ArticlePDF AvailableLiterature Review

Flavonoid-statin interactions causing myopathy and the possible significance of OATP transport, CYP450 metabolism and mevalonate synthesis

Authors:

Abstract

3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase inhibitors, statins, are a primary treatment for hyperlipidemic cardiovascular diseases which are a leading global cause of death. Statin therapy is life saving and discontinuation due to adverse events such as myotoxicity may lead to unfavourable outcomes. There is no known mechanism for statin-induced myotoxicity although it is theorized that it is due to inhibition of downstream products of the HMG-CoA pathway. It is known that drug-drug interactions with conventional medicines exacerbate the risk of statin-induced myotoxicity, though little attention has been paid to herb-drug interactions with complementary medicines. Flavonoids are a class of phytochemicals which can be purchased as high dose supplements. There is evidence that flavonoids can raise statin plasma levels, increasing the risk of statin-induced myopathy. This could be due to pharmacokinetic interactions involving hepatic cytochrome 450 (CYP450) metabolism and organic anion transporter (OATP) absorption. There is also the potential for flavonoids to directly and indirectly inhibit HMG-CoA reductase which could contraindicate statin-therapy. This review aims to discuss what is currently known about the potential for high dose flavonoids to interact with the hepatic CYP450 metabolism, OATP uptake of statins or their ability to interact with HMG-CoA reductase. Flavonoids of particular interest will be covered and the difficulties of examining herbal products will be discussed throughout.
Flavonoid-statin interactions causing myopathy and the possible significance of
OATP transport, CYP450 metabolism and mevalonate synthesis
Joshua Zechner1, Susan M. Britza1, Rachael Farrington1, Roger W. Byard1,2, and Ian F. Musgrave1
1. Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
2. Forensic Science SA, Adelaide, SA, 5000, Australia
Corresponding Author:
Mr Joshua Zechner,
Faculty of Medical Science
Level 3 Medical School South Building,
The University of Adelaide, Frome Road,
Adelaide 5005, Australia
Email: joshua.zechner@adelalaide.edu.au
Word count: 4281
Abstract: 213
Figure count: 5
Table count: 1
Abstract:
3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase inhibitors, statins, are a primary treatment for hyperlipidemic
cardiovascular diseases which are a leading global cause of death. Statin therapy is life saving and discontinuation due to adverse
events such as myotoxicity may lead to unfavourable outcomes. There is no known mechanism for statin-induced myotoxicity
although it is theorised that it is due to inhibition of downstream products of the HMG-CoA pathway. It is known that drug-drug
interactions with conventional medicines exacerbate the risk of statin-induced myotoxicity, though little attention has been paid to
herb-drug interactions with complementary medicines. Flavonoids are a class of phytochemicals which can be purchased as high
dose supplements. There is evidence that flavonoids can raise statin plasma levels, increasing the risk of statin-induced myopathy.
This could be due to pharmacokinetic interactions involving hepatic cytochrome 450 (CYP450) metabolism and organic anion
transporter (OATP) absorption. There is also the potential for flavonoids to directly and indirectly inhibit HMG-CoA reductase
which could contraindicate statin-therapy. This review aims to discuss what is currently known about the potential for high dose
flavonoids to interact with the hepatic CYP450 metabolism, OATP uptake of statins or their ability to interact with HMG-CoA
reductase. Flavonoids of particular interest will be covered and the difficulties of examining herbal products will be discussed
throughout.
Keywords:
CYP450; OATP; Statins; Flavonoids; Myopathy; Herbal Supplements
3
1. Introduction:
Hypercholesterolaemia is a condition characterised by high plasma levels of low-density lipoproteins (LDL) and low levels of
high-density lipoproteins. It is associated with a high risk of forming atherosclerotic plaques, thus resulting in potentially fatal
cardiovascular diseases[1]. The primary treatment for hypercholesteremia is statin therapy[2, 3]. Statins act by inhibiting the rate
limiting enzyme 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase which causes reduced hepatic biosynthesis of
HMG-CoA into mevalonate, a crucial step in cholesterol synthesis[4]. By decreasing hepatic cholesterol LDL receptors are
induced through a feedback loop and therefore increased LDL uptake within the liver occurs[4, 5]. This results in reduced serum
levels of LDL, making statins an effective treatment for hypercholesterolaemia[6].
Cardiovascular diseases are estimated to cause 31% of deaths globally per year making them the leading cause of death according
to the World Health Organization[7]. To combat this, statin therapy has become the most commonly prescribed drugs in the
world. For example, in Australia statins are used by approximately 25% of the population aged 65-years or over, with atorvastatin,
rosuvastatin and simvastatin being the most prescribed[2, 8]. Patient mortality is estimated to be decreased by 31% and 40% for
stroke and myocardial infarction, respectively, by statin therapy[9, 10].
Statins are well tolerated but there are rare cases of statin-induced side effects such as myopathies which may progress to
potentially fatal rhabdomyolysis[11-15]. Myopathy is defined as muscular symptoms, such as pain and tenderness with elevation
of creatinine kinase plasma levels ≥10x the upper normal limit; rhabdomyolysis may be defined as muscular symptoms with renal
impairment and creatine kinase plasma levels ≥40x the upper normal limit[12, 15]. The incidence of statin-induced
rhabdomyolysis has been estimated to be approximately 0.40-1.10 in 10,000 person years[11-15]. Most statins have a comparable
potential for developing myopathy or rhabdomyolysis, however, cerivastatin which was removed from the market in 2001, has
been associated with more cases of myotoxicities[12, 14, 16]. The risk for statin-induced myotoxicity is increased with higher
blood plasma concentrations[13, 17].
2. Flavonoids:
Flavonoids are a broad class of phytochemicals containing many subclasses such as flavones, flavonols, isoflavones, flavanones,
chalcones and flavan-3-ols (Figure 1)[18]. These are abundant in nature, commonly occurring in plant material which includes
fruits, vegetables, and complementary medicines (Table 1)[18].
4
Figure 1: Core flavonoid structure and 10 structures of different flavonoids. Adapted from Panche et al[18], Fan et al[19]
and Cione et al[20].
Within Australia it is estimated that 47.8%, 9.5% and 7.5% of people will use either vitamin/mineral supplements, western or
Chinese herbals or flower essences respectively, which are likely to contain flavonoids, this data could be considered to be
broadly representative of the western experience[21, 22]. The geriatric population seems to be the most vulnerable to flavonoid-
statin interactions. Not only are they the primary age group receiving statin therapy, but they are also more likely to be using
complementary medicines with approximately 25% reporting having used some form of complementary medicine in 2017[21,
23].
The average dietary daily intake of total flavonoids in Australia has been estimated to be 626 ± 579mg a day[24]. Dietary
flavonoids could modulate the pharmacokinetics of statins as reviewed by Peluso et al[25] but there has been few investigations
concerning the potential effects of co-administering high dose herbal supplements with statins, which can contain up to ≥1000mg
of flavonoids[26, 27].
Plasma concentrations achieved when taking high dose flavonoids are poorly understood even though high dose supplements are
readily available by over-the-counter sales. The studies that do exist do not investigate long term usage and co-administration with
other drugs.
5
Table 1: List of flavonoids, their subclass, and some examples of flavonoid-containing plant sources
Flavonoid
Subclass
Plant
References
Apigenin
Flavone
Celery and garlic
Hollman and Arts[28].
Miean and Mohamed[29].
Biochanin A
Isoflavone
Red clover (Trifolium
pratense) and chickpeas
Křížová et al[30].
Gao et al[31].
Epigallocatechin Gallate
Flavan-3-ol
Green tea (Camellia sinensis)
and cocoa
Cione et al[20].
Genistein
Isoflavone
Soybean and chickpeas
Spagnuolo et al[32].
Hesperitin
Flavanone
Grapefruit, orange, lemon, and
bergamot
Gattuso et al[33].
Kaempferol
Flavonol
Spinach, cabbage, and
cauliflower
Sultana and Anwar[34].
Licochalcone A
Chalcone
Chinese liquorice (Glycyrrhiza
inflata)
Fan et al[19].
Luteolin
Flavone
Broccoli and bird’s eye chilli
Miean and Mohamed[29].
Naringenin
Flavanone
Grapefruit, orange, lemon, and
bergamot
Alam et al[35].
Gattuso et al[33].
Quercetin
Flavonol
Peas, onion, and apple
Sultana and Anwar[34].
Hollman and Arts[28].
3. Pharmacokinetic interactions:
3.1 CYP450 interactions
The statins simvastatin, lovastatin and atorvastatin are likely to be subject to drug-drug or herb-drug interactions involving
cytochrome 3A4 (CYP3A4) modulation[36, 37]. CYP3A4 inhibitors are known to raise the plasma concentrations of simvastatin
and atorvastatin[36, 37]. By reducing the metabolism of statins, CYP3A4 inhibitors increase statin systemic exposure and
therefore increase the chance for statin-induced myopathy[36-40]. This idea has been largely accepted by clinicians and
researchers though there are inconsistencies in the data. A large cohort study found a relative risk of 2.17 for hospitalisation due to
statin-induced rhabdomyolysis when concomitantly prescribed the potent CYP3A4 inhibiting antibiotics clarithromycin and
erythromycin compared to the non-inhibitor antibiotic azithromycin[38]. It should be noted that clarithromycin also exhibits
6
strong inhibition of organic anion transporting polypeptide 1B1 (OATP1B1) however the primary statin taken in this study was
atorvastatin, so the primary interaction was attributed to CYP3A4 inhibition[38, 41].
In contrast, a large retrospective study concluded that there was no significant difference for developing statin-induced myopathy
in the presence of a CYP3A4 inhibitor compared to statin therapy alone, this was regardless of the statin being a CYP3A4
substrate or not[42]. The primary concomitant drug taken in this cohort was diltiazem, a moderate CYP3A4 inhibitor that has
negligible effects on OATP1B1[42, 43]. Rowan et al[42] may have observed no significant differences between statin co-
administration and statin monotherapy as diltiazem might be too weak of a CYP3A4 inhibitor to cause significant adverse
reactions with statins or because statins are more likely to interact with inhibitors that target both CYP3A4 and transporters[42,
43]. For example, the in-vitro IC50 for CYP3A4 by diltiazem is reported to be between 110-127µM while the in-vitro IC50 for
CYP3A4 by clarithromycin IC50 is reported to be 56±5µM[44, 45]. This would need to be confirmed through further research.
Neither study investigated the concomitant use of herbal compounds[41, 42].
