Open Access Article
Nicole Tosi†
a,
Nicola Luigi Bragazzi†
*a,
Cristiana Mignogna
a,
Mirko Treccani
a,
Letizia Bresciani
a,
David Vauzour
b,
Giovanni Malerba
c,
Valeria Barili
d,
Davide Martorana
ef,
Daniele Del Rio
ag and
Pedro Mena
ag
aHuman Nutrition Unit, Department of Food and Drug, University of Parma, Parma, Italy. E-mail: nicolaluigi.bragazzi@unipr.it
bNorwich Medical School, University of East Anglia, Norwich, UK
cGM Lab, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Verona, Italy
dMedical Genetics, Department of Medicine and Surgery, University of Parma, Parma, Italy
eMedical Genetics Unit, Department of Onco-Hematology, University Hospital of Parma, Parma, Italy
fCoreLab Unit, Research Center, University Hospital of Parma, Parma, Italy
gMicrobiome Research Hub, University of Parma, Parma, Italy
First published on 19th November 2025
Response to (poly)phenol intake is highly variable among subjects, and genetic variants may contribute to such variability. However, evidence addressing this assumption is currently lacking. To address such shortcomings, we systematically reviewed the current literature and selected twelve studies looking at associations between the inter-individual variability in (poly)phenol bioavailability and metabolism and single nucleotide polymorphisms (SNPs) in candidate genes involved in (poly)phenol ADME (absorption, distribution, metabolism, and excretion). In total, 88 SNPs in 33 genes were studied, of which slightly more than half (n = 17) were related to drug/xenobiotic metabolism. More specifically, two were involved in absorption, seven in phase I metabolism, four in phase II metabolism, and four in excretion. The remaining 16 genes were related to steroid hormone metabolism and activity. Considering genes specifically related to (poly)phenol ADME, 16 SNPs showed significant modifying effects on urinary and/or plasma levels of phenolic metabolites and/or on their kinetic parameters. However, it was not possible to associate a particular genetic variant with a change in (poly)phenol-related ADME. Only a few studies applied stringent statistical criteria and recruited sufficiently large and diverse samples to reach solid and reliable conclusions. As such, studies employing larger samples, leveraging integrative bioinformatics approaches and genome-wide linkage, are warranted.
Contrasting findings reported in the literature derive from the influence of various factors leading to high inter-individual variability in (poly)phenol bioavailability,11,12 which is the main determinant of their effectiveness and physiological response, influencing health outcomes. Factors determining differences in the absorption, distribution, metabolism, and excretion (ADME) process can be exogenous, including the variability of (poly)phenol content in foods,13 the high degree of diversification of chemical structures,14,15 and the complex interplay of (poly)phenols with other components of the diet (fibre, proteins, etc.), as well as the food matrix.16 However, endogenous/individual factors are considered the principal source of heterogeneity: among them, personal characteristics (age, sex, ethnicity), lifestyle (dietary habits, smoking, physical activity, medication), pathophysiological status, gut microbiota, and genetic background.17–20
In the case of the bioavailability and metabolism of (poly)phenols, genetic variations and microbiota composition and activity are the major contributors to variability.11,12 Single Nucleotide Polymorphisms (SNPs) represent a source of genetic variation that may, at least partially, explain differences in the response to dietary (poly)phenol exposure.21 A further layer of variability is related to the gut microbiota, which can be considered a key player in a variety of individual physiological processes and an important determinant of metabolic phenotypes (also known as metabotypes).22,23 The human microbiome has been linked with host genetic variants, and dysbiosis has been associated with several diseases. As such, human genetic variants are expected to play a relevant role in explaining various complex and overlapping (patho)physiological phenomena, ranging from food intake to the composition of the microbiome and their impacts on food metabolism and human health.24 However, to the best of our knowledge, information is scarce concerning the influence of genetic variants on the bioavailability and metabolism of dietary (poly)phenols and consequently on the response to dietary (poly)phenol exposure.
Often found in glycosylated forms, (poly)phenols are partially hydrolyzed by human enzymes in the upper gastrointestinal tract, releasing aglycones that can be absorbed and conjugated by phase II enzymes in enterocytes and hepatocytes.1–3 However, most (poly)phenols are not absorbed in the small intestine and reach the colon intact, where they are modified by the gut microbiota into smaller catabolites. Gut microbial catabolites are more easily absorbed and can be conjugated by phase II enzymes in colonocytes and hepatocytes.1–3 As such, the molecules present in the circulation and potentially bioavailable for target tissues are phase II metabolites of both human and microbial-human origin, which are finally excreted through urine. SNPs in genes involved in (poly)phenol ADME, including transporters, glycosidases, and phase II enzymes (sulfotransferases or SULTs, UDP-glucuronosyltransferases or UGTs, and catechol-O-methyltransferase or COMT), could have an impact on the amount and type of metabolites excreted18 (Fig. 1).
A better understanding of the genetic component would enable and enhance the translation of our current knowledge about these bioactive compounds into more effective and personalised dietary advice. A recently published review found that more research on the factors that underlie the differences in the response to (poly)phenol exposure is urgently needed.10 Therefore, the present study was undertaken to fill this knowledge gap, with the aim of systematically identifying and appraising genetic factors associated with the inter-individual variability in (poly)phenol ADME after their dietary intake and assessing their causal relationships with (poly)phenol bioavailability and metabolism.
The “Medical Subject Headings” (MeSH) thesaurus, a controlled, hierarchically-organised vocabulary produced by the National Library of Medicine (NLM), was leveraged. When appropriate, the wild-card option (i.e., truncated words) was used. (Poly)phenol compound names were obtained from Phenol-Explorer, the most comprehensive database on (poly)phenols.25 (Poly)phenol metabolite names were obtained from PhytoHub, a freely available electronic database containing detailed information about dietary phytochemicals and their human and animal metabolites. The full string, which was modified and adapted according to each database, is reported in SI Table 1. Moreover, the “Human Genome Epidemiology Literature Finder” (HuGE Navigator Database) based at the “Public Health Genomics and Precision Health Knowledge Base” (PHGKB v8.5) was mined. Extensive cross-referencing was applied. Finally, target journals were hand-searched: these journals were selected because they represent the most frequently publishing outlets in the fields of nutritional genomics, nutrikinetics, and (poly)phenol research. Their inclusion ensured that our manual search complemented database screening and minimized the risk of missing relevant studies at the intersection of (poly)phenol metabolism and genetics. The literature search was conducted by two researchers independently and is updated as of September 24, 2023. Further details are reported in Table 1 and SI Table 1.
