subjects. Clin Sci (Lond) 2000;99(3):233–8. [PubMed] [Google Scholar]
- 70.Liu J, et al. Gly389Arg polymorphism of beta1-adrenergic receptor is associated with the cardiovascular response to metoprolol. Clin Pharmacol Ther. 2003;74(4):372–9. doi: 10.1016/S0009-9236(03)00224-8. [DOI] [PubMed] [Google Scholar]
- 71.Sofowora GG, et al. A common beta1-adrenergic receptor polymorphism (Arg389Gly) affects blood pressure response to beta-blockade. Clin Pharmacol Ther. 2003;73(4):366–71. doi: 10.1016/s0009-9236(02)17734-4. [DOI] [PubMed] [Google Scholar]
- 72.Liu J, et al. beta1-Adrenergic receptor polymorphisms influence the response to metoprolol monotherapy in patients with essential hypertension. Clin Pharmacol Ther. 2006;80(1):23–32. doi: 10.1016/j.clpt.2006.03.004. [DOI] [PubMed] [Google Scholar]
- 73.Johnson JA, et al. Beta 1-adrenergic receptor polymorphisms and antihypertensive response to metoprolol. Clin Pharmacol Ther. 2003;74(1):44–52. doi: 10.1016/S0009-9236(03)00068-7. [DOI] [PubMed] [Google Scholar]
- 74.Zateyshchikov DA, et al. Association of CYP2D6 and ADRB1 genes with hypotensive and antichronotropic action of betaxolol in patients with arterial hypertension. Fundam Clin Pharmacol. 2007;21(4):437–43. doi: 10.1111/j.1472-8206.2007.00518.x. [DOI] [PubMed] [Google Scholar]
- 75.Filigheddu F, et al. Genetic polymorphisms of the beta-adrenergic system: association with essential hypertension and response to beta-blockade. Pharmacogenomics J. 2004;4(3):154–60. doi: 10.1038/sj.tpj.6500247. [DOI] [PubMed] [Google Scholar]
- 76.Kurland L, et al. Angiotensinogen gene polymorphisms: relationship to blood pressure response to antihypertensive treatment. Results from the Swedish Irbesartan Left Ventricular Hypertrophy Investigation vs Atenolol (SILVHIA) trial. Am J Hypertens. 2004;17(1):8–13. doi: 10.1016/j.amjhyper.2003.09.009. [DOI] [PubMed] [Google Scholar]
- 77.Kurland L, et al. Angiotensin converting enzyme gene polymorphism predicts blood pressure response to angiotensin II receptor type 1 antagonist treatment in hypertensive patients. J Hypertens. 2001;19(10):1783–7. doi: 10.1097/00004872-200110000-00012. [DOI] [PubMed] [Google Scholar]
- 78.Dudley C, et al. Prediction of patient responses to antihypertensive drugs using genetic polymorphisms: investigation of renin-angiotensin system genes. J Hypertens. 1996;14(2):259–62. doi: 10.1097/00004872-199602000-00016. [DOI] [PubMed] [Google Scholar]
- 79.Harrap SB, et al. The ACE gene I/D polymorphism is not associated with the blood pressure and cardiovascular benefits of ACE inhibition. Hypertension. 2003;42(3):297–303. doi: 10.1161/01.HYP.0000088322.85804.96. [DOI] [PubMed] [Google Scholar]
- 80.Hingorani AD, et al. Renin-angiotensin system gene polymorphisms influence bloodpressure and the response to angiotensin converting enzyme inhibition. J Hypertens. 1995;13(12 Pt 2):1602–9. [PubMed] [Google Scholar]
- 81.Kurland L, et al. Aldosterone synthase (CYP11B2) -344 C/T polymorphism is related to antihypertensive response: result from the Swedish Irbesartan Left Ventricular Hypertrophy Investigation versus Atenolol (SILVHIA) trial. Am J Hypertens. 2002;15(5):389–93. doi: 10.1016/s0895-7061(02)02256-2. [DOI] [PubMed] [Google Scholar]
- 82.Redon J, et al. Renin-angiotensin system gene polymorphisms: relationship with blood pressure and microalbuminuria in telmisartan-treated hypertensive patients. Pharmacogenomics J. 2005;5(1):14–20. doi: 10.1038/sj.tpj.6500280. [DOI] [PubMed] [Google Scholar]
