1,103
Views
5
CrossRef citations to date
0
Altmetric
Review Article

Genetic and ethnic modulation of cardiovascular toxicity of vascular endothelial growth factor inhibitors

, , &
Pages 46-56 | Received 07 Jul 2017, Accepted 13 Sep 2017, Published online: 27 Sep 2017

Abstract

Vascular endothelial growth factor (VEGF) inhibitors, including monoclonal antibodies and tyrosine kinase inhibitors (TKIs), are important as anticancer treatments through curbing tumour angiogenesis and growth. VEGF inhibitors have significant cardiovascular effects. By blocking VEGF receptors, ligands, or signal pathways, VEGF inhibitors disturb the balance between vasodilation and vasoconstriction, undermine endothelial cell integrity, and activate cardiomyocyte apoptosis. VEGF inhibitors increase risks of hypertension, heart failure, thromboembolism and arrhythmia. Genetic and geographic studies showed that genetic polymorphisms likely play significant predictive or prognostic roles in cardiovascular toxicity associated with VEGF inhibitors. This review updates current understandings of VEGF inhibitors on cardiovascular toxicity, explores potential mechanisms, and clarifies whether genetic or ethnic factors contribute to their adverse effects.

    Key Messages

  • VEGF inhibitors disturb the balance between vasodilation and vasoconstriction, undermine endothelial cell integrity and activate cardiomyocyte apoptosis.

  • VEGF inhibitors increase risks of hypertension, heart failure, thromboembolism and arrhythmia.

  • Genetic and geographic studies showed that genetic polymorphisms likely play significant predictive or prognostic roles in cardiovascular toxicity associated with VEGF inhibitors.

Introduction

Onco-cardiology is critical for optimal cancer care, since cancer and cardiovascular diseases (CVDs) often coexist in middle-aged and senior patients. Anticancer therapies may aggravate CVD, and their side effects may cause cancer treatments to fail. Angiogenesis inhibitors are novel agents that inhibit tumour growth and angiogenesis by blocking vascular endothelial growth factor (VEGF) receptors or interfering in their signal pathways [Citation1]. VEGF monoclonal antibodies, such as bevacizumab [Citation2], and tyrosine kinase inhibitors (TKIs) with anti-VEGF activities, such as sunitinib [Citation3], sorafenib [Citation4], pazopanib [Citation5] and axitinib [Citation6], were proven to prolong the survival of cancer patients with various advanced or metastatic malignant solid cancers (). However, VEGF can promote endothelial proliferation, angiogenesis, and the production of nitric oxide (NO) and prostacyclin, and reduce endothelin-1 [Citation7,Citation8]. Theoretically, anti-VEGF agents can also undermine normal angiogenesis and cardiovascular homeostasis.

Table 1. Vascular endothelial growth factor inhibitors.

Genetic and geographic studies showed that genetic polymorphisms or ethnic differences probably play significant predictive or prognostic roles in cardiovascular toxicity associated with VEGF inhibitors. Genetic variants may increase or reduce the potential toxicity of VEGF inhibitors, influencing their short- or long-term treatment outcomes. Accordingly, genetic monitoring may optimize the application of VEGF inhibitors in cancer treatment. This review updates current understandings of VEGF inhibitors on cardiovascular toxicity, explores potential mechanisms and clarifies whether genetic or ethnic factors contribute to their adverse effects.

Methodology

We examined the publications in PubMed from 2004 to 2017 by combining the keywords “vascular endothelial growth factor” and “cancer”, and identified associated publications with the focus on VEGF inhibitors by the keywords of “angiogenesis inhibitor”, “monoclonal antibody”, “tyrosine kinase inhibitor”, “protein kinase inhibitor”, “bevacizumab”, “aflibercept”, “ramucirumab”, “sunitinib,” “sorafenib, “pazopanib”, “axitinib”, “vandetanib”, “regorafenib”, “cabozantinib”, or “lenvatinib”, respectively. We also searched literatures in PubMed for VEGF inhibitor-induced cardiotoxicities by the keywords of “hypertension”, “heart failure”, “thromboembolism”, “cardiac ischemia”, “arrhythmia” or “cardiovascular toxicity” with or without related to “genetic polymorphism” and “racial”. In the 925 publications screened, we excluded case reports, disease reviews, editorials, duplicates, non-English articles and the studies not related to adult cancer therapy or VEGF-inhibitor cardiovascular toxicity, but additionally included some references from the published clinical trials. Under this setting, a total of 398 articles were included and reviewed.

The grading system of cardiovascular toxicity of VEGF inhibitor in this review and associated clinical trials is based on Common Terminology Criteria for Adverse Events version 1, 2, 3 or 4 [Citation9]. Grade 3 or higher represents high grade of adverse events. The grading criteria of hypertension or raised blood pressure is as followed: grade 1, systolic blood pressure (BP) 120-139 mmHg or diastolic BP 80-89 mmHg; grade 2, recurrent or persistent (≥24 h) systolic BP 140-159 mmHg or diastolic BP 90-99 mmHg, or symptomatic diastolic BP increases by>20 mmHg, and monotherapy might be indicated; grade 3, systolic BP ≥ 160 mmHg or diastolic BP ≥ 100 mmHg and more than one drug or more intensive therapy than previously might be indicated; grade 4, life-threatening consequences, such as malignant hypertension, transient or permanent neurologic deficit and hypertensive crisis, and urgent intervention might be indicated [Citation9]. The grading criteria of QTc prolongation is as followed: grade 1, QTc 450-480 ms; grade 2, QTc 481-500 ms; grade 3, QTc ≥ 501 ms on at least two separate electrical leads; grade 4, QTc ≥ 501 ms or > 60 ms change from baseline and torsade de pointes or polymorphic ventricular tachycardia or signs/symptoms of serious arrhythmia [Citation9].

Cellular and molecular mechanisms of cardiovascular toxicity of VEGF inhibitors

VEGFs promote endothelial cell proliferation, induce two vasodilators, NO, and prostacyclin, and reduce the vasoconstrictor, endothelin-1 () [Citation7,Citation8]. By blocking VEGFs, their receptors, or signal pathways, VEGF inhibitors undermine endothelial cell integrity and the balance between vasodilation and vasoconstriction () [Citation7,Citation8]. Endothelial cells are important components for maintaining vascular homeostasis and avoiding haemorrhaging and thrombosis. Thus, VEGF inhibitors interfere with normal vascular homeostasis, leading to higher tendencies for hypertension and thrombosis [Citation7,Citation8]. Previous studies reported similar manifestations between cardiovascular toxicity associated with VEGF inhibitors and pregnancy-associated cardiovascular complications, such as preeclampsia and peripartum cardiomyopathies [Citation10,Citation11]. VEGF inhibitors induce a syndrome-like preeclampsia, including hypertension, proteinuria and deteriorating renal function. Renal biopsies of such patients revealed thrombotic microangiopathy, a feature similar to preeclampsia [Citation12–14]. In addition, soluble feline McDonough sarcoma (FMS)-related tyrosine kinase 1 (FLT1), also called soluble VEGF receptor 1 (VEGFR1), can cause preeclampsia and peripartum cardiomyopathies [Citation15,Citation16]. Like VEGF inhibitors, FLT1, secreted by the placenta in late gestation, blocks the VEGF signal pathway [Citation16], indicating a potential causal relation between VEGF inhibitors and their cardiovascular toxicity.

Figure 1. Vascular endothelial growth factor (VEGF) and its associated signal pathways maintain vascular homeostasis through promoting endothelial cell proliferation, inducing two vasodilators, nitric oxide and prostacyclin, and reducing endothelin-1, a vasoconstrictor.

Figure 1. Vascular endothelial growth factor (VEGF) and its associated signal pathways maintain vascular homeostasis through promoting endothelial cell proliferation, inducing two vasodilators, nitric oxide and prostacyclin, and reducing endothelin-1, a vasoconstrictor.

Figure 2. By blocking vascular endothelial growth factors (VEGFs), their receptors, or signal pathways, VEGF inhibitors undermine endothelial cell integrity and the balance between vasodilation and vasoconstriction, leading to hypertension, cardiac ischemia, thromboembolism, heart failure and microangiopathy.

Figure 2. By blocking vascular endothelial growth factors (VEGFs), their receptors, or signal pathways, VEGF inhibitors undermine endothelial cell integrity and the balance between vasodilation and vasoconstriction, leading to hypertension, cardiac ischemia, thromboembolism, heart failure and microangiopathy.

VEGFs also play pivotal roles in cardiomyocyte protection. VEGFs protect against doxorubicin-induced apoptosis of cardiomyocytes through upregulating the pro-survival Akt/nuclear factor (NF)-κB/bcl-2 signalling pathway [Citation17]. Overexpression of VEGFs in doxorubicin-induced heart failure (HF) increases cardiac mitochondrial respiration, thereby preserving the left ventricular volume [Citation18]. In pacing-induced dilated cardiomyopathy, VEGFs activate the antiapoptotic regulator, Akt, thereby enhancing cardiomyocyte survival [Citation19]. VEGFs also abolish hypoxia-induced apoptotic factors, such as cleaved caspases-3, -8 and -9, as well as Bax in neonatal cardiomyocytes [Citation20]. Therefore, it is possible that VEGF inhibitors undermine these protective effects of VEGFs and activate cardiomyocyte apoptosis, leading to cardiac systolic dysfunction and HF.

While VEGF monoclonal antibodies mainly focus on VEGF receptors or ligands, TKIs not only block the VEGF signal pathway through targeting VEGF receptors (VEGFRs) and platelet-derived growth factor receptor, but also block multiple other kinases or signal pathways associated with tumour growth and inflammation, such as KIT kinases, colony stimulating factor 1 receptor (CSF1R), rearranged during transfection (RET), rapidly accelerated fibrosarcoma (RAF), fibroblast growth factor receptor (FGFR) and so on (). This multi-targeted property of TKIs may expand or strengthen their anticancer efficacy, but may also increase potential cardiovascular toxicity, such as HF [Citation21,Citation22].

Although the mechanisms of reduced left ventricular systolic function are not clear, it is hypothesized that TKIs inhibit multiple off-target kinase activities and the VEGF signalling pathway, leading to cardiac dysfunction [Citation23,Citation24]. Sunitinib was shown to inhibit ribosomal S6 kinase and AMP-activated protein kinase (AMPK), promoting ATP depletion and potential cardiomyocyte apoptosis [Citation23,Citation24]. Sorafenib, through blocking RAF1, inhibits the extracellular signal-regulated kinase kinase cascade, undermining regulation of cardiomyocyte survival, especially under stress [Citation23]. Currently, it remains unclear whether inhibition of RAF1 by sorafenib also activates two stress-induced, proapoptotic pathways associated with apoptosis, viz., signal-regulating kinase 1 and mammalian sterile 20 kinase 2 (MST2), which could lead to even more profound effects of sorafenib on cardiomyocyte apoptosis and potential HF [Citation23]. These potential detrimental effects of TKIs on regulating cardiomyocyte apoptosis and survival as well as energy utilization were hypothesized to cause cardiac dysfunction and HF.

Furthermore, VEGF inhibitors may also induce arrhythmia. The mechanism of drug-induced QTc prolongation was speculated to be a direct interaction between the studied drugs and the human ether-a-go-go-related gene (hERG) potassium channels, interfering with delayed rectifier potassium currents by delaying cardiac repolarization [Citation25–28]. In preclinical studies, sunitinib and vandetanib were found to have a significant interaction with the hERG channel [Citation29]. In addition, cardiac ischemia and HF induced by TKIs can cause QTc prolongation [Citation30–32], thus the electrophysiological effects of sunitinib and vandetanib on the hERG channel may aggravate the risk of ventricular arrhythmia related to QTc prolongation in cancer patients.

Although there are limited clinical reports about atrial fibrillation (AF) associated with TKIs, sunitinib was reported to inhibit AMPK [Citation23], a compensatory pathway activated by AF [Citation33]. Activation of AMPK by AF helps maintain atrial calcium handling and atrial contractions [Citation33]. AMPK was hypothesized to protect paroxysmal AF from progressing into chronic AF, while fractional AMPK phosphorylation increased with paroxysmal AF and decreased with persistent AF [Citation33]. Therefore, further studies are needed to clarify this potential interaction and relationship between sunitinib and AF through blocking AMPK.