| Search items | Details |
|---|---|
| Search string components | (Poly)phenols |
| Dietary exposure/intake | |
| Human metabolism | |
| SNPs | |
| Databases/registries/repositories mined/searched | PubMed/MEDLINE, Scopus, WoS, EMBASE, CAS/ACS, HuGE Navigator Database (PHGKB v8.5) |
| Inclusion criteria | P (humans) |
| I/E (exposed to dietary (poly)phenols) | |
| C (stratified according to their genotype status) | |
| O (quantification of (poly)phenol-related plasma and/or urinary metabolites by any separation methodology/technique of analytical chemistry) | |
| S (any original study, observational/interventional, retrospective/prospective, cross-sectional/longitudinal, randomised/non-randomised, parallel/cross-over, placebo/non-placebo-controlled, pilot, etc.) | |
| Exclusion criteria | P (in vitro studies- cellular-, in silico/computational studies) |
| I/E (exposed to nutrients other than (poly)phenols) | |
| C (not providing information about genotype status) | |
| O (not quantifying (poly)phenol-related metabolites) | |
| S (reviews of any type) | |
| Time filter | None applied |
| Language filter | None applied |
| Target journals hand-searched | Biochem. Pharmacol.; Br. J. Nutr.; Cancer Epidemiol. Biomarkers Prev.; Eur. J. Pharmacol.; Genes Nutr.; Int. J. Mol. Epidemiol. Genet.; Mol. Nutr. Food Res.; Nutrients; PLoS One |
Studies were excluded if studying non-human populations (P), i.e., if designed as in vitro models (like cellular models), animal models, or in silico/computational models. Also, articles were not retained in the present review if study subjects were exposed (E) to dietary compounds other than (poly)phenols or if, in the case of exposure to dietary (poly)phenols, (poly)phenol-related metabolites were not analytically characterised/measured (O). Another cause of exclusion was given by other studied outcomes, including changes in dietary habits and eating patterns (e.g. energy intake), anthropometric characteristics (body weight, body mass index (BMI), waist circumference), or metabolic responses (for instance, glucose and insulin levels, or circulating leptin, ghrelin, or adiponectin). Finally, studies were excluded if designed as technical notes reporting new methodologies or advancements in techniques of analytical chemistry but narrowly focused on the technical aspects, letters to the editor, editorials, commentaries, and expert opinions, or not containing sufficient quantitative details (S). Reviews (of any type, narrative, systematic, or meta-analysis) were also excluded but were, however, scanned to increase the chance of not missing content relevant to the present review.
A specifically designed Excel spreadsheet was utilised. Two researchers independently extracted the data. Further details about the search strategy adopted in the present systematic review are reported in Table 1 and SI Table 1.
513 items, which remained at 5310 after removing the duplicates. A total of 5288 items were discarded based on title and/or abstract. Twenty-two studies were scrutinised in full text. Based on the inclusion and exclusion criteria mentioned above, ten studies28–37 were “excluded with reason” (SI Table 2). Finally, twelve studies were retained in the present systematic review, overviewed in Table 2 and detailed in SI Tables 3 and 4.38–49 The flowchart adopted to manage the study retrieval and inclusion/exclusion is depicted in Fig. 2.
| Study | Study design | Country | Sample size | SNPs | Dietary (poly)phenols | (Poly)phenol source | Intervention duration | Impact of SNPs |
|---|---|---|---|---|---|---|---|---|
| BMI indicates body mass index (kg m−2). | ||||||||
Brown et al. (2011) 38 |
Single-centre, placebo-controlled, double-blinded, cross-over study with age- and BMI-matched groups | UK | 64 subjects aged 40–69 years, 100% males, overweight or obese | COMT (rs4680) | Flavan-3-ols (mainly catechins and gallocatechins) | Green tea | 6 weeks | Lower concentrations in homozygous GG individuals |
Chang et al. (2019) 39 |
Randomised cross-over study | USA | 252 subjects aged 59.4 ± 6.2 years, 100% females, BMI 30.5 ± 7.6 | 60 functional SNPs in 29 genes (ARPC1A, BAIAP2L1, BMF, COMT, CYP17A1, CYP19A1, CYP1A1, CYP1B1, CYP3A4, CYP3A5, ESR1, GCKR, HHEX, HSD17B1, JMJD1C, LHCGR, NR2F2, PRMT6, SHBG, SLCO1B1, SULT1A1, TDGF3, TSPYL5, UGT1A1, ZBTB10, ZKSCAN5, ZNF652) | Lignans | Ground brown flaxseed | 6 weeks | Impact of SNPs on enterolactone excretion |
Fraga et al. (2022) 40 |
Non-randomised, pre-post design study | Brazil | 46 subjects aged 26.26 ± 4.50 years [19–38 years], 20 males (43.5%) and 26 females (56.5%), BMI 23.23 ± 2.54 [18.50–29.9] | SULT1A1 (rs3760091, rs4788068), SULT1C4 (rs1402467), ABCC2 (rs8187710) | Flavanones (mainly hesperetin and naringenin) | Pasteurised orange juice (Citrus sinensis) | 24 hours | Significant (p < 0.05) relationship between SNPs and high excretion of phase II flavanone metabolites |
Hong et al. (2012) 41 |
Cross-sectional, ancillary sub-analysis of a population-based study | South Korea | 1391 healthy subjects aged 52.0 years, 758 males (54.5%) and 633 females (45.5%), BMI 24.9 | HACE1 (rs6927608, rs4946645, rs11759010, rs17065302, and rs4245525) | Equol | Soy | Not applicable – observational | Significant effects of five SNPs clustered in the 6q21 region, the most significant of which was rs6927608 (allele A>C) |
| Inoue-Choi et al. (2010)42 | Cross-sectional sub-analysis nested within the Shanghai Cohort Study | China | 660 subjects aged 56.7 ± 5.0 years, 100% males | COMT (rs4680) | Flavan-3-ols (mainly catechins and gallocatechins) | Green tea | Not applicable – observational | Urinary tea catechins were lower by 35–45% among men carrying the LL genotype (p = 0.007). When consuming greater amount of green tea (≥5 g day−1), urinary tea catechins did not differ according to COMT genotype |
| Lorenz et al. (2014)43 | Non-randomised, pre-post design study | Germany | 24 subjects, 10 males (41.7%) and 14 females (58.3%) | COMT (rs6269, rs4633, rs4680, rs4818) | EGCG | Pure compound | 2 hours | None |
| Miller et al. (2012)44 | Pilot, non-placebo-controlled trial | UK | 20 overweight and obese individuals, aged 54.6 ± 3.2 years [18–70 years], 14 males (70.0%) and 6 females (30.0%) | COMT (rs4680) | Flavan-3-ols (mainly catechins and gallocatechins) | Green tea | 8 hours | None |
| Miller et al. (2012)45 | Randomised, double-blinded, placebo-controlled, crossover trial, with age- and BMI-matched groups | UK | 47 overweight/obese adults, 100% males | COMT (rs4680) | Flavan-3-ols (mainly catechins and gallocatechins) | Green tea | 24 hours | None |
| Momma et al. (2023)46 | Cross-sectional, ancillary sub-analysis of a population-based study | UK | 16 672 subjects, 9020 females (54.1%) and 7652 males (45.9%) |
PON1 (rs662, rs854560, rs705379, rs705381, and rs854572) | Flavan-3-ols | Diet (not better characterised) | Not applicable – observational | No differences in the sum of 5-(4′-hydroxyphenyl)-γ-valerolactone-3′-sulfate and 5-(4′-hydroxyphenyl)-γ-valerolactone-3′-glucuronide metabolites in men, and only small differences among rs662 and rs705381 alleles in women with no clinical and physiological impact |
| Muhrez, et al. (2017)47 | Metabolomics study | France | 53 subjects aged 23 years [20–59 years], 24M males (45.3%) and 29 females (54.7%) | ABCC2/MRP2 (rs717620, rs3740066, rs2273697) | NA | Free diet | Not applicable – observational | SNPs associated with some (poly)phenol low-molecular weight urinary metabolites |
| Scholl et al. (2018)48 | Population-based, open, one-arm nutrikinetic study | Germany | 84 subjects aged 24.5 ± 3.9 years [19–49 years], 30 males (35.7%) and 54 females (64.3%), BMI 22.7 ± 2.6 | MRP2/ABCC2 (rs2273697, rs3740066, rs717620, rs8187710), OATP1B1/SLCO1B1 (rs4149056, rs2306283, rs11045819), Pgp/ABCB1 (rs1045642, rs1128503, rs2032582), COMT (rs4680), SULT1A1 (rs750155), UGT1A1 (rs8175347) | Flavan-3-ols (mainly catechins and gallocatechins) | Green tea | 5 days | Impact of SNPs of MRP2 and OATP1B1 |
| Wakeling and Ford (2012)49 | Non-randomised, pre-post design study | UK | 100 subjects aged 33.0 ± 9.3 years [18–50 years], 100% females, BMI 23.4 ± 3.4 | UGT1A1 (rs8175347), CBG (rs358231), ABCG2 (rs2231142), and ABCC2 (rs2273697), LPH (rs3754689) | Isoflavones (mainly glycitein, daidzein and genistein) | Soy | 24 hours | Impact of SNPs on isoflavone excretion |
42 and 2023.46 Sample sizes ranged from 20
44 to 16
672
46 subjects (median 74), totalling 19
413 participants. The age of the participants varied from 18
44 to 75 years.39 Five studies38,39,42,45,49 recruited populations consisting of males38,42,45 or females39,49 only. In the remaining seven studies,40,41,43,44,46–48 the percentage of male subjects ranged from 35.7%
48 to 70.0%.44 Of note, only two studies46,48 reported and analysed data stratified according to sex/gender.