- 83.Ortlepp JR, et al. Variants of the CYP11B2 gene predict response to therapy with candesartan. Eur J Pharmacol. 2002;445(1-2):151–2. doi: 10.1016/s0014-2999(02)01766-1. [DOI] [PubMed] [Google Scholar]
- 84.Cheung BM, et al. Meta-analysis of large randomized controlled trials to evaluate the impact of statins on cardiovascular outcomes. Br J Clin Pharmacol. 2004;57(5):640–51. doi: 10.1111/j.1365-2125.2003.02060.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Chasman DI, et al. Pharmacogenetic study of statin therapy and cholesterol reduction. JAMA. 2004;291(23):2821–7. doi: 10.1001/jama.291.23.2821. [DOI] [PubMed] [Google Scholar]
- 86.Thompson JF, et al. An association study of 43 SNPs in 16 candidate genes with atorvastatin response. Pharmacogenomics J. 2005;5(6):352–8. doi: 10.1038/sj.tpj.6500328. [DOI] [PubMed] [Google Scholar]
- 87.Hannuksela ML, et al. Relation of polymorphisms in the cholesteryl ester transfer protein gene to transfer protein activity and plasma lipoprotein levels in alcohol drinkers. Atherosclerosis. 1994;110(1):35–44. doi: 10.1016/0021-9150(94)90065-5. [DOI] [PubMed] [Google Scholar]
- 88.Kuivenhoven JA, et al. The role of a common variant of the cholesteryl ester transfer protein gene in the progression of coronary atherosclerosis. The Regression Growth Evaluation Statin Study Group. N Engl J Med. 1998;338(2):86–93. doi: 10.1056/NEJM199801083380203. [DOI] [PubMed] [Google Scholar]
- 89.de Grooth GJ, et al. The cholesteryl ester transfer protein (CETP) TaqIB polymorphism in the cholesterol and recurrent events study: no interaction with the response to pravastatin therapy and no effects on cardiovascular outcome: a prospective analysis of the CETP TaqIB polymorphism on cardiovascular outcome and interaction with cholesterol-lowering therapy. J Am Coll Cardiol. 2004;43(5):854–7. doi: 10.1016/j.jacc.2003.08.056. [DOI] [PubMed] [Google Scholar]
- 90.Freeman DJ, et al. A polymorphism of the cholesteryl ester transfer protein gene predicts cardiovascular events in non-smokers in the West of Scotland Coronary Prevention Study. Eur. Heart J. 2003;24(20):1833–42. doi: 10.1016/j.ehj.2003.07.001. [DOI] [PubMed] [Google Scholar]
- 91.Klerkx AH, et al. Cholesteryl ester transfer protein concentration is associated with progression of atherosclerosis and response to pravastatin in men with coronary artery disease (REGRESS) Eur J Clin Invest. 2004;34(1):21–8. doi: 10.1111/j.1365-2362.2004.01281.x. [DOI] [PubMed] [Google Scholar]
- 92.Marschang P, et al. Plasma cholesteryl ester transfer protein concentrations predict cardiovascular events in patients with coronary artery disease treated with pravastatin. J Intern Med. 2006;260(2):151–9. doi: 10.1111/j.1365-2796.2006.01674.x. [DOI] [PubMed] [Google Scholar]
- 93.Carlquist JF, et al. The cholesteryl ester transfer protein Taq1B gene polymorphism predicts clinical benefit of statin therapy in patients with significant coronary artery disease. Am Heart J. 2003;146(6):1007–14. doi: 10.1016/S0002-8703(03)00501-5. [DOI] [PubMed] [Google Scholar]
- 94.Boekholdt SM, et al. Cholesteryl ester transfer protein TaqIB variant, high-density lipoprotein cholesterol levels, cardiovascular risk, and efficacy of pravastatin treatment: individual patient meta-analysis of 13,677 subjects. Circulation. 2005;111(3):278–87. doi: 10.1161/01.CIR.0000153341.46271.40. [DOI] [PubMed] [Google Scholar]
