- Research
- Open access
- Published:
Pharmacogenomic insights into atorvastatin and rosuvastatin adverse effects: a prospective observational study in the UAE’s multiethnic population
Human Genomics volume 19, Article number: 44 (2025)
Abstract
Background
Statins are essential for managing cardiovascular disease (CVD), but adverse effects often lead to treatment discontinuation and non-adherence, underscoring the need for personalized approaches. This study aimed to evaluate the influence of pharmacogenomic (PGx) variants and demographic factors on statin-associated adverse effects in a multiethnic cohort from the United Arab Emirates (UAE).
Methods
This sub-analysis of the EmHeart Study included 675 patients using rosuvastatin or atorvastatin. Patients were genotyped for SLCO1B1 and ABCG2 actionable variants using real-time PCR. Data on demographics, comorbidities, and statin use were extracted from electronic health records. Adverse events, including statin-associated muscle symptoms (SAMS) and liver enzyme elevation, were tracked over 12 months. Associations were analyzed using chi-square tests and logistic regression.
Results
Rosuvastatin users carrying the ABCG2 rs2231142 variant had a threefold increased risk of liver enzyme elevation, particularly among East Asian patients (P < 0.005). Atorvastatin users with the SLCO1B1 rs4149056 variant exhibited a twofold increased risk of SAMS, with higher rates observed in females and Arabs (P < 0.05). The combination of rosuvastatin with ezetimibe further exacerbated risks of SAMS and liver enzyme elevation.
Conclusion
This study highlights the importance of genetic testing and demographic factors, such as ethnicity and gender, in tailoring statin therapy to minimize adverse effects. Despite extensive research on PGx-guided statin prescribing, clinical implementation remains limited. Integrating PGx testing into routine practice and enhancing physician awareness of genetic and demographic risk factors can improve the safety, efficacy, and adherence of lipid-lowering therapies in diverse populations.
Introduction
Cardiovascular diseases (CVDs) are major and leading global and regional causes of morbidity and mortality. The estimated number of deaths caused by CVDs globally is approximately 17.9 million each year, representing ~ 32% of all global deaths [1]. In the United Arab Emirates (UAE), a country noted for its multiethnic and highly heterogeneous population, with Emiratis constituting only about 11% of the total [2]., CVDs are a significant health concern [3]. These diseases are responsible for approximately 40% of all deaths in the country, highlighting the significant healthcare challenges posed by demographic diversity [4]. The high prevalence of cardiovascular diseases has placed considerable economic and social strains on the country [3].
Statins (3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors) are the most prescribed medications for managing and preventing CVDs by lowering Low-density lipoprotein cholesterol (LDL-C), both in the UAE and globally [5]. The 2019 ESC/EAS guidelines strongly emphasized aggressively lowering LDL-C to reduce the risk of atherosclerotic cardiovascular disease (ASCVD) [6]. Lowering LDL is not merely a treatment goal but a fundamental aspect of the comprehensive management approach that targets multiple risk factors, including lipid levels, blood sugar, blood pressure, weight, and lifestyle habits [7]. Adherence to LDL-lowering therapies, such as statins, plays a crucial role in achieving the objectives of this integrated strategy, which aims to reduce cardiovascular events through coordinated interventions addressing these risk factors [8]. A study conducted in the UAE on the use of statins among 3,066 Emirati patients hospitalized with acute coronary syndrome (ACS) found that, despite cardiologists’ recommendations for statin usage before or after experiencing ACS, only 58.1% of patients were on statins before or within 90 days following the incident [9]. These statistics underscore the need for broader use of statins among this demographic and highlight a significant gap between current prescribing behaviors and the established guidelines for Emirati patients [9].
Although statins are generally safe and well tolerated, they can sometimes be associated with side events of variable severity. The most reported side event is statin-associated muscle symptoms (SAMS), affecting about 27% of statin users [10]. SAMS can lead to noncompliance and premature discontinuation of the medication [11]. Hence, it is crucial to identify patients with true SAMS to ensure optimal statin use and to study the confounding factors affecting its incidence [12].
In this study, we utilized the well-established evidence linking pharmacogenomic variants in SLCO1B1, which encodes a solute carrier transport molecule essential for liver uptake of statins. Variants that have significant evidence of associations with statin pharmacokinetics (rs4149056 (521T > C), rs2306283 (388 A > G)) were selected [13, 14]. Additionally, we considered variant rs2231142 (421 C > A) in the ATP-binding cassette sub-family G member 2 (ABCG2) for its impact on rosuvastatin pharmacokinetics and the development of side effects [15], which led to the latest CPIC guidelines [16]. This manuscript reports an observational real-world study that explored the prevalence of SAMS within our cardiovascular disease cohort (EmHeart Study). The full description of the study can be found elsewhere [17]. It includes an ethnically diverse and heterogeneous patient population with varied medical histories, ethnicities, and nationalities. By analyzing data from such a population, we aimed to comprehensively understand these side events and their potential risk factors, including age, gender, BMI, ethnicity, genetic factors, combination therapy, duration of treatment, and statin intensity. Although genetic and other factors have been extensively studied in simvastatin users [18, 19], they remain less explored in those taking atorvastatin and rosuvastatin, highlighting the significance of this study. Identifying these associations can contribute to developing a more personalized approach to statin therapy. This approach would balance the established benefits of statin use in CVD prevention with minimizing potential adverse events for individual patients. Ultimately, this research seeks to optimize the risk-benefit profile of statin therapy, ensuring a safer and more effective CVD prevention strategy at the global multiethnic levels.