Most studies (67%) were conducted in Europe – five in the UK,38,44–46,49 two in Germany,43,48 and one in France.47 Two studies were carried out in the Americas – one in the USA39 and one in Brazil40 – and two in Asia, one in China,42 and one in South Korea.41 As such, information was primarily provided for European/Caucasian ancestry, with only one study investigating African ancestry along with Caucasian ancestry39 and other two focusing exclusively on Asiatic ancestry.41,42
Concerning BMI, no information was provided in four studies.42,43,46,47 When reported, BMI ranged from 18.5 to 38.0 kg m−2, meaning that a range of populations – normal weight, overweight, and obese – have been investigated in the studies included in the present systematic review, with a higher representation of the latter.
672 out of approximately 19
500—come from a single observational study.46 Additionally, a significant portion (1391 participants) were recruited from another observational study.41 Of the interventional studies, only three38,39,45 were randomised, double-blinded, cross-over investigations. Furthermore, four studies40,43,44,49 were single acute intake, one-arm, non-placebo-controlled investigations. Another study48 was designed as a population-based, open, one-arm nutrikinetic study. Finally, the duration of the dietary intervention varied from a few hours/one day40,49 to six weeks,38,39 thus including both acute and chronic trials.
Concerning smoking status, no information was provided for three studies.43,46,47 Four studies39,41,42,48 included smokers, whereas all the other studies excluded smokers from enrolment. When reported/recruited, the rate of current smokers varied considerably from 19.8%
39 to 60%.42 Regarding alcohol intake, no information was provided for five studies.39,43,46,47,49 When reported, the rate of habitual alcohol consumers ranged from 5.9%
41 to 89.3%.48 All the other studies excluded alcohol consumers or asked participants to refrain from alcohol consumption from 24
44 to 48 hours45 before the investigation. Concerning exercise and physical activity, no information was provided for eight studies.39,41–44,46,47,49
Finally, there was a considerable degree of heterogeneity also in the reporting of drugs and contraceptives/hormone therapy. Information about the former variable was not disclosed in five studies,41–43,46,47 whereas all the other studies excluded patients on drugs from enrolment. Concerning the latter variable, only one study48 considered this parameter, with 44.0% and 56.0% of the sample recruited ever and never using contraceptives, respectively. Of note, the use of contraceptives was found to impact dietary (poly)phenol metabolism.
![]() | ||
| Fig. 3 Number of studies investigating a certain (poly)phenol source (on the left) or (poly)phenol class (on the right). | ||
Dietary assessment was performed mainly by carrying out a 24-hour dietary recall. In one study,39 a series of 12 telephone-administered 24-hour dietary recalls were assigned randomly throughout the 5-month active intervention period. Only one study,42 which was observational, utilised a “Food Frequency Questionnaire” (FFQ) that included 45 food groups or items representing commonly consumed local foods to assess eating habits in terms of dietary patterns. Of note, only in one study38 participants were prospectively required to record dietary and daily activity information.
A median value of 1 gene per study was investigated: the range of genes under study went from one38,42–47 to 25.39 Seven studies38,42–47 explored one single gene, one study40 three genes, another study49 five genes, and a further study48 six genes. Finally, one study39 appraised 25 genes. Of note, a single study39 was responsible for most (76%) of the genes investigated, and, despite some degree of overlapping, no gene was studied by all the articles included in the present systematic review (Fig. 4). It is worth stressing that a study41 did not adopt the candidate gene approach, adopting, instead, a genome-wide exploration unbiased from an a priori selection of any preferential gene.
![]() | ||
| Fig. 4 Network-based representation of the overlapping genes involved in (poly)phenol adme among studies included in the present systematic review. Only interconnected genes are shown. | ||
Of the 33 genes studied, slightly more than half (n = 17, 51.5%) were related to drug/xenobiotic metabolism (Table 3). More specifically, two (11.8%; 6.1% of the entire set of genes) were involved in absorption, seven (41.2%; 21.2% of the overall set of genes) in phase I metabolism, four (23.5%; 12.1% of all genes) in phase II metabolism, and four (23.5%; 12.1% of the entire list of genes) in excretion. The remaining 16 genes (48.5%) were related, to varying degrees, to steroid hormone metabolism and activity (Fig. 5).