Many flavonoids have inhibited CYP3A4 in-vitro at comparable concentrations to clarithromycin. Naringenin was shown to
inhibit CYP450 metabolism of simvastatin with a Ki=25.48±10.10µM[46]. Biochanin A and genistein non-competitively inhibited
CYP3A4 with an IC50=65.11±3.97 and ~24.4µM respectively[47, 48]. Quercetin non-competitively inhibits CYP3A4 with an
IC50=5.5±0.7µM, making it one of the most potent inhibitors among the other flavonoids[49]. Epigallocatechin gallate (EGCG)
non-competitively inhibited CYP3A4 and CYP2C9 with IC50=23.7±1.54µM and 39.1±22.46µM, this would suggest that EGCG
would be likely to interact with fluvastatin, rosuvastatin, pravastatin and pitavastatin[50].
There are a limited number of studies investigating the co-administration of high dose flavonoids with statins in humans. A study
in rats receiving intravenous EGCG at 5mg/kg found that the area under the curve (AUC) of simvastatin and its metabolite
simvastatin acid were raised by 2.2 and 1.4-fold respectively[48]. This was likely due to EGCG inhibiting both CYP3A4
metabolism and OATP-uptake[48]. Similarly, 16 male humans co-administered 500mg of quercetin and pravastatin for 14 days,
had an increase in AUC of 1.24-fold compared to pravastatin alone[51]. The authors of this study did not investigate the
significance of CYP3A4, but it seems likely that OATP1B1 or CYP2C9 inhibition are the primary factors leading to increased
pravastatin serum levels as it is not susceptible to CYP3A4 interactions[37, 51].
3.2 OATP interactions
While the inhibition of CYP450 metabolism of statins has historically been the primary focus when investigating statin-induced
myopathy in more recent years inhibition of OATP uptake has been identified as an important risk factor. The significance of
inhibited OATP1B1 function in statin-induced myopathy has been highlighted in large genome studies. Those with the rs4149056
single nucleotide polymorphism within the SLCO1B1 gene that encodes OATP1B1 have reduced OATP1B1 statin uptake and
therefore reduced hepatic clearance of statins, with simvastatin being the most affected [17, 52]. Those taking 40mg of simvastatin
7
every day who were homozygous for this polymorphism had an odds ratio of 4.5 (CI 2.6-7.7) for developing myopathy, compared
to those without[17]. The review by Niemi[52] summarises that those homozygous with this polymorphism had an AUC increase
after a single dose of simvastatin, pitavastatin, atorvastatin, pravastatin or rosuvastatin by 2.21-fold, 1.62-1.91 fold, 1.44-fold,
0.57-1.30 fold, 0.62-1.17 fold respectively[52]. There was no significant effect by fluvastatin[52].
Much like the genomic studies, large cohort studies have identified the dangers of concomitant use of OATP1B1 inhibitors with
statins. For example, Gemfibrozil is an OATP1B1 inhibitor that has minimal effects on CYP3A4 and a potent inhibitory effect on
CYP2C9[53, 54]. It was found that those taking gemfibrozil with a statin had a calculated incidence rate ratio of 11.93 (CI 3.96-
35.93) for being hospitalised due to rhabdomyolysis compared to those taking a statin alone[55]. These results were similar to
those of Graham et al previously[13].
Recently a study investigating the effects of 99 flavonoids on OATP1B1 uptake of statins found that 8 flavonoids significantly
inhibited the uptake of fluvastatin[19]. The three most potent inhibitors were licochalcone A (IC50 = 7.96 μM), luteolin
(IC50 = 22.03 μM) and biochanin A (IC50 = 22.28 μM)[19]. Wang et al[56] found the most potent inhibitors of OATP1B1-
mediated uptake of [3H]dehydroepiandrosterone sulphate, to be biochanin A, genistein and EGCG with IC50=11.3±3.22µM,
14.9±3.76µM and 14.1±1.44 µM respectively. They also found the mechanism to be non-competitive inhibition[56]. Mandery et
al[57] found the flavonoids quercetin, kaempferol and apigenin exhibited competitive inhibition of OATP1B1. The Ki values for
quercetin, kaempferol and apigenin for the OATP1B1-mediated uptake of bromosulfophthalein were 8.8±0.8µM, 32.4±2.2µM and
53.4±2.1µM, respectively[57]. The authors[57] repeated these tests on the uptake of atorvastatin and interestingly found that the
Ki for apigenin had become 0.6±0.2µM while quercetin and kaempferol had remained relatively unchanged[57]. They suggest that
apigenin may form different inhibitor-receptor complexes dependent on the substrates. Further studies should be done to
investigate if flavonoids have differing inhibitory affinities with different statins.
It is difficult to say what flavonoids pose a greater risk for developing statin-induced myopathy until further studies are conducted
investigating flavonoid interactions for all statins. Though the pleiotropic inhibitory effects of flavonoids would put them at risk of
interacting with a range of statins.
4. Statin pharmacokinetics and drug interactions:
Plasma concentrations of statins are a result of the balance between cellular uptake and metabolic breakdown. Most statins rely on
OATP uptake, for cellular uptake and are metabolised by CYP450 in the liver[58, 59]. Simvastatin, lovastatin, and atorvastatin are
CYP3A4 substrates, while fluvastatin, rosuvastatin, pravastatin and pitavastatin are CYP2C9 substrates[37, 40, 59, 60].
Hydrophilic statins such as rosuvastatin and pravastatin are primarily dependent on OATP1B1 for entry into cells while lipophilic
statins can permeate through cell walls, though OATP1B1 still plays a role[58, 60].
8
Genetic polymorphisms with reduced CYP450 metabolism or OATP1B1 uptake, or concomitant use of drugs that inhibit these
processes can result in increased plasma statin concentrations and myopathy[13, 17, 36, 37, 39, 40]. For example, the fibrate,
gemfibrozil and the macrolide antibiotic, clarithromycin (OATP inhibitor and combined OATP and CYP450 inhibitor,
respectively) induced rhabdomyolysis when co-administered with statins[13, 38, 55].
5. Mechanisms of myotoxicity:
Currently no mechanism for statin-induced myopathy has been established though there are many hypotheses. Two of the leading
hypothesised mechanisms include dysfunctional mitochondria or decreased prenylation due to the inhibition of the HMG-CoA
pathway[60-62].
Evidence for the dysfunctional mitochondrial theory is linked to decreased coenzyme Q10 (CoQ10) levels which disrupt
mitochondria within skeletal muscle, releasing free radicals and thus resulting in myotoxicity (Figure 3)[61]. CoQ10 is a
downstream product of the HMG-CoA pathway which plays an important role in electron transport within the mitochondrial
respiratory chain[61]. It has been noted that there is a decrease in CoQ10 in serum levels of patients with muscle diseases[63].
Some studies as reviewed by Deichmann, Lavie and Andrews[61] have reported that statin users have had decreased CoQ10
serum levels and have suggested that this is a potential mechanism for statin-induced myotoxicity others have not found a link
between statin induced falls in CoQ10 serum levels and mitochondrial dysfunction. Many small-scale studies supplementing
CoQ10 within those suffering from statin-induced myotoxicity produced inconsistent results and therefore it has been accepted
that there is currently insufficient data to state whether CoQ10 is an underlying mechanism to statin-induced myopathy[61, 62].
Figure 2: Decreased CoQ10 disrupting mitochondria in skeletal muscle cell, hypothetically causing myotoxicity[61]
9
The hypothesis for statin inhibition of protein prenylation as a cause of myotoxicity is associated with decreased levels of
Geranylgeranyl pyrophosphate (GGPP). GGPP is a downstream product in the HMG-CoA pathway and plays a role in prenylation
of proteins which are vital for cell function[64]. In-vitro studies have found that statin use will inhibit GGPP activity and therefore
disrupt GTPases such as RhoA and Rab which require prenylation for cell membrane anchorage (Figure 3)[65, 66]. RhoA plays a
role in the cytoskeleton of myocytes by allowing actin to form stress fibres, while Rab is important in regulating vesicle transport
within the cytoplasm, both vital roles for cell survival (Figure 5)[66-68]. Sakamoto[66] and colleagues identified that fluvastatin
and pravastatin-inhibition of Rab lead to morphological changes of myocyte organelles including mitochondria thus ultimately
causing cell death, moreover, these morphological changes were attenuated in the presence of GGPP. Itagaki et al[65] found that
simvastatin and fluvastatin, but not pravastatin, induced skeletal muscle apoptosis in rats by caspase-3 activation. The authors
observed that RhoA had been displaced from the cell membrane into the cytosol and theorize that this may be an underlying
mechanism for the observed statin-induced myotoxicity as both apoptosis and RhoA displacement were attenuated in the presence
of GGPP[65].
Figure 3: Decreased GGPP resulting in decreased Rab and RhoA prenylation hypothetically resulting in decreased vesicle
membrane anchorage or actin stress fibre formation and cytoskeleton organisation respectively which both ultimately lead
to myotoxicity[65-67].
10
6. Pharmacodynamic interactions:
Flavonoids have been studied as potential HMG-CoA reductase inhibitors with the expected outcome that they may have less
adverse reactions than statins while effectively treating hypercholesteremia[69]. It is proposed, however, that the inhibition of the
HMG-CoA pathway may lead to myotoxic events which is why careful attention should be paid to high dose flavonoid intake and
muscle toxicities in animal and human trials. Currently there are some studies trying to elucidate the relationship of flavonoids
with HMG-CoA through molecular docking in-silico[70-72].
The flavonoids quercetin, EGCG and biochanin A inhibit HMG-CoA reductase directly or indirectly[70, 71, 73]. EGCG potently
binds to HMG-CoA reductase in liver microsomes with a Kd of 77.0 ± 12.0 nM through an allosteric site on HMG-CoA
reductase[71]. When EGCG is co-administered with pravastatin, a synergistic interaction achieves greater inhibition than
monotherapy with either compound[71]. Quercetin was found to inhibit HMG-CoA reductase by binding directly to the substrate
binding site with a more moderate binding affinity (Kd=38.8±13.2µM) in cultures of the human liver cancer cell line known as
HepG2 cells[70]. Whereas biochanin A indirectly inhibits HMG-CoA reductase activity by decreasing levels of mRNA for HMG-
CoA reductase and sterol regulatory element-binding protein 2, an important stimulus for HMG-CoA reductase activity[73].
Conjugate forms of the flavonoids naringenin and hesperetin contain HMG-CoA like moieties and modelling suggests are capable
of directly binding to the substrate site of HMG-CoA reductase resulting in inhibition, though their efficacy compared to statins is
yet to be determined experimentally[72].