Methods
Patient characteristics
Eligible participants were adult patients aged 18 years or older who had been prescribed either atorvastatin or rosuvastatin, agreed to participate in a 12-month follow-up study, and signed an informed consent form. Exclusion criteria for the study were as follows: patients who were pregnant or breastfeeding; those who planned to use the study drug for less than seven days; individuals with hepatic insufficiency, defined as alanine aminotransferase (ALT) or aspartate aminotransferase (AST) levels more than twice the upper limit of the normal range, or those with liver cirrhosis; patients with renal insufficiency, where serum creatinine was more than 1.5 times the upper limit of the normal range, or serum creatinine exceeded 2.5 mg/dl in the last seven days, or those with severe renal function impairment requiring dialysis; subjects with an active tumor, a recent cancer diagnosis, or those undergoing chemotherapy treatment; patients with hematologic issues, such as a white blood cell count less than 2 x \(\:{10}^{9}\)/L, or platelet counts under 100 x\(\:{10}^{9}\)/L and patients suffering from sepsis. We initially enrolled 712 patients. To minimize confounding, 20 atorvastatin users and 17 rosuvastatin users were excluded due to underlying musculoskeletal disorders, hypothyroidism, or chronic pain syndromes. The final cohort included 675 patients who were eligible for the 12-month follow-up analysis.
All the recruited patients have been genotyped for SLCO1B1 rs4149056 (521T > C), SLCO1B1 rs2306283 (388 A > G), and ABCG2 rs2231142 (421 C > A) variants as part of the EmHeart Study.
Data collection, ethical approvals, and Follow-up
The study was conducted per the declaration of Helsinki and received approval from The Abu Dhabi Health Research and Technology Ethical Committee, reference numbers (DOH/CVDC/2020/1187), (DOH/CVDC/2021/1519), (DOH/CVDC/2022/1458), (MCME.CR.213.MAIN.2021), and (SNA/FA/2020-14). All the baseline and follow-up data were recorded using the Castor-EDC software (Netherlands) (www.castoredc.com). Castor EDC is a web-based platform designed to capture data entered via an electronic Case Report Form (eCRF) at clinical sites, which is then uploaded into a centralized database. Access to the software was limited to authorized users who log in using their credentials, ensuring the confidentiality of study participant data. Additionally, this tool simplified the process of exporting and analyzing the data, enhancing the efficiency and accuracy of research analysis.
Clinical information was gathered from electronic medical records (EMR); data collected included the reasons for prescribing statins, such as cardiovascular disease, and comorbid conditions that could influence the risk of SAMS, like thyroid disorders, were identified using the International Classification of Diseases (ICD). Medication details, such as levothyroxine prescriptions, helped confirm diagnoses like hypothyroidism. Demographics like age, gender, height, weight, smoking status, alcohol consumption, ethnicity, concomitant medications, family history, and comorbidities. Besides laboratory results, prescriptions, and noted drug allergies. Follow-ups were made by reviewing the medical records and conducting phone calls to confirm the occurrence of SAMS within one year of recruitment. Outcomes include the occurrence of SAMS while using atorvastatin or rosuvastatin (the only two statins prescribed in UAE).
Sample collection and DNA isolation
Peripheral blood samples (3 mL) were collected in EDTA-containing vacutainer tubes (BD Inc., UK) and stored at -20 °C until further analysis. Genomic DNA isolation from whole blood was achieved using the FlexiGene® DNA kit (Qiagen, Germany) or the QIAamp® DNA kit (Qiagen, Germany), following the manufacturer’s instructions. DNA quality and quantity were evaluated using a Nanodrop One Spectrophotometer (Thermo Fisher Scientific, USA).
Genotyping of genetic variants
Genotyping for three specific genetic variants was performed using TaqMan® SNP assays for SLCO1B1 rs4149056 (521T > C), SLCO1B1 rs2306283 (388 A > G), ABCG2 rs2231142 (421 C > A) (catalogue numbers C__8898463_10, C_30633988_10, and C_15854163_70, respectively) in conjunction with TaqMan SNP master mix on a Realtime-QuantStudio 7 Flex PCR machine (Applied Biosystems, ThermoFisher Scientific). The resulting genotypes were validated using TaqMan Genotyper software version 1.6.0 (Applied Biosystems, ThermoFisher Scientific). The genotypes of a representative sample were confirmed by Sanger sequencing, which showed 100% concordance.
Statin Associated Muscle Symptoms (SAMS)
The diagnosis of statin-induced myalgia and other milder forms of statin-associated muscle symptoms (SAMS) relies predominantly on clinical criteria due to the absence of specific diagnostic tests [20]. These criteria recognize symptoms such as muscle pain, cramps, and weakness as common indicators of SAMS. Symptoms typically affect large muscle groups like the thighs, buttocks, back, and shoulders, presenting bilaterally and early after initiating or increasing the statin dosage. Contrarily, muscle cramps often occur unilaterally and might affect smaller muscles in the hands and feet, with symptoms commonly emerging about one month after starting treatment. We applied the SAMS-CI tool [21], which gives a score indicating the probability of the muscle pain symptoms being induced by statins. However, in this study, we included all patients who suffered from SAMS regardless of the values of these scores. We have evaluated the EMR for other medical conditions that could cause muscle pain, such as hypothyroidism, vitamin D deficiency, or underlying muscle disease. Moreover, we asked the patients via phone calls if the pain was synchronous with excessive physical work, changes in exercise patterns, concurrent illnesses, hypothyroidism, or underlying muscle diseases. Those patients were excluded from the SAMS diagnosis.
Transaminase elevation
Using medical records, we checked the laboratory results for alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels. The study’s clinical laboratory data focused on three key measures: AST, ALT, and creatinine, which were adjusted for age, sex, race, and creatinine level to control for potential confounders affecting liver enzyme levels. For analytical purposes, the value closest to the genotyping date for each variable was used. Transaminase elevation was defined as ALT levels greater than three times the upper limit of normal, a threshold that helps identify significant hepatocellular injury potentially related to statin use. Physicians specifically noted cases where elevated liver enzymes were attributed probably to statin use, leading to the cessation of statin therapy to monitor enzyme levels, thus confirming the clinical pathway followed in response to suspected adverse drug reactions.
Statistical analysis
The statistical analysis was designed to investigate the associations between statin use and the incidence of adverse effects, specifically SAMS and transaminase elevation. Initially, univariate analyses, including Chi-square tests, explored the frequencies and comparison of adverse events between patients treated with atorvastatin and rosuvastatin. Then we investigated factors affecting the occurrence of adverse events including genetic variations. This step helped identify potential variables of interest based on the observed distributions in the cohort.
