Next Article in Journal
Moringa (Moringa oleifera) Leaf Attenuates the High-Cholesterol Diet-Induced Adverse Events in Zebrafish: A 12-Week Dietary Intervention Resulted in an Anti-Obese Effect and Blood Lipid-Lowering Properties
Next Article in Special Issue
Efficacy of SGLT2 Inhibitors, GLP-1 Receptor Agonists, DPP-4 Inhibitors, and Sulfonylureas on Moderate-to-Severe COPD Exacerbations Among Patients with Type 2 Diabetes: A Systematic Review and Network Meta-Analysis
Previous Article in Journal
Bee Venom Proteins Enhance Proton Absorption by Membranes Composed of Phospholipids of the Myelin Sheath and Endoplasmic Reticulum: Pharmacological Relevance
Previous Article in Special Issue
Empagliflozin Protects Against Oxidative Stress in the Diabetic Brain by Inducing H2S Formation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Pharmacogenetics and Molecular Ancestry of SLC22A1, SLC22A2, SLC22A3, ABCB1, CYP2C8, CYP2C9, and CYP2C19 in Ecuadorian Subjects with Type 2 Diabetes Mellitus

by
Adiel Ortega-Ayala
1,2,3,4,†,
Carla González de la Cruz
2,3,4,†,
Lorena Mora
5,6,
Mauro Bonilla
6,
Leandro Tana
6,
Fernanda Rodrigues-Soares
2,3,4,7,
Pedro Dorado
2,4,
Adrián LLerena
2,3,4,* and
Enrique Terán
4,6,*
1
Department of Pharmacology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
2
Personalised Medicine and Mental Health Unit, University Institute for Biosanitary Research of Extremadura (INUBE), 06080 Badajoz, Spain
3
Pharmacogenomics and Personalized Medicine Unit, Badajoz University Hospital, Extremadura Health Service SES, 06006 Badajoz, Spain
4
RIBEF Red Iberoamericana de Farmacogenégica y Farmacogenómica SIFF, 06080 Badajoz, Spain
5
Laboratorio Clínico, Dispensario Central del Instituto Ecuatoriano de Seguridad Social (IESS), Quito 170901, Ecuador
6
Colegio de Ciencias de la Salud, Universidad San Francisco de Quito (USFQ), Quito 170901, Ecuador
7
Department of Pathology, Genetic and Evolution, Universidade Federal do Triângulo Mineiro, Uberaba 38025-350, Brazil
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2025, 18(9), 1335; https://doi.org/10.3390/ph18091335
Submission received: 6 August 2025 / Revised: 1 September 2025 / Accepted: 3 September 2025 / Published: 5 September 2025

Abstract

Background/Objectives: In Ecuador, the prevalence of type 2 diabetes mellitus (T2DM) is the second leading cause of death after ischemic heart disease. Genetic variability in protein-coding genes, single nucleotide variants (SNVs), influences the response to antidiabetic drugs. The frequency of SNVs varies among different populations, so studying the ancestral proportions among SNVs is important for personalized medicine in the treatment of T2DM. This study aimed to evaluate the distribution of Native American, European, and African (NATAM, EUR, and AFR) ancestry in 23 allelic variants of the seven genes that encode the relevant enzymes that metabolize antidiabetic drugs in an Ecuadorian population. Methods: Twenty-three allelic variants of seven genes were analyzed in 297 patients with T2DM from Ecuador, and the molecular ancestry of the samples was analyzed considering three ancestral groups, NATAM, EUR, and AFR using 90 ancestry informative markers (AIMs). Allele and ancestry distributions were analyzed using Spearman’s correlation. Results: The Ecuadorian population presents NATAM (61.33%), EUR (34.48%), and AFR (2.60%) ancestry components. CYP2C8*1 and CYP2C9*1 were positively related to NATAM ancestry, while CYP2C8*4 and CYP2C9*2 were positively related to EUR ancestry. CYP2C19*17 was positively correlated to AFR ancestry. The correlation of SLC22A1 variants such as A in rs594709 was positively correlated with NATAM, while GAT in rs72552763 was positive for EUR. The G variant of rs628031 of the SLC22A1 gene was positively correlated with NATAM and negatively correlated with EUR. The C variant of rs2076828 of the SLC22A3 gene was positively correlated with NATAM ancestry. Conclusions: In the Ecuadorian population, a predominance of Native American ancestry has been observed. Among the allelic variants related to enzymes that metabolize antidiabetic drugs, a relationship has been observed between this ancestral component and variants of the CYP2C8*1, CYP2C9*1, SLC22A1 (rs594709 and rs628031), and SLC22A3 (rs2076828) genes. This information is fundamental for the development of strategies for the implementation of personalized medicine programs for Latin American patients.

Graphical Abstract

1. Introduction

Type 2 diabetes mellitus (T2DM) accounts for a major public health problem worldwide due to its high prevalence, increasing incidence, high morbidity and mortality rates, and considerable healthcare costs. T2DM currently affects more than 500 million people, and the healthcare burden is unevenly distributed, disproportionately affecting low- and middle-income countries, which account for approximately 80% of the global diabetic population [1].
According to the International Diabetes Federation, the highest rates of T2DM incidence in young populations are found among Canadian First Nations, Native Americans, Indigenous Australians, and Afro-Latin Americans. In contrast, the lowest incidence rates are found among Europeans and white Americans [2]. These differences are attributed to interethnic variability, genetic predisposition, and disparities in access to healthcare [2].
It is estimated that approximately 40% of people with T2DM in Latin America are undiagnosed, making it difficult to accurately estimate the costs to healthcare systems of treating and caring for patients with T2DM [3].
In Ecuador, the prevalence of diabetes is 7.1% [4], and it is the second leading cause of death after ischemic heart disease, for which T2DM is also a risk factor [5].
Metformin remains the first-line pharmacological treatment for T2DM. However, when therapeutic goals are not achieved with monotherapy, combination therapy is recommended. The most effective agents used in combination include pioglitazone, sodium-glucose cotransporter 2 inhibitors (SGLT2i), glucagon-like peptide-1 receptor agonists (GLP-1), and sulfonylureas (SU) [6].
Despite the wide range of pharmacological treatments available, interindividual variability in response to drugs remains a significant challenge. This variability is largely influenced by variations in the genes involved in the pharmacokinetics and pharmacodynamics of antidiabetic drugs, accounting for 20–30% of these differences in pharmacological response [7].
Genetic variation not only affects therapeutic efficacy but can also contribute to the occurrence of adverse drug reactions (ADRs). Specifically, 59% of drugs commonly associated with ADRs are metabolized by enzymes encoded by allelic variants with altered function [8]. This is important, as it has now been shown that overall mortality due to adverse events has tripled between 2001 and 2019 [9]. Specifically, hypoglycemic agents are one of the drugs of greatest concern in the occurrence of ADRs [10].
The mechanism of action of metformin is mainly through the inhibition of hepatic glucose production, and its cellular uptake and distribution depend on organic cation transporters (OCTs), particularly OCT1, OCT2, OCT3, and P-glycoprotein (P-gp). These transporters are encoded by highly polymorphic genes SLC22A1, SLC22A2, SLC22A3, and ABCB1, respectively [11], which are associated with the therapeutic response to metformin and play a fundamental role in its pharmacokinetics [12]. An association has been demonstrated between genetic variants of SLC22A1 and the onset of gastrointestinal ADRs in diabetic patients treated with metformin [13,14].
Drugs such as SU and thiazolidinediones (TZD) are metabolized mainly by cytochrome P450 (CYP450) enzymes. Specifically, SUs are mainly metabolized by CYP2C9 and CYP2C19 [15], while TZDs are metabolized by CYP2C8 [16]. Several studies have described an association between reduced CYP2C9 function and an increased risk of hypoglycemia secondary to SU use [17,18]. In the case of TZDs, there are studies that report the appearance of edema or lower weight gain in patients carrying the CYP2C8*3 genetic variant compared to wild-type patients [19]. Therefore, in managing the disease, the goal is to identify the most appropriate treatment for each patient, for which it is essential to incorporate the analysis of pharmacogenetic biomarkers into clinical practice, both for disease prediction and for pharmacological monitoring [20].
In sum, seven genes have been described as the most relevant for the pharmacogenetics of antidiabetic drugs: CYP2C8 for TZDs, CYP2C9 and CYP2C19 for SU, and SLC22A1, SLC22A2, SLC22A3, and ABCB1 for metformin.
An association has been demonstrated between genetic variants relevant to drug response and the ethnogeographic distribution of the population. Therefore, information on the distribution of these variants is important for guiding specific therapies for each population [21,22]. However, to date, no pharmacogenetic studies have specifically addressed the influence of the unique genetic ancestry of the Ecuadorian population diagnosed with T2DM. Most pharmacogenetic investigations aiming to optimize antidiabetic drug prescription have been conducted predominantly in European populations, potentially limiting the applicability of their findings to the Ecuadorian context [23]. This fact is especially relevant in populations with weak health systems, which are often accompanied by a shortage of medicines for the treatment of relevant diseases such as T2DM [24].
As in other populations in South America, the Ecuadorian population is multicultural and multiethnic, resulting from multiple migratory events and admixture over more than 525 years. This means that this population is characterized by the continuous mixing of European, Amerindian, and African individuals, giving rise to great ethnic diversity [25]. At present, Ecuador comprises three main ethnic groups: the mestizo population, Native Amerindian, and Afro-Ecuadorian. The mestizo population represents approximately 85% of the total population and is a mixed group of European and Amerindian descent (NATAM) [25].
The study of population-specific genetic variants, with the aim of providing evidence to optimize the clinical implementation of pharmacogenetics in Latin American populations, has been the ultimate goal of the RIBEF (Ibero-American Network of Pharmacogenetics and Pharmacogenomics). This has involved initial studies of self-reported ancestry in individuals [26], followed by subsequent analyses of the molecular ancestry of the population [27]. The main objective is to develop strategies for the implementation of pharmacogenetics adapted to the Latin American population [21,22]. In a previous study in Mexico [28,29], the RIBEF studies were initiated in diabetic patients, one of the most prevalent diseases in this region that causes relevant health problems, finding relevant information regarding the relevance of the ancestral component, pharmacogenetic polymorphisms and the response to antidiabetic drug treatment. In this Mexican DMT2 study, the percentage of NATAM ancestry was 65.48% and was positively correlated with CYP2C8*1, CYP2C9*1, SLC22A1 (rs594709), and SLC22A3 (rs2076828). This study moves the question from North America to the most relevant populations in South America, the Andean populations studied in Ecuador. Diabetes is associated in part with poverty-related health problems, which is sometimes related to autochthonous American populations, contributing to the maintenance of the poverty–disease circuit. The main aim is therefore whether there is any peculiarity in terms of pharmacogenetic variants that would allow the optimization of personalized medicine programs in Latin American autochthonous populations [26,27]. In other words, if the Native American component (NATAM) is related to the presence of certain pharmacogenetic variables in diabetic patients from Ecuador in the South American region of the Andes in order to develop Personalized Medicine programs for T2DM patients.
This present study aims to evaluate the distribution of Native American (NATAM), European (EUR), and African (AFR) ancestry in Ecuadorian T2DM patients and its correlation with 19 variants of the seven genes (CYP2C8, CYP2C9, CYP2C19, SLC22A1, SLC22A2, SLC22A3, and ABCB1) that encode the relevant enzymes and transporters implicated in the pharmacogenetics of antidiabetic drugs.

2. Results

2.1. Genotype and Allelic Frequencies

This study analyzed 297 patients with T2DM from Ecuador. The diplotype and allele frequencies of the cytochromes CYP2C8, CYP2C9, and CYP2C19, as well as the “activity score”, are summarized in Table S1, as well as the allelic and genotypic frequencies and activity score distribution by CYP2C8, CYP2C9, and CYP2C19 within a sample of Ecuadorian T2DM patients (n = 297). The allelic and genotypic frequencies of the SNVs in the SLC22A1, SLC22A2, SLC22A3, and ABCB1 genes are summarized in Table S2.
SNV allelic and genotypic frequencies in SLC22A1, SLC22A2, SLC22A3, and ABCB1 (n = 297) are reported. All analyzed alleles were in Hardy–Weinberg equilibrium, except for SNV rs594709 in the SLC22A1 gene (p = 0.030).

2.2. Description of Ancestry in Ecuadorian Patients with T2DM

The proportion of NATAM, EUR, and AFR ancestry was determined in 294 Ecuadorian patients. The clusters from which ancestry proportions were estimated are shown in Figure 1. In the analysis of the median and the 25th and 75th percentiles of ancestry proportions in our sample, we found that the highest proportion corresponded to NATAM ancestry [61.33% (51.59–70.61)], followed by EUR ancestry [34.48% (25.55–42.36)] and AFR ancestry [2.60% (0.00–7.45)].

2.3. Ancestry Inference Among CYP2C8, CYP2C9, and CYP2C19 Diplotypes and Activity Scores

The proportion of NATAM, EUR, and AFR ancestries by groups of individuals with different diplotypes and activity scores of the cytochromes CYP2C8, CYP2C9, and CYP2C19 is summarized in Table 1.