Hypothetically if the inhibition of mevalonate synthesis is a risk factor for developing myopathy, high doses of flavonoids may be
contraindicated with statin therapy. Further studies are needed to evaluate combined therapy of flavonoids and statins, measuring
HMG-CoA reductase activity and testing whether these findings are constrained to in-vitro or in-silico settings.
7. Statin adverse events and phytochemical interactions:
Complementary medicines have been scrutinised less closely than conventional medicines for adverse reactions. The Australian
population has a high level of use of complementary medicines with the local industry estimated to be worth billions of dollars[21,
74]. Herbal medicines are one of the many forms of complementary medicines with 9.5% of Australians reporting use of herbal
products over a 12-month period[21]. Certain foods and herbal supplements have been shown to have severe adverse effects when
taken concomitantly with drugs, with the classic example being grapefruit juice[75]. It is known that consuming grapefruit with
either simvastatin, lovastatin or atorvastatin can raise statin serum levels, increasing the risk for statin-induced myopathies[75].
Citrus fruit juices such as grapefruit and lemon juice inhibit CYP3A4, and more recently has been shown to inhibit OATP1B1[57,
11
75, 76]. While grapefruit juice inhibition of CYP3A4 is due to furanocoumarins37, inhibition of CYP3A4 by other citrus fruit
juices and OATP1B1 by fruit juices generally has been attributed to a class of phytochemicals known as flavonoids[57, 76].
There are approximately 5000 known flavonoids that are found in foods and herbal supplements[77]. Flavonoids have been
investigated for potential therapeutic properties for a range of medical conditions and events such as cancer, menopause, and
cardiovascular disease[78, 79]. However, these herbal supplements need to be approached with caution as very little is known
about their pharmacological impact and how flavonoids may interact with medications at high dosages.
In terms of statins In-vitro studies have demonstrated that some flavonoids can inhibit both CYP450 and OATP1B1 (Figure 4)
which may have implications for statin pharmacokinetics, possibly leading to adverse events when concomitantly used with
statins at large doses[47, 56, 57, 80-82]. In vitro studies suggesting interactions are reinforced by animal and human trials that
have reported increased statin blood concentrations in those taking flavonoids[46, 48, 51, 57, 75].
Figure 4: Flavonoids inhibit hepatic OATP1B1 uptake and CYP450 metabolism of statins raising statin blood
concentration levels and potentially leading to muscle toxicity
Additionally, flavonoids can inhibit HMG-CoA reductase to varying degrees which is a proposed pathway in the development of
statin-induced myopathy, as described in the previous section, suggesting the potential for pharmacodynamic interactions (Figure
5)[70, 71, 73].
12
Figure 5: Statins and flavonoids both inhibit HMG-CoA reductase inhibiting coenzyme Q10 and geranylgeranyl
pyrophosphate which may lead to muscle toxicity
Finally, it has also been reported that polypharmacy of flavonoids increases their bioavailability, thus increasing systemic
exposure[81, 83].
Flavonoid rich foods and statin interactions have been reviewed by Peluso et al[25] but currently there is a lack of data
surrounding concomitant use of statins with herbal supplements, specifically those with high doses of flavonoids that inhibit both
CYP3A4 and OATP1B1 and the potential resultant adverse events. This review will summarise what is currently known about
flavonoids potential to interact with statins through hepatic OATP1B1 and CYP450 modulation as well as through the inhibition
of cholesterol synthesis. P-glycoprotein may also play a role but will not be covered in this review[84, 85].
8. Plasma flavonoid concentrations achievable via supplements:
Although flavonoids can potentially affect both the pharmacodynamics and pharmacokinetics of statins, the question arises as to
whether oral consumption of supplements will achieve the required concentrations in the body to produce adverse effects. EGCG
and quercetin are common flavonoids used in supplements likely to be consumed by people on statins, possibly even combined,
and examining their plasma concentrations may give some insight into the potential risk of interactions.
In fasting subjects EGCG reached a peak blood plasma level of 7.4 ± 3.6 µM after consuming a single dose catechin mixture
containing 1,200 mg EGCG[86]. Fed subjects achieved a lower plasma concentration of 2 ± 1.7 µM with the same dose[86]. The
13
high variability seen in this study is mirrored in another study following a large cohort taking either 0, 500mg or 1000mg of
quercetin as twice daily doses over 12-weeks saw large interindividual variation with peak blood plasma concentrations ranging
from 0-2.98 µM[87]. As the variations appeared to be idiosyncratic the authors speculated that differing transporter genotypes or
dietary intakes may play a role[87].
In terms of the potential inhibition of statin metabolism, peak EGCG concentrations in fasted subjects are 3 times lower than the
IC50 for CYP3A4, and peak fed concentrations around 10 times less, suggesting acute ingestion of even high levels of
supplements by themselves will not greatly alter statin concentrations[50, 86]. Chronic consumption of quercetin supplements
achieves plasma levels around the Ki for CYP3A4 (2.98 µM compared with Ki of 5.5 µM)[87, 88].
In contrast EGCG may produce plasma levels sufficient to inhibit OATP (7.4 µM vs IC50 of 14.1 µM) while quercetin was well
below the Ki for OATP1 (53.4 µM)[56, 86]. Nonetheless, administration of 500 mg quercetin once a day for 14 days with
pravastatin increased Cmax of pravastatin by 131%[51]. As many quercetin supplements are dosed at 500mg/day or greater
showing the potential for induction of adverse effects.
Furthermore, flavonoids are often given in combinations which may increase their impact of statin metabolism.
9. Herbal medicine composition and plasma flavonoid concentrations:
Herbal medicines are very complex as they may contain hundreds to thousands of compounds. It is often difficult to discern what
precise compounds are present and physiologically active due to collectively-labelled ingredients. For example, flavonoid
supplements may have their ingredients broadly listed as flavonoids or bioflavonoids, making it difficult for researchers to identify
potentially harmful ingredients.
To add further complexity, flavonoids occur with additional sugar moieties (this structural form is known as either a glucoside or
a glycoside) in plant material which have modified absorption within the intestine compared to their aglycone counter parts[89,
90]. An example of this was observed when human subjects consumed 300mg of a quercetin glucoside a naturally occurring
conjugate form of quercetin, quercetin-4’-O-β-D-glucoside[91]. This resulted in mean peak plasma quercetin levels of
9.72±1.38µM which is approximately three times higher than Jin et al[87] observed, as quercetin in its glucoside or glycoside
form appears to have favourable absorption in the intestinal membrane[87, 89-91].
Additionally, there is evidence that polypharmacy of flavonoids can increase flavonoid concentrations, further complicating
flavonoid-statin interactions. In-vitro it was observed that the amount of biochanin A present in the human colorectal
adenocarcinoma cell line, Caco-2 cells and HepG2 cells was increased by 1.51 and 3.06 times respectively in the presence of
quercetin and EGCG respectively, with significantly decreased conjugation in the caco-2 cells[81]. In-vivo it was found that
EGCG and quercetin promoted enterohepatic recycling of biochanin A, therefore increasing its systemic exposure[81]. The
14
authors proposed that multiple mechanisms, including inhibition of conjugation, CYP450s and efflux transporters involved in the
metabolism and excretion of biochanin A were responsible[81]. This is of importance as these flavonoids commonly occur within
the diet and have been investigated for treating cardiovascular diseases, potentially increasing the risk of being combined with
statins[92-94].
The more realistic approach of combined oral administration of flavonoids that commonly occur together in the diet or in
supplements highlights that flavonoid blood plasma concentrations may reach higher than expected level due to pharmacokinetic
interactions amongst each other[81]. Further research investigating whether flavonoids with high inhibitor constants for pathways
not only important for statin pharmacokinetics, but also other flavonoid pharmacokinetics are needed to see if the polypharmacy
of flavonoids can result in flavonoid-flavonoid interactions thus potentiating statin-flavonoid interactions.
10. Discussion:
Statins are the primary treatment for hypercholesterolaemia and the cessation of therapy due to unexplained statin intolerance
could be life threatening[8-10]. There is a risk of flavonoids being co-administered with statins as not only are they abundant in
the diet, but high dose supplements are also available for several conditions including cardiovascular diseases[69, 78, 79]. Despite
little evidence for them acting therapeutically.
Older Australians appear to be the most vulnerable to flavonoid-statin interactions as they are likely to use both statins and
supplements (such as bioflavonoid antioxidants) which can contain concentrations of flavonoids capable of inhibiting the uptake
and metabolism of statins[8, 21]. There are many studies focusing on statin-drug reactions, but it is uncommon for them to include
herbal supplement usage[13, 38, 42, 55]. Supplement intake should certainly be recorded while on life-saving medications to
allow retrospective studies to investigate all possible causes for discontinuation.
Though limited in size, several human trials investigating statin-flavonoid use found increased statin serum levels with some
statins, potentially raising the risk for statin-induced myopathy[48, 51]. Furthermore, while flavonoids such as quercetin, luteolin,
EGCG and biochanin A have been studied there are many more flavonoids that have not been studied available for purchase at
higher dosages than the studied flavonoids that have produced statin interactions.
Raised statin plasma levels are likely due to flavonoids inhibiting CYP3A4, CYP2C9 and OATP1B1[19, 46-50, 56, 57]. Among
the flavonoids studied, the most potent inhibitor for OATP1B1 was licochalcone with an IC50 = 7.96μM. Licochalcone is not sold
as a pure supplement but is present in Chinese liquorice (Glycyrrhiza inflata) herbal supplements.
Quercetin was the most potent inhibitor of CYP3A4 with an IC50 of 5.5±0.7µM while EGCG also inhibited CYP2C9 with an IC50
of 39.1±22.46µM [19, 49, 50]. The IC50 concentration for quercetin on CYP3A4 can be achieved in plasma after oral ingestion of
300mg of quercetin-4’-O-β-D-glucoside[91]. As many quercetin containing supplements have between 250-800 mg of quercetin
15
per dose there is significant potential for CYP3A4 inhibition. There is a lack of data pertaining to whether the IC50 values of
licochalcone and EGCG can be achieved while taking high dose supplements. Furthermore, data on the effect of supplements
containing multiple flavonoids is not available even though synergistic interactions are possible.
These flavonoid IC50 values are comparable to that of the known OATP1B1 and CYP3A4 inhibitor clarithromycin and thus
suggest a high risk of clinically important interactions, especially in those whose genetic background puts them at risk. Further
investigations should evaluate a range of flavonoids with each statin to find high risk flavonoid-statin combinations.
Additionally, the inhibition of HMG-CoA reductase may lead to pharmacodynamic interactions between flavonoids and statins.