Subsequently, multivariate logistic regression models were employed to elucidate further the relationships between patient demographics, genetic profiles, and the occurrence of adverse events. These models adjusted for multiple confounding factors such as age, sex, BMI, and ethnicity to ensure the robustness of the results. Variables with a p-value < 0.1 in the univariate analysis were entered into the multivariate logistic regression model to identify independent predictors of adverse events. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to quantify the impact of various predictors, including the intensity of statin therapy and the presence of pharmacogenomic variants like SLCO1B1 rs4149056 and ABCG2 rs2231142. Statistical significance was established at a p-value of less than 0.05, using SPSS (version 29.0, IBM Corporation, Armonk, NY, USA) for all analyses.
Results
Our cohort included 675 patients treated with statins (352 with atorvastatin and 323 with rosuvastatin) as monotherapy or in combination with ezetimibe (N = 264). The average age of our cohort was 52.43 years (± 11.52). Table 1 lists the demographics of patients and indications for statins.
Genotyping results were analyzed, and Table 2 lists the minor allele frequencies (MAF) of the studied variants among the different ethnicities of the study cohort.
The frequencies of adverse events were compared between atorvastatin and rosuvastatin users. Table 3 lists these frequencies and the p-value of the Chi-square test, illustrating the significance of differences between the observed frequencies. A significant difference was detected in the frequencies of gastrointestinal (GI) symptoms and elevation in liver enzymes.
SAMS were the most frequent side effects prevalent in our cohort affecting around 30% of statins users. Although the overall incidence of SAMS did not significantly differ between atorvastatin and rosuvastatin users, we analyzed them separately due to differences in pharmacogenomic associations and co-medication profiles (e.g., ezetimibe use), which could affect the risk of adverse effects in a drug-specific manner. The percentage of patients and odds ratios of SAMS in each group, with 95% confidence intervals and p-values, are listed in Table 4 for the atorvastatin group and Table 5 for the rosuvastatin group. Gender, BMI, Arab ethnicity, and carrying the minor allele at SLCO1B1:rs4149056 were all significant variables in the atorvastatin group. In comparison, a similar analysis yielded different significant variables in the rosuvastatin group (Table 5). The combination of ezetimibe therapy and ABCG2 variant was significant and nearly significant in this group, respectively.
Thereafter, regression analysis revealed that SLCO1B1 rs4149056 was the most significant predictor of SAMS among atorvastatin users as shown in Table 6. In contrast, in the rosuvastatin group, ezetimibe co-therapy emerged as a significant contributor while the ABCG2 rs2231142 variant showed a trend toward significance (Table 7). Elevated transaminase levels were significantly associated with the use of combination ezetimibe therapy and the ABCG2 variant in rosuvastatin users, as shown in Table 8. These findings were further supported by regression analysis (Table 9).
Finally, we stratified the incidence of transaminase elevation according to ethnicity, hypothesizing that some side effects may occur more frequently in some ethnic groups. East Asians demonstrated a higher susceptibility to these adverse events, with 36.30% of the liver enzyme elevation group being East Asian, of whom 73.7% were carriers of the ABCG2 rs2231142 variant. Additionally, in the SAMS group, 52% of participants were Arab (25% of whom carried the variant), and 21.80% were East Asian, with 51.85% carrying the variant. The frequencies of elevated liver enzymes and SAMS are plotted in different ethnic groups (Fig. 1). The percentage of ABCG2: rs2231142 (421 C > A) was further illustrated on the same plot to illustrate the positive trend between the carrier status of this variant and ethnic group.
Discussion
This observational real-time cohort study provides valuable insights into the comparative safety profiles of atorvastatin and rosuvastatin in a multiethnic population. It focuses on statin-associated muscle symptoms (SAMS), the most reported adverse event, and transaminase elevation, surprisingly more frequently observed among rosuvastatin users in our cohort. The study highlights differences in these adverse events across a multi-ethnic heterogeneous population. It evaluates confounding factors such as age, gender, obesity, ethnicity, genetic variants, duration of statin therapy, and combination therapies.
The structural differences between atorvastatin and rosuvastatin significantly impact their pharmacological properties [22]. Our results indicate that gastrointestinal symptoms are significantly more common in rosuvastatin patients than atorvastatin patients. Specifically, 10.2% of rosuvastatin users experienced gastrointestinal symptoms compared to 4.5% of atorvastatin users (P = 0.0035). The administration of rosuvastatin in mice leads to significant alterations in gut microbiota composition, bile acid metabolism, and local gene expression profiles related to inflammation and gut homeostasis [23]. Another study showed that rosuvastatin has a limited effect on human gut microbiome composition [24]. Roy and colleagues compared high-dose atorvastatin (80 mg/day) and rosuvastatin (40 mg/day) among post-percutaneous coronary intervention (PCI) patients, focusing on their effects on gastroesophageal reflux disease (GERD) and gastritis. It was found that GERD and gastritis occurred in 2.18% of patients taking atorvastatin versus 4.83% in those taking rosuvastatin during a three-month follow-up [25]. Our data revealed that statin therapy led to a 10.51% increased risk of developing diabetes, with a 12.7% increased risk in rosuvastatin users, and 8.5% in the case of atorvastatin (P = 0.0730).
Additionally, elevated liver enzymes were observed in 13.6% of rosuvastatin users compared to 3.4% of atorvastatin users (P < 0.0001). This rate of elevated liver enzymes observed in rosuvastatin users in our study is notably higher than typical rates reported in the literature, which generally consider clinically significant transaminase increases with statins to be rare. This finding suggests that genetic predispositions or demographic factors may elevate the risk in certain populations. These results are contrary to those of a significant drug-level network meta-analysis of 165,534 individuals, which indicated that atorvastatin (OR, 2.55; 95% CI 1.71–3.74) was more likely to cause transaminase elevation than rosuvastatin (OR, 1.59; 95%CI, 1.02–2.50) [26]. The discrepancy may be due to the multi-ethnic composition of our cohort, particularly since most of the transaminase elevation cases were linked to East Asians carrying the ABCG2 rs2231142 minor allele. So rosuvastatin might still be safer for specific ethnicities regarding transaminase elevation and atorvastatin might be safer for East Asians. This was discussed in our previous whole exome analysis for an Emirati cohort, and we stated that the integration of population-specific pharmacogenomic data into medical decisions to optimize drug efficacy while minimizing adverse effects is an appealing approach in personalized medicine [27]. Additionally, the smaller sample size of our study compared to the meta-analysis could have influenced these findings. Moreover, transaminase elevation in statin users is influenced by multiple factors [28]. Although our study included comprehensive medical reviews, it faced limitations in fully assessing all potential contributors to transaminase elevations. Specifically, we were unable to evaluate the impact of alcohol consumption, over-the-counter medications (e.g., acetaminophen), and herbal products on these laboratory parameters.
Our analysis detected no significant association between SAMS and transaminase elevation across different age groups. Although randomized controlled trials (RCTs) assessing the safety of rosuvastatin remain scarce. The JUPITER trial, which involved 5,695 participants aged over 70 years at enrolment, reported no specific safety concerns for this age group compared to younger individuals [29]. Another recent study showed that the relationship between cholesterol levels and cardiovascular mortality is more significant in younger individuals compared to older ones [30].
Our study specifically examined the impact of combining ezetimibe with rosuvastatin among 323 users and found significant effects on liver enzyme levels. Notably, ethnicity and the use of ezetimibe were prominent factors. The combination of rosuvastatin and ezetimibe showed higher odds of transaminase elevation when compared to statin monotherapy. A recent study by González-Iglesias et al. found that ABCG2 polymorphisms had a greater impact on rosuvastatin levels than SLCO1B1 variants, supporting our findings on ABCG2 rs2231142. However, they also reported that ezetimibe co-administration lowered rosuvastatin levels, contrasting with our observed increase in SAMS and liver enzyme elevation [31]. This may reflect complex pharmacokinetic-pharmacodynamic interactions, where lower plasma levels don’t always mean lower toxicity. Tissue distribution, transporter inhibition, pharmacodynamic effects, and genetic and clinical factors may influence outcomes. Further studies are needed to clarify these mechanisms. Zhu and colleagues conducted a metanalysis (N = 3,105), and they found no significant difference in the safety profile, specifically, transaminase elevation between double-dose statin monotherapy and combination therapy with ezetimibe [32]. The safety and tolerability of ezetimibe/rosuvastatin therapy were comparable with those of rosuvastatin monotherapy, as indicated by Hong and colleagues [33]. The SaveSAMS trial compares the impact of intensive statin monotherapy versus combination therapy on SAMS and transaminase elevation in elderly patients with established atherosclerotic cardiovascular disease. Their results have not yet been published [34].
The presence of the ABCG2 rs2231142 variant was also notable, particularly in the context of elevated liver enzymes (O.R = 3.481, P = 0.0005). This suggests that genetic screening could play a crucial role in choosing the appropriate statin for each patient, in this case, avoiding rosuvastatin as this variant influences its excretion [35]. In contrast, factors such as age, gender, obesity, smoking, and alcohol consumption did not significantly impact liver enzyme levels, suggesting a more complex interplay that warrants further investigation to understand the mechanisms behind statin-related liver enzyme elevation. Although asymptomatic elevation of liver enzymes does not require discontinuation of statin therapy according to the FDA guidelines [36], many studies have shown that patients are often concerned about these elevations, which can contribute to discontinuation [37] and lower adherence rates [38]. This concern is especially significant because mild enzyme elevations have been associated with higher rates of statin discontinuation against clinical recommendations. While elevated transaminase levels can have various causes, it is worth noting that the ABCG2 rs2231142 (421 C > A) variant, which has been linked to higher uric acid levels, is more frequent in certain populations [39]. Although we aimed to exclude patients with symptomatic gout, some participants carried the minor allele and therefore had a higher risk of hyperuricemia. However, there is limited evidence linking hyperuricemia itself to transaminase elevation, suggesting that other factors may also contribute to liver enzyme changes in these individuals.
Arab patients exhibited lower rates of enzyme elevation compared to non-Arabs. Furthermore, that might be due to the low frequency of ABCG2 rs2231142 among Arabs, which explains fewer cases of gout hyperuricemia and elevated liver enzyme cases, which drove us to the same conclusion of the suitability of rosuvastatin for individuals of Emirati origin [27]. These findings are consistent with previous studies that have reported a higher incidence of gastrointestinal issues and liver enzyme elevations with rosuvastatin, potentially due to its greater potency and different metabolic pathways [40]. A study published in 2016 highlighted that the maximum dose of rosuvastatin should be restricted to 20 mg in East Asians to mitigate the risk of liver enzyme elevation and rhabdomyolysis [41]. This recommendation stems from the observation that East Asians have approximately double the plasma levels of rosuvastatin compared to Western populations at the same dosage [42]. Nevertheless, the current study did not identify an association between higher doses of rosuvastatin and the Incidence of adverse events (SAMS or transaminase elevation).
The analysis of SAMS revealed that female gender, Arab ethnicity, and the SLCO1B1 rs4149056 variant are significant risk factors for developing SAMS while using atorvastatin. The odds ratio for SAMS in females was 1.7857 (P = 0.0281), indicating a more than 1.5-fold increase in risk compared to males. Gender-related differences are evident in the impact of SLCO1B1 genetic variation [43]. Female patients with the SLCO1B1 c.521T > C variant are more likely to experience higher cholesterol levels, reduced statin efficacy, increased SAMS, potentially influenced by variability in OATP1B1 activity [44] and smaller volumes of distribution relative to males [18]. Arab ethnicity was associated with a higher risk (O.R = 2.129, P = 0.0022), which may be due to genetic predispositions or cultural differences in reporting symptoms. The SLCO1B1 rs4149056 variant also showed a significant association (O.R = 2.202, P = 0.0063), corroborating previous findings that link this variant to SAMS [14, 45, 46]. A study published in the Lancet 2022 elucidates that although a marginal increase in muscle pain occurs initially with statin use, most muscle-related complaints are not attributable to statins [47]. Importantly, the minor risk of SAMS is substantially outweighed by the cardiovascular advantages of statin therapy.