- 95.Sing K, et al. Lipoprotein lipase gene mutations, plasma lipid levels, progression/regression of coronary atherosclerosis, response to therapy, and future clinical events. Lipoproteins and Coronary Atherosclerosis Study. Atherosclerosis. 1999;144(2):435–42. doi: 10.1016/s0021-9150(99)00004-0. [DOI] [PubMed] [Google Scholar]
- 96.Lahoz C, et al. The -514C/T polymorphism of the hepatic lipase gene significantly modulates the HDL-cholesterol response to statin treatment. Atherosclerosis. 2005;182(1):129–34. doi: 10.1016/j.atherosclerosis.2005.02.001. [DOI] [PubMed] [Google Scholar]
- 97.Ordovas JM, et al. Effect of apolipoprotein E and A-IV phenotypes on the low density lipoprotein response to HMG CoA reductase inhibitor therapy. Atherosclerosis. 1995;113(2):157–66. doi: 10.1016/0021-9150(94)05439-p. [DOI] [PubMed] [Google Scholar]
- 98. Pedro-Botet J, et al. Apolipoprotein E genotype affects plasma lipid response to atorvastatin in a gender specific mannerApolipoprotein E genotype affects plasma lipid response to atorvastatin in a gender specific manner . Atherosclerosis. 2001;158(1):183–93. doi: 10.1016/s0021-9150(01)00410-5. [DOI] [PubMed] [Google Scholar]
- 99. Ballantyne CM, et al. Apolipoprotein E genotypes and response of plasma lipids and progression-regression of coronary atherosclerosis to lipid-lowering drug therapyApolipoprotein E genotypes and response of plasma lipids and progression-regression of coronary atherosclerosis to lipid-lowering drug therapy. J Am Coll Cardiol. 2000;36(5):1572–8. doi: 10.1016/s0735-1097(00)00918-9. [DOI] [PubMed] [Google Scholar]
- 100.Gerdes LU, et al. The apolipoprotein epsilon4 allele determines prognosis and the effect on prognosis of simvastatin in survivors of myocardial infarction: a substudy of the Scandinavian simvastatin survival study. Circulation. 2000;101(12):1366–71. doi: 10.1161/01.cir.101.12.1366. [DOI] [PubMed] [Google Scholar]
- 101.Maitland-van der Zee AH, et al. The effectiveness of hydroxy-methylglutaryl coenzyme A reductase inhibitors (statins) in the elderly is not influenced by apolipoprotein E genotype. Pharmacogenetics. 2002;12(8):647–53. doi: 10.1097/00008571-200211000-00009. [DOI] [PubMed] [Google Scholar]
- 102.Deakin S, et al. Simvastatin modulates expression of the PON1 gene and increases serum paraoxonase: a role for sterol regulatory element-binding protein-2. Arterioscler Thromb Vasc Biol. 2003;23(11):2083–9. doi: 10.1161/01.ATV.0000096207.01487.36. [DOI] [PubMed] [Google Scholar]
- 103.Malin R, et al. Paraoxonase genotype modifies the effect of pravastatin on high-density lipoprotein cholesterol. Pharmacogenetics. 2001;11(7):625–33. doi: 10.1097/00008571-200110000-00009. [DOI] [PubMed] [Google Scholar]
- 104.Turban S, et al. A prospective study of paraoxonase gene Q/R192 polymorphism and severity, progression and regression of coronary atherosclerosis, plasma lipid levels, clinical events and response to fluvastatin. Atherosclerosis. 2001;154(3):633–40. doi: 10.1016/s0021-9150(00)00495-0. [DOI] [PubMed] [Google Scholar]
- 105.Yokoyama C, et al. SREBP-1, a basic-helix-loop-helix-leucine zipper protein that controls transcription of the low density lipoprotein receptor gene. Cell. 1993;75(1):187–97. [PubMed] [Google Scholar]