2.3.1. CYP2C8

Comparing the ancestry proportions among the different CYP2C8 diplotypes, we found statistically significant differences for NATAM ancestry (p = 0.002) and EUR ancestry (p = 0.005). Post hoc analysis of CYP2C8 diplotypes revealed that, for NATAM ancestry, the *1/*1 diplotype was significantly different*1/*3 (pBonferroni = 0.034) and *1/*4 (pBonferroni = 0.034). According to these results, patients with the *1/*1 diplotype had a higher proportion of NATAM ancestry compared to those with the *1/*3 and *1/*4 diplotypes (Figure 2A). In the analysis by CYP2C8 activity score groups (Figure 2D), patients categorized with an activity score of 2 had a higher proportion of NATAM ancestry compared to those with a score of 1.5 (p < 0.001). Similarly, patients carrying the CYP2C8 *1/*4 diplotype exhibited a higher proportion of EUR ancestry compared to those with the *1/*1 diplotype (pBonferroni = 0.030; Figure 2B). In the activity score analysis (Figure 2E), patients with an activity score of 2 showed a lower proportion of EUR ancestry compared to those with a score of 1.5 (p = 0.003). No differences were found in AFR ancestry proportions among CYP2C8 diplotypes (Figure 2C) or activity scores (Figure 2F).

2.3.2. CYP2C9

In the ancestry inference analysis among CYP2C9 diplotypes and activity scores, differences were found only for NATAM ancestry (Table 1). In the post hoc analysis, patients carrying the CYP2C9 *1/*1 diplotype showed a higher proportion of NATAM ancestry compared to those with the CYP2C9 *1/*2 diplotype (pBonferroni = 0.021) (Figure 3A). Similarly, patients categorized with an activity score of 2 had a higher proportion of NATAM ancestry compared to those with an activity score of 1.5 (Figure 3D).

2.3.3. CYP2C19

In the ancestry inference analysis among CYP2C19 diplotypes and activity score, statistical significance was found only for AFR ancestry. In the post hoc analysis of CYP2C19 diplotypes (Figure 4C), patients carrying the CYP2C19 *1/*17 diplotype had a higher proportion of AFR ancestry compared to those with CYP2C19 *1/*1 (pBonferroni = 0.049) and CYP2C19 *1/*2 (pBonferroni = 0.037). Despite classifying a CYP2C19 phenotype based on the alleles carried by the patients, we decided to establish an activity score similar to the current CYP2D6 phenotype classification, as has been proposed in previous studies [31]. Moreover, in the activity score analysis (Figure 4F), patients categorized as UM (ultra-rapid metabolizers) had a higher proportion of AFR ancestry compared to those with activity scores of 2 (pBonferroni = 0.049) and 1 (pBonferroni = 0.037).

2.4. Ancestry Inference Among Transporter SNVs

The inference of NATAM, EUR, and AFR ancestry proportions across groups of individuals with different genotypes of the transporters OCT1, OCT2, OCT3, and p-gp, encoded by the SLC22A1, SLC22A2, SLC22A3, and ABCB1 genes, respectively, is summarized in Table 2. Statistical significance was found only in the distribution of NATAM, EUR, and/or AFR ancestry for variants in SLC22A1 (rs72552763, rs594709, rs628031) and SLC22A3 (rs2076828).

2.4.1. SLC22A1

For the SLC22A1 rs72552763 variant, differences in NATAM (p = 0.022) and EUR (p = 0.017) ancestry proportions were found. Post hoc analysis showed that patients with the del/del genotype had a higher proportion of NATAM ancestry compared to those with the GAT/GAT genotype (pBonferroni = 0.044) (Figure 5A). Regarding EUR ancestry, patients with the GAT/GAT genotype had a higher EUR ancestry proportion than those with del/del (pBonferroni = 0.034) (Figure 5B). For SLC22A1 rs594709, ancestry differences were found in NATAM (p = 0.040) and EUR (p = 0.013). Post hoc analysis revealed that patients with the AA genotype had higher NATAM ancestry than those with AG (pBonferroni = 0.034) (Figure 5D). In contrast, EUR ancestry was higher in patients with the AG genotype compared to those with AA (pBonferroni = 0.046) (Figure 5E).
For rs628031, differences were found in NATAM (p = 0.042) and EUR (p = 0.040) ancestry proportions. Post hoc analysis of NATAM ancestry (Figure 5G) showed that patients with the GG genotype had a higher proportion of NATAM ancestry compared to GA carriers (pBonferroni = 0.036). Conversely, patients with the GA genotype had higher EUR ancestry compared to GG carriers (pBonferroni = 0.044) (Figure 5H).

2.4.2. SLC22A3

Statistically significant differences in NATAM (p < 0.001) and EUR (p < 0.001) ancestry proportions were found for individuals with different genotypes of SLC22A3 rs2076828. Post hoc analysis revealed that patients with the CC genotype had higher NATAM ancestry (Figure 5J) than those with the CG genotype (pBonferroni < 0.001). Conversely, CG genotype carriers had higher EUR ancestry (Figure 5K) compared to CC carriers (pBonferroni = 0.001).

2.5. Correlation Analysis Between Genetic Variants and Ancestry Proportion

To explore correlations between alleles and NATAM, EUR, and AFR ancestry proportions, we conducted Spearman correlation analyses for each cytochrome and transporter allele found to be statistically significant in inferential analyses. Allelic correlation and ancestry proportion analyses were performed for CYP2C8, CYP2C9, CYP2C19, and variants of SLC22A1 (rs72552763, rs594709, rs628031) and SLC22A3 (rs2076828).

2.5.1. Correlation Analysis for CYP2C8 Variants

The findings of the correlation analysis between alleles (wt, *3, *4) and CYP2C8 activity score with NATAM, EUR, and AFR ancestry are summarized in Table S3. Statistically significant correlations were observed between certain CYP2C8 alleles and NATAM (wt, *3, *4) or EUR (wt and *4) ancestry, but not AFR.
A positive correlation was found between the CYP2C8 wt allele and NATAM ancestry (Rho = 0.193, p < 0.001), and negative correlations were observed for CYP2C8*3 (Rho = −0.140, p = 0.015) and *4 (Rho = −0.137, p = 0.018). EUR ancestry showed a negative correlation with the wt allele (Rho = −0.170, p = 0.003) and a positive correlation with CYP2C8*4 (Rho = 0.142, p = 0.014).

2.5.2. Correlation Analysis for CYP2C9 Variants

The correlation analysis between alleles (wt, *2, *3) and CYP2C9 activity score with NATAM, EUR, and AFR ancestry is summarized in Table S4. NATAM ancestry showed a positive correlation with the wt allele (Rho = 0.163, p = 0.005) and a negative correlation with the CYP2C9*2 allele (Rho = −0.141, p = 0.015). EUR ancestry showed a negative correlation with the wt allele (Rho = −0.127, p = 0.028) and a positive correlation with CYP2C9*2 (Rho = 0.121, p = 0.038). AFR ancestry showed a negative correlation with the wt allele (Rho = −0.130, p = 0.025).

2.5.3. Correlation Analysis for CYP2C19 Variants

The correlation analysis between alleles (wt, *2, *4, *17) and CYP2C19 activity score is summarized in Table S5. Correlation between ancestry proportion and allelic frequency in CYP2C19 variants. Statistically significant findings were observed only for the CYP2C19*17 allele, which was negatively correlated with NATAM ancestry (Rho = −0.146, p = 0.012) and positively correlated with AFR ancestry (Rho = 0.174, p = 0.002).
To analyze the correlation between enzymatic activity of CYP2C8, CYP2C9, and CYP2C19, a scatter plot was generated for activity scores and ancestry proportions (NATAM, EUR, AFR), adjusted with a linear regression line (Figure 6). For CYP2C8 activity scores (1.5 and 2), a positive correlation with NATAM ancestry (Rho = 0.193, p < 0.001) and a negative correlation with EUR ancestry (Rho = −0.170, p = 0.003) were found (Figure 6A). For CYP2C9 enzymatic activity (Figure 6B), a positive correlation was found with NATAM ancestry (Rho = 0.161, p = 0.005), and negative correlations with EUR ancestry (Rho = −0.124, p = 0.032) and AFR ancestry (Rho = −0.135, p = 0.022). For CYP2C19, a positive correlation was observed between activity score and AFR ancestry (Rho = 0.145, p = 0.012).

2.5.4. Correlation Analysis for Transporters SNVs

The correlation analysis between ancestry proportions (NATAM, EUR, AFR) and SNV alleles (rs72552763, rs594709, rs628031, rs2076828) is summarized in Table S6. For NATAM ancestry, positive correlations were found with the A allele of rs594709 (Rho = 0.147, p = 0.011), the G allele of rs628031 (Rho = 0.147, p = 0.011), and the C allele of rs2076828 (Rho = 0.261, p < 0.001), while a negative correlation was observed with the GAT allele of rs72552763 (Rho = −0.157, p = 0.006). For EUR ancestry, a positive correlation was found with the GAT allele of rs72552763 (Rho = 0.162, p = 0.005), and negative correlations with the A allele of rs594709 (Rho = −0.169, p = 0.003), the G allele of rs628031 (Rho = −0.148, p = 0.010), and the C allele of rs2076828 (Rho = −0.230, p < 0.001). Only the C allele of rs2076828 was negatively correlated with AFR ancestry (Rho = −0.127, p = 0.029).

3. Discussion

3.1. Ancestry of the Ecuadorian Population

Similar to other Latin American populations, the Ecuadorian population is defined as multiethnic and multicultural, with a complex demographic history of admixture involving Europeans, Native Americans [32], and individuals of African descent since the Spanish colonization. This admixture has shaped the current patterns of ethnic diversity in the population [33].
Within this historical context, our results indicate that the studied population has an ancestry composition of 61.33% NATAM, 34.48% EUR, and 2.60% AFR. These findings are consistent with recent studies reporting a predominance of NATAM ancestry in the Ecuadorian population [33,34].
When compared to other Latin American populations, a high proportion of NATAM ancestry has also been observed in Mexico [28] and Peru [35]. In contrast, populations from Colombia [36] and Brazil [37] exhibit a predominance of EUR ancestry. Caribbean populations, such as those in the Dominican Republic and Puerto Rico, on the other hand, present a significant proportion of AFR ancestry [38,39]. This genetic diversity is associated with interindividual variability in response to pharmacological therapies [40]. This constitutes a current challenge in pharmacology, as therapeutic response to various treatments has been shown to vary substantially across global populations [41]. One of the contributing factors is the frequency distribution of pharmacogenetic variants among different ethnic groups [33].

3.2. Ancestry and Pharmacogenetics of CYP450

3.2.1. CYP2C8

In the analysis of molecular ancestry and genetic variants of CYP2C8, significant differences were found in relation to the NATAM and EUR ancestral components. Specifically, carriers of the *1 allele exhibited predominantly NATAM ancestry, which was less prevalent among carriers of the *3 and *4 alleles. These results suggest a positive association between the CYP2C8*1 variant and NATAM ancestry, while the CYP2C8*4 variant may correlate with EUR ancestry. None of the alleles showed a significant association with AFR ancestry.
The allele frequencies of CYP2C8*3 and CYP2C8*4 in the studied population were 5.21% and 1.51%, respectively. These values are slightly lower than previously reported frequencies, where CYP2C8*3 was found at 8.1% in the Ecuadorian population [42]. In contrast, public databases report a somewhat higher frequency of this variant in European populations (~10%) [43].
CYP2C8 variability may significantly impact the pharmacokinetics, therapeutic response, and toxicity of a wide range of drugs, including antidiabetics [44]. Notably, the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD) recommend the use of TZDs, particularly pioglitazone and rosiglitazone, for the treatment of diabetes [16]. These drugs are primarily metabolized by CYP2C8 [16]. The CYP2C8*4 variant is considered a loss-of-function allele; however, the enzymatic activity of CYP2C8*3 remains unclear, as it has been shown to be substrate-specific. Some studies have reported no reduction in pioglitazone metabolism, whereas others have demonstrated increased metabolism of TZDs and a 36% reduction in rosiglitazone plasma concentration among healthy carriers of CYP2C8*3. This reduction may lead to diminished therapeutic efficacy, with higher levels of glycated hemoglobin observed in CYP2C8*3 carriers compared to *1/*1 individuals [16]. Additionally, adverse effects such as edema and weight gain have been reported more frequently in T2DM patients carrying the *3 allele compared to wild-type individuals [19].
Currently, no specific pharmacogenetic clinical guidelines exist for CYP2C8. However, this enzyme is included alongside CYP2C9 in the pharmacogenetic guideline for non-steroidal anti-inflammatory drugs (NSAIDs) [45] because the CYP2C8*3 allele is in strong linkage disequilibrium with the CYP2C9*2 allele [46,47]. Consequently, for CYP2C8 activity classification, we adopted the same system used for CYP2C9 [45], where CYP2C8*3 and CYP2C8*4 are assigned an activity score of 1.5. More recently, an alternative phenotypic classification for CYP2C8 has been proposed, defining UMs as *3/*3 individuals, rapid metabolizers (RMs) as *1/*3, normal metabolizers (NMs) as *1/*1, intermediate metabolizers (IMs) as *1/*4, and poor metabolizers (PMs) as *4/*4 [48]. This discrepancy arises from the previously mentioned substrate-specific activity of CYP2C8, leading to ongoing debate regarding the functional classification of CYP2C8*3. In fact, one study reported that depending on whether CYP2C8*3 is considered a normal- or decreased-function allele, the proportion of NMs in Ecuador could range from 100% to approximately 90%, with the remaining 10% being classified as IMs [49].
Given the absence of clinical guidelines for CYP2C8 genotyping in diabetic patients, we propose incorporating ancestry as a key factor to enhance therapeutic precision and reduce adverse reactions to hypoglycemic agents—one of the most significant concerns for current diabetic populations [10]. Therefore, due to the high proportion of NATAM ancestry in the Ecuadorian population and its association with the CYP2C8*1 variant (wild type), the present results suggest that TZDs may represent a safe and effective treatment option for Ecuadorian patients with T2DM. This supports the consideration of ethnogeographic background in CYP2C8 variability profiling.