| Gene | Protein | Function | SNP | Study |
|---|---|---|---|---|
| SNPs that have a significant modifying effect on (poly)phenol ADME are in bold. The study analysing each SNP is also indicated. | ||||
| CBG/GBA3 | Cytosolic β-glucosidase | Deglycosilation – absorption | rs358231 | 49 |
| LCT | Lactase-phlorizin hydrolase | rs3754689 | 49 | |
| ABCC2/MRP2 | ATP-binding cassette C2/multidrug resistance-associated protein 2 | Efflux – excretion | rs2273697 | 47–49 |
| rs717620 | 47 and 48 | |||
| rs3740066 | 47 and 48 | |||
| rs8187710 | 40 and 48 | |||
| ABCG2/BCRP | ATP-binding cassette G2/breast cancer resistance protein | rs2231142 | 49 | |
| ABCB1/PGP/MDR1 | ATP-binding cassette B1/P-glycoprotein 1/multidrug resistant-associated protein 1 | rs1045642 | 48 | |
| rs1128503 | 48 | |||
| rs2032582 | 48 | |||
| OATP1B1/SLCO1B1 | Organic anion transporting polypeptide 1B1/solute carrier organic anion transporter 1B1 | Uptake – excretion | rs4149056 | 39 and 48 |
| rs11045819 | 48 | |||
| rs2306283 | 48 | |||
| COMT | Catechol-O-methyltransferase | Methylation – phase II metabolism | rs4680 | 38, 39, 42–45 and 48 |
| rs6269 | 39 and 43 | |||
| rs4633 | 39 and 43 | |||
| rs4818 | 43 | |||
| rs4646312 | 39 | |||
| UGT1A1 | UDP-glucuronosyltransferase 1A1 | Glucuronidation – phase II metabolism | rs8175347 | 48 and 49 |
| rs4124874 | 39 | |||
| rs10929302 | 39 | |||
| rs887829 | 39 | |||
| rs6742078 | 39 | |||
| SULT1A1 | Sulfotransferase 1A1 | Sulfation – phase II metabolism | rs26528 | 39 |
| rs8049439 | 39 | |||
| rs4788068 | 40 | |||
| rs3760091 | 40 | |||
| rs750155 | 48 | |||
| SULT1C4 | Sulfotransferase 1E1 | rs1402467 | 40 | |
| PON1 | Paraoxonase 1 | Hydrolysis – phase I metabolism | rs662 | 46 |
| rs854560 | 46 | |||
| rs705379 | 46 | |||
| rs705381 | 46 | |||
| rs854572 | 46 | |||
![]() | ||
| Fig. 5 Number of genes with a certain role in (poly)phenol ADME or other roles (on the left), and times that genes with a certain role were studied (on the right). | ||
A median value of 5 SNPs per study was analysed: the range of SNPs went from one SNP38,42,44 to sixty.39 Three studies38,42,44 studied the impact of a single SNP, one study47 the effects of three SNPs, two studies,40,43 and four studies41,45,46,49 of five SNPs. Finally, one study48 and a further study39 explored the effects of panels of 13 and 60 SNPs, respectively. Of note, a single study39 was responsible for the majority (68%) of the SNPs investigated among 9 genes (Fig. 6). Considering genes specifically related to (poly)phenol ADME, 16 SNPs showed significant modifying effects on urinary and/or plasma levels of phenolic metabolites and/or on their kinetic parameters (Table 4).
| Gene | SNPs | Impact of SNPs | Study |
|---|---|---|---|
| COMT | rs4680 | Lower 24 h urinary excretion of EGC, EC, 4′-O-methyl-EGC, 3′,4′-diHPVL, 3′,4′,5′-triHPVL in AA genotype group (low activity COMT) | 42 |
| COMT | rs4680 | Lower 24 h urinary excretion of EGC and 4′-O-methyl-EGC in GG genotype group (high activity COMT) | 38 |
| COMT | rs4680 | Higher urinary excretion of 4′-O-methyl-EGC in GG genotype group (high activity COMT) during the first 5.5 h | 44 |
| SLCO1B1 | rs4149056, rs2306283 | Plasma pharmacokinetics and/or relative bioavailability of EGCG and EGC | 48 |
| COMT | rs4680 | ||
| MRP2 | rs717620, rs3740066 | ||
| UGT1A1 | rs8175347 | ||
| SULT1A1 | rs4788068, rs3760091 | Higher urinary excretion of phase II conjugates of hesperetin and naringenin | 40 |
| SULT1C4 | rs1402467 | ||
| MRP2 | rs8187710 | ||
| UGT1A1 | rs8175347 | Relative/absolute, individual/total urinary excretion of isoflavone metabolites, and/or sulfate-to-glucuronide ratio | 49 |
| CBG | rs358231 | ||
| MRP2 | rs2273697 | ||
| BCRP | rs2231142 | ||
| MRP2 | rs717620, rs3740066, rs2273697 | Urinary excretion of 1,3-dihydroxybenzene, 2-(3′,4′-dihydroxyphenyl)acetic acid, 3′,4′-dihydroxycinnamic acid, 3-(3′-hydroxyphenyl)propanoic acid, 4′-hydroxyhippuric acid | 47 |
| SLCO1B1 | rs4149056 | Urinary excretion of enterolactone, with an effect depending on ethnicity (European or African ancestry women) | 39 |
| COMT | rs4633 | ||
| PON1 | rs661, rs705381 | Urinary excretion of the sum of sulfated and glucuronidated forms of 3′,4′-dihydroxyphenyl-γ-valerolactone in women | 46 |
![]() | ||
| Fig. 6 Network-based representation of the overlapping SNPs involved in (poly)phenol adme among studies included in the present systematic review. Only interconnected SNPs are shown. | ||
The functional annotation of SNPs and related genes in terms of biological processes, molecular functions, and cellular components is pictorially shown in SI Fig. 1 and 2. Forty-five biological processes (SI Table 5 and SI Fig. 1), 19 molecular functions (SI Table 6 and SI Fig. 2), and one cellular component (SI Table 7) could be identified. The locations of SNPs along the human genome are displayed in Fig. 7 and SI Table 8.
In most cases, SNPs had been chosen and tested by the authors based on knowledge derived from previous in vitro, animal or in silico/computational studies or from clinical/epidemiological studies. Only one study39 devised a systematic methodology for selecting SNPs, integrating data-mining (by surveying the HuGE Navigator Database) and text mining (by comprehensively looking for all published genome-wide association studies, GWAS, of genes coding proteins involved in the metabolism of endo- and xenobiotics, across any phenotype).
In most studies,42–45,47–49 genotyping was carried out by TaqMan assay, which utilises the 5′ nuclease activity of Taq polymerase to generate a fluorescent signal during polymerase chain reaction (PCR), or similar techniques. In one study,40 “Kompetitive Allele Specific” PCR (KASP) and agarose gel electrophoresis were utilised, which enabled a fast, cheap, but high-quality and highly reliable bi-allelic SNP characterisation. In another study,38 genotyping was conducted by direct sequencing. Finally, one study39 genotyped by multiplexing (utilizing the MassARRAY technology and iPLEX Gold assay). Of note, only one study41 performed a GWAS. Only one study46 did not provide any detail about the genotyping technique.
Furthermore, two studies44,45 included only homozygous variant individuals for a specific variant of the COMT gene, excluding those heterozygous. Finally, concerning genetic assumptions, they were generally met. Only two studies40,43 found evidence of linkage disequilibrium and violation of the Hardy–Weinberg equilibrium, at least for a SNP and in a sub-group of the population recruited.
Goodness-of-fit statistics were rarely reported.47,48 In one study,47 the goodness of fit of the models was estimated by computing R2 and Q2 by 7-fold internal cross-validation, and the analysis of variance (ANOVA) of the cross-validated residuals (or CV-ANOVA). In a pharmacokinetics/nutrikinetics study,48 model selection was based on the objective function value, several goodness-of-fit indicators and plots, and a visual predictive check. A stepwise covariate model-building strategy was leveraged by utilising forward inclusion of covariates followed by backward elimination.