- 106.Sakakura Y, et al. Sterol regulatory element-binding proteins induce an entire pathway of cholesterol synthesis. Biochem. Biophys Res Commun. 2001;286(1):176–83. doi: 10.1006/bbrc.2001.5375. [DOI] [PubMed] [Google Scholar]
- 107.Marian AJ, et al. Interactions between angiotensin-I converting enzyme insertion/deletion polymorphism and response of plasma lipids and coronary atherosclerosis to treatment with fluvastatin: the lipoprotein and coronary atherosclerosis study. J. Am Coll Cardiol. 2000;35(1):89–95. doi: 10.1016/s0735-1097(99)00535-5. [DOI] [PubMed] [Google Scholar]
- 108.Maitland-van der Zee AH, et al. Absence of an interaction between the angiotensinconverting enzyme insertion-deletion polymorphism and pravastatin on cardiovascular disease in high-risk hypertensive patients: the Genetics of Hypertension-Associated Treatment (GenHAT) study. Am Heart J. 2007;153(1):54–8. doi: 10.1016/j.ahj.2006.10.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Bray PF, et al. The platelet Pl(A2) and angiotensin-converting enzyme (ACE) D allele polymorphisms and the risk of recurrent events after acute myocardial infarction. Am J Cardiol. 2001;88(4):347–52. doi: 10.1016/s0002-9149(01)01677-0. [DOI] [PubMed] [Google Scholar]
- 110.Maitland-van der Zee AH, et al. Effectiveness of HMG-CoA reductase inhibitors is modified by the ACE insertion deletion polymorphism. Atherosclerosis. 2004;175(2):377–9. doi: 10.1016/j.atherosclerosis.2004.04.009. [DOI] [PubMed] [Google Scholar]
- 111.Boekholdt SM, et al. Variants of toll-like receptor 4 modify the efficacy of statin therapy and the risk of cardiovascular events. Circulation. 2003;107(19):2416–21. doi: 10.1161/01.CIR.0000068311.40161.28. [DOI] [PubMed] [Google Scholar]
- 112.Holloway JW, Yang IA, Ye S. Variation in the toll-like receptor 4 gene and susceptibility to myocardial infarction. Pharmacogenet Genomics. 2005;15(1):15–21. doi: 10.1097/01213011-200501000-00003. [DOI] [PubMed] [Google Scholar]
- 113.Tachibana-Iimori R, et al. Effect of genetic polymorphism of OATP-C (SLCO1B1) on lipid-lowering response to HMG-CoA reductase inhibitors. Drug Metab Pharmacokinet. 2004;19(5):375–80. doi: 10.2133/dmpk.19.375. [DOI] [PubMed] [Google Scholar]
- 114.Mwinyi J, et al. Evidence for inverse effects of OATP-C (SLC21A6) 5 and 1b haplotypes on pravastatin kinetics. Clin Pharmacol Ther. 2004;75(5):415–21. doi: 10.1016/j.clpt.2003.12.016. [DOI] [PubMed] [Google Scholar]
- 115.Igel M, et al. Impact of the SLCO1B1 polymorphism on the pharmacokinetics and lipidlowering efficacy of multiple-dose pravastatin. Clin Pharmacol Ther. 2006;79(5):419–26. doi: 10.1016/j.clpt.2006.01.010. [DOI] [PubMed] [Google Scholar]
- 116.Niemi M, et al. Acute effects of pravastatin on cholesterol synthesis are associated with SLCO1B1 (encoding OATP1B1) haplotype *17. Pharmacogenet Genomics. 2005;15(5):303–9. doi: 10.1097/01213011-200505000-00005. [DOI] [PubMed] [Google Scholar]
- 117.Kajinami K, et al. Polymorphisms in the multidrug resistance-1 (MDR1) gene influence the response to atorvastatin treatment in a gender-specific manner. Am J Cardiol. 2004;93(8):1046–50. doi: 10.1016/j.amjcard.2004.01.014. [DOI] [PubMed] [Google Scholar]
- 118.Fiegenbaum M, et al. The role of common variants of ABCB1, CYP3A4, and CYP3A5 genes in lipid-lowering efficacy and safety of simvastatin treatment. Clin Pharmacol Ther. 2005;78(5):551–8. doi: 10.1016/j.clpt.2005.08.003. [DOI] [PubMed] [Google Scholar]