3.2.2. CYP2C9

In the analysis of the relationship between molecular ancestry and CYP2C9 genetic variants, significant differences were observed exclusively with the NATAM ancestral component. A correlation was found between *1/*1 individuals and higher NATAM ancestry compared to *1/*2 individuals. Consequently, individuals with a genotype-inferred phenotype classified as NMs exhibited a greater proportion of NATAM ancestry than those classified as IMs. These findings support previous studies demonstrating that the CYP2C9*2 variant is more prevalent among individuals with EUR ancestry, while the CYP2C9*3 variant is commonly found in South Asian populations [50].
In the Ecuadorian population, the allele frequencies of CYP2C9*2 and CYP2C9*3 were 5.23% and 2.36%, respectively, resulting in an estimated prevalence of approximately 15% for intermediate metabolizers. These relatively low frequencies are consistent with earlier findings, where the *2 and *3 allele frequencies were reported as 2.44% and 2.83%, respectively [42,51], with a comparable IM prevalence of about 14% [51]. However, our results indicate a slightly higher frequency than that reported in a more recent study [50]. As in previous research, no PMs were identified in the population [51].
Similar frequencies of CYP2C9*2 and CYP2C9*3 have been reported in other Latin American populations, including 4.6% and 6.2% in Peru, and 3.83% and 2.82% in a Mexican T2DM population [28]. However, the highest reported frequencies of these variants in South America are found in Uruguay, Colombia, and Brazil [50], which is consistent with the greater EUR ancestry observed in those populations.
According to the Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines, the enzymatic activity of CYP2C9, and therefore the classification of individuals as NMs (*1/*1), IMs (*1/*2, *1/*3, *2/*2), or PMs (*2/*3, *3/*3), is determined by the presence of the *1, *2, and *3 alleles. These classifications are particularly relevant for the prescription of drugs such as NSAIDs [45], phenytoin [52], and warfarin [53]. Previous studies have reported adverse reactions to SUs in carriers of CYP2C9 decreased-function alleles, who are more likely to experience secondary hypoglycemia [18]. This finding is supported by a recent meta-analysis showing that T2DM patients carrying CYP2C9*2 had a higher risk of hypoglycemic episodes due to reduced metabolic activity, ultimately compromising SU efficacy [54].
Nonetheless, no consensus has been reached regarding the impact of CYP2C9 genotype on SU efficacy. Some studies report better therapeutic outcomes in *1/*1 patients compared to *1/*3 and *3/*3 carriers [55], which would favor the Ecuadorian population due to its high frequency of CYP2C9*1. Other studies suggest that CYP2C9*3 carriers may achieve better glycemic control, possibly due to prolonged drug half-life [56,57]. This inconsistency in findings may stem from several factors, including study design heterogeneity, differences in hypoglycemia definitions, patient age, the specific SU assessed, or sample size limitations [58]. Despite these discrepancies, SU-induced hypoglycemia remains a current challenge in T2DM management, as it can lead to adverse effects ranging from mild discomfort or treatment withdrawal to serious morbidity [59].
Therefore, genotyping CYP2C9 variants specific to each population may be crucial, particularly during the initiation of SU therapy, in order to optimize treatment efficacy and reduce the risk of ADRs.

3.2.3. CYP2C19

In the inference analysis between the CYP2C19 gene and molecular ancestry, significant differences were observed only with the AFR ancestral component. Additionally, in the diplotype analysis, individuals carrying the CYP2C19 *1/*17 genotype exhibited a higher proportion of AFR ancestry compared to CYP2C19 *1/*1 and CYP2C19 *1/*2 individuals. These findings are consistent with previous studies reporting that the increased function CYP2C19*17 allele and this UM phenotype are negatively associated with NATAM ancestry, helping to explain the variability in allele frequencies across Latin America [27], and positively associated with EUR and AFR ancestry [38].
In the studied population, the frequencies of CYP2C19*2 and CYP2C19*17 were 11.14% and 6.25%, respectively. These frequencies support previous findings reporting a loss-of-function (*2) allele frequency of approximately 12% in Ecuador, and an increased-function (*17) allele frequency of 2.10% [60]. Other studies have reported slightly higher CYP2C19*17 frequencies (9.5%) in Ecuador [61], although these are still lower than those reported in European or African populations [62].
Based on CYP2C19 genotypes, the extrapolated phenotypic distribution was 1.35% PMs, 11.82% IMs, and approximately 20% UMs. Interestingly, the observed frequency of UMs is notably lower than previously reported values (41.4%), while the PM frequency was higher (0.7%) [42]. Although SUs are primarily metabolized by CYP2C9, CYP2C19 also contributes to their metabolism [15]. A study conducted in China proposed that CYP2C19 may play an even more significant role than CYP2C9 in the metabolism of gliclazide [63], suggesting increased efficacy and reduced ADRs. However, that study did not report the presence of CYP2C19*17 carriers [64].
According to CPIC guidelines for CYP2C19 metabolized drugs, such as clopidogrel [65], tricyclic antidepressants [66], and selective serotonin reuptake inhibitors (SSRIs) [67], the *1 allele is classified as normal function, *17 as increased function, and *2 and *4 as decreased function alleles. These guidelines provide drug dosing recommendations and strategies to minimize ADRs based on genotype-inferred phenotypes.
Despite the absence of pharmacogenetic guidelines for antidiabetic drugs, SU remains part of combination therapy with metformin when glycemic control is not adequately achieved [6]. Since these drugs are metabolized by highly polymorphic enzymes such as CYP2C9 and CYP2C19, variability in enzyme function can lead to reduced drug efficacy and the occurrence of ADRs, including severe secondary hypoglycemia [18].

3.3. Ancestry and OCTs

Our results from the analysis of ancestry distribution in variants of the OCTs show that the ancestral component is unevenly distributed among the genotypes of variants rs72552763, rs594709, and rs628031 of the SLC22A1 gene and the rs2076828 variant of the SLC22A3 gene. These findings contrast with previously obtained results by RIBEF collaboration in a study conducted in Mexican patients diagnosed with T2DM [28], where differences were only found in the distribution of the ancestry proportion among the genotypes of variants rs2076828 and rs628031.

3.3.1. SLC22A1

In the allelic correlation and ancestry proportion analysis, we found that the GAT allele of the rs72552763 variant is negatively correlated with NATAM ancestry and positively correlated with EUR ancestry. These findings are consistent with databases such as the 1000 Genomes Project, where the highest frequency of the deletion is found in populations from the Americas, specifically Mexicans from Los Angeles, California, and Peruvians from Lima [68], with results very similar to those in our sample and those reported in Mexican mestizos diagnosed with T2DM [28]. These findings suggest that populations in the Americas may have a higher prevalence of the variant allele of rs72552763, which in turn suggests possible pharmacogenetic implications. Some studies have reported implications in the pharmacokinetics of metformin associated with the presence of the del allele of rs72552763, among which are a decrease in the hepatic volume of distribution of metformin in del/del genotype carriers compared with carriers of the GAT allele [69], a decrease in metformin steady-state in T2DM patients carrying the variant, including rs72552763 [70], as well as pharmacokinetic alterations of other cationic drugs, such as decreased transport of ranitidine [71] and reduced clearance of morphine [72] observed in del/del genotype carriers. This suggests a possible decrease in OCT-1 transporter function in the presence of the del allele of rs72552763, which may have clinical implications, particularly in terms of therapeutic efficacy and the occurrence of adverse reactions associated with metformin or cationic drugs. Some studies have reported higher HbA1c levels in Mexican T2DM patients treated with metformin who carry the del allele [29]. Furthermore, in a longitudinal analysis conducted in Mexican T2DM patients treated with metformin monotherapy, the del allele has also been associated with a shorter time to HbA1c non-control compared with patients carrying the GAT/GAT genotype [73]. Similarly, it has been reported that patients carrying the GAT/del genotype have a higher risk of metformin-associated adverse reactions in Egyptian patients diagnosed with T2DM [74], where the frequency of the del allele is particularly low [68].
Our findings on rs594709 suggest a positive correlation between the presence of the A allele and NATAM ancestry and a negative correlation of the A allele with EUR ancestry. These findings are consistent with what has been reported in populations from the Americas, where the A allele is the most frequent (78%), especially in Mexicans and Peruvians, where this figure reaches 86.7% and 90.0%, respectively [68].
Studies conducted in diabetic patients considering the possible clinical implications of rs594709 are limited, and no association has been reported with the time to HbA1c non-control in Mexican T2DM patients [73]. However, in a study where an adjustment was made for another variant such as rs2289669, specifically with the G allele, it was found that the presence of the AA genotype of rs594709 and the G allele of rs2289669 may be associated with a decrease in fasting glucose, without having a significant impact on HbA1c [75]. Another study conducted in Mexican T2DM patients followed for 12 months found no differences in HbA1c levels among rs594709 genotypes; however, after adjusting for sex, time since diabetes diagnosis, and abdominal circumference, it was found that patients carrying the GG genotype showed higher HbA1c levels compared with carriers of the A allele [76]. Based on the above, although we found a significant correlation between NATAM, EUR ancestry, and the A allele of rs594709, the clinical implication of this variant in the clinical pharmacogenetics of diabetes may be modest.
The findings in this work regarding rs628031 suggest that the G allele is positively correlated with NATAM ancestry and negatively with EUR ancestry, consistent with what has been reported in the 1000 Genomes Project, as the population in the Americas has the highest prevalence of the G allele, reaching 88.3% and 90.0% in Mexicans and Peruvians, respectively [68]. Additionally, these frequencies are similar to those found in our study as well as in other studies conducted in the Mexican population [28].
The effect of the rs628031 variant on h-OCT1 transporter function has been studied, finding that the loss of transporter function with the presence of the rs628031 (M420V) variant is approximately 10% [77], conditioning a possible increase in metformin concentration, therapeutic efficacy, and adverse reactions in carriers of the A variant. However, no impact has been found on metformin Css (metformin steady-state concentration) among the different genotypes of rs628031 [78]. The clinical impact of this variant has been studied in Han Chinese patients treated with metformin, where AA carriers showed a greater reduction in HbA1c [79]. However, in a study conducted in diabetic patients from northern Mexico, it was found using an over-dominant model that carriers of GG+AA genotypes of rs628031 have higher HbA1c levels compared with GA carriers [80].

3.3.2. SLC22A2

In this study, we analyzed the rs316019 variant of SLC22A2; however, no significant correlation was found between rs316019 alleles and the proportion of NATAM, EUR, or AFR ancestry. According to data from the 1000 Genomes Project, the minor allele frequency in the American population, specifically in Mexico, is 5.5% and in Peru 5.3%, values comparable to that observed in the present work (3.5%) for the A allele of rs316019. In contrast, in Europe this frequency ranges between 6.1% and 13.1% [68]. Although OCT2 is responsible for most of the renal clearance of metformin [81], some studies have not found an association with the drug’s therapeutic response, or suggest that the effect may be small [18]. Another study reported that the AUC0–48h of metformin tends to be higher in individuals carrying the wild-type genotype compared to rs316019 heterozygotes; however, these differences were not statistically significant [82]. In contrast, other studies have identified an association between metformin therapeutic response and codominant models involving rs316019 of SLC22A2 and rs12943590 of SLC47A2, as these variants appear to influence HbA1c levels [83]. Nevertheless, the low prevalence of rs316019 and the modest effect reported in metformin users suggest that its relevance at the population level is limited.

3.3.3. SLC22A3

In this work, we found a positive correlation between the C allele of rs2076828 and NATAM ancestry, while the correlation was negative with EUR and AFR ancestry. This finding is consistent with what has been reported in the 1000 Genomes Project database, as the population with the highest frequency of the C allele belongs to the American continent [68].
Through an in vitro assay that used luciferase activity as a biomarker of gene expression, it was determined that the presence of the G allele of rs2076828 was associated with significantly lower luciferase activity compared with the reference C allele. In addition, it was observed that the minor G allele is associated with a reduced response to metformin in healthy subjects; however, the presence of the rs2076828 variant does not seem to have a significant effect on the pharmacokinetic parameters of metformin [84]. This may be because the OCT3 transporter is predominantly expressed in skeletal muscle, where, through increased AMP-Activated Protein Kinase (AMPK) activity in muscle, metformin improves glucose uptake and increases muscle glycogen [85]. In turn, OCT3 expression is moderate in the basolateral membrane of the proximal convoluted tubule, participating in metformin elimination; however, its role is less important than that of OCT2, which plays a key role in the excretion of pharmacological and endogenous substrates [86]. In this sense, it is possible that in populations with a high prevalence of the G allele, the therapeutic response to metformin may be reduced.

