Next Article in Journal
Family Meals and Social Eating Behavior and Their Association with Disordered Eating among Spanish Adolescents: The EHDLA Study
Previous Article in Journal
The Association between Grazing and Food Addiction: The Italian Version of the Repetitive Eating Questionnaire (Rep(Eat)-Q) and Its Relationships with Food Addiction Criteria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Drug–Nutrient Interactions of 45 Vitamins, Minerals, Trace Elements, and Associated Dietary Compounds with Acetylsalicylic Acid and Warfarin—A Review of the Literature

by
David Renaud
1,2,3,*,
Alexander Höller
4 and
Miriam Michel
5
1
DIU MAPS, Fundamental and Biomedical Sciences, Paris-Cité University, 75006 Paris, France
2
DIU MAPS, Health Sciences Faculty, Universidad Europea Miguel de Cervantes, 47012 Valladolid, Spain
3
Fundacja Recover, 05-124 Skrzeszew, Poland
4
Department of Nutrition and Dietetics, University Hospital Innsbruck, 6020 Innsbruck, Austria
5
Department of Child and Adolescent Health, Division of Pediatrics III—Cardiology, Pulmonology, Allergology and Cystic Fibrosis, Medical University of Innsbruck, 6020 Innsbruck, Austria
*
Author to whom correspondence should be addressed.
Nutrients 2024, 16(7), 950; https://doi.org/10.3390/nu16070950
Submission received: 5 March 2024 / Revised: 19 March 2024 / Accepted: 22 March 2024 / Published: 26 March 2024
(This article belongs to the Section Micronutrients and Human Health)

Abstract

:
In cardiology, acetylsalicylic acid (ASA) and warfarin are among the most commonly used prophylactic therapies against thromboembolic events. Drug–drug interactions are generally well-known. Less known are the drug–nutrient interactions (DNIs), impeding drug absorption and altering micronutritional status. ASA and warfarin might influence the micronutritional status of patients through different mechanisms such as binding or modification of binding properties of ligands, absorption, transport, cellular use or concentration, or excretion. Our article reviews the drug–nutrient interactions that alter micronutritional status. Some of these mechanisms could be investigated with the aim to potentiate the drug effects. DNIs are seen occasionally in ASA and warfarin and could be managed through simple strategies such as risk stratification of DNIs on an individual patient basis; micronutritional status assessment as part of the medical history; extensive use of the drug–interaction probability scale to reference little-known interactions, and application of a personal, predictive, and preventive medical model using omics.

1. Introduction

1.1. Drug–Nutrient Interactions (DNIs)

Classically, pharmacological interactions (drug–drug) are considered, as well as interactions with the diet (food–drug) [1,2,3,4] or with phytotherapeutic agents (herb–drug) [5,6,7,8]. Little attention is given to DNI, which typically is 100-fold less researched than drug–drug interaction, but still clinically relevant [9,10,11,12,13]. Several classifications of DNIs are in use [12]. Medication can impact the patient’s micronutritional status—levels of essential micronutrients for proper physiological functioning—either directly or indirectly [9]. These mechanisms may be physicochemical, physiological, or pathophysiological [11,14]. There are four types of DNIs [15]: (1) bio-inactivation; (2) altered absorption type; (3) altered effect; (4) altered excretion [9,11,16,17].
Medication can directly compete with micronutrients when they use the same metabolic and transport pathways, modifying the pharmacokinetics or micronutrient status [18,19,20] (absorption, distribution, metabolism, excretion) [21]. Physiochemical interactions are mostly represented by a chelation, causing loss of a nutrient and lower drug activity. Medication can directly induce a physiological change affecting the micronutrient [10,22]. Medication can indirectly affect patient health and micronutritional status [23,24,25]. Physiological mechanisms include drug-induced changes in appetite, digestion, or renal transformation, but also drug-induced micronutritional status [23]. Pathophysiological mechanisms occur when the drug impairs nutrient uptake or usage. A typical case is when the drug alters absorption of a nutrient or when drug toxicity inhibits a metabolic process [26]. These interactions may vary in different tissues, with tissue-specific interactions [11,27]. Many DNIs are mutual—for instance, the drug affects micronutritional status, and the micronutrient affects drug metabolism, contributing to adverse drug effects (Figure 1) [28,29,30].
DNIs are not always negative [31,32]. As Thurnham defined, a well-balanced diet should contain ‘the adequate micronutrients to sustain body functions, but also a wide range of non-nutrients to optimize phase I (oxidation) and phase II (conjugation) metabolic processes’ [28]. These interactions can also be harvested to potentiate or reduce side effects of a medication [33]. Some drug regimens may improve micronutritional status [34,35]. DNIs can cause clinically visible (short-term) life-threatening health disorders [36] but also cause undetected long-term aging-associated health issues [37,38]. The mechanism behind age-associated chronic disease is hypothesized by the Triage theory of Bruce Ames [39]: “When micronutrient availability is limited, functions required for short-term survival and reproduction take precedence over functions whose loss can be tolerated”. This hypothesis was applied to vitamin K [40,41], with proteins required for short-term survival like blood clotting served first, and selenium, with selenoproteins [42].

1.2. Quality of Evidence–Clinical Speculation or Theoretical Major Burden?

A relatively small list of DNIs is documented [12]. Potential interactions have often yet to be evaluated for their incidence and clinical significance. Evaluation of drug safety is mostly about drug–drug interactions [43]. DNIs are not addressed during the drug development phase [44]. As stated in the handbook of drug–nutrient interactions [45], most data come either traced from pharmacological studies or reports from clinicians. As for pharmacological studies, DNIs are sourced from pharmacokinetic data from studies with mostly small numbers of subjects, focusing only on the clinical relevance of the interaction, with some focusing on the mechanism behind the interaction. As for clinical reports, most data come from case series and anecdotal reports. Very few studies were conducted with control groups. Some studies lack clarity on the baseline status of micronutrients, which is important for describing DNIs accurately. Many data come from animal models and cannot be extrapolated into clinical recommendations. The overall recommendation in the field of DNIs is to consider the risk of interaction on an individual patient basis. Large-scale recommendations cannot be made without larger studies. It is expected to improve over time in a similar way drug–drug interactions are now widely recognized. Historically, drug–drug interactions appeared in the early 1960s [46]. From 1970, the Swedish drug regulatory agency required pharmaceutical companies to issue annual reviews of drug interactions [46]. Knowledge on DNIs in the education of medical doctors and pharmacists is still scarce. When DNIs are included in the education of pharmacists, the recognition of their importance is growing, and students have reported encountering DNIs on a weekly basis [47].

1.3. Acetylsalicylic Acid (ASA)—An Overview

The antiplatelet drug ASA is the most used pharmacon worldwide [48]. The mechanism of ASA involves the inhibition of prostaglandin synthesis [49,50] through acetylation of the platelet cyclooxygenase–isozymes (COX) at functionally important amino acids (COX-1, isoform of the platelet enzyme, at serine530 [51]; COX-2, isoform at serine516) [52,53]. This prevents arachidonic acid [54] from accessing the catalytic site of the enzyme at tyrosine385 [55] and results in irreversible inhibition of cyclic prostanoids formation (platelet-dependent thromboxane A2 (TXA2), prostaglandins, and prostacyclin [53]). ASA preferably inhibits COX-1, with a 150–200 times higher affinity for COX-1 than for COX-2 [49,53]. It is debated if ASA might have antithrombotic properties [56] through modulation of clotting factors such as thrombin, fibrinogen, factor XIII, and tissue plasminogen [57]. Other mechanisms are discussed in the literature, such as changes in endothelial iron status [58]. ASA efficiency may be impacted by polymorphisms of MDRP1 and methylation status [59]. Low-dose ASA is used for both primary and secondary prevention of cardiovascular disease [60], with varying success [61]. Side effects like upper gastrointestinal injury are described in the literature [62].

1.4. Warfarin—An Overview

The anticoagulant drug warfarin is a widely used drug for prophylaxis [63]. Its effect relies on blocking the function of the vitamin K epoxide reductase complex (VKOR) in the liver, then inhibiting the K-dependent coagulation factors II, VII, IX, and X [64]. Warfarin also inhibits endogenous production of protein C and protein S [65]. The main indication of warfarin is to prevent deep-vein thrombosis, thromboembolic events, and reduce the risk of stroke in patients with atrial fibrillation, valvular disease, or an artificial heart valve [66]. The drug is a mixture of R and S-enantiomers, with the latter being the most potent. Warfarin is metabolized in the liver by the cytochrome P450 (CYP) pathway [67,68,69,70]. Among the six enzymes of the CYP family [71], four are involved in metabolizing warfarin. The S-enantiomer is mostly metabolized through the CYP2C9 enzyme to 7-hydroxywarfarin [68] and partially by CYP3A4 [69,72]. The R-enantiomer is metabolized by CYP1A2 to 6- and 8-hydroxywarfarin [68] and CYP3A4 to 10-hydroxywarfarin [68]. CYP2C8, CYP2C18, and CYP2C19 are also involved [72,73,74,75]. Most cytochrome P450 enzymes are known to be polymorphic [76], with more than 2000 variants described [77]. CYP2C9 is one of the most present CYPs in the liver, and its polymorphisms are of clinical relevance [78,79,80]. In contrast, the importance of CYP2C19 genotype variations is debated in pharmacokinetics of warfarin enantiomers [81]. VKOR and CYP polymorphisms may not be the only explanation for the dose variability of warfarin. Gut microbiota status may modulate warfarin efficiency, as it produces endogenous vitamin K [82,83]. Other genetic polymorphisms, such as CYP4F2 [84] or ABCB1 [85], are also involved. One haplotype of ABCB1 was overrepresented among patients who had a stable INR with a low dose warfarin.. Warfarin requires periodic functional monitoring, with International Normalized Ratio (INR) monitoring having become the standard [86]. INR response is known to be influenced by diet, liver function, comorbidities, drug interactions [87], as well as unpredictable patient-specific responses, partially due to polymorphisms of CYP [88,89,90,91].

2. Methods

Our literature review covers DNIs between 45 nutrients and ASA and warfarin, published in English, French, Russian, German, and Danish from 1936 to 2023 on Pubmed. As a first step, we searched using the keywords ‘acetylsalicylic acid, ‘salicylate’, and ‘salicylate therapy’, ‘aspirin’ OR ‘warfarin’, AND [micronutrient] AND ‘interaction’. As a second step, we searched using the keywords ‘absorption’, ‘transport’, ‘metabolism’, ‘excretion’ for ‘acetylsalicylic acid’ OR ‘aspirin’ OR ‘warfarin’ AND [micronutrient].

3. Defining the “Hidden Hunger” Essentiality of Micronutrients

Micronutrient deficiency is sometimes called the ‘hidden hunger’ [92], underlining the importance of micronutrients for health. The term ‘micronutrients‘ is generally used to define essential vitamins, minerals, and trace elements required to sustain basic physiologic functions [93]. Micronutrients are necessary as cofactors for vital enzymatic reactions [94]. Severe deficiencies might cause or aggravate clinical symptoms [36,95].
Vitamins are classified as water-soluble or fat-soluble. Water-soluble vitamins are thiamine (B1) [96,97,98,99], riboflavin (B2) [96,100,101], niacin (B3) [96], pantothenic acid (B5) [96], pyridoxine (B6) [102], biotin (B7) [103], folate (B9) [104,105], cobalamins (B12) [106], and ascorbic acid (C) [107,108,109]. Fat-soluble vitamins are retinol (A), α-, β-, γ- and δ-tocopherols/α-, β-, γ-, and δ-tocotrienols (E) [110,111,112], calciferols (D), and phylloquinone/menaquinone (K). Minerals [113] are calcium [114], phosphorous [115], magnesium [116], sodium [117], potassium [118], and chloride [119]. Trace elements [113] are iron [120,121], copper [122,123,124,125], zinc [126,127], selenium [42,128,129], and iodine [130]. This list is nonexhaustive and the essentiality of some nutrients is still debated [113]. The American Institute of Medicine published recommendations for dietary reference intake for thiamine (B1), riboflavin (B2), niacin (B3), vitamin B6, folate (B9), cobalamins as methylcobalamin, hydroxycobalamin, cyanocobalamin (B12) [131], pantothenic acid (B5), biotin (B7), and choline [132]. It also published recommendations of dietary reference intake for vitamin A, vitamin K, arsenic, boron, chromium, copper, iodine, iron, manganese, molybdenum, nickel, silicon, vanadium, and zinc [133], as well as recommendations for vitamin C, vitamin E, selenium, and carotenoids [134]. A list of 10 extra compounds was proposed as they ensure the proper function of longevity proteins, and a shortage of these compounds may result in age-associated chronic disease and cumulative insidious damages [135]. This list of associated dietary compounds includes taurine [136,137,138], ergothioneine [139,140,141], pyrroloquinoline quinone (PQQ) [142,143], queuine [144], carotenoids such as lutein [145], zeaxanthin [145], lycopene [116,146], α-carotene [145], β-carotene [145], β-cryptoxanthin [145,147,148], and astaxanthin [149,150]. Choline is also considered an essential nutrient [135,151,152]. Phenolics are equally considered as vital human dietary components [153]. Other important nutrients are needed to allow for proper biological function. For instance, two macronutrients, fatty acids [154,155,156] (some being essential) and dietary amino acids [157,158], are also critical for metabolism [159]. Choline will be excluded as its interactions relate to a dietary choline metabolite produced by the gut microbiota [160]. Phenolics will be excluded as their interactions are more within the field of drug–herb interactions [161]. Our review will consider exclusively ‘micronutrient’ in the broadest meaning possible, including vitamins, minerals, trace elements, and selected associated dietary compounds (taurine, ergothioneine, PQQ, queuine).

4. ASA and DNIs

4.1. Reported ASA DNIs

4.1.1. Water-Soluble Vitamins

Thiamine (B1)

Increased thiamine urinary excretion. Based on one human case report (five patients at therapeutic doses for rheumatic fever) [162], and two animal models [163,164], ASA may increase urinary excretion of thiamine (Table 1). Clinical relevance was discussed elsewhere, suggesting prophylactic thiamine supplementation [165]. A short period of ASA intake increased thiamin excretion, while long-term use decreased excretion. According to Cleland, this might be due to a loss of body stores during the previous period of increased excretion (Figure 2).

Niacin (B3)

ASA modulating prostaglandins to reduce niacin-flush. Inhibitory drugs of cyclooxygenase reduce flushing by mediating the formation of prostaglandins [166,167,168], improving quality of life in the therapeutic supplementation of niacin [169]. The vascular factor of niacin-flushing is mediated by the hydroxycarboxylic acid receptor 2 (HCA2) and involves cyclooxygenase-mediated formation of prostaglandin D2 and E2 [170,171]. The mechanism was seen in humans (32 healthy subjects) receiving niacin and ASA (80 or 325 mg dose) on four separate visits that were at least 24 h apart [172] (Table 1). A higher dose did not provide additional benefits in another human study (42 healthy subjects, 650 mg ASA daily for four consecutive days) [173] (Figure 2).

Folate (B9)

Multifactorial folate renal excretion. Initially seen in patients with rheumatoid arthritis [174,175,176], ASA was found to interact with folate. Increased urinary excretion, significant fall in serum levels due to competition of binding sites on serum proteins [177,178,179], and interference with folate–oenzyme metabolism are three suspected mechanisms explaining the interaction [180]. ASA may slightly increase urinary excretion of folate based on one human case report (one healthy woman) [177]. Several reports indicate that 70% of patients with rheumatoid arthritis have decreased serum folate, with ASA-induced alterations in serum folate binding [179] (Table 1) (Figure 2).
Fall in folate serum concentration. In Lawrence et al., ASA induced a significant reversible fall in serum folate. This finding might be generalized to non-steroidal anti-inflammatory drugs but not to drugs having only antipyretic or analgesic properties [180,181]. One study challenges if the interaction applies to low-dose ASA as well [182]. Indeed, all studies finding interactions between ASA and folate had high intakes of ASA (for instance, 650 mg ASA every 4 h for 3 days [177]) (Figure 2).
Potentiating ASA through folate supplementation. One article reviewed a mechanism that may potentiate ASA through high-dose folate in acute coronary syndromes and other diseases associated with increased platelet oxidative stress, namely modulation of nitric oxide synthase [183] (Figure 2).

Cobalamins (B12)

Altered absorption of vitamin B12. In a study on 255 patients with cardiovascular diseases, vitamin B12 deficiency was strongly related to the use of ASA [184]. This proposed mechanism could involve reduced absorption due to side effects of ASA on stomach mucosa and reduced secretion of the intrinsic factor [184,185,186,187]. The authors found a protective effect of Helicobacter Pylori on vitamin B12 absorption, supposedly from increased gastric acidity and an increased level of free vitamin B12. Generally, it is seen that Helicobacter is not protective but rather a cause of alterations. Helicobacter Pylori is generally seen as an important factor in the complex [188] cobalamins absorption [189]. Interestingly, unpublished data from a study on B vitamins for improved cognitive functioning in older people with mild cognitive impairment did not find any interaction between ASA and vitamin B12 [190]. Details on interactions of cyanocobalamin, methylcobalamin, and hydroxycobalamin with ASA are not available (Figure 2).

Ascorbic Acid (C)

Unaffected absorption of ASA. Through multi-spectra and voltametric studies, ASA was found to influence vitamin C binding with human serum albumin, but this influence does not affect the absorption of ASA [191].
Decreased absorption of vitamin C [22]. In animal models (guinea pigs and rats), ASA decreased gastrointestinal absorption of vitamin C [192,193], but the effect was thought to be different in humans due to the different absorption mechanisms between animals and humans [194]. A recent spectroscopy study revealed a mechanism that may reduce ascorbic acid absorption in concomitant administration of ASA and vitamin C [191]. Zhang et al. recommended against taking vitamin C concomitantly with ASA, since it was found to reducevitamin C absorption (Figure 2).
Decreased intragastric and serum vitamin C levels [13,28,178,195]. In a double-blind study of 45 healthy human subjects, gastric mucosa and gastric juice was found to be among the largest depots of ascorbic acid, with 25 times the serum concentration (Table 1) [196]. A clinically high dose of ASA (800 mg ASA three times per day for 6 days) lowered ascorbic acid concentration by 10% in gastric mucosa. This decrease may be due to ASA-induced mucosal damage rather than decreased absorption [192], relating to the ascorbic acid gastroprotection effect in co-administration [13,197,198]. Whereas in the case of rheumatoid fever, high doses of ASA interact with ascorbic acid [195], the effect of chronic low-dose ASA on ascorbic acid status is unclear [13].
Increased urinary excretion of vitamin C [22]. The interaction was found in both animal (guinea pigs) [199,200] and human studies (case report of three children, 4–6 years old [201])—(Table 1). In humans, renal excretion stabilizes under chronic use of ASA, with a maximum excretion rate on the 6th day [202]. Enough ascorbic acid is retained for antiscorbutic effects, but there is a 114 percent impact on plasma and leukocytes concentration [199,202]. The derease in leukocyte ascorbic concentration was described in pediatrics as inhibition of vitamin C storage in leukocyte labile store [203]. Loh et al. therefore suggest prophylactic supplementation of ascorbic acid with chronic use of ASA [202] (Figure 2).
Limited impact on specific neuronal Cox-2 inhibition [204]. The neuroprotective effects of ascorbic acid might be related to a local Cox-2 inhibitory effect. Other studies found limited Cox-2 inhibition by ascorbic acid [205,206], but these findings cannot be extrapolated to overall Cox-2 [204].

4.1.2. Fat-Soluble Vitamins

Tocopherols/Tocotrienols (E)

Potentiation of antiplatelet effect with tocopherols and tocotrienols. α-tocopherol has a mild antiplatelet and antioxidant effect [207,208,209,210]. While a sole α-tocopherol supplement is unlikely to have a clinically relevant antiplatelet effect [211], in conjunction with ASA, it might become relevant, increasing the risk of hemorrhage [212] or enhancing a preventive treatment regimen in patients with transient ischemic attacks [213]. Mixed tocopherols might be even more potent [214] (Figure 2).
To harvest this cumulative mechanism and potentiate ASA, the concomitant use of tocopherols and tocotrienols was studied in a double-blind, randomized study on 100 patients (52 patients on ASA + vitamin E, 48 on ASA only) with transient ischemic attacks, minor strokes, or residual ischemic neurologic deficits, with highly significant reduction in platelet adhesiveness (2.7 × 105 platelets adherent/cm2 for ASA + E–4.4 × 105 platelets adherent/cm2 for ASA only) and significant reduction in the incidence of ischemic events in patients in the vitamin E plus aspirin group compared to the aspirin only group (three fatal events in ASA + E–6 in ASA only, one recurrent ischemic attack in ASA + E, two in ASA only, p < 0.05), with no significant difference in the incidence of hemorrhagic stroke between groups (three in both groups over 2 year period) (Table 1) [213]. High dose tocopherols (300 mg/day) are prone to antiplatelet interactions with ASA [215].
Gastric protection against ASA-induced damage. In an animal model (Sprague–Dawley rats), α-tocopherol showed gastric protection of the mucosa through suspected lipid peroxidation inhibition [216]. Tocotrienols are equally effective [217]. Another animal model study suggests that γ-tocopherol exhibits stronger lipid peroxidation inhibition than α-tocopherol and thus provides better gastric protection [218] (Figure 2).

4.1.3. Minerals

Sodium

Decreased excretion at high ASA dose. Prostaglandins mediate renal sodium excretion and extracellular fluid volume regulation [219]. The effect may diminish the chronic use of nonsteroidal anti-inflammatory drugs [220]. Nevertheless, at high doses, decreased renal excretion might have a clinical impact on the patient (i.e., altered blood pressure) [221]. In adults, the dose-dependent renal effect of ASA may be considered from 80 mg per day [222] (Table 1) (Figure 2).

4.1.4. Trace Elements

Iron

Increased risk of anemia. Using data from the Framingham study [223], Fleming et al. isolated data and correlated chronic ASA use (more than seven times a week) with lower serum ferritin levels (a decrease by 21 to 50%) [224] (Table 1), without being able to distinguish latent anemia from an anti-inflammatory response. Another cohort study in Denmark showed similar results [225] (Table 1). A recent study on the elderly using ASA found a fall in ferritin and an increased risk of anemia [226]. Another study analyzed the interaction of ASA and iron in cases of anemia without overt bleeding, with mixed results due to the quality of available data [227]. ASA is irritating to the gastric mucosa [62]. One commentary suggests using chronic ASA-induced iron loss as an anticancer mechanism [228]. It is also known that under certain conditions, ASA can bind with iron [229,230], even though no literature describes this in vivo (Figure 2).

4.1.5. Associated Dietary Compounds

Taurine

Booster of antiplatelet effect. A human study (49 healthy adults, 24–45 years old, free of any medication for at least 15 days, no control group) found further inhibition of platelet aggregation with taurine supplementation [231] (Table 1). Taurine inhibits the aggregation induced by adenosine diphosphate [232]. Hence, coadministration of ASA and taurine might have some benefits in antithrombotic therapy [233], with a possible effect on ASA resistance [231].
Possible gastric protection against ASA-induced damages. In an animal model (Wistar albino rats, 220–300 g), taurine was hypothesized to protect the gastric mucosa [234].
Alteration of excretion. In a human study (six patients treated for rheumatic arthritis), ASA lowered urinary taurine excretion, evidenced by taurine clearance (urinary excretion divided by serum taurine concentration) and a mild rise in taurine serum levels. This mechanism might also be found in healthy subjects [235] (Table 1).
Table 1. Summary of clinically relevant DNIs with ASA. ↑ increase, ↓ decrease.
Table 1. Summary of clinically relevant DNIs with ASA. ↑ increase, ↓ decrease.
NutrimentEffect on Nutrient Status or FunctionHuman StudiesReferences
NumberStudy DesignNumber of PatientsDosageResult
thiamine (B1)↑ excretion1case report5647–1943 mg/day aspirin (ASA) for 5 daysmean urinary excretion thiamine ↑ 50%[154]
niacin (B3)↓ flush (PGD2/PGE2 modulation)2Interventional—4 groups, 1 control31placebo—placebo;
80 mg ASA—500 mg B3;
325 mg ASA—500 mg B3;
Placebo—500 mg B3
↓ warmth,
↓ flushing,
↓ itching,
↓ tingling,
no difference between 325 mg and 650 mg ASA
[164]
interventional—3 groups, 1 control42Placebo—500 mg B3;
325 mg ASA—500 mg B3;
650 mg ASA—500 mg B3
[165]
folate (B9)↑ excretion3observational—37 patients, 59 controls37non indicated high-dose ASA (rhumatoid arthritis treatment)65% (24/37) subnormal folate serum level
(<150 mcg/mL)
[166]
observational1650 mg ASA every 4 h, for 3 dayssubnormal serum folate level[169]
observational182.1–3 g/day ASA, from 1 day to chronic usesubnormal serum folate level (<5 ng/mL)[171]
cobalamins (B12)↓ absorption1descriptive cross-sectional observational study255low dose ASA for secondary prevention of ischemic heart disease①14% patients
<150 pmol/L serum B12
② 30% patients
150–250 pmol/L serum B12
[176]
ascorbic acid (C)↓ C intragastric concentration1interventional—randomized, double-blind, parallel group453 × 80 mg ASA for 6 days↓ gastric mucosa concentration per 10%[188]
↑ urinary excretion1case report3162 mg ASA 2 times at 3 days interval↑ urinary excretion[193]
↓ C leukocyte concentration1interventional10600 mg ASA, 500 mg C↓ C leukocyte concentration by 114%[195]
tocopherols/tocotrienols (E)antiplatelet potentiation of ASA1interventional100325 mg ASA and 400 IU α-tocopherol during 2 yearsplatelet adhesion reduced by 40% ASA + α-tocopherol group[205]
sodium↓ urinary excretion1interventional—2 groups, 1 control16placebo and 160 mg ASA Group 1, 80 mg and 320 mg ASA Group 2interacting with ACE Inhibitors from 80 mg[214]
iron↓ serum ferritin2Interventional—from Framingham heart study, 4 groups913number of ASA per week: non-user, 1–6, 7, >7↓ 25% serum ferritin from >7 ASA per week than non users[215]
interventional—multiple cohorts170 on ASA arm; 1146 placebo armdose ASA unavailable—based on medical history↓Lower serum ferritin (median 136 mcg/L ASA, 169 mcg/L)[216]
↑ anemia1interventional19,114placebo and 100 mg ASAincreased incidence of anemia and decline in ferritin[217]
taurinepotentiation of ASA1interventional49400 mg and 1600 mg/day taurine for 14 daysdecreased aggregability through alteration in TXA2 release and GSH[223]
↓ urinary excretion1interventional6high dose ASA for rhumatoid arthritisIncreased excretion of taurine[227]

4.2. Questionable ASA DNIs

4.2.1. Water-Soluble Vitamins

Riboflavin (B2)

No known interaction. In a study of the complexation of molecules (riboflavin, sodium salicylate, caffeine) in aqueous solution [236], it was found that a combination of riboflavin and sodium salicylate (an analogue of ASA) changed the biological activity of riboflavin. While the relation between caffeine and riboflavin is studied in the context of cutaneous melanoma [237], the relation between salicylates and riboflavin is currently unknown. In a human study (49 patients with migraine), 23 patients received 400 mg of riboflavin with 75 mg of ASA daily. No side effects were seen, except in one patient who withdrew from the study for gastric intolerance [238].

Niacin (B3)

Niacin antiplatelet and fibrinogen effect. Niacin has a direct inhibitory effect on platelet aggregation, corresponding with the metabolism of niacin in 12 human normal blood donors [239]. An hour after an oral dose of niacin, collagen and arachidonic acid-induced platelet aggregation showed a significant reduction, but not so after 12 h. In peripheral vascular disease, niacin has been shown to affect fibrinogen levels [240], which are highly correlated with changes in LDL-c [241]. The clinical significance thereof is unknown.

Pantothenic Acid (B5)

No known interaction between ASA and pantothenic acid. The antiplatelet effect of a biologically active intermediate is to be considered, although its clinical significance is unknown.
Pantethine antiplatelet effect. Pantethine is a biologically active intermediate in the production of Coenzyme A, derived from pantothenic acid (vitamin B5). It is used as a supplement [242]. In a human study on 31 diabetic patients with hyperlipidemia, pantethine had mild antiplatelet aggregation properties [243].

4.2.2. Fat-Soluble Vitamins

Retinol (A)

No interaction in animal model. A study [244] was done on 32 rats on the chronic administration of phenobarbitone, ASA (250 mg/kg/day), and oxytetracycline. The ASA group had normal vitamin A serum levels, comparable to that seen in the control group.
Unclear gastric protection. Several animal model studies suggested a cytoprotective effect for drug-induced gastric mucosal lesions [245,246,247]. 200 mg of ASA-induced gastric mucosal injury benefited from vitamin A administration [247]. But, in a double-blind, placebo-controlled study, no gastric protection was found with β-carotene [248], a provitamin A [249].

4.2.3. Minerals

Phosphorous

Increased urinary excretion. The animal study on the calcemic and calciuric effect also monitored phosphorous excretion and discovered a significant increase from single dose ASA, but no alteration of excretion was found in chronic use [250].

Calcium

Reduced serum and urinary calcium. A study performed on normocalciuric rats showed reduced serum and urinary calcium after a single dose of ASA (−19.6% in serum), as well as with chronic ASA use (−20.8% in serum) [250]. The study suggests, by comparison with indometacin, two distinct mechanisms behind the observed effects–one being prostaglandin inhibition and the other remaining unidentified.

Magnesium

Insignificant magnesium serum increase and decrease of magnesium urinary excretion. Gomaa et al. [250] found an increase in magnesium levels, but it was insignificant compared to normal magnesium levels after a single dose of ASA. There was no alteration of magnesium excretion under chronic ASA. Magnesium bioavailability varies greatly depending on galenics [251,252].

Potassium

No known interaction between ASA and potassium. Conducting a PubMed search (55 results), no relevant studies appeared when the keywords used were ‘acetylsalicylic acid’, ‘aspirin’ and ‘potassium’ and ‘interaction’.

4.2.4. Trace Elements

Copper

Interaction between ASA and copper unclear. Conducting a PubMed search (12 results), only Brumas et al. [253] appeared to be relevant when the keywords used were ‘ASA’, ‘ASA’ with ‘Copper’ and ‘Interaction’. ASA can bind with transition elements such as copper [229,230], and this interaction was found to potentiate the anti-inflammatory effect of ASA [254].

Zinc

No clinically significant interaction between ASA and zinc. In our PubMed search (19 results), no relevant studies appeared when the keywords used were ‘acetylsalicylic acid, ‘aspirin’ and ‘zinc’. ASA can bind with transition elements such as zinc [229,230].

Selenium

Interaction unclear. In a study on patients with ASA-sensitive asthma, lowered enzymatic activity of glutathione peroxidase activity was correlated with lowered serum selenium levels [255]. Inflammation is partially regulated by selenium and selenoproteins through the modulation of eicosanoid biosynthesis [256]. In an animal model, dietary selenium intake did not influence serum or renal excretion [257]. Selenium impacts the arachidonic acid cascade and inhibits the production of thromboxane A2 [258,259].

Chromium

Increased absorption of chromium on rat model with ASA. Two mechanisms are suspected: ASA can bind with transition elements, such as chromium [229,230], but increased absorption could also relate to competition for binding sites on ligands in intestinal absorption between chromium and other metals, like zinc and iron [260,261]. Another suspected mechanism is the inhibition of gastrointestinal prostaglandin synthesis [262].

4.2.5. Associated Dietary Compounds

Lycopene

Unclear additive antiplatelet effect. Lycopene has an antithrombotic and antiplatelet effect [263,264,265]. One study compared platelet aggregation in vitro between ASA and concentrated lycopene and found a synergistic effect at 4 μmol/L of lycopene supplementation daily but not at higher concentrations [266]. For comparison, the median plasma lycopene in an observational study of plasma lycopene concentrations in 111 participants in a trial involving β-carotene was 0.59 μmol/L [267].

α-Carotene

No known interaction between ASA and α-carotene. In a PubMed search (five results), no relevant studies appeared when the keywords used were ‘acetylsalicylic acid, ‘aspirin’ and ‘alpha-carotene’. α-carotene is a provitamin A, contributing to 12–35% of newly converted vitamin A [268]. In an animal model, vitamin A reduced ASA-induced gastric injury [247], but a double-blind placebo-controlled trial on β-carotene, another provitamin A, showed no gastric protection [248].

β-Carotene

No gastric protection against ASA-induced injury. Beta-carotene is a provitamin A and a scavenger antioxidant. In a human double-blind placebo-controlled trial on the effect of chronic β-carotene supplementation (6 months) on the response to acute ASA injury (12 subjects, single oral dose of 650 mg of ASA dissolved in 60 mL of water), endoscopic control found mucosal lesions three hours post-administration in supplemented patients, as well as in the control group [248]. Vitamin A was found to be protective, but not β-carotene, as a provitamin A.

β-Cryptoxanthin

No known interaction between ASA and β-cryptoxanthin. PubMed yielded one irrelevant paper using the keywords ‘acetylsalicylic acid’, ‘aspirin’ with ‘beta-cryptoxanthin’. β-cryptoxanthin is a provitamin A [147,148]. In an animal model, ASA-induced gastric injury benefited from vitamin A [247], but a double-blind placebo-controlled trial on β-carotene, another provitamin A, showed no gastric protection [248].

Astaxanthin

No interaction between ASA and astaxanthin. To test a synthetic astaxanthin derivative, a human study tested the interaction between ASA and astaxanthin on 12 ASA-free subjects and eights subjects treated with ASA. The 25 tested biomarkers of platelet, coagulation, or fibrinolytic activity were unaffected by astaxanthin intake in both groups [269].

4.3. ASA and Metabolic Misuse of Micronutrients

Metabolic misuse. ASA is an electrophoretic uncoupler of mitochondrial oxidative phosphorylation [26]. By reducing the electric potential at the inner mitochondrial membrane below 200 mV (negative inside), it inhibits ATP synthase (complex V) and increases the accumulation of several-fold drugs inside the mitochondria, leading to alterations in processes in the mitochondria matrix [270]. This same membrane potential mechanism makes ASA induce cell death through modulation of the voltage-dependent anion channel [271]. A decrease in bioenergetic efficiency is double-faced: it may be beneficial to cellular function through the reduction of reactive oxidative species production and the balancing of nutrient availability [272], but it may also contribute to increasing mitochondrial dysfunction and toxicity [273], which couldaccount for a significant proportion of adverse effects of prescription drugs [26]. Micronutrients involved in bioenergetics. such as iron, zinc, biotin, vitamin B6, pantothenic acid, and copper [274], would then be required with an increased need—as mitochondria produce around 40 kg of ATP each day [270]. Compensatory mechanisms were revealed in animals [275], and inadequate micronutrient intake would result in further mitochondrial decay [39,274,276]. The clinical relevance of the mechanism in humans has yet to be assessed.

4.4. Unstudied DNIs with ASA

The following elements did not yield any results through PubMed, using ‘acetylsalicylic acid’ and ‘aspirin’ and the respective substance as keywords. Further searches adding ‘absorption’, ‘transport’, ‘metabolism’, or ‘excretion’ yielded no relevant results: biotin (B7)—22 results; chloride—61; sulfur—6; iodine—16; manganese—40; molybdenum—6; fluoride—53; arsenic—52; boron—14; nickel—22; silicon—69; vanadium—7. Interactions between ASA and these elements were not reported.

5. Warfarin and DNIs

5.1. Reported Warfarin DNIs

5.1.1. Water-Soluble Vitamins

Niacin (B3)

One case report on synergistic effect of warfarin and niacin. Niacin has an impact on fibrinogen [240,241], plasminogen inhibitor type 1 (PAI-1) [277], and lipoprotein(a) [278]. One case report found a probable interaction between warfarin and one week‘s intake of high dose niacin (1000 mg) with critically elevated INR (12.3) [279] (Table 2). The case report found no previous occurrence in the literature but offered four hypothetical mechanisms leading to interactions, suggesting careful monitoring with supplementation (Figure 3).

Folate (B9)

Secondary drug–diet interaction inducing folate deficiency. Studies failed to find a relation between homocysteinemia and warfarin levels, but they suspect a drug–diet interaction. Green leafy vegetables are among the important sources of folate [280], as are strawberries [281], whole grain, egg yolk, liver, and citrus fruit [282]. While not being part of an appropriate clinical practice for patients under warfarin [283,284], 68% of patients in a retrospective cohort study involving 317 patients reported being advised to limit or avoid vitamin-K rich foods [285], such as green leafy vegetables. Avoidance of green leafy vegetables might cause a folate deficiency in most patients, as early as after 6 months of therapy with warfarin [286,287] (Table 2) (Figure 3).
No association of serum folate levels with bleeding. In a longitudinal cohort human study on 719 patients taking long-term warfarin, high homocysteine levels (≥17.2 μmol/L) and serum folate (≥11.9 nmol/L) were not associated with bleeding events, but both were associated with cardiovascular events (myocardial infarction, ischemic stroke, peripheral arterial emboli) [288] (Table 2).
Increased clearance of S-7-hydroxywarfarin but of no clinical relevance. Consistent with the lack of association between serum folate levels and bleeding, folic acid supplementation increased clearance of S-7-hydroxywarfarin but did not translate into a clinical change; INR and warfarin dose change being not significant [289] (Table 2).

Ascorbic Acid (C)

Conflicting interaction between high-dose vitamin C and warfarin. The literature is conflicting [290]. Case reports from the 1970s published in JAMA [291,292,293,294,295] initiated animal [296] and human [297,298] studies contradicting any relevant interaction. Still, case reports are published showing interactions between ascorbic acid and warfarin (with INR climbing from 1.1 to 15.4 within 2 days after the discontinuation of vitamin C) [299]. Despite a lack of data to support or refute the interaction, one study hypothesizes an interaction through a common dietary source of vitamin C and vitamin K [298] (Table 2) (Figure 3).

5.1.2. Fat-Soluble Vitamins

Retinol (A)

Possible interaction with high levels of vitamin A. One case report on 13 patients under warfarin on interactions between warfarin and mango fruit [300] (Table 2) referenced a literature article on the interaction between vitamin A supplementation and warfarin [301]. One in vitro analysis of interactions between retinols and cytochrome P450 found that a high retinol dose could inhibit CYP2C19 [302], one of the CYPs responsible for the 7-hydroxylation of the warfarin R-isomer [72] (Figure 3).

Tocopherols/Tocotrienols (E)

Potentiation of warfarin with vitamin E. α-tocopherols has a known mild antiplatelet effect [207,208,209] and has been discussed to be at risk of interaction with warfarin [303]. It was initially found as non-interacting [304] on a double-blind study comprising 25 patients. In more recent studies, serum vitamin E levels were predictive of hemorrhagic events, especially in vitamin K-deficient patients (one interventional study on 12 cardiology patients receiving long-term warfarin, with mild-to-moderate prothrombin times (range, 16.0–21.5 s), and one retrospective observational study on 566 consecutive patients) [305,306] (Table 2). A high dose of tocopherols (300 mg/day) is related to interactions with warfarin [215]. Interactions between vitamin E and vitamin K activity are established, but the precise metabolic pathway is still under discussion [307] (Figure 3).

Calciferols (D)

Vitamin D status and chronic warfarin therapy. In a human study (40 patients with deep vein thrombosis or pulmonary embolism and vitamin D deficiency (<20 ng/mL), oral doses of 50,000 IU vitamin D3 per week for 8 weeks enhanced the anticoagulant effect of warfarin and reduced the maintenance dose requirement [308]. In another human study (89 subjects), only 25 subjects had a normal 25-hydroxyvitamin D level (over 30 ng/mL); 43 patients had an insufficient level (21–29 ng/mL), and 21 had a deficient level (<20 ng/mL). A weak significant association between these levels and the warfarin sensitivity index suggests an interaction [309] (Table 2). Early studies are finding a role of vitamin D in the pathogenesis of thrombosis through the modulation of tissue factor and/or production of cytokines [310]. While no study explicitly analyzed the role of ABCB1 protein in the drug–nutrient interaction between warfarin and D vitamin, it is notable that the ABCB1 protein contributes to vitamin D absorption [311], and its polymorphism has an impact on warfarin dose maintenance [85] (Figure 3).
Vitamin D status and increased risk of arterial calcification. In an animal study, massive artery calcification appeared with high doses of vitamin D [312]. The study describes that high doses of vitamin D might elevate serum calcium levels, and serum calcium elevation might correlate with the onset of artery calcification. The study also considers the role of elevated phosphate serum levels. Warfarin inhibits the activity of the matrix Gla protein as a calcification inhibitor [40] (Figure 3).

K Vitamin

Warfarin is known for the inhibition of hepatic vitamin K epoxide reductase, inhibiting the reduction of vitamin K required for carboxylation of factors II, VII, IX, and X [64]. It is expected that this will decrease the gamma–carboxylation of 17 vitamin K-dependent proteins [313]. As reviewed by McCann and Ames [40], the then 14 known vitamin K-dependent proteins are categorized into three groups: four anticoagulation factors—prothrombin (factor II), factor VII, factor IX, and factor X—believed to all be mostly gamma–carboxylated in the liver; three anticoagulation regulatory proteins—protein C, protein S, and protein Z—mainly gamma–carboxylated in the liver, but also in other tissues; and Matrix GLA protein, osteocalcin, Gas6, Tgfbi, periostin, and proline-rich, Gla proteins 1–4. The report by McCann and Ames is one of the two demonstrating the triage theory in micronutrition [39,42]. Vitamin K is preferentially transported into the liver, where the most urgently required proteins are metabolized, related to coagulation functions [314]. Only when there is hepatic vitamin K sufficiency is vitamin K transported to extrahepatic tissues [315]. Lack of menaquinone-7-trans (the vitamin K2, MK-7-trans form) results in the inactivation of extrahepatic vitamin K-dependent proteins such as matrix GLA protein [316,317] and osteocalcin [318]. The role of the ABCB1 protein in K vitamin transintestinal efflux has been explored in animal models [319]. While the ABCB1 protein also interacts with warfarin [85], it is currently unknown if this has any clinical significance (Figure 3).
Bone mineral density reduction. Studies are not consistent in design, and the impact of antivitamin K on bone mineral density is controversial [320,321]. A pediatric study (70 children with chronic conditions requiring warfarin therapy) reports low bone mineral density (<2.0) in 13% of the patients. Body mass index (BMI) and growth hormone deficiency were identified as risk factors for bone mineral density reduction [322] (Table 2). In an adult study of patients with rheumatic valvular disease (70 patients with mechanical valve replacement), a marked reduction in BMD was found with long-term warfarin use [323] (Table 2). A systematic review and meta-analysis found an increased risk of fractures from warfarin therapy [324], consistent with the inhibition of osteocalcin, the most abundant non-collagenous protein in the bone [325] (Figure 3).
Arterial calcification and atherosclerosis. The use of warfarin is associated with an increase in systemic calcification, including arterial calcification [326], through the inhibition of matrix GLA protein. The etiology of vascular calcification is complex and involves several factors [327]. The long-term effects are debated and include valvular stenosis, stroke, and kidney disease [328]. Valvular and arterial calcification is a major challenge for children under antivitamin K, such as Fontan patients with high and intermediate thromboembolic risk [329]. One study suggested that the extract of Ginkgo Bilola EGB761 might be able to alleviate warfarin-induced aortic valve calcification [330]. This result cannot be transposed to all forms of Ginkgo Biloba, as literature reports have documented serious interactions [331]. Specific supplementation of menaquinone-7-trans (K2, MK7-trans) could specifically target Matrix GLA protein function [316,317], but it is impractical on a clinical level, as low doses of MK-7 supplementation significantly influenced INR values (with a mean value of INR dropping by 40%) [332] (Table 2) (Figure 3).

5.1.3. Minerals

In a review of drug–herb and DNIs with warfarin, it is recommended to delay the administration of warfarin and minerals/trace elements by two hours due to the theorical risk of warfarin reducing absorption by binding [333].

Magnesium

Serum magnesium as a factor in stabilizing INR. In a human study (169 patients, 18–70 years old, under warfarin therapy), serum magnesium levels are a factor stabilizing INR (1.8 ± 0.2 mg/dL in non-stable INR group, 2.0 ± 0.1 mg/dL in stable INR group; p < 0.001) [334] (Table 2). Magnesium (II) is an important component of the coagulation cascade, influencing factor IX or VIII [335,336] (Figure 3).

Potassium

Secondary drug–diet-nutrient interaction. Low vitamin K foods can be high in potassium [337], which can be detrimental in cases of chronic kidney disease (CKD) [338], as those patients are at risk of hyperkalemia.

5.1.4. Associated Dietary Compounds

Astaxanthin

Reported interaction with astaxanthin supplementation. One case report (one patient with ischemic stroke) assessed the probability of a relation between warfarin and the supplement as plausible [339] (Table 2). Astaxanthin mildly inhibits CYP2C19 [340] at a level unlikely to cause interaction. The mechanism is unclear (Figure 3).
Table 2. Summary of clinically relevant DNIs with warfarin. ↑ increase.
Table 2. Summary of clinically relevant DNIs with warfarin. ↑ increase.
NutrimentEffect on Nutrient
Status or Function
Human StudiesReferences
NumberStudy DesignNumber of PatientsDosageResult
niacin (B3)synergistic effect1case report12.5 mg warfarin/day + 1000 mg Niacin/dayINR jumped from 18 months stable INR 2.0–2.9 to 12.3 in a week[271]
folate (B9)no association with bleeding1longitudinal cohort71986% patients in INR 2.0–3.5no association[277]
↑ clearance of S-7-hydroxywarfarin1interventional245 mg/day B9 supplementationnon significant changes in dose and INR[278]
dietary-induced Folate deficiency1observational114dose unavailableimpaired folate status in as little as 6 months[275]
ascorbic acid (C)no interaction8observational57mean 3.3 mg/day warfarinno significant INR change between C vitamin and warfarin dose[287]
retinol (A)possible interaction2case reports13daily mango intake from 1–6 in 2 days to one monthexact mechanism unknown, suspected A vitamin intake through mango[289]
tocopherols/tocotrienols (E)potentiate with vitamin E3observational566dose unavailablehigher serum E predictive of hemorrhagic events[294]
calciferol (D)low 25-OH D status1observational89INR 2.0–3.5 for 3 months from at least 3 consecutive visits25/89 normal (>30 ng/mL);
43/89 subclinical deficiency (21–29 ng/mL);
21/89 clinical deficiency
(<20 ng/mL)
[298]
K vitaminvascular calcification4 quoted in McCann/Ames MGLA inhibition can cause vascular calcification[313]
bone density reduction—pediatrics1observational70more than a year on warfarin13% of patients with BMD < 2.0[309]
bone density reduction—adults1observational70more than a year,
1.25–8.75 mg warfarin/day
Significant decrease in lumbar spine BMD[310]
MK7influence INR interventional1810, 20, 45 mcg/day K2 MK-7mean lowering of INR of 40% at 10 microg, 60% at 20 microg[319]
magnesiumstabilize INR1observational—2 groups, stable and unstable INR169variousMg significantly lower in unstable patients, most influential INR stabilization factor in the study[321]
astaxanthininfluence INR1case report1warfarin 3 mg, astaxanthin 16 mgINR jumped from 1.4 to 10.38. Probable relationship through the scale[327]

5.2. Questionable Warfarin DNIs

5.2.1. Water-Soluble Vitamins

Riboflavin (B2)

No clinically relevant interaction between warfarin and riboflavin. Flavoproteins are involved [341] in the enzymes required for the biosynthesis of the hepatic vitamin K-dependent clotting factors in an animal model [342]. But, flavin deficiency did not impact vitamin K 2,3-epoxide reductase activity, as the latter is not a flavoprotein, nor did it impact the inhibition of the vitamin K epoxide reductase complex by warfarin [342].

Cobalamins (B12)

No interaction between serum cobalamins and warfarin intake. In a human study of 114 patients under warfarin, after 6 months of treatment with warfarin, no significant variation in plasma vitamin B12 levels could be seen [287].

5.2.2. Minerals

In a review of drug–herb and DNIs with warfarin, it is recommended to separate the administration of warfarin and minerals (and trace elements) by two hours due to the theorical risk of warfarin reducing absorption by binding [333].

Calcium

No interaction with calcium urinary excretion. In a human study (11 men on warfarin for at least 90 days), no difference in calcium urinary excretion was seen between the patients taking warfarin and the control group, as well as between people taking warfarin and people who had stopped warfarin, suggesting that there is no important role of a vitamin K-dependent mechanism in renal calcium excretion [343].

Magnesium

Mixed results with respect to absorption by magnesium antacids. In an in vitro model of drug interaction in the gut, absorption of warfarin was lowered by 20% in the presence of magnesium trisilicate [344]. In a human study (12 healthy subjects, 23–32 years old), magnesium hydroxide had no impact on the absorption of warfarin [345]. Different magnesium galenics have different bioavailabilities and may explain different study results [251,346].

Sodium

Clinically used warfarin is a salt [347] to eliminate trace impurities from the amorphous form of warfarin [348]. In a PubMed search (1148 results), no relevant studies appeared when the keywords were ‘warfarin’ with ‘sodium’.

5.2.3. Trace Elements

In a review of drug–herb and DNIs with warfarin, it is recommended to separate the administration of warfarin and minerals (and trace elements) by two hours [333].

Iron

Interaction between warfarin and iron unclear. Stenton et al. mentioned a theorical risk of decreased absorption of warfarin and iron through ligand binding [333]. Through a PubMed search, no case reports of warfarin and iron interaction appeared. Like for ASA, iron status might be impacted by bleeding. This was the hypothesis raised by a human study on increased iron requirement in patients on hemodialysis and antiplatelet therapy or warfarin [349]. Conversely, iron deficiency might alter warfarin metabolization through cytochrome P450, a heme-containing enzyme [350,351].

5.2.4. Associated Dietary Compounds

Lutein

No known interaction between warfarin and lutein. In one study reviewing carotenoids‘ potential of inhibition of cytochrome P450, lutein was found to have no inhibitory effect on studied CYPs [340].

Zeaxanthin

No known interaction between warfarin and zeaxanthin. Using PubMed, no relevant studies appeared when the keywords used were ‘warfarin’ with ‘zeaxanthin’. Zeaxanthin is a mild inhibitor of CYP3A4/5 [340], one of the CYPs responsible for the 10-hydroxylation of the warfarin R-isomer, but at a level unlikely to cause interaction.

Lycopene

No known interaction between warfarin and lycopene. No study investigated any synergistic effect with warfarin.

β-Carotene

No known interaction between warfarin and β-carotene. By PubMed search, no relevant studies appeared when the keywords used were ‘warfarin’ with ‘β-carotene’. It is debated if high β-carotene levels can lower α-tocopherol levels [303], which hypothetically could alter platelet aggregation.

β-Cryptoxanthin

No known interaction between warfarin and β-cryptoxanthin. In a PubMed search, no study appeared when the keywords used were ‘warfarin’ with ‘β-cryptoxanthin’. β-cryptoxanthin is a mild inhibitor of CYP2C8 [340], one of the CYPs responsible for the 7-hydroxylation of the warfarin R-isomer, but at a level unlikely to cause interactions.

5.3. Coenzyme Q10

Coenzyme Q10 is a critical component of the mitochondrial respiratory chain, transporting electrons to complex III in the electron transport chain [352]. It is also an important antioxidant [353,354]. It is, from a chemistry perspective, an analogue of vitamin K [355,356]. Through this analogy, it (in particular, the R-enantiomer [357]) may be interacting with warfarin through cytochrome P450 [358]. There are several case reports on the interaction between coenzyme Q10 and warfarin [359,360,361]. Nevertheless, a randomized, double-blind, placebo-controlled, cross-over trial found no clinical significance of interaction in the coenzyme Q10 supplement (100 mg daily) and warfarin therapy [362]. In a meta-analysis, anticoagulants were found to be associated with increased death from all causes [363]. Induced mitochondrial dysfunction caused by oral anticoagulation, such as warfarin interacting with coenzyme Q10, is questioned [364,365] (Figure 3).

5.4. Unstudied DNIs with Warfarin

With respect to the following elements, PubMed searches yielded no results using ‘warfarin’ and the element’s name. Further research adding ‘absorption’, ‘transport’, ‘metabolism’, or ‘excretion’ yielded no relevant results: thiamine (B1)—2 results; pantothenic acid (B5)—0; pyridoxine (B6)—5; biotin (B7)—16; chloride—14; sulfur—0; copper—16; zinc—23; selenium—8; iodine—73; chromium—16; manganese—0; molybdenum—0; fluoride—7; arsenic—12; boron—3; nickel—8; silicon—36; vanadium—1; taurine—3; ergothioneine—0; pyrroloquinoline quinone—0; queuine—0; α-carotene—0.

6. Discussion

The daily dietary intake of nutrients (from macronutrients (fat, protein carbohydrates), and micronutrients (vitamins, minerals, trace elements)) plays an essential role in human health. The daily consumption of nutrients is not uniform, displaying intra- and interindividual variation. Intraindividual variation reflects the temporal fluctuations in nutrient intake experienced by an individual, influenced by factors such as meals, timing, appetite, lifestyle adjustments, as well as biological ones (absorption, transport, metabolism, and excretion influenced by the physiology and the pathophysiology specific to each individual).
Polymorphisms affecting the absorption, transport, metabolism, and excretion of micronutrients such as vitamins, minerals, trace elements, and other cofactors play a crucial role in complex biochemistry processes. These interactions occur at various stages, from ingestion to the final reabsorption in the excretion system. Micronutrients, such as vitamins and minerals, often work synergistically or competitively with one another, impacting their absorption and bioavailability—the balance between copper/zinc/iron is a classic example [366]. Additionally, the presence of various dietary components, such as fiber, and macronutrients, such as carbohydrates, fats, or proteins, can further modulate the absorption of essential nutrients. Gut microbiota might also influence the occurrence of interactions through the modulation of drug efficiency [367,368,369]. Nutrition research has made significant discoveries, highlighting the complexity of nutrition and the importance of a well-formulated diet to ensure optimal nutrient absorption, transport, metabolism, excretion of nutrients, as well as non-nutrients required to optimize phase I and phase II metabolic processes [28], gut microbiome status, and overall health.
DNIs are a challenge for clinicians. Available data are limited and are mostly based on case reports or post-marketing observational studies [44]. Nevertheless, it is recognized that DNIs may influence health outcomes in vulnerable populations, such as elderly, obese, critically ill, transplant recipients, patients receiving enteral or parenteral nutrition, chronically diseased patients, or patients under polypharmacy [44]. Polypharmacy is generally defined as five medications for an adult [370] or two medications at the same time (for either more than a day or more than a month [371]) for an infant. In the elderly, the number of medications is associated with poorer nutritional status [372]. Polypharmacy increases the risk of drug–drug interactions, making the overall nutritional picture harder to understand, with its cumulative and synergistic effects [373]. Pediatric patients are to be equally considered [374,375], especially vulnerable groups—for example, patients with complex congenital heart disease, such as patients with Fontan circulation [40,376,377,378,379,380,381,382], on chronic polypharmacy. Warfarin-induced arterial calcification, amplified by growth and vitamin D, is challenging the widespread use of ASA and antivitamin K on the pediatric population [312,383]. Among the high-risk medications for DNIs is warfarin [384]. It is likely that drugs with a high risk of common drug–drug interactions are also of concern [44], including, among others, drugs with a low therapeutic range and drugs interacting with CYP450 [385].
Furthermore, when listing the limitations of DNI literature, clinical importance should be considered. Micronutritional deficiencies are common in western countries as well as developing countries [36,374]. DNIs and polypharmacy theoretically increase the risks of micronutritional deficiencies, enhancing the risk of adverse effect on chronically ill people with impaired nutritional status [9].
Not all micronutritional deficiencies have evident symptoms and remain undiagnosed [98,386]. Unclear symptoms of many micronutrient deficiencies [387] might hide behind the pathophysiology and course of the disease.
Making the global picture even more complex, not all micronutritional deficiencies have immediate pathophysiologic effects. In triage theory [39], it is suspected that the limited availability of micronutrients can cause an increase in chronic diseases. On an evolutionary premise, short-term survival for reproduction is favored over long-term health [37,38,388]. The triage theory has been demonstrated from a biochemical perspective and in animal models, with K vitamin-dependent proteins [40,314] and selenoproteins [42], where mild micronutritional deficiencies can cause insidious damage, accelerating the deterioration of chronic diseases [386]. The issue might be particularly concerning for the pediatric population with chronic illnesses, as this would lead to accelerated deterioration of the disease with micronutritional deficiencies, or conversely, slowing down the deterioration process [135]. Thus, it is important that clinicians pay more attention to nutritional status as part of a normal medical assessment of each patient.
Several approaches might improve the patient’s clinical outcome.
From the pharmacist and clinician’s view, systematically considering the DNIs.
From the clinician’s perspective, stratifying the risk of interactions as part of the patient assessment or drug regimen review process, on an individual patient basis, as suggested by Boullata [44,389].
As a patient-centered approach, doing a micronutritional assessment as part of the medical examination for vulnerable populations, even using supplementation [390]. This can be done through medical history and methods of questioning (anamnesis) [391], as well as appropriate biomarker assessment on an individual patient basis [92].
When facing a possible DNI, clinician can use a drug-interaction probability scale designed to consider the causal relationship [392]. This scale is already in use by clinicians for the evaluation and reporting of DNIs [44,91,393,394] (Figure 4).
Karadima et al. suggested a major change [9]—a healthcare model using predictive, preventive, personalized medicine [395], taking advantage of omics technologies [396].
Through clinical [397,398,399] use of genomics [400,401], nutrigenetics [402], proteomics [398,403], metabolomics [404,405], and bioinformatics [406], clinicians may detect signs of diseases at a preventive stage, as well as stratify the risk of DNI [9]. Omics can be used clinically to assess the efficiency of micronutritional intervention [407,408] or the functionality of systems such as antioxidative defenses [409]. Omics could bring new insights into the importance of personalized therapeutic nutrition [410]. Drug–nutrient–genome interactions are being explored in cardiology [411]. Polymorphisms in the vitamin K epoxide reductase complex (VKOR) have been found to impact the interaction between warfarin and K vitamin through drug–nutrient–genome analysis [412].

7. Limitations

Even though ASA and warfarin are among the oldest and most prescribed drugs worldwide, the review showed clear limitations.
Analyzing the DNIs demonstrated a lack of unified methodology and unified dose. Human cohort studies are relatively small. Some ASA–nutrient interaction studies’ designs were meant for rheumatic arthritis patients receiving high-dose ASA, which cannot be extrapolated to cardiovascular protective low-dose ASA.
Several protocols were realized on animal models only, questioning if the effects can be extrapolated to humans. The difference in gastrointestinal absorption of vitamin C between guinea pigs and humans illustrates that this is not always the case.
For a non-negligible amount of micronutrients (15 out of 45 for ASA; more than 30 out of 45 for warfarin, including all trace elements), we are currently missing data and thus cannot include or exclude a DNI.
Interactions between retinoids/carotenoids and ASA were also barely explored. Sometimes, even proper diagnostic tests for marginal micronutritional deficiencies are unavailable, like in trace elements [113]. Moreover, not all polymorphisms of the main mechanisms behind antiplatelet and/or antithrombotic effects of ASA and warfarin have been identified. Additionally, polypharmacy could equally change DNIs, particularly with synergistic mechanisms.
All these limitations do not detract from the intermediate objective of this review—paving the way for risk stratification for DNIs and resulting micronutritional deficiencies with the chronic use of ASA or warfarin.

8. Conclusions

DNIs are reported occasionally in both warfarin and ASA users. A limitation of this assessment is the scarcity of available data. Potential DNIs could be managed through various strategies.

Author Contributions

Conceptualization, D.R. and M.M.; investigation, D.R. and M.M.; resources, M.M.; writing—original draft preparation, D.R.; writing—review and editing, M.M. and A.H.; supervision, M.M.; project administration, D.R. and M.M.; funding acquisition, D.R. All authors have read and agreed to the published version of the manuscript.

Funding

M.M. is receiving a grant (KLI-1036B) by the FWF.

Acknowledgments

This work is dedicated to the lovely Bernadette-Marie, born with HLHS.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Bushra, R.; Aslam, N.; Khan, A. Food Drug Interactions. Oman Med. J. 2011, 26, 77–83. [Google Scholar] [CrossRef] [PubMed]
  2. Vuong, M.; González Aragón, C.; Montarroyos, S.S. Common Food and Drug Interactions. Pediatr. Rev. 2023, 44, 68–80. [Google Scholar] [CrossRef] [PubMed]
  3. Deng, J.; Zhu, X.; Chen, Z.; Fan, C.H.; Kwan, H.S.; Wong, C.H.; Shek, K.Y.; Zuo, Z.; Lam, T.N. A Review of Food–Drug Interactions on Oral Drug Absorption. Drugs 2017, 77, 1833–1855. [Google Scholar] [CrossRef] [PubMed]
  4. Roe, D.A. Diet and Drug Interactions; Springer: Dordrecht, The Netherlands, 1989. [Google Scholar] [CrossRef]
  5. Fugh-Berman, A. Herb-Drug Interactions. Lancet 2000, 355, 134–138. [Google Scholar] [CrossRef] [PubMed]
  6. Hu, Z.; Yang, X.; Ho, P.C.L.; Chan, S.Y.; Heng, P.W.S.; Chan, E.; Duan, W.; Koh, H.L.; Zhou, S. Herb-Drug Interactions: A Literature Review. Drugs 2005, 65, 1239–1282. [Google Scholar] [CrossRef] [PubMed]
  7. Izzo, A.A.; Ernst, E. Interactions Between Herbal Medicines and Prescribed Drugs: A Systematic Review. Drugs 2001, 61, 2163–2175. [Google Scholar] [CrossRef] [PubMed]
  8. Izzo, A.A.; Di Carlo, G.; Borrelli, F.; Ernst, E. Cardiovascular Pharmacotherapy and Herbal Medicines: The Risk of Drug Interaction. Int. J. Cardiol. 2005, 98, 1–14. [Google Scholar] [CrossRef]
  9. Karadima, V.; Kraniotou, C.; Bellos, G.; Tsangaris, G.T. Drug-Micronutrient Interactions: Food for Thought and Thought for Action. EPMA J. 2016, 7, 10. [Google Scholar] [CrossRef] [PubMed]
  10. Samaras, D.; Samaras, N.; Lang, P.-O.; Genton, L.; Frangos, E.; Pichard, C. Effects of Widely Used Drugs on Micronutrients: A Story Rarely Told. Nutrition 2013, 29, 605–610. [Google Scholar] [CrossRef]
  11. Chan, L. Drug-Nutrient Interactions. J. Parenter. Enter. Nutr. 2013, 37, 450–459. [Google Scholar] [CrossRef]
  12. Santos, C.A.; Boullata, J.I. An Approach to Evaluating Drug-Nutrient Interactions. Pharmacotherapy 2005, 25, 1789–1800. [Google Scholar] [CrossRef] [PubMed]
  13. Mohn, E.; Kern, H.; Saltzman, E.; Mitmesser, S.; McKay, D. Evidence of Drug–Nutrient Interactions with Chronic Use of Commonly Prescribed Medications: An Update. Pharmaceutics 2018, 10, 36. [Google Scholar] [CrossRef] [PubMed]
  14. Roe, D.A. Drug and Food Interactions as They Affect the Nutrition of Older Individuals. Aging Milan Italy 1993, 5, 51–53. [Google Scholar] [PubMed]
  15. Chan, L.-N. Drug-Nutrient Interaction in Clinical Nutrition. Curr. Opin. Clin. Nutr. Metab. Care 2002, 5, 327–332. [Google Scholar] [CrossRef] [PubMed]
  16. Gröber, U. Interactions between drugs and micronutrients. Med. Monatsschr. Pharm. 2006, 29, 26–35. [Google Scholar] [CrossRef] [PubMed]
  17. Gunturu, S.G.; Dharmarajan, T.S.; Dharmarajan, T.S.; Dharmarajan, T.S.; Dharmarajan, T.S. Drug–Nutrient Interactions. In Geriatric Gastroenterology; Pitchumoni, C.S., Dharmarajan, T.S., Eds.; Springer: New York, NY, USA, 2012; pp. 89–98. [Google Scholar] [CrossRef]
  18. Boullata, J.I. Influence of Medication on Nutritional Status. In Preventive Nutrition; Bendich, A., Deckelbaum, R.J., Eds.; Humana Press: Totowa, NJ, USA, 2005; pp. 833–868. [Google Scholar] [CrossRef]
  19. Said, H.M.; Redha, R.; Nylander, W. Biotin Transport in the Human Intestine: Inhibition by Anticonvulsant Drugs. Am. J. Clin. Nutr. 1989, 49, 127–131. [Google Scholar] [CrossRef] [PubMed]
  20. Mock, D.M.; Dyken, M.E. Biotin Catabolism Is Accelerated in Adults Receiving Long-Term Therapy with Anticonvulsants. Neurology 1997, 49, 1444–1447. [Google Scholar] [CrossRef] [PubMed]
  21. Lombardi, L.R.; Kreys, E.; Gerry, S.; Boullata, J.I. Nutrition in the Age of Polypharmacy. In Preventive Nutrition; Bendich, A., Deckelbaum, R.J., Eds.; Humana Press: Totowa, NJ, USA, 2010; pp. 79–123. [Google Scholar] [CrossRef]
  22. Prescott, J.D.; Drake, V.J.; Stevens, J.F. Medications and Micronutrients: Identifying Clinically Relevant Interactions and Addressing Nutritional Needs. J. Pharm. Technol. 2018, 34, 216–230. [Google Scholar] [CrossRef] [PubMed]
  23. Boullata, J.I. Drug and Nutrition Interactions: Not Just Food for Thought. J. Clin. Pharm. Ther. 2013, 38, 269–271. [Google Scholar] [CrossRef] [PubMed]
  24. Genser, D. Food and Drug Interaction: Consequences for the Nutrition/Health Status. Ann. Nutr. Metab. 2008, 52 (Suppl. S1), 29–32. [Google Scholar] [CrossRef] [PubMed]
  25. White, R. Drugs and Nutrition: How Side Effects Can Influence Nutritional Intake. Proc. Nutr. Soc. 2010, 69, 558–564. [Google Scholar] [CrossRef] [PubMed]
  26. Wallace, K.B. Drug-Induced Mitochondrial Neuropathy in Children: A Conceptual Framework for Critical Windows of Development. J. Child Neurol. 2014, 29, 1241–1248. [Google Scholar] [CrossRef]
  27. Mason, P. Important Drug–Nutrient Interactions. Proc. Nutr. Soc. 2010, 69, 551–557. [Google Scholar] [CrossRef] [PubMed]
  28. Thurnham, D.I. An Overview of Interactions between Micronutrients and of Micronutrients with Drugs, Genes and Immune Mechanisms. Nutr. Res. Rev. 2004, 17, 211–240. [Google Scholar] [CrossRef] [PubMed]
  29. Polk, R.E.; Healy, D.P.; Sahai, J.; Drwal, L.; Racht, E. Effect of Ferrous Sulfate and Multivitamins with Zinc on Absorption of Ciprofloxacin in Normal Volunteers. Antimicrob. Agents Chemother. 1989, 33, 1841–1844. [Google Scholar] [CrossRef] [PubMed]
  30. Campbell, N.; Kara, M.; Hasinoff, B.; Haddara, W.; McKay, D. Norfloxacin Interaction with Antacids and Minerals. Br. J. Clin. Pharmacol. 1992, 33, 115–116. [Google Scholar] [CrossRef] [PubMed]
  31. Btaiche, I.F.; Sweet, B.V.; Kraft, M.D. Positive Drug–Nutrient Interactions. In Handbook of Drug-Nutrient Interactions; Boullata, J.I., Armenti, V.T., Eds.; Humana Press: Totowa, NJ, USA, 2009; pp. 303–339. [Google Scholar] [CrossRef]
  32. Proell, P.; Congpuong, K.; Reinthaler, F.F.; Wernsdorfer, G.; Wiedermann, U.; Wernsdorfer, W.H. Synergism between Pyronaridine and Retinol in Plasmodium Falciparum in Vitro. Wien. Klin. Wochenschr. 2009, 121 (Suppl. S3), 32. [Google Scholar] [CrossRef] [PubMed]
  33. Byshchevskił, A.S.; Galiyan, S.L.; Vakulin, A.A.; Dement’eva, I.A.; SAbirov, A.X. [Influence of vitamins-antioxidants on antiaggregative effect of acetylsalicylic acid]. Ukr. Biokhimicheskii Zhurnal 1978 1997, 69, 93–98. [Google Scholar]
  34. Drain, P.K.; Kupka, R.; Mugusi, F.; Fawzi, W.W. Micronutrients in HIV-Positive Persons Receiving Highly Active Antiretroviral Therapy. Am. J. Clin. Nutr. 2007, 85, 333–345. [Google Scholar] [CrossRef] [PubMed]
  35. Nose, S.; Wasa, M.; Tazuke, Y.; Owari, M.; Fukuzawa, M. Cisplatin Upregulates Glutamine Transport in Human Intestinal Epithelial Cells: The Protective Mechanism of Glutamine on Intestinal Mucosa After Chemotherapy. J. Parenter. Enter. Nutr. 2010, 34, 530–537. [Google Scholar] [CrossRef] [PubMed]
  36. Bailey, R.L.; West, K.P., Jr.; Black, R.E. The Epidemiology of Global Micronutrient Deficiencies. Ann. Nutr. Metab. 2015, 66 (Suppl. S2), 22–33. [Google Scholar] [CrossRef] [PubMed]
  37. Kirkwood, T.B.L. Understanding Ageing from an Evolutionary Perspective: 4th Key Symposium: Understanding Ageing. J. Intern. Med. 2008, 263, 117–127. [Google Scholar] [CrossRef] [PubMed]
  38. Kirkwood, T.B.L.; Austad, S.N. Why Do We Age? Nature 2000, 408, 233–238. [Google Scholar] [CrossRef]
  39. Ames, B.N. Low Micronutrient Intake May Accelerate the Degenerative Diseases of Aging through Allocation of Scarce Micronutrients by Triage. Proc. Natl. Acad. Sci. USA 2006, 103, 17589–17594. [Google Scholar] [CrossRef] [PubMed]
  40. McCann, J.C.; Ames, B.N. Vitamin K, an Example of Triage Theory: Is Micronutrient Inadequacy Linked to Diseases of Aging? Am. J. Clin. Nutr. 2009, 90, 889–907. [Google Scholar] [CrossRef] [PubMed]
  41. Kaźmierczak-Barańska, J.; Karwowski, B.T. Vitamin K Contribution to DNA Damage—Advantage or Disadvantage? A Human Health Response. Nutrients 2022, 14, 4219. [Google Scholar] [CrossRef] [PubMed]
  42. McCann, J.C.; Ames, B.N. Adaptive Dysfunction of Selenoproteins from the Perspective of the Triage Theory: Why Modest Selenium Deficiency May Increase Risk of Diseases of Aging. FASEB J. 2011, 25, 1793–1814. [Google Scholar] [CrossRef]
  43. Péter, S.; Navis, G.; de Borst, M.H.; von Schacky, C.; van Orten-Luiten, A.C.B.; Zhernakova, A.; Witkamp, R.F.; Janse, A.; Weber, P.; Bakker, S.J.L.; et al. Public Health Relevance of Drug–Nutrition Interactions. Eur. J. Nutr. 2017, 56 (Suppl. S2), 23–36. [Google Scholar] [CrossRef] [PubMed]
  44. Boullata, J.I.; Hudson, L.M. Drug–Nutrient Interactions: A Broad View with Implications for Practice. J. Acad. Nutr. Diet. 2012, 112, 506–517. [Google Scholar] [CrossRef] [PubMed]
  45. Boullata, J.I.; Armenti, V.T. (Eds.) Handbook of Drug-Nutrient Interactions; Humana Press: Totowa, NJ, USA, 2010. [Google Scholar] [CrossRef]
  46. Sjöqvist, F.; Böttiger, Y. Historical Perspectives: Drug Interactions—It All Began with Cheese. J. Intern. Med. 2010, 268, 512–515. [Google Scholar] [CrossRef] [PubMed]
  47. Drummond, D.; Kirby, A.N.; Gamston, C.E.; Westrick, S.; Saunders, J.; Lloyd, K.B. Impact of Interdisciplinary Education on Pharmacy Student Knowledge and Comfort with Counseling on Drug-Nutrient Interactions. Curr. Pharm. Teach. Learn. 2021, 13, 382–390. [Google Scholar] [CrossRef] [PubMed]
  48. Desborough, M.J.R.; Keeling, D.M. The Aspirin Story—From Willow to Wonder Drug. Br. J. Haematol. 2017, 177, 674–683. [Google Scholar] [CrossRef] [PubMed]
  49. Tanasescu, S.; Lévesque, H.; Thuillez, C. Pharmacologie de l’aspirine. Rev. Médecine Interne 2000, 21, S18–S26. [Google Scholar] [CrossRef]
  50. Bjorkman, D.J. The Effect of Aspirin and Nonsteroidal Anti-Inflammatory Drugs on Prostaglandins. Am. J. Med. 1998, 105, 8S–12S. [Google Scholar] [CrossRef] [PubMed]
  51. Shimokawa, T.; Smith, W.L. Prostaglandin Endoperoxide Synthase. The Aspirin Acetylation Region. J. Biol. Chem. 1992, 267, 12387–12392. [Google Scholar] [CrossRef] [PubMed]
  52. Tacconelli, S.; Contursi, A.; Falcone, L.; Mucci, M.; D’Agostino, I.; Fullone, R.; Sacco, A.; Zucchelli, M.; Bruno, A.; Ballerini, P.; et al. Characterization of Cyclooxygenase-2 Acetylation and Prostanoid Inhibition by Aspirin in Cellular Systems. Biochem. Pharmacol. 2020, 178, 114094. [Google Scholar] [CrossRef] [PubMed]
  53. Schrör, K. Aspirin and Platelets: The Antiplatelet Action of Aspirin and Its Role in Thrombosis Treatment and Prophylaxis. Semin. Thromb. Hemost. 1997, 23, 349–356. [Google Scholar] [CrossRef] [PubMed]
  54. Vane, J.R. Inhibition of Prostaglandin Synthesis as a Mechanism of Action for Aspirin-like Drugs. Nat. New Biol. 1971, 231, 232–235. [Google Scholar] [CrossRef] [PubMed]
  55. Hochgesang, G.P.; Rowlinson, S.W.; Marnett, L.J. Tyrosine-385 Is Critical for Acetylation of Cyclooxygenase-2 by Aspirin. J. Am. Chem. Soc. 2000, 122, 6514–6515. [Google Scholar] [CrossRef]
  56. Ajjan, R.A.; Standeven, K.F.; Khanbhai, M.; Phoenix, F.; Gersh, K.C.; Weisel, J.W.; Kearney, M.T.; Ariëns, R.A.S.; Grant, P.J. Effects of Aspirin on Clot Structure and Fibrinolysis Using a Novel In Vitro Cellular System. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 712–717. [Google Scholar] [CrossRef] [PubMed]
  57. Undas, A.; Brummel-Ziedins, K.E.; Mann, K.G. Antithrombotic Properties of Aspirin and Resistance to Aspirin: Beyond Strictly Antiplatelet Actions. Blood 2007, 109, 2285–2292. [Google Scholar] [CrossRef]
  58. Oberle, S.; Polte, T.; Abate, A.; Podhaisky, H.-P.; Schröder, H. Aspirin Increases Ferritin Synthesis in Endothelial Cells: A Novel Antioxidant Pathway. Circ. Res. 1998, 82, 1016–1020. [Google Scholar] [CrossRef] [PubMed]
  59. Xu, L.; Wang, Y. Combined Influence of ABCB1 Genetic Polymorphism and DNA Methylation on Aspirin Resistance in Chinese Ischemic Stroke Patients. Acta Neurol. Belg. 2022, 122, 1057–1064. [Google Scholar] [CrossRef] [PubMed]
  60. Angiolillo, D.J.; Capodanno, D. Aspirin for Primary Prevention of Cardiovascular Disease in the 21st Century: A Review of the Evidence. Am. J. Cardiol. 2021, 144, S15–S22. [Google Scholar] [CrossRef]
  61. Undas, A.; Sydor, W.J.; Brummel, K.; Musial, J.; Mann, K.G.; Szczeklik, A. Aspirin Alters the Cardioprotective Effects of the Factor XIII Val34Leu Polymorphism. Circulation 2003, 107, 17–20. [Google Scholar] [CrossRef]
  62. Dubois, R.W.; Melmed, G.Y.; Henning, J.M.; Bernal, M. Risk of Upper Gastrointestinal Injury and Events in Patients Treated with Cyclooxygenase (COX)-1/COX-2 Nonsteroidal Antiinflammatory Drugs (NSAIDs), COX-2 Selective NSAIDs, and Gastroprotective Cotherapy: An Appraisal of the Literature. JCR J. Clin. Rheumatol. 2004, 10, 178–189. [Google Scholar] [CrossRef] [PubMed]
  63. Fuentes, A.; Pineda, M.; Venkata, K. Comprehension of Top 200 Prescribed Drugs in the US as a Resource for Pharmacy Teaching, Training and Practice. Pharmacy 2018, 6, 43. [Google Scholar] [CrossRef] [PubMed]
  64. Ansell, J.; Hirsh, J.; Hylek, E.; Jacobson, A.; Crowther, M.; Palareti, G. Pharmacology and Management of the Vitamin K Antagonists. Chest 2008, 133, 160S–198S. [Google Scholar] [CrossRef]
  65. Esmon, C.T.; Vigano-D’Angelo, S.; D’Angelo, A.; Comp, P.C. Anticoagulation Proteins C and S. Adv. Exp. Med. Biol. 1987, 214, 47–54. [Google Scholar] [CrossRef]
  66. Lim, G.B. Warfarin: From Rat Poison to Clinical Use. Nat. Rev. Cardiol. 2017. [Google Scholar] [CrossRef]
  67. Jaffer, I.H.; Weitz, J.I. Antithrombotic Drugs. In Hematology; Elsevier: Amsterdam, The Netherlands, 2018; pp. 2168–2188. [Google Scholar] [CrossRef]
  68. Kaminsky, L.S.; Zhang, Z.-Y. Human P450 Metabolism of Warfarin. Pharmacol. Ther. 1997, 73, 67–74. [Google Scholar] [CrossRef] [PubMed]
  69. Takahashi, H.; Echizen, H. Pharmacogenetics of Warfarin Elimination and Its Clinical Implications. Clin. Pharmacokinet. 2001, 40, 587–603. [Google Scholar] [CrossRef] [PubMed]
  70. Takahashi, H.; Kashima, T.; Nomizo, Y.; Muramoto, N.; Shimizu, T.; Nasu, K.; Kubota, T.; Kimura, S.; Echizen, H. Metabolism of Warfarin Enantiomers in Japanese Patients with Heart Disease Having Different CYP2C9 and CYP2C19 Genotypes*. Clin. Pharmacol. Ther. 1998, 63, 519–528. [Google Scholar] [CrossRef] [PubMed]
  71. Guengerich, F.P. Characterization of Human Cytochrome P450 Enzymes. FASEB J. 1992, 6, 745–748. [Google Scholar] [CrossRef] [PubMed]
  72. Norwood, D.A.; Parke, C.K.; Rappa, L.R. A Comprehensive Review of Potential Warfarin-Fruit Interactions. J. Pharm. Pract. 2015, 28, 561–571. [Google Scholar] [CrossRef] [PubMed]
  73. Okey, A.B. Enzyme Induction in the Cytochrome P-450 System. Pharmacol. Ther. 1990, 45, 241–298. [Google Scholar] [CrossRef] [PubMed]
  74. Rettie, A.E.; Korzekwa, K.R.; Kunze, K.L.; Lawrence, R.F.; Eddy, A.C.; Aoyama, T.; Gelboin, H.V.; Gonzalez, F.J.; Trager, W.F. Hydroxylation of Warfarin by Human cDNA-Expressed Cytochrome P-450: A Role for P-4502C9 in the Etiology of (S)-Warfarin-Drug Interactions. Chem. Res. Toxicol. 1992, 5, 54–59. [Google Scholar] [CrossRef]
  75. Eble, J.N.; West, B.D.; Link, K.P. A Comparison of the Isomers of Warfarin. Biochem. Pharmacol. 1966, 15, 1003–1006. [Google Scholar] [CrossRef]
  76. Ingelman-Sundberg, M. Polymorphism of Cytochrome P450 and Xenobiotic Toxicity. Toxicology 2002, 181–182, 447–452. [Google Scholar] [CrossRef] [PubMed]
  77. Preissner, S.C.; Hoffmann, M.F.; Preissner, R.; Dunkel, M.; Gewiess, A.; Preissner, S. Polymorphic Cytochrome P450 Enzymes (CYPs) and Their Role in Personalized Therapy. PLoS ONE 2013, 8, e82562. [Google Scholar] [CrossRef] [PubMed]
  78. Wang, B.; Wang, J.; Huang, S.-Q.; Su, H.-H.; Zhou, S.-F. Genetic Polymorphism of the Human Cytochrome P450 2C9 Gene and Its Clinical Significance. Curr. Drug Metab. 2009, 10, 781–834. [Google Scholar] [CrossRef] [PubMed]
  79. Herman, D.; Locatelli, I.; Grabnar, I.; Peternel, P.; Stegnar, M.; Mrhar, A.; Breskvar, K.; Dolzan, V. Influence of CYP2C9 Polymorphisms, Demographic Factors and Concomitant Drug Therapy on Warfarin Metabolism and Maintenance Dose. Pharmacogenomics J. 2005, 5, 193–202. [Google Scholar] [CrossRef] [PubMed]
  80. Lindh, J.; Lundgren, S.; Holm, L.; Alfredsson, L.; Rane, A. Several-Fold Increase in Risk of Overanticoagulation by CYP2C9 Mutations. Clin. Pharmacol. Ther. 2005, 78, 540–550. [Google Scholar] [CrossRef] [PubMed]
  81. Uno, T.; Sugimoto, K.; Sugawara, K.; Tateishi, T. The Effect of CYP2C19 Genotypes on the Pharmacokinetics of Warfarin Enantiomers. J. Clin. Pharm. Ther. 2008, 33, 67–73. [Google Scholar] [CrossRef] [PubMed]
  82. Karl, J.P.; Meydani, M.; Barnett, J.B.; Vanegas, S.M.; Barger, K.; Fu, X.; Goldin, B.; Kane, A.; Rasmussen, H.; Vangay, P.; et al. Fecal Concentrations of Bacterially Derived Vitamin K Forms Are Associated with Gut Microbiota Composition but Not Plasma or Fecal Cytokine Concentrations in Healthy Adults. Am. J. Clin. Nutr. 2017, 106, 1052–1061. [Google Scholar] [CrossRef] [PubMed]
  83. Aydin, S. Can Vitamin K Synthesis Altered by Dysbiosis of Microbiota Be Blamed in the Etiopathogenesis of Venous Thrombosis? Biosci. Microbiota Food Health 2017, 36, 73–74. [Google Scholar] [CrossRef] [PubMed]
  84. Caldwell, M.D.; Awad, T.; Johnson, J.A.; Gage, B.F.; Falkowski, M.; Gardina, P.; Hubbard, J.; Turpaz, Y.; Langaee, T.Y.; Eby, C.; et al. CYP4F2 Genetic Variant Alters Required Warfarin Dose. Blood 2008, 111, 4106–4112. [Google Scholar] [CrossRef] [PubMed]
  85. Wadelius, M.; Sörlin, K.; Wallerman, O.; Karlsson, J.; Yue, Q.-Y.; Magnusson, P.K.E.; Wadelius, C.; Melhus, H. Warfarin Sensitivity Related to CYP2C9, CYP3A5, ABCB1 (MDR1) and Other Factors. Pharmacogenomics J. 2004, 4, 40–48. [Google Scholar] [CrossRef] [PubMed]
  86. Kuruvilla, M.; Gurk-Turner, C. A Review of Warfarin Dosing and Monitoring. Bayl. Univ. Med. Cent. Proc. 2001, 14, 305–306. [Google Scholar] [CrossRef] [PubMed]
  87. Davidson, M.H. Does Differing Metabolism by Cytochrome P450 Have Clinical Importance? Curr. Atheroscler. Rep. 2000, 2, 14–19. [Google Scholar] [CrossRef] [PubMed]
  88. Gage, B.; Eby, C.; Johnson, J.; Deych, E.; Rieder, M.; Ridker, P.; Milligan, P.; Grice, G.; Lenzini, P.; Rettie, A.; et al. Use of Pharmacogenetic and Clinical Factors to Predict the Therapeutic Dose of Warfarin. Clin. Pharmacol. Ther. 2008, 84, 326–331. [Google Scholar] [CrossRef] [PubMed]
  89. Di Minno, A.; Frigerio, B.; Spadarella, G.; Ravani, A.; Sansaro, D.; Amato, M.; Kitzmiller, J.P.; Pepi, M.; Tremoli, E.; Baldassarre, D. Old and New Oral Anticoagulants: Food, Herbal Medicines and Drug Interactions. Blood Rev. 2017, 31, 193–203. [Google Scholar] [CrossRef] [PubMed]
  90. Holbrook, A.M. Systematic Overview of Warfarin and Its Drug and Food Interactions. Arch. Intern. Med. 2005, 165, 1095. [Google Scholar] [CrossRef] [PubMed]
  91. Hurren, K.M.; Lewis, C.L. Probable Interaction between Warfarin and Bee Pollen. Am. J. Health Syst. Pharm. 2010, 67, 2034–2037. [Google Scholar] [CrossRef] [PubMed]
  92. Luo, H.; Geng, J.; Zeiler, M.; Nieckula, E.; Sandalinas, F.; Williams, A.; Young, M.F.; Suchdev, P.S. A Practical Guide to Adjust Micronutrient Biomarkers for Inflammation Using the BRINDA Method. J. Nutr. 2023, 153, 1265–1272. [Google Scholar] [CrossRef] [PubMed]
  93. Morris, A.L.; Mohiuddin, S.S. Biochemistry, Nutrients. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  94. McCormick, D.B. Micronutrient Cofactor Research with Extensions to Applications. Nutr. Res. Rev. 2002, 15, 245–262. [Google Scholar] [CrossRef] [PubMed]
  95. Biesalski Hans, K.; Jana, T. Micronutrients in the Life Cycle: Requirements and Sufficient Supply. NFS J. 2018, 11, 1–11. [Google Scholar] [CrossRef]
  96. Hrubša, M.; Siatka, T.; Nejmanová, I.; Vopršalová, M.; Kujovská Krčmová, L.; Matoušová, K.; Javorská, L.; Macáková, K.; Mercolini, L.; Remião, F.; et al. Biological Properties of Vitamins of the B-Complex, Part 1: Vitamins B1, B2, B3, and B5. Nutrients 2022, 14, 484. [Google Scholar] [CrossRef] [PubMed]
  97. Lonsdale, D. Thiamin(e): The Spark of Life. In Water Soluble Vitamins; Stanger, O., Ed.; Subcellular Biochemistry; Springer: Dordrecht, The Netherlands, 2012; Volume 56, pp. 199–227. [Google Scholar] [CrossRef]
  98. Marrs, C.; Lonsdale, D. Hiding in Plain Sight: Modern Thiamine Deficiency. Cells 2021, 10, 2595. [Google Scholar] [CrossRef] [PubMed]
  99. Thiamine Deficiency Disease, Dysautonomia, and High Calorie Malnutrition. ScienceDirect. Available online: http://www.sciencedirect.com:5070/book/9780128103876/thiamine-deficiency-disease-dysautonomia-and-high-calorie-malnutrition (accessed on 4 October 2023).
  100. Lienhart, W.-D.; Gudipati, V.; Macheroux, P. The Human Flavoproteome. Arch. Biochem. Biophys. 2013, 535, 150–162. [Google Scholar] [CrossRef]
  101. Balasubramaniam, S.; Christodoulou, J.; Rahman, S. Disorders of Riboflavin Metabolism. J. Inherit. Metab. Dis. 2019, 42, 608–619. [Google Scholar] [CrossRef] [PubMed]
  102. Hellmann, H.; Mooney, S. Vitamin B6: A Molecule for Human Health? Molecules 2010, 15, 442–459. [Google Scholar] [CrossRef] [PubMed]
  103. Zempleni, J.; Wijeratne, S.S.K.; Hassan, Y.I. Biotin. BioFactors 2009, 35, 36–46. [Google Scholar] [CrossRef] [PubMed]
  104. Ebara, S. Nutritional Role of Folate. Congenit. Anom. 2017, 57, 138–141. [Google Scholar] [CrossRef] [PubMed]
  105. Shane, B. Folate and Vitamin B12 Metabolism: Overview and Interaction with Riboflavin, Vitamin B6, and Polymorphisms. Food Nutr. Bull. 2008, 29 (Suppl. S2), S5–S16. [Google Scholar] [CrossRef]
  106. O’Leary, F.; Samman, S. Vitamin B12 in Health and Disease. Nutrients 2010, 2, 299–316. [Google Scholar] [CrossRef] [PubMed]
  107. Chambial, S.; Dwivedi, S.; Shukla, K.K.; John, P.J.; Sharma, P. Vitamin C in Disease Prevention and Cure: An Overview. Indian J. Clin. Biochem. 2013, 28, 314–328. [Google Scholar] [CrossRef]
  108. Carr, A.; Maggini, S. Vitamin C and Immune Function. Nutrients 2017, 9, 1211. [Google Scholar] [CrossRef] [PubMed]
  109. Naidu, K.A. Vitamin C in Human Health and Disease Is Still a Mystery? An Overview. Nutr. J. 2003, 2, 7. [Google Scholar] [CrossRef] [PubMed]
  110. Peh, H.Y.; Tan, W.S.D.; Liao, W.; Wong, W.S.F. Vitamin E Therapy beyond Cancer: Tocopherol versus Tocotrienol. Pharmacol. Ther. 2016, 162, 152–169. [Google Scholar] [CrossRef]
  111. Sen, C.K.; Khanna, S.; Roy, S. Tocotrienols: Vitamin E beyond Tocopherols. Life Sci. 2006, 78, 2088–2098. [Google Scholar] [CrossRef] [PubMed]
  112. Sen, C.K.; Khanna, S.; Rink, C.; Roy, S. Tocotrienols: The Emerging Face of Natural Vitamin E. In Vitamins & Hormones; Elsevier: Amsterdam, The Netherlands, 2007; Volume 76, pp. 203–261. [Google Scholar] [CrossRef]
  113. Jackson, M.J. Diagnosis and Detection of Deficiencies of Micronutrients: Minerals. Br. Med. Bull. 1999, 55, 634–642. [Google Scholar] [CrossRef] [PubMed]
  114. Peacock, M. Calcium Metabolism in Health and Disease. Clin. J. Am. Soc. Nephrol. 2010, 5 (Suppl. S1), S23–S30. [Google Scholar] [CrossRef]
  115. Peacock, M. Phosphate Metabolism in Health and Disease. Calcif. Tissue Int. 2021, 108, 3–15. [Google Scholar] [CrossRef]
  116. Swaminathan, R. Magnesium Metabolism and Its Disorders. Clin. Biochem. Rev. 2003, 24, 47–66. [Google Scholar] [PubMed]
  117. Strazzullo, P.; Leclercq, C. Sodium. Adv. Nutr. 2014, 5, 188–190. [Google Scholar] [CrossRef] [PubMed]
  118. Weaver, C.M. Potassium and Health. Adv. Nutr. 2013, 4, 368S–377S. [Google Scholar] [CrossRef] [PubMed]
  119. Berend, K.; Van Hulsteijn, L.H.; Gans, R.O.B. Chloride: The Queen of Electrolytes? Eur. J. Intern. Med. 2012, 23, 203–211. [Google Scholar] [CrossRef] [PubMed]
  120. Dev, S.; Babitt, J.L. Overview of Iron Metabolism in Health and Disease. Hemodial. Int. 2017, 21 (Suppl. S1), S6–S20. [Google Scholar] [CrossRef] [PubMed]
  121. Abbaspour, N.; Hurrell, R.; Kelishadi, R. Review on Iron and Its Importance for Human Health. J. Res. Med. Sci. Off. J. Isfahan Univ. Med. Sci. 2014, 19, 164–174. [Google Scholar]
  122. Uriu-Adams, J.Y.; Keen, C.L. Copper, Oxidative Stress, and Human Health. Mol. Aspects Med. 2005, 26, 268–298. [Google Scholar] [CrossRef] [PubMed]
  123. DiNicolantonio, J.J.; Mangan, D.; O’Keefe, J.H. Copper Deficiency May Be a Leading Cause of Ischaemic Heart Disease. Open Heart 2018, 5, e000784. [Google Scholar] [CrossRef] [PubMed]
  124. Wapnir, R.A. Copper Absorption and Bioavailability. Am. J. Clin. Nutr. 1998, 67, 1054S–1060S. [Google Scholar] [CrossRef] [PubMed]
  125. Zheng, L.; Han, P.; Liu, J.; Li, R.; Yin, W.; Wang, T.; Zhang, W.; James Kang, Y. Role of Copper in Regression of Cardiac Hypertrophy. Pharmacol. Ther. 2015, 148, 66–84. [Google Scholar] [CrossRef] [PubMed]
  126. Chasapis, C.T.; Loutsidou, A.C.; Spiliopoulou, C.A.; Stefanidou, M.E. Zinc and Human Health: An Update. Arch. Toxicol. 2012, 86, 521–534. [Google Scholar] [CrossRef]
  127. Maret, W.; Sandstead, H.H. Zinc Requirements and the Risks and Benefits of Zinc Supplementation. J. Trace Elem. Med. Biol. 2006, 20, 3–18. [Google Scholar] [CrossRef] [PubMed]
  128. Brown, K.; Arthur, J. Selenium, Selenoproteins and Human Health: A Review. Public Health Nutr. 2001, 4, 593–599. [Google Scholar] [CrossRef] [PubMed]
  129. Rayman, M.P. The Importance of Selenium to Human Health. Lancet 2000, 356, 233–241. [Google Scholar] [CrossRef] [PubMed]
  130. Hatch-McChesney, A.; Lieberman, H.R. Iodine and Iodine Deficiency: A Comprehensive Review of a Re-Emerging Issue. Nutrients 2022, 14, 3474. [Google Scholar] [CrossRef] [PubMed]
  131. Spence, J.D.; Yi, Q.; Hankey, G.J.B. Vitamins in Stroke Prevention: Time to Reconsider. Lancet Neurol. 2017, 16, 750–760. [Google Scholar] [CrossRef] [PubMed]
  132. Institute of Medicine, Food and Nutrition Board; Subcommittee on Upper Reference Levels of Nutrients; Standing Committee on the Scientific Evaluation of Dietary Reference Intakes and its Panel on Folate, Other B Vitamins, and Choline. Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and Choline; National Academies Press: Washington, DC, USA, 1998; p. 6015. [Google Scholar] [CrossRef]
  133. Panel on Micronutrients, Food and Nutrition Board. Dietary Reference Intakes for Vitamin A, Vitamin K, Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc; National Academies Press: Washington, DC, USA, 2001; p. 10026. [Google Scholar] [CrossRef]
  134. Panel on Dietary Antioxidants and Related Compounds; Subcommittee on Upper Reference Levels of Nutrients; Subcommittee on Interpretation and Uses of Dietary Reference Intakes; Standing Committee on the Scientific Evaluation of Dietary Reference Intakes; Food and Nutrition Board; Institute of Medicine. Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids; National Academies Press: Washington, DC, USA, 2000; p. 9810. [Google Scholar] [CrossRef]
  135. Ames, B.N. Prolonging Healthy Aging: Longevity Vitamins and Proteins. Proc. Natl. Acad. Sci. USA 2018, 115, 10836–10844. [Google Scholar] [CrossRef] [PubMed]
  136. Huxtable, R.J. Physiological Actions of Taurine. Physiol. Rev. 1992, 72, 101–163. [Google Scholar] [CrossRef] [PubMed]
  137. Jong, C.J.; Azuma, J.; Schaffer, S. Mechanism Underlying the Antioxidant Activity of Taurine: Prevention of Mitochondrial Oxidant Production. Amino Acids 2012, 42, 2223–2232. [Google Scholar] [CrossRef] [PubMed]
  138. Hansen, S.; Andersen, M.; Cornett, C.; Gradinaru, R.; Grunnet, N. A Role for Taurine in Mitochondrial Function. J. Biomed. Sci. 2010, 17 (Suppl. S1), S23. [Google Scholar] [CrossRef] [PubMed]
  139. Melville, D.B. Ergothioneine. In Vitamins & Hormones; Elsevier: Amsterdam, The Netherlands, 1959; Volume 17, pp. 155–204. [Google Scholar] [CrossRef]
  140. Cheah, I.K.; Tang, R.M.Y.; Yew, T.S.Z.; Lim, K.H.C.; Halliwell, B. Administration of Pure Ergothioneine to Healthy Human Subjects: Uptake, Metabolism, and Effects on Biomarkers of Oxidative Damage and Inflammation. Antioxid. Redox Signal. 2017, 26, 193–206. [Google Scholar] [CrossRef] [PubMed]
  141. Cheah, I.K.; Halliwell, B. Ergothioneine, Recent Developments. Redox Biol. 2021, 42, 101868. [Google Scholar] [CrossRef] [PubMed]
  142. Jonscher, K.R.; Chowanadisai, W.; Rucker, R.B. Pyrroloquinoline-Quinone Is More Than an Antioxidant: A Vitamin-like Accessory Factor Important in Health and Disease Prevention. Biomolecules 2021, 11, 1441. [Google Scholar] [CrossRef] [PubMed]
  143. Harris, C.B.; Chowanadisai, W.; Mishchuk, D.O.; Satre, M.A.; Slupsky, C.M.; Rucker, R.B. Dietary Pyrroloquinoline Quinone (PQQ) Alters Indicators of Inflammation and Mitochondrial-Related Metabolism in Human Subjects. J. Nutr. Biochem. 2013, 24, 2076–2084. [Google Scholar] [CrossRef] [PubMed]
  144. Fergus, C.; Barnes, D.; Alqasem, M.; Kelly, V. The Queuine Micronutrient: Charting a Course from Microbe to Man. Nutrients 2015, 7, 2897–2929. [Google Scholar] [CrossRef] [PubMed]
  145. Rodriguez-Concepcion, M.; Avalos, J.; Bonet, M.L.; Boronat, A.; Gomez-Gomez, L.; Hornero-Mendez, D.; Limon, M.C.; Meléndez-Martínez, A.J.; Olmedilla-Alonso, B.; Palou, A.; et al. A Global Perspective on Carotenoids: Metabolism, Biotechnology, and Benefits for Nutrition and Health. Prog. Lipid Res. 2018, 70, 62–93. [Google Scholar] [CrossRef] [PubMed]
  146. Przybylska, S. Lycopene—A Bioactive Carotenoid Offering Multiple Health Benefits: A Review. Int. J. Food Sci. Technol. 2020, 55, 11–32. [Google Scholar] [CrossRef]
  147. Burri, B.J.; La Frano, M.R.; Zhu, C. Absorption, Metabolism, and Functions of β-Cryptoxanthin. Nutr. Rev. 2016, 74, 69–82. [Google Scholar] [CrossRef] [PubMed]
  148. Burri, B.J. Beta-Cryptoxanthin as a Source of Vitamin A: Beta-Cryptoxanthin as a Source of Vitamin A. J. Sci. Food Agric. 2015, 95, 1786–1794. [Google Scholar] [CrossRef] [PubMed]
  149. Higuera-Ciapara, I.; Félix-Valenzuela, L.; Goycoolea, F.M. Astaxanthin: A Review of Its Chemistry and Applications. Crit. Rev. Food Sci. Nutr. 2006, 46, 185–196. [Google Scholar] [CrossRef] [PubMed]
  150. Donoso, A.; González-Durán, J.; Muñoz, A.A.; González, P.A.; Agurto-Muñoz, C. Therapeutic Uses of Natural Astaxanthin: An Evidence-Based Review Focused on Human Clinical Trials. Pharmacol. Res. 2021, 166, 105479. [Google Scholar] [CrossRef] [PubMed]
  151. Zeisel, S.H.; Da Costa, K.-A. Choline: An Essential Nutrient for Public Health. Nutr. Rev. 2009, 67, 615–623. [Google Scholar] [CrossRef]
  152. Wallace, T.C.; Blusztajn, J.K.; Caudill, M.A.; Klatt, K.C.; Natker, E.; Zeisel, S.H.; Zelman, K.M. Choline: The Underconsumed and Underappreciated Essential Nutrient. Nutr. Today 2018, 53, 240–253. [Google Scholar] [CrossRef]
  153. Kumar, N.; Goel, N. Phenolic Acids: Natural Versatile Molecules with Promising Therapeutic Applications. Biotechnol. Rep. 2019, 24, e00370. [Google Scholar] [CrossRef] [PubMed]
  154. Das, U. Essential Fatty Acids—A Review. Curr. Pharm. Biotechnol. 2006, 7, 467–482. [Google Scholar] [CrossRef] [PubMed]
  155. Das, U.N. Essential Fatty Acids: Biochemistry, Physiology and Pathology. Biotechnol. J. 2006, 1, 420–439. [Google Scholar] [CrossRef] [PubMed]
  156. Cholewski, M.; Tomczykowa, M.; Tomczyk, M. A Comprehensive Review of Chemistry, Sources and Bioavailability of Omega-3 Fatty Acids. Nutrients 2018, 10, 1662. [Google Scholar] [CrossRef]
  157. Wu, G. Amino Acids: Metabolism, Functions, and Nutrition. Amino Acids 2009, 37, 1–17. [Google Scholar] [CrossRef] [PubMed]
  158. Babygirija, R.; Lamming, D.W. The Regulation of Healthspan and Lifespan by Dietary Amino Acids. Transl. Med. Aging 2021, 5, 17–30. [Google Scholar] [CrossRef] [PubMed]
  159. Hou, Y.; Wu, G. Nutritionally Nonessential Amino Acids: A Misnomer in Nutritional Sciences. Adv. Nutr. 2017, 8, 137–139. [Google Scholar] [CrossRef] [PubMed]
  160. Zhu, W.; Wang, Z.; Tang, W.H.W.; Hazen, S.L. Gut Microbe-Generated Trimethylamine N-Oxide from Dietary Choline Is Prothrombotic in Subjects. Circulation 2017, 135, 1671–1673. [Google Scholar] [CrossRef] [PubMed]
  161. Kyselova, Z. Toxicological Aspects of the Use of Phenolic Compounds in Disease Prevention. Interdiscip. Toxicol. 2011, 4, 173–183. [Google Scholar] [CrossRef]
  162. Cleland, J.B. The Effect of Salicylates (A) on the estimation of thiamine by the thiochrome method, (B) on the excretion of thiamine. Aust. J. Exp. Biol. Med. Sci. 1943, 21, 153–158. [Google Scholar] [CrossRef]
  163. Cleland, J.B. The effect of salicylates and thiamine deficiency on the thiamine content of rat tissue. Aust. J. Exp. Biol. Med. Sci. 1946, 24, 227–230. [Google Scholar] [CrossRef]
  164. Mehlman, M.A.; Tobin, R.B.; Madappally, M.M.; Hahn, H.K.J. Mode of Action of Aspirin. J. Biol. Chem. 1971, 246, 1618–1622. [Google Scholar] [CrossRef]
  165. Maggioni, G.F. Salicylate Therapy in Children. Arch. Dis. Child. 1948, 23, 40–47. [Google Scholar] [CrossRef]
  166. Phillips, W. Is cutaneous flushing prostaglandin mediated? Lancet 1981, 317, 754–756. [Google Scholar] [CrossRef]
  167. Parson, H.K.; Harati, H.; Cooper, D.; Vinik, A.I. Role of Prostaglandin D2 and the Autonomic Nervous System in Niacin-Induced Flushing: PGD2 and ANS in Niacin-Induced Flushing. J. Diabetes 2013, 5, 59–67. [Google Scholar] [CrossRef] [PubMed]
  168. Cheng, K.; Wu, T.-J.; Wu, K.K.; Sturino, C.; Metters, K.; Gottesdiener, K.; Wright, S.D.; Wang, Z.; O’Neill, G.; Lai, E.; et al. Antagonism of the Prostaglandin D2 Receptor 1 Suppresses Nicotinic Acid-Induced Vasodilation in Mice and Humans. Proc. Natl. Acad. Sci. USA 2006, 103, 6682–6687. [Google Scholar] [CrossRef] [PubMed]
  169. Jiang, P.; Parreno, R.; Thakkar, R.B.; Krause, S.; Doan, T.T.; Padley, R.J. Abstract P59: Aspirin Improves the Impact of Niacin-Associated Flushing on Quality of Life Measures. Circ. Cardiovasc. Qual. Outcomes 2011, 4 (Suppl. S1), AP59. [Google Scholar] [CrossRef]
  170. Offermanns, S. Heating Up the Cutaneous Flushing Response. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1122–1123. [Google Scholar] [CrossRef] [PubMed]
  171. Kamanna, V.S.; Ganji, S.H.; Kashyap, M.L. The Mechanism and Mitigation of Niacin-Induced Flushing. Int. J. Clin. Pract. 2009, 63, 1369–1377. [Google Scholar] [CrossRef] [PubMed]
  172. Whelan, A.M.; Price, S.O.; Fowler, S.F.; Hainer, B.L. The Effect of Aspirin on Niacin-Induced Cutaneous Reactions. J. Fam. Pract. 1992, 34, 165–168. [Google Scholar]
  173. Jungnickel, P.W.; Maloley, P.A.; Tuin, E.L.; Peddicord, T.E.; Campbell, J.R. Effect of Two Aspirin Pretreatment Regimens on Niacin-Induced Cutaneous Reactions. J. Gen. Intern. Med. 1997, 12, 591–596. [Google Scholar] [CrossRef] [PubMed]
  174. Omer, A.; Mowat, A.G. Nature of Anaemia in Rheumatoid Arthritis. IX. Folate Metabolism in Patients with Rheumatoid Arthritis. Ann. Rheum. Dis. 1968, 27, 414–424. [Google Scholar] [CrossRef] [PubMed]
  175. Deller, D.J.; Urban, E.; Ibbotson, R.N.; Horwood, J.; Milazzo, S.; Robson, H.N. Folic-Acid Deficiency in Rheumatoid Arthritis: Relation of Levels of Serum Folic-Acid Activity of Treatment with Phenylbutazone. BMJ 1966, 1, 765–767. [Google Scholar] [CrossRef] [PubMed]
  176. Gough, K.R.; McCarthy, C.; Read, A.E.; Mollin, D.L.; Waters, A.H. Folic-Acid Deficiency in Rheumatoid Arthritis. BMJ 1964, 1, 212–217. [Google Scholar] [CrossRef] [PubMed]
  177. Lawrence, V.A.; Loewenstein, J.E.; Eichner, E.R. Aspirin and Folate Binding: In Vivo and in Vitro Studies of Serum Binding and Urinary Excretion of Endogenous Folate. J. Lab. Clin. Med. 1984, 103, 944–948. [Google Scholar]
  178. Alonso-Aperte, E.; Varela-Moreiras, G. Drugs-Nutrient Interactions: A Potential Problem during Adolescence. Eur. J. Clin. Nutr. 2000, 54 (Suppl. S1), S69–S74. [Google Scholar] [CrossRef] [PubMed]
  179. Alter, H.J.; Zvaifler, N.J.; Rath, C.E. Interrelationship of Rheumatoid Arthritis, Folic Acid, and Aspirin. Blood 1971, 38, 405–416. [Google Scholar] [CrossRef] [PubMed]
  180. Baggott, J.E.; Morgan, S.L.; Ha, T.; Vaughn, W.H.; Hine, R.J. Inhibition of Folate-Dependent Enzymes by Non-Steroidal Anti-Inflammatory Drugs. Biochem. J. 1992, 282, 197–202. [Google Scholar] [CrossRef] [PubMed]
  181. Ha, T.; Morgan, S.L.; Vaughn, W.H.; Eto, I.; Baggott, J.E. Detection of Inhibition of 5-Aminoimidazole-4-Carboxamide Ribotide Transformylase by Thioinosinic Acid and Azathioprine by a New Colorimetric Assay. Biochem. J. 1990, 272, 339–342. [Google Scholar] [CrossRef] [PubMed]
  182. Ho, G.Y.F.; Xue, X.; Cushman, M.; McKeown-Eyssen, G.; Sandler, R.S.; Ahnen, D.J.; Barry, E.L.; Saibil, F.; Bresalier, R.S.; Rohan, T.E.; et al. Antagonistic Effects of Aspirin and Folic Acid on Inflammation Markers and Subsequent Risk of Recurrent Colorectal Adenomas. JNCI J. Natl. Cancer Inst. 2009, 101, 1650–1654. [Google Scholar] [CrossRef] [PubMed]
  183. McCarty, M.F. High-Dose Folate May Improve Platelet Function in Acute Coronary Syndrome and Other Pathologies Associated with Increased Platelet Oxidative Stress. Med. Hypotheses 2007, 69, 12–19. [Google Scholar] [CrossRef] [PubMed]
  184. Van Oijen, M.G.H.; Laheij, R.J.F.; Peters, W.H.M.; Jansen, J.B.M.J.; Verheugt, F.W.A. Association of Aspirin Use with Vitamin B12 Deficiency (Results of the BACH Study). Am. J. Cardiol. 2004, 94, 975–977. [Google Scholar] [CrossRef] [PubMed]
  185. Hunt, A.; Harrington, D.; Robinson, S. Vitamin B12 Deficiency. BMJ 2014, 349 (Suppl. S1), g5226. [Google Scholar] [CrossRef] [PubMed]
  186. Carabotti, M.; Annibale, B.; Lahner, E. Common Pitfalls in the Management of Patients with Micronutrient Deficiency: Keep in Mind the Stomach. Nutrients 2021, 13, 208. [Google Scholar] [CrossRef] [PubMed]
  187. Carmel, R. Malabsorption of Food Cobalamin. Baillieres Clin. Haematol. 1995, 8, 639–655. [Google Scholar] [CrossRef] [PubMed]
  188. Andres, E. Vitamin B12 (Cobalamin) Deficiency in Elderly Patients. Can. Med. Assoc. J. 2004, 171, 251–259. [Google Scholar] [CrossRef] [PubMed]
  189. Annibale, B.; Capurso, G.; Delle Fave, G. Consequences of Helicobacter Pylori Infection on the Absorption of Micronutrients. Dig. Liver Dis. 2002, 34, S72–S77. [Google Scholar] [CrossRef] [PubMed]
  190. Wu, Y.; Smith, A.D.; Refsum, H.; Kwok, T. Effectiveness of B Vitamins and Their Interactions with Aspirin in Improving Cognitive Functioning in Older People with Mild Cognitive Impairment: Pooled Post-Hoc Analyses of Two Randomized Trials. J. Nutr. Health Aging 2021, 25, 1154–1160. [Google Scholar] [CrossRef] [PubMed]
  191. Zhang, Q.; Zhu, Z.; Ni, Y. Interaction between Aspirin and Vitamin C with Human Serum Albumin as Binary and Ternary Systems. Spectrochim. Acta. A Mol. Biomol. Spectrosc. 2020, 236, 118356. [Google Scholar] [CrossRef] [PubMed]
  192. Basu, T.K. Vitamin C-Aspirin Interactions. Int. J. Vitam. Nutr. Res. Suppl. Int. Z. Vitam.-Ernahrungsforschung Suppl. 1982, 23, 83–90. [Google Scholar]
  193. Hollander, D.; Dadufalza, V.D.; Fairchild, P.A. Intestinal Absorption of Aspirin. Influence of pH, Taurocholate, Ascorbate, and Ethanol. J. Lab. Clin. Med. 1981, 98, 591–598. [Google Scholar] [PubMed]
  194. Nelson, E.W.; Lane, H.; Fabri, P.J.; Scott, B. Demonstration of Saturation Kinetics in the Intestinal Absorption of Vitamin C in Man and the Guinea Pig. J. Clin. Pharmacol. 1978, 18, 325–335. [Google Scholar] [CrossRef]
  195. Sahud, M. Effect of aspirin ingestion on ascorbic-acid levels in rheumatoid arthritis. Lancet 1971, 297, 937–938. [Google Scholar] [CrossRef]
  196. Schulz, H.-U.; Schürer, M.; Krupp, S.; Dammann, H.-G.; Timm, J.; Gessner, U. Effects of Acetylsalicylic Acid on Ascorbic Acid Concentrations in Plasma, Gastric Mucosa, Gastric Juice and Urine a Double-Blind Study in Healthy Subjects. Int. J. Clin. Pharmacol. Ther. 2004, 42, 481–487. [Google Scholar] [CrossRef] [PubMed]
  197. Konturek, P.C.; Kania, J.; Hahn, E.G.; Konturek, J.W. Ascorbic Acid Attenuates Aspirin-Induced Gastric Damage: Role of Inducible Nitric Oxide Synthase. J. Physiol. Pharmacol. Off. J. Pol. Physiol. Soc. 2006, 57 (Suppl. S5), 125–136. [Google Scholar]
  198. Pohle, T.; Brzozowski, T.; Becker, J.C.; Van der Voort, I.R.; Markmann, A.; Konturek, S.J.; Moniczewski, A.; Domschke, W.; Konturek, J.W. Role of Reactive Oxygen Metabolites in Aspirin-Induced Gastric Damage in Humans: Gastroprotection by Vitamin C. Aliment. Pharmacol. Ther. 2001, 15, 677–687. [Google Scholar] [CrossRef] [PubMed]
  199. Daş, N.; Nebioğlu, S. Vitamin C Aspirin Interactions in Laboratory Animals. J. Clin. Pharm. Ther. 1992, 17, 343–346. [Google Scholar] [CrossRef] [PubMed]
  200. Spitzer, J.M.; Shapiro, S. The Effect of Salicylate Medication upon the Urinary Excretion of Vitamin C. Am. J. Dig. Dis. 1948, 15, 80–84. [Google Scholar] [CrossRef] [PubMed]
  201. Daniels, A.L.; Everson, G.J. Influence of Acetylsalicylic Acid (Aspirin) on Urinary Excretion of Ascorbic Acid. Exp. Biol. Med. 1936, 35, 20–24. [Google Scholar] [CrossRef]
  202. Loh, H.S.; Watters, K.; Wilson, C.W.M. The Effects of Aspirin on the Metabolic Availability of Ascorbic Acid in Human Beings. J. Clin. Pharmacol. New Drugs 1973, 13, 480–486. [Google Scholar] [CrossRef] [PubMed]
  203. Oiness, K.N. Nutritional Consequences of Drugs Used in Pediatrics. Clin. Pediatr. 1985, 24, 417–420. [Google Scholar] [CrossRef] [PubMed]
  204. Candelario-Jalil, E.; Akundi, R.S.; Bhatia, H.S.; Lieb, K.; Appel, K.; Muñoz, E.; Hüll, M.; Fiebich, B.L. Ascorbic Acid Enhances the Inhibitory Effect of Aspirin on Neuronal Cyclooxygenase-2-Mediated Prostaglandin E2 Production. J. Neuroimmunol. 2006, 174, 39–51. [Google Scholar] [CrossRef] [PubMed]
  205. Han, S.-S.; Kim, K.; Hahm, E.-R.; Lee, S.J.; Surh, Y.-J.; Park, H.K.; Kim, W.S.; Jung, C.W.; Lee, M.H.; Park, K.; et al. L-Ascorbic Acid Represses Constitutive Activation of NF-kappaB and COX-2 Expression in Human Acute Myeloid Leukemia, HL-60. J. Cell. Biochem. 2004, 93, 257–270. [Google Scholar] [CrossRef]
  206. Sánchez-Moreno, C.; Paniagua, M.; Madrid, A.; Martín, A. Protective Effect of Vitamin C against the Ethanol Mediated Toxic Effects on Human Brain Glial Cells. J. Nutr. Biochem. 2003, 14, 606–613. [Google Scholar] [CrossRef] [PubMed]
  207. Freedman, J.E.; Farhat, J.H.; Loscalzo, J.; Keaney, J.F. α-Tocopherol Inhibits Aggregation of Human Platelets by a Protein Kinase C–Dependent Mechanism. Circulation 1996, 94, 2434–2440. [Google Scholar] [CrossRef] [PubMed]
  208. Steiner, M.; Anastasi, J. Vitamin E. An Inhibitor of the Platelet Release Reaction. J. Clin. Investig. 1976, 57, 732–737. [Google Scholar] [CrossRef] [PubMed]
  209. Agradi, E.; Petroni, A.; Socini, A.; Galli, C. In Vitro Effects of Synthetic Antioxidants and Vitamin E on Arachidonic Acid Metabolism and Thromboxane Formation in Human Platelets and on Platelet Aggregation. Prostaglandins 1981, 22, 255–266. [Google Scholar] [CrossRef] [PubMed]
  210. González-Correa, J.A.; Arrebola, M.M.; Guerrero, A.; Cañada, M.J.; Muñoz Marín, J.; De La Cuesta, F.S.; De La Cruz, J.P. Antioxidant and Antiplatelet Effects of the Alpha-Tocopherol–Aspirin Combination in Type 1-like Diabetic Rats. Life Sci. 2006, 79, 1405–1412. [Google Scholar] [CrossRef]
  211. Vatassery, G.; Morley, J.; Kuskowski, M. Vitamin E in Plasma and Platelets of Human Diabetic Patients and Control Subjects. Am. J. Clin. Nutr. 1983, 37, 641–644. [Google Scholar] [CrossRef] [PubMed]
  212. Liede, K.E.; Haukka, J.K.; Saxén, L.M.; Heinonen, O.P. Increased Tendency towards Gingival Bleeding Caused by Joint Effect of α-Tocopherol Supplementation and Acetylsalicylic Acid. Ann. Med. 1998, 30, 542–546. [Google Scholar] [CrossRef] [PubMed]
  213. Steiner, M.; Glantz, M.; Lekos, A. Vitamin E plus Aspirin Compared with Aspirin Alone in Patients with Transient Ischemic Attacks. Am. J. Clin. Nutr. 1995, 62, 1381S–1384S. [Google Scholar] [CrossRef] [PubMed]
  214. Liu, M.; Wallmon, A.; Olsson-Mortlock, C.; Wallin, R.; Saldeen, T. Mixed Tocopherols Inhibit Platelet Aggregation in Humans: Potential Mechanisms123. Am. J. Clin. Nutr. 2003, 77, 700–706. [Google Scholar] [CrossRef] [PubMed]
  215. Podszun, M.; Frank, J. Vitamin E—Drug Interactions: Molecular Basis and Clinical Relevance. Nutr. Res. Rev. 2014, 27, 215–231. [Google Scholar] [CrossRef] [PubMed]
  216. Sugimoto, N.; Yoshida, N.; Yoshikawa, T.; Nakamuara, Y.; Ichikawa, H.; Naito, Y.; Kondo, M. Effect of Vitamin E on Aspirin-Induced Gastric Mucosal Injury in Rats. Dig. Dis. Sci. 2000, 45, 599–605. [Google Scholar] [CrossRef] [PubMed]
  217. Nafeeza, M.I.; Fauzee, A.M.; Kamsiah, J.; Gapor, M.T. Comparative Effects of a Tocotrienol-Rich Fraction and Tocopherol in Aspirin-Induced Gastric Lesions in Rats. Asia Pac. J. Clin. Nutr. 2002, 11, 309–313. [Google Scholar] [CrossRef] [PubMed]
  218. Jiang, Q.; Moreland, M.; Ames, B.N.; Yin, X. A Combination of Aspirin and γ-Tocopherol Is Superior to That of Aspirin and α-Tocopherol in Anti-Inflammatory Action and Attenuation of Aspirin-Induced Adverse Effects. J. Nutr. Biochem. 2009, 20, 894–900. [Google Scholar] [CrossRef] [PubMed]
  219. Susic, D.; Sparks, J.C. Effects of Aspirin on Renal Sodium Excretion, Blood Pressure, and Plasma and Extracellular Fluid Volume in Salt-Loaded Rats. Prostaglandins 1975, 10, 825–831. [Google Scholar] [CrossRef] [PubMed]
  220. Kadokawa, T.; Hosoki, K.; Takeyama, K.; Minato, H.; Shimizu, M. Effects of Nonsteroidal Anti-Inflammatory Drugs (NSAID) on Renal Excretion of Sodium and Water, and on Body Fluid Volume in Rats. J. Pharmacol. Exp. Ther. 1979, 209, 219–224. [Google Scholar] [PubMed]
  221. Costa, A.C.; Reina-Couto, M.; Albino-Teixeira, A.; Sousa, T. Aspirin and Blood Pressure: Effects When Used Alone or in Combination with Antihypertensive Drugs. Rev. Port. Cardiol. 2017, 36, 551–567. [Google Scholar] [CrossRef] [PubMed]
  222. Juhlin, T.; Jönsson, B.A.G.; Höglund, P. Renal Effects of Aspirin Are Clearly Dose-Dependent and Are of Clinical Importance from a Dose of 160 Mg. Eur. J. Heart Fail. 2008, 10, 892–898. [Google Scholar] [CrossRef] [PubMed]
  223. Mahmood, S.S.; Levy, D.; Vasan, R.S.; Wang, T.J. The Framingham Heart Study and the Epidemiology of Cardiovascular Disease: A Historical Perspective. Lancet 2014, 383, 999–1008. [Google Scholar] [CrossRef]
  224. Fleming, D.J.; Jacques, P.F.; Massaro, J.M.; D’Agostino, R.B., Sr.; Wilson, P.W.; Wood, R.J. Aspirin Intake and the Use of Serum Ferritin as a Measure of Iron Status. Am. J. Clin. Nutr. 2001, 74, 219–226. [Google Scholar] [CrossRef] [PubMed]
  225. Milman, N.; Ovesen, L.; Byg, K.-E.; Graudal, N. Iron Status in Danes Updated 1994. I: Prevalence of Iron Deficiency and Iron Overload in 1332 Men Aged 40–70 Years. Influence of Blood Donation, Alcohol Intake, and Iron Supplementation. Ann. Hematol. 1999, 78, 393–400. [Google Scholar] [CrossRef] [PubMed]
  226. McQuilten, Z.K.; Thao, L.T.P.; Pasricha, S.-R.; Artz, A.S.; Bailey, M.; Chan, A.T.; Cohen, H.J.; Lockery, J.E.; Murray, A.M.; Nelson, M.R.; et al. Effect of Low-Dose Aspirin Versus Placebo on Incidence of Anemia in the Elderly: A Secondary Analysis of the Aspirin in Reducing Events in the Elderly Trial. Ann. Intern. Med. 2023, 176, 913–921. [Google Scholar] [CrossRef] [PubMed]
  227. Gaskell, H.; Derry, S.; Moore, R.A. Is There an Association between Low Dose Aspirin and Anemia (without Overt Bleeding)?: Narrative Review. BMC Geriatr. 2010, 10, 71. [Google Scholar] [CrossRef] [PubMed]
  228. Mascitelli, L.; Goldstein, M.R. Aspirin-Associated Iron Loss: An Anticancer Mechanism Even in the Short Term? Am. J. Med. 2013, 126, e11. [Google Scholar] [CrossRef] [PubMed]
  229. Martell, A.E.; Smith, R.M. Other Organic Ligands; Springer: Boston, MA, USA, 1977. [Google Scholar] [CrossRef]
  230. Davis, M.L.; Seaborn, C.D.; Stoecker, B.J. Effects of Over-the-Counter Drugs on 51chromium Retention and Urinary Excretion in Rats. Nutr. Res. 1995, 15, 201–210. [Google Scholar] [CrossRef]
  231. Franconi, F.; Miceli, M.; Bennardini, F.; Mattana, A.; Covarrubias, J.; Seghieri, G. Taurine Potentiates the Antiaggregatory Action of Aspirin and Indomethacin. In Taurine; Lombardini, J.B., Schaffer, S.W., Azuma, J., Eds.; Advances in Experimental Medicine and Biology; Springer: Boston, MA, USA, 1992; Volume 315, pp. 181–186. [Google Scholar] [CrossRef]
  232. Kurachi, M.; Hongoh, K.; Watanabe, A.; Aihara, H. Suppression of Bronchial Response to Platelet Activating Factor Following Taurine Administration. In The Biology of Taurine; Huxtable, R.J., Franconi, F., Giotti, A., Eds.; Advances in Experimental Medicine and Biology; Springer: Boston, MA, USA, 1987; Volume 217, pp. 189–198. [Google Scholar] [CrossRef]
  233. Roşca, A.E.; Vlădăreanu, A.-M.; Mirica, R.; Anghel-Timaru, C.-M.; Mititelu, A.; Popescu, B.O.; Căruntu, C.; Voiculescu, S.E.; Gologan, Ş.; Onisâi, M.; et al. Taurine and Its Derivatives: Analysis of the Inhibitory Effect on Platelet Function and Their Antithrombotic Potential. J. Clin. Med. 2022, 11, 666. [Google Scholar] [CrossRef]
  234. Kimura, T.; Yamashita, S.; Kim, K.S.; Sezaki, H. Electrophysiological Approach to the Action of Taurine on Rat Gastric Mucosa. J. Pharmacobiodyn. 1982, 5, 495–500. [Google Scholar] [CrossRef] [PubMed]
  235. Rylance, H.J.; Myhal, D.R. Taurine Excretion and the Influence of Drugs. Clin. Chim. Acta 1971, 35, 159–164. [Google Scholar] [CrossRef] [PubMed]
  236. Baranovskii, S.F.; Bolotin, P.A. Association of Riboflavin, Caffeine, and Sodium Salicylate in Aqueous Solution. J. Appl. Spectrosc. 2007, 74, 211–218. [Google Scholar] [CrossRef]
  237. Scurachio, R.S.; Mattiucci, F.; Santos, W.G.; Skibsted, L.H.; Cardoso, D.R. Caffeine Metabolites Not Caffeine Protect against Riboflavin Photosensitized Oxidative Damage Related to Skin and Eye Health. J. Photochem. Photobiol. B 2016, 163, 277–283. [Google Scholar] [CrossRef] [PubMed]
  238. Schoenen, J.; Lenaerts, M.; Bastings, E. High-Dose Riboflavin as a Prophylactic Treatment of Migraine: Results of an Open Pilot Study. Cephalalgia 1994, 14, 328–329. [Google Scholar] [CrossRef] [PubMed]
  239. Norgard, N.R.B.; Bacon, N.; Agosti, M. Oral Administration of Sustained Release Niacin Inhibits Platelet Aggregation. Curr. Clin. Pharmacol. 2016, 11, 43–46. [Google Scholar] [CrossRef] [PubMed]
  240. Chesney, C.M.; Elam, M.B.; Herd, J.A.; Davis, K.B.; Garg, R.; Hunninghake, D.; Kennedy, J.W.; Applegate, W.B. Effect of Niacin, Warfarin, and Antioxidant Therapy on Coagulation Parameters in Patients with Peripheral Arterial Disease in the Arterial Disease Multiple Intervention Trial (ADMIT). Am. Heart J. 2000, 140, 631–636. [Google Scholar] [CrossRef] [PubMed]
  241. Philipp, C.S.; Cisar, L.A.; Saidi, P.; Kostis, J.B. Effect of Niacin Supplementation on Fibrinogen Levels in Patients with Peripheral Vascular Disease. Am. J. Cardiol. 1998, 82, 697–699. [Google Scholar] [CrossRef] [PubMed]
  242. Rumberger, J.A.; Napolitano, J.; Azumano, I.; Kamiya, T.; Evans, M. Pantethine, a Derivative of Vitamin B5 Used as a Nutritional Supplement, Favorably Alters Low-Density Lipoprotein Cholesterol Metabolism in Low– to Moderate–Cardiovascular Risk North American Subjects: A Triple-Blinded Placebo and Diet-Controlled Investigation. Nutr. Res. 2011, 31, 608–615. [Google Scholar] [CrossRef] [PubMed]
  243. Hiramatsu, K.; Nozaki, H.; Arimori, S. Influence of pantethine on platelet volume, microviscosity, lipid composition and functions in diabetes mellitus with hyperlipidemia. Tokai J. Exp. Clin. Med. 1981, 6, 49–57. [Google Scholar] [PubMed]
  244. Maqbool, T.; Hashmi, A.S.; Shah, B.H. Effect of Chronic Administration of Aspirin, Phenobarbitone and Oxytetracycline on the Plasma Levels of Vitamin A in Albino Rats (Partially Retrieved on a Server of the World Health Organization). JPMA J. Pak. Med. Assoc. 1990, 40, 89–90. [Google Scholar] [PubMed]
  245. Jávor, T.; Bata, M.; Lovász, L.; Morón, F.; Nagy, L.; Patty, I.; Szabolcs, J.; Tárnok, F.; Tóth, G.; Mózsik, G. Gastric Cytoprotective Effects of Vitamin A and Other Carotenoids. Int. J. Tissue React. 1983, 5, 289–296. [Google Scholar] [PubMed]
  246. Mózsik, G.; Jávor, T.; Tóth, G.; Zsoldos, T.; Tigyi, A. Interrelationships between the Gastric Cytoprotective Effects of Vitamin A and Beta-Carotene and the Gastric Mucosal Superoxide Dismutase Activity in Rats. Acta Physiol. Hung. 1984, 64, 315–318. [Google Scholar] [PubMed]
  247. Jávor, T.; Tárnok, F.; Past, T.; Nagy, S. Cytoprotective Effect of Free Radical Scavengers against Mucosal Damage Produced by Different Antirheumatic Drugs. Int. J. Tissue React. 1986, 8, 35–40. [Google Scholar] [PubMed]
  248. Moses, F.M.; Kikendall, J.W.; Bowen, P.; Young, T.R. Chronic β-Carotene Supplementation Does Not Alter the Gastric Mucosal Response to Acute Aspirin Ingestion. J. Clin. Gastroenterol. 1990, 12, 267–270. [Google Scholar] [CrossRef] [PubMed]
  249. Olson, J.A. Provitamin A Function of Carotenoids: The Conversion of β-Carotene into Vitamin A. J. Nutr. 1989, 119, 105–108. [Google Scholar] [CrossRef] [PubMed]
  250. Gomaa, A.A.; Hassan, H.A.; Ghaneimah, S.A. Effect of Aspirin and Indomethacin on the Serum and Urinary Calcium, Magnesium and Phosphate. Pharmacol. Res. 1990, 22, 59–70. [Google Scholar] [CrossRef] [PubMed]
  251. Bennati, G.; Cirino, M.; Benericetti, G.; Maximova, N.; Zanier, M.; Pigato, F.; Parzianello, A.; Maestro, A.; Barbi, E.; Zanon, D. Compounded Effervescent Magnesium for Familial Hypomagnesemia: A Case Report. Pharmaceuticals 2023, 16, 785. [Google Scholar] [CrossRef] [PubMed]
  252. Think Healthy Group, Inc. Bioavailability of Single-Dose Magnesium Salts; Clinical Trial Registration NCT04139928; Think Healthy Group, Inc.: Washington, DC, USA, 2020. Available online: https://clinicaltrials.gov/ct2/show/NCT04139928 (accessed on 11 July 2022).
  253. Brumas, V. Copper(II) Interactions with Nonsteroidal Antiinflammatory Agents. I. Salicylic Acid and Acetylsalicylic Acid. J. Inorg. Biochem. 1995, 57, 191–207. [Google Scholar] [CrossRef] [PubMed]
  254. Chohan, Z.H.; Iqbal, M.S.; Iqbal, H.S.; Scozzafava, A.; Supuran, C.T. Transition Metal Acetylsalicylates and Their Anti-Inflammatory Activity. J. Enzyme Inhib. Med. Chem. 2002, 17, 87–91. [Google Scholar] [CrossRef] [PubMed]
  255. Pearson, D.J.; Suarez-Mendez, V.J.; Day, J.P.; Miller, P.F. Selenium Status in Relation to Reduced Glutathione Peroxidase Activity in Aspirin-Sensitive Asthma. Clin. Exp. Allergy 1991, 21, 203–208. [Google Scholar] [CrossRef] [PubMed]
  256. Mattmiller, S.A.; Carlson, B.A.; Sordillo, L.M. Regulation of Inflammation by Selenium and Selenoproteins: Impact on Eicosanoid Biosynthesis. J. Nutr. Sci. 2013, 2, e28. [Google Scholar] [CrossRef] [PubMed]
  257. Birt, D.F.; Julius, A.D. Dietary Fat and Selenium Effects on Ex Vivo Prostaglandin Production in Rat Colon, Kidney, and Blood. Nutr. Cancer 1986, 8, 117–123. [Google Scholar] [CrossRef] [PubMed]
  258. Eisenmann, C.J.; Miller, R.K. The Effect of Selenium Compounds (Selenite, Selenate, Ebselen) on the Production of Thromboxane and Prostacyclin by the Human Term Placenta in Vitro. Toxicol. Appl. Pharmacol. 1995, 135, 18–24. [Google Scholar] [CrossRef] [PubMed]
  259. Perona, G.; Schiavon, R.; Guidi, G.C.; Veneri, D.; Minuz, P. Selenium Dependent Glutathione Peroxidase: A Physiological Regulatory System for Platelet Function. Thromb. Haemost. 1990, 64, 312–318. [Google Scholar] [CrossRef] [PubMed]
  260. Ducros, V. Chromium Metabolism: A Literature Review. Biol. Trace Elem. Res. 1992, 32, 65–77. [Google Scholar] [CrossRef] [PubMed]
  261. Hopkins, L.L.; Schwarz, K. Chromium (III) Binding to Serum Proteins, Specifically Siderophilin. Biochim. Biophys. Acta BBA-Gen. Subj. 1964, 90, 484–491. [Google Scholar] [CrossRef] [PubMed]
  262. Sparks, K.D.; Whitenack, M.L.; Stoecker, B.J.; Adeleye, B.O.; Sangiah, S. Aspirin and Buffering Compounds Affect the Absorption and Retention of 51chromium. J. Am. Diet. Assoc. 1995, 95, A18. [Google Scholar] [CrossRef]
  263. Yamamoto, J.; Taka, T.; Yamada, K.; Ijiri, Y.; Murakami, M.; Hirata, Y.; Naemura, A.; Hashimoto, M.; Yamashita, T.; Oiwa, K.; et al. Tomatoes Have Natural Anti-Thrombotic Effects. Br. J. Nutr. 2003, 90, 1031–1038. [Google Scholar] [CrossRef]
  264. Das, R.K.; Datta, T.; Biswas, D.; Duss, R.; O’Kennedy, N.; Duttaroy, A.K. Evaluation of the Equivalence of Different Intakes of Fruitflow in Affecting Platelet Aggregation and Thrombin Generation Capacity in a Randomized, Double-Blinded Pilot Study in Male Subjects. BMC Nutr. 2021, 7, 80. [Google Scholar] [CrossRef] [PubMed]
  265. Chen, H.; Zhang, S.; Wang, H.; Bao, L.; Wu, W.; Qi, R. Fruitflow Inhibits Platelet Function by Suppressing Akt/GSK3β, Syk/PLCγ2 and P38 MAPK Phosphorylation in Collagen-Stimulated Platelets. BMC Complement. Med. Ther. 2022, 22, 75. [Google Scholar] [CrossRef] [PubMed]
  266. Sawardekar, S.; Patel, T.; Uchil, D. Comparative Evaluation of Antiplatelet Effect of Lycopene with Aspirin and the Effect of Their Combination on Platelet Aggregation: An in Vitro Study. Indian J. Pharmacol. 2016, 48, 26. [Google Scholar] [CrossRef] [PubMed]
  267. Mayne, S.T.; Cartmel, B.; Silva, F.; Kim, C.S.; Fallon, B.G.; Briskin, K.; Zheng, T.; Baum, M.; Shor-Posner, G.; Goodwin, W.J. Plasma Lycopene Concentrations in Humans Are Determined by Lycopene Intake, Plasma Cholesterol Concentrations and Selected Demographic Factors. J. Nutr. 1999, 129, 849–854. [Google Scholar] [CrossRef] [PubMed]
  268. Cooperstone, J.L.; Goetz, H.J.; Riedl, K.M.; Harrison, E.H.; Schwartz, S.J.; Kopec, R.E. Relative Contribution of α-Carotene to Postprandial Vitamin A Concentrations in Healthy Humans after Carrot Consumption. Am. J. Clin. Nutr. 2017, 106, 59–66. [Google Scholar] [CrossRef] [PubMed]
  269. Serebruany, V.; Malinin, A.; Goodin, T.; Pashkow, F. The In Vitro Effects of Xancor, a Synthetic Astaxanthine Derivative, on Hemostatic Biomarkers in Aspirin-Naïve and Aspirin-Treated Subjects with Multiple Risk Factors for Vascular Disease. Am. J. Ther. 2010, 17, 125–132. [Google Scholar] [CrossRef] [PubMed]
  270. Neupane, P.; Bhuju, S.; Thapa, N.; Bhattarai, H.K. ATP Synthase: Structure, Function and Inhibition. Biomol. Concepts 2019, 10, 1–10. [Google Scholar] [CrossRef] [PubMed]
  271. Tewari, D.; Majumdar, D.; Vallabhaneni, S.; Bera, A.K. Aspirin Induces Cell Death by Directly Modulating Mitochondrial Voltage-Dependent Anion Channel (VDAC). Sci. Rep. 2017, 7, 45184. [Google Scholar] [CrossRef] [PubMed]
  272. Liesa, M.; Shirihai, O.S. Mitochondrial Dynamics in the Regulation of Nutrient Utilization and Energy Expenditure. Cell Metab. 2013, 17, 491–506. [Google Scholar] [CrossRef] [PubMed]
  273. Wesselink, E.; Koekkoek, W.A.C.; Grefte, S.; Witkamp, R.F.; Van Zanten, A.R.H. Feeding Mitochondria: Potential Role of Nutritional Components to Improve Critical Illness Convalescence. Clin. Nutr. 2019, 38, 982–995. [Google Scholar] [CrossRef] [PubMed]
  274. Ames, B.N. Delaying the Mitochondrial Decay of Aging. Ann. N. Y. Acad. Sci. 2004, 1019, 406–411. [Google Scholar] [CrossRef] [PubMed]
  275. Mackerer, C.R.; Mehlman, M.A.; Tobin, R.B. Effects of Chronic Acetylsalicylate Administration on Several Nutritional and Biochemical Parameters in Rats Fed Diets of Varied Thiamin Content. Biochem. Med. 1973, 8, 51–60. [Google Scholar] [CrossRef]
  276. Ames, B.N.; Atamna, H.; Killilea, D.W. Mineral and Vitamin Deficiencies Can Accelerate the Mitochondrial Decay of Aging. Mol. Aspects Med. 2005, 26, 363–378. [Google Scholar] [CrossRef] [PubMed]
  277. Brown, S.L.; Sobel, B.E.; Fujii, S. Attenuation of the Synthesis of Plasminogen Activator Inhibitor Type 1 by Niacin: A Potential Link Between Lipid Lowering and Fibrinolysis. Circulation 1995, 92, 767–772. [Google Scholar] [CrossRef]
  278. Johansson, J.O.; Egberg, N.; Asplund-Carlson, A.; Carlson, L.A. Nicotinic Acid Treatment Shifts the Fibrinolytic Balance Favourably and Decreases Plasma Fibrinogen in Hypertriglyceridaemic Men. J. Cardiovasc. Risk 1997, 4, 165–171. [Google Scholar] [CrossRef]
  279. Christopher, A. Critically Elevated INR in a Patient on Warfarin After Increase in Extended-Release Niacin Dose. Ann. Pharmacother. 2011, 45, 1450. [Google Scholar] [CrossRef]
  280. Shohag, M.J.I.; Wei, Y.; Yu, N.; Lu, L.; Zhang, J.; He, Z.; Patring, J.; Yang, X. Folate Content and Composition of Vegetables Commonly Consumed in China. J. Food Sci. 2012, 77, H239–H245. [Google Scholar] [CrossRef] [PubMed]
  281. Tulipani, S.; Romandini, S.; Suarez, J.M.A.; Capocasa, F.; Mezzetti, B.; Battino, M.; Busco, F.; Bamonti, F.; Novembrino, C. Folate Content in Different Strawberry Genotypes and Folate Status in Healthy Subjects after Strawberry Consumption. BioFactors 2008, 34, 47–55. [Google Scholar] [CrossRef] [PubMed]
  282. Krajcovicová-Kudlácková, M.; Valachovicová, M.; Blazícek, P. Seasonal Folate Serum Concentrations at Different Nutrition. Cent. Eur. J. Public Health 2013, 21, 36–38. [Google Scholar] [CrossRef] [PubMed]
  283. Leblanc, C.; Presse, N.; Lalonde, G.; Dumas, S.; Ferland, G. Higher Vitamin K Intake Is Associated with Better INR Control and a Decreased Need for INR Tests in Long-Term Warfarin Therapy. Thromb. Res. 2014, 134, 210–212. [Google Scholar] [CrossRef] [PubMed]
  284. Ferland, G.; Chahine, S.; Presse, N.; Dube, M.-P.; Nigam, A.; Blostein, M.; deDenus, S.; Perreault, S.; Tardif, J.-C. Increasing Dietary Vitamin K Intake Stabilizes Anticoagulation Therapy in Warfarin-Treated Patients with a History of Instability: A 24-Week Randomized Controlled Trial (OR36-04-19). Curr. Dev. Nutr. 2019, 3 (Suppl. S1), nzz035.OR36-04-19. [Google Scholar] [CrossRef]
  285. Leblanc, C.; Dubé, M.-P.; Presse, N.; Dumas, S.; Nguyen, M.; Rouleau-Mailloux, É.; Perreault, S.; Ferland, G. Avoidance of Vitamin K-Rich Foods Is Common among Warfarin Users and Translates into Lower Usual Vitamin K Intakes. J. Acad. Nutr. Diet. 2016, 116, 1000–1007. [Google Scholar] [CrossRef] [PubMed]
  286. Sobczyńska-Malefora, A.J.; Harrington, D.; Rangarajan, S.; Kovacs, J.-A.; Shearer, M.J.; Savidge, G.F. Hyperhomocysteinemia and B-Vitamin Status after Discontinuation of Oral Anticoagulation Therapy in Patients with a History of Venous Thromboembolism. Clin. Chem. Lab. Med. 2003, 41, 1493–1497. [Google Scholar] [CrossRef] [PubMed]
  287. Sobczyńska-Malefora, A.; Harrington, D.J.; Lomer, M.C.; Pettitt, C.; Hamilton, S.; Rangarajan, S.; Shearer, M.J. Erythrocyte Folate and 5-Methyltetrahydrofolate Levels Decline during 6 Months of Oral Anticoagulation with Warfarin. Blood Coagul. Fibrinolysis 2009, 20, 297–302. [Google Scholar] [CrossRef] [PubMed]
  288. Lind, M.; Jansson, J.-H.; Nilsson, T.K.; Johansson, L. High Homocysteine and Low Folate Plasma Concentrations Are Associated with Cardiovascular Events but Not Bleeding during Warfarin Treatment. Clin. Chem. Lab. Med. CCLM 2016, 54, 1981–1986. [Google Scholar] [CrossRef] [PubMed]
  289. Muszkat, M.; Bialer, O.; Blotnick, S.; Adar, L.; Xie, H.-G.; Ufer, M.; Cascorbi, I.; Caraco, Y. Effects of Folic Acid Supplementation on the Pharmacokinetics and Anticoagulant Effect of Warfarin: An Open-Label, Prospective Study of Long-Term Administration in Adults. Clin. Ther. 2010, 32, 347–356. [Google Scholar] [CrossRef] [PubMed]
  290. Harris, J.E. Interaction of Dietary Factors with Oral Anticoagulants: Review and Applications. J. Am. Diet. Assoc. 1995, 95, 580–584. [Google Scholar] [CrossRef] [PubMed]
  291. Deckert, F.W. Ascorbic Acid and Warfarin. JAMA 1973, 223, 440. [Google Scholar] [CrossRef] [PubMed]
  292. Rosenthal, G. Interaction of Ascorbic Acid and Warfarin. JAMA J. Am. Med. Assoc. 1971, 215, 1671. [Google Scholar] [CrossRef]
  293. Smith, E.C.; Skalski, R.J.; Johnson, G.C.; Rossi, G.V. Interaction of Ascorbic Acid and Warfarin. JAMA 1972, 221, 1166. [Google Scholar] [CrossRef]
  294. Hume, R.; Johnstone, J.M.; Weyers, E. Interaction of Ascorbic Acid and Warfarin. JAMA 1972, 219, 1479. [Google Scholar] [CrossRef] [PubMed]
  295. Dedichen, J. The Effect of Ascorbic Acid given to Patients on Chronic Anticoagulant Therapy. Boll. Soc. Ital. Cardiol. 1973, 18, 690–692. [Google Scholar] [PubMed]
  296. Weintraub, M.; Griner, P.F. Warfarin and Ascorbic Acid: Lack of Evidence for a Drug Interaction. Toxicol. Appl. Pharmacol. 1974, 28, 53–56. [Google Scholar] [CrossRef] [PubMed]
  297. Feetam, C.L.; Leach, R.H.; Meynell, M.J. Lack of a Clinically Important Interaction between Warfarin and Ascorbic Acid. Toxicol. Appl. Pharmacol. 1975, 31, 544–547. [Google Scholar] [CrossRef]
  298. Wynne, H.; Khan, T.; Avery, P.; Wood, P.; Ward, A.; Kamali, F. Dietary Related Plasma Vitamin C Concentration Has No Effect on Anticoagulation Response to Warfarin. Thromb. Res. 2006, 118, 501–504. [Google Scholar] [CrossRef] [PubMed]
  299. Sattar, A.; Willman, J.E.; Kolluri, R. Possible Warfarin Resistance Due to Interaction with Ascorbic Acid: Case Report and Literature Review. Am. J. Health-Syst. Pharm. AJHP Off. J. Am. Soc. Health-Syst. Pharm. 2013, 70, 782–786. [Google Scholar] [CrossRef] [PubMed]
  300. Monterrey-Rodríguez, J.; Feliú, J.F.; Rivera-Miranda, G.C. Interaction between Warfarin and Mango Fruit. Ann. Pharmacother. 2002, 36, 940–941. [Google Scholar] [CrossRef] [PubMed]
  301. Vitamin Supplements. Med. Lett. Drugs Ther. 1985, 27, 66–68.
  302. Yamazaki, H. Effects of Arachidonic Acid, Prostaglandins, Retinol, Retinoic Acid and Cholecalciferol on Xenobiotic Oxidations Catalysed by Human Cytochrome P450 Enzymes. Xenobiotica 1999, 29, 231–241. [Google Scholar] [CrossRef] [PubMed]
  303. Garewal, H.S.; Diplock, A.T. How ???Safe??? Are Antioxidant Vitamins? Drug Saf. 1995, 13, 8–14. [Google Scholar] [CrossRef] [PubMed]
  304. Kim, J.M.; White, R.H. Effect of Vitamin E on the Anticoagulant Response to Warfarin. Am. J. Cardiol. 1996, 77, 545–546. [Google Scholar] [CrossRef] [PubMed]
  305. Pastori, D.; Carnevale, R.; Cangemi, R.; Saliola, M.; Nocella, C.; Bartimoccia, S.; Vicario, T.; Farcomeni, A.; Violi, F.; Pignatelli, P. Vitamin E Serum Levels and Bleeding Risk in Patients Receiving Oral Anticoagulant Therapy: A Retrospective Cohort Study. J. Am. Heart Assoc. 2013, 2, e000364. [Google Scholar] [CrossRef] [PubMed]
  306. Corrigan, J.J. The effect of vitamin e on warfarin-induced vitamin k deficiency*. Ann. N. Y. Acad. Sci. 1982, 393, 361–368. [Google Scholar] [CrossRef] [PubMed]
  307. Traber, M.G. Vitamin E and K Interactions--a 50-Year-Old Problem. Nutr. Rev. 2008, 66, 624–629. [Google Scholar] [CrossRef]
  308. Hejazi, M.E.; Modarresi-Ghazani, F.; Hamishehkar, H.; Mesgari-Abbasi, M.; Dousti, S.; Entezari-Maleki, T. The Effect of Treatment of Vitamin D Deficiency on the Level of P-Selectin and Hs-CRP in Patients with Thromboembolism: A Pilot Randomized Clinical Trial. J. Clin. Pharmacol. 2017, 57, 40–47. [Google Scholar] [CrossRef]
  309. Khansari, N.; Bagheri, M.; Homayounfar, S.; Poorolajal, J.; Mehrpooya, M. Influence of Vitamin D Status on the Maintenance Dose of Warfarin in Patients Receiving Chronic Warfarin Therapy. Cardiol. Ther. 2022, 11, 421–432. [Google Scholar] [CrossRef] [PubMed]
  310. Mohammad, S.; Mishra, A.; Ashraf, M.Z. Emerging Role of Vitamin D and Its Associated Molecules in Pathways Related to Pathogenesis of Thrombosis. Biomolecules 2019, 9, 649. [Google Scholar] [CrossRef] [PubMed]
  311. Margier, M.; Collet, X.; le May, C.; Desmarchelier, C.; André, F.; Lebrun, C.; Defoort, C.; Bluteau, A.; Borel, P.; Lespine, A.; et al. ABCB1 (P-Glycoprotein) Regulates Vitamin D Absorption and Contributes to Its Transintestinal Efflux. FASEB J. 2019, 33, 2084–2094. [Google Scholar] [CrossRef] [PubMed]
  312. Price, P.A.; Faus, S.A.; Williamson, M.K. Warfarin-Induced Artery Calcification Is Accelerated by Growth and Vitamin D. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 317–327. [Google Scholar] [CrossRef] [PubMed]
  313. Card, D.J.; Gorska, R.; Harrington, D.J. Laboratory Assessment of Vitamin K Status. J. Clin. Pathol. 2020, 73, 70–75. [Google Scholar] [CrossRef] [PubMed]
  314. Donaldson, C.J.; Harrington, D.J. Therapeutic Warfarin Use and the Extrahepatic Functions of Vitamin K-Dependent Proteins. Br. J. Biomed. Sci. 2017, 74, 163–169. [Google Scholar] [CrossRef] [PubMed]
  315. Theuwissen, E.; Smit, E.; Vermeer, C. The Role of Vitamin K in Soft-Tissue Calcification. Adv. Nutr. 2012, 3, 166–173. [Google Scholar] [CrossRef] [PubMed]
  316. Roumeliotis, S.; Dounousi, E.; Eleftheriadis, T.; Liakopoulos, V. Association of the Inactive Circulating Matrix Gla Protein with Vitamin K Intake, Calcification, Mortality, and Cardiovascular Disease: A Review. Int. J. Mol. Sci. 2019, 20, 628. [Google Scholar] [CrossRef] [PubMed]
  317. Cirilli, I.; Orlando, P.; Silvestri, S.; Marcheggiani, F.; Dludla, P.V.; Kaesler, N.; Tiano, L. Carboxylative Efficacy of Trans and Cis MK7 and Comparison with Other Vitamin K Isomers. BioFactors Oxf. Engl. 2022, 48, 1129–1136. [Google Scholar] [CrossRef] [PubMed]
  318. Sato, T.; Inaba, N.; Yamashita, T. MK-7 and Its Effects on Bone Quality and Strength. Nutrients 2020, 12, 965. [Google Scholar] [CrossRef]
  319. Margier, M.; Le May, C.; Antoine, T.; Halimi, C.; Nowicki, M.; Lespine, A.; Reboul, E. P-Glycoprotein (ABCB1) Is Involved in Vitamin K Efflux. Food Chem. 2021, 343, 128510. [Google Scholar] [CrossRef] [PubMed]
  320. Fawzy, A.M.; Lip, G.Y.H. Warfarin and Increased Fracture Risk? Answering the Big Question. Age Ageing 2022, 51, afab263. [Google Scholar] [CrossRef] [PubMed]
  321. Trost, S.; Tesfaye, N.; Harindhanavudhi, T. The Interplay between Bone and Heart Health as Reflected in Medication Effects: A Narrative Review. Womens Health 2023, 19, 174550572311655. [Google Scholar] [CrossRef] [PubMed]
  322. Avila, M.L.; Pullenayegum, E.; Williams, S.; Shammas, A.; Stimec, J.; Sochett, E.; Marr, K.; Brandão, L.R. Timing of Low Bone Mineral Density and Predictors of Bone Mineral Density Trajectory in Children on Long-Term Warfarin: A Longitudinal Study. Osteoporos. Int. 2016, 27, 1547–1557. [Google Scholar] [CrossRef] [PubMed]
  323. Rezaieyazdi, Z.; Falsoleiman, H.; Khajehdaluee, M.; Saghafi, M.; Mokhtari-Amirmajdi, E. Reduced Bone Density in Patients on Long-Term Warfarin. Int. J. Rheum. Dis. 2009, 12, 130–135. [Google Scholar] [CrossRef] [PubMed]
  324. Huang, H.-K.; Peng, C.C.-H.; Lin, S.-M.; Munir, K.M.; Chang, R.H.-E.; Wu, B.B.-C.; Liu, P.P.-S.; Hsu, J.-Y.; Loh, C.-H.; Tu, Y.-K. Fracture Risks in Patients Treated with Different Oral Anticoagulants: A Systematic Review and Meta-Analysis. J. Am. Heart Assoc. 2021, 10, e019618. [Google Scholar] [CrossRef] [PubMed]
  325. Sugiyama, T.; Kugimiya, F.; Kono, S.; Kim, Y.T.; Oda, H. Warfarin Use and Fracture Risk: An Evidence-Based Mechanistic Insight. Osteoporos. Int. 2015, 26, 1231–1232. [Google Scholar] [CrossRef] [PubMed]
  326. Poterucha, T.J.; Goldhaber, S.Z. Warfarin and Vascular Calcification. Am. J. Med. 2016, 129, 635.e1–635.e4. [Google Scholar] [CrossRef]
  327. Lee, S.J.; Lee, I.-K.; Jeon, J.-H. Vascular Calcification—New Insights into Its Mechanism. Int. J. Mol. Sci. 2020, 21, 2685. [Google Scholar] [CrossRef]
  328. Elango, K.; Javaid, A.; Khetarpal, B.K.; Ramalingam, S.; Kolandaivel, K.P.; Gunasekaran, K.; Ahsan, C. The Effects of Warfarin and Direct Oral Anticoagulants on Systemic Vascular Calcification: A Review. Cells 2021, 10, 773. [Google Scholar] [CrossRef] [PubMed]
  329. Anticoagulation Algorithm for Fontan Patients. American College of Cardiology. Available online: https://www.acc.org/Latest-in-Cardiology/Articles/2023/04/05/14/10/Anticoagulation-Algorithm-For-Fontan-Patients (accessed on 10 October 2023).
  330. Liu, J.; Liu, C.; Qian, C.; Abela, G.; Sun, W.; Kong, X. Ginkgo Biloba Extract EGB761 Alleviates Warfarin-Induced Aortic Valve Calcification Through the BMP2/Smad1/5/Runx2 Signaling Pathway. J. Cardiovasc. Pharmacol. 2021, 78, 411–421. [Google Scholar] [CrossRef] [PubMed]
  331. Haemorrhage Due to Ginkgo Biloba? Prescrire Int. 2008, 17, 19.
  332. Theuwissen, E.; Teunissen, K.J.; Spronk, H.M.H.; Hamulyák, K.; Ten Cate, H.; Shearer, M.J.; Vermeer, C.; Schurgers, L.J. Effect of Low-Dose Supplements of Menaquinone-7 (Vitamin K2) on the Stability of Oral Anticoagulant Treatment: Dose-Response Relationship in Healthy Volunteers. J. Thromb. Haemost. JTH 2013, 11, 1085–1092. [Google Scholar] [CrossRef] [PubMed]
  333. Stenton, S.B.; Bungard, T.J.; Ackman, M.L. Interactions between Warfarin and Herbal Products, Minerals, and Vitamins: A Pharmacist’s Guide. Can. J. Hosp. Pharm. 2001, 54, 186–192. [Google Scholar] [CrossRef]
  334. Ardahanli, I.; Akhan, O.; Celik, M. The Effect of Serum Magnesium Level on Stable Anticoagulation in Patients Using Warfarin for Various Cardiac Indications. Biol. Trace Elem. Res. 2022, 200, 4297–4302. [Google Scholar] [CrossRef] [PubMed]
  335. Sekiya, F.; Yoshida, M.; Yamashita, T.; Morita, T. Magnesium(II) Is a Crucial Constituent of the Blood Coagulation Cascade. J. Biol. Chem. 1996, 271, 8541–8544. [Google Scholar] [CrossRef] [PubMed]
  336. Gajsiewicz, J.M.; Nuzzio, K.M.; Rienstra, C.M.; Morrissey, J.H. Tissue Factor Residues That Modulate Magnesium-Dependent Rate Enhancements of the Tissue Factor/Factor VIIa Complex. Biochemistry 2015, 54, 4665–4671. [Google Scholar] [CrossRef] [PubMed]
  337. Han, H. The Importance of Vitamin K Monitoring with Warfarin (Coumadin) Use in Chronic Kidney Disease Patients. J. Ren. Nutr. 2015, 25, e7–e8. [Google Scholar] [CrossRef] [PubMed]
  338. Brookes, E.M.; Snider, J.; Hart, G.K.; Robbins, R.; Power, D.A. Serum Potassium Abnormalities in Chronic Kidney Disease: Prevalence, Patient Characteristics and Clinical Outcomes. Intern. Med. J. 2021, 51, 1906–1918. [Google Scholar] [CrossRef] [PubMed]
  339. Santiyanon, N.; Yeephu, S. Interaction between Warfarin and Astaxanthin: A Case Report. J. Cardiol. Cases 2019, 19, 173–175. [Google Scholar] [CrossRef]
  340. Zheng, Y.F.; Bae, S.H.; Kwon, M.J.; Park, J.B.; Choi, H.D.; Shin, W.G.; Bae, S.K. Inhibitory Effects of Astaxanthin, β-Cryptoxanthin, Canthaxanthin, Lutein, and Zeaxanthin on Cytochrome P450 Enzyme Activities. Food Chem. Toxicol. 2013, 59, 78–85. [Google Scholar] [CrossRef]
  341. Suttie, J.W.; Jackson, C.M. Prothrombin Structure, Activation, and Biosynthesis. Physiol. Rev. 1977, 57, 1–70. [Google Scholar] [CrossRef] [PubMed]
  342. Preusch, P.C.; Suttie, J.W. Vitamin K-Dependent Reactions in Rat Liver: Role of Flavoproteins. J. Nutr. 1981, 111, 2087–2097. [Google Scholar] [CrossRef]
  343. Worcester, E.M.; Sebastian, J.L.; Hiatt, J.G.; Beshensky, A.M.; Sadowski, J.A. The Effect of Warfarin on Urine Calcium Oxalate Crystal Growth Inhibition and Urinary Excretion of Calcium and Nephrocalcin. Calcif. Tissue Int. 1993, 53, 242–248. [Google Scholar] [CrossRef] [PubMed]
  344. McElnay, J.C.; Harron, D.W.; D’Arcy, P.F.; Collier, P.S. The Interaction of Warfarin with Antacid Constituents in the Gut. Experientia 1979, 35, 1359–1360. [Google Scholar] [CrossRef] [PubMed]
  345. Ambre, J.J.; Fischer, L.J. Effect of Coadministration of Aluminum and Magnesium Hydroxides on Absorption of Anticoagulants in Man. Clin. Pharmacol. Ther. 1973, 14, 231–237. [Google Scholar] [CrossRef] [PubMed]
  346. Blancquaert, L.; Vervaet, C.; Derave, W. Predicting and Testing Bioavailability of Magnesium Supplements. Nutrients 2019, 11, 1663. [Google Scholar] [CrossRef] [PubMed]
  347. PubChem. Warfarin Sodium. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/16204922 (accessed on 10 October 2023).
  348. DailyMed—WARFARIN SODIUM Tablet. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=36a919ce-7a21-40c0-9af5-2299bf2b1f97 (accessed on 10 October 2023).
  349. Flint, S.; Taylor, E.; Beavis, J.; Becker, G.J.; Pedagogos, E. Increased Iron Requirement in Hemodialysis Patients on Antiplatelet Agents or Warfarin. Nephron Clin. Pract. 2009, 113, c38–c45. [Google Scholar] [CrossRef] [PubMed]
  350. Nicholls, D.P.; Shepherd, A.M.M. Altered warfarin metabolism in iron-deficient patient. Lancet 1978, 312, 215–216. [Google Scholar] [CrossRef] [PubMed]
  351. Gilardi, G.; Di Nardo, G. Heme Iron Centers in Cytochrome P450: Structure and Catalytic Activity. Rend. Lincei 2017, 28 (Suppl. S1), 159–167. [Google Scholar] [CrossRef]
  352. Hernández-Camacho, J.D.; Bernier, M.; López-Lluch, G.; Navas, P. Coenzyme Q10 Supplementation in Aging and Disease. Front. Physiol. 2018, 9, 44. [Google Scholar] [CrossRef]
  353. Bentinger, M.; Brismar, K.; Dallner, G. The Antioxidant Role of Coenzyme Q. Mitochondrion 2007, 7, S41–S50. [Google Scholar] [CrossRef] [PubMed]
  354. Sangsefidi, Z.S.; Yaghoubi, F.; Hajiahmadi, S.; Hosseinzadeh, M. The Effect of Coenzyme Q10 Supplementation on Oxidative Stress: A Systematic Review and Meta-Analysis of Randomized Controlled Clinical Trials. Food Sci. Nutr. 2020, 8, 1766–1776. [Google Scholar] [CrossRef] [PubMed]
  355. Combs, A.B.; Porter, T.H.; Folkers, K. Anticoagulant Activity of a Naphthoquinone Analog of Vitamin K and an Inhibitor of Coenzyme Q10- Enzyme Systems. Res. Commun. Chem. Pathol. Pharmacol. 1976, 13, 109–114. [Google Scholar] [PubMed]
  356. Morton, R.A. Ubiquinones, plastoquinones and vitamins K. Biol. Rev. 1971, 46, 47–96. [Google Scholar] [CrossRef] [PubMed]
  357. Zhou, S.; Chan, E. Effect of Ubidecarenone on Warfarin Anticoagulation and Pharmacokinetics of Warfarin Enantiomers in Rats. Drug Metabol. Drug Interact. 2001, 18, 99–122. [Google Scholar] [CrossRef] [PubMed]
  358. Zhou, Q.; Zhou, S.; Chan, E. Effect of Coenzyme Q10 on Warfarin Hydroxylation in Rat and Human Liver Microsomes. Curr. Drug Metab. 2005, 6, 67–81. [Google Scholar] [CrossRef] [PubMed]
  359. Heck, A.M.; DeWitt, B.A.; Lukes, A.L. Potential Interactions between Alternative Therapies and Warfarin. Am. J. Health-Syst. Pharm. AJHP Off. J. Am. Soc. Health-Syst. Pharm. 2000, 57, 1221–1227; quiz 1228–1230. [Google Scholar] [CrossRef]
  360. Spigset, O. Reduced Effect of Warfarin Caused by Ubidecarenone. Lancet 1994, 344, 1372–1373. [Google Scholar] [CrossRef] [PubMed]
  361. Landbo, C.; Almdal, T.P. [Interaction between warfarin and coenzyme Q10]. Ugeskr. Laeger 1998, 160, 3226–3227. [Google Scholar] [PubMed]
  362. Engelsen, J.; Nielsen, J.D.; Hansen, K.F.W. [Effect of Coenzyme Q10 and Ginkgo biloba on warfarin dosage in patients on long-term warfarin treatment. A randomized, double-blind, placebo-controlled cross-over trial]. Ugeskr. Laeger 2003, 165, 1868–1871. [Google Scholar] [PubMed]
  363. Odén, A.; Fahlén, M. Oral Anticoagulation and Risk of Death: A Medical Record Linkage Study. BMJ 2002, 325, 1073–1075. [Google Scholar] [CrossRef] [PubMed]
  364. Fiddian-Green, R.G. Did the Anticoagulants Cause Deaths by Inducing Mitochondrial Dysfuinction? 2023. Available online: https://www.bmj.com/rapid-response/2011/10/29/did-anticoagulants-cause-deaths-inducing-mitochondrial-dysfuinction (accessed on 20 March 2024).
  365. Bețiu, A.M.; Noveanu, L.; Hâncu, I.M.; Lascu, A.; Petrescu, L.; Maack, C.; Elmér, E.; Muntean, D.M. Mitochondrial Effects of Common Cardiovascular Medications: The Good, the Bad and the Mixed. Int. J. Mol. Sci. 2022, 23, 13653. [Google Scholar] [CrossRef] [PubMed]
  366. Szabo, R.; Bodolea, C.; Mocan, T. Iron, Copper, and Zinc Homeostasis: Physiology, Physiopathology, and Nanomediated Applications. Nanomaterials 2021, 11, 2958. [Google Scholar] [CrossRef]
  367. Hitchings, R.; Kelly, L. Predicting and Understanding the Human Microbiome’s Impact on Pharmacology. Trends Pharmacol. Sci. 2019, 40, 495–505. [Google Scholar] [CrossRef] [PubMed]
  368. Sharma, A.; Buschmann, M.M.; Gilbert, J.A. Pharmacomicrobiomics: The Holy Grail to Variability in Drug Response? Clin. Pharmacol. Ther. 2019, 106, 317–328. [Google Scholar] [CrossRef]
  369. Pryor, R.; Martinez-Martinez, D.; Quintaneiro, L.; Cabreiro, F. The Role of the Microbiome in Drug Response. Annu. Rev. Pharmacol. Toxicol. 2020, 60, 417–435. [Google Scholar] [CrossRef]
  370. Masnoon, N.; Shakib, S.; Kalisch-Ellett, L.; Caughey, G.E. What Is Polypharmacy? A Systematic Review of Definitions. BMC Geriatr. 2017, 17, 230. [Google Scholar] [CrossRef] [PubMed]
  371. Bakaki, P.M.; Horace, A.; Dawson, N.; Winterstein, A.; Waldron, J.; Staley, J.; Pestana Knight, E.M.; Meropol, S.B.; Liu, R.; Johnson, H.; et al. Defining Pediatric Polypharmacy: A Scoping Review. PLoS ONE 2018, 13, e0208047. [Google Scholar] [CrossRef] [PubMed]
  372. Kok, W.E.; Haverkort, E.B.; Algra, Y.A.; Mollema, J.; Hollaar, V.R.Y.; Naumann, E.; Schueren, M.A.E.; de van der Jerković-Ćosić, K. The Association between Polypharmacy and Malnutrition(Risk) in Older People: A Systematic Review. Clin. Nutr. ESPEN 2022, 49, 163–171. [Google Scholar] [CrossRef] [PubMed]
  373. Doan, J.; Zakrzewski-Jakubiak, H.; Roy, J.; Turgeon, J.; Tannenbaum, C. Prevalence and Risk of Potential Cytochrome P450-Mediated Drug-Drug Interactions in Older Hospitalized Patients with Polypharmacy. Ann. Pharmacother. 2013, 47, 324–332. [Google Scholar] [CrossRef] [PubMed]
  374. Kaganov, B.; Caroli, M.; Mazur, A.; Singhal, A.; Vania, A. Suboptimal Micronutrient Intake among Children in Europe. Nutrients 2015, 7, 3524–3535. [Google Scholar] [CrossRef]
  375. Kordas, K.; Lönnerdal, B.; Stoltzfus, R.J. Interactions between Nutrition and Environmental Exposures: Effects on Health Outcomes in Women and Children. J. Nutr. 2007, 137, 2794–2797. [Google Scholar] [CrossRef] [PubMed]
  376. Avitabile, C.M.; Leonard, M.B.; Zemel, B.S.; Brodsky, J.L.; Lee, D.; Dodds, K.; Hayden-Rush, C.; Whitehead, K.K.; Goldmuntz, E.; Paridon, S.M.; et al. Lean Mass Deficits, Vitamin D Status and Exercise Capacity in Children and Young Adults after Fontan Palliation. Heart 2014, 100, 1702–1707. [Google Scholar] [CrossRef] [PubMed]
  377. Bruun, D.E.R. Vitamin D and Calcium Intake in Children with Fontan Circulation—In Relationship to Vitamin D Status and Bone Mineral Density. Master’s Thesis, University of Oslo, Oslo, Norway, 2021. [Google Scholar]
  378. Charpie, J.R. Vitamin C May Improve Endothelial Function and Exercise Capacity in Functional Single Ventricle Patients after Fontan Palliation; Clinical Ttrial Registration NCT00974025. 2017. Available online: https://clinicaltrials.gov/ct2/show/NCT00974025 (accessed on 8 May 2022).
  379. Diab, S.G.; Godang, K.; Müller, L.O.; Almaas, R.; Lange, C.; Brunvand, L.; Hansen, K.M.; Myhre, A.G.; Døhlen, G.; Thaulow, E.; et al. Progressive Loss of Bone Mass in Children with Fontan Circulation. Congenit. Heart Dis. 2019, 14, 996–1004. [Google Scholar] [CrossRef]
  380. Renaud, D.; Scholl-Bürgi, S.; Karall, D.; Michel, M. Comparative Metabolomics in Single Ventricle Patients after Fontan Palliation: A Strong Case for a Targeted Metabolic Therapy. Metabolites 2023, 13, 932. [Google Scholar] [CrossRef] [PubMed]
  381. Michel, M.; Dubowy, K.-O.; Entenmann, A.; Karall, D.; Adam, M.G.; Zlamy, M.; Odri Komazec, I.; Geiger, R.; Niederwanger, C.; Salvador, C.; et al. Targeted Metabolomic Analysis of Serum Amino Acids in the Adult Fontan Patient with a Dominant Left Ventricle. Sci. Rep. 2020, 10, 8930. [Google Scholar] [CrossRef] [PubMed]
  382. Michel, M.; Dubowy, K.-O.; Zlamy, M.; Karall, D.; Adam, M.G.; Entenmann, A.; Keller, M.A.; Koch, J.; Odri Komazec, I.; Geiger, R.; et al. Targeted Metabolomic Analysis of Serum Phospholipid and Acylcarnitine in the Adult Fontan Patient with a Dominant Left Ventricle. Ther. Adv. Chronic Dis. 2020, 11, 204062232091603. [Google Scholar] [CrossRef] [PubMed]
  383. Schranz, D.; Voelkel, N.F. “Nihilism” of Chronic Heart Failure Therapy in Children and Why Effective Therapy Is Withheld. Eur. J. Pediatr. 2016, 175, 445–455. [Google Scholar] [CrossRef]
  384. Nutescu, E.A.; Shapiro, N.L.; Ibrahim, S.; West, P. Warfarin and Its Interactions with Foods, Herbs and Other Dietary Supplements. Expert Opin. Drug Saf. 2006, 5, 433–451. [Google Scholar] [CrossRef] [PubMed]
  385. Mallet, L.; Spinewine, A.; Huang, A. The Challenge of Managing Drug Interactions in Elderly People. Lancet 2007, 370, 185–191. [Google Scholar] [CrossRef] [PubMed]
  386. Lonsdale, D. Thiamine and Magnesium Deficiencies: Keys to Disease. Med. Hypotheses 2015, 84, 129–134. [Google Scholar] [CrossRef] [PubMed]
  387. Stevens, G.A.; Beal, T.; Mbuya, M.N.N.; Luo, H.; Neufeld, L.M.; Addo, O.Y.; Adu-Afarwuah, S.; Alayón, S.; Bhutta, Z.; Brown, K.H.; et al. Micronutrient Deficiencies among Preschool-Aged Children and Women of Reproductive Age Worldwide: A Pooled Analysis of Individual-Level Data from Population-Representative Surveys. Lancet Glob. Health 2022, 10, e1590–e1599. [Google Scholar] [CrossRef] [PubMed]
  388. Kirkwood, T.B.; Rose, M.R. Evolution of Senescence: Late Survival Sacrificed for Reproduction. Philos. Trans. R. Soc. Lond. B Biol. Sci. 1991, 332, 15–24. [Google Scholar] [CrossRef] [PubMed]
  389. Lamers, Y. Approaches to Improving Micronutrient Status Assessment at the Population Level. Proc. Nutr. Soc. 2019, 78, 170–176. [Google Scholar] [CrossRef] [PubMed]
  390. Ervin, R.B.; Kennedy-Stephenson, J. Mineral Intakes of Elderly Adult Supplement and Non-Supplement Users in the Third National Health and Nutrition Examination Survey. J. Nutr. 2002, 132, 3422–3427. [Google Scholar] [CrossRef] [PubMed]
  391. Grüne, S. Anamnese und körperliche Untersuchung. DMW-Dtsch. Med. Wochenschr. 2015, 141, 24–27. [Google Scholar] [CrossRef] [PubMed]
  392. Horn, J.R.; Hansten, P.D.; Chan, L.-N. Proposal for a New Tool to Evaluate Drug Interaction Cases. Ann. Pharmacother. 2007, 41, 674–680. [Google Scholar] [CrossRef] [PubMed]
  393. Komperda, K.E. Potential Interaction Between Pomegranate Juice and Warfarin. Pharmacotherapy 2009, 29, 1002–1006. [Google Scholar] [CrossRef] [PubMed]
  394. Hanselin, M.R.; Vande Griend, J.P.; Linnebur, S.A. INR Elevation with Maitake Extract in Combination with Warfarin. Ann. Pharmacother. 2010, 44, 223–224. [Google Scholar] [CrossRef] [PubMed]
  395. Sadkovsky, I.A.; Golubnitschaja, O.; Mandrik, M.A.; Studneva, M.A.; Abe, H.; Schroeder, H.; Antonova, E.N.; Betsou, F.; Bodrova, T.A.; Payne, K.; et al. PPPM (Predictive, Preventive and Personalized Medicine) as a New Model of the National and International Healthcare Services and Thus a Promising Strategy to Prevent a Disease: From Basics to Practice. Int. J. Clin. Med. 2014, 05, 855–870. [Google Scholar] [CrossRef]
  396. McColl, E.R.; Asthana, R.; Paine, M.F.; Piquette-Miller, M. The Age of Omics-Driven Precision Medicine. Clin. Pharmacol. Ther. 2019, 106, 477–481. [Google Scholar] [CrossRef] [PubMed]
  397. Krier, J.B.; Kalia, S.S.; Green, R.C. Genomic Sequencing in Clinical Practice: Applications, Challenges, and Opportunities. Dialogues Clin. Neurosci. 2016, 18, 299–312. [Google Scholar] [CrossRef] [PubMed]
  398. Al-Amrani, S.; Al-Jabri, Z.; Al-Zaabi, A.; Alshekaili, J.; Al-Khabori, M. Proteomics: Concepts and Applications in Human Medicine. World J. Biol. Chem. 2021, 12, 57–69. [Google Scholar] [CrossRef] [PubMed]
  399. Kirwan, J.A. Translating Metabolomics into Clinical Practice. Nat. Rev. Bioeng. 2023, 1, 228–229. [Google Scholar] [CrossRef]
  400. Archibald, J. Genomics: A Very Short Introduction; Oxford University Press: Oxford, UK, 2018; Volume 1. [Google Scholar] [CrossRef]
  401. Ghosh, D.; Skinner, M.A.; Laing, W.A. Pharmacogenomics and Nutrigenomics: Synergies and Differences. Eur. J. Clin. Nutr. 2007, 61, 567–574. [Google Scholar] [CrossRef]
  402. Subbiah, M.T.R. Nutrigenetics and Nutraceuticals: The next Wave Riding on Personalized Medicine. Transl. Res. J. Lab. Clin. Med. 2007, 149, 55–61. [Google Scholar] [CrossRef] [PubMed]
  403. Wilkins, M.R.; Sanchez, J.-C.; Gooley, A.A.; Appel, R.D.; Humphery-Smith, I.; Hochstrasser, D.F.; Williams, K.L. Progress with Proteome Projects: Why All Proteins Expressed by a Genome Should Be Identified and How to Do It. Biotechnol. Genet. Eng. Rev. 1996, 13, 19–50. [Google Scholar] [CrossRef] [PubMed]
  404. Clish, C.B. Metabolomics: An Emerging but Powerful Tool for Precision Medicine. Mol. Case Stud. 2015, 1, a000588. [Google Scholar] [CrossRef]
  405. Michel, M.; Laser, K.T.; Dubowy, K.-O.; Scholl-Bürgi, S.; Michel, E. Metabolomics and Random Forests in Patients with Complex Congenital Heart Disease. Front. Cardiovasc. Med. 2022, 9, 994068. [Google Scholar] [CrossRef]
  406. Patient-Specific Modelling & Bioinformatics in PPPM. EPMA J. 2011, 2 (Suppl. S1), 181–187. [CrossRef]
  407. Steg, A.; Oczkowicz, M.; Smołucha, G. Omics as a Tool to Help Determine the Effectiveness of Supplements. Nutrients 2022, 14, 5305. [Google Scholar] [CrossRef] [PubMed]
  408. McClorry, S.; Slupsky, C.M.; Lind, T.; Karlsland Åkeson, P.; Hernell, O.; Öhlund, I. Effectiveness of Vitamin D Supplementation in Swedish Children May Be Negatively Impacted by BMI and Serum Fructose. J. Nutr. Biochem. 2020, 75, 108251. [Google Scholar] [CrossRef] [PubMed]
  409. von Arnim, C.A.F.; Dismar, S.; Ott-Renzer, C.S.; Noeth, N.; Ludolph, A.C.; Biesalski, H.K. Micronutrients Supplementation and Nutritional Status in Cognitively Impaired Elderly Persons: A Two-Month Open Label Pilot Study. Nutr. J. 2013, 12, 148. [Google Scholar] [CrossRef] [PubMed]
  410. Ortea, I. Foodomics in Health: Advanced Techniques for Studying the Bioactive Role of Foods. TrAC Trends Anal. Chem. 2022, 150, 116589. [Google Scholar] [CrossRef]
  411. Stouras, I.; Papaioannou, T.G.; Tsioufis, K.; Eliopoulos, A.G.; Sanoudou, D. The Challenge and Importance of Integrating Drug–Nutrient–Genome Interactions in Personalized Cardiovascular Healthcare. J. Pers. Med. 2022, 12, 513. [Google Scholar] [CrossRef] [PubMed]
  412. Saito, R.; Takeda, K.; Yamamoto, K.; Nakagawa, A.; Aoki, H.; Fujibayashi, K.; Wakasa, M.; Motoyama, A.; Iwadare, M.; Ishida, R.; et al. Nutri-Pharmacogenomics of Warfarin Anticoagulation Therapy: VKORC1 Genotype-Dependent Influence of Dietary Vitamin K Intake. J. Thromb. Thrombolysis 2014, 38, 105–114. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Bidirectional relationship of DNIs. Reproduced from Karadima et al. [9].
Figure 1. Bidirectional relationship of DNIs. Reproduced from Karadima et al. [9].
Nutrients 16 00950 g001
Figure 2. Selected DNIs with ASA. Created with biorender.com, accessed on 20 March 2024.
Figure 2. Selected DNIs with ASA. Created with biorender.com, accessed on 20 March 2024.
Nutrients 16 00950 g002
Figure 3. Selected DNIs with warfarin. Created with biorender.com, accessed on 20 March 2024.
Figure 3. Selected DNIs with warfarin. Created with biorender.com, accessed on 20 March 2024.
Nutrients 16 00950 g003
Figure 4. Drug Interaction Probability Scale. taken from Horn et al. [392].
Figure 4. Drug Interaction Probability Scale. taken from Horn et al. [392].
Nutrients 16 00950 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Renaud, D.; Höller, A.; Michel, M. Potential Drug–Nutrient Interactions of 45 Vitamins, Minerals, Trace Elements, and Associated Dietary Compounds with Acetylsalicylic Acid and Warfarin—A Review of the Literature. Nutrients 2024, 16, 950. https://doi.org/10.3390/nu16070950

AMA Style

Renaud D, Höller A, Michel M. Potential Drug–Nutrient Interactions of 45 Vitamins, Minerals, Trace Elements, and Associated Dietary Compounds with Acetylsalicylic Acid and Warfarin—A Review of the Literature. Nutrients. 2024; 16(7):950. https://doi.org/10.3390/nu16070950

Chicago/Turabian Style

Renaud, David, Alexander Höller, and Miriam Michel. 2024. "Potential Drug–Nutrient Interactions of 45 Vitamins, Minerals, Trace Elements, and Associated Dietary Compounds with Acetylsalicylic Acid and Warfarin—A Review of the Literature" Nutrients 16, no. 7: 950. https://doi.org/10.3390/nu16070950

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

Article Metrics

Back to TopTop