The downstream products of mevalonate, CoQ10 and GGPP have been theorized to play a role in statin-induced myopathy[61, 65,
66]. The flavonoids quercetin, EGCG and Biochanin A as well as conjugates of naringenin and hesperitin have all been shown to
either directly or indirectly inhibit HMG-CoA reductase[70-73]. EGCG seems to be of major concern as it has a high binding
affinity of Kd=77.0 ± 12.0nM and can act synergistically with pravastatin[71]. Hypothetically EGCG could inhibit CoQ10 and
GGPP which could imply that the EGCG is capable of myopathy itself or promoting statin-induced myopathies. This would need
to be investigated in myocyte cultures in the presence and absence of either CoQ10 or GGPP. Quercetin and the conjugates of
naringenin and hesperitin bind directly to the substrate binding site of HMG-CoA reductase which may competitively inhibit
statin binding, but this would require further testing to confirm[70, 72].
Some of the major difficulties researchers experience when exploring the safety of herbal supplements is unclear labelling of the
ingredients present in supplements as well as there being many different compounds present in these products. There are very few
regulations surrounding ingredients lists for herbal supplements of flavonoids allowing for collective-labels such as flavonoids
and bioflavonoids which makes it difficult to identify harmful flavonoids in these supplements. Flavonoids can occur in different
compositions such as aglycones, glucosides and glycosides which can have differing bioavailabilities as mentioned above[89-91].
There is also evidence that flavonoid-flavonoid interactions can occur such as those seen in the Moon and Morris’[81] study
showing that biochanin A in the presence of quercetin and EGCG produced higher plasma levels than biochanin alone.
In conclusion, herbal supplements should be scrutinised more closely when there is evidence of physiological activity but lack of
evidence for safety, especially when co-administered with conventional medicines. Flavonoid dietary intake may not be an issue
with statins. However, high dose supplements that greatly exceed the average dietary flavonoid intake are available and their
concomitant use with statins remains to be fully understood[30]. Furthermore, there is evidence that suggests clinically significant
interactions are possible.
Author contributions:
J. Z. prepared and wrote the first draft. S. M. B., R. F., R.W.B., and I. F. M. critically revised the draft. All authors approved the
final version.
16
Declaration of competing interest:
The authors declare that there are no conflicts of interest.
Acknowledgements:
Supported by the University of Adelaide Postgraduate Student Support (project ID: 15116917). J. Z. holds a University of
Adelaide PhD scholarship. The University of Adelaide did not direct the conduct or interpretation of this research.
References:
1. Gofman JW & Lindgren F (1950). The role of lipids and lipoproteins in atherosclerosis. Science 111, 166-71. DOI:
10.1126/science.111.2877.166.
2. Department of Health (2020). PBS Expenditure and Prescriptions Report 1 July 2019 to 30 June 2020. Accessed January
2021 [http://www.pbs.gov.au/info/browse/statistics].
3. Schaiff RAB, Moe RM & Krichbaum DW (2008). An overview of cholesterol management. Am Health Drug Benefits 1,
39-48.
4. Alberts AW, Chen J, Kuron G, Hunt V, Huff J, Hoffman C, Rothrock J, Lopez M, Joshua H, Harris E, Patchett A,
Monaghan R, Currie S, Stapley E, Albers-Schonberg G, Hensens O, Hirshfield J, Hoogsteen K, Liesch J & Springer J
(1980). Mevinolin: a highly potent competitive inhibitor of hydroxymethylglutaryl-coenzyme A reductase and a
cholesterol-lowering agent. Proc Natl Acad Sci U S A 77, 3957-61. DOI: 10.1073/pnas.77.7.3957.
5. Bilheimer DW, Grundy SM, Brown MS & Goldstein JL (1983). Mevinolin and colestipol stimulate receptor-mediated
clearance of low density lipoprotein from plasma in familial hypercholesterolemia heterozygotes. Proc Natl Acad Sci U S
A 80, 4124-8. DOI: 10.1073/pnas.80.13.4124.
6. Mabuchi H, Haba T, Tatami R, Miyamoto S, Sakai Y, Wakasugi T, Watanabe A, Koizumi J & Takeda R (1981). Effect
of an inhibitor of 3-hydroxy-3-methyglutaryl coenzyme A reductase on serum lipoproteins and ubiquinone-10-levels in
patients with familial hypercholesterolemia. N Engl J Med 305, 478-82. DOI: 10.1056/nejm198108273050902.
7. World Health Organization (2017). Cardiovascular diseases (CVDs). Accessed February 2021
[https://www.who.int/en/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds)].
8. Ofori-Asenso R, Ilomaki J, Zomer E, Curtis AJ, Zoungas S & Liew D (2018). A 10-Year Trend in Statin Use Among
Older Adults in Australia: an Analysis Using National Pharmacy Claims Data. Cardiovasc Drugs Ther 32, 265-72. DOI:
10.1007/s10557-018-6794-x.
17
9. Penning-van Beest FJ, Termorshuizen F, Goettsch WG, Klungel OH, Kastelein JJ & Herings RM (2007). Adherence to
evidence-based statin guidelines reduces the risk of hospitalizations for acute myocardial infarction by 40%: a cohort
study. Eur Heart J 28, 154-9. DOI: 10.1093/eurheartj/ehl391.
10. Sacks FM, Pfeffer MA, Moye LA, Rouleau JL, Rutherford JD, Cole TG, Brown L, Warnica JW, Arnold JM, Wun CC,
Davis BR & Braunwald E (1996). The effect of pravastatin on coronary events after myocardial infarction in patients
with average cholesterol levels. Cholesterol and Recurrent Events Trial investigators. N Engl J Med 335, 1001-9. DOI:
10.1056/nejm199610033351401.
11. Coste J, Billionnet C, Rudnichi A, Pouchot J, Dray-Spira R, Giral P & Zureik M (2019). Statins for primary prevention
and rhabdomyolysis: A nationwide cohort study in France. Eur J Prev Cardiol 26, 512-21. DOI:
10.1177/2047487318776831.
12. Garcia-Rodriguez LA, Masso-Gonzalez EL, Wallander MA & Johansson S (2008). The safety of rosuvastatin in
comparison with other statins in over 100,000 statin users in UK primary care. Pharmacoepidemiol Drug Saf 17, 943-52.
DOI: 10.1002/pds.1603.
13. Graham DJ, Staffa JA, Shatin D, Andrade SE, Schech SD, La Grenade L, Gurwitz JH, Chan KA, Goodman MJ & Platt R
(2004). Incidence of hospitalized rhabdomyolysis in patients treated with lipid-lowering drugs. Jama 292, 2585-90. DOI:
10.1001/jama.292.21.2585.
14. Chang JT, Staffa JA, Parks M & Green L (2004). Rhabdomyolysis with HMG-CoA reductase inhibitors and gemfibrozil
combination therapy. Pharmacoepidemiol Drug Saf 13, 417-26. DOI: 10.1002/pds.977.
15. Stroes ES, Thompson PD, Corsini A, Vladutiu GD, Raal FJ, Ray KK, Roden M, Stein E, Tokgözoğlu L, Nordestgaard
BG, Bruckert E, De Backer G, Krauss RM, Laufs U, Santos RD, Hegele RA, Hovingh GK, Leiter LA, Mach F, März W,
Newman CB, Wiklund O, Jacobson TA, Catapano AL, Chapman MJ & Ginsberg HN (2015). Statin-associated muscle
symptoms: impact on statin therapy-European Atherosclerosis Society Consensus Panel Statement on Assessment,
Aetiology and Management. Eur Heart J 36, 1012-22. DOI: 10.1093/eurheartj/ehv043.
16. Furberg CD & Pitt B (2001). Withdrawal of cerivastatin from the world market. Curr Control Trials Cardiovasc Med 2,
205-7. DOI: 10.1186/cvm-2-5-205.
17. Link E, Parish S, Armitage J, Bowman L, Heath S, Matsuda F, Gut I, Lathrop M & Collins R (2008). SLCO1B1 variants
and statin-induced myopathy--a genomewide study. N Engl J Med 359, 789-99. DOI: 10.1056/NEJMoa0801936.
18. Panche AN, Diwan AD & Chandra SR (2016). Flavonoids: an overview. J Nutr Sci 5, e47. DOI: 10.1017/jns.2016.41.
19. Fan X, Bai J, Hu M, Xu Y, Zhao S, Sun Y, Wang B, Hu J & Li Y (2020). Drug interaction study of flavonoids toward
OATP1B1 and their 3D structure activity relationship analysis for predicting hepatoprotective effects. Toxicology
152445. DOI: 10.1016/j.tox.2020.152445.
18
20. Cione E, La Torre C, Cannataro R, Caroleo MC, Plastina P & Gallelli L (2019). Quercetin, Epigallocatechin Gallate,
Curcumin, and Resveratrol: From Dietary Sources to Human MicroRNA Modulation. Molecules (Basel, Switzerland) 25,
63. DOI: 10.3390/molecules25010063.
21. Steel A, McIntyre E, Harnett J, Foley H, Adams J, Sibbritt D, Wardle J & Frawley J (2018). Complementary medicine
use in the Australian population: Results of a nationally-representative cross-sectional survey. Sci Rep 8, 17325. DOI:
10.1038/s41598-018-35508-y.
22. Ventola CL (2010). Current Issues Regarding Complementary and Alternative Medicine (CAM) in the United States:
Part 1: The Widespread Use of CAM and the Need for Better-Informed Health Care Professionals to Provide Patient
Counseling. P T 35, 461-8.
23. Ofori-Asenso R, Ilomaki J, Zomer E, Curtis AJ, Zoungas S & Liew D (2018). A 10-Year Trend in Statin Use Among
Older Adults in Australia: an Analysis Using National Pharmacy Claims Data. Cardiovasc Drugs Ther 32, 265-272.
DOI: 10.1007/s10557-018-6794-x.
24. Murphy KJ, Walker KM, Dyer KA & Bryan J (2019). Estimation of daily intake of flavonoids and major food sources in
middle-aged Australian men and women. Nutr Res 61, 64-81. DOI: 10.1016/j.nutres.2018.10.006.
25. Peluso I, Palmery M & Serafini M (2015). Association of flavonoid-rich foods and statins in the management of
hypercholesterolemia: a dangerous or helpful combination? Curr Drug Metab 16, 833-46. DOI:
10.2174/1389200216666151015113828.
26. Galati G & O'Brien PJ (2004). Potential toxicity of flavonoids and other dietary phenolics: significance for their
chemopreventive and anticancer properties. Free Radic Biol Med 37, 287-303. DOI:
10.1016/j.freeradbiomed.2004.04.034.