3.3.4. ABCB1

In this study, we analyzed three ABCB1 polymorphisms (rs1045642, rs2032582, and rs1128503) but found no evidence of correlation between the alleles or genotype distributions of these variants and the proportion of NATAM, EUR, or AFR ancestry. ABCB1 encodes P-gp, a transporter protein that facilitates the efflux of metformin [87]. However, to the best of our knowledge, the influence of these variants on the clinical efficacy of metformin remains limited. The absence of statistically significant differences in ancestry proportions, combined with the lack of correlation between these genotypes and ancestry, reinforces the notion that not all genetic variants with potential pharmacogenetic relevance necessarily have population-level or structural significance.
The characterization of genetic variants in Latin American populations associated with drug response is essential for identifying groups at risk of potential toxicity when administered specific medications. This knowledge is particularly valuable when translating clinical data from foreign populations to local contexts [88], as previous studies have reported associations between T2DM and non-European ancestry in Latino populations [89,90]. In fact, determining the frequencies of these molecular variants in Latin American populations, particularly in healthy individuals, will allow for the assessment of the safety and efficacy of antidiabetic drugs in these populations.

4. Materials and Methods

4.1. Study Design

This was an observational study conducted in accordance with the Declaration of Helsinki, and it was approved by the Bioethics Committee at the Universidad San Francisco de Quito (2017–077T–06/27/2017). All participants gave written informed consent.

4.2. Inclusion and Exclusion Criteria

The patients recruited for this study were diagnosed with T2DM and undergoing chronic treatment with SU or metformin. Patients of both genders aged between 35 and 65 years were included. Exclusion criteria were as follows: patients diagnosed with type 1 diabetes, as well as those with a recognized genetic syndrome of insulin resistance; chronic gastrointestinal diseases associated with malabsorption such as chronic pancreatitis, alcoholism, or drug abuse; pregnant women; kidney, liver, or thyroid disease, and concomitant treatment with corticosteroids or estrogens.

4.3. Data Collection

This study analyzed 297 patients with T2DM attending to the outpatient’s clinic at the “Dispensario Central”, which belongs to the Social Security Institute in Quito, Ecuador between September 2017 and August 2018.

4.4. Genotyping Procedure

The DNA was isolated and purified from blood samples using a QIAmp DNA extraction kit (Qiagen, Hilden, Germany). Genotyping for the different CYP450 and transport variants (Table S7: Analysis allelic variants with their respective PCR-TR probes across Ecuadorian T2DM patients (n = 297) was performed using commercially available genomic DNA Taqman® assays (Applied Biosystems, Foster City, CA, USA). Genotypes were assigned according to the presence of “key” SNVs associated with the relevant alleles (Table S7). All assays included positive (heterozygous and/or homozygous) and negative (no DNA) control samples from previous studies of our group. Plates were read with an ABI 7300 real-time PCR system (Applied Biosystems, Foster City, CA, USA), and the following thermocycling conditions were applied for all assays: 10 min for initial denaturation at 95 °C, followed by 40 denaturation cycles of 15 s at 92 °C and annealing at 60 °C for 1 min. Allele discrimination lasted 30 s at 60 °C. Genotype-based phenotype predictions were assigned to CYP2C8 and CYP2C9 as follows: individuals carrying either two non-functional alleles or one non-functional allele plus a reduced function allele were PMs, with an assigned activity score of 0–0.5. IMs were those individuals with either two reduced function alleles or a normal function allele plus a reduced function or non-functional allele, with an activity score of 1–1.5. NMs carry two normal function alleles, with an activity score of 2 [91]. There is no current activity score assignment consensus about CYP2C19. Individuals are rather classified according to their CYP2C19 genotype; however, we employed a classification of the different metabolizer phenotypes: PMs carry two non-functional alleles, activity score 0; IMs carry one normal function allele plus one non-functional allele, or an increased function allele plus one non-functional allele, activity score 1–1.5; NMs carry two normal function alleles, activity score 2; rapid (gRMs) and UMs carry one normal function allele plus an increased function allele or two increased function alleles, respectively. These two latter groups are integrated into a single UM with an activity score of >2 [65]. This phenotype group classification based on genotype and/or activity score has been developed from published CPIC [65,91].

4.5. Genomic Ancestry Analysis

Individual genomic ancestry was determined in 297 individuals from Ecuador. Out of these, 3 individuals did not register all 90 ancestry markers; thus, no ancestry proportion was determined thereby. The AFR, EUR, and NATAM components were inferred by genotyping 90 ancestry informative markers (AIMs) from the same panel used in previous studies [28,38] AIMs genotyping was performed at the National Genotyping Centre (CEGEN) at Santiago de Compostela, Spain, using the Sequenom (San Diego, CA, USA) platform. Individuals from the three parental populations were also inserted in the final database: 114 Spaniards and 296 Peruvian Native Americans from RIBEF-CEIBA [27] and 209 African Yoruba individuals from the 1000 Genomes Project [30]. The complete databases were transformed to ped map format using GLU 1.0b2 software (https://code.google.com/archive/p/glu-genetics/; accessed on 25 October 2024) and then analyzed in the Admixture software [92] in an unsupervised mode, assuming a tri-hybrid model (k = 3).

4.6. Statistical Analysis

The statistical analysis and figures were generated using R-4.2.0 (available at: https://www.R-project.org/; accessed on 18 February 2025). The analysis was carried out across three distinct phases: (i) descriptive, (ii) inferential, and (iii) correlation.

4.6.1. Descriptive Analysis

A description of allelic and genotypic frequencies was carried out for the variants under investigation. In the case of CYP2C8, CYP2C9, and CYP2C9, activity scores are also reported. The Hardy–Weinberg equilibrium was determined through Pearson’s Chi-squared test (Table S1: Allelic and genotypic frequencies and activity score distribution by CYP2C8, CYP2C9, and CYP2C19 within a sample of Ecuadorian T2DM patients (n = 297) and Table S2: SNV allelic and genotypic frequencies in SLC22A1, SLC22A2, SLC22A3, and ABCB1 (n = 297)).

4.6.2. Inferential Analysis

Ancestry proportion of NATAM, EUR, and AFR components were described and grouped according to the SNVs analyzed. In the case of CYP2C8, CYP2C9, and CYP2C19, ancestry proportions were grouped according to each activity score. Normality tests were performed through Shapiro–Wilk or Kolmogorov–Smirnov test. Inferential analyses were performed through Kruskal–Wallis test and Mann–Whitney’s U test for the case of 2 independent groups. Adjustment of p-values for multiple comparisons was performed employing the Bonferroni correction method and False Discovery Rate through Benjamini–Hochberg test. The significance value was p < 0.05.

4.6.3. Correlation Analysis

Across statistically significant transporters and cytochromes (CYP2C8, CYP2C9, CYP2C19, rs72552763, rs594709, rs628031, and rs2076828), Spearman’s rank correlation coefficient was determined by either the genotype’s allele or each individual’s diplotype (0, 0.5, and 1) and the ancestry proportion (NATAM, EUR, or AFR). The significance value was p < 0.05. A Spearman’s correlation analysis was performed between ancestry proportion and the activity score of CYP2C8, CYP2C9, and CYP2C19. A scatter plot with a regression line and 95% confidence interval was generated using the function lm() in stats package (v4.2.0), through method = (lm) from geom_smooth in ggplot2 (v3.5.1). Spearman’s coefficient correlation was reported, and the significance value was p < 0.05.

5. Conclusions

Among the studied Ecuadorian population of T2DM patients, the proportion of NATAM ancestry (61.3%) is higher than the EUR (34.5%) or AFR component, which is of very little relevance (2.6%%). Regarding the relevance of this ancestral component of the native population of Ecuador (NATAM), CYP2C8 and CYP2C9 (*1/*1 diplotype) and activity score of 2 have been related. No reference has been found for CYP2C19 (which does appear for the AFR component). In relation to the transport-related NATAM ancestry, it has been related to SLC22A1 (rs594709 and rs628031) and SLC22A3 (rs2076828) genes. An association was not found between SLC22A2 and ABCB1 with NATAM ancestry.
The results of this study highlight the relevance of considering genomic ancestry in the implementation of clinical pharmacogenetics for T2DM to ensure patients receive safe and effective antidiabetic treatment. By identifying differences among genotypes of the main drug-metabolizing enzymes and transporters involved in antidiabetic drug disposition, and by detecting a correlation between the distribution of these variants and the assignment of the activity score for drugs such as metformin, TZDs, and SUs, this study underscores the importance of a personalized medicine approach to T2DM in Ecuador.

The Limitations of the Study

In future studies, we aim to analyze the clinical relevance in patients diagnosed with T2DM by collecting clinical, biochemical, and pharmacological parameters, which were not included in the present study. Furthermore, we intend to increase the study sample size by incorporating diverse ethnic groups from across Ecuador, rather than limiting the population to patients from Quito.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph18091335/s1, Table S1. Allelic and genotypic frequencies and activity score distribution by CYP2C8, CYP2C9, and CYP2C19 within a sample of Ecuadorian T2DM patients (n = 297). Table S2. SNV allelic and genotypic frequencies in SLC22A1, SLC22A2, SLC22A3, and ABCB1 (n = 297). Table S3. Correlation between ancestry proportion and allelic frequency in CYP2C8 variants. Table S4. Correlation between ancestry proportion and allelic frequency in CYP2C9 variants Table S5. Correlation between ancestry proportion and allelic frequency in CYP2C19 variants Table S6. Correlation between ancestry proportion and allelic frequency in SNVs in SLC22A1 and SLC22A3. Table S7. Analysis of allelic variants with their respective Taqman PCR-RT assays, enzymatic activity.

Author Contributions

Conceptualization, A.L. and E.T.; methodology, A.L., E.T., P.D., and F.R.-S.; software, A.O.-A.; validation, P.D., F.R.-S., A.O.-A., C.G.d.l.C., L.M., M.B. and L.T.; formal analysis, P.D., F.R.-S. and A.O.-A.; investigation, A.O.-A., C.G.d.l.C., L.M., M.B. and L.T.; resources, C.G.d.l.C., L.M., M.B. and L.T.; data curation, P.D., F.R.-S., A.O.-A., L.M., M.B. and L.T.; writing—original draft preparation, P.D., A.O.-A. and C.G.d.l.C.; writing—review and editing, A.O.-A., C.G.d.l.C., F.R.-S., P.D., E.T. and A.L.; visualization, A.O.-A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Agencia Extremeña de Cooperación Internacional para el Desarrollo (AEXCID) grant number 24IA001 (Junta de Extremadura, Spain); by Universidad San Francisco de Quito grant number HUBI 5459; and F.R.S. was supported by Conselho Nacional de Desenvolvimento Científico e Tecnológico (Brazil) grant number 200824/2024-4.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Bioethics Committee at the Universidad San Francisco de Quito (2017-077T–06/27/2017).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The original contributions presented in this study are included in the article/Supplementary Material. Further inquiries can be directed to the corresponding authors.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ABCB1ATP Binding Cassette Subfamily B Member 1
ADRsAdverse drug reactions
AFRAfrican
AIMsAncestry informative markers
CPICClinical Pharmacogenetics Implementation Consortium Guidelines
CYP450Cytochrome P450
T2DMType 2 diabetes mellitus
EUREuropean
IMIntermediate metabolizer
NATAMNative American
NMNormal metabolizer
OCTOrganic cation transporters
PMPoor metabolizer
P-gpP-glycoprotein
SNVSingle nucleotide allelic variant
SUSulfonylureas
TZDThiazolidinediones