- 119.Mangravite LM, Thorn CF, Krauss RM. Clinical implications of pharmacogenomics of statin treatment. Pharmacogenomics J. 2006;6(6):360–74. doi: 10.1038/sj.tpj.6500384. [DOI] [PubMed] [Google Scholar]
- 120.Mangravite LM, Krauss RM. Pharmacogenomics of statin response. Curr Opin Lipidol. 2007;18(4):409–14. doi: 10.1097/MOL.0b013e328235a5a2. [DOI] [PubMed] [Google Scholar]
- 121.McKinney BA, et al. Machine learning for detecting gene-gene interactions: a review. Appl Bioinformatics. 2006;5(2):77–88. doi: 10.2165/00822942-200605020-00002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Angiolillo DJ, et al. Lack of association between the P2Y12 receptor gene polymorphism and platelet response to clopidogrel in patients with coronary artery disease. Thromb Res. 2005;116(6):491–7. doi: 10.1016/j.thromres.2005.03.001. [DOI] [PubMed] [Google Scholar]
- 123.Schwartz GL, et al. Interacting effects of gender and genotype on blood pressure response to hydrochlorothiazide. Kidney Int. 2002;62(5):1718–23. doi: 10.1046/j.1523-1755.2002.00624.x. [DOI] [PubMed] [Google Scholar]
- 124.Todd GP, et al. Relation between changes in blood pressure and serum ACE activity after a single dose of enalapril and ACE genotype in healthy subjects. Br J Clin Pharmacol. 1995;39(2):131–4. doi: 10.1111/j.1365-2125.1995.tb04419.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Nakano Y, et al. Angiotensin I-converting enzyme gene polymorphism and acute response to captopril in essential hypertension. Am J Hypertens. 1997;10(9 Pt 1):1064–8. doi: 10.1016/s0895-7061(97)00279-3. [DOI] [PubMed] [Google Scholar]
- 126.Kohno M, et al. Association between angiotensin-converting enzyme gene polymorphisms and regression of left ventricular hypertrophy in patients treated with angiotensinconverting enzyme inhibitors. Am J Med. 1999;106(5):544–9. doi: 10.1016/s0002-9343(99)00067-4. [DOI] [PubMed] [Google Scholar]
- 127.Penno G, et al. Effect of angiotensin-converting enzyme (ACE) gene polymorphism on progression of renal disease and the influence of ACE inhibition in IDDM patients: findings from the EUCLID Randomized Controlled Trial. EURODIAB Controlled Trial of Lisinopril in IDDM. Diabetes. 1998;47(9):1507–11. doi: 10.2337/diabetes.47.9.1507. [DOI] [PubMed] [Google Scholar]
- 128.Yu H, Zhang Y, Liu G. Relationship between polymorphism of the angiotensinconverting enzyme gene and the response to angiotensin-converting enzyme inhibition in hypertensive patients. Hypertens Res. 2003;26(11):881–6. doi: 10.1291/hypres.26.881. [DOI] [PubMed] [Google Scholar]
- 129.Yu Y, et al. Associations of baseline blood pressure levels and efficacy of Benazepril treatment with interaction of alpha-adducin and ACE gene polymorphisms in hypertensives. Clin Exp Hypertens. 2005;27(1):83–94. doi: 10.1081/ceh-200044273. [DOI] [PubMed] [Google Scholar]
- 130.van der Kleij FG, et al. ACE polymorphism does not determine short-term renal response to ACE-inhibition in proteinuric patients. Nephrol Dial Transplant. 1997;12(2):42–6. [PubMed] [Google Scholar]
- 131.Moriyama T, et al. Association of angiotensin I-converting enzyme gene polymorphism with susceptibility to antiproteinuric effect of angiotensin I-converting enzyme inhibitors in patients with proteinuria. J Am Soc Nephrol. 1995;6(6):1676–8. doi: 10.1681/ASN.V661676. [DOI] [PubMed] [Google Scholar]
- 132.Cannella G, et al. Angiotensin-converting enzyme gene polymorphism and reversibility of uremic left ventricular hypertrophy following long-term antihypertensive therapy. Kidney Int. 1998;54(2):618–26. doi: 10.1046/j.1523-1755.1998.00027.x. [DOI] [PubMed] [Google Scholar]
- 133.O’Toole L, et al. Effect of the insertion/deletion polymorphism of the angiotensinconverting enzyme gene on response to angiotensin-converting enzyme inhibitors in patients with heart failure. J Cardiovasc Pharmacol. 1998;32(6):988–94. doi: 10.1097/00005344-199812000-00017. [DOI] [PubMed] [Google Scholar]
- 134.Mizuiri S, et al. Renal hemodynamic changes induced by captopril and angiotensinconverting enzyme gene polymorphism. Nephron. 1997;75(3):310–4. doi: 10.1159/000189554. [DOI] [PubMed] [Google Scholar]
- 135.Ohmichi N, et al. Relationship between the response to the angiotensin converting enzyme inhibitor imidapril and the angiotensin converting enzyme genotype. Am J Hypertens. 1997;10(8):951–5. doi: 10.1016/s0895-7061(97)00121-0. [DOI] [PubMed] [Google Scholar]
- 136.Weekers L, et al. Modulation of the renal response to ACE inhibition by ACE insertion/deletion polymorphism during hyperglycemia in normotensive, normoalbuminuric type 1 diabetic patients. Diabetes. 2005;54(10):2961–7. doi: 10.2337/diabetes.54.10.2961. [DOI] [PubMed] [Google Scholar]
- 137. Suwelack B, et al. Impact of ACE polymorphism on renal allograft function, blood pressure, and proteinuria under ACE inhibitionImpact of ACE polymorphism on renal allograft function, blood pressure, and proteinuria under ACE inhibition. Transplant Proc. 2002;34(5):1763–6. doi: 10.1016/s0041-1345(02)03058-0. [DOI] [PubMed] [Google Scholar]
- 138.Stavroulakis GA, et al. Predicting response to chronic antihypertensive treatment with fosinopril: the role of angiotensin-converting enzyme gene polymorphism. Cardiovasc Drugs Ther. 2000;14(4):427–32. doi: 10.1023/a:1007820401377. [DOI] [PubMed] [Google Scholar]
- 139.Hernandez D, et al. Regression of left ventricular hypertrophy by lisinopril after renal transplantation: role of ACE gene polymorphism. Kidney Int. 2000;58(2):889–97. doi: 10.1046/j.1523-1755.2000.00239.x. [DOI] [PubMed] [Google Scholar]
- 140.Mondorf UF, et al. Contribution of angiotensin I converting enzyme gene polymorphism and angiotensinogen gene polymorphism to blood pressure regulation in essential hypertension. Am J Hypertens. 1998;11(2):174–83. doi: 10.1016/s0895-7061(97)00402-0. [DOI] [PubMed] [Google Scholar]
- 141.Yu H, et al. T1198C polymorphism of the angiotensinogen gene and antihypertensive response to angiotensin-converting enzyme inhibitors. Hypertens Res. 2005;28(12):981–6. doi: 10.1291/hypres.28.981. [DOI] [PubMed] [Google Scholar]
- 142.Su X, et al. Association between angiotensinogen, angiotensin II receptor genes, and blood pressure response to an angiotensin-converting enzyme inhibitor. Circulation. 2007;115(6):725–32. doi: 10.1161/CIRCULATIONAHA.106.642058. [DOI] [PubMed] [Google Scholar]
- 143.Bis JC, et al. Angiotensinogen Met235Thr polymorphism, angiotensin-converting enzyme inhibitor therapy, and the risk of nonfatal stroke or myocardial infarction in hypertensive patients. Am J Hypertens. 2003;16(12):1011–7. doi: 10.1016/j.amjhyper.2003.07.018. [DOI] [PubMed] [Google Scholar]
- 144.Miller JA, Thai K, Scholey JW. Angiotensin II type 1 receptor gene polymorphism predicts response to losartan and angiotensin II. Kidney Int. 1999;56(6):2173–80. doi: 10.1046/j.1523-1755.1999.00770.x. [DOI] [PubMed] [Google Scholar]