27. Kent K, Charlton KE, Russell J, Mitchell P & Flood VM (2015). Estimation of Flavonoid Intake in Older Australians:
Secondary Data Analysis of the Blue Mountains Eye Study. J Nutr Gerontol Geriatr 34, 388-98. DOI:
10.1080/21551197.2015.1088917.
28. Hollman PCH & Arts ICW (2000). Flavonols, flavones and flavanols nature, occurrence and dietary burden. J Sci Food
Agric 80, 1081-1093. DOI: https://doi.org/10.1002/(SICI)1097-0010(20000515)80:7<1081::AID-JSFA566>3.0.CO;2-G.
29. Miean KH & Mohamed S (2001). Flavonoid (myricetin, quercetin, kaempferol, luteolin, and apigenin) content of edible
tropical plants. J Agric Food Chem 49, 3106-12. DOI: 10.1021/jf000892m.
30. Křížová L, Dadáková K, Kašparovská J & Kašparovský T (2019). Isoflavones. Molecules 24, 1076. DOI:
10.3390/molecules24061076.
31. Gao Y, Yao Y, Zhu Y & Ren G (2015). Isoflavone content and composition in chickpea (Cicer arietinum L.) sprouts
germinated under different conditions. J Agric Food Chem 63, 2701-7. DOI: 10.1021/jf5057524.
19
32. Spagnuolo C, Russo GL, Orhan IE, Habtemariam S, Daglia M, Sureda A, Nabavi SF, Devi KP, Loizzo MR, Tundis R &
Nabavi SM (2015). Genistein and cancer: current status, challenges, and future directions. Adv Nutr 6, 408-19. DOI:
10.3945/an.114.008052.
33. Gattuso G, Barreca D, Gargiulli C, Leuzzi U & Caristi C (2007). Flavonoid composition of Citrus juices. Molecules 12,
1641-73. DOI: 10.3390/12081641.
34. Sultana B & Anwar F (2008). Flavonols (kaempeferol, quercetin, myricetin) contents of selected fruits, vegetables and
medicinal plants. Food Chem 108, 879-84. DOI: 10.1016/j.foodchem.2007.11.053.
35. Alam MA, Subhan N, Rahman MM, Uddin SJ, Reza HM & Sarker SD (2014). Effect of citrus flavonoids, naringin and
naringenin, on metabolic syndrome and their mechanisms of action. Adv Nutr 5, 404-417. DOI: 10.3945/an.113.005603.
36. Kantola T, Kivisto KT & Neuvonen PJ (1998). Effect of itraconazole on the pharmacokinetics of atorvastatin. Clin
Pharmacol Ther 64, 58-65. DOI: 10.1016/s0009-9236(98)90023-6.
37. Neuvonen PJ, Kantola T & Kivisto KT (1998). Simvastatin but not pravastatin is very susceptible to interaction with the
CYP3A4 inhibitor itraconazole. Clin Pharmacol Ther 63, 332-41. DOI: 10.1016/s0009-9236(98)90165-5.
38. Patel AM, Shariff S, Bailey DG, Juurlink DN, Gandhi S, Mamdani M, Gomes T, Fleet J, Hwang YJ & Garg AX (2013).
Statin toxicity from macrolide antibiotic coprescription: a population-based cohort study. Ann Intern Med 158, 869-76.
DOI: 10.7326/0003-4819-158-12-201306180-00004.
39. Neuvonen PJ & Jalava KM (1996). Itraconazole drastically increases plasma concentrations of lovastatin and lovastatin
acid. Clin Pharmacol Ther 60, 54-61. DOI: 10.1016/s0009-9236(96)90167-8.
40. Kivisto KT, Kantola T & Neuvonen PJ (1998). Different effects of itraconazole on the pharmacokinetics of fluvastatin
and lovastatin. Br J Clin Pharmacol 46, 49-53. DOI: 10.1046/j.1365-2125.1998.00034.x.
41. Abu Mellal A, Hussain N & Said AS (2019). The clinical significance of statins-macrolides interaction: comprehensive
review of in vivo studies, case reports, and population studies. Ther Clin Risk Manag 15, 921-36. DOI:
10.2147/tcrm.S214938.
42. Rowan CG, Brunelli SM, Munson J, Flory J, Reese PP, Hennessy S, Lewis J, Mines D, Barrett JS, Bilker W & Strom BL
(2012). Clinical importance of the drug interaction between statins and CYP3A4 inhibitors: a retrospective cohort study
in The Health Improvement Network. Pharmacoepidemiol Drug Saf 21, 494-506. DOI: 10.1002/pds.3199.
43. Tamraz B, Fukushima H, Wolfe AR, Kaspera R, Totah RA, Floyd JS, Ma B, Chu C, Marciante KD, Heckbert SR, Psaty
BM, Kroetz DL & Kwok P-Y (2013). OATP1B1-related drug-drug and drug-gene interactions as potential risk factors
for cerivastatin-induced rhabdomyolysis. Pharmacogenet Genomics 23, 355-64. DOI: 10.1097/FPC.0b013e3283620c3b.
44. Yeo KR & Yeo WW (2001). Inhibitory effects of verapamil and diltiazem on simvastatin metabolism in human liver
microsomes. Br J Clin Pharmacol 51, 461-70. DOI: 10.1046/j.1365-2125.2001.01386.x.
20
45. Zhao XJ, Koyama E & Ishizaki T (1999). An in vitro study on the metabolism and possible drug interactions of
rokitamycin, a macrolide antibiotic, using human liver microsomes. Drug Metab Dispos 27, 776-85.
46. Le Goff N, Koffel JC, Vandenschrieck S, Jung L & Ubeaud G (2002). Comparison of in vitro hepatic models for the
prediction of metabolic interaction between simvastatin and naringenin. Eur J Drug Metab Pharmacokinet 27, 233-41.
DOI: 10.1007/bf03192333.
47. Kopecna-Zapletalova M, Krasulova K, Anzenbacher P, Hodek P & Anzenbacherova E (2017). Interaction of
isoflavonoids with human liver microsomal cytochromes P450: inhibition of CYP enzyme activities. Xenobiotica 47,
324-31. DOI: 10.1080/00498254.2016.1195028.
48. Yang W, Zhang Q, Yang Y, Xu J, Fan A, Yang CS, Li N, Lu Y, Chen J, Zhao D, Aa J & Chen X (2017).
Epigallocatechin-3-gallate decreases the transport and metabolism of simvastatin in rats. Xenobiotica 47, 86-92. DOI:
10.3109/00498254.2016.1159747.
49. Elbarbry F, Ung A & Abdelkawy K (2018). Studying the Inhibitory Effect of Quercetin and Thymoquinone on Human
Cytochrome P450 Enzyme Activities. Pharmacogn Mag 13, 895-99. DOI: 10.4103/0973-1296.224342.
50. Satoh T, Fujisawa H, Nakamura A, Takahashi N & Watanabe K (2016). Inhibitory Effects of Eight Green Tea Catechins
on Cytochrome P450 1A2, 2C9, 2D6, and 3A4 Activities. J Pharm Pharm Sci 19, 188-97. DOI: 10.18433/j3ms5c.
51. Wu LX, Guo CX, Chen WQ, Yu J, Qu Q, Chen Y, Tan ZR, Wang G, Fan L, Li Q, Zhang W & Zhou HH (2012).
Inhibition of the organic anion-transporting polypeptide 1B1 by quercetin: an in vitro and in vivo assessment. Br J Clin
Pharmacol 73, 750-7. DOI: 10.1111/j.1365-2125.2011.04150.x.
52. Niemi M (2010). Transporter pharmacogenetics and statin toxicity. Clin Pharmacol Ther 87, 130-3. DOI:
10.1038/clpt.2009.197.
53. Nakagomi-Hagihara R, Nakai D, Tokui T, Abe T & Ikeda T (2007). Gemfibrozil and its glucuronide inhibit the hepatic
uptake of pravastatin mediated by OATP1B1. Xenobiotica 37, 474-86. DOI: 10.1080/00498250701278442.
54. Wen X, Wang JS, Backman JT, Kivistö KT & Neuvonen PJ (2001). Gemfibrozil is a potent inhibitor of human
cytochrome P450 2C9. Drug Metab Dispos 29, 1359-61.
55. Amend KL, Landon J, Thyagarajan V, Niemcryk S & McAfee A (2011). Incidence of hospitalized rhabdomyolysis with
statin and fibrate use in an insured US population. Ann Pharmacother 45, 1230-9. DOI: 10.1345/aph.1Q110.
56. Wang X, Wolkoff AW & Morris ME (2005). Flavonoids as a novel class of human organic anion-transporting
polypeptide OATP1B1 (OATP-C) modulators. Drug Metab Dispos 33, 1666-72. DOI: 10.1124/dmd.105.005926.
57. Mandery K, Balk B, Bujok K, Schmidt I, Fromm MF & Glaeser H (2012). Inhibition of hepatic uptake transporters by
flavonoids. Eur J Pharm Sci 46, 79-85. DOI: https://doi.org/10.1016/j.ejps.2012.02.014.
21
58. Kunze A, Huwyler J, Camenisch G & Poller B (2014). Prediction of organic anion-transporting polypeptide 1B1- and
1B3-mediated hepatic uptake of statins based on transporter protein expression and activity data. Drug Metab Dispos 42,
1514-21. DOI: 10.1124/dmd.114.058412.
59. Schachter M (2005). Chemical, pharmacokinetic and pharmacodynamic properties of statins: an update. Fundam Clin
Pharmacol 19, 117-25. DOI: 10.1111/j.1472-8206.2004.00299.x.
60. Turner RM & Pirmohamed M (2019). Statin-Related Myotoxicity: A Comprehensive Review of Pharmacokinetic,
Pharmacogenomic and Muscle Components. J Clin Med 9, DOI: 10.3390/jcm9010022.
61. Deichmann R, Lavie C & Andrews S (2010). Coenzyme q10 and statin-induced mitochondrial dysfunction. Ochsner J
10, 16-21.
62. Marcoff L & Thompson PD (2007). The role of coenzyme Q10 in statin-associated myopathy: a systematic review. J Am
Coll Cardiol 49, 2231-7. DOI: 10.1016/j.jacc.2007.02.049.
63. Quinzii CM & Hirano M (2011). Primary and secondary CoQ(10) deficiencies in humans. Biofactors 37, 361-5. DOI:
10.1002/biof.155.
64. Zhao Y, Wu T-Y, Zhao M-F & Li C-J (2020). The balance of protein farnesylation and geranylgeranylation during the
progression of nonalcoholic fatty liver disease. The Journal of biological chemistry 295, 5152-62. DOI:
10.1074/jbc.REV119.008897.
65. Itagaki M, Takaguri A, Kano S, Kaneta S, Ichihara K & Satoh K (2009). Possible mechanisms underlying statin-induced
skeletal muscle toxicity in L6 fibroblasts and in rats. J Pharmacol Sci 109, 94-101. DOI: 10.1254/jphs.08238fp.
66. Sakamoto K, Honda T, Yokoya S, Waguri S & Kimura J (2007). Rab-small GTPases are involved in fluvastatin and
pravastatin-induced vacuolation in rat skeletal myofibers. Faseb J 21, 4087-94. DOI: 10.1096/fj.07-8713com.
67. Takai Y, Sasaki T & Matozaki T (2001). Small GTP-binding proteins. Physiol Rev 81, 153-208. DOI:
10.1152/physrev.2001.81.1.153.
68. Ridley AJ & Hall A (1992). The small GTP-binding protein rho regulates the assembly of focal adhesions and actin
stress fibers in response to growth factors. Cell 70, 389-99. DOI: 10.1016/0092-8674(92)90163-7.
69. Sung JH, Lee SJ, Park KH & Moon TW (2004). Isoflavones inhibit 3-hydroxy-3-methylglutaryl coenzyme A reductase
in vitro. Biosci Biotechnol Biochem 68, 428-32. DOI: 10.1271/bbb.68.428.
70. Cuccioloni M, Bonfili L, Mozzicafreddo M, Cecarini V, Pettinari R, Condello F, Pettinari C, Marchetti F, Angeletti M &
Eleuteri AM (2016). A ruthenium derivative of quercetin with enhanced cholesterol-lowering activity. RSC Advances 6,
39636-41. DOI: 10.1039/C6RA06403E.
71. Cuccioloni M, Mozzicafreddo M, Spina M, Tran CN, Falconi M, Eleuteri AM & Angeletti M (2011). Epigallocatechin-
3-gallate potently inhibits the in vitro activity of hydroxy-3-methyl-glutaryl-CoA reductase. J Lipid Res 52, 897-907.
DOI: 10.1194/jlr.M011817.
22
72. Leopoldini M, Malaj N, Toscano M, Sindona G & Russo N (2010). On the inhibitor effects of bergamot juice flavonoids
binding to the 3-hydroxy-3-methylglutaryl-CoA reductase (HMGR) enzyme. J Agric Food Chem 58, 10768-73. DOI:
10.1021/jf102576j.
73. Karimi N, Ghadimi D & Fathi M (2020). The Inhibitory Effect of Biochanin A on Hepatic Cholesterol Biosynthesis in
High Glucose-Induced Steatosis in HepG2 Cells. RABMS 6, 167-85.
74. Xue CC, Zhang AL, Lin V, Da Costa C & Story DF (2007). Complementary and alternative medicine use in Australia: a
national population-based survey. J Altern Complement Med 13, 643-50. DOI: 10.1089/acm.2006.6355.
75. Bailey DG, Dresser G & Arnold JM (2013). Grapefruit-medication interactions: forbidden fruit or avoidable
consequences? Cmaj 185, 309-16. DOI: 10.1503/cmaj.120951.
76. Baltes MR, Dubois JG & Hanocq M (2001). Application to drug-food interactions of living cells as in vitro model
expressing cytochrome P450 activity: enzyme inhibition by lemon juice. Talanta 54, 983-7. DOI: 10.1016/s0039-
9140(01)00368-x.
77. Kawser Hossain M, Abdal Dayem A, Han J, Yin Y, Kim K, Kumar Saha S, Yang GM, Choi HY & Cho SG (2016).
Molecular Mechanisms of the Anti-Obesity and Anti-Diabetic Properties of Flavonoids. Int J Mol Sci 17, 569. DOI:
10.3390/ijms17040569.
78. Setchell KD & Cassidy A (1999). Dietary isoflavones: biological effects and relevance to human health. J Nutr 129, 758-
67. DOI: 10.1093/jn/129.3.758S.
79. de Camargo AC, Favero BT, Morzelle MC, Franchin M, Alvarez-Parrilla E, de la Rosa LA, Geraldi MV, Marostica
Junior MR, Shahidi F & Schwember AR (2019). Is Chickpea a Potential Substitute for Soybean? Phenolic Bioactives and
Potential Health Benefits. Int J Mol Sci 20, DOI: 10.3390/ijms20112644.
80. Fan L, Zhang W, Guo D, Tan ZR, Xu P, Li Q, Liu YZ, Zhang L, He TY, Hu DL, Wang D & Zhou HH (2008). The effect
of herbal medicine baicalin on pharmacokinetics of rosuvastatin, substrate of organic anion-transporting polypeptide
1B1. Clin Pharmacol Ther 83, 471-6. DOI: 10.1038/sj.clpt.6100318.
81. Moon YJ & Morris ME (2007). Pharmacokinetics and bioavailability of the bioflavonoid biochanin A: effects of
quercetin and EGCG on biochanin A disposition in rats. Mol Pharm 4, 865-72. DOI: 10.1021/mp7000928.
82. Izumi S, Nozaki Y, Maeda K, Komori T, Takenaka O, Kusuhara H & Sugiyama Y (2015). Investigation of the impact of
substrate selection on in vitro organic anion transporting polypeptide 1B1 inhibition profiles for the prediction of drug-
drug interactions. Drug Metab Dispos 43, 235-47. DOI: 10.1124/dmd.114.059105.
83. Ness J, Johnson D & Nisly N (2003). "Polyherbacy": herbal supplements as a form of polypharmacy in older adults. J
Gerontol A Biol Sci Med Sci 58, M478. DOI: 10.1093/gerona/58.5.m478.
84. Holtzman CW, Wiggins BS & Spinler SA (2006). Role of P-glycoprotein in statin drug interactions. Pharmacotherapy
26, 1601-7. DOI: 10.1592/phco.26.11.1601.
23
85. Mohana S, Ganesan M, Agilan B, Karthikeyan R, Srithar G, Beaulah Mary R, Ananthakrishnan D, Velmurugan D,
Rajendra Prasad N & Ambudkar SV (2016). Screening dietary flavonoids for the reversal of P-glycoprotein-mediated
multidrug resistance in cancer. Mol Biosyst 12, 2458-70. DOI: 10.1039/c6mb00187d.
86. Chow HH, Hakim IA, Vining DR, Crowell JA, Ranger-Moore J, Chew WM, Celaya CA, Rodney SR, Hara Y & Alberts
DS (2005). Effects of dosing condition on the oral bioavailability of green tea catechins after single-dose administration
of Polyphenon E in healthy individuals. Clin Cancer Res 11, 4627-33. DOI: 10.1158/1078-0432.Ccr-04-2549.
87. Jin F, Nieman DC, Shanely RA, Knab AM, Austin MD & Sha W (2010). The variable plasma quercetin response to 12-
week quercetin supplementation in humans. Eur J Clin Nutr 64, 692-7. DOI: 10.1038/ejcn.2010.91.
88. Elbarbry F, Ung A & Abdelkawy K (2018). Studying the Inhibitory Effect of Quercetin and Thymoquinone on Human
Cytochrome P450 Enzyme Activities. Pharmacogn Mag 13, 895-899. DOI: 10.4103/0973-1296.224342.
89. Aziz AA, Edwards CA, Lean ME & Crozier A (1998). Absorption and excretion of conjugated flavonols, including
quercetin-4'-O-beta-glucoside and isorhamnetin-4'-O-beta-glucoside by human volunteers after the consumption of
onions. Free Radic Res 29, 257-69. DOI: 10.1080/10715769800300291.
90. Murota K, Nakamura Y & Uehara M (2018). Flavonoid metabolism: the interaction of metabolites and gut microbiota.
Biosci Biotechnol Biochem 82, 600-10. DOI: 10.1080/09168451.2018.1444467.
91. Hubbard GP, Wolffram S, Lovegrove JA & Gibbins JM (2004). Ingestion of quercetin inhibits platelet aggregation and
essential components of the collagen-stimulated platelet activation pathway in humans. J Thromb Haemost 2, 2138-45.
DOI: 10.1111/j.1538-7836.2004.01067.x.
92. Patel RV, Mistry BM, Shinde SK, Syed R, Singh V & Shin HS (2018). Therapeutic potential of quercetin as a
cardiovascular agent. Eur J Med Chem 155, 889-904. DOI: 10.1016/j.ejmech.2018.06.053.
93. Babu PV & Liu D (2008). Green tea catechins and cardiovascular health: an update. Curr Med Chem 15, 1840-50. DOI:
10.2174/092986708785132979.
94. Yu X-H, Chen J-J, Deng W-Y, Xu X-D, Liu Q-X, Shi M-W & Ren K (2020). Biochanin A Mitigates Atherosclerosis by
Inhibiting Lipid Accumulation and Inflammatory Response. Oxid Med Cell Longev 2020, 15. DOI:
10.1155/2020/8965047.
... 43 There is evidence that traditional Chinese medicine flavonoids can raise plasma levels of statins and increase the risk of statin induced myopathy by interacting with the liver CYP450 enzyme, OATP uptake, or HMG-CoA reductase. 44 A large body of evidence shows that the benefits of statin therapy far outweigh any actual or perceived risks. 45 Whether the adverse effects of statins are caused by the effects of statins or nocebo is still debated. ...
Article
Full-text available
Surveillance of drug safety is an important aspect in the routine medical care. Adverse events caused by real-world drug utilization has become one of the leading causes of death and an urgent issue in the field of toxicology. Cardiovascular disease is now the leading cause of fatal diseases in most countries, especially in the elderly population who often suffer from multiple diseases and need long-term multidrug therapy. Among which, statins have been widely used to lower bad cholesterol and regress coronary plaque mainly in patients with hyperlipidemia and atherosclerotic cardiovascular diseases (ASCVD). Although the real-world benefits of statins are significant, different degrees and types of adverse drug reactions (ADR) such as liver dysfunction and muscle injury, have a great impact on the original treatment regimens as well as the quality of life. This review describes the epidemiology, mechanisms, early identification and post-intervention of statin-associated liver dysfunction and muscle injury based on the updated clinical evidence. It provides systematic and comprehensive guidance and necessary supplement for the clinical safety of statin use in cardiovascular diseases.