References

  1. Puig-García, M.; Caicedo-Montaño, C.; Márquez-Figueroa, M.; Chilet-Rosell, E.; Montalvo-Villacis, G.; Benazizi-Dahbi, I.; Peralta, A.; Torres-Castillo, A.L.; Parker, L.A. Prevalence and gender disparities of type 2 diabetes mellitus and obesity in Esmeraldas, Ecuador: A population-based survey in a hard-to-reach setting. Int. J. Equity Health 2023, 22, 124. [Google Scholar] [CrossRef]
  2. Global Clinical Practice Recommendations-International Diabetes Federation. Available online: https://idf.org/what-we-do/education/idf-clinical-practice-recommendations-for-type-2-diabetes-2025/ (accessed on 18 July 2025).
  3. López-Jaramillo, P.; Barbosa, E.; Molina, D.I.; Sanchez, R.; Diaz, M.; Camacho, P.A.; Lanas, F.; Pasquel, M.; Accini, J.L.; Ponte-Negretti, C.I.; et al. Latin American Consensus on the Management of Hypertension in the Patient with Diabetes and the Metabolic Syndrome. J. Hypertens. 2019, 37, 1126–1147. [Google Scholar] [CrossRef]
  4. Ecuador Refuerza su Compromiso en la Lucha Contra la Diabetes–Ministerio de Salud Pública. Available online: https://www.salud.gob.ec/ecuador-refuerza-su-compromiso-en-la-lucha-contra-la-diabetes/ (accessed on 18 July 2025).
  5. Martin-Delgado, J.; Tovar, C.; Pazmiño, I.; Briones, A.; Carrillo, I.; Guilabert, M.; Mira, J.J. Indicators for Adequate Diabetes Care for the Indigenous Communities of Ecuador. Health Expect. 2022, 25, 3315–3325. [Google Scholar] [CrossRef]
  6. American Diabetes Association Professional Practice Committee. Pharmacologic Approaches to Glycemic Treatment: Standards of Care in Diabetes. Diabetes Care 2025, 48 (Suppl. S1), S181–S206. [Google Scholar] [CrossRef] [PubMed]
  7. Zhou, Y.; Lauschke, V.M. Population Pharmacogenomics: An Update on Ethnogeographic Differences and Opportunities for Precision Public Health. Hum. Genet. 2022, 141, 1113–1136. [Google Scholar] [CrossRef] [PubMed]
  8. Eichelbaum, M.; Ingelman-Sundberg, M.; Evans, W.E. Pharmacogenomics and individualized drug therapy. Annu. Rev. Med. 2006, 57, 119–137. [Google Scholar] [CrossRef] [PubMed]
  9. Koyama, T.; Iinuma, S.; Yamamoto, M.; Niimura, T.; Osaki, Y.; Nishimura, S.; Harada, K.; Zamami, Y.; Hagiya, H. International Trends in Adverse Drug Event-Related Mortality from 2001 to 2019: An Analysis of the World Health Organization Mortality Database from 54 Countries. Drug Saf. 2024, 47, 237–249. [Google Scholar] [CrossRef]
  10. Ducoffe, A.R.; York, A.; Hu, D.J.; Perfetto, D.; Kerns, R.D. National action plan for adverse drug event prevention: Recommendations for safer outpatient opioid use. Pain Med. 2016, 17, 2291–2304. [Google Scholar] [CrossRef][Green Version]
  11. Damanhouri, Z.A.; Alkreathy, H.M.; Alharbi, F.A.; Abualhamail, H.; Ahmad, M.S. A Review of the Impact of Pharmacogenetics and Metabolomics on the Efficacy of Metformin in Type 2 Diabetes. Int. J. Med. Sci. 2023, 20, 142–150. [Google Scholar] [CrossRef]
  12. Peng, A.; Gong, C.; Xu, Y.; Liang, X.; Chen, X.; Hong, W.; Yan, J. Association between Organic Cation Transporter Genetic Polymorphisms and Metformin Response and Intolerance in T2DM Individuals: A Systematic Review and Meta-Analysis. Front. Public Health 2023, 11, 1183879. [Google Scholar] [CrossRef]
  13. Dujic, T.; Zhou, K.; Donnelly, L.A.; Tavendale, R.; Palmer, C.N.A.; Pearson, E.R. Association of organic cation transporter 1 with intolerance to metformin in type 2 diabetes: A GoDARTS study. Diabetes 2015, 64, 1786–1793. [Google Scholar] [CrossRef] [PubMed]
  14. Dujic, T.; Causevic, A.; Bego, T.; Malenica, M.; Velija-Asimi, Z.; Pearson, E.R.; Semiz, S. Organic Cation Transporter 1 Variants and Gastrointestinal Side Effects of Metformin in Patients with Type 2 Diabetes. Diabet. Med. 2016, 33, 511–551. [Google Scholar] [CrossRef]
  15. Wang, K.; Yang, A.; Shi, M.; Tam, C.C.H.; Lau, E.S.H.; Fan, B.; Lim, C.K.P.; Lee, H.M.; Kong, A.P.S.; Luk, A.O.Y.; et al. CYP2C19 Loss-of-function Polymorphisms Are Associated with Reduced Risk of Sulfonylurea Treatment Failure in Chinese Patients with Type 2 Diabetes. Clin. Pharmacol. Ther. 2022, 111, 461–469. [Google Scholar] [CrossRef] [PubMed]
  16. Dawed, A.Y.; Donnelly, L.; Tavendale, R.; Carr, F.; Leese, G.; Palmer, C.N.; Pearson, E.R.; Zhou, K. CYP2C8 and SLCO1B1 Variants and Therapeutic Response to Thiazolidinediones in Patients With Type 2 Diabetes. Diabetes Care 2016, 39, 1902–1908. [Google Scholar] [CrossRef]
  17. Gökalp, O.; Gunes, A.; Cam, H.; Cure, E.; Aydın, O.; Tamer, M.N.; Scordo, M.G.; Dahl, M.L. Mild hypoglycaemic attacks induced by sulphonylureas related to CYP2C9, CYP2C19 and CYP2C8 polymorphisms in routine clinical setting. Eur. J. Clin. Pharmacol. 2011, 67, 1223–1229. [Google Scholar] [CrossRef]
  18. Dujic, T.; Zhou, K.; Donnelly, L.A.; Leese, G.; Palmer, C.N.A.; Pearson, E.R. Interaction between variants in the CYP2C9 and POR genes and the risk of sulfonylurea-induced hypoglycaemia: A GoDARTS Study. Diabetes Obes. Metab. 2018, 20, 211–214. [Google Scholar] [CrossRef]
  19. Baye, A.M.; Fanta, T.G.; Siddiqui, M.K.; Dawed, A.Y. The Genetics of Adverse Drug Outcomes in Type 2 Diabetes: A Systematic Review. Front. Genet. 2021, 12, 675053. [Google Scholar] [CrossRef]
  20. Terán, E.; Cuautle, P.; Tana, L.; Rodríguez, N.; Bonilla, M.; Molina, J.; Llerena, A. Farmacogenética de la Diabetes Mellitus en Latinoamérica: Una Perspectiva desde la Red Iberoamericana de Farmacogenética. In Farmacogenómica y Medicina Personalizada en Latinoamérica, 1st ed.; Quiñonez, L., Redal, M.A., Eds.; Editorial Académica Española: London, UK, 2020; pp. 443–452. [Google Scholar]
  21. Peñas-Lledó, E.; Terán, E.; Sosa-Macías, M.; Galaviz-Hernández, C.; Gil, J.P.; Nair, S.; Diwakar, S.; Hernández, I.; Lara-Riegos, J.; Ramírez-Roa, R.; et al. Challenges and Opportunities for Clinical Pharmacogenetic Research Studies in Resource-limited Settings: Conclusions From the Council for International Organizations of Medical Sciences-Ibero-American Network of Pharmacogenetics and Pharmacogenomics Meeting. Clin. Ther. 2020, 42, 1595–1610.e5. [Google Scholar]
  22. Sosa-Macías, M.; Teran, E.; Waters, W.; Fors, M.M.; Altamirano, C.; Jung-Cook, H.; Galaviz-Hernández, C.; López-López, M.; Remírez, D.; Moya, G.E.; et al. Pharmacogenetics and ethnicity: Relevance for clinical implementation, clinical trials, pharmacovigilance and drug regulation in Latin America. Pharmacogenomics 2016, 17, 1741–1747. [Google Scholar] [CrossRef]
  23. Lteif, C.; Gawronski, B.E.; Cicali, E.J.; Martinez, K.A.; Newsom, K.J.; Starostik, P.; Cavallari, L.H.; Duarte, J.D. Development of an Ancestrally Inclusive Preemptive Pharmacogenetic Testing Panel. Clin. Transl. Sci. 2025, 18, e70230. [Google Scholar] [CrossRef] [PubMed]
  24. Hernandez, S.; Hindorff, L.A.; Morales, J.; Ramos, E.M.; Manolio, T.A. Patterns of pharmacogenetic variation in nine biogeographic groups. Clin. Transl. Sci. 2024, 17, e70017. [Google Scholar] [CrossRef] [PubMed]
  25. Santangelo, R.; González-Andrade, F.; Børsting, C.; Torroni, A.; Pereira, V.; Morling, N. Analysis of ancestry informative markers in three main ethnic groups from Ecuador supports a trihybrid origin of Ecuadorians. Forensic Sci. Int. Genet. 2017, 31, 29–33. [Google Scholar] [CrossRef]
  26. Naranjo, M.G.; Rodrigues-Soares, F.; Peñas-Lledó, E.M.; Tarazona-Santos, E.; Fariñas, H.; Rodeiro, I.; Terán, E.; Grazina, M.; Moya, G.E.; López-López, M.; et al. CEIBA-Consortium of the Ibero-American Network of Pharmacogenetics and Pharmacogenomics RIBEF, Interethnic Variability in CYP2D6, CYP2C9, and CYP2C19 Genes and Predicted Drug Metabolism Phenotypes Among 6060 Ibero- and Native Americans: RIBEF-CEIBA Consortium Report on Population Pharmacogenomics. Omics A J. Integr. Biol. 2018, 22, 575–588. [Google Scholar]
  27. Rodrigues-Soares, F.; Peñas-Lledó, E.M.; Tarazona-Santos, E.; Sosa-Macías, M.; Terán, E.; López-López, M.; Rodeiro, I.; Moya, G.E.; Calzadilla, L.R.; Ramírez-Roa, R.; et al. RIBEF Ibero-American Network of Pharmacogenetics and Pharmacogenomics, Genomic Ancestry, CYP2D6, CYP2C9, and CYP2C19 Among Latin Americans. Clin. Pharmacol. Ther. 2020, 107, 257–268. [Google Scholar] [CrossRef] [PubMed]
  28. Ortega-Ayala, A.; de la Cruz, C.G.; Dorado, P.; Rodrigues-Soares, F.; Castillo-Nájera, F.; LLerena, A.; Molina-Guarneros, J. Molecular Ancestry Across Allelic Variants of SLC22A1, SLC22A2, SLC22A3, ABCB1, CYP2C8, CYP2C9, and CYP2C19 in Mexican-Mestizo DMT2 Patients. Biomedicines 2025, 13, 1156. [Google Scholar] [CrossRef]
  29. Ortega-Ayala, A.; De Andrés, F.; Llerena, A.; Bartolo-Montiel, C.M.; Molina-Guarneros, J.A. Impact of SLC22A1 variants rs622342 and rs72552763 on HbA1c and metformin plasmatic concentration levels in patients with type 2 diabetes mellitus. Biomed. Rep. 2024, 21, 117. [Google Scholar] [CrossRef]
  30. 1000 Genomes Project Consortium; Auton, A.; Brooks, L.D.; Durbin, R.M.; Garrison, E.P.; Kang, H.M.; Korbel, J.O.; Marchini, J.L.; McCarthy, S.; McVean, G.A.; et al. A global reference for human genetic variation. Nature 2015, 526, 68–74. [Google Scholar] [CrossRef]
  31. Peñas-Lledó, E.M.; Guillaume, S.; de Andrés, F.; Cortés-Martínez, A.; Dubois, J.; Kahn, J.P.; Leboyer, M.; Olié, E.; LLerena, A.; Courtet, P. A one-year follow-up study of treatment-compliant suicide attempt survivors: Relationship of CYP2D6-CYP2C19 and polypharmacy with suicide reattempts. Transl. Psychiatry 2022, 12, 451. [Google Scholar] [CrossRef]
  32. Homburger, J.R.; Moreno-Estrada, A.; Gignoux, C.R.; Nelson, D.; Sanchez, E.; Ortiz-Tello, P.; Pons-Estel, B.A.; Acevedo-Vasquez, E.; Miranda, P.; Langefeld, C.D.; et al. Genomic insights into the ancestry and demographic history of South America. PLoS Genet. 2015, 11, e1005602. [Google Scholar] [CrossRef]
  33. Farinango, C.; Gallardo-Cóndor, J.; Freire-Paspuel, B.; Flores-Espinoza, R.; Jaramillo-Koupermann, G.; López-Cortés, A.; Burgos, G.; Tejera, E.; Cabrera-Andrade, A. Genetic variations of the DPYD gene and its relationship with ancestry proportions in different Ecuadorian trihybrid populations. J. Pers. Med. 2022, 12, 950. [Google Scholar] [CrossRef]
  34. Nagar, S.D.; Conley, A.B.; Chande, A.T.; Rishishwar, L.; Sharma, S.; Mariño-Ramírez, L.; Aguinaga-Romero, G.; González-Andrade, F.; Jordan, I.K. Genetic ancestry and ethnic identity in Ecuador. HGG Adv. 2021, 2, 100050. [Google Scholar] [CrossRef] [PubMed]
  35. Sandoval, J.R.; Salazar-Granara, A.; Acosta, O.; Castillo-Herrera, W.; Fujita, R.; Pena, S.D.; Santos, F.R. Tracing the genomic ancestry of Peruvians reveals a major legacy of pre-Columbian ancestors. J. Hum. Genet. 2013, 58, 627–634. [Google Scholar] [CrossRef]
  36. Mariño-Ramírez, L.; Sharma, S.; Hamilton, J.M.; Nguyen, T.L.; Gupta, S.; Natarajan, A.V.; Nagar, S.D.; Menuey, J.L.; Chen, W.A.; Sánchez-Gómez, A.; et al. The Consortium for Genomic Diversity, Ancestry, and Health in Colombia (CÓDIGO), building local capacity in genomics, bioinformatics, and precision medicine. bioRxiv 2025. [Google Scholar] [CrossRef]
  37. Pena, S.D.J.; Santos, F.R.; Tarazona-Santos, E. Genetic admixture in Brazil. Am. J. Med. Genet. Part C Semin. Med. Genet. 2020, 184, 928–938. [Google Scholar] [CrossRef]
  38. Guevara, M.; Rodrigues-Soares, F.; de la Cruz, C.G.; de Andrés, F.; Rodríguez, E.; Peñas-Lledó, E.; LLerena, A. CEIBA Consortium of the Ibero-American Network of Pharmacogenetics and Pharmacogenomics RIBEF. Afro-Latin American Pharmacogenetics of CYP2D6, CYP2C9, and CYP2C19 in Dominicans: A Study from the RIBEF-CEIBA Consortium. Pharmaceutics 2024, 16, 1399. [Google Scholar] [CrossRef]
  39. Nieves-Colón, M.A.; Ulrich, E.C.; Chen, L.; Torres Colón, G.A.; Rivera Clemente, M.; La Corporación Piñones Se Integra (COPI); Benn Torres, J. Genetic ancestry in Puerto Rican Afro-descendants illustrates diverse histories of African diasporic populations. Am. J. Biol. Anthropol. 2024, 185, e25029. [Google Scholar] [CrossRef]
  40. Sosa-Macías, M.; Fricke-Galindo, I.; Fariñas, H.; Monterde, L.; Ruiz-Cruz, E.D.; Molina-Guarneros, J.; Tarazona-Santos, E.; Rodrigues-Soares, F.; Galaviz-Hernández, C.; Peñas-Lledó, E.; et al. Pharmacogenetics, ethnicity, treatment and health in Latin American populations. Pharmacogenomics 2023, 24, 489–492. [Google Scholar] [CrossRef]
  41. Zhang, J.; Litvinova, M.; Liang, Y.; Wang, Y.; Wang, W.; Zhao, S.; Wu, Q.; Merler, S.; Viboud, C.; Vespignani, A.; et al. Changes in contact patterns shape the dynamics of the COVID-19 outbreak in China. Science 2020, 368, 1481–1486. [Google Scholar] [CrossRef] [PubMed]
  42. Vicente, J.; González-Andrade, F.; Soriano, A.; Fanlo, A.; Martínez-Jarreta, B.; Sinués, B. Genetic polymorphisms of CYP2C8, CYP2C9 and CYP2C19 in Ecuadorian Mestizo and Spaniard populations: A comparative study. Mol. Biol. Rep. 2014, 41, 1267–1272. [Google Scholar] [CrossRef] [PubMed]