- 145.Ye P, Shang Y, Ding X. The influence of apolipoprotein B and E gene polymorphisms on the response to simvastatin therapy in patients with hyperlipidemia. Chin Med Sci J. 2003;18(1):9–13. [PubMed] [Google Scholar]
- 146. Nestel P, et al. A comparative study of the efficacy of simvastatin and gemfibrozil in combined hyperlipoproteinemia: prediction of response by baseline lipids, apo E genotype, lipoprotein(a) and insulinA comparative study of the efficacy of simvastatin and gemfibrozil in combined hyperlipoproteinemia: prediction of response by baseline lipids, apo E genotype, lipoprotein(a) and insulin. Atherosclerosis. 1997;129(2):231–9. doi: 10.1016/s0021-9150(96)06031-5. [DOI] [PubMed] [Google Scholar]
- 147.Takane H, et al. Pharmacogenetic determinants of variability in lipid-lowering response to pravastatin therapy. J Hum Genet. 2006;51(9):822–6. doi: 10.1007/s10038-006-0025-1. [DOI] [PubMed] [Google Scholar]
- 148.Pena R, et al. Effect of apoE genotype on the hypolipidaemic response to pravastatin in an outpatient setting. J Intern Med. 2002;251(6):518–25. doi: 10.1046/j.1365-2796.2002.00991.x. [DOI] [PubMed] [Google Scholar]
- 149.Sanllehy C, et al. Lack of interaction of apolipoprotein E phenotype with the lipoprotein response to lovastatin or gemfibrozil in patients with primary hypercholesterolemia. Metabolism. 1998;47(5):560–5. doi: 10.1016/s0026-0495(98)90240-2. [DOI] [PubMed] [Google Scholar]
- 150.Ojala JP, et al. Effect of apolipoprotein E polymorphism and XbaI polymorphism of apolipoprotein B on response to lovastatin treatment in familial and non-familial hypercholesterolaemia. J Intern Med. 1991;230(5):397–405. doi: 10.1111/j.1365-2796.1991.tb00464.x. [DOI] [PubMed] [Google Scholar]
- 151.Tavintharan S, et al. Apolipoprotein E genotype affects the response to lipid-lowering therapy in Chinese patients with type 2 diabetes mellitus. Diabetes Obes Metab. 2007;9(1):81–6. doi: 10.1111/j.1463-1326.2006.00577.x. [DOI] [PubMed] [Google Scholar]
- 152.Zuccaro P, et al. Tolerability of statins is not linked to CYP450 polymorphisms, but reduced CYP2D6 metabolism improves cholesteraemic response to simvastatin and fluvastatin. Pharmacol Res. 2007;55(4):310–7. doi: 10.1016/j.phrs.2006.12.009. [DOI] [PubMed] [Google Scholar]
- 153.Maitland-van der Zee AH, et al. Apolipoprotein-E polymorphism and response to pravastatin in men with coronary artery disease (REGRESS) Acta Cardiol. 2006;61(3):327–31. doi: 10.2143/AC.61.3.2014836. [DOI] [PubMed] [Google Scholar]
- 154. Ballantyne CM. Current and future aims of lipid-lowering therapy: changing paradigms and lessons from the Heart Protection Study on standards of efficacy and safetyCurrent and future aims of lipid-lowering therapy: changing paradigms and lessons from the Heart Protection Study on standards of efficacy and safety. Am J Cardiol. 2003;92(4B):3K–9K. doi: 10.1016/s0002-9149(03)00768-9. [DOI] [PubMed] [Google Scholar]
- 155.Salazar LA, et al. Lipid-lowering response of the HMG-CoA reductase inhibitor fluvastatin is influenced by polymorphisms in the low-density lipoprotein receptor gene in Brazilian patients with primary hypercholesterolemia. J Clin Lab Anal. 2000;14(3):125–31. doi: 10.1002/(SICI)1098-2825(2000)14:3<125::AID-JCLA7>3.0.CO;2-S. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Lahoz C, et al. Baseline levels of low-density lipoprotein cholesterol and lipoprotein (a) and the AvaII polymorphism of the low-density lipoprotein receptor gene influence the response of low-density lipoprotein cholesterol to pravastatin treatment. Metabolism. 2005;54(6):741–7. doi: 10.1016/j.metabol.2004.12.020. [DOI] [PubMed] [Google Scholar]