While more studies are warranted, a preliminary conclusion suggests that rosuvastatin should not be prescribed at high doses to East Asians. We noticed that physicians in the UAE do not consider ethnicity when prescribing statins. Instead, moderate-intensity atorvastatin might be a better choice for them. This recommendation is particularly pertinent because the MAF of the ABCG2 rs2231142 (421 C > A) variant is approximately 30% in East Asians. In contrast, it is about 10.3% in Europeans and much lower in African populations at around 2.7% (gnomad.broadinstitute.org). It is contributing to increased sensitivity to rosuvastatin and its adverse events.
The study highlights the need to re-evaluate the choice of statin-type therapy in patients who experience statin-related events, as most patients can tolerate long-term statin use if rechallenged [48]. Clinicians may consider evaluating known risk factors, including genetic markers, when choosing among statins rather than relying on empirical trial-and-error approaches. This approach is vital for preventing avoidable cardiovascular events and deaths that could result from the permanent cessation of statin therapy. Based on the factors discussed in this study, it is possible to prevent discontinuation due to side effects from the beginning by choosing the appropriate statin for each individual. These factors include gender and SLCO1B1 genotyping among atorvastatin users, ezetimibe therapy, and ABCG2 for rosuvastatin. Atorvastatin might be safer for individuals with fatty livers and other liver risk factors. A trial to implement pharmacogenetic testing in the healthcare system in the UAE, conducted from 2021 to 2024, is underway [17]. We are currently analyzing the data collected to refine further and tailor our approaches to statin therapy based on genetic profiles. Longitudinal studies are also needed to assess personalized statin therapy’s long-term safety and efficacy. In addition to transporter gene variants, other factors, such as CYP3A enzyme activity, also influence statins plasma levels and metabolism, as highlighted [49]. Including CYP3A activity in future analyses could provide a more comprehensive understanding of pharmacokinetic variability and aid in optimizing personalized dosing strategies for atorvastatin and rosuvastatin.
Study limitations
While this study provides valuable insights, several limitations must be acknowledged. First, the observational design restricts our ability to draw causal inferences. Additionally, the reliance on self-reported symptoms may introduce reporting bias. Future research should validate these findings through randomized controlled trials and investigate the mechanisms underlying the observed genetic associations. We excluded patients whose muscle pain could be attributed to factors such as recent strenuous physical activity, changes in exercise patterns, concurrent illnesses, hypothyroidism, or underlying muscle conditions. However, for some patients who did not return for follow-up visits, we relied solely on medical records. Furthermore, our data on muscle pain relied on phone calls and patient-reported symptoms, which could introduce subjectivity. Secondly, our approach to diagnosing statin-associated muscle symptoms (SAMS) may not effectively differentiate subjective reports from objectively confirmed cases, potentially impacting the accuracy of muscle symptom prevalence estimates related to statin use. Thirdly, Creatine kinase (CK) was not included as a muscle damage indicator, as CK lab tests were unavailable in the medical records for our cohort. Future studies could incorporate CK measurements to assess muscle-related effects with statins better. Fourth, high stratification and multiple testing may limit the generalizability and robustness of some findings. Fifth, although our study included comprehensive medical reviews, limitations existed in assessing all potential contributors to transaminase elevations. Specifically, we were unable to evaluate the impact of over-the-counter medications (e.g., acetaminophen) and herbal products on these laboratory values. Finally, although our analysis focused on SLCO1B1 and ABCG2 variants supported by strong evidence of association with statins pharmacokinetics, we acknowledge that the inclusion of additional polymorphisms in the same genes may offer a more comprehensive evaluation of transporter functionality.
Conclusions
Our study highlights significant differences in adverse effects between atorvastatin and rosuvastatin, influenced by genetic variants and demographic factors. Given that genetic factors are the most confounding elements affecting the incidence of adverse events, these findings support genetic screening to guide statin therapy and minimize adverse effects, paving the way for more personalized and effective management of dyslipidemia in diverse populations. By integrating these findings into clinical practice, healthcare providers can enhance patient outcomes and reduce the incidence of statin-related adverse events, ultimately improving the quality of care for patients with cardiovascular disease. While our study highlights associations between specific genetic variants (such as SLCO1B1 c.521T > C and ABCG2 rs2231142) and adverse reactions to statin therapy, it is possible that these associations are also influenced by ethnicity-related factors beyond the tested genetic markers. Ethnic groups may carry additional genetic variations or environmental exposures that impact drug metabolism and response, which were not specifically examined in this study. Therefore, while our findings suggest genetic influences on statin intolerance, they may not fully account for the complexities of interethnic variability. Further research involving broader genetic testing and multiethnic analyses is warranted to better understand these associations and to refine pharmacogenomic recommendations for diverse populations.
Data availability
The data from the current study (EMHEART) are not publicly available due to regulations of the local ethical committee but are available from the corresponding author upon reasonable request.
References
Cardiovascular diseases (CVDs)– Key facts [Internet]. 2021 Jun. Available from: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds).
Elessawy FM. The Consequences of Population Growth on the Demographic Characteristics of Abu Dhabi City in the United Arab Emirates. aeb [Internet]. 2020 [cited 2024 Jun 27];8:268–76. Available from: http://www.hrpub.org/journals/article_info.php?aid=9497
Al-Shamsi S. Performance of the Framingham coronary heart disease risk score for predicting 10-year cardiac risk in adult United Arab Emirates nationals without diabetes: a retrospective cohort study. BMC Fam Pract [Internet]. 2020 [cited 2024 Jun 26];21:175. Available from: https://biomedcentral-bmcfampract.publicaciones.saludcastillayleon.es/articles/10.1186/s12875-020-01246-2
Ghader N, Al-Yateem N, Dalibalta S, Razzak HA, Rahman SA, Al Matrooshi F et al. Cardiovascular health research priorities in the United Arab Emirates. Front Public Health [Internet]. 2023 [cited 2024 Jun 26];11:1130716. Available from: https://www.frontiersin.org/articles/10.3389/fpubh.2023.1130716/full
Roth GA, Mensah GA, Fuster V. The Global Burden of Cardiovascular Diseases and Risks. Journal of the American College of Cardiology [Internet]. 2020 [cited 2024 Jun 27];76:2980–1. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0735109720378037
Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L et al. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. European Heart Journal [Internet]. 2020 [cited 2024 Sep 23];41:111–88. Available from: https://academic.oup.com/eurheartj/article/41/1/111/5556353
Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation [Internet]. 2019 [cited 2024 Jun 26];139. Available from: https://www.ahajournals.org/doi/10.1161/CIR.0000000000000624
Hassanein M, Sabbour H, Al Awadi F, Abusnana S, Afandi B, Al Kaabi J et al. Cardiometabolic Guidelines: Cardiovascular Risk Assessment and Management in Patients with Dysglycemia. Dubai Diabetes Endocrinol J [Internet]. 2023 [cited 2024 Sep 23];29:67–88. Available from: https://doiorg.publicaciones.saludcastillayleon.es/10.1159/000531107
Shehab A, Bhagavathula A. Statin therapy and low-density lipoprotein cholesterol reduction after acute coronary syndrome: Insights from the United Arab Emirates. Heart Views [Internet]. 2020 [cited 2024 Jun 26];21:80. Available from: https://journals.lww.com/10.4103/HEARTVIEWS.HEARTVIEWS_115_19
Reith C, Baigent C, Blackwell L, Emberson J, Spata E, Davies K et al. Effect of statin therapy on muscle symptoms: an individual participant data meta-analysis of large-scale, randomised, double-blind trials. The Lancet [Internet]. 2022 [cited 2024 Jun 21];400:832–45. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0140673622015458
Bradley CK, Wang TY, Li S, Robinson JG, Roger VL, Goldberg AC et al. Patient-Reported Reasons for Declining or Discontinuing Statin Therapy: Insights From the PALM Registry. JAHA [Internet]. 2019 [cited 2024 Jun 27];8:e011765. Available from: https://www.ahajournals.org/doi/10.1161/JAHA.118.011765
Pulipati VP, Davidson MH. Recent advances and emerging therapies in management of dyslipidemias. Trends in Cardiovascular Medicine [Internet]. 2021 [cited 2024 Oct 1];31:419–24. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1050173820301122
Link PS, Armitage, Masuda D, Collins. SLCO1B1 Variants and Statin-Induced Myopathy — A Genomewide Study. N Engl J Med [Internet]. 2008 [cited 2024 Jul 5];359:789–99. Available from: http://www.nejm.org/doi/abs/10.1056/NEJMoa0801936
Voora D, Shah SH, Spasojevic I, Ali S, Reed CR, Salisbury BA et al. The SLCO1B1*5Genetic Variant Is Associated With Statin-Induced Side Effects. Journal of the American College of Cardiology [Internet]. 2009 [cited 2024 Jun 21];54:1609–16. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0735109709017136
Silveira AMR, Duarte GHB, Fernandes AMADP, Garcia PHD, Vieira NR, Antonio MA et al. Serum Predose Metabolic Profiling for Prediction of Rosuvastatin Pharmacokinetic Parameters in Healthy Volunteers. Front Pharmacol [Internet]. 2021 [cited 2024 Jun 28];12:752960. Available from: https://www.frontiersin.org/articles/10.3389/fphar.2021.752960/full
Cooper-DeHoff RM, Niemi M, Ramsey LB, Luzum JA, Tarkiainen EK, Straka RJ et al. The Clinical Pharmacogenetics Implementation Consortium Guideline for SLCO1B1, ABCG2, and CYP2C9 genotypes and Statin‐Associated Musculoskeletal Symptoms. Clin Pharma and Therapeutics [Internet]. 2022 [cited 2024 Jun 27];111:1007–21. Available from: https://ascpt.onlinelibrary.wiley.com/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cpt.2557
Al-Mahayri ZN, Khasawneh LQ, Alqasrawi MN, Altoum SM, Jamil G, Badawi S et al. Pharmacogenomics implementation in cardiovascular disease in a highly diverse population: initial findings and lessons learned from a pilot study in United Arab Emirates. Hum Genomics [Internet]. 2022 [cited 2024 Jul 3];16:42. Available from: https://biomedcentral-humgenomics.publicaciones.saludcastillayleon.es/articles/10.1186/s40246-022-00417-9
Wilke RA, Ramsey LB, Johnson SG, Maxwell WD, McLeod HL, Voora D et al. The Clinical Pharmacogenomics Implementation Consortium: CPIC Guideline for SLCO1B1 and Simvastatin-Induced Myopathy. Clin Pharmacol Ther [Internet]. 2012 [cited 2024 Oct 10];92:112–7. Available from: https://doiorg.publicaciones.saludcastillayleon.es/10.1038/clpt.2012.57
Ramsey LB, Johnson SG, Caudle KE, Haidar CE, Voora D, Wilke RA et al. The Clinical Pharmacogenetics Implementation Consortium Guideline for SLCO1B1 and Simvastatin-Induced Myopathy: 2014 Update. Clin Pharmacol Ther [Internet]. 2014 [cited 2025 Jan 17];96:423–8. Available from: https://onlinelibrary.wiley.com/doi/10.1038/clpt.2014.125
Rosenson RS, Baker SK, Jacobson TA, Kopecky SL, Parker BA. An assessment by the Statin Muscle Safety Task Force: 2014 update. Journal of Clinical Lipidology [Internet]. 2014 [cited 2024 Jun 26];8:S58–71. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1933287414001603
Rosenson RS, Miller K, Bayliss M, Sanchez RJ, Baccara-Dinet MT, Chibedi-De-Roche D et al. The Statin-Associated Muscle Symptom Clinical Index (SAMS-CI): Revision for Clinical Use, Content Validation, and Inter-rater Reliability. Cardiovasc Drugs Ther [Internet]. 2017 [cited 2024 Jun 26];31:179–86. Available from: http://link.springer.com/10.1007/s10557-017-6723-4
Schachter M. Chemical, pharmacokinetic and pharmacodynamic properties of statins: an update. Fundamemntal Clinical Pharma [Internet]. 2005 [cited 2024 Jun 20];19:117–25. Available from: https://onlinelibrary.wiley.com/doi/10.1111/j.1472-8206.2004.00299.x
Nolan JA, Skuse P, Govindarajan K, Patterson E, Konstantinidou N, Casey PG et al. The influence of rosuvastatin on the gastrointestinal microbiota and host gene expression profiles. American Journal of Physiology-Gastrointestinal and Liver Physiology [Internet]. 2017 [cited 2024 Jun 21];312:G488–97. Available from: https://www.physiology.org/doi/10.1152/ajpgi.00149.2016
Kummen M, Solberg OG, Storm-Larsen C, Holm K, Ragnarsson A, Trøseid M et al. Rosuvastatin alters the genetic composition of the human gut microbiome. Sci Rep [Internet]. 2020 [cited 2024 Jun 21];10:5397. Available from: https://www.nature.com/articles/s41598-020-62261-y
Roy D, Mahapatra T, Manna K, Kar A, Rana MS, Roy A et al. Comparing effectiveness of high-dose Atorvastatin and Rosuvastatin among patients undergone Percutaneous Coronary Interventions: A non-concurrent cohort study in India. Den Uil C, editor. PLoS ONE [Internet]. 2020 [cited 2024 Jun 23];15:e0233230. Available from: https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0233230
Naci H, Brugts J, Ades T, Comparative Tolerability. and Harms of Individual Statins: A Study-Level Network Meta-Analysis of 246 955 Participants From 135 Randomized, Controlled Trials. Circ: Cardiovascular Quality and Outcomes [Internet]. 2013 [cited 2024 Jun 21];6:390–9. Available from: https://www.ahajournals.org/doi/10.1161/CIRCOUTCOMES.111.000071
Alqasrawi MN, Al-Mahayri ZN, Alblooshi H, Alsafar H, Ali BR. Utilizing Pharmacogenomic Data for a Safer Use of Statins among theEmirati Population. CVP [Internet]. 2024 [cited 2024 Jun 21];22:218–29. Available from: https://www.eurekaselect.com/226224/article
Ward NC, Watts GF, Eckel RH. Statin Toxicity: Mechanistic Insights and Clinical Implications. Circ Res [Internet]. 2019 [cited 2023 Dec 29];124:328–50. Available from: https://www.ahajournals.org/doi/10.1161/CIRCRESAHA.118.312782
Glynn RJ. Rosuvastatin for Primary Prevention in Older Persons With Elevated C-Reactive Protein and Low to Average Low-Density Lipoprotein Cholesterol Levels: Exploratory Analysis of a Randomized Trial. Ann Intern Med [Internet]. 2010 [cited 2024 Jun 21];152:488. Available from: http://annals.org/article.aspx?doi=10.7326/0003-4819-152-8-201004200-00005
Wang J, Ye Y, Chen X, Hu X, Peng Y. Trends in statin use for the primary prevention of atherosclerotic cardiovascular disease among US adults by demographic characteristics, 1999–2020. Eur J Clin Pharmacol [Internet]. 2024 [cited 2024 Jun 27]; Available from: https://link.springer.com/10.1007/s00228-024-03699-1
González-Iglesias E, Méndez-Ponce C, Ochoa D, Román M, Mejía-Abril G, Martín-Vilchez S et al. Effect of Genetic Variants on Rosuvastatin Pharmacokinetics in Healthy Volunteers: Involvement of ABCG2, SLCO1B1 and NAT2. IJMS [Internet]. 2024 [cited 2025 Apr 8];26:260. Available from: https://www.mdpi.com/1422-0067/26/1/260
Zhu Y, Hu H, Yang J, Yao Q, Xu H, Yu Y et al. The efficacy and safety of statin in combination with ezetimibe compared with double-dose statin in patients with high cardiovascular risk: A meta-analysis. Bosn J of Basic Med Sci [Internet]. 2019 [cited 2024 Jun 21]; Available from: https://www.bjbms.org/ojs/index.php/bjbms/article/view/4437
Hong SJ, Jeong HS, Ahn JC, Cha D-H, Won KH, Kim W et al. A Phase III, Multicenter, Randomized, Double-blind, Active Comparator Clinical Trial to Compare the Efficacy and Safety of Combination Therapy With Ezetimibe and Rosuvastatin Versus Rosuvastatin Monotherapy in Patients With Hypercholesterolemia: I-ROSETTE (Ildong Rosuvastatin & Ezetimibe for Hypercholesterolemia) Randomized Controlled Trial. Clinical Therapeutics [Internet]. 2018 [cited 2024 Jun 21];40:226–241.e4. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0149291818300079
Cha J-J, Hong SJ, Kim JH, Lim S, Joo HJ, Park JH et al. Effect of rosuvastatin 20 mg versus rosuvastatin 5 mg plus ezetimibe on statin side-effects in elderly patients with atherosclerotic cardiovascular disease: Rationale and design of a randomized, controlled SaveSAMS trial. American Heart Journal [Internet]. 2023 [cited 2024 Jun 21];261:45–50. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0002870323000649
Lehtisalo M, Taskinen S, Tarkiainen EK, Neuvonen M, Viinamäki J, Paile-Hyvärinen M et al. A comprehensive pharmacogenomic study indicates roles for SLCO1B1, ABCG2 and SLCO2B1 in rosuvastatin pharmacokinetics. Brit J Clinical Pharma [Internet]. 2023 [cited 2024 Jun 24];89:242–52. Available from: https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bcp.15485
U.S. Food and Drug Administration. Cholesterol-Lowering Drugs Get Labeling Changes [Internet]. 2012 Feb. Available from: https://www.fda.gov/drugs/special-features/cholesterol-lowering-drugs-get-labeling-changes
Thompson PD, Panza G, Zaleski A, Taylor B. Statin-Associated Side Effects. Journal of the American College of Cardiology [Internet]. 2016 [cited 2024 Nov 11];67:2395–410. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0735109716016922
Brinton EA. Understanding Patient Adherence and Concerns with STatins and MedicatION Discussions With Physicians (ACTION): A survey on the patient perspective of dialogue with healthcare providers regarding statin therapy. Clinical Cardiology [Internet]. 2018 [cited 2024 Nov 11];41:710–20. Available from: https://onlinelibrary.wiley.com/doi/10.1002/clc.22975
Hoque KM, Dixon EE, Lewis RM, Allan J, Gamble GD, Phipps-Green AJ et al. The ABCG2 Q141K hyperuricemia and gout associated variant illuminates the physiology of human urate excretion. Nat Commun [Internet]. 2020 [cited 2024 Jun 21];11:2767. Available from: https://www.nature.com/articles/s41467-020-16525-w
Collins R, Reith C, Emberson J, Armitage J, Baigent C, Blackwell L et al. Interpretation of the evidence for the efficacy and safety of statin therapy. The Lancet [Internet]. 2016 [cited 2024 Jun 27];388:2532–61. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0140673616313575
Benes LB, Bassi NS, Davidson MH. The Risk of Hepatotoxicity, New Onset Diabetes and Rhabdomyolysis in the Era of High-Intensity Statin Therapy: Does Statin Type Matter? Progress in Cardiovascular Diseases [Internet]. 2016 [cited 2024 Jun 25];59:145–52. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0033062016300779
Tomlinson B, Chow E. The maximum dose of atorvastatin and simvastatin as well as rosuvastatin should be restricted in East Asians. Hong Kong Med J [Internet]. 2024 [cited 2024 Jun 8]; Available from: https://www.hkmj.org/abstracts/v30n2/184.htm
Taniguchi T, Zanetti-Yabur A, Wang P, Usyk M, Burk RD, Wolkoff AW. Interindividual Diversity in Expression of Organic Anion Uptake Transporters in Normal and Cirrhotic Human Liver. Hepatol Commun [Internet]. 2020 [cited 2024 Oct 10];4:739–52. Available from: https://journals.lww.com/10.1002/hep4.1489
Neuvonen M, Tornio A, Hirvensalo P, Backman JT, Niemi M. Performance of Plasma Coproporphyrin I and III as OATP1B1 Biomarkers in Humans. Clin Pharma and Therapeutics [Internet]. 2021 [cited 2024 Oct 10];110:1622–32. Available from: https://ascpt.onlinelibrary.wiley.com/doi/10.1002/cpt.2429
Brunham LR, Lansberg PJ, Zhang L, Miao F, Carter C, Hovingh GK et al. Differential effect of the rs4149056 variant in SLCO1B1 on myopathy associated with simvastatin and atorvastatin. Pharmacogenomics J [Internet]. 2012 [cited 2024 Jun 21];12:233–7. Available from: https://www.nature.com/articles/tpj201092
Lu B, Sun L, Seraydarian M, Hoffmann TJ, Medina MW, Risch N et al. Effect of SLCO1B1 T521C on Statin-Related Myotoxicity With Use of Lovastatin and Atorvastatin. Clin Pharma and Therapeutics [Internet]. 2021 [cited 2024 Jun 21];110:733–40. Available from: https://ascpt.onlinelibrary.wiley.com/doi/10.1002/cpt.2337
Marcus ME, Manne-Goehler J, Theilmann M, Farzadfar F, Moghaddam SS, Keykhaei M et al. Use of statins for the prevention of cardiovascular disease in 41 low-income and middle-income countries: a cross-sectional study of nationally representative, individual-level data. The Lancet Global Health [Internet]. 2022 [cited 2023 Dec 29];10:e369–79. Available from: https://linkinghub.elsevier.com/retrieve/pii/S2214109X21005519
Zhang H, Plutzky J, Skentzos S, Morrison F, Mar P, Shubina M et al. Discontinuation of Statins in Routine Care Settings.
DeGorter MK, Tirona RG, Schwarz UI, Choi Y-H, Dresser GK, Suskin N et al. Clinical and Pharmacogenetic Predictors of Circulating Atorvastatin and Rosuvastatin Concentrations in Routine Clinical Care. Circ Cardiovasc Genet [Internet]. 2013 [cited 2024 Nov 11];6:400–8. Available from: https://www.ahajournals.org/doi/10.1161/CIRCGENETICS.113.000099
Acknowledgements
The authors would like to thank the UAE Ministry of Education (MOE) for the generous funding of this project.
Funding
This research is supported by a collaborative research grant from the UAE Ministry of Education (grant number 1570604941; UAEU21M139). We also thank UAEU for supporting M.N.A. through a Ph.D. tuition waiver fellowship.
Author information
Authors and Affiliations
Contributions
Authorship Contribution Mais N. Alqasrawi: Conceptualization, Data curation, Formal analysis, Visualization, Investigation, Methodology & Writing the original draft. Zeina N. Al‑Mahayri: Conceptualization, Validation, Methodology & Writing– review and editing. Areej S. AlBawa’neh: Data curation, Investigation, Formal analysis & writing the original draft. Lubna Q. Khasawneh: Data curation & Investigation. Lilas Dabaghie: Data curation. Sahar M. Altoum: Data curation. Dana Hamza: Investigation. Virendra Misra: Investigation & Writing– review and editing, Husam Ouda: Investigation& Writing– review and editing. Salahdein Aburuz: Methodology, Formal Analysis & Writing– review and editing, Fatima Al-Maskari: Conceptualization, Funding acquisition & Writing– review and editing. Juma AlKaabi: Conceptualization, Funding acquisition & Writing– review and editing, George P. Patrinos: Conceptualization, Funding acquisition & Writing– review and editing. Bassam R. Ali: Conceptualization, Funding acquisition, Writing– review and editing, Data Curation, supervision, validation &Project administration.All co-authors contributed to the final manuscript and approved it.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
This study was conducted in compliance with the Declaration of Helsinki and was approved by the Abu Dhabi Health Research and Technology Ethical Committee (Institutional Review Board - IRB) (Ref. DOH/CVDC/2020/1187). All participants provided written informed consent before enrolment in the study.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Alqasrawi, M.N., Al‑Mahayri, Z.N., AlBawa’neh, A.S. et al. Pharmacogenomic insights into atorvastatin and rosuvastatin adverse effects: a prospective observational study in the UAE’s multiethnic population. Hum Genomics 19, 44 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40246-025-00753-6
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40246-025-00753-6