  43. ‘Pharmfreq’. Available online: https://www.pharmfreq.com/?_inputs_&gene=%22CYP2C8%22&allele=%22*3%22&go=1&mapping_method=%22Countries%22 (accessed on 8 July 2025).
  44. Chan, A.T.; Zauber, A.G.; Hsu, M.; Breazna, A.; Hunter, D.J.; Rosenstein, R.B.; Eagle, C.J.; Hawk, E.T.; Bertagnolli, M.M. Cytochrome P450 2C9 variants influence response to celecoxib for prevention of colorectal adenoma. Gastroenterology 2009, 136, 2127–2136.e1. [Google Scholar] [CrossRef]
  45. Theken, K.N.; Lee, C.R.; Gong, L.; Caudle, K.E.; Formea, C.M.; Gaedigk, A.; Klein, T.E.; Agúndez, J.A.G.; Grosser, T. Clinical Pharmacogenetics Implementation Consortium Guideline (CPIC) for CYP2C9 and Nonsteroidal Anti-Inflammatory Drugs. Clin. Pharmacol. Ther. 2020, 108, 191–200. [Google Scholar] [CrossRef]
  46. Dorado, P.; Cavaco, I.; Cáceres, M.C.; Piedade, R.; Ribeiro, V.; LLerena, A. Relationship between CYP2C8 genotypes and diclofenac 5-hydroxylation in healthy Spanish volunteers. Eur. J. Clin. Pharmacol. 2008, 64, 967–970. [Google Scholar] [CrossRef]
  47. Ochoa, D.; Prieto-Pérez, R.; Román, M.; Talegón, M.; Rivas, A.; Galicia, I.; Abad-Santos, F. Effect of Gender and CYP2C9 and CYP2C8 Polymorphisms on the Pharmacokinetics of Ibuprofen Enantiomers. Pharmacogenomics 2015, 16, 939–948. [Google Scholar] [CrossRef]
  48. Campodónico, D.M.; Zubiaur, P.; Soria-Chacartegui, P.; Casajús, A.; Villapalos-García, G.; Navares-Gómez, M.; Gómez-Fernández, A.; Parra-Garcés, R.; Mejía-Abril, G.; Román, M.; et al. CYP2C8*3 and *4 Define CYP2C8 Phenotype: An Approach with the Substrate Cinitapride. Clin. Transl. Sci. 2022, 15, 2613–2624. [Google Scholar] [CrossRef]
  49. Céspedes-Garro, C.; Fricke-Galindo, I.; Naranjo, M.E.; Rodrigues-Soares, F.; Fariñas, H.; de Andrés, F.; López-López, M.; Peñas-Lledó, E.M.; LLerena, A. Worldwide interethnic variability and geographical distribution of CYP2C9 genotypes and phenotypes. Expert. Opin. Drug Metab. Toxicol. 2015, 11, 1893–1905. [Google Scholar] [CrossRef] [PubMed]
  50. Zhou, Y.; Nevosadová, L.; Eliasson, E.; Lauschke, V.M. Global distribution of functionally important CYP2C9 alleles and their inferred metabolic consequences. Hum. Genom. 2023, 17, 4–13. [Google Scholar] [CrossRef]
  51. Dorado, P.; Beltrán, L.J.; MacHín, E.; Peñas-Lledó, E.M.; Terán, E.; Llerena, A. Losartan hydroxylation phenotype in an Ecuadorian population: Influence of CYP2C9 genetic polymorphism, habits and gender. Pharmacogenomics 2012, 13, 1711–1717. [Google Scholar] [CrossRef] [PubMed]
  52. Karnes, J.H.; Rettie, A.E.; Somogyi, A.A.; Huddart, R.; Fohner, A.E.; Formea, C.M.; Ta Michael Lee, M.; Llerena, A.; Whirl-Carrillo, M.; Klein, T.E.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2C9 and HLA-B Genotypes and Phenytoin Dosing: 2020 Update. Clin. Pharmacol. Ther. 2021, 109, 302–309. [Google Scholar] [CrossRef]
  53. Johnson, J.A.; Caudle, K.E.; Gong, L.; Whirl-Carrillo, M.; Stein, C.M.; Scott, S.A.; Lee, M.T.; Gage, B.F.; Kimmel, S.E.; Perera, M.A.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Pharmacogenetics-Guided Warfarin Dosing: 2017 Update. Clin. Pharmacol. Ther. 2017, 102, 397–404. [Google Scholar] [CrossRef] [PubMed]
  54. Yee, J.; Heo, Y.; Kim, H.; Yoon, H.Y.; Song, G.; Gwak, H.S. Association Between the CYP2C9 Genotype and Hypoglycemia Among Patients With Type 2 Diabetes Receiving Sulfonylurea Treatment: A Meta-analysis. Clin. Ther. 2021, 43, 836–843.e4. [Google Scholar] [CrossRef]
  55. Koren, S.; Koren, R. Any Polymorphisms of CYP2C9 Affects the Biochemical Profile of Diabetic Patients Receiving Glibenclamide. Clin. Med. Biochem. Open Access 2015, 1, 1–4. [Google Scholar] [CrossRef]
  56. Salam, R.F.A.; Zeyada, R.; Osman, N.A. Effect of CYP2C9 gene polymorphisms on response to treatment with sulfonylureas in a cohort of Egyptian type 2 diabetes mellitus patients. Comp. Clin. Path 2014, 23, 341–346. [Google Scholar] [CrossRef]
  57. Castelán-Martínez, O.D.; Hoyo-Vadillo, C.; Bazán-Soto, T.B.; Cruz, M.; Tesoro-Cruz, E.; Valladares-Salgado, A. CYP2C9*3 gene variant contributes independently to glycaemic control in patients with type 2 diabetes treated with glibenclamide. J. Clin. Pharm. Ther. 2018, 43, 768–774. [Google Scholar] [CrossRef] [PubMed]
  58. Jan, A.; Saeed, M.; Mothana, R.A.; Muhammad, T.; Rahman, N.; Alanzi, A.R.; Akbar, R. Association of CYP2C9*2 Allele with Sulphonylurea-Induced Hypoglycaemia in Type 2 Diabetes Mellitus Patients: A Pharmacogenetic Study in Pakistani Pashtun Population. Biomedicines 2023, 11, 2282. [Google Scholar] [CrossRef]
  59. Scheen, A.J. Sulphonylureas in the management of type 2 diabetes: To be or not to be? Diabetes Epidemiol. Manag. 2021, 1, 100002. [Google Scholar] [CrossRef]
  60. Alonso Llorente, A.; Salgado Garrido, J.; Teijido Hermida, Ó.; González Andrade, F.; Valiente Martín, A.; Fanlo Villacampa, A.J.; Vicente Romero, J. Genetic Polymorphisms of CYP2C19 in Ecuadorian Population: An Interethnic Approach. Heliyon 2024, 10, e28566. [Google Scholar] [CrossRef]
  61. De Andrés, F.; Terán, S.; Hernández, F.; Terán, E.; Llerena, A. To Genotype or Phenotype for Personalized Medicine? CYP450 Drug Metabolizing Enzyme Genotype-Phenotype Concordance and Discordance in the Ecuadorian Population. Omi. A J. Integr. Biol. 2016, 20, 699–710. [Google Scholar] [CrossRef]
  62. Fricke-Galindo, I.; Jung-Cook, H.; Llerena, A.; López-López, M. Interethnic Variability of Pharmacogenetic Biomarkers in Mexican Healthy Volunteers: A Report from the RIBEF (Ibero-American Network of Pharmacogenetics and Pharmacogenomics). Drug Metab. Pers. Ther. 2016, 31, 61–81. [Google Scholar] [CrossRef]
  63. Zhang, Y.; Si, D.; Chen, X.; Lin, N.; Guo, Y.; Zhou, H.; Zhong, D. Influence of CYP2C9 and CYP2C19 Genetic Polymorphisms on Pharmacokinetics of Gliclazide MR in Chinese Subjects. Br. J. Clin. Pharmacol. 2007, 64, 67–74. [Google Scholar] [CrossRef]
  64. Tan, B.; Zhang, Y.F.; Chen, X.Y.; Zhao, X.H.; Li, G.X.; Zhong, D.F. The effects of CYP2C9 and CYP2C19 genetic polymorphisms on the pharmacokinetics and pharmacodynamics of glipizide in Chinese subjects. Eur. J. Clin. Pharmacol. 2010, 66, 145–151. [Google Scholar] [CrossRef]
  65. Lee, C.R.; Luzum, J.A.; Sangkuhl, K.; Gammal, R.S.; Sabatine, M.S.; Stein, C.M.; Kisor, D.F.; Limdi, N.A.; Lee, Y.M.; Scott, S.A.; et al. Clinical Pharmacogenetics Implementation Consortium Guideline for CYP2C19 Genotype and Clopidogrel Therapy: 2022 Update. Clin. Pharmacol. Ther. 2022, 112, 959–967. [Google Scholar] [CrossRef] [PubMed]
  66. Hicks, J.K.; Sangkuhl, K.; Swen, J.J.; Ellingrod, V.L.; Müller, D.J.; Shimoda, K.; Bishop, J.R.; Kharasch, E.D.; Skaar, T.C.; Gaedigk, A.; et al. Clinical Pharmacogenetics Implementation Consortium Guideline (CPIC) for CYP2D6 and CYP2C19 Genotypes and Dosing of Tricyclic Antidepressants: 2016 Update. Clin. Pharmacol. Ther. 2017, 102, 37–44. [Google Scholar] [CrossRef]
  67. Bousman, C.A.; Stevenson, J.M.; Ramsey, L.B.; Sangkuhl, K.; Hicks, J.K.; Strawn, J.R.; Singh, A.B.; Ruaño, G.; Mueller, D.J.; Tsermpini, E.E.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2D6, CYP2C19, CYP2B6, SLC6A4, and HTR2A Genotypes and Serotonin Reuptake Inhibitor Antidepressants. Clin. Pharmacol. Ther. 2023, 114, 51–68. [Google Scholar] [CrossRef]
  68. Dyer, S.C.; Austine-Orimoloye, O.; Azov, A.G.; Barba, M.; Barnes, I.; Barrera-Enriquez, V.P.; Becker, A.; Bennett, R.; Beracochea, M.; Berry, A.; et al. Ensembl 2025. Nucleic Acids Res. 2025, 53, D948–D957. [Google Scholar] [CrossRef]
  69. Sundelin, E.; Gormsen, L.C.; Jensen, J.B.; Vendelbo, M.H.; Jakobsen, S.; Munk, O.L.; Christensen, M.; Brøsen, K.; Frøkiaer, J.; Jessen, N. Genetic Polymorphisms in Organic Cation Transporter 1 Attenuates Hepatic Metformin Exposure in Humans. Clin. Pharmacol. Ther. 2017, 102, 841–848. [Google Scholar] [CrossRef]
  70. Christensen, M.M.H.; Brasch-Andersen, C.; Green, H.; Nielsen, F.; Damkier, P.; Beck-Nielsen, H.; Brosen, K. The pharmacogenetics of metformin and its impact on plasma metformin steady-state levels and glycosylated hemoglobin A1c. Pharmacogenet. Genom. 2011, 21, 837–850. [Google Scholar] [CrossRef]
  71. Meyer, M.J.; Seitz, T.; Brockmöller, J.; Tzvetkov, M.V. Effects of genetic polymorphisms on the OCT1 and OCT2-mediated uptake of ranitidine. PLoS ONE 2017, 12, e0189521. [Google Scholar] [CrossRef]
  72. Fukuda, T.; Chidambaran, V.; Mizuno, T.; Venkatasubramanian, R.; Ngamprasertwong, P.; Olbrecht, V.; Esslinger, H.R.; Vinks, A.A.; Sadhasivam, S. OCT1 Genetic Variants Influence the Pharmacokinetics of Morphine in Children. Pharmacogenomics 2013, 14, 1141–1151. [Google Scholar] [CrossRef] [PubMed]
  73. Ortega-Ayala, A.; De Andrés, F.; Llerena, A.; Bartolo-Montiel, C.M.; Acosta-Altamirano, G.; Molina-Guarneros, J.A. Longitudinal Assessment of SNPs rs72552763 and rs622342 in SLC22A1 over HbA1c Control among Mexican-Mestizo Diabetic Type 2 Patients. Front. Pharmacol. 2024, 15, 1433519. [Google Scholar] [CrossRef]
  74. Ahmed, A.; Elsadek, H.M.; Shalaby, S.M.; Elnahas, H.M. Association of SLC22A1, SLC47A1, and KCNJ11 polymorphisms with efficacy and safety of metformin and sulfonylurea combination therapy in Egyptian patients with type 2 diabetes. Res. Pharm. Sci. 2023, 18, 614–625. [Google Scholar] [CrossRef] [PubMed]
  75. Xiao, D.; Guo, Y.; Li, X.; Yin, J.Y.; Zheng, W.; Qiu, X.W.; Xiao, L.; Liu, R.R.; Wang, S.Y.; Gong, W.J.; et al. The Impacts of SLC22A1 rs594709 and SLC47A1 rs2289669 Polymorphisms on Metformin Therapeutic Efficacy in Chinese Type 2 Diabetes Patients. Int. J. Endocrinol. 2016, 2016, 4350712. [Google Scholar] [CrossRef] [PubMed]
  76. Reséndiz-Abarca, C.A.; Flores-Alfaro, E.; Suárez-Sánchez, F.; Cruz, M.; Valladares-Salgado, A.; Del Carmen Alarcón-Romero, L.; Vázquez-Moreno, M.A.; Wacher-Rodarte, N.A.; Gómez-Zamudio, J.H. Altered Glycemic Control Associated with Polymorphisms in the SLC22A1 (OCT1) Gene in a Mexican Population with Type 2 Diabetes Mellitus Treated with Metformin: A Cohort Study. J. Clin. Pharmacol. 2019, 59, 1384–1390. [Google Scholar] [CrossRef]
  77. Zhang, S.; Zhu, A.; Kong, F.; Chen, J.; Lan, B.; He, G.; Gao, K.; Cheng, L.; Sun, X.; Yan, C.; et al. Structural Insights into Human Organic Cation Transporter 1 Transport and Inhibition. Cell Discov. 2024, 10, 30. [Google Scholar] [CrossRef]
  78. Ningrum, V.D.A.; Sadewa, A.H.; Ikawati, Z.; Yuliwulandari, R.; Ikhsan, M.R.; Fajriyah, R. The influence of metformin transporter gene SLC22A1 and SLC47A1 variants on steady-state pharmacokinetics and glycemic response. PLoS ONE 2022, 17, e0271410. [Google Scholar] [CrossRef]
  79. Zhou, Y.; Ye, W.; Wang, Y.; Jiang, Z.; Meng, X.; Xiao, Q.; Zhao, Q.; Yan, J. Genetic Variants of OCT1 Influence Glycemic Response to Metformin in Han Chinese Patients with Type-2 Diabetes Mellitus in Shanghai. Int. J. Clin. Exp. Pathol. 2015, 8, 9533–9542. [Google Scholar]
  80. Moreno-González, J.G.; Reza-López, S.A.; González-Rodríguez, E.; Siqueiros-Cendón, T.S.; Escareño Contreras, A.; Rascón-Cruz, Q.; Leal-Berumen, I. Genetic Variants of SLC22A1 rs628031 and rs622342 and Glycemic Control in T2DM Patients from Northern Mexico. Genes 2025, 16, 139. [Google Scholar] [CrossRef]
  81. Emami-Riedmaier, A.; Schaeffeler, E.; Nies, A.T.; Mörike, K.; Schwab, M. Stratified medicine for the use of antidiabetic medication in treatment of type II diabetes and cancer: Where do we go from here? J. Intern. Med. 2015, 277, 235–247. [Google Scholar] [CrossRef]
  82. Santoro, A.B.; Botton, M.R.; Struchiner, C.J.; Suarez-Kurtz, G. Influence of pharmacogenetic polymorphisms and demographic variables on metformin pharmacokinetics in an admixed Brazilian cohort. Br. J. Clin. Pharmacol. 2018, 84, 987–996. [Google Scholar] [CrossRef] [PubMed]
  83. Phani, N.M.; Vohra, M.; Kakar, A.; Adhikari, P.; Nagri, S.K.; D’Souza, S.C.; Umakanth, S.; Satyamoorthy, K.; Rai, P.S. Implication of critical pharmacokinetic gene variants on therapeutic response to metformin in Type 2 diabetes. Pharmacogenomics 2018, 19, 905–911. [Google Scholar] [CrossRef]
  84. Chen, E.C.; Liang, X.; Yee, S.W.; Geier, E.G.; Stocker, S.L.; Chen, L.; Giacomini, K.M. Targeted Disruption of Organic Cation Transporter 3 Attenuates the Pharmacologic Response to Metformin. Mol. Pharmacol. 2015, 88, 75–83. [Google Scholar] [CrossRef] [PubMed]
  85. Musi, N.; Hirshman, M.F.; Nygren, J.; Svanfeldt, M.; Bavenholm, P.; Rooyackers, O.; Zhou, G.; Williamson, J.M.; Ljunqvist, O.; Efendic, S.; et al. Metformin Increases AMP-Activated Protein Kinase Activity in Skeletal Muscle of Subjects with Type 2 Diabetes. Diabetes 2002, 51, 2074–2081. [Google Scholar] [CrossRef]