... Multidrug and toxin extrusion (MATE) 1 and MATE2-K proteins, expressed on the brush border membrane, play a role in the active renal secretion of drugs in the kidney [12]. These nine transporters (P-gp, BCRP, OATP1B1, OATP1B3, OAT1, OAT3, OCT2, MATE1, and MATE2-K) are primarily associated with drug interactions, and the co-administration of inhibitor drugs for transporters with substrate drugs may induce serious adverse events or treatment failure [13,14]. ...
Article
Full-text available
Understanding possible follow-up actions on in vitro findings helps determine the necessity of labeling for drug interactions. We analyzed information for in vitro findings on transporter-mediated interactions of drugs approved by the U.S. Food and Drug Administration's Center for Drug Evaluation and Research for the last five years (i.e., 2017-2021) and their follow-up actions for labeling. Higher R values than the pre-defined cut-off were observed with 3.7-39.1% inhibitor drugs in a simple prediction. Among these drugs, 16-41.7% were labeled with their potential drug interactions, while results of supporting studies or scientific rationales were submitted for the other drugs leading to no interaction labeling. In vitro transporter substrates were reported with 1.7-67.6% of drugs. The interaction labels for these substrate drugs were observed in up to 40% of drugs, while the other drugs were not labeled on the drug interactions with claims for their low interaction potential, evidenced by clinical studies or scientific rationales. The systematic and comprehensive analysis in this study will provide insight into the management of in vitro findings for transporter substrate or inhibitor drugs.
Article
Full-text available
Cardiovascular disease (CVD) is a disease that causes death in the world. Behavioral risk factors are crucial in dealing with CVD disease. High levels of cholesterol in the blood are associated with hypercholesterolemia. It can increase the risk of CVD disease. The HMG-CoA reductase enzyme is an enzyme that plays a role in cholesterol synthesis. It will convert the HMG-CoA compound into the mevalonate. Inhibition of this enzyme can reduce cholesterol synthesis because it inhibits the formation of mevalonate, which is the initial stage of cholesterol synthesis. Herbal compounds are compounds produced by plants. It has good benefits for the body. Flavonoid compounds can be beneficial for health. Molecular docking is a method used to determine the binding affinity and interaction values of the enzyme protein interaction with the ligand to be bound. The results of Apigenin binding affinity values is -7.7 kcal/mol, Luteolin is -8 kcal/mol, Quercetin is -8.2 kcal/mol, Kaempferol is -7.4 kcal/mol, Phloretin is -6.8 kcal/mol, Chalconaringenin is -6.9 kcal/mol, Cyanidin is -7.9 kcal/mol, Delphinidin is -7.8 kcal/mol, Hesperetin is -7.7 kcal/mol, Narigenin is -7.8 kcal/mol, Daidzein is -6.8 kcal/mol, Genistein is -7 kcal/mol, Rutin is -9.2 kcal/mol, Taxifolin is -8 kcal /mol, Diosmetin is -7.6 kcal/mol, and its native ligand Rosuvastatin is -8.5 kcal/mol. Flavonoid derivative compounds can bind to the HMG-CoA reductase enzyme. They can be candidates for antihyperlipidemia drugs.
Article
Machine learning (ML) has shown a great promise in predicting toxicity of small molecules. However, the availability of data for such predictions is often limited. Because of the unsatisfactory performance of models trained on a single toxicity endpoint, we collected toxic small molecules with multiple toxicity endpoints from previous study. The dataset comprises 27 toxic endpoints categorized into seven toxicity classes, namely, carcinogenicity and mutagenicity, acute oral toxicity, respiratory toxicity, irritation and corrosion, cardiotoxicity, CYP450, and endocrine disruption. In addition, a binary classification Common‐Toxicity task was added based on the aforementioned dataset. To improve the performance of the models, we added marketed drugs as negative samples. This study presents a toxicity predictive model, ToxMPNN, based on the message passing neural network (MPNN) architecture, aiming to predict the toxicity of small molecules. The results demonstrate that ToxMPNN outperforms other models in capturing toxic features within the molecular structure, resulting in more precise predictions with the ROC_AUC testing score of 0.886 for the Toxicity_drug dataset. Furthermore, it was observed that adding marketed drugs as negative samples not only improves the predictive performance of the binary classification Common‐Toxicity task but also enhances the stability of the model prediction. It shows that the graph‐based deep learning (DL) algorithms in this study can be used as a trustworthy and effective tool to assess small molecule toxicity in the development of new drugs.
Article
Atorvastatin, an effective lipid-lowering drug, could reduce the risks of morbidity and mortality of cardiovascular diseases. Patients with cardiovascular diseases often use atorvastatin along with berberine. Atorvastatin is the substrate of CYP3A4 and P-gp. However, berberine is the inhibitor. The combination might lead to DDIs. The aim of this study was to assess the effect of berberine on pharmacokinetics and pharmacodynamics of atorvastatin in rats.Plasma concentrations of atorvastatin with or without berberine were determined by HPLC. Pharmacokinetics parameters were calculated and used to evaluate pharmacokinetics interactions. The effect of berberine on pharmacodynamics of atorvastatin was investigated by detecting blood lipid, SOD, MDA, GSH-Px, AST, ALT, and liver histopathology.Cmax, tmax, and AUC0-t of atorvastatin in combination group significantly increased both in normal and model rats (p < 0.01). The increase of t1/2, AUC0-t in model rats was more significant than that in normal rats (p < 0.05). Pharmacodynamics indexes in treatment groups were significantly improved, especially combination group (p < 0.05). Moreover, it could be found that there is a significant recovery in liver histopathology.In conclusion, berberine could affect pharmacokinetics of atorvastatin, enhance lipid-lowering effect and improve liver injury in rats. More attention should be paid to plasma exposure in clinical to avoid adverse reactions.
Article
In humans, approximately 70% of drugs are eliminated through the liver. This process is governed by the concerted action of membrane transporters and metabolic enzymes. Transporters mediating hepatocellular uptake of drugs belong to the SLC (Solute carrier) superfamily of transporters. Drug efflux either toward the portal vein or into the bile is mainly mediated by active transporters of the ABC (ATP Binding Cassette) family. Alteration in the function and/or expression of liver transporters due to mutations, disease conditions, or co-administration of drugs or food components can result in altered pharmacokinetics. On the other hand, drugs or food components interacting with liver transporters may also interfere with liver function (e.g., bile acid homeostasis) and may even cause liver toxicity. Accordingly, certain transporters of the liver should be investigated already at an early stage of drug development. Most frequently radioactive probes are applied in these drug-transporter interaction tests. However, fluorescent probes are cost-effective and sensitive alternatives to radioligands, and are gaining wider application in drug-transporter interaction tests. In our review, we summarize our current understanding about hepatocyte ABC and SLC transporters affected by drug interactions. We provide an update of the available fluorescent and fluorogenic/activable probes applicable in in vitro or in vivo testing of these ABC and SLC transporters, including near-infrared transporter probes especially suitable for in vivo imaging. Furthermore, our review gives a comprehensive overview of the available fluorescence-based methods, not directly relying on the transport of the probe, suitable for the investigation of hepatic ABC or SLC-type drug transporters.
Article
Full-text available
Luteolin and naringenin are flavonoids found in various foods/beverages and present in certain dietary supplements. After a high intake of these flavonoids, their sulfate and glucuronide conjugates reach micromolar concentrations in the bloodstream. Some pharmacokinetic interactions of luteolin and naringenin have been investigated in previous studies; however, only limited data are available in regard to their metabolites. In this study, we aimed to investigate the interactions of the sulfate and glucuronic acid conjugates of luteolin and naringenin with human serum albumin, cytochrome P450 (CYP2C9, 2C19, and 3A4) enzymes, and organic anion transporting polypeptide (OATP1B1 and OATP2B1) transporters. Our main findings are as follows: (1) Sulfate conjugates formed more stable complexes with albumin than the parent flavonoids. (2) Luteolin and naringenin conjugates showed no or only weak inhibitory action on the CYP enzymes examined. (3) Certain conjugates of luteolin and naringenin are potent inhibitors of OATP1B1 and/or OATP2B1 enzymes. (4) Conjugated metabolites of luteolin and naringenin may play an important role in the pharmacokinetic interactions of these flavonoids.
Article
Full-text available
Biochanin A (BCA), a dietary isoflavone extracted from red clover and cabbage, has been shown to antagonize hypertension and myocardial ischemia/reperfusion injury. However, very little is known about its role in atherogenesis. The aim of this study was to observe the effects of BCA on atherosclerosis and explore the underlying mechanisms. Our results showed that administration of BCA promoted reverse cholesterol transport (RCT), improved plasma lipid profile, and decreased serum proinflammatory cytokine levels and atherosclerotic lesion area in apoE-/- mice fed a Western diet. In THP-1 macrophage-derived foam cells, treatment with BCA upregulated ATP-binding cassette (ABC) transporter A1 (ABCA1) and ABCG1 expression and facilitated subsequent cholesterol efflux and diminished intracellular cholesterol contents by activating the peroxisome proliferator-activated receptor γ (PPARγ)/liver X receptor α (LXRα) and PPARγ/heme oxygenase 1 (HO-1) pathways. BCA also activated these two signaling pathways to inhibit the secretion of proinflammatory cytokines. Taken together, these findings suggest that BCA is protective against atherosclerosis by inhibiting lipid accumulation and inflammatory response through the PPARγ/LXRα and PPARγ/HO-1 pathways. BCA may be an attractive drug for the prevention and treatment of atherosclerotic cardiovascular disease.
Article
Full-text available
Protein prenylation is an essential posttranslational modification and includes protein farnesylation and geranylgeranylation using farnesyl diphosphate (FPP) or geranylgeranyl diphosphate (GGPP) as substrates, respectively. Geranylgeranyl diphosphate synthase (GGPPS) is a branchpoint enzyme in the mevalonate (MVA) pathway that affects the ratio of FPP to GGPP. Abnormal GGPPS expression and activity can therefore disrupt the balance of farnesylation and geranylgeranylation and alter the ratio between farnesylated and geranylgeranylated proteins. This change is associated with the progression of nonalcoholic fatty liver disease (NAFLD), a condition characterized by hepatic fat overload. Of note, differential accumulation of farnesylated and geranylgeranylated proteins has been associated with differential stages of NAFLD and NAFLD-associated liver fibrosis. In this review, we summarize key aspects of protein prenylation as well as advances that have uncovered the regulation of associated metabolic patterns and signaling pathways, such as Ras GTPase signaling, involved in NAFLD progression. Additionally, we discuss unique opportunities for targeting prenylation in NAFLD/hepatocellular carcinoma (HCC) with agents such as statins and bisphosphonates (BPs) to improve clinical outcomes.