The enzyme catechol-O-methyltransferase (COMT), encoded by a gene located at 22q11.21 (genomic coordinates, 22: 19
941
772–19
969
975), is responsible for catalysing a major metabolic pathway of flavonoids and their gut microbiota metabolites, namely their O-methylation, which is the transfer of a methyl group from S-adenosylmethionine (SAM) to one of the hydroxyl groups of catechols, to produce O-methylated catechols and S-adenosyl-L-homocysteine (SAH). This biotransformation mainly occurs in the intestinal tract, liver, and kidney.55 Two isoforms of COMT have been demonstrated to exist: a soluble form of the protein, which is the predominant one in most tissues, and a membrane-bound form. The genetic polymorphism rs4680 is a common missense variant (G to A base change), resulting in a valine-to-methionine amino acid substitution at position 108 in the soluble protein and at position 158 in the membrane-bound form. The allele rs4680-A, which can be found in insulin-resistant and type 2 diabetics, seems to alter the function of the COMT enzyme by decreasing its thermostability and resulting in a 30–40% reduction of its activity.55
Six studies38,42–45,48 explored the impact of COMT-related genetic variations on the metabolism of green tea (gallo)catechins. In the study by Brown et al.,38 the authors recruited 83 subjects who consumed decaffeinated green tea extract (about six to eight cups of moderate-strength green tea daily). Plasma concentrations of EGCG were undetectable at baseline and remained undetectable after dietary supplementation in the placebo group but increased in the experimental group. Urinary excretion over 24 h of EGC and 4′-O-methyl-EGC was similar for the two groups at baseline, but post-intervention increased in the experimental group by 28-fold and 34-fold, respectively. Mean urinary concentrations of EGC and 4′-O-methyl-EGC were significantly lower (p < 0.01) in individuals homozygous for the high-activity COMT G-allele (rs4680-G), potentially reflecting increased metabolic flux and a more rapid turnover and conversion to downstream metabolites compared to individuals carrying at least one copy of the low-activity COMT A-allele.
In the study by Inoue-Choi et al.,42 660 subjects, self-identified as daily green tea drinkers, were recruited from four small, geographically defined communities from the Greater Shanghai metropolitan area, and stratified according to COMT genotypes (rs4680): 343 carried the homozygous wild-type genotype (GG), 49 the homozygous variant genotype (AA), and 268 the heterozygous genotype (GA). The enrolled subjects not only geographically but also genetically belonged to the Asian area, in accordance with frequencies reported in the database (f(G) = 0.72 and f(A) = 0.27). Comparing the three COMT genotype groups, nominal statistically significant differences could be found for the five (poly)phenol metabolites under study when considering each one individually (EGC, EC, 4′-O-methyl-EGC, 5-(3′,4′-dihydroxyphenyl)-γ-valerolactone, 5-(3′,4′,5′-trihydroxyphenyl)-γ-valerolactone). Considering all the metabolites altogether, the difference was highly significant (p = 0.007). More specifically, the authors found that those carrying the homozygous low-activity genotype had lower urinary levels of five tea (poly)phenol metabolites by 35–45%, compared to those with the wild type high-activity genotype or the heterozygous variant genotype (who had, on the contrary, comparable levels of urinary metabolites). The authors speculated that carriers of the homozygous genotype could retain more green tea (poly)phenols in their bodies and benefit more from green tea intake. Of note, when stratifying according to daily green tea consumption, low green tea consumers (<5 g day−1) had lower urinary metabolite concentrations in both homozygous low-activity and heterozygous genotype groups, compared with the high-activity homozygous wild-type genotype, whilst the difference between high- and intermediate-activity COMT genotypes (genotypes GG and GA) tended to disappear with increasing green tea consumption (p = 0.18).
Conversely, Lorenz et al.43 found no impact of COMT genotype on EGCG plasma levels. The authors explored the enzymatic activities of four functional COMT SNPs determined in red blood cells in a sample of 24 healthy human volunteers supplemented with pure EGCG. The analysed SNPs were rs4680 (472G>A, Val158Met), rs6269 (1-98A>G), rs4633 (186C>T, His62=) and rs4818 (408C>G, Leu136=). EGCG plasma levels and COMT enzyme activities in erythrocytes were measured before and two hours after intervention. While enzymatic COMT activities were affected by the COMT SNPs (p < 0.001), EGCG plasma levels significantly increased after the intervention and were not influenced by any of the COMT SNPs and different enzyme activities. Furthermore, EGCG ingestion did not result in impairment of COMT activity, which significantly increased by 24% after EGCG consumption.
Similarly, the study by Miller et al.44 found no influence of COMT genotype. The authors recruited 20 participants who were exposed to decaffeinated green tea extract. Maximum plasma concentrations of 1.09 μM, 405, 331, 160, and 77.1 nM were reached for EGCG, EGC, EC, ECG, and 4′-O-methyl EGCG at 81.5, 98.5, 99.0, 85.5 and 96.5 min, respectively. Bimodal and single-peaked curves could be observed for the non-gallated and gallated green tea catechins, respectively, probably reflecting a meal effect from the high-carbohydrate breakfast administered 1 hour after the capsule consumption. It is hypothesised that galloylated species, different from non-galloylated ones, may form complexes with the food components, resulting in a single peak being observed. Other possible explanations could be a depletion in glucuronidation precursors in the fasted status. No statistically significant differences based on COMT genotype could be found in terms of maximum concentration, time to reach maximum concentration, AUC, EGCG half-life absorption, and EGCG half-life elimination. However, a trend towards a greater maximum concentration and AUC response in the plasma concentration curve for 4′-O-methyl-EGCG in the COMT AA genotype group could be noted. Also, a tendency towards a higher maximum plasma concentration for the AA genotype for EC, EGC and ECG was detected, along with higher median concentrations of 4′′-O-methyl-EGCG, which was detected and quantified only in a few samples.
The authors were able to replicate their findings in another, more extensive study45 with 47 participants. A genotypic effect was observed for urinary 4′-O-methyl-EGC during the first 5.5 h, with the COMT GG group exhibiting a greater concentration (p = 0.049) than the other 2 genotypes, even if marginally. No differences could be found for the remaining 18.5 h collection or for total EGC during the 5.5 h and 18.5 h collections. Finally, no difference in concentrations of plasma metabolites (C, EC, EGC, ECG, EGCG, 4′-O-methyl-EGCG, 4′′-O-methyl EGCG, 4′,4′′-O-dimethyl-EGCG) could be detected. According to the authors, these findings are coherent with the hypothesis of a slower methylation enzymatic function in the COMT AA genotype individuals, as well as with a differential preference for methylation position (“regioselectivity”) induced by the genetic variation, which could explain why, in the COMT GG group, secondary and tertiary methylation products, such as 4′,4′′-O-dimethyl EGCG and 3′,4′,4′′-O-trimethyl EGCG, are more likely to be detected with respect to primary methylation products.44,45 On the other hand, all these differences are, overall, slight and not statistically significant, suggesting that green tea (gallo)catechins, having low bioavailability and being poorly absorbed in the small intestine and quickly eliminated, may have better enzyme–substrate interactions and binding affinity than endogenous substrates, which could counteract, at least partly, the effects of the polymorphisms.
Whilst COMT genotype as a potential genetic factor mediating green tea exposure/intake and metabolic response has been the most investigated, other studied SNPs are related to genes coding members of the family of drug transporters, which include uptake (Organic Anion Transporting Polypeptide type 1B1, SLCO1B1) and efflux (Multi-drug Resistance-associated Protein type 2, ABCC2) transporters.56 The former is specifically expressed in the liver, on the basolateral membrane of hepatocytes, whereas the latter is an ATP-binding cassette protein and is expressed in several human tissues, being of particular importance for intestinal drug absorption and hepatic drug elimination.