  86. Zazuli, Z.; Duin, N.J.C.B.; Jansen, K.; Vijverberg, S.J.H.; Maitland-Van der Zee, A.H.; Masereeuw, R. The impact of genetic polymorphisms in organic cation transporters on renal drug disposition. Int. J. Mol. Sci. 2020, 21, 6627. [Google Scholar] [CrossRef]
  87. Hemauer, S.J.; Nanovskaya, T.N.; Abdel-Rahman, S.Z.; Patrikeeva, S.L.; Hankins, G.D.V.; Ahmed, M.S. Modulation of human placental P-glycoprotein expression and activity by MDR1 gene polymorphisms. Biochem. Pharmacol. 2010, 79, 921–925. [Google Scholar] [CrossRef]
  88. Gallardo-Cóndor, J.; Naranjo, P.; Atarihuana, S.; Coello, D.; Guevara-Ramírez, P.; Flores-Espinoza, R.; Burgos, G.; López-Cortés, A.; Cabrera-Andrade, A. Population-Specific Distribution of TPMT Deficiency Variants and Ancestry Proportions in Ecuadorian Ethnic Groups: Towards Personalized Medicine. Ther. Clin. Risk Manag. 2023, 19, 1005–1018. [Google Scholar] [CrossRef]
  89. Florez, J.C.; Price, A.L.; Campbell, D.; Riba, L.; Parra, M.V.; Yu, F.; Duque, C.; Saxena, R.; Gallego, N.; Tello-Ruiz, M.; et al. Strong Association of Socioeconomic Status with Genetic Ancestry in Latinos: Implications for Admixture Studies of Type 2 Diabetes. Diabetologia 2009, 52, 1528–1536. [Google Scholar] [CrossRef] [PubMed]
  90. Chande, A.T.; Rishishwar, L.; Conley, A.B.; Valderrama-Aguirre, A.; Medina-Rivas, M.A.; Jordan, I.K. Ancestry Effects on Type 2 Diabetes Genetic Risk Inference in Hispanic/Latino Populations. BMC Med. Genet. 2020, 21 (Suppl. S2), 132. [Google Scholar] [CrossRef] [PubMed]
  91. Cooper-DeHoff, R.M.; Niemi, M.; Ramsey, L.B.; Luzum, J.A.; Tarkiainen, E.K.; Straka, R.J.; Gong, L.; Tuteja, S.; Wilke, R.A.; Wadelius, M.; et al. The Clinical Pharmacogenetics Implementation Consortium Guideline for SLCO1B1, ABCG2, and CYP2C9 Genotypes and Statin-Associated Musculoskeletal Symptoms. Clin. Pharmacol. Ther. 2022, 111, 1007–1021. [Google Scholar] [CrossRef] [PubMed]
  92. Alexander, D.H.; Novembre, J.; Lange, K. Fast model-based estimation of ancestry in unrelated individuals. Genome Res. 2009, 19, 1655–1664. [Google Scholar] [CrossRef]
Figure 1. Individual ancestry proportions (k = 3) of Native Americans (n = 296), Spaniards (n = 114) from previous studies [27], Yoruba from 1000 Genomes (AFR, n = 209) [30], and Ecuadorian T2DM patients (n = 294).
Figure 1. Individual ancestry proportions (k = 3) of Native Americans (n = 296), Spaniards (n = 114) from previous studies [27], Yoruba from 1000 Genomes (AFR, n = 209) [30], and Ecuadorian T2DM patients (n = 294).
Pharmaceuticals 18 01335 g001
Figure 2. Comparison of ancestry proportions (NATAM, EUR, and AFR) among Ecuadorian T2DM patients grouped by diplotype and activity score of CYP2C8. Panels (AC) show grouping by CYP2C8 diplotypes (*1/*1, *1/*3, *1/*4). Panels (DF) show grouping by CYP2C8 activity score (1.5 and 2). The p-value corresponds to the post hoc test with Bonferroni adjustment. * Statistical significance (p < 0.05); ns: not significant.
Figure 2. Comparison of ancestry proportions (NATAM, EUR, and AFR) among Ecuadorian T2DM patients grouped by diplotype and activity score of CYP2C8. Panels (AC) show grouping by CYP2C8 diplotypes (*1/*1, *1/*3, *1/*4). Panels (DF) show grouping by CYP2C8 activity score (1.5 and 2). The p-value corresponds to the post hoc test with Bonferroni adjustment. * Statistical significance (p < 0.05); ns: not significant.
Pharmaceuticals 18 01335 g002
Figure 3. Comparison of ancestry proportion (NATAM, EUR, and AFR) among Ecuadorian T2DM patients grouped by diplotype and activity score of CYP2C9. Panels (AC) show grouping by CYP2C9 diplotypes (*1/*1, *1/*2, *1/*3, *2/*2). Panels (DF) show grouping by CYP2C9 activity score (1, 1.5, and 2). The p-value corresponds to the post hoc test with Bonferroni adjustment. * Statistical significance (p < 0.05); ns: not significant.
Figure 3. Comparison of ancestry proportion (NATAM, EUR, and AFR) among Ecuadorian T2DM patients grouped by diplotype and activity score of CYP2C9. Panels (AC) show grouping by CYP2C9 diplotypes (*1/*1, *1/*2, *1/*3, *2/*2). Panels (DF) show grouping by CYP2C9 activity score (1, 1.5, and 2). The p-value corresponds to the post hoc test with Bonferroni adjustment. * Statistical significance (p < 0.05); ns: not significant.
Pharmaceuticals 18 01335 g003
Figure 4. Comparison of ancestry proportion (NATAM, EUR, and AFR) among Ecuadorian T2DM patients grouped by diplotype and activity score of CYP2C19. Panels (AC) show grouping by CYP2C19 diplotypes (*1/*1, *1/*17, *1/*2, *17/*17, *2/*17, and *2/*2). Panels (DF) show grouping by CYP2C19 activity score (PM, 1, 1.5, 2, and UM). The p-value corresponds to the post hoc test with Bonferroni adjustment. * Statistical significance (p < 0.05); ns: not significant.
Figure 4. Comparison of ancestry proportion (NATAM, EUR, and AFR) among Ecuadorian T2DM patients grouped by diplotype and activity score of CYP2C19. Panels (AC) show grouping by CYP2C19 diplotypes (*1/*1, *1/*17, *1/*2, *17/*17, *2/*17, and *2/*2). Panels (DF) show grouping by CYP2C19 activity score (PM, 1, 1.5, 2, and UM). The p-value corresponds to the post hoc test with Bonferroni adjustment. * Statistical significance (p < 0.05); ns: not significant.
Pharmaceuticals 18 01335 g004
Figure 5. Comparison of ancestry proportion (NATAM, EUR, and AFR) among Ecuadorian T2DM patients grouped by genotypes of SLC variants. Panels (AC): Grouping by genotypes of rs72552763 in SLC22A1. Panels (DF): Grouping by genotypes of rs594709 in SLC22A1. Panels (GI): Grouping by genotypes of rs628031 in SLC22A1. Panels (JL): Grouping by genotypes of rs2076828 in SLC22A3. * Statistical significance (p < 0.05); ns: not significant.
Figure 5. Comparison of ancestry proportion (NATAM, EUR, and AFR) among Ecuadorian T2DM patients grouped by genotypes of SLC variants. Panels (AC): Grouping by genotypes of rs72552763 in SLC22A1. Panels (DF): Grouping by genotypes of rs594709 in SLC22A1. Panels (GI): Grouping by genotypes of rs628031 in SLC22A1. Panels (JL): Grouping by genotypes of rs2076828 in SLC22A3. * Statistical significance (p < 0.05); ns: not significant.
Pharmaceuticals 18 01335 g005
Figure 6. Scatter plot of the activity score of cytochromes encoded by CYP2C8, CYP2C9, and CYP2C19, according to ancestry proportion (NATAM, EUR, and AFR). Lines represent the fitted line from a linear regression model, and grey areas represent the 95% confidence interval. The statistics shown correspond to Spearman’s ordinal correlation for each ancestry proportion. Panels (AC): Plots corresponding to CYP2C8, CYP2C9, and CYP2C19, respectively. Panels (DF) correspond to the faceted scatter plots according to ancestry proportion in CYP2C8, CYP2C9, and CYP2C19, respectively. Red points correspond to NATAM ancestry, yellow points correspond to EUR ancestry, and blue points correspond to AFR ancestry. * Statistical significance (p < 0.05).
Figure 6. Scatter plot of the activity score of cytochromes encoded by CYP2C8, CYP2C9, and CYP2C19, according to ancestry proportion (NATAM, EUR, and AFR). Lines represent the fitted line from a linear regression model, and grey areas represent the 95% confidence interval. The statistics shown correspond to Spearman’s ordinal correlation for each ancestry proportion. Panels (AC): Plots corresponding to CYP2C8, CYP2C9, and CYP2C19, respectively. Panels (DF) correspond to the faceted scatter plots according to ancestry proportion in CYP2C8, CYP2C9, and CYP2C19, respectively. Red points correspond to NATAM ancestry, yellow points correspond to EUR ancestry, and blue points correspond to AFR ancestry. * Statistical significance (p < 0.05).
Pharmaceuticals 18 01335 g006
Table 1. Distribution of ancestry percentage across different genotypes and activity scores of CYP2C8, CYP2C9, and CYP2C19 in Ecuadorian patients diagnosed with T2DM (n = 294).
Table 1. Distribution of ancestry percentage across different genotypes and activity scores of CYP2C8, CYP2C9, and CYP2C19 in Ecuadorian patients diagnosed with T2DM (n = 294).
NATAMEURAFR NATAMEURAFR
CYP2C8 Activity score
*1/*162.61
(52.71–71.70)
33.88
(24.35–41.58)
2.48
(0.00–7.31)
1---
*1/*355.41
(46.78–63.68)
38.58
(31.13–47.70)
5.01
(0.77–8.92)
1.554.83
(46.85–62.21)
40.23
(32.32–48.16)
4.58
(0.77–8.92)
*1/*449.98
(48.43–53.26)
43.80
(40.05–48.37)
2.54
(1.55–8.87)
262.61
(52.71–71.70)
33.88
(24.35–41.58)
2.48
(0.00–7.31)
*3/*4---
pKW0.002 *0.005 *0.262pU<0.001 *0.003 *0.109
CYP2C9
*1/*162.55
(52.67–71.49)
34.09
(24.34–41.68)
2.40
(0.00–7.06)
156.36
(48.72–64.91)
34.89
(29.90–45.35)
6.61
(1.72–8.45)
*1/*255.51
(45.76–64.34)
38.58
(30.19–47.06)
5.01
(0.68–9.01)
1.555.51
(45.76–64.34)
38.58
(30.19–47.07)
5.01
(0.68–9.01)
*1/*356.63
(49.29–65.18)
34.72
(29.79–41.99)
7.18
(2.18–8.59)
262.55
(52.66–71.49)
34.09
(24.34–41.68)
2.40
(0.00–7.06)
*2/*2---
*2/*3---
pKW0.031 *0.1320.053pKW0.021 *0.0970.070
CYP2C19
*1/*162.42
(51.50–72.79)
33.63
(24.07–42.50)
2.44
(0.00–6.60)
PM60.54
(53.34–68.82)
31.93
(29.27–35.16)
5.50
(1.90–9.47)
*1/*260.94
(54.63–67.25)
34.44
(28.14–41.31)
1.89
(0.00–5.97)
160.94
(54.63–67.25)
34.44
(28.14–41.31)
1.89
(0.00–5.97)
*1/*4---1.5---
*1/*1758.93
(50.04–64.08)
36.27
(30.13–44.09)
6.92
(0.58–10.27)
262.42
(51.50–72.79)
33.63
(24.07–42.50)
2.44
(0.00–6.60)
*2/*260.54
(53.34–68.82)
31.93
(29.27–35.16)
5.50
(1.90–9.47)
UM57.31
(50.10–63.82)
36.33
(30.15–43.71)
7.06
(0.92–10.19)
*17/*17---
*2/*17---
pKW0.1830.6010.025 *pKW0.1830.6010.025*
KW, Kruskal–Wallis test; U Mann–Whitney’s U test; * Statistical significance (p < 0.05). Showing median and interquartile ranges (p25–p75). PM: Poor metabolizers; UM: Ultra-rapid metabolizers.
Table 2. Ancestry distribution percentage across different genotypes of SLC22A1, SLC22A2, SLC22A3, and ABCB1 among Ecuadorian T2DM patients (n = 294).
Table 2. Ancestry distribution percentage across different genotypes of SLC22A1, SLC22A2, SLC22A3, and ABCB1 among Ecuadorian T2DM patients (n = 294).
GeneIDGenotypeNATAMEURAFR
SLC22A1rs72552763GAT/GAT59.61 (49.30–67.77)35.66 (27.29–44.39)3.13 (0.00–7.51)
GAT/del61.96 (52.55–72.83)34.42 (22.95–41.17)2.56 (0.00–7.55)
del/del64.98 (60.31–76.51)30.11 (21.94–36.37)1.89 (0.00–6.70)
pKW0.022 *0.017 *0.842
rs622342A/A60.46 (49.38–67.91)35.53 (26.25–43.23)2.70 (0.00–7.86)
A/C60.28 (51.47–71.26)35.30 (24.37–42.77)2.77 (0.00–7.35)
C/C64.98 (60.31–76.51)30.11 (21.94–36.37)1.89 (0.00–6.70)
PKW0.2470.2430.397
rs12208357C/C61.41 (51.78–70.61)34.44 (25.55–42.08)2.62 (0.00–7.39)
C/T51.21 (44.62–64.92)44.73 (34.13–48.11)0.66 (0.93–8.83)
T/T---
pU0.3390.2650.884
rs2282143C/C61.41 (51.78–70.61)34.44 (25.55–42.08)2.62 (0.00–7.39)
C/T51.21 (44.62–64.92)44.73 (34.13–48.11)0.93 (0.66–8.83)
T/T---
pU0.3390.2650.884
rs594709A/A62.58 (52.90–71.94)33.17 (22.91–41.83)2.57 (0.00–7.76)
A/G58.46 (49.93–66.18)37.13 (27.70–45.61)3.38 (0.00–6.57)
G/G56.09 (42.93–68.57)41.27 (31.42–53.12)0.00 (0.00–1.31)
pKW0.040 *0.013 *0.298
rs683369C/C61.10 (51.52–70.60)34.53 (24.27–42.47)2.60 (0.00–7.68)
C/G61.96 (52.16–70.55)34.13 (27.19–41.71)3.14 (0.00–6.61)
G/G--
pU0.8430.6250.849
rs628031G/G62.21 (52.85–72.00)33.63 (23.17–42.00)2.47 (0.00–7.64)
G/A57.96 (49.51–65.48)38.47 (29.54–45.10)4.02 (0.00–6.96)
A/A67.71 (44.48–71.17)32.28 (28.82–50.25)0.00 (0.00–0.19)
pKW0.042 *0.040 *0.143
SLC22A2rs316019C/C61.97 (51.93–70.79)34.33 (25.55–41.90)2.54 (0.00–7.36)
C/A54.99 (46.89–66.91)39.39 (26.97–44.43)5.26 (1.03–10.60)
A/A---
pU0.1630.3690.120
SLC22A3rs2076828C/C63.16 (53.56–72.81)32.89 (22.99–40.97)2.31 (0.00–6.61)
C/G55.40 (47.32–65.61)39.52 (31.02–47.14)4.32 (0.00–8.31)
G/G53.25 (46.62–57.56)40.07 (38.26–43.06)7.32 (2.70–9.14)
pKW<0.001 *<0.001 *0.560
ABCB1rs2032582G/G57.27 (49.30–65.98)36.21 (28.09–44.97)3.57 (0.00–7.35)
G/A60.11 (47.98–67.88)36.29 (28.13–47.83)3.84 (1.77–9.51)
A/A---
G/T61.33 (50.31–70.44)34.39 (26.05–42.52)2.10 (0.00–7.51)
T/T62.73 (54.13–72.24)33.70 (24.05–41.62)2.22 (0.00–6.44)
T/A66.02 (60.04–71.07)30.90 (23.53–35.07)3.09 (0.00–7.99)
pKW0.1390.1810.623
rs1128503C/C57.93 (49.23–65.52)36.37 (30.03–44.99)3.59 (0.00–7.19)
C/T60.95 (50.37–70.38)33.88 (25.74–41.68)2.31 (0.00–7.75)
T/T63.16 (53.14–72.79)33.18 (23.40–41.76)2.21 (0.00–6.56)
pKW0.0720.1270.481
rs1045642C/C58.04 (49.91–67.67)34.72 (28.91–44.13)3.48 (0.00–6.96)
C/T61.26 (50.63–69.90)34.42 (25.96–41.58)2.40 (0.00–7.69)
T/T63.16 (53.27–72.44)32.72 (22.87–42.43)2.14 (0.00–5.50)
pKW0.2060.3180.730
KW Kruskal–Wallis test; U Mann–Whitney’s U test; * Statistical significance (p < 0.05). Showing median and interquartile ranges (p25–p75).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ortega-Ayala, A.; de la Cruz, C.G.; Mora, L.; Bonilla, M.; Tana, L.; Rodrigues-Soares, F.; Dorado, P.; LLerena, A.; Terán, E. Pharmacogenetics and Molecular Ancestry of SLC22A1, SLC22A2, SLC22A3, ABCB1, CYP2C8, CYP2C9, and CYP2C19 in Ecuadorian Subjects with Type 2 Diabetes Mellitus. Pharmaceuticals 2025, 18, 1335. https://doi.org/10.3390/ph18091335