Article
Full-text available
Epidemiologic studies suggest that dietary polyphenol intake is associated with a lower incidence of several non-communicable diseases. Although several foods contain complex mixtures of polyphenols, numerous factors can affect their content. Besides the well-known capability of these molecules to act as antioxidants, they are able to interact with cell-signaling pathways, modulating gene expression, influencing the activity of transcription factors, and modulating microRNAs. Here we deeply describe four polyphenols used as nutritional supplements: quercetin, resveratrol, epigallocatechin gallate (ECGC), and curcumin, summarizing the current knowledge about them, spanning from dietary sources to the epigenetic capabilities of these compounds on microRNA modulation.
Article
Full-text available
Statins are a cornerstone in the pharmacological prevention of cardiovascular disease. Although generally well tolerated, a small subset of patients experience statin-related myotoxicity (SRM). SRM is heterogeneous in presentation; phenotypes include the relatively more common myalgias, infrequent myopathies, and rare rhabdomyolysis. Very rarely, statins induce an anti-HMGCR positive immune-mediated necrotizing myopathy. Diagnosing SRM in clinical practice can be challenging, particularly for mild SRM that is frequently due to alternative aetiologies and the nocebo effect. Nevertheless, SRM can directly harm patients and lead to statin discontinuation/non-adherence, which increases the risk of cardiovascular events. Several factors increase systemic statin exposure and predispose to SRM, including advanced age, concomitant medications, and the nonsynonymous variant, rs4149056, in SLCO1B1, which encodes the hepatic sinusoidal transporter, OATP1B1. Increased exposure of skeletal muscle to statins increases the risk of mitochondrial dysfunction, calcium signalling disruption, reduced prenylation, atrogin-1 mediated atrophy and pro-apoptotic signalling. Rare variants in several metabolic myopathy genes including CACNA1S, CPT2, LPIN1, PYGM and RYR1 increase myopathy/rhabdomyolysis risk following statin exposure. The immune system is implicated in both conventional statin intolerance/myotoxicity via LILRB5 rs12975366, and a strong association exists between HLA-DRB1*11:01 and anti-HMGCR positive myopathy. Epigenetic factors (miR-499-5p, miR-145) have also been implicated in statin myotoxicity. SRM remains a challenge to the safe and effective use of statins, although consensus strategies to manage SRM have been proposed. Further research is required, including stringent phenotyping of mild SRM through N-of-1 trials coupled to systems pharmacology omics- approaches to identify novel risk factors and provide mechanistic insight.
Article
Full-text available
The objectives of this article were to review the mechanism and clinical significance of statins-macrolides interaction, determine which combination has the highest risk for the interaction, and identify key patients' risk factors for the interaction in relation to the development of muscle toxicity. A literature review was conducted in PubMed and Embase (1946 to December 2018) using combined terms: statins - as group and individual agents, macrolides - as group and individual agents, drug interaction, muscle toxicity, rhabdomyolysis, CYP3A4 inhibitors, and OAT1B inhibitors, with forward and backward citation tracking. Relevant English language in vivo studies in healthy volunteers, case reports, and population studies were included. The interaction between statins and macrolides depends on the type of statin and macrolide used. The mechanism of the interaction is due to macrolides' inhibition of CYP3A4 isoenzyme and OAT1B transporter causing increased exposure to statins. The correlation of this increased statin's exposure to the development of muscle toxicity could not be established, unless the patient had other risk factors such as advanced age, cardiovascular diseases, renal impairment, diabetes, and the concomitant use of other CYP3A4 inhibitors. Simvastatin, lovastatin, and to lesser extent atorvastatin are the statins most affected by this interaction. Rosuvastatin, fluvastatin, and pravastatin are not significantly affected by this interaction. Telithromycin, clarithromycin, and erythromycin are the most "offending" macrolides, while azithromycin appears to be safe to use with statins. This review presented a clear description of the clinical significance of this interaction in real practice. Also, it provided health care professionals with clear suggestions and recommendations on how to overcome this interaction. In conclusion, understanding the different characteristics of each statin and macrolide, as well as key patients' risk factors, will enable health care providers to utilize both groups effectively without compromising patient safety.
Article
Full-text available
Legume seeds are rich sources of protein, fiber, and minerals. In addition, their phenolic compounds as secondary metabolites render health benefits beyond basic nutrition. Lowering apolipoprotein B secretion from HepG2 cells and decreasing the level of low-density lipoprotein (LDL)-cholesterol oxidation are mechanisms related to the prevention of cardiovascular diseases (CVD). Likewise, low-level chronic inflammation and related disorders of the immune system are clinical predictors of cardiovascular pathology. Furthermore, DNA-damage signaling and repair are crucial pathways to the etiology of human cancers. Along CVD and cancer, the prevalence of obesity and diabetes is constantly increasing. Screening the ability of polyphenols in inactivating digestive enzymes is a good option in pre-clinical studies. In addition, in vivo studies support the role of polyphenols in the prevention and/or management of diabetes and obesity. Soybean, a well-recognized source of phenolic isoflavones, exerts health benefits by decreasing oxidative stress and inflammation related to the above-mentioned chronic ailments. Similar to soybeans, chickpeas are good sources of nutrients and phenolic compounds, especially isoflavones. This review summarizes the potential of chickpea as a substitute for soybean in terms of health beneficial outcomes. Therefore, this contribution may guide the industry in manufacturing functional foods and/or ingredients by using an undervalued feedstock.
Article
Full-text available
Phytoestrogens are naturally occurring nonsteroidal phenolic plant compounds that, due to their molecular structure and size, resemble vertebrate steroids estrogens. This review is focused on plant flavonoids isoflavones, which are ranked among the most estrogenic compounds. The main dietary sources of isoflavones for humans are soybean and soybean products, which contain mainly daidzein and genistein. When they are consumed, they exert estrogenic and/or antiestrogenic effects. Isoflavones are considered chemoprotective and can be used as an alternative therapy for a wide range of hormonal disorders, including several cancer types, namely breast cancer and prostate cancer, cardiovascular diseases, osteoporosis, or menopausal symptoms. On the other hand, isoflavones may also be considered endocrine disruptors with possible negative influences on the state of health in a certain part of the population or on the environment. This review deals with isoflavone classification, structure, and occurrence, with their metabolism, biological, and health effects in humans and animals, and with their utilization and potential risks.
Article
Full-text available
Abstract In order to describe the prevalence and characteristics of complementary medicine (CM) practice and product use by Australians, we conducted a cross-sectional online survey with Australian adults aged 18 and over. Rates of consultation with CM practitioners, and use of CM products and practices were assessed. The sample (n = 2,019) was broadly representative of the Australian population. Prevalence of any CM use was 63.1%, with 36% consulting a CM practitioner and 52.8% using any CM product or practice. Bodywork therapists were the most commonly consulted CM practitioners (massage therapists 20.7%, chiropractors 12.6%, yoga teachers 8.9%) and homeopaths were the least commonly consulted (3.4%). Almost half of respondents (47.8%) used vitamin/mineral supplements, while relaxation techniques/meditation were the most common practice (15.8%). CM users were more likely to be female, have a chronic disease diagnosis, no private health insurance, a higher education level, and not be looking for work. Prevalence of CM use in Australia has remained consistently high, demonstrating that CM is an established part of contemporary health management practices within the general population. It is critical that health policy makers and health care providers acknowledge CM in their attempts to ensure optimal public health and patient outcomes.
Article
Organic anion transporting polypeptide 1B1 (OATP1B1), a liver-specific uptake transporter, was associated with drug induced liver injury (DILI). Screening and identifying potent OATP1B1 inhibitors with little toxicity is of great value in reducing OATP1B1-mediated DILI. Flavonoids are a group of polyphenols ubiquitously present in vegetables, fruits and herbal products, some of them were reported to produce transporter-mediated DDI. Our objective was to investigate potential inhibitors of OATP1B1 from 99 flavonoids, and to assess the hepatoprotective effects on bosentan induced liver injury. Eight flavonoids, including biochanin A, hispidulin, isoliquiritigenin, isosinensetin, kaempferol, licochalcone A, luteolin and sinensetin exhibited significant inhibition (>50%) on OATP1B1 in OATP1B1-HEK293 cells, which reduced the OATP1B1-mediated influx of methotrexate, accordingly decreased its cytotoxicity in OATP1B1-HEK293 cells and increased its AUC0−t in different extents in rats, from 28.27% to 82.71%. In bosentan-induced rat liver injury models, 8 flavonoids reduced the levels of serum total bile acid (TBA) and the liver concentration of bosentan in different degrees. Among them, kaempferol decreased the concentration most significantly, by 54.17%, which indicated that flavonoids may alleviate bosentan-induced liver injury by inhibiting OATP1B1-mediated bosentan uptake. Furthermore, the pharmacophore model indicated the hydrogen bond acceptors and hydrogen bond donors may play critical role in the potency of flavonoids inhibition on OATP1B1. Taken together, our findings would provide helpful information for predicting the potential risks of flavonoid-containing food/herb-drug interactions in humans and alleviating bosentan -induced liver injury by OATP1B1 regulation.
Article
Flavonoid consumption has reported health benefits such as reducing cardiovascular disease risk factors, improving endothelial function, and delaying age-related cognitive decline. However, there are little dietary intake data for Australians, which limit our ability to make dietary recommendations to increase intakes to a level where health benefits are seen. The aim of this cross-sectional study was to determine the intake of flavonoids, flavonoid classes, and flavonoid subclasses of 1183 Australians aged 39 to 65 years using a validated 215-item food frequency questionnaire. Based on limited global flavonoid intake data, flavanols are the major dietary flavonoid and are found predominantly in tea and cocoa. As Australians are large tea drinkers, we anticipated that flavanols would be the major flavonoid in the Australian diet. The flavonoid content of foods was determined using a combination of the United States Department of Agriculture Databases and the Phenol-Explorer Database. One-way analysis of variance was undertaken to examine differences between flavonoid intake between men and women. Total flavonoid intake was 626 ± 579 mg/d. Men and women consumed 566 ± 559 mg and 660 ± 588 mg of total flavonoids per day, respectively. Thearubigin accounted for 58% of the flavonoid intake. Women consumed more total flavonoids, thearubigins (both P <.01), anthocyanidins (P <.0001), flavan-3-ols, flavones, and flavonols (all P <.05) than men, whereas men consumed more flavanones than women (P =.01). There was no difference between sexes for the consumption of isoflavones. The data indicated that flavan-3-ols, predominantly thearubigin from tea, were the main flavonoid consumed by Australians. This information contributes to population flavonoid intakes, which should be considered when exploring flavonoid and health relationships.