In the nutrikinetics study by Scholl et al.,48 84 healthy participants took green tea extract capsules for 5 days. On day 5, plasma profiles for EGCG, EGC and ECG were obtained by collecting venous blood samples 0.5, 1, 2, 3, 4, 5, 7, and 9 hours after the last green tea extract capsule consumption. A substantial between-subject variability in pharmacokinetics was found, with maximum plasma concentrations varying more than 6-fold (6.1, 7.7 and 6.6-fold for EGCG, EGC and ECG, respectively). For EGCG and ECG, the highest inter-individual variabilities concerned the zero-and first-order absorption processes, whilst for EGC, the highest interindividual variability regarded the central volume of distribution and the intercompartmental distribution. The AUCs of EGCG, EGC and ECG were 877.9 (360.8–1576.5), 35.1 (8.0–87.4), and 183.6 (55.5–364.6) μg L−1 × h respectively, and the elimination half-lives were 2.6 (1.8–3.8), 3.9 (0.9–10.7) and 1.8 (0.8–2.9) hours, respectively. Metabolite concentrations were found to decline in a biexponential fashion. Genetic polymorphisms in genes of the drug transporters ABCC2 and SLCO1B1 were found to explain, at least partly, the high variability in pharmacokinetic parameters. More specifically, carriers of the C allele of ABCC2 rs717620 (-24C>T) exhibited 26% less EGCG relative bioavailability than carriers of the variant allele T (p = 0.00721). Additionally, total body clearance of EGCG was lower in carriers of the C allele of rs4149056 (521T>C, Val174Ala) (p = 0.01212). Concerning EGC, its pharmacokinetics was affected by MRP2 rs3740066 (3972C>T, Ile1324=) (p = 0.00153, a higher intercompartmental clearance in carriers of the C allele), OATP1B1 rs2306283 (388A>G, Asn130Asp) (p = 0.00163, a 35% reduction in total body clearance in carriers of the A allele), ACBC2 rs717620 (-24C>T) (p = 0.01654, a reduction in central volume of distribution in carriers of the T allele), UGT1A*28 or rs8175347 (p = 0.01795, a 26% reduction in total body clearance in wildtype carriers), and COMT rs4680 (p = 0.02350, a 24% reduction in relative bioavailability in carriers of the low-activity AA genotype). Of note is that habitual tea drinking was not found to impact the kinetics of any of the metabolites measured.
In the study by Chang et al.,39 ground brown flaxseed consumption for 6 weeks in post-menopausal women resulted in increased urinary enterolactone excretion, which was higher in European ancestry women. Associations between urinary phytoestrogen enterolactone excretion and 70 functional polymorphisms in 29 steroid hormone metabolizing genes, including genes putatively involved in (poly)phenol excretion and phase II metabolism (SLCO1B1, COMT, UGT1A1, SULT1A1), were explored. In women of European ancestry, 12% and 17% of the variation in baseline and post-intervention excretion levels was explained by genetic variants. In African ancestry women, this percentage was 16% and 13%, respectively. These associations were more marked among women of African ancestry than among those of European ancestry. However, these associations were only nominal (p < 0.05), failing to achieve the statistical significance threshold after correction for multiple comparisons. Of note, no SNPs were associated in both ethnic groups.
Wakeling and Ford49 recruited a sample of 100 pre-menopausal women. The participants in the “Soy Isoflavone Metabolism Study” consumed a commercial soy supplement as a single bolus dose. The authors measured the urinary levels of isoflavone metabolites (aglycones, 7-glucuronides, and sulfates of genistein, daidzein, and glycitein) and their relationships with UGT1A1*28 promoter polymorphism and SNPs in other genes involved in absorption and excretion (GBA3, LCT, ABCC2, and ABCG2). ABCG2/BCRP (Breast Cancer Resistance Protein) is an efflux transporter belonging to the ATP-binding cassette family and is present in the luminal membrane of the intestine. Urine was collected over 24 h. Large inter-individual differences in isoflavone recovery (mean 39% [range 11–89%], eightfold variation) and metabolites could be observed, with glucuronides representing the primary metabolites (72% of total). No statistically significant association between the UGT1A1*28 polymorphism (rs8175347) and net recovery in the urine of any individual isoflavone metabolite under study could be detected. However, the polymorphism showed other significant associations. In more detail, the UGT1A1*28 minor allele was found to be positively associated with the percentage of glycitein excreted in urine as sulfate (p = 0.046), and, after excluding 5 participants with both minor alleles (A allele) of GBA3 and ABCG2, due to a statistically significant interaction (p = 0.025), it was positively associated with the percentage of glycitein as sulfate in urine (p = 0.014), negatively with percentage of glycitein as glucuronide (p = 0.028), positively with combined isoflavones as sulfate (p = 0.035), and positively with sulphate-to-glucuronide ratio for combined isoflavones (p = 0.036). Finally, SNPs in GBA3 (rs358231), ABCG2 (rs2231142), and ABCC2 (rs2273697) were also found to correlate with differences in isoflavone metabolites in urine. In detail, GBA3 rs358231 (1368T>A, Tyr456SeM) was associated with the percentage of total isoflavone recovered in urine as glucuronide (p = 0.035), ABCG2 rs2231142 (421C>A, Lys141Gln) was negatively associated with the percentage of total isoflavone recovered in urine as glucuronide (p = 0.042), and ABCC2 rs2273697 (1249C>A, Val141Ile) was associated with total glycitein excretion (p = 0.034) and total genistein excretion (p = 0.019).
Equol, or 4′,7-isoflavandiol, is a nonsteroidal estrogen derived from the metabolism of isoflavones by the gut microbiota.65 Equol production occurs in approximately 25–30% of the adult population of Western countries, while this percentage reaches 50–60% when considering adults from Japan, China, and Korea or Western adult vegetarians. These differences are due to variations in the intestinal bacterial composition,66 whereas the contribution of the genetic makeup of the individual is less known. Inter-individual differences in equol production could explain the differential impact of soy intake on health-related outcomes, especially cardiovascular health.65,67,68 Hong et al.41 attempted to fill in this knowledge gap, by performing a GWAS of the equol-producing phenotype in a sample of 1391 Koreans in the context of free diet. In the study population, 70.1% of the participants were equol-producers and exhibited significantly lower blood pressure than nonproducers. Five SNPs identified in the HACE1 gene (HECT Domain And Ankyrin Repeat Containing E3 Ubiquitin Protein Ligase 1) were significantly associated with equol production, the most significant of which was rs6927608: more specifically, individuals with a minor allele of this SNP (allele C) were less likely to produce equol and displayed more elevated systolic blood pressure values. HACE1 plays a crucial role in a variety of biological functions and processes, including tumour suppression, by hindering tumour cell proliferation and facilitating the programmed cell death of tumour cells.69 Moreover, it safeguards the heart, mitigates against oxidative stress, and regulates cellular dynamics.70 The authors speculated that HACE1 could be involved in host intestinal immune responses and the maintenance of the pool of equol-producing bacteria, thus mediating the impact of the equol-producing phenotype on heart health.41
Fraga and coauthors40 investigated the impact of four SNPs in three genes involved in phase II metabolism or excretion (SULT1A1, SULT1C4, ABCC2). A total of 46 volunteers ingested a single dose of orange juice. Based on the urinary excretion of phase II hesperetin and naringenin metabolites, 25 and 21 subjects were categorised as high- and low-excretors, respectively. A positive, statistically significant relationship between three out of the four SNPs under study (SULT1A1 rs4788068, SULT1C4 rs1402467 and ABCC2 rs8187710) and the excretion of phase II flavanone metabolites was found.
Another study46 explored the impact of polymorphisms in the gene PON1 on the performance of an emerging biomarker of flavan-3-ol intake, namely the sum of two gut microbial metabolites specific for this class of flavonoids: 5-(4′-hydroxyphenyl)-γ-valerolactone-3′-sulfate and 5-(4′-hydroxyphenyl)-γ-valerolactone-3′-glucuronide. The authors hypothesised the role of a family of human serum proteins known as paraoxonases (PONs) in catalysing the hydrolysis of PVLs into phenylvaleric acids (PVAs), thus affecting the levels of circulating PVLs and so their suitability as nutritional biomarkers. The study first consisted of a single acute intake, single arm, non-randomised intervention part, in which 13 healthy male volunteers were recruited and administered a flavan-3-ol-containing drink consisting of a fruit-flavoured beverage mix prepared with a flavan-3-ol-containing cocoa extract. A rapid transformation of PVLs into PVAs could be observed in serum samples collected ex vivo, with a half-life of approximately 9.8 ± 0.3 minutes. This conversion was mediated by the PON1 and PON3 isoforms, with PON enzymes also involved in processing the phase II metabolites of PVLs. This evidence was confirmed collecting and analysing urine samples: the majority of PVAs metabolites were conjugated in the same positions and with the same moieties (methyl groups, sulfates, and glucuronides) as their PVL precursors. In the second part of the study, an ancillary analysis of the EPIC-Norfolk cohort study, five common PON1 genetic variations (rs662, rs854560, rs705379, rs705381, rs854572) were investigated to determine their impact on the urinary levels of a specific biomarker of flavan-3-ol intake (in a context of free diet), showing a limited influence on the inter-individual variations in biomarker levels. Although two SNPs were found to contribute to inter-individual differences, the overall genetic variation did not affect the performance of the biomarker, suggesting that the benefits of considering PON1 SNPs are negligible. As pointed out by the authors, further studies are needed to better understand the role of PON in flavan-3-ol metabolism and assess the impact of genetic variants on PVL excretion. Such studies should consider the great inter-individual variability in flavan-3-ol metabolism and in the subsequent PVL production, as reported in the literature.72–75
When considering observational studies or studies performed in a context of free diet, the association between (poly)phenol metabolites and specific SNPs could be affected by some biases, including the background diet, the time of urinary collection, and the convergence between metabolic pathways of (poly)phenols and other endogenous and exogenous aromatic compounds. Regarding background diet, a correction for (poly)phenol intake should be considered, since it can influence metabolite excretion hiding potential associations between metabolites and SNPs. Another potential bias could emerge when the urine collection time (with respect to the time of (poly)phenol exposure) is not clearly assessed. Finally, when evaluating the excretion of low-molecular weight microbial metabolites, their heterogeneous origin should be pointed out, since they can derive from endogenous and exogenous sources other than (poly)phenols,76 potentially influencing the relationship between their levels in biological fluids and the presence of genetic variants. Therefore, the best way to assess the association between (poly)phenol ADME and genetic background is to perform controlled intervention trials assessing the participants’ diet and collecting 24 h urine samples, which are more informative than morning spot urine.
Finally, concerning funding/sponsorship, no information was provided for one study.48 For the remaining studies, funding/sponsorship was provided by national councils/institutes,40,42,43,49 non-profit/public organisations,47 or companies with significant commercial interests in food products and/or beverages.38,44–46 In a study,39 one of the authors served as a consultant/advisory board member of the council of a body/institution with significant commercial interests in food products and/or beverages.
39 (8.3%) reached a high overall level of evidence, while 6
38,40,41,43,45,48 (50.0%) were moderate and 5
42,44,46,47,49 (41.7%) low. In terms of methodological quality, 2
39,48 (16.7%) studies were rated high, 9
38,40–47 (75.0%) moderate, and 1
49 (8.3%) low, whereas for study design, 4
38,39,41,45 (33.3%) were considered high, 5
40,43,44,48,49 (41.7%) moderate, and 3
42,46,47 (25.0%) low. Population directness and generalizability were high in 1
46 (8.3%), moderate in 6
39–41,43,45,48 (50.0%), and low in 5
38,42,44,47,49 (41.7%) of the studies. Regarding study directness (relatedness), 8
38–40,42,44,45,48,49 (66.7%) showed a high degree of relevance and 4
41,43,46,47 (33.3%) a moderate one. Statistical precision was high in 3
39,41,46 (25.0%), moderate in 6
38,40,42,47–49 (50.0%), and low in 3
43–45 (25.0%) of the studies, while consistency of results was uniformly moderate across all studies (12/12; 100%). Correction for plausible confounding was high in 5
38–40,45,48 (41.7%), moderate in 2
42,43 (16.7%), and low in 5
41,44,46,47,49 (41.7%), whereas effect sizes were moderate in 7
38–41,47–49 (58.3%) and low in 5
42–46 (41.7%), with no study achieving a high effect size. Publication or funding bias was moderate in 5
38,39,44–46 (41.7%) and low in the remaining 7
40–43,47–49 (58.3%). Biological plausibility was consistently moderate across all studies (12/12; 100%). Nutrient–dose response was low in 11
38–41,43–49 (91.7%) and moderate in only 1
42 (8.3%), while allele–dose response was moderate in 4
38,40,48,49 (33.3%) and low in 8
39,41–47 (66.7%).
Half of the studies investigated green tea flavan-3-ol ADME, using green tea, green tea extract, or pure EGCG as sources. Other studies explored the bioavailability and metabolism of flavanones, isoflavones, and lignans, present in orange juice, soy supplement, and ground brown flaxseed, respectively. Only one study analysed the impact of an unrestricted diet on the phenolic metabolome, and two other studies investigated the metabolism of flavan-3-ols and isoflavones deriving from a free diet, considering their specific gut microbial metabolites. These findings indicate that the investigation of the association between the variability in (poly)phenol ADME and genetic differences is limited and needs more research, focusing on other classes. Moreover, gallocatechins, flavanones, isoflavones, and lignans are not ubiquitous classes but characterize specific foods, being their intake in the daily diet restricted, suggesting that the sources considered in the present review are not so representative from a dietary point of view. Therefore, the study of (poly)phenol ADME and related SNPs should be widened by considering other classes, including flavan-3-ols of different origin from green tea, flavonols, and cinnamic acids, among others.
A total of 88 SNPs in 33 different genes were studied, a half of them related to xenobiotic metabolism and the remaining ones related to steroid hormone metabolism/activity and analysed in relation to phytoestrogens. In particular, two genes are involved in absorption (CBG/GBA and LCT), seven in phase I metabolism (PON1, CYP1A1, CYP1B1, CYP3A4, CYP3A5, CYP17A1, CYP19A1), four in phase II metabolism (COMT, UGT1A1, SULT1A1, SULT1C4) and four in excretion (ABCC2/MRP2, ABCG2/BCRP, ABCB1/PGP/MDR1, OATP1B1/SCLO1B1). Considering the genes analysed in the studies and their role in ADME process, phase I metabolism was the most representative phase (7 genes), followed by excretion (4 genes) and phase II metabolism (4 genes). This was unexpected as phase I metabolism is a very limited pathway when it comes to (poly)phenol metabolism, driven mainly by microbial catabolism and phase II conjugations.1–3 Going into detail, 6 genes coding for isoforms of cytochrome P450 were analysed in a single study39 investigating the impact of SNPs on phytoestrogen levels after lignan consumption. Taking into account the number of times a gene was studied in all works, the most studied phase resulted to be phase II metabolism, followed by excretion and phase I metabolism. Absorption was the least represented and studied phase, with only two genes studied in one work. These results are consistent with the crucial role of conjugation in the metabolism of (poly)phenols.
Considering the metabolites that were influenced by genetic variability, many of them were phase II conjugates, including methylated forms of gallocatechins (4′-O-methyl-EGC), sulfated and glucuronidated forms of isoflavones (glycitein, genistein, and daidzein), and sulfated, glucuronidated, sulfo-glucuronidated, and diglucuronidated forms of flavanones (naringenin and hesperetin). Other metabolites included algycones of (gallo)catechins (EGCG, EGC, EC) and isoflavones (glycitein, genistein, and daidzein), phenolic acids (3′,4′-dihydroxycinnamic acid, 4′-hydroxyhippuric acid), and gut microbiota catabolites (5-(3′,4′-dihydroxyphenyl)-γ-valerolactone, 5-(3′,4′,5′-trihydroxyphenyl)-γ-valerolactone, 5-(4′-hydroxyphenyl)-γ-valerolactone-3′-sulfate, 5-(4′-hydroxyphenyl)-γ-valerolactone-3′-glucuronide, enterolactone, equol, 3′,4′-dihydroxyphenylacetic acid, benzene-1,3-diol).
A total of 16 SNPs in genes involved in (poly)phenol ADME showed a significant modifying effect on urinary and/or plasma levels of phenolic metabolites and/or on their kinetic parameters. Regarding phase II metabolism, COMT genetic variants were analysed mainly in relation to gallocatechins, UGT1A1 genetic variants were associated with isoflavone phase II metabolites, and SULT1A1/SULT1C4 were explored together with flavanone phase II metabolites. The most studied genetic variation was rs4680 in the COMT gene. Glucosidases involved in (poly)phenol absorption (GBA3 and LCT) were analysed only in relation to soy isoflavones, while influx/efflux transporters involved in (poly)phenol excretion were investigated with all sources of (poly)phenols, suggesting their importance in determining (poly)phenol bioavailability. Finally, genetic variations of PON1 were analysed to determine their impact on the urinary levels of the two most important phenyl-gamma-valerolactone conjugates, used as biomarkers of flavan-3-ol intake. However, there was no consensus among studies allowing the association of a particular genetic variant with a reduction/increase of a specific metabolite in urine or plasma or with kinetic parameters.
The findings are conflicting and can only be partially reconciled by considering differences in methods and protocols. Most studies were conducted in controlled settings to minimize biases and confounders, but these do not always reflect real-world conditions, particularly in eating patterns. For example, Inoue-Choi et al.42 captured the wide variability in green tea consumption, with urinary samples timed based on pharmacokinetic studies rather than laboratory schedules. Other studies in this review may lack ecological validity. Additionally, some had small sample sizes, lacked replication, and did not adjust for confounders like seasonal food intake. SNP variations by ethnicity suggest more diverse populations are needed, with systems genetics or GWAS approaches offering promise.
Our understanding of how genetic variations influence the metabolism of dietary (poly)phenols is limited due to the lack of studies and knowledge about the functional effects of relevant SNPs. Specifically, we still do not fully grasp the extent of phase II metabolism, both locally within enterocytes and systemically in the liver. Given the complexity of dietary exposure, reductionist approaches fail to capture its heterogeneity. New integrative methodologies, combining in vivo and in vitro studies, wet-lab and computational techniques, nutritional epidemiology, and mathematical modelling (pharmacokinetics/nutrikinetics), are needed.77 These approaches can help uncover how specific genotypes affect the metabolism and health outcomes of ingested (poly)phenols, enabling personalized dietary recommendations.78,79 Most studied polymorphisms have shown minimal impact on metabolism, and polygenic scores may help clarify ADME genotypes. Additionally, differences in gastrointestinal motility and gut microbiota likely contribute to variability in plasma concentrations and urinary excretion of (poly)phenols.
The present systematic review has several strengths, including (i) high methodological rigor, (ii) transparency and reproducibility, (iii) extensive, comprehensive literature search conducted on several databases enhanced by cross-referencing and target journal hand-search, a critical approach, quality appraisal, and (iv) complemented by bioinformatics analyses. On the other hand, it has a few shortcomings that should be properly acknowledged, including the relatively small number of studies included and the high degree of heterogeneity that hinders a meta-analysis. Additionally, exploring the connection between genetic variants and (poly)phenol bioavailability is difficult for several reasons: (i) the absence of validated and reliable biomarkers of intake for most classes of (poly)phenols, which makes it hard to identify key phenolic metabolites that explain how genetics contribute to the variability in their bioavailability; (ii) the presence of confounding factors, such as gut microbiota composition and activity, which can affect the variation in (poly)phenol ADME among individuals, especially when considering gut microbial metabolites; and (iii) the fact that personalized nutrition approaches with (poly)phenols should not only consider genetics but also incorporate other factors influencing variability like age, sex, gut microbiota, dietary habits, and more. In particular, the gut microbiota composition and activity in the main determinant of the heterogeneity in (poly)phenol colonic catabolism, acting as a possible confounder when analysing associations between SNPs and gut microbial metabolites.
The research in the field should be increased and improved in different ways: (i) considering (poly)phenol classes that are more representative of the population daily intake, such as flavan-3-ols, flavonols, anthocyanins and cinnamic acids; (ii) investigating less explored phases of the metabolism (absorption, distribution, excretion); (iii) increasing the sample size and exploring more diverse populations from a genetic point of view (ethnicity); (iv) considering confounding factors (sex, age, smoking, alcohol, drugs, habitual diet) and acknowledging the factors that could contribute to the inter-individual variability in (poly)phenol bioavailability (gut microbiome, lifestyle, pathophysiological status), potentially hiding associations between ADME and SNPs; (v) highlighting possible limitations that could hinder the analysis, including the lack of reliable and validated biomarkers of (poly)phenol intake for most classes; (vi) analysing more genes and SNPs in relation to a specific (poly)phenol class, also implementing untargeted approaches such as GWAS and new methodologies such as systems genetics and polygenic risk scores.
Supplementary information (SI) is available. See DOI: https://doi.org/10.1039/d5fo03349g.
Footnote |
| † They share co-first authorship. |
| This journal is © The Royal Society of Chemistry 2025 |