VEGF inhibitor-induced hypertension

Hypertension is the most common cardiovascular toxicity of VEGF inhibitors. One meta-analysis of 12,949 patients treated with or without bevacizumab for various solid tumours from 19 randomized controlled trials (RCTs) revealed that the incidence of all cases of significantly elevated blood pressure was 24%, while high-grade elevated blood pressure was 8% [Citation34]. Bevacizumab increased the risk of significantly elevated blood pressure (relative risk [RR]: 5.38, 95% confidence interval [CI]: 3.6 3 ∼ 7.97) [Citation34]. A dose-dependent association was also identified. The RRs for significantly elevated blood pressure associated with low-dose (2.5 mg/kg per week) and high-dose bevacizumab (5 mg/kg per week) were 4.11 (95% CI: 2.4 9 ∼ 6.78) and 7.17 (95% CI: 3.9 1 ∼ 13.13), respectively [Citation34]. VEGF TKIs, such as sunitinib and sorafenib, also have similar incidences and risks of significantly elevated blood pressure. One meta-analysis of 4999 patients treated with or without sunitinib from 13 prospective trials revealed that the incidence of all cases of elevated blood pressure was 21.6%, while high-grade elevated blood pressure was 6.8% [Citation35]. Sunitinib significantly increased the risk of high-grade elevated blood pressure (RR: 22.72, 95% CI: 4.4 8 ∼ 115.29) [Citation35]. Another meta-analysis of 4599 patients using sorafenib showed that the incidence of all cases of elevated blood pressure was 23.4%, while high-grade elevated blood pressure was 5.7% [Citation36]. Other TKIs and monoclonal antibodies, including pazopanib [Citation37], axitinib [Citation6], lenvatinib [Citation38], ramucirumab [Citation39] and aflibercept [Citation40], all showed similar or even higher incidences of elevated blood pressure in phase III trials or meta-analyses.

Currently, it is still controversial as to whether a higher risk of hypertension induced by VEGF inhibitors predicts better efficacy and outcomes of cancer treatment. Previous studies showed that hypertension induced by VEGF inhibitors was associated with better treatment outcomes with sunitinib for renal cell carcinoma [Citation41,Citation42], sorafenib for hepatocellular carcinoma [Citation43], and bevacizumab for non-small cell lung cancer, colorectal cancer, ovarian cancer, and breast cancer [Citation44–48]. However, one systemic review of 6487 patients from seven phase III trials of bevacizumab for various cancers found no significant association between survival outcomes and early development of hypertension (blood pressure increased by>20 mmHg systolic or>10 mmHg diastolic within the first 60 days of treatment) [Citation49]. One retrospective study of 337 patients treated with pazopanib for non-adipocytic soft-tissue sarcomas showed no correlation between hypertension and treatment efficacy within 5 weeks of treatment [Citation50]. Thus, further investigations are needed to clarify these clinical implications between the risk of hypertension and treatment efficacy.

VEGF inhibitor-induced thromboembolisms

Thromboembolisms are another potential cardiovascular toxicity of VEGF inhibitors. VEGF inhibitors, including monoclonal antibodies and TKIs, have a significant association with arterial thromboembolisms, including cerebrovascular and cardiovascular ischemia or infarction [Citation51–54]. In one meta-analysis of 1745 patients from five RCTs for various cancers, compared to chemotherapy alone, chemotherapy with bevacizumab was associated with a higher risk of arterial thromboembolic events (ATEs) (hazard ratio [HR]: 2.0, 95% CI: 1.0 5 ∼ 3.75), but not with venous thromboembolic events (VTEs) [Citation51]. Two other larger meta-analyses also revealed that bevacizumab increased the risk of ATEs with no dose-dependent relation between low- and high-dose bevacizumab [Citation52,Citation53]. Of these two meta-analyses, one study involving 12,617 patients from 20 RCTs revealed that bevacizumab increased the risk of ATEs (RR: 1.44, 95% CI: 1.0 8 ∼ 1.91), with a higher risk of cardiac ischemia (RR: 2.14, 95% CI: 1.1 2 ∼ 4.08) and no significant risk of cerebral ischemia (RR: 1.37, 95% CI: 0.6 7 ∼ 2.79) [Citation53]. The other study with 13,026 patients from 20 RCTs also showed that bevacizumab increased the risk of ATEs (RR: 1.46, 95% CI: 1.1 1 ∼ 1.93) but provided no further information about different types of ATE [Citation52]. Recently, two newly introduced VEGF monoclonal antibodies, ramucirumab and aflibercept, were also reported to induce higher incidences of arterial embolism, compared to chemotherapy alone in their phase III trials [Citation39,Citation40]. VEGF-targeted TKIs are also associated with higher risks or incidences of ATEs in clinical trials or meta-analyses. One meta-analysis of 10,225 subjects from 10 studies revealed that sunitinib and sorafenib brought a higher risk of ATEs (RR: 3.03, 95% CI: 1.2 5 ∼ 7.37) [Citation54]. One phase III trial of 392 patients treated with or without lenvatinib for differentiated thyroid cancer refractory to radioiodine also revealed a higher incidence of ATEs (5% vs. 2%) [Citation38].

The risk of VTEs with VEGF inhibitors remains controversial. Although one meta-analysis of 7956 patients from 15 RCTs in 2008 revealed a higher risk of VTEs with bevacizumab [Citation55], another meta-analysis of 1745 patients from five RCTs in 2007 showed no significant association between VTEs and bevacizumab [Citation51]. One larger recent meta-analysis of 6055 patients from 10 RCTs in 2011 also showed no significant association between VTEs and bevacizumab [Citation56]. The author of the 2011 study speculated that the difference in results among these trials probably came from different study designs and methodologies [Citation56]. While the 2011 study pooled individual patient data to reduce the limitation of a meta-analysis of aggregated data, the 2008 study developed its meta-analysis by summarizing rates of VTEs and included trials which did not report all-grade VTEs and did not distinguish between venous and arterial events [Citation56]. As for TKIs, one recent meta-analysis of 7441 patients from 17 RCTs in 2013 showed no significant association between VTEs and TKIs [Citation57]. However, further clarifying investigations are still needed to resolve these conflicting reports.

VEGF inhibitor-induced HF

The use of VEGF TKIs is commonly associated with asymptomatic reduction in left ventricular systolic function and symptomatic HF [Citation21,Citation22]. In contrast, bevacizumab-associated HF was infrequently reported in some trials of advanced breast cancer with paclitaxel, capecitabine, or anthracycline, an indication for which it is not now approved [Citation58].

One meta-analysis involving 10,647 patients treated with different TKIs from 16 phase III trials and 5 phase II trials revealed that the incidence of all-grade HF was 2.39%, while that of high-grade HF was 1.19% [Citation22]. TKIs increased risks of both all-grade (RR: 2.69, 95% CI: 1.8 6 ∼ 3.87) and high-grade HF (CHF; RR: 1.65, 95% CI: 0.7 3 ∼ 3.70) [Citation22]. Although it is hypothesized that more specific TKIs possibly had lower cardiac toxicity [Citation24], this analysis found that more specific TKIs, such as axitinib, and non-specific TKIs, including sunitinib, sorafenib, vandetanib and pazopanib, seemed to have similar risks of HF [Citation22]. One retrospective study of 75 patients treated with sunitinib suggested that the incidences and impacts of TKI-associated HF were probably higher in the real world (28% for a ≥10% reduction in the left ventricular ejection fraction (LVEF); 19% for a ≥15% reduction in LVEF; and 8% for clinical HF), which was significantly higher than the risk reported in clinical trials [Citation59].

VEGF inhibitor-induced cardiac arrhythmogenesis

VEGF inhibitors were reported to induce arrhythmia in clinical trials and post-market reports. Bevacizumab was found to induce supraventricular arrhythmia and conduction disorders in one retrospective analysis, whereas bevacizumab-induced cardiac ischemia was hypothesized to induce these arrhythmic events [Citation60]. Moreover, interactions of TKIs with the hERG channel [Citation29] and AMPK pathway [Citation23] or genesis of cardiac ischemia [Citation54] and HF (22) by TKIs also increase potential arrhythmic risks.

Vandetanib and sunitinib were reported to possess greater arrhythmic risks. Moreover, it was reported that there was a dose-dependent relation about QTc prolongation induced by vandetanib [Citation61] and sunitinib [Citation62]. One meta-analysis of a total of 6548 patients from 18 clinical trials for arrhythmic events of TKIs, including sunitinib, pazopanib, axitinib and vandetanib, revealed that patients receiving TKIs had a significantly higher risk of all-grade (RR: 8.66, 95% CI: 4.9 2 ∼ 15.2) and high-grade (RR: 2.69, 95% CI: 1.3 3 ∼ 5.44) QTc prolongation than those receiving no TKIs [Citation61]. However, in that study, sunitinib and vandetanib were two major contributors to the significantly increased risk of QTc prolongation. That study included only two trials of pazopanib with a total of 806 patents and only one trial of axitinib with 630 patients, leading to limited study power for pazopanib and axitinib [Citation61]. Therefore, the arrhythmic risks of other TKIs, including sorafenib, pazopanib, axitinib, cabozantinib, ponatinib and regorafenib, remain unclear [Citation29,Citation61].

Ethnic and genetic modulation of VEGF inhibitors-induced hypertension

Ethnic differences are common in pharmacological treatment. Previous studies revealed that there was an ethnic difference in VEGF inhibitors-induced hypertension. One prospective observational cohort study including 227 African American patients and 1589 Caucasian patients with metastatic colorectal cancer receiving first-line bevacizumab therapy showed African Americans (13.7%) had a higher rate of worsening hypertension than Caucasian patients (8.9%), but had similar rates of arterial (1.8% vs. 2.3%) and venous (5.3% vs. 4.8%) thromboembolism [Citation63]. African Americans are known to have the highest incidence, prevalence, and severity of hypertension among different ethnic groups [Citation64–66], the cardiovascular toxicity of VEGF inhibitor may potentiate cardiovascular risks for this group, although clinical reports are limited.

One meta-analysis of 4308 patients with lung cancer showed that non-Asians had a higher risk of severe bleeding (RR: 2.17, 95% CI: 1.1 4 ∼ 4.15) and thromboembolisms (RR: 3.65, 95% CI: 1.8 9 ∼ 7.04) and a lower risk of severe proteinuria (RR: 0.43, 95% CI: 0.2 8 ∼ 0.66) than Asian populations, while severe hypertension and haemoptysis were similar in the two groups [Citation67]. In one phase III trial of 723 patients treated with axitinib or sorafenib for renal cell carcinoma, a higher incidence of all-grade or high-grade hypertension occurred in a Japanese population (axitinib: all-grade 64%, high-grade 44%; sorafenib: all-grade 62%, high-grade 45%) than in the overall population (axitinib: all-grade 40%, high-grade 16%; sorafenib: all-grade 29%, high-grade 11%) [Citation68].

One multinational, prospective study of 325 Asians and 4046 non-Asians treated with sunitinib for renal cell carcinoma revealed that Asians (23%; 4%) had a similar incidence of all-grade and high-grade hypertension to non-Asians (21%; 5%), but Asians treated in Asia had a higher incidence (30%) of all-grade hypertension than Asians treated outside Asia (8%) [Citation69]. The conclusion of that study suggested that even environmental factors should be considered with this difference, since these factors are difficult to control and evaluate [Citation69]. One pharmacokinetics study of axitinib for Japanese, Chinese and Caucasians suggested that there was no significant pharmacokinetic difference between Asians and Caucasians [Citation70], possibly indicating that clinical differences in VEGF therapy were based on other types of genetic polymorphisms rather than metabolic differences.

Genetic polymorphisms are also important factors regulating pharmacological responses and treatment outcomes. Some genetic studies showed that genetic polymorphisms or markers were able to predict bevacizumab- or sunitinib-induced hypertension () [Citation46, Citation71–78]. Genetic polymorphisms, such as SV2C(71) and VEGF-A c.*237C>T [Citation72], were more associated with bevacizumab-induced hypertension. VEGF-634 CC polymorphism was associated with higher risks of both bevacizumab-induced hypertension and thromboembolism [Citation73]. On the other hand, VEGF-1498 TT [Citation46], VEGF-2578 C/C [Citation74] and VEGF-936 C/C [Citation74] were associated with less high-grade bevacizumab-induced hypertension. In addition, Schneider et al. examined VEGF genotypes, hypertension and overall survival in metastatic breast cancer patients treated with bevacizumab [Citation46]. They found that the VEGF-2578AA genotype was associated with superior survival and a higher incidence of hypertension [Citation46]. Sibertin-Blanc et al. also found VEGF-A c.*237C>T was associated with better survival and higher risk of bevacizumab-induced hypertension in metastatic colorectal cancer [Citation72]. These findings indicated that higher risk of hypertension could be a surrogate marker for treatment outcome.

Table 2. Potential effects of genetic polymorphism on hypertension induced by vascular endothelial growth factor inhibitors.

Genetic markers, such as EGLN3, EGF and WNK1, are more closely associated with bevacizumab-induced hypertension, while KDR was associated with fewer hypertensive events [Citation75]. WNK kinases regulate sodium-chloride co-transporters in distal renal tubules [Citation79]. Mutation of the WNK1 gene causes overexpression of co-transporters, leading to Gordon’s syndrome, a Mendelian disease with hypertension and hyperkalaemia [Citation80]. Polymorphisms of the WNK1 gene are also associated with hypertension [Citation81] and the treatment response to thiazide diuretics [Citation82]. In addition, the ECF gene encodes epidermal growth factors (EGFs). It is known that the EGF receptor may increase blood pressure through influencing glomerular arterioles and sodium reabsorption in the kidneys [Citation83]. Furthermore, both the EGLN3 and KDR genes are involved in the VEGF-A pathway. The KDR protein is a VEGF receptor. It was reported that polymorphisms of the KDR gene decrease VEGF-A’s binding ability to the KDR protein, increasing the risk of coronary artery disease [Citation84]. EGLN3 is a member of the Egg-laying Nine (EGLN) gene family, which regulates hypoxia-inducible transcription factors (HIFs) and cellular oxygen homeostasis [Citation85]. HIFs also increase several gene expressions related to angiogenesis, including VEGF [Citation85]. Thus, it is reasonable that these gene markers would play certain roles in the development or prediction of bevacizumab-induced hypertension.

Polymorphisms in VEGF ACG haplotypes −2578A>C, −460C>T and −405C>G are more closely associated with sunitinib-induced hypertension [Citation78]. Clinical genetic studies reported that ACG haplotypes are probably associated with ischemic heart disease and coronary artery disease [Citation86–88]. Therefore, ACG haplotypes may also contribute to sunitinib-induced hypertension through influencing the VEGF signal pathway. However, the biologic roles of these ACG haplotypes remain controversial. Some reports suggested that ACG haplotypes−2578A,−460C and 405G were associated with reduced levels of serum VEGF [Citation89,Citation90], while other reports found no association between serum VEGF levels and haplotype−2578A [Citation91]. Additionally, these genetic polymorphisms may also be differently distributed between different ethnic groups, leading to different risks of VEGF-associated cardiovascular toxicity. One Korean study showed that VEGF SNP-634 G/G was associated with a higher risk of sunitinib-induced hypertension [Citation77], while it was reported that polymorphisms of VEGF-634 were associated with serum VEGF levels and its production [Citation92,Citation93]. Another Korean study found that the genetic polymorphism, ABCG2 421C>A, increased the risk of sunitinib-related toxicity, such as thrombocytopenia, neutropenia and hand-foot syndrome [Citation76]. It was reported that the ABCG2 transporter, a multidrug transporter, can be inhibited by binding to sunitinib, and also decreases the extrusion of sunitinib [Citation94–96]. Polymorphisms of the ABCG2 gene are also associated with the function and amount of ABCG2 [Citation96,Citation97], leading to potential differences in inhibitory effects of sunitinib. In addition, a higher frequency of 421C>A was found in Asians [Citation98,Citation99]. Thus, ethnic differences in genetic polymorphisms might also contribute to different presentations of VEGF toxicity. However, some of these genetic findings provide conflicting evidence. VEGF-634 CC was associated with higher or lower bevacizumab-induced hypertension in different studies [Citation46,Citation73]. More studies are still needed to clarify the underlying mechanisms and correlations before these clinical and genetic findings can be applied in clinical practice.

In conclusion, VEGF inhibitors disturb the balance between vasodilation and vasoconstriction, undermine endothelial cell integrity, and interact with off-target pathways, increasing potential risks of hypertension, thromboembolisms, HF and arrhythmias. Current VEGF research has not completely elucidated the underlying mechanisms of long-term impacts on and genetic or ethnic predictors of cardiovascular toxicity. Future clinical and genetic studies are needed to resolve these issues.

Disclosure statement

The authors report no conflicts of interest.

References

  • Ferrara N, Adamis AP. Ten years of anti-vascular endothelial growth factor therapy. Nat Rev Drug Discov. 2016;15:385–403.
  • Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350:2335–2342.
  • Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007;356:115–124.
  • Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–390.
  • Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. JCO. 2010;28:1061–1068.
  • Rini BI, Escudier B, Tomczak P, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet (London, England). 2011;378:1931–1939.
  • Li W, Croce K, Steensma DP, et al. Vascular and metabolic implications of novel targeted cancer therapies: focus on kinase inhibitors. J Am Coll Cardiol. 2015;66:1160–1178.
  • Lankhorst S, Saleh L, Danser AJ, et al. Etiology of angiogenesis inhibition-related hypertension. Curr Opin Pharmacol. 2015;21:7–13.
  • Common Terminology Criteria for Adverse Events v4.0. National Cancer Institute; 2009.
  • Bellinger AM, Arteaga CL, Force T, et al. Cardio-oncology: how new targeted cancer therapies and precision medicine can inform cardiovascular discovery. Circulation. 2015;132:2248–2258.
  • Moslehi JJ. Cardiovascular toxic effects of targeted cancer therapies. N Engl J Med. 2016;375:1457–1467.
  • Eremina V, Jefferson JA, Kowalewska J, et al. VEGF inhibition and renal thrombotic microangiopathy. N Engl J Med. 2008;358:1129–1136.
  • Patel TV, Morgan JA, Demetri GD, et al. A preeclampsia-like syndrome characterized by reversible hypertension and proteinuria induced by the multitargeted kinase inhibitors sunitinib and sorafenib. J Natl Cancer Inst. 2008;100:282–284.
  • Vigneau C, Lorcy N, Dolley-Hitze T, et al. All anti-vascular endothelial growth factor drugs can induce ‘pre-eclampsia-like syndrome’: a RARe study. Nephrol Dial Transplant. 2014;29:325–332.
  • Powe CE, Levine RJ, Karumanchi SA. Preeclampsia, a disease of the maternal endothelium: the role of antiangiogenic factors and implications for later cardiovascular disease. Circulation. 2011;123:2856–2869.
  • Patten IS, Rana S, Shahul S, et al. Cardiac angiogenic imbalance leads to peripartum cardiomyopathy. Nature. 2012;485:333–338.
  • Overexpression of vascular endothelial growth factor 165 (VEGF165) protects cardiomyocytes against doxorubicin-induced apoptosis. J Chemother. 2010;22:402–406.
  • Räsänen M, Degerman J, Nissinen TA, et al. VEGF-B gene therapy inhibits doxorubicin-induced cardiotoxicity by endothelial protection. Proc Natl Acad Sci USA. 2016;113:13144–13149.
  • Pepe M, Mamdani M, Zentilin L, et al. Intramyocardial VEGF-B167 gene delivery delays the progression towards congestive failure in dogs with pacing-induced dilated cardiomyopathy. Circ Res. 2010;106:1893–1903.
  • Zhao T, Zhao W, Meng W, et al. VEGF-C/VEGFR-3 pathway promotes myocyte hypertrophy and survival in the infarcted myocardium. Am J Transl Res. 2015;7:697–709.
  • Di Lisi D, Madonna R, Zito C, et al. Anticancer therapy-induced vascular toxicity: VEGF inhibition and beyond. Int J Cardiol. 2017;227:11–17.
  • Ghatalia P, Morgan CJ, Je Y, et al. Congestive heart failure with vascular endothelial growth factor receptor tyrosine kinase inhibitors. Critic Rev Oncol/Hematol. 2015;94:228–237.
  • Force T, Krause DS, Van Etten RA. Molecular mechanisms of cardiotoxicity of tyrosine kinase inhibition. Nat Rev Cancer. 2007;7:332–344.
  • Cheng H, Force T. Why do kinase inhibitors cause cardiotoxicity and what can be done about it? Prog Cardiovasc Dis. 2010;53:114–120.
  • Sanguinetti MC, Mitcheson JS. Predicting drug-hERG channel interactions that cause acquired long QT syndrome. Trends Pharmacol Sci. 2005;26:119–124.
  • Katchman AN, Koerner J, Tosaka T, et al. Comparative evaluation of HERG currents and QT intervals following challenge with suspected torsadogenic and nontorsadogenic drugs. J Pharmacol Exp Ther. 2006;316:1098–1106.
  • Yang T, Roden DM. Extracellular potassium modulation of drug block of IKr. Implications for torsade de pointes and reverse use-dependence. Circulation. 1996;93:407–411.
  • Kupershmidt S, Yang IC, Hayashi K, et al. The IKr drug response is modulated by KCR1 in transfected cardiac and noncardiac cell lines. FASEB J. 2003;17:2263–2265.
  • Shah RR, Morganroth J, Shah DR. Cardiovascular safety of tyrosine kinase inhibitors: with a special focus on cardiac repolarisation (QT interval). Drug Saf. 2013;36:295–316.
  • Kramer B, Brill M, Bruhn A, et al. Relationship between the degree of coronary artery disease and of left ventricular function and the duration of the QT-interval in ECG. Eur Heart J. 1986;7:14–24.
  • Hart G. Cellular electrophysiology in cardiac hypertrophy and failure. Cardiovasc Res. 1994;28:933–946.
  • Davey PP, Barlow C, Hart G. Prolongation of the QT interval in heart failure occurs at low but not at high heart rates. Clin Sci. 2000;98:603–610.
  • Harada M, Tadevosyan A, Qi X, et al. Atrial fibrillation activates AMP-dependent protein kinase and its regulation of cellular calcium handling: potential role in metabolic adaptation and prevention of progression. J Am Coll Cardiol. 2015;66:47–58.
  • An MM, Zou Z, Shen H, et al. Incidence and risk of significantly raised blood pressure in cancer patients treated with bevacizumab: an updated meta-analysis. Eur J Clin Pharmacol. 2010;66:813–821.
  • Zhu X, Stergiopoulos K, Wu S. Risk of hypertension and renal dysfunction with an angiogenesis inhibitor sunitinib: systematic review and meta-analysis. Acta Oncologica (Stockholm, Sweden). 2009;48:9–17.
  • Wu S, Chen JJ, Kudelka A, et al. Incidence and risk of hypertension with sorafenib in patients with cancer: a systematic review and meta-analysis. Lancet Oncol. 2008;9:117–123.
  • Qi WX, Lin F, Sun YJ, et al. Incidence and risk of hypertension with pazopanib in patients with cancer: a meta-analysis. Cancer Chemother Pharmacol. 2013;71:431–439.
  • Schlumberger M, Tahara M, Wirth LJ, et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. N Engl J Med. 2015;372:621–630.
  • Fuchs CS, Tomasek J, Yong CJ, et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet (London, England). 2014;383:31–39.
  • Van Cutsem E, Tabernero J, Lakomy R, et al. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. JCO. 2012;30:3499–3506.
  • Donskov F, Michaelson MD, Puzanov I, et al. Sunitinib-associated hypertension and neutropenia as efficacy biomarkers in metastatic renal cell carcinoma patients. Br J Cancer. 2015;113:1571–1580.
  • Rini BI, Cohen DP, Lu DR, et al. Hypertension as a biomarker of efficacy in patients with metastatic renal cell carcinoma treated with sunitinib. J Natl Cancer Inst. 2011;103:763–773.
  • Estfan B, Byrne M, Kim R. Sorafenib in advanced hepatocellular carcinoma: hypertension as a potential surrogate marker for efficacy. Am J Clin Oncol. 2013;36:319–324.
  • Mir O, Coriat R, Cabanes L, et al. An observational study of bevacizumab-induced hypertension as a clinical biomarker of antitumor activity. The Oncologist. 2011;16:1325–1332.
  • Dahlberg SE, Sandler AB, Brahmer JR, et al. Clinical course of advanced non-small-cell lung cancer patients experiencing hypertension during treatment with bevacizumab in combination with carboplatin and paclitaxel on ECOG 4599. JCO. 2010;28:949–954.
  • Schneider BP, Wang M, Radovich M, et al. Association of vascular endothelial growth factor and vascular endothelial growth factor receptor-2 genetic polymorphisms with outcome in a trial of paclitaxel compared with paclitaxel plus bevacizumab in advanced breast cancer: ECOG 2100. JCO. 2008;26:4672–4678.
  • Ryanne Wu R, Lindenberg PA, Slack R, et al. Evaluation of hypertension as a marker of bevacizumab efficacy. J Gastrointest Cancer. 2009;40:101–108.
  • Scartozzi M, Galizia E, Chiorrini S, et al. Arterial hypertension correlates with clinical outcome in colorectal cancer patients treated with first-line bevacizumab. Ann Oncol. 2009;20:227–230.
  • Hurwitz HI, Douglas PS, Middleton JP, et al. Analysis of early hypertension and clinical outcome with bevacizumab: results from seven phase III studies. The Oncologist. 2013;18:273–280.
  • Duffaud F, Sleijfer S, Litiere S, et al. Hypertension (HTN) as a potential biomarker of efficacy in pazopanib-treated patients with advanced non-adipocytic soft tissue sarcoma. A retrospective study based on european organisation for research and treatment of cancer (EORTC) 62043 and 62072 trials. Eur J Cancer (Oxford, England: 1990). 2015;51:2615–2623.
  • Scappaticci FA, Skillings JR, Holden SN, et al. Arterial thromboembolic events in patients with metastatic carcinoma treated with chemotherapy and bevacizumab. J Natl Cancer Inst. 2007;99:1232–1239.
  • Schutz FA, Je Y, Azzi GR, et al. Bevacizumab increases the risk of arterial ischemia: a large study in cancer patients with a focus on different subgroup outcomes. Ann Oncol. 2011;22:1404–1412.
  • Ranpura V, Hapani S, Chuang J, et al. Risk of cardiac ischemia and arterial thromboembolic events with the angiogenesis inhibitor bevacizumab in cancer patients: a meta-analysis of randomized controlled trials. Acta Oncologica (Stockholm, Sweden). 2010;49:287–297.
  • Choueiri TK, Schutz FA, Je Y, et al. Risk of arterial thromboembolic events with sunitinib and sorafenib: a systematic review and meta-analysis of clinical trials. JCO. 2010;28:2280–2285.
  • Nalluri SR, Chu D, Keresztes R, et al. Risk of venous thromboembolism with the angiogenesis inhibitor bevacizumab in cancer patients: a meta-analysis. JAMA. 2008;300:2277–2285.
  • Hurwitz HI, Saltz LB, Van Cutsem E, et al. Venous thromboembolic events with chemotherapy plus bevacizumab: a pooled analysis of patients in randomized phase II and III studies. JCO. 2011;29:1757–1764.
  • Sonpavde G, Je Y, Schutz F, et al. Venous thromboembolic events with vascular endothelial growth factor receptor tyrosine kinase inhibitors: a systematic review and meta-analysis of randomized clinical trials. Crit Rev Oncol Hematol. 2013;87:80–89.
  • Choueiri TK, Mayer EL, Je Y, et al. Congestive heart failure risk in patients with breast cancer treated with bevacizumab. JCO. 2011;29:632–638.
  • Chu TF, Rupnick MA, Kerkela R, et al. Cardiotoxicity associated with tyrosine kinase inhibitor sunitinib. Lancet (London, England). 2007;370:2011–2019.
  • Shamloo BK, Chhabra P, Freedman AN, et al. Novel adverse events of bevacizumab in the US FDA adverse event reporting system database: a disproportionality analysis. Drug Safety. 2012;35:507–518.
  • Ghatalia P, Je Y, Kaymakcalan MD, et al. QTc interval prolongation with vascular endothelial growth factor receptor tyrosine kinase inhibitors. Br J Cancer. 2015;112:296–305.
  • Bello CL, Mulay M, Huang X, et al. Electrocardiographic characterization of the QTc interval in patients with advanced solid tumors: pharmacokinetic- pharmacodynamic evaluation of sunitinib. Clin Cancer Res. 2009;15:7045–7052.
  • Polite BN, Sing A, Sargent DJ, et al. Exploring racial differences in outcome and treatment for metastatic colorectal cancer: results from a large prospective observational cohort study (BRiTE). Cancer. 2012;118:1083–1090.
  • Hajjar I, Kotchen TA. Trends in prevalence, awareness, treatment, and control of hypertension in the United States, 1988-2000. JAMA. 2003;290:199–206.
  • Selassie A, Wagner CS, Laken ML, et al. Progression is accelerated from prehypertension to hypertension in blacks. Hypertension (Dallas, TX: 1979). 2011;58:579–587.
  • Carson AP, Howard G, Burke GL, et al. Ethnic differences in hypertension incidence among middle-aged and older adults: the multi-ethnic study of atherosclerosis. Hypertension (Dallas, TX: 1979). 2011;57:1101–1107.
  • Chen Z, Zhong B, Lun X, et al. Specific safety profile of bevacizumab in asian patients with advanced NSCLC: a meta-analysis. Medicine. 2015;94:e975.
  • Ueda T, Uemura H, Tomita Y, et al. Efficacy and safety of axitinib versus sorafenib in metastatic renal cell carcinoma: subgroup analysis of Japanese patients from the global randomized Phase 3 AXIS trial. Japanese J Clin Oncol. 2013;43:616–628.
  • Lee SH, Bang YJ, Mainwaring P, et al. Sunitinib in metastatic renal cell carcinoma: an ethnic Asian subpopulation analysis for safety and efficacy. Asia-Pac J Clin Oncol. 2014;10:237–245.
  • Chen Y, Suzuki A, Tortorici MA, et al. Axitinib plasma pharmacokinetics and ethnic differences. Invest New Drugs. 2015;33:521–532.
  • Schneider BP, Li L, Shen F, et al. Genetic variant predicts bevacizumab-induced hypertension in ECOG-5103 and ECOG-2100. Br J Cancer. 2014;111:1241–1248.
  • Sibertin-Blanc C, Mancini J, Fabre A, et al. Vascular endothelial growth factor a c.*237C > T polymorphism is associated with bevacizumab efficacy and related hypertension in metastatic colorectal cancer. Digest Liver Dis. 2015;47:331–337.
  • Etienne-Grimaldi MC, Formento P, Degeorges A, et al. Prospective analysis of the impact of VEGF-A gene polymorphisms on the pharmacodynamics of bevacizumab-based therapy in metastatic breast cancer patients. Br J Clin Pharmacol. 2011;71:921–928.
  • Morita S, Uehara K, Nakayama G, et al. Association between bevacizumab-related hypertension and vascular endothelial growth factor (VEGF) gene polymorphisms in Japanese patients with metastatic colorectal cancer. Cancer Chemother Pharmacol. 2013;71:405–411.
  • Lambrechts D, Moisse M, Delmar P, et al. Genetic markers of bevacizumab-induced hypertension. Angiogenesis. 2014;17:685–694.
  • Kim HR, Park HS, Kwon WS, et al. Pharmacogenetic determinants associated with sunitinib-induced toxicity and ethnic difference in Korean metastatic renal cell carcinoma patients. Cancer Chemother Pharmacol. 2013;72:825–835.
  • Kim JJVS, Elson P, Rini BI, Ganapathi MK, Ganapathi R. VEGF single nucleotide polymorphisms (SNPs) and correlation to sunitinib-induced hypertension (HTN) in metastatic renal cell carcinoma (mRCC) patients (pts). J Clin Oncol. 2009;27(15S Suppl):5005.
  • Eechoute K, van der Veldt AA, Oosting S, et al. Polymorphisms in endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF) predict sunitinib-induced hypertension. Clin Pharmacol Ther. 2012;92:503–510.
  • Huang CL, Kuo E. Mechanisms of disease: WNK-ing at the mechanism of salt-sensitive hypertension. Nat Clin Pract Nephrol. 2007;3:623–630.
  • Wilson FH, Disse-Nicodeme S, Choate KA, et al. Human hypertension caused by mutations in WNK kinases. Science (New York, NY). 2001;293:1107–1112.
  • Newhouse S, Farrall M, Wallace C, Hoti M, Burke B, Howard P, etet al. Polymorphisms in the WNK1 gene are associated with blood pressure variation and urinary potassium excretion. PloS One. 2009;4:e5003.
  • Turner ST, Schwartz GL, Chapman AB, et al. WNK1 kinase polymorphism and blood pressure response to a thiazide diuretic. Hypertension (Dallas, TX: 1979). 2005;46:758–765.
  • Staruschenko A, Palygin O, Ilatovskaya DV, et al. Epidermal growth factors in the kidney and relationship to hypertension. Am J Physiol Renal Physiol. 2013;305:F12–F20.
  • Wang Y, Zheng Y, Zhang W, et al. Polymorphisms of KDRGene are associated with coronary heart disease. J Am College Cardiol. 2007;50:760–767.
  • Kant R, Bali A, Singh N, et al. Prolyl 4 hydroxylase: a critical target in the pathophysiology of diseases. Korean J Physiol Pharmacol. 2013;17:111–120.
  • Chen Y, Dawes PT, Packham JC, et al. Interaction between smoking and polymorphism in the promoter region of the vegfa gene is associated with ischemic heart disease and myocardial infarction in rheumatoid arthritis. J Rheumatol. 2011;38:802–809.
  • Biselli PM, Guerzoni AR, de Godoy MF, et al. Vascular endothelial growth factor genetic variability and coronary artery disease in Brazilian population. Heart Vessels. 2008;23:371–375.
  • Howell WM, Ali S, Rose-Zerilli MJ, et al. VEGF polymorphisms and severity of atherosclerosis. J Med Genet. 2005;42:485–490.
  • Shahbazi M, Fryer AA, Pravica V, et al. Vascular endothelial growth factor gene polymorphisms are associated with acute renal allograft rejection. J Am Soc Nephrol. 2002;13:260–264.
  • Steffensen KD, Waldstrøm M, Brandslund I, et al. The relationship of VEGF polymorphisms with serum VEGF levels and progression-free survival in patients with epithelial ovarian cancer. Gynecol Oncol. 2010;117:109–116.
  • Prior SJ, Hagberg JM, Paton CM, Douglass LW, Brown MD, McLenithan JC, etet al. DNA sequence variation in the promoter region of the VEGF gene impacts VEGF gene expression and maximal oxygen consumption. Am J Physiol. 2006;290:H1848–H1H55.
  • Oh SY, Kwon HC, Kim SH, et al. The relationship of vascular endothelial growth factor gene polymorphisms and clinical outcome in advanced gastric cancer patients treated with FOLFOX: VEGF polymorphism in gastric cancer. BMC Cancer. 2013;13:43.
  • Awata T, Inoue K, Kurihara S, et al. A common polymorphism in the 5’-untranslated region of the VEGF gene is associated with diabetic retinopathy in type 2 diabetes. Diabetes. 2002;51:1635–1639.
  • Ozvegy-Laczka C, Cserepes J, Elkind NB, et al. Tyrosine kinase inhibitor resistance in cancer: role of ABC multidrug transporters. Drug Resist Updates. 2005;8:15–26.
  • Ozvegy-Laczka C, Hegedus T, Varady G, et al. High-affinity interaction of tyrosine kinase inhibitors with the ABCG2 multidrug transporter. Mol Pharmacol. 2004;65:1485–1495.
  • Shukla S, Robey RW, Bates SE, et al. Sunitinib (Sutent, SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ATP-binding cassette (ABC) transporters P-glycoprotein (ABCB1) and ABCG2. Drug Metabol Dispos. 2009;37:359–365.
  • Cascorbi I. Role of pharmacogenetics of ATP-binding cassette transporters in the pharmacokinetics of drugs. Pharmacol Ther. 2006;112:457–473.
  • Kim KA, Joo HJ, Park JY. ABCG2 polymorphisms, 34G > A and 421C > A in a Korean population: analysis and a comprehensive comparison with other populations. J Clin Pharm Ther. 2010;35:705–712.
  • Keskitalo JE, Pasanen MK, Neuvonen PJ, et al. Different effects of the ABCG2 c.421C > A SNP on the pharmacokinetics of fluvastatin, pravastatin and simvastatin. Pharmacogenomics. 2009;10:1617–1624.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.