- 157.Guzman EC, et al. Association of the apolipoprotein B gene polymorphisms with cholesterol levels and response to fluvastatin in Brazilian individuals with high risk for coronary heart disease. Clin Chem Lab Med. 2000;38(8):731–6. doi: 10.1515/CCLM.2000.103. [DOI] [PubMed] [Google Scholar]
- 158.Lahoz C, et al. Apo A-I promoter polymorphism influences basal HDL-cholesterol and its response to pravastatin therapy. Atherosclerosis. 2003;168(2):289–95. doi: 10.1016/s0021-9150(03)00094-7. [DOI] [PubMed] [Google Scholar]
- 159.Sorkin SC, et al. APOA1 polymorphisms are associated with variations in serum triglyceride concentrations in hypercholesterolemic individuals. Clin Chem Lab Med. 2005;43(12):1339–45. doi: 10.1515/CCLM.2005.229. [DOI] [PubMed] [Google Scholar]
- 160.van Venrooij FV, et al. Common cholesteryl ester transfer protein gene polymorphisms and the effect of atorvastatin therapy in type 2 diabetes. Diabetes Care. 2003;26(4):1216–23. doi: 10.2337/diacare.26.4.1216. [DOI] [PubMed] [Google Scholar]
- 161.Winkelmann BR, et al. Haplotypes of the cholesteryl ester transfer protein gene predict lipid-modifying response to statin therapy. Pharmacogenomics J. 2003;3(5):284–96. doi: 10.1038/sj.tpj.6500195. [DOI] [PubMed] [Google Scholar]
- 162. de Grooth GJ, et al. The relationship between cholesteryl ester transfer protein levels and risk factor profile in patients with familial hypercholesterolemiaThe relationship between cholesteryl ester transfer protein levels and risk factor profile in patients with familial hypercholesterolemia. Atherosclerosis. 2004;173(2):261–7. doi: 10.1016/j.atherosclerosis.2003.11.020. [DOI] [PubMed] [Google Scholar]
- 163.Kajinami K, et al. A promoter polymorphism in cholesterol 7alpha-hydroxylase interacts with apolipoprotein E genotype in the LDL-lowering response to atorvastatin. Atherosclerosis. 2005;180(2):407–15. doi: 10.1016/j.atherosclerosis.2004.12.019. [DOI] [PubMed] [Google Scholar]
- 164.Salek L, et al. Effects of SREBF-1a and SCAP polymorphisms on plasma levels of lipids, severity, progression and regression of coronary atherosclerosis and response to therapy with fluvastatin. J Mol Med. 2002;80(11):737–44. doi: 10.1007/s00109-002-0381-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Fiegenbaum M, et al. Determinants of variable response to simvastatin treatment: the role of common variants of SCAP, SREBF-1a and SREBF-2 genes. Pharmacogenomics J. 2005;5(6):359–64. doi: 10.1038/sj.tpj.6500334. [DOI] [PubMed] [Google Scholar]
- 166.Fan YM, et al. Effects of pravastatin therapy on serum lipids and coronary reactivity ar not associated with SREBP cleavage-activating protein polymorphism in healthy young men. Clin Genet. 2001;60(4):319–21. doi: 10.1034/j.1399-0004.2001.600413.x. [DOI] [PubMed] [Google Scholar]
- 167.Rodrigues AC, et al. High baseline serum total and LDL cholesterol levels are associated with MDR1 haplotypes in Brazilian hypercholesterolemic individuals of European descent. Braz J Med Biol Res. 2005;38(9):1389–97. doi: 10.1590/s0100-879x2005000900014. [DOI] [PubMed] [Google Scholar]