AMA Style

Ortega-Ayala A, de la Cruz CG, Mora L, Bonilla M, Tana L, Rodrigues-Soares F, Dorado P, LLerena A, Terán E. Pharmacogenetics and Molecular Ancestry of SLC22A1, SLC22A2, SLC22A3, ABCB1, CYP2C8, CYP2C9, and CYP2C19 in Ecuadorian Subjects with Type 2 Diabetes Mellitus. Pharmaceuticals. 2025; 18(9):1335. https://doi.org/10.3390/ph18091335

Chicago/Turabian Style

Ortega-Ayala, Adiel, Carla González de la Cruz, Lorena Mora, Mauro Bonilla, Leandro Tana, Fernanda Rodrigues-Soares, Pedro Dorado, Adrián LLerena, and Enrique Terán. 2025. "Pharmacogenetics and Molecular Ancestry of SLC22A1, SLC22A2, SLC22A3, ABCB1, CYP2C8, CYP2C9, and CYP2C19 in Ecuadorian Subjects with Type 2 Diabetes Mellitus" Pharmaceuticals 18, no. 9: 1335. https://doi.org/10.3390/ph18091335

APA Style

Ortega-Ayala, A., de la Cruz, C. G., Mora, L., Bonilla, M., Tana, L., Rodrigues-Soares, F., Dorado, P., LLerena, A., & Terán, E. (2025). Pharmacogenetics and Molecular Ancestry of SLC22A1, SLC22A2, SLC22A3, ABCB1, CYP2C8, CYP2C9, and CYP2C19 in Ecuadorian Subjects with Type 2 Diabetes Mellitus. Pharmaceuticals, 18(9), 1335. https://doi.org/10.3390/ph18091335

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop