Next Article in Journal
Inhibition of Liquid–Liquid Phase Separation for Breaking the Solubility Barrier of Amorphous Solid Dispersions to Improve Oral Absorption of Naftopidil
Next Article in Special Issue
Effects of Mangiferin on LPS-Induced Inflammation and SARS-CoV-2 Viral Adsorption in Human Lung Cells
Previous Article in Journal
PLLA Composites Combined with Delivery System of Bioactive Agents for Anti-Inflammation and Re-Endothelialization
Previous Article in Special Issue
Gossypol and Its Natural Derivatives: Multitargeted Phytochemicals as Potential Drug Candidates for Oncologic Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oral Pharmacokinetics of Hydroxycinnamic Acids: An Updated Review

by
Kleyton Santos Veras
,
Flávia Nathiely Silveira Fachel
,
Bibiana Verlindo de Araújo
,
Helder Ferreira Teixeira
and
Letícia Scherer Koester
*
Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Avenida Ipiranga, 2752, Porto Alegre 90610-000, Rio Grande do Sul, Brazil
*
Author to whom correspondence should be addressed.
Pharmaceutics 2022, 14(12), 2663; https://doi.org/10.3390/pharmaceutics14122663
Submission received: 19 October 2022 / Revised: 22 November 2022 / Accepted: 25 November 2022 / Published: 30 November 2022
(This article belongs to the Special Issue Recent Advances in Natural Product Drugs)

Abstract

:
Hydroxycinnamic acids (HCAs) such as caffeic acid (CA), chlorogenic acid (CGA), coumaric acid (COA) isomers, ferulic acid (FA) and rosmarinic acid (RA) are natural phenolic acids with widespread distribution in vegetal foods and well-documented pharmacological activities. However, the low bioavailability of HCAs impairs their administration by the oral route. The present review addresses new findings and important factors/obstacles for their oral administration, which were unexplored in the reviews published a decade ago concerning the bioavailability of phenolic acids. Based on this, the article aims to perform an updated review of the water solubility and gastrointestinal stability of HCAs, as well as describe their oral absorption, distribution, metabolism and excretion (ADME) processes by in vitro, ex vivo, in situ and in vivo methods.

1. Introduction

Hydroxycinnamic acids (HCAs) are a large class of natural phenolic compounds that present cinnamic acid as the basic chemical structure. The HCA class originates from the hydroxylation, methylation, or esterification of cinnamic acid with quinic acid or 3,4-dihydroxyphenillatic acid and is represented by caffeic acid (CA), chlorogenic acids (CGAs), meta (m-), ortho (o-) and para (p-) coumaric acid (COA), ferulic acid (FA) and rosmarinic acid (RA) (Figure 1). CGAs can be subdivided into caffeoylquinic acids, feruloylquinic acids and dicaffeoylquinic acids, of which 5-caffeoylquinic acid is frequently referred to as the main CGA [1,2,3].
HCAs have a wide distribution in the food consumed globally, including coffee, food herbs, red wine and whole grains, which have been related to improving the quality of health [4,5,6,7,8]. Additionally, their isolated forms have gained interest in the pharmaceutical field due to their countless pharmacological activities demonstrated by in vitro and in vivo studies, such as analgesic, antibacterial, anti-cancer, anti-diabetic, antifungal, anti-hyperlipidemic, anti-hypertension, anti-inflammatory, antimutagenic, anti-obesity, antioxidant, anti-tyrosinase, immunomodulatory, neuroprotective and photoprotective. On the other hand, their positive effects after oral intake depend upon their bioavailability in the body [5,7,9,10,11,12,13].
Reviews reporting the oral bioavailability of HCAs are available in the literature. Despite the richness of outcomes related to these reviews, important factors for the overall oral bioavailability of HCAs, such as water solubility, stability and physicochemical properties are not fully explored in general. In addition, the reviews do not offer a clear division of the type of approaches applied in the studies, and new findings have been published in the last 10 years [13,14,15].
In this context, this review addresses studies related to aqueous solubility, stability and the oral absorption, distribution, metabolism and elimination (ADME) processes of HCAs evaluated by in vitro, ex vivo, in situ, and in vivo methods. The data are arranged separately when appropriate, according to the factor evaluated and the method applied, in order to create an easy assessment that could be used as a guide for future research targeting the oral administration of HCAs. Studies published before 2010 years have only been described where they are lacking in previously published reviews or where they contain relevant results for the current context.

2. Water Solubility of Hydroxycinnamic Acids (HCAs)

Water solubility plays a major role in the oral administration of drugs since it must be in solution to be absorbed in the gastrointestinal tract [16]. Drugs that present water solubility lower than 0.1 mg/mL are classified as poorly soluble according to Horter and Dressmann [17]. Based on the data ascribed in Table 1, all HCAs exhibited water solubility values higher than 0.1 mg/mL, characterizing them as water-soluble compounds. However, it is important to take into account that the strength of the highest drug product should be soluble in 250 mL or less of aqueous media over a pH range of 1 to 6.8 at 37 °C ± 1 °C for a drug substance to be considered highly soluble [18]. Unfortunately, HCAs are not considered pharmaceutical drugs and do not have a defined dose, preventing their Biopharmaceutical classification (BCS), but CA, CGA and RA have been investigated in clinical trials in doses of 300, 200–560 and 10–150 mg, respectively [19,20,21,22,23,24,25,26,27].
CA and FA are the HCAs with more available water solubility data, and some contrasting results have been reported for them. The respective water solubilities of CA and FA at pH 7.2 and 7.4 were stated to be 6.510 and 0.178 mg/mL, and 6.630 and 5.420 mg/mL, respectively, showing a difference of 36.57- and 1.22-fold for a narrow pH range [28,29]. Furthermore, Rastogi and Jana [28] related a higher water solubility for CA at pH 6.8 (0.188 mg/mL) than at pH 7.4 (0.178 mg/mL), when the opposite would be expected (Table 1) since water solubility is a pH-dependent physicochemical property that will increase with a rise in pH for acid compounds, such as HCAs, because of the ionization set by the pKa [30].
More pharmacokinetic and pharmacodynamic studies are necessary in order to establish the therapeutic doses of HCAs and classify them into the BCS to estimate the impact of their water solubility on oral bioavailability, which remains an open factor.
Table 1. Physicochemical properties of HCAs.
Table 1. Physicochemical properties of HCAs.
HCA MW
(g/mol)
Water Solubility
(mg/mL) a
pKaLog PLog D
(pH 7.4)
PSA (Å)HBD HBA RB References
CA 180 0.178 (pH 7.4)
0.188 (pH 6.5)
0.300 (25 °C)
0.420 (pH 3)
0.550 (pH 3.42/15 °C)
0.980 (pH 3.37/25 °C)
1.230 (pH 3.34/30 °C)
1.770 (37 °C)
2.040 (pH 3.25/40 °C)
2.920 (pH 3.17/50 °C)
6.510 (pH 7.2)
4.36–4.70
7.60–9.46
11.17–11.85
0.93–4.60 −1.78–1.7477.75–81.003 4 2 [28,29,31,32,33,34,35,36,37,38,39,40,41]
CGA 354 40.000 (25 °C) 3.33
7.8–8.26
−0.45 −3.91–3.57164.746 9 5 [31,40,42,43,44,45,46]
m-COA 164 1.540 (25 °C) 4.48–4.60
10.35–10.39
1.83 −1.34 57.53 2 b 3 b 2 b [38,39,47,48,49]
o-COA 164 0.490 (25 °C) 4.00–4.13
9.58–9.60
1.5 N.A. 57.53 2 3 2 b [47,48,49,50]
p-COA 164 0.344 (25 °C)
0.700 (37 °C)
4.36–4.70
8.84–9.92
1.43–4.45 −1.32 56.20–57.50 2 3 2 [32,33,38,40,47,51,52]
FA 194 0.333 (25 °C)
0.454 (25 °C)
0.570 (pH 3.54/15 °C)
0.710 (pH 3)
0.780 (pH 3.46/25 °C)
0.920 (pH 3.40/30 °C)
0.950 (pH 3.40/30 °C)
1.490 (37 °C)
1.760 (pH 3.31/40 °C)
2.190 (pH 3.18/50 °C)
5.420 (pH 7.4)
6.630 (pH 7.2)
4.58–4.96
9.68–9.99
1.25–4.12
−1.38–1.23
66.802 4 3 [29,32,33,36,38,39,40,41,50,51,52,53,54]
RA 360 1.800 (pH 1.2/ 25 °C) 2.92
8.36
9.56
10.62
1.63–1.78 −2.45 144.52–145.005 8 7 [39,55,56,57,58]
a Some studies did not relate the pH or temperature used in the solubility assay. b Data not available in the literature and obtained from SwissADME software [59]. N.A. not available in the literature or SwissADME software. MW: Molecular weight. Log P: partition coefficient. log D: distribution coefficient. PSA: polar surface area. HBD: number of hydrogen bond donor. HBA: number of hydrogen bond acceptor. RB: rotatable bonds.

3. Stability of Hydroxycinnamic Acids (HCAs)

Stability in simulated gastrointestinal medium and plasma are assays that allow the loss of content catalyzed by the environmental conditions to be estimated [60,61]. Simulated gastrointestinal stability studies for CA and RA, in isolated form, were performed in two phases: gastric and intestinal [62,63,64,65,66]. The studies demonstrated that CA and RA present a content recovery higher than 98% after the gastric phase, while the recovered fraction decreased to 46–75% and 69–75%, respectively, in intestinal conditions [62,64]. CGA exhibited stability of 48% after all simulated gastrointestinal assays [67]. The lower content of CA, RA and CGA after the gastrointestinal assay was ascribed to the low chemical stability of phenolic compounds in the alkali medium of the intestinal phase [62,64,67]. FA is stable in the gastric environment [68]; nevertheless, there are no reports about its stability in intestinal conditions. The substantial loss of content could be the first obstacle for the oral delivery of HCAs.
Conversely, Ren et al. [42] showed that the gastric and intestinal phases chemically affect the stability of CGA, while Costa et al. [65] and Zoric et al. [63] reported a significant loss of RA in the gastric phase when compared with the intestinal phase. The effect of other compounds in the digestive solution on the stability of CGA was also investigated by employing a mixture of three phenolic compounds plus CGA. The CGA content in the mixture composed of cyanidin 3-rutinoside, quercetin-3-rutinoside, catechin and CGA was 94.9%, while for the mixture composed of quercetin, quercetin 3-O-glucoside, rutin and CGA had a value of 11.5% [69,70], suggesting that the matrix had an influence on the stability of CGA.
In addition to evaluating the stability of compounds, the simulated gastrointestinal medium was also employed to analyze the free fraction of CA, CGA and FA from food, since they can interact chemically with amino acids, peptides and proteins. Simulated proteolytic digestibility revealed a low release of HCAs from proteins, a fact that decreases the availability of their free forms for absorption [71].
Stability in plasma is crucial to maintain the drug in a desirable concentration in the body and to promote its pharmacological effect [61]. The stability of CGA in rat and human plasma and phosphate buffer (pH 7.4) solution at 37 °C revealed the formation of a substance with a similar fragmentation pattern to CGA, identified as its isomer. The pH-dependent isomerization reaction of CGA is well-described in the literature; thus, it is more probable that its isomerization was due to the alkali pH rather than an enzymatic reaction [72,73]. RA was stable in rat plasma and phosphate buffer at 37 °C, while a de-esterification reaction occurred in human plasma, forming CA (<4%) [72].
Despite the absence of specific plasma stability analyses for CA, p-COA and FA in the literature, bioanalytical validation studies indicated that CA was 70% stable in rabbit plasma for 24 h at 25 °C [74], and p-COA and FA were stable for 4 h and 24 h, respectively, in rat plasma at room temperature [75,76]. Studies relating to the gastrointestinal stability of COA isomers and the plasma stability of m- and o-COA isomers were not found in the literature.

4. Absorption, Distribution, Metabolism and Elimination (ADME) of Hydroxycinnamic Acids (HCAs)

4.1. In Vitro Studies

4.1.1. Absorption

In vitro methods are extensively used in the screening of drugs, aiming to elucidate their oral absorption mechanism and the fraction captured or permeated, presenting advantages over in vivo methods, such as lower cost and faster analysis. Artificial membrane-based and cell culture-based methods are among the in vitro methods used, such as the parallel artificial membrane permeability assay (PAMPA), and 2/4/A1, Caco-2, IEC-18, HT29-MTX, MDCK, T7 cell lines, and Caco-2/HT29-MTX, Caco-2/Raji B, Caco-2/HT29/Raji B co-cultures, respectively [77,78,79].
PAMPA is a method that mimics the transcellular passive intestinal permeability of drugs and enables their diffusion in different types of lipids and pH to be estimated [80]. Apparent permeability coefficient (Papp) values higher than 10.0 × 10−6 cm/s, between 5.0 and 10.0 × 10−6 cm/s and lower than 5.0 × 10−6 cm/s classify a drug as highly, moderately and slowly absorbed, respectively [28].
The PAMPA of CA at three concentrations indicated a Papp value lower than 4 × 10−6 cm/s [28]. In another study, an even lower Papp value, expressed as the logarithm of Papp (log Papp = −5.52 or Papp~3 × 10−6 cm/s), was found for CA, CGA and RA in the pH range 4.0–8.0. These Papp values mean that less than 1% of each HCA was transported toward the receptor compartment [81,82,83]. A higher transport rate was only observed for FA at pH 4.0, for which the log Papp was −4.77 (Papp~17 × 10−6 cm/s); over pH 4.0, the Papp values were equal to the other HCAs [81,82].
Transcellular passive diffusion is a process affected by the physicochemical properties of the drug, including the molecular weight (MW), partition coefficient (log P), distribution coefficient (log D), number of hydrogen bond donors (HBDs) and acceptors (HBAs), pKa, number of rotatable bonds (RBs) and polar surface area (PSA) [77,81,82,84]. Based on the different physicochemical properties, some drug oral absorption prediction rules are used in medicinal chemistry research and the pharmaceutical industry, such as the rules of Egan et al. [85], Ghose et al. [86], Lipinski et al. [87], Muegge et al. [88], Oprea [89], Veber et al. [90] and Zmuidinavicius et al. [91], and the HCAs violate them in one or more parameters (Figure 2).
CGA violates all prediction rules, displaying the highest number of violations, followed by RA > CA > FA. FA appears to be the most favorable HCA to permeate by transcellular passive diffusion, which is visualized at pH 4.0 by PAMPA at least. Van de Waterbeemd et al. [92] affirmed that a drug with log D higher than one unit at pH 7.4 (pKa-dependent property) is more easily transported by transcellular passive diffusion. In this sense, the higher Papp of FA over the other HCAs is justified, since it presents the highest first pKa and log D 7.4 values (Table 1). The COA isomers demonstrate violations in the prediction rules; however, there are no studies in the literature about their transcellular passive diffusion.
It is important to point out that the oral absorption of a drug is the sum of passive and carrier-mediated influx and efflux transporters [93]. In this context, the in vitro cell culture-based techniques are more adequate to evaluate the absorption of drugs, since they express influx and efflux transporters. Aside from that, they allow the intestinal and hepatic metabolism by phase 1 and 2 metabolic enzymes to be estimated [94,95].
Permeability studies in the Caco-2 cell line demonstrated that FA, m- and p-COA in the presence of an inward-direct proton gradient are transported by the transcellular mechanism through monocarboxylate transporters (MCT), with a linear concentration-dependence; in the absence of this proton gradient, their transport occurs by passive diffusion [96,97,98]. CA, CGA and RA are mainly transported by the paracellular mechanism, since they do not have a polarised flux. However, it has been shown that the transport of CA and RA also occurs, to a lesser extent, by transcellular mechanisms through MCT and organic anion-transporting polypeptide (OATP) transporters [59,99,100].
The intracellular accumulation of the HCAs was <2%, and the rate of transport, defined as the amount permeated toward the basolateral side, followed the order: FA (3.42–30.52%) > m-COA (1.34–13.74%) > p-COA (1.55–10.87%) > CA (0.10–1.57%) > RA (0.03–1.30%) > CGA (0.10–0.30%) [59,96,97,98,99,101,102]. The results expressed as Papp (apical → basolateral) (cm/s) were similar: FA (10.0 × 10−6) > o-COA (6.0 × 10−6) > m-COA = p-COA (5.0 × 10−6) > CA (1.40–2.12 × 10−6) > CGA (0.38–0.86 × 10−6) > RA (0.20–0.86 × 10−6). These data indicate that FA and COA isomers show the highest permeability when compared to CA, CGA and RA. Variations in the amount permeated and Papp is caused by differences in the experiment time or the presence or absence of a proton gradient [28,48,102,103,104,105]. Recently, Mortelé et al. [106] reported a Papp of 2.42 × 10−6 cm/s for CGA, which is substantially higher than the other values found for this HCA.
Studies performed with vegetal matrices containing CA, p-COA and RA showed an increase in their rate of transport, while a negative effect was reported for CGA. The vegetal matrices were composed of numerous substances that have different degrees of affinity by the transporters, which could change the permeability of the HCAs [45,100,102,107,108].
The transport of CA, CGA, p-COA and FA was also investigated in other cell lines [45,109,110]. CGA in gastric cells presented an absorption twice as high as in Caco-2 cells, a finding associated with the acid pH of the medium [45]. The paracellular transport is influenced by the charge state of the drug, in which the uncharged state is desirable [111]. Based on the pKa of CGA (Table 1), the acid pH (pH 3.0) of gastric cells maintained more than 50% of CGA in the uncharged state, whereas more than 50% of CGA would be in a negatively charged state in Caco-2 cells (pH 7.4).
In a Caco-2/HT29-MTX co-culture, a higher amount of FA permeated in the presence of a proton gradient. The transport was linear and independent of concentration, a characteristic of the transcellular passive diffusion mechanism [109], contrasting with the outcomes reported by Konishi and Shimizu [96]. Additionally, in the T84 cell line, among CA, CGA, p-COA and FA, only FA was detected on the basolateral side, demonstrating that it is the most permeable HCA and confirming the results shown by the PAMPA and Caco-2 cell methods [81,82,96,110].
Efflux transporters, mainly P-glycoprotein (P-gp), multidrug resistance-associated protein 2 (MRP2) and breast cancer resistance protein (BCRP), also have a significant contribution with respect to drug transport [112]. The efflux ratio was lower than two for CA, CGA, FA and RA, suggesting that they are not substrates for efflux transporters [103,105]. Nonetheless, for CGA, this hypothesis is refuted by the use of P-gp, MRP2 and BCRP inhibitors that improve its absorption [106,113]. In addition, CA and RA showed a concentration-dependent inhibitory or inducer activity on P-gp, MRP2 and BCRP [114,115,116,117,118], while FA expressed only an inhibitory activity on P-gp [119,120].

4.1.2. Metabolism

Cytochrome P450 isoenzymes (CYP), catechol-O-methyltransferase (COMT), UDP-glucuronosyltransferases (UGTs) and sulfotransferases (SULTs) are the main enzymes present in the cells of the body responsible for drug metabolism. Enzymes present in Caco-2 cells metabolized CA to CA glucuronide, CA methyl-glucuronide, CA sulfate, FA and isoferulic acid. CGA underwent isomerization, hydrolysis (formation of CA), sulfonation and methylation (formation of feruloylquinic acid). p-COA was very stable, presenting just traces of its glucuronidated, methylated and sulfated forms. FA produced FA glucuronide, FA sulfate and dihydroferulic acid. These similar metabolites were also detected in the Caco-2/HT29-MTX co-culture and T84 cell line. RA underwent isomerization, hydrolysis (formation of CA) and methylation. The appearance of HCA metabolites after incubation with Caco-2 cells reveals that the intestine would be the first site of their metabolism. Quantitatively, the free forms of CA (69.56–94.6%), CGA (73.1–94.7%), p-COA (~100%), FA (~95.50–95.62%) and RA (88.25%) remained at a higher concentration than the metabolites [101,104,109,110,121,122,123].
Data about the metabolism of HCAs in HepG2 cell are available for CA, CGA, FA and RA. The free HCAs were the predominant forms (>63.8%); compared to the Caco-2 cells, extra metabolites were only detected for RA, which was identified as RA glucuronide, RA methyl-glucuronide and FA [122,123]. Moreover, liver microsomes produced 14 metabolites from RA, which were glutathione conjugates and glucuronidated forms [124]. The incubation of CA and FA with isolated hepatocytes and liver microsomes revealed the formation of dihydrocaffeic acid, dihydroferulic acid and CA glutathione conjugates [125,126]. Additionally, in human intestinal and liver S9 homogenates, the predominant pathway of metabolization for CA and FA was sulfation (>95%), while glucuronidation was extremely low [127].
The cell culture-based methods identified a large number of HCA metabolites, which sometimes differed qualitatively and quantitatively, demonstrating that they express distinct metabolization pathways [121,122,123,125,127]. Regardless of the lack of homogeneity in some results, the in vitro methods remain the first choice in elucidating the mechanism involved in the absorption and metabolism of drugs. Figure 3 illustrates a scheme for the oral absorption of RA based on the gastrointestinal stability, Caco-2 cell permeability and HepG2 cell metabolism studies, predicting a bioavailable fraction lower than 1% [62,102,122].
Microbial metabolism in the intestinal tract also has a considerable effect on the available fraction for absorption when the drugs are administered by the oral route. The in vitro fermentation of CA and CGA carried out with human fecal microbiota, revealed that the HCAs evaluated over a time interval of 0.5–2 h were undetectable and the metabolite 3-hydroxyphenylpropionic acid (3-HPPA) was identified for both. The maximum concentration (Cmax) of 3-HPPA was reached at 2 h; afterward, it was also completely degraded [128,129]. Another study reported that the 6 h incubation of CGA with human fecal microbiota produced 11 metabolites; among them, dihydroxycaffeic acid, dihydroxyferulic acid and 3-HPPA comprised 75–83% [130].
FA incubated with rat fecal microbiota apparently had a slower degradation during 48 h, forming two metabolites: dihydroxyferulic acid and 3-methoxyl-4-hydroxybenzenepropanoic acid [131]. For RA, the probiotic strain Lactobacillus johnsonii isolated from the human intestinal microbiota completely hydrolyzed the RA into CA and 3,4-dihydroxyphenyllactic acid after 4 h [132].
In summary, the in vitro PAMPA and Caco-2 cell experiments indicated that the physicochemical properties of HCAs can limit their absorption across the gastrointestinal membranes, with FA being the most absorbed compound among the HCAs. The effect of efflux transporters on the total absorption of CGA was also confirmed, in addition to its physicochemical properties. On the other hand, microbiota, intestinal and hepatic metabolisms could considerably reduce the systemic amount of HCAs.

4.2. Ex Vivo Studies

Compared to the in vitro assays, ex vivo methods provide more distinctive features of the intestinal tissue structure [133]. The rate of transport of CA, CGA and p-COA in the porcine intestinal mucosa (cecal pole) was 3.7, 1.9 and 3.3%, respectively [134]. In the porcine jejunal segment, the amount of CA and CGA absorbed was lower than 1.5%. For CGA, the transport kinetic was linear and non-saturable, which is consistent with the data from in vitro methods [99,135]. The recovery of FA in the rat ascending or descending colon was statistically similar, reaching values of 4.66% and 4.76%, respectively [109].
Only CGA and p-COA transport were considered in the different segments of the gastrointestinal tract employing the same tissue source, with CGA being more permeable in the duodenum than in the ileum, jejunum and colon, while p-COA was transported more in the jejunum and ileum than in the cecum, colon and stomach [136,137]. When the rate of transport was analyzed in tissues from different animal species and intestinal segments, the results were not consistent. This is exemplified by CA, which showed greater transport in porcine ileum, whereas the highest rate in rats was found in the jejunum [134,135,138].
An increase in CGA transport was achieved due to the synergic action of the extract components [137] and the use of a P-gp inhibitor [135]. The transport of RA contained in a vegetal matrix was evaluated in rat jejunum mucosa, demonstrating an increase in transport with an increase in the vegetal matrix concentration. Nonetheless, neither the initial RA content nor the relative amount transported is reported [139].
With regard to metabolism, the glucuronidated form of CA was found to be in a higher concentration than the free form [138]. CGA was metabolized to its isomers and CGA glucuronide, and also hydrolyzed to CA [135]. p-COA did not present any metabolites, while FA produced the same metabolites as described in the in vitro studies, Section 4.1.2 [109,138]. There are no studies in the literature reporting the use of ex vivo methods to evaluate the absorption of m- and o-COA isomers.
The quantitative data obtained from ex vivo methods are not comparable, due to the use of different animals and tissue types; most of the studies did not reveal the amount transported relative to the initial dose. Despite this, from the available data, the highest absorption of FA and metabolization of HCAs was perceived in accordance with the in vitro methods.

4.3. In Situ Studies

In situ methods offer advantages such as an intact gastrointestinal mucosa, and the presence of a nervous system and blood flow, which are not possible to obtain in the in vitro-based cell and ex vivo studies, getting closer to the in vivo mechanisms [133]. In order to avoid the use of the term recurrent, all in-situ gastric absorption studies described in this section were carried out with the pylorus ligation.
From the gastric route, 89.7% of free CA was measured in the portal vein after 5 min of administration, revealing its fast and considerable absorption. A decline in the concentration of free CA was observed in the abdominal aorta (39.1%), which indicated that CA underwent metabolism in the liver [140]. In contrast, its perfusion in the duodenal and jejunal plus ileal segments showed that only 12.4% and 19.5% of CA was absorbed, respectively, displaying a lower absorbed fraction when compared to the gastric assay, regardless of hepatic metabolism [141,142]. The identification of FA and isoferulic acid in the perfusion samples provides evidence of the metabolism of CA in the passage through the intestinal membranes [142].
Studies for CGA demonstrated that its absorption is independent of concentration, a characteristic of substances transported by passive mechanisms [42,99,137]. When administered in the stomach, 9.43% and 4.57% of CGA were recovered in the gastric vein and aorta in free form, respectively, without the detection of metabolites [143]. Konishi et al. [140] confirmed its low gastric absorption and the reduced concentration in the abdominal aorta, evidencing its hepatic metabolization. In addition, 8% was absorbed through CGA perfusion in the jejunal plus ileal segment, and isoferulic acid was identified as a product of its intestinal metabolism [142].
The rate of absorption of CGA was very close in the stomach and intestine: the lack of significant difference between them was posteriorly demonstrated by Ren et al. [42], who reported the respective concentrations of 7.7% and 7.9% for the gastric and intestinal experiments. When assessing the degree of permeability according to intestinal segments, the duodenum was found to be more permeable than the ileum, in accordance with the ex vivo assay [137].
The gastric administration of p-COA promoted the absorption of 73.3% in the portal vein and 57.5% of the remaining content in the abdominal artery in the free form. In contrast to the other HCAs, the decrease in the free form was accompanied by the metabolized forms, but the reduction in the latter occurred to a lesser extent [140]. Intestinal in situ experiments were not found for p-COA.
Two in situ gastric studies of FA demonstrated its high absorption rate in the free form [68,140]. The amounts collected in the portal vein in the free, glucuronidated, sulfated and sulfoglucuronidated forms were 49.2%, 2.9%, 2.6% and 43.1%, respectively. However, a decrease in the FA-free form (6.2%) was observed in the samples from the celiac artery, while the FA-metabolized forms increased (93.8%), a factor derived from its hepatic metabolism and biliary excretion [68]. This was corroborated by Konishi et al. [140] who reported the presence of 62.8% free FA in the portal vein, while its concentration fell to 18.4% in the abdominal artery. Based on the free FA collected in the urine (4.6%), probable renal metabolism would also be responsible for the concentration decline [68]. It is important to highlight that these two studies revealed a 2.97-fold difference in the amount of free FA after hepatic metabolism [68,140].
In the intestinal perfusion, 60% of FA detected in the mesenteric vein was in the free form and 40% was in the metabolized form. A 2.4-fold reduction was observed in the sample collected in the abdominal aorta, assuring hepatic metabolization [144]. Comparing these results with those reported previously indicates that the absorption of FA is higher in the intestine than in the stomach, and the intestine is the first site of metabolism, followed by the liver [68,140,144].
FA contained in a semipurified sample was transported in the jejunal plus ileal segment in a manner directly proportional to its perfused concentration, presenting a non-saturable mechanism [145]. The rate absorbed in the intestinal segment was 56.1%, and no metabolites were detected after intestinal passage, in contrast with the data previously described. Nevertheless, after metabolism in the liver, no free FA was found in the bloodstream [138,144,145]. This section does not discuss the m- and o-COA isomers and RA due to the lack of studies in the literature.
The analysis of all in situ data showed that the free fraction of p-COA (57.5%) was superior to CA (39.1%), FA (6.2–18.4%) and CGA (4.57%) after hepatic metabolism, denoting the impact of metabolization on the bioavailability of HCAs.

4.4. In Vivo Studies

Reviews reporting the oral pharmacokinetics of HCAs within vegetal matrices are already available in the literature [14,15]. Based on this and the finding that other compounds can modify the pharmacokinetic processes of HCAs, as seen in the in vitro and ex vivo assessments, only studies with isolated HCAs were selected for review in this section. The in vivo pharmacokinetic parameter data are provided in Table 2.

4.4.1. Absorption

The in vivo pharmacokinetic data revealed that the HCAs presented rapid absorption after oral administration, with absorption t1/2 and Tmax ranging between 0.07 and 0.08 h and 0.03 and 1.5 h, respectively (Table 2). For the [3–14C] CA, the highest plasma radioactivity was visualized at 0–1 h [163]. The short time to reach the Cmax suggests that HCA compounds, in part, are absorbed in the gastric environment. In fact, the in situ gastric studies described above indicated that more than 60% of CA, p-COA and FA were transported to the portal vein after 5 min [140].
The rate of absorption obtained from the area under the curve (AUC)/dose relationship increased in the following order: CGA < RA < p-COA < FA < CA (Table 2). This order demonstrates that CA was more absorbed than FA and p-COA, data that were not observed in the in vitro, ex vivo and in situ evaluations [96,99,138,142,144]. The measure of AUC is affected by several factors, including the blood-sampling site. A comparative study revealed a significant increase in the AUC value for blood samples collected from the carotid artery compared to the caudal vein [164]. The respective sampling sites that presented the highest AUC for CA and FA in pharmacokinetic studies were the carotid artery and the caudal vein [146,158], a fact that, in part, could justify the higher rate of absorption exhibited for CA. Furthermore, the AUC for p-COA and FA was not extrapolated to infinite time [153,158]. The AUC/dose relationship based on the study by Kim et al., (2020) [155] was excluded as the dose was not defined by the weight.

4.4.2. Distribution

The distribution of a drug through the body is dependent on its physicochemical properties and plasma protein binding [165]. Among the HCAs with the available volume of distribution (Vd) data (Table 2), FA presented the highest value, suggesting a wide distribution [158]. In fact, FA exhibited a two-compartmental pharmacokinetic profile, denoting a distribution phase prior to elimination [156]. The Vd values for CA, CGA and RA were clearly lower than FA but also indicated a tissue distribution, since the values were superior to the real animal body volume. Additionally, the t1/2α of CA and FA revealed that their distribution is slower than their absorption when orally administered (Table 2) [141,146,149,156,160,166].
Considering the physicochemical characteristics given in Table 1, FA is the most lipophilic HCA and its plasma binding protein value (73.5%) is higher than CA (66%) and CGA (25.6%) [150,167]. Together, these data reveal that adequate physicochemical properties are more important to the partition toward tissues than only the plasma-free fraction for HCAs. The lower lipophilicity and higher binding protein value (91.4%) of RA could explain its smaller distribution compared to FA [168].
The specific tissues reached by CA were the kidneys, liver, muscles, lungs, heart, spleen and testes. The kidney (3.2%) and liver (0.3%) showed the highest concentration of CA, while the rate of CA was equal to or lower than 0.1% for the other tissues [163]. De Oliveira et al. [151] described CGA as being distributed through the kidney, liver and muscles, but quantitative analyses were not performed. Broader and quantitative tissue distribution of CGA was carried out by Chen et al. [152], who observed a decrease in the AUC in the following order: liver > kidney > heart > spleen > lung. For FA, the AUC of the kidney represented 76% of its tissue distribution. Minority AUCs were measured in the liver, lung, heart, spleen and brain [158]. Oral pharmacokinetic studies for p-COA and RA did not describe their tissue distribution.

4.4.3. Metabolism

The metabolism of HCAs is catalyzed mainly by hydrolysis and conjugation by COMT, UGTs and SULTs [148,151,158,159,161,163,169]. A drastic decrease in the AUC and Cmax measured from the portal vein and the abdominal artery was observed for CA, p-COA and RA. The reduction in free RA was accompanied by an increase in its metabolites, indicating additional metabolism by the liver. On the other hand, the concentration of free and metabolized forms of CA and p-COA declined simultaneously [147,153].
CA was metabolised to CA 3- or 4-O-glucuronide, CA 3- or 4-O-sulfate, CA sulfoglucuronide, FA, FA 4-O-glucuronide, FA 4-O-sulfate, isoferulic acid and isoferulic acid 3-O-sulfate [148,163,169]. The plasma concentrations of free FA and isoferulic acid were lower than free CA, while its glucuronidated (26 µM) and sulfoglucuronidated (~12 µM) forms had higher plasma concentrations than free CA (1.2 µM) [148,169]. These quantitative results were shown in the ex vivo studies and in the majority of the in vitro assessments [122,123,138].
The CGA metabolites present in the plasma were CGA glucuronide, CA, CA glucuronide, hippuric acid, FA sulfate, feruloylquinic acid isomers and isoferulic acid sulfate. The plasma Cmax value for the metabolites was 2-fold lower than the free CGA. Other metabolites were also detected in the tissues, such as CA sulfate, dihydrocaffeic acid, FA, FA sulfoglucuronide and isoferulic acid sulfoglucuronide [151]. The preceding pharmacokinetic evaluation did not detect the presence of CGA in the plasma, even in a higher dose, and only three CGA metabolites were identified: CA glucuronide, CA sulfoglucuronide and FA sulfoglucuronide [169]. Compared to the in vitro and ex vivo studies, the in vivo assays distinguished a higher number of metabolites, but the presence of CGA isomers was not reported in either the plasma or the tissues [104,123,138,151].
FA glucuronide and FA sulfate were the metabolites found for FA, while CA, FA and m-COA were detected in the plasma for RA, as well as their glucuronidation, sulfation and methylation products [158,159,161,170]. Most of the FA was detected in its sulfated forms, which is also described for the human intestinal and liver S9 homogenates and Caco-2 cells, while FA glucuronide was the metabolite found in the highest concentration in HepG2 cells [101,109,121,122,123,127,158]. For RA, the ratio between the AUC of the glucuronidated/free RA forms was 3.2, indicating that more than 75% of RA in the bloodstream was metabolized, while its hydrolysis occurred to a lesser extent [159,161,162]. Indeed, the experiments in the Caco-2 cells, HepG2 cells and liver microsome showed the glucuronidation and hydrolysis of RA, but the concentration of metabolized RA was considerably lower than the free RA [122,124,161].
The metabolites found in the in vivo evaluations are qualitatively supported by the in vitro, ex vivo and in situ assays. The lack of one or other metabolite can be seen when comparing the studies, which can probably be related to the fact that full identification of the HCA metabolites was not the main objective of the studies.

4.4.4. Elimination

Drug elimination is divided into two main processes: biotransformation and excretion. Biotransformation is the process in which the drug is converted to a metabolite, while excretion is the removal of the drug in the intact state or free form. The pharmacokinetic parameters that describe drug elimination are clearance, t1/2 and t1/2β [165]. Based on these parameters, it is possible to affirm that the HCAs have a short elimination when orally administered (Table 2), which is consistent with their elevated distribution into elimination organs [152,158,163].
The rate of urinary excretion for free CA, p-COA and FA was higher in the first 6 h of urine collection, reaching 10.2%, 17.4% and 3%, respectively, after 48 h. CGA in its free form had a negligible urinary excretion of 0.04% in the same period [171]. On the other hand, RA was excreted more at the later collection points (8–18 h), showing a total rate of 0.44% [159]. Studies carried out for a reduced time (8–24 h) exhibited rate values even higher than the 48 h study for CA (9.9%), CGA (0.07–0.499%), p-COA (24%) and FA (5.4%) [136,150,151,163,172].
The renal clearance of CA (0.0157 L/h kg) and CGA (0.147–0.292 L/h kg) demonstrated that CA is eliminated basically by biotransformation, while the hepatic extraction ratio would be responsible for 23.1–28.2% of CGA elimination [146,150]. The concentration of HCA metabolites found in the urine was higher than their unchanged forms. The CA metabolites were structurally similar to those identified in the plasma [163]. For p-COA, its conjugated forms were detected without distinguishing types, while the RA metabolites recovered in urine were CA, CA glucuronide and sulfate, m-COA, m-COA glucuronide and sulfate, RA glucuronide and sulfate, methyl-RA, methyl-RA glucuronide and sulfate, FA, and FA glucuronide and sulfate [159,171].
Interestingly, even though the plasma concentration of free CGA was greater than its metabolites and it had low hepatic excretion, the ratio between the metabolites and the free form of CGA was 121, implying that CGA underwent renal metabolism. In addition, extra CGA metabolites were reported in the urine than in the plasma, such as free isoferulic acid, m- and p-COA, and 3-HPPA [68,150,151,171]. For FA, the urinary excretion of FA conjugates achieved a rate of 70.2%, of which FA glucuronide represented 5.1% [171,172].
The total urinary excretion, the sum of free and metabolized forms relative to the dose was higher for FA (73.2%) than CA (61.6%), p-COA (54.1%), RA (5.47%) and CGA (4.9%), demonstrating that FA was the most absorbed HCA, which is supported by the in vitro and ex vivo methods [159,171]. Nonetheless, its extensive biotransformation, illustrated by in situ and in vivo studies, decreases the bioavailable fraction, which is reflected in the low free concentration in plasma and urine. Taking only the free form of HCAs into account, p-COA exhibited the highest concentration in urine [68,171].

4.4.5. Absolute Oral Bioavailability

The absolute oral bioavailability data demonstrated that p-COA was the most bioavailable HCA, followed by CA, RA and CGA (Table 2), in accordance with the rate of urinary excretion [171]. No data were found in the literature for FA; however, based on the results of the in situ and in vivo experiments, it possibly presents an absolute oral bioavailability intermediate to CA and RA. The in vivo data for RA were close to that described in the in vitro studies (Figure 3) [160]. However, the in vivo loss of RA after liver biotransformation was extremely high compared to the in vitro approach [122,161].
A decline in absolute oral bioavailability with the elevation of the dose was related to RA (Table 2). Analysis of the AUCs per dose revealed a lack of proportionality, indicating that RA has a non-linear pharmacokinetic feature [160]. The existence of dose proportionality was assumed for all doses tested for CGA, although a considerable reduction in renal clearance occurred with the increase in dose [150].
Some considerations are necessary with respect to the approach selected to analyze the pharmacokinetic profiles. Fourteen studies used a non-compartmental approach [75,105,147,148,149,150,151,152,154,157,158,159,160,161], while three reported a bicompartmental behavior [141,146,156]. The adoption of different analysis approaches produces misleading pharmacokinetic data, promoting the heterogeneity of results in the literature, which could hamper their use for clinical purposes [165]. Overall, the in vivo studies verified that the HCAs are quickly absorbed, and widely distributed, with physicochemical properties directly affecting their distribution, undergoing massive metabolization, and presenting fast elimination and low oral bioavailability.

5. Strategies to Improve Oral Bioavailability

Although out of the scope of this review, it is worth mentioning that recent reviews have gathered the technological strategies employed to improve the bioavailability of RA and FA, which are the most studied HCAs in this field. However, only a few studies have addressed their oral administration and evaluated the performance of the strategy in vivo [173,174,175].
Zhang et al. evaluated the effect of a nanostructured lipid carrier and solid lipid nanoparticles on the oral bioavailability of FA against its aqueous form, demonstrating that both formulations were able to improve the Cmax and AUC of HCA. The in vivo performance of the nanostructured lipid carrier gave Cmax and AUC parameters that were 1.32-fold and 1.38-fold better than solid lipid nanoparticles, a fact that was associated with the different release behavior presented for both formulations [157].
The incorporation of FA in a self-microemulsifying drug delivery system increased the AUC 1.73-fold when compared to its administration in an aqueous form. Additionally, it was observed that the emulsified FA had a higher body distribution, even in the brain [158].
The formulation of RA in a phospholipid complex and phospholipid complex oil solution promoted an increase of 47% and 225% in the Cmax and 120% and 291% in the AUC, respectively, when compared with an aqueous RA solution [105]. Once again, the differences between the formulations were ascribed to the release, since the lower release of RA in the phospholipid complex oil solution decreased its pre-systemic metabolism and improved the systemic fraction.
Yang et al. demonstrated that the co-administration of RA with piperine, a glucuronidation inhibitor, increased the relative bioavailability of RA 1.24-, 1.32-, 2.02- and 2.26-fold in the presence of inhibitor at concentrations of 20, 40, 60 and 80 mg/kg, compared to the administration of RA [161].
With the exception of the data by Yang et al., [161] all technological strategies that have been explored to improve the oral bioavailability of FA and RA are lipid-based delivery systems. In general, these lipid-based systems promoted a higher bioavailability of HCAs by enhancing their stability in the gastrointestinal tract by modifying the release and improving the lipid solubility, facilitating their transport through the gastrointestinal mucosa and, to some extent, promoting lymphatic absorption [105,157,158].
Despite the available in vivo studies focusing only on one type of system, most of the technological approaches described in the reviews could be easily applied to oral administration, such as cyclodextrin complexes, solid dispersions, and polymeric nano and microparticles [173,174,175], demonstrating an unexplored field of research.

6. Conclusions

CA and FA are the HCAs with the most data in the literature describing factors and processes related to oral bioavailability by different approaches, while only a few results can be found for the m- and o-COA isomers. The majority of investigations in recent pharmacokinetic studies have involved CGA, p-COA and RA.
The data described in this review allow us to affirm that water solubility is apparently not a limiting step for the oral absorption of HCAs, but more studies are needed to better understand the effect of this factor on their oral bioavailability. On the other hand, a substantial loss of HCA content was observed in the gastrointestinal environment, which decreases the fraction available for absorption.
In vitro absorption by passive and carrier-mediated transporters was inexpressive for most HCAs and was influenced by their physicochemical properties, highlighting that the permeation across gastrointestinal membranes is another issue for their absorption. In addition to the absorption process, in vitro, in situ, ex vivo and in vivo methods showed the great impact of pre-systemic metabolism on the bioavailable fraction. The sum of all these factors is responsible for the clear low absolute oral bioavailability values found.
It is important to mention that the in vitro, ex vivo, in situ and in vivo methods, in general, showed a qualitative correlation in their results; however, some discrepancies were identified quantitatively. Additionally, each approach presents limitations and variables, requiring extreme care when interpreting the data.
Overall, the results described in this review highlight the main factors/obstacles that impact the oral administration of HCAs and can be a guide to the development of delivery systems to overcome the issues of HCAs and produce a more successful therapy. Some recent reviews have shown several technological approaches that could be applied to the oral delivery of HCAs, but lipid-based delivery systems and their in vivo oral administration were the main focus of the investigation, revealing new potential opportunities.

Author Contributions

K.S.V.: Conceptualization, Investigation, Writing—Original Draft, Visualization. F.N.S.F.: Writing—Review and Editing. B.V.d.A.: Writing—Review and Editing. H.F.T.: Writing—Review and Editing. L.S.K.: Conceptualization, Investigation, Writing—Review and Editing, Supervision, Funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior—Brasil (CAPES)—Finance Code 001, Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq—grant 317458/2021-3).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Razzaghi-Asl, N.; Garrido, J.; Khazraei, H.; Borges, F.; Firuzi, O. Antioxidant Properties of Hydroxycinnamic Acids: A Review of Structure-Activity Relationships. Curr. Med. Chem. 2013, 20, 4436–4450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Rocha, L.D.; Monteiro, M.C.; Teodoro, A.J. Anticancer Properties of Hydroxycinnamic Acids—A Review. Cancer Clin. Oncol. 2012, 1, 109–121. [Google Scholar] [CrossRef]
  3. Taofiq, O.; González-Paramás, A.M.; Barreiro, M.F.; Ferreira, I.C.F.R.; McPhee, D.J. Hydroxycinnamic Acids and Their Derivatives: Cosmeceutical Significance, Challenges and Future Perspectives, a Review. Molecules 2017, 22, 281. [Google Scholar] [CrossRef] [PubMed]
  4. Amoah, S.K.S.; Sandjo, L.P.; Kratz, J.M.; Biavatti, M.W. Rosmarinic Acid—Pharmaceutical and Clinical Aspects. Planta Med. 2016, 82, 388–406. [Google Scholar] [CrossRef] [Green Version]
  5. Pei, K.; Ou, J.; Huang, J.; Ou, S. P-Coumaric Acid and Its Conjugates: Dietary Sources, Pharmacokinetic Properties and Biological Activities. J. Sci. Food Agric. 2016, 96, 2952–2962. [Google Scholar] [CrossRef]
  6. Tajik, N.; Tajik, M.; Mack, I.; Enck, P. The Potential Effects of Chlorogenic Acid, the Main Phenolic Components in Coffee, on Health: A Comprehensive Review of the Literature. Eur. J. Nutr. 2017, 56, 2215–2244. [Google Scholar] [CrossRef]
  7. Magnani, C.; Isaac, V.L.B.; Correa, M.A.; Salgado, H.R.N. Caffeic Acid: A Review of Its Potential Use in Medications and Cosmetics. Anal. Methods 2014, 6, 3203–3210. [Google Scholar] [CrossRef]
  8. Kumar, N.; Pruthi, V. Potential Applications of Ferulic Acid from Natural Sources. Biotechnol. Rep. 2014, 4, 86–93. [Google Scholar] [CrossRef] [Green Version]
  9. Alagawany, M.; Abd El-Hack, M.E.; Farag, M.R.; Gopi, M.; Karthik, K.; Malik, Y.S.; Dhama, K. Rosmarinic Acid: Modes of Action, Medicinal Values and Health Benefits. Anim. Health Res. Rev. 2017, 18, 1–10. [Google Scholar] [CrossRef]
  10. Fachel, F.N.S.; Schuh, R.S.; Veras, K.S.; Bassani, V.L.; Koester, L.S.; Henriques, A.T.; Braganhol, E.; Teixeira, H.F. An Overview of the Neuroprotective Potential of Rosmarinic Acid and Its Association with Nanotechnology-Based Delivery Systems: A Novel Approach to Treating Neurodegenerative Disorders. Neurochem. Int. 2019, 122, 47–58. [Google Scholar] [CrossRef]
  11. Naveed, M.; Hejazi, V.; Abbas, M.; Kamboh, A.A.; Khan, G.J.; Shumzaid, M.; Ahmad, F.; Babazadeh, D.; FangFang, X.; Modarresi-Ghazani, F.; et al. Chlorogenic Acid (CGA): A Pharmacological Review and Call for Further Research. Biomed. Pharmacother. 2018, 97, 67–74. [Google Scholar] [CrossRef] [PubMed]
  12. Mancuso, C.; Santangelo, R. Ferulic Acid: Pharmacological and Toxicological Aspects. Food Chem. Toxicol. 2014, 65, 185–195. [Google Scholar] [CrossRef] [PubMed]
  13. Selka, A.; Moutombi, F.J.N.; Jean-François, J.; Touaibia, M. Hydroxycinnamic Acids and Their Related Synthetic Analogs: An Update of Pharmacological Activities. Mini-Rev. Med. Chem. 2021, 21, 1516–1544. [Google Scholar] [CrossRef] [PubMed]
  14. Lafay, S.; Gil-Izquierdo, A. Bioavailability of Phenolic Acids. Phytochem. Rev. 2008, 7, 301–311. [Google Scholar] [CrossRef]
  15. Zhao, Z.; Moghadasian, M.H. Bioavailability of Hydroxycinnamates: A Brief Review of in Vivo and in Vitro Studies. Phytochem. Rev. 2010, 9, 133–145. [Google Scholar] [CrossRef]
  16. Savjani, K.T.; Gajjar, A.K.; Savjani, J.K. Drug Solubility: Importance and Enhancement Techniques. ISRN Pharm. 2012, 2012, 195727. [Google Scholar] [CrossRef] [Green Version]
  17. Horter, D.; Dressman, J.B. Influence of Physiochemical Properties on Dissolution of Drugs. Adv. Drug Deliv. Rev. 1997, 25, 3–14. [Google Scholar] [CrossRef]
  18. United States Department of Health and Human Services; Food and Drug Administration. Guidance for Industry—Waiver of In Vivo Bioavailability and Bioequivalence Studies for Immediate-Release Solid Oral Dosage Forms Based on a Biopharmaceutics Classification System; Center for Drug Evaluation and Research: Silver Spring, MD, USA, 2017. [Google Scholar]
  19. United States National Library of Medicine. The Efficacy and Safety of Caffeic Acid for Esophageal Cancer (NCT03070262). Available online: ClinicalTrials.gov/show/NCT03070262 (accessed on 27 May 2022).
  20. United States National Library of Medicine. The Effects of Coffee Main Constituents (Caffeine and Chlorogenic Acid) Supplementation on Inflammatory, Metabolic Factors, Hepatic Steatosis and Fibrosis in None-Alcoholic Fatty Liver Patients with Type 2 Diabetes (NCT02929901). Available online: ClinicalTrials.gov/show/NCT02929901 (accessed on 27 May 2022).
  21. United States National Library of Medicine. Effect of Chlorogenic Acid on Patients with Impaired Glucose Tolerance (NCT02621060). Available online: ClinicalTrials.gov/show/NCT02621060 (accessed on 27 May 2022).
  22. United States National Library of Medicine. Caffeic Acid Combining High-Dose Dexamethasone in Management of ITP. Available online: ClinicalTrials.gov/show/NCT02556814 (accessed on 27 May 2022).
  23. United States National Library of Medicine. Dietary Polyphenols and Glycation in Renal Insufficiency (NCT02524938). Available online: ClinicalTrials.gov/show/NCT02524938 (accessed on 27 May 2022).
  24. United States National Library of Medicine. Evaluation of Effects of Green Coffee Bean Extract (GCE) on Physiological and Psychological Variables (NCT02388672). Available online: ClinicalTrials.gov/show/NCT02388672 (accessed on 27 May 2022).
  25. United States National Library of Medicine. Caffeic Acid Tablets as a Second-Line Therapy for ITP (NCT02351622). Available online: ClinicalTrials.gov/show/NCT02351622 (accessed on 27 May 2022).
  26. United States National Library of Medicine. Efficacy of Spearmint Tea in Relieving Osteoarthritis of the Knee (NCT01380015). Available online: ClinicalTrials.gov/show/NCT01380015 (accessed on 27 May 2022).
  27. United States National Library of Medicine. Mint Tea for the Treatment of Nasal Polyps (NCT00465543). Available online: ClinicalTrials.gov/show/NCT00465543 (accessed on 27 May 2022).
  28. Rastogi, H.; Jana, S. Evaluation of Physicochemical Properties and Intestinal Permeability of Six Dietary Polyphenols in Human Intestinal Colon Adenocarcinoma Caco-2 Cells. Eur. J. Drug Metab. Pharmacokinet. 2016, 41, 33–43. [Google Scholar] [CrossRef]
  29. Saija, A.; Tomaino, A.; Trombetta, D.; De Pasquale, A.; Uccella, N.; Barbuzzi, T.; Paolino, D.; Bonina, F. In Vitro and in Vivo Evaluation of Caffeic and Ferulic Acids as Topical Photoprotective Agents. Int. J. Pharm. 2000, 199, 39–47. [Google Scholar] [CrossRef]
  30. Box, K.; Comer, J. Using Measured PKa, LogP and Solubility to Investigate Supersaturation and Predict BCS Class. Curr. Drug Metab. 2008, 9, 869–878. [Google Scholar] [CrossRef]
  31. de Campos Buzzi, F.; Franzoi, C.L.; Antonini, G.; Fracasso, M.; Filho, V.C.; Yunes, R.A.; Niero, R. Antinociceptive Properties of Caffeic Acid Derivatives in Mice. Eur. J. Med. Chem. 2009, 44, 4596–4602. [Google Scholar] [CrossRef] [PubMed]
  32. Kfoury, M.; Landy, D.; Auezova, L.; Greige-Gerges, H.; Fourmentin, S. Effect of Cyclodextrin Complexation on Phenylpropanoids’ Solubility and Antioxidant Activity. Beilstein J. Org. Chem. 2014, 10, 2322–2331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Beneduci, A.; Furia, E.; Russo, N.; Marino, T. Complexation Behaviour of Caffeic, Ferulic and p-Coumaric Acids towards Aluminium Cations: A Combined Experimental and Theoretical Approach. New J. Chem. 2017, 41, 5182–5190. [Google Scholar] [CrossRef]
  34. Kfoury, M.; Geagea, C.; Ruellan, S.; Greige-Gerges, H.; Fourmentin, S. Effect of Cyclodextrin and Cosolvent on the Solubility and Antioxidant Activity of Caffeic Acid. Food Chem. 2019, 278, 163–169. [Google Scholar] [CrossRef]
  35. Cheng, J.C.; Dai, F.; Zhou, B.; Yang, L.; Liu, Z.L. Antioxidant Activity of Hydroxycinnamic Acid Derivatives in Human Low Density Lipoprotein: Mechanism and Structure-Activity Relationship. Food Chem. 2007, 104, 132–139. [Google Scholar] [CrossRef]
  36. Divakar, S.; Maheswaran, M.M. Structural Studies on Inclusion Compounds of β-Cyclodextrin with Some Substituted Phenols. J. Incl. Phenom. Mol. Recognit. Chem. 1997, 27, 113–126. [Google Scholar] [CrossRef]
  37. Im, N.R.; Kim, K.M.; Young, S.J.; Park, S.N. Physical Characteristics and in Vitro Skin Permeation of Elastic Liposomes Loaded with Caffeic Acid-Hydroxypropyl-β-Cyclodextrin. Korean J. Chem. Eng. 2016, 33, 2738–2746. [Google Scholar] [CrossRef]
  38. Nile, S.H.; Ko, E.Y.; Kim, D.H.; Keum, Y.S. Screening of Ferulic Acid Related Compounds as Inhibitors of Xanthine Oxidase and Cyclooxygenase-2 with Anti-Inflammatory Activity. Braz. J. Pharmacogn. 2016, 26, 50–55. [Google Scholar] [CrossRef] [Green Version]
  39. Adomako-Bonsu, A.G.; Chan, S.L.; Pratten, M.; Fry, J.R. Antioxidant Activity of Rosmarinic Acid and Its Principal Metabolites in Chemical and Cellular Systems: Importance of Physico-Chemical Characteristics. Toxicol. Vitro 2017, 40, 248–255. [Google Scholar] [CrossRef]
  40. Váňová, J.; Liimatta, L.J.; Česla, P.; Wiedmer, S.K. Determination of Distribution Constants of Antioxidants by Electrokinetic Chromatography. Cogent Chem. 2017, 3, 1–11. [Google Scholar] [CrossRef]
  41. Mota, F.L.; Queimada, A.J.; Pinho, S.P.; Macedo, E.A. Aqueous Solubility of Some Natural Phenolic Compounds. Ind. Eng. Chem. Res. 2008, 47, 5182–5189. [Google Scholar] [CrossRef] [Green Version]
  42. Ren, J.; Jiang, X.; Li, C.; Li, K.; Chen, Z.; Ma, G. Absorptive Profile of Chlorogenic Acid in Rats. Pharmazie 2007, 62, 689–692. [Google Scholar] [CrossRef] [PubMed]
  43. Álvarez-Parrilla, E.; Palos, R.; De La Rosa, L.A.; Frontana-Uribe, B.A.; González-Aguilar, G.A.; Machi, L.; Ayala-Zavala, J.F. Formation of Two 1:1 Chlorogenic Acid: β-Cyclodextrin Complexes at PH 5: Spectroscopic, Thermodynamic and Voltammetric Study. J. Mex. Chem. Soc. 2010, 54, 103–110. [Google Scholar] [CrossRef]
  44. Sanchez, M.B.; Miranda-Perez, E.; Verjan, J.C.G.; de los Angeles Fortis Barrera, M.; Perez-Ramos, J.; Alarcon-Aguilar, F.J. Potential of the Chlorogenic Acid as Multitarget Agent: Insulin-Secretagogue and PPAR α/γ Dual Agonist. Biomed. Pharmacother. 2017, 94, 169–175. [Google Scholar] [CrossRef] [PubMed]
  45. Farrell, T.L.; Dew, T.P.; Poquet, L.; Hanson, P.; Williamson, G. Absorption and Metabolism of Chlorogenic Acids in Cultured Gastric Epithelial Monolayers. Drug Metab. Dispos. 2011, 39, 2338–2346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Hakkou, Z.; Maciuk, A.; Leblais, V.; Bouanani, N.E.; Mekhfi, H.; Bnouham, M.; Aziz, M.; Ziyyat, A.; Rauf, A.; Hadda, T.B.; et al. Antihypertensive and Vasodilator Effects of Methanolic Extract of Inula Viscosa: Biological Evaluation and POM Analysis of Cynarin, Chlorogenic Acid as Potential Hypertensive. Biomed. Pharmacother. 2017, 93, 62–69. [Google Scholar] [CrossRef]
  47. Benvidi, A.; Dadras, A.; Abbasi, S.; Tezerjani, M.D.; Rezaeinasab, M.; Tabaraki, R.; Namazian, M. Experimental and Computational Study of the PKa of Coumaric Acid Derivatives. J. Chin. Chem. Soc. 2019, 66, 589–593. [Google Scholar] [CrossRef]
  48. Stȩpnik, K.E.; Malinowska, I.; Rój, E. In Vitro and in Silico Determination of Oral, Jejunum and Caco-2 Human Absorption of Fatty Acids and Polyphenols. Micellar Liquid Chromatography. Talanta 2014, 130, 265–273. [Google Scholar] [CrossRef]
  49. Šmidovnik, A.; Stražišar, M.; Jazbec, P.; Fir, M.M.; Prošek, M. Effect of Complexation Cyclodextrins with Phenolic Acids and Coenzyme Q10on Their Physico-Chemical Properties and Bioavailability. Acta Chim. Slov. 2010, 57, 9–16. [Google Scholar]
  50. Dey, P.; Sarkar, I.; Dutta, S.; Saha, M.R.; Chaudhuri, T.K. Correlative Metabolomic Fingerprinting and Molecular Docking Studies of Dermatological Phytotherapeutics of South-Eastern Himalaya. J. Tradit. Complement. Med. 2019, 9, 243–248. [Google Scholar] [CrossRef]
  51. Jablonsky, M.; Haz, A.; Sladkova, A.; Strizincova, P.; Skulcova, A.; Majova, V.; Jablonsky, J. Nutraceuticals as Phenolic Bioactive Compounds Analysis of Softwood Bark and Their Possibilities of Industry Applications. J. Hyg. Eng. Des. 2019, 26, 93–99. [Google Scholar]
  52. Ota, A.; Abramovič, H.; Abram, V.; Poklar Ulrih, N. Interactions of P-Coumaric, Caffeic and Ferulic Acids and Their Styrenes with Model Lipid Membranes. Food Chem. 2011, 125, 1256–1261. [Google Scholar] [CrossRef]
  53. Monti, D.; Tampucci, S.; Chetoni, P.; Burgalassi, S.; Saino, V.; Centini, M.; Staltari, L.; Anselmi, C. Permeation and Distribution of Ferulic Acid and Its α-Cyclodextrin Complex from Different Formulations in Hairless Rat Skin. AAPS PharmSciTech 2011, 12, 514–520. [Google Scholar] [CrossRef] [PubMed]
  54. Han, X.; Zhang, Z.; Shen, H.; Zheng, J.; Zhang, G. Comparison of Structures, Physicochemical Properties and in Vitro Bioactivity between Ferulic Acid-β-Cyclodextrin Conjugate and the Corresponding Inclusion Complex. Food Res. Int. 2019, 125, 108619. [Google Scholar] [CrossRef] [PubMed]
  55. Nath, L.S.; Khan, S.A.; Ahmad, A. Computer Aided Screening of Natural Products in Search of Lead Molecules for Design and Development of Potent Anti-Inflammatory Agents. Sch. Acad. J. Pharm. 2014, 3, 496–503. [Google Scholar]
  56. El-Boshy, M.; Header, E.; ElSawy, N.; Basalamah, M.; Mubarak, M.; Hadda, T. Studies on the Constituents of Rosmarinus Officinalis and Their Synergistic Effect in Experimental Diabetic Rats. J. Investig. Biochem. 2015, 4, 36. [Google Scholar] [CrossRef]
  57. Al Danaf, N.; Melhem, R.A.; Assaf, K.I.; Nau, W.M.; Patra, D. Photophysical Properties of Neutral and Dissociated Forms of Rosmarinic Acid. J. Lumin. 2016, 175, 50–56. [Google Scholar] [CrossRef]
  58. Veras, K.S.; Silveira Fachel, F.N.; Delagustin, M.G.; Teixeira, H.F.; Barcellos, T.; Henriques, A.T.; Bassani, V.L.; Koester, L.S. Complexation of Rosmarinic Acid with Hydroxypropyl-β-Cyclodextrin and Methyl-β-Cyclodextrin: Formation of 2:1 Complexes with Improved Antioxidant Activity. J. Mol. Struct. 2019, 1195, 582–590. [Google Scholar] [CrossRef]
  59. Daina, A.; Michielin, O.; Zoete, V. SwissADME: A Free Web Tool to Evaluate Pharmacokinetics, Drug-Likeness and Medicinal Chemistry Friendliness of Small Molecules; Springer Science and Business Media LLC: Berlin/Heidelberg, Germany, 2017. [Google Scholar]
  60. Abuhelwa, A.Y.; Williams, D.B.; Upton, R.N.; Foster, D.J.R. Food, Gastrointestinal PH, and Models of Oral Drug Absorption. Eur. J. Pharm. Biopharm. 2017, 112, 234–248. [Google Scholar] [CrossRef]
  61. Di, L.; Kerns, E.H.; Hong, Y.; Chen, H. Development and Application of High Throughput Plasma Stability Assay for Drug Discovery. Int. J. Pharm. 2005, 297, 110–119. [Google Scholar] [CrossRef]
  62. Gayoso, L.; Claerbout, A.S.; Calvo, M.I.; Cavero, R.Y.; Astiasarán, I.; Ansorena, D. Bioaccessibility of Rutin, Caffeic Acid and Rosmarinic Acid: Influence of the in Vitro Gastrointestinal Digestion Models. J. Funct. Foods 2016, 26, 428–438. [Google Scholar] [CrossRef]
  63. Zorić, Z.; Markić, J.; Pedisić, S.; Bučevic-Popović, V.; Generalic-Mekinić, I.; Grebenar, K.; Kulišić-Bilušic, T. Stability of Rosmarinic Acid in Aqueous Extracts from Different Lamiaceae Species after in Vitro Digestion with Human Gastrointestinal Enzymes. Food Technol. Biotechnol. 2016, 54, 97–102. [Google Scholar] [CrossRef] [PubMed]
  64. Porfírio, S.; Falé, P.L.V.; Madeira, P.J.A.; Florêncio, M.H.; Ascensão, L.; Serralheiro, M.L.M. Antiacetylcholinesterase and Antioxidant Activities of Plectranthus Barbatus Tea, after in Vitro Gastrointestinal Metabolism. Food Chem. 2010, 122, 179–187. [Google Scholar] [CrossRef]
  65. Costa, P.; Grevenstuk, T.; Rosa da Costa, A.M.; Gonçalves, S.; Romano, A. Antioxidant and Anti-Cholinesterase Activities of Lavandula Viridis L’Hér Extracts after in Vitro Gastrointestinal Digestion. Ind. Crops Prod. 2014, 55, 83–89. [Google Scholar] [CrossRef]
  66. Tagliazucchi, D.; Verzelloni, E.; Bertolini, D.; Conte, A. In Vitro Bio-Accessibility and Antioxidant Activity of Grape Polyphenols. Food Chem. 2010, 120, 599–606. [Google Scholar] [CrossRef]
  67. D’Antuono, I.; Garbetta, A.; Linsalata, V.; Minervini, F.; Cardinali, A. Polyphenols from Artichoke Heads (Cynara cardunculus (L.) Subsp. Scolymus Hayek): In Vitro Bio-Accessibility, Intestinal Uptake and Bioavailability. Food Funct. 2015, 6, 1268–1277. [Google Scholar] [CrossRef] [PubMed]
  68. Zhao, Z.; Egashira, Y.; Sanada, H. Ferulic Acid Is Quickly Absorbed from Rat Stomach as the Free Form and Then Conjugated Mainly in Liver. J. Nutr. 2004, 134, 3083–3088. [Google Scholar] [CrossRef] [Green Version]
  69. Bermúdez-Soto, M.J.; Tomás-Barberán, F.A.; García-Conesa, M.T. Stability of Polyphenols in Chokeberry (Aronia melanocarpa) Subjected to in Vitro Gastric and Pancreatic Digestion. Food Chem. 2007, 102, 865–874. [Google Scholar] [CrossRef]
  70. Siracusa, L.; Kulisic-Bilusic, T.; Politeo, O.; Krause, I.; Dejanovic, B.; Ruberto, G. Phenolic Composition and Antioxidant Activity of Aqueous Infusions from Capparis spinosa L. and Crithmum maritimum L. before and after Submission to a Two-Step in Vitro Digestion Model. J. Agric. Food Chem. 2011, 59, 12453–12459. [Google Scholar] [CrossRef]
  71. Budryn, G.; Pałecz, B.; Rachwał-Rosiak, D.; Oracz, J.; Zaczyńska, D.; Belica, S.; Navarro-González, I.; Meseguer, J.M.V.; Pérez-Sánchez, H. Effect of Inclusion of Hydroxycinnamic and Chlorogenic Acids from Green Coffee Bean in β-Cyclodextrin on Their Interactions with Whey, Egg White and Soy Protein Isolates. Food Chem. 2015, 168, 276–287. [Google Scholar] [CrossRef]
  72. Celli, N.; Dragani, L.K.; Murzilli, S.; Pagliani, T.; Poggi, A. In Vitro and in Vivo Stability of Caffeic Acid Phenethyl Ester, a Bioactive Compound of Propolis. J. Agric. Food Chem. 2007, 55, 3398–3407. [Google Scholar] [CrossRef]
  73. Narita, Y.; Inouye, K. Degradation Kinetics of Chlorogenic Acid at Various PH Values and Effects of Ascorbic Acid and Epigallocatechin Gallate on Its Stability under Alkaline Conditions. J. Agric. Food Chem. 2013, 61, 966–972. [Google Scholar] [CrossRef] [PubMed]
  74. Uang, Y.S.; Kang, F.L.; Hsu, K.Y. Determination of Caffeic Acid in Rabbit Plasma by High-Performance Liquid Chromatography. J. Chromatogr. B Biomed. Sci. Appl. 1995, 673, 43–49. [Google Scholar] [CrossRef] [PubMed]
  75. Cui, Y.; Li, Q.; Zhang, M.; Liu, Z.; Yin, W.; Liu, W.; Chen, X.; Bi, K. LC-MS Determination and Pharmacokinetics of p-Coumaric Acid in Rat Plasma after Oral Administration of p-Coumaric Acid and Freeze-Dried Red Wine. J. Agric. Food Chem. 2010, 58, 12083–12088. [Google Scholar] [CrossRef] [PubMed]
  76. He, B.; Li, Q.; Jia, Y.; Zhao, L.; Xiao, F.; Lv, C.; Xu, H.; Chen, X.; Bi, K. A UFLC-MS/MS Method for Simultaneous Quantitation of Spinosin, Mangiferin and Ferulic Acid in Rat Plasma: Application to a Comparative Pharmacokinetic Study in Normal and Insomnic Rats. J. Mass Spectrom. 2012, 47, 1333–1340. [Google Scholar] [CrossRef]
  77. Kerns, E.H.; Di, L.; Petusky, S.; Farris, M.; Ley, R.; Jupp, P. Combined Application of Parallel Artificial Membrane Permeability Assay and Caco-2 Permeability Assays in Drug Discovery. J. Pharm. Sci. 2004, 93, 1440–1453. [Google Scholar] [CrossRef]
  78. Sarmento, B.; Andrade, F.; Da Silva, S.B.; Rodrigues, F.; Das Neves, J.; Ferreira, D. Cell-Based in Vitro Models for Predicting Drug Permeability. Expert Opin. Drug Metab. Toxicol. 2012, 8, 607–621. [Google Scholar] [CrossRef]
  79. De Souza, J.; Freitas, Z.M.F.; Storpirtis, S. Modelos in Vitro Para Determinação Da Absorção de Fármacos e Previsão Da Relação Dissolução/Absorção. Rev. Bras. Ciências Farm. 2007, 43, 515–527. [Google Scholar] [CrossRef] [Green Version]
  80. Avdeef, A. The Rise of PAMPA. Expert Opin. Drug Metab. Toxicol. 2005, 1, 325–342. [Google Scholar] [CrossRef]
  81. Petit, C.; Bujard, A.; Skalicka-Woźniak, K.; Cretton, S.; Houriet, J.; Christen, P.; Carrupt, P.A.; Wolfender, J.L. Prediction of the Passive Intestinal Absorption of Medicinal Plant Extract Constituents with the Parallel Artificial Membrane Permeability Assay (PAMPA). Planta Med. 2016, 82, 424–431. [Google Scholar] [CrossRef] [Green Version]
  82. Petit, C.; Ceccarelli, M.; Cretton, S.; Houriet, J.; Skalicka-Woźniak, K.; Christen, P.; Carrupt, P.A.; Goracci, L.; Wolfender, J.L. Passive Intestinal Absorption of Representative Plant Secondary Metabolites: A Physicochemical Study. Planta Med. 2017, 83, 718–726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Avdeef, A. Absorption and Drug Development: Solubility, Permeability, and Charge State, 2nd ed.; John Wiley & Sons: Hoboken, NJ, USA, 2012. [Google Scholar]
  84. Li, D.; Kerns, E. Drug-Like Properties: Concepts, Structure Design and Methods from ADME to Toxicity Optimization, 2nd ed.; Academic Press: Cambridge, MA, USA, 2016. [Google Scholar]
  85. Egan, W.J.; Merz, K.M.; Baldwin, J.J. Prediction of Drug Absorption Using Multivariate Statistics. J. Med. Chem. 2000, 43, 3867–3877. [Google Scholar] [CrossRef]
  86. Ghose, A.K.; Viswanadhan, V.N.; Wendoloski, J.J. A Knowledge-Based Approach in Designing Combinatorial or Medicinal Chemistry Libraries for Drug Discovery. 1. A Qualitative and Quantitative Characterization of Known Drug Databases. J. Comb. Chem. 1999, 1, 55–68. [Google Scholar] [CrossRef] [PubMed]
  87. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and Computational Approaches to Estimate Solubility and Permeability in Drug Discovery and Development Settings. Adv. Drug Deliv. Rev. 1997, 23, 3–25. [Google Scholar] [CrossRef]
  88. Muegge, I.; Heald, S.L.; Brittelli, D. Simple Selection Criteria for Drug-like Chemical Matter. J. Med. Chem. 2001, 44, 1841–1846. [Google Scholar] [CrossRef] [PubMed]
  89. Oprea, T.I. Property Distribution of Drug-Related Chemical Databases. J. Comput. Aided Mol. Des. 2000, 14, 251–264. [Google Scholar] [CrossRef]
  90. Veber, D.F.; Johnson, S.R.; Cheng, H.Y.; Smith, B.R.; Ward, K.W.; Kopple, K.D. Molecular Properties That Influence the Oral Bioavailability of Drug Candidates. J. Med. Chem. 2002, 45, 2615–2623. [Google Scholar] [CrossRef] [PubMed]
  91. Zmuidinavicius, D.; Didziapetris, R.; Japertas, P.; Avdeef, A.; Petrauskas, A. Classification Structure-Activity Relations (C-SAR) in Prediction of Human Intestinal Absorption. J. Pharm. Sci. 2003, 92, 621–633. [Google Scholar] [CrossRef] [PubMed]
  92. Van de Waterbeemd, H.; Lennernas, H.; Artursson, P. Drug Bioavailability: Estimation of Solubility, Permeability, Absorption and Bioavailability, 1st ed.; Wiley-VCH: Weinheim, Germany, 2003. [Google Scholar]
  93. Sugano, K.; Kansy, M.; Artursson, P.; Avdeef, A.; Bendels, S.; Di, L.; Ecker, G.F.; Faller, B.; Fischer, H.; Gerebtzoff, G.; et al. Coexistence of Passive and Carrier-Mediated Processes in Drug Transport. Nat. Rev. Drug Discov. 2010, 9, 597–614. [Google Scholar] [CrossRef] [PubMed]
  94. Sun, H.; Chow, E.C.Y.; Liu, S.; Du, Y.; Pang, K.S. The Caco-2 Cell Monolayer: Usefulness and Limitations. Expert Opin. Drug Metab. Toxicol. 2008, 4, 395–411. [Google Scholar] [CrossRef]
  95. Gomez-Lechon, M.; Donato, M.; Lahoz, A.; Castell, J. Cell Lines: A Tool for In Vitro Drug Metabolism Studies. Curr. Drug Metab. 2008, 9, 1–11. [Google Scholar] [CrossRef] [PubMed]
  96. Konishi, Y.; Shimizu, M. Transepithelial Transport of Ferulic Acid by Monocarboxylic Acid Transporter in Caco-2 Cell Monolayers. Biosci. Biotechnol. Biochem. 2003, 67, 856–862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Konishi, Y.; Kobayashi, S.; Shimizu, M. Transepithelial Transport of P-Coumaric Acid and Gallic Acid in Caco-2 Cell Monolayers. Biosci. Biotechnol. Biochem. 2003, 67, 2317–2324. [Google Scholar] [CrossRef]
  98. Konishi, Y.; Kobayashi, S. Microbial Metabolites of Ingested Caffeic Acid Are Absorbed by the Monocarboxylic Acid Transporter (MCT) in Intestinal Caco-2 Cell Monolayers. J. Agric. Food Chem. 2004, 52, 6418–6424. [Google Scholar] [CrossRef] [PubMed]
  99. Konishi, Y.; Kobayashi, S. Transepithelial Transport of Chlorogenic Acid, Caffeic Acid, and Their Colonic Metabolites in Intestinal Caco-2 Cell Monolayers. J. Agric. Food Chem. 2004, 52, 2518–2526. [Google Scholar] [CrossRef] [PubMed]
  100. Falé, P.L.; Ascensão, L.; Serralheiro, M.L.M. Effect of Luteolin and Apigenin on Rosmarinic Acid Bioavailability in Caco-2 Cell Monolayers. Food Funct. 2013, 4, 426–431. [Google Scholar] [CrossRef]
  101. Monente, C.; Ludwig, I.A.; Stalmach, A.; De Peña, M.P.; Cid, C.; Crozier, A. In Vitro Studies on the Stability in the Proximal Gastrointestinal Tract and Bioaccessibility in Caco-2 Cells of Chlorogenic Acids from Spent Coffee Grounds. Int. J. Food Sci. Nutr. 2015, 66, 657–664. [Google Scholar] [CrossRef]
  102. Qiang, Z.; Ye, Z.; Hauck, C.; Murphy, P.A.; McCoy, J.-A.; Widrlechner, M.P.; Reddy, M.B.; Hendrich, S. Permeability of Rosmarinic Acid in Prunella Vulgaris and Ursolic Acid in Salvia Officinalis Extracts across Caco-2 Cell Monolayers. J. Ethnopharmacol. 2011, 137, 1107–1112. [Google Scholar] [CrossRef] [Green Version]
  103. Farrell, T.L.; Poquet, L.; Dew, T.P.; Barber, S.; Williamson, G. Predicting Phenolic Acid Absorption in Caco-2 Cells: A Theoretical Permeability Model and Mechanistic Study. Drug Metab. Dispos. 2012, 40, 397–406. [Google Scholar] [CrossRef]
  104. Gómez-Juaristi, M.; Sarria, B.; Goya, L.; Bravo-Clemente, L.; Mateos, R. Experimental Confounding Factors Affecting Stability, Transport and Metabolism of Flavanols and Hydroxycinnamic Acids in Caco-2 Cells. Food Res. Int. 2020, 129, 108797. [Google Scholar] [CrossRef]
  105. Yang, J.H.; Zhang, L.; Li, J.S.; Chen, L.H.; Zheng, Q.; Chen, T.; Chen, Z.P.; Fu, T.M.; Di, L.Q. Enhanced Oral Bioavailability and Prophylactic Effects on Oxidative Stress and Hepatic Damage of an Oil Solution Containing a Rosmarinic Acid-Phospholipid Complex. J. Funct. Foods 2015, 19, 63–73. [Google Scholar] [CrossRef]
  106. Mortelé, O.; Jörissen, J.; Spacova, I.; Lebeer, S.; van Nuijs, A.L.N.; Hermans, N. Demonstrating the Involvement of an Active Efflux Mechanism in the Intestinal Absorption of Chlorogenic Acid and Quinic Acid Using a Caco-2 Bidirectional Permeability Assay. Food Funct. 2021, 12, 417–425. [Google Scholar] [CrossRef] [PubMed]
  107. Villalva, M.; Jaime, L.; Aguado, E.; Nieto, J.A.; Reglero, G.; Santoyo, S. Anti-Inflammatory and Antioxidant Activities from the Basolateral Fraction of Caco-2 Cells Exposed to a Rosmarinic Acid Enriched Extract. J. Agric. Food Chem. 2018, 66, 1167–1174. [Google Scholar] [CrossRef] [PubMed]
  108. Lee, H.J.; Cha, K.H.; Kim, C.Y.; Nho, C.W.; Pan, C.H. Bioavailability of Hydroxycinnamic Acids from Crepidiastrum Denticulatum Using Simulated Digestion and Caco-2 Intestinal Cells. J. Agric. Food Chem. 2014, 62, 5290–5295. [Google Scholar] [CrossRef]
  109. Poquet, L.; Clifford, M.N.; Williamson, G. Transport and Metabolism of Ferulic Acid through the Colonic Epithelium. Drug Metab. Dispos. 2008, 36, 190–197. [Google Scholar] [CrossRef]
  110. Bergmann, H.; Rogoll, D.; Scheppach, W.; Melcher, R.; Richling, E. The Ussing Type Chamber Model to Study the Intestinal Transport and Modulation of Specific Tight-Junction Genes Using a Colonic Cell Line. Mol. Nutr. Food Res. 2009, 53, 1211–1225. [Google Scholar] [CrossRef]
  111. Karlsson, J.; Ungell, A.L.; Gråsjö, J.; Artursson, P. Paracellular Drug Transport across Intestinal Epithelia: Influence of Charge and Induced Water Flux. Eur. J. Pharm. Sci. 1999, 9, 47–56. [Google Scholar] [CrossRef]
  112. Giacomini, K.M.; Huang, S.M.; Tweedie, D.J.; Benet, L.Z.; Brouwer, K.L.R.; Chu, X.; Dahlin, A.; Evers, R.; Fischer, V.; Hillgren, K.M.; et al. Membrane Transporters in Drug Development. Nat. Rev. Drug Discov. 2010, 9, 215–236. [Google Scholar] [CrossRef]
  113. Zhou, W.; Shan, J.; Wang, S.; Cai, B.; Di, L. Transepithelial Transport of Phenolic Acids in Flos Lonicerae Japonicae in Intestinal Caco-2 Cell Monolayers. Food Funct. 2015, 6, 3072–3080. [Google Scholar] [CrossRef]
  114. Li, F.-R.; Fu, Y.-Y.; Jiang, D.-H.; Wu, Z.; Zhou, Y.-J.; Guo, L.; Dong, Z.-M.; Wang, Z.-Z. Reversal Effect of Rosmarinic Acid on Multidrug Resistance in SGC7901/Adr Cell. J. Asian Nat. Prod. Res. 2013, 15, 276–285. [Google Scholar] [CrossRef]
  115. Hong, Y.J.; Yang, S.Y.; Nam, M.H.; Koo, Y.C.; Lee, K.W. Caffeic Acid Inhibits the Uptake of 2-Amino-1-Methyl-6-Phenylimidazo-[4,5-b]Pyridine (PhIP) by Inducing the Efflux Transporters Expression in Caco-2 Cells. Biol. Pharm. Bull. 2015, 38, 201–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Teng, Y.; Wang, C.C.N.; Liao, W.; Lan, Y.; Hung, C. Caffeic Acid Attenuates Multi-Drug Resistance in Cancer Cells by Inhibiting Efflux Function of Human P-Glycoprotein. Molecules 2020, 25, 247. [Google Scholar] [CrossRef] [Green Version]
  117. Wu, J.; Zhu, Y.; Li, F.; Zhang, G.; Shi, J.; Ou, R.; Tong, Y. Spica Prunellae and Its Marker Compound Rosmarinic Acid Induced the Expression of Efflux Transporters through Activation of Nrf2-Mediated Signaling Pathway in HepG2 Cells. J. Ethnopharmacol. 2016, 193, 1–11. [Google Scholar] [CrossRef] [PubMed]
  118. Li, M.; Yin, D.; Li, J.; Shao, F.; Zhang, Q.; Jiang, Q.; Zhang, M.; Yang, Y. Rosmarinic Acid, the Active Component of Salvia Miltiorrhizae, Improves Gliquidone Transport by Regulating the Expression and Function of P-Gp and BCRP in Caco-2 Cells. Pharmazie 2020, 75, 18–22. [Google Scholar] [CrossRef] [PubMed]
  119. Muthusamy, G.; Balupillai, A.; Ramasamy, K.; Shanmugam, M.; Gunaseelan, S.; Mary, B.; Prasad, N.R. Ferulic Acid Reverses ABCB1-Mediated Paclitaxel Resistance in MDR Cell Lines. Eur. J. Pharmacol. 2016, 786, 194–203. [Google Scholar] [CrossRef] [PubMed]
  120. Muthusamy, G.; Gunaseelan, S.; Prasad, N.R. Ferulic Acid Reverses P-Glycoprotein-Mediated Multidrug Resistance via Inhibition of PI3K/Akt/NF-ΚB Signaling Pathway. J. Nutr. Biochem. 2019, 63, 62–71. [Google Scholar] [CrossRef] [PubMed]
  121. Kern, S.M.; Bennett, R.N.; Needs, P.W.; Mellon, F.A.; Kroon, P.A.; Garcia-Conesa, M.T. Characterization of Metabolites of Hydroxycinnamates in the in Vitro Model of Human Small Intestinal Epithelium Caco-2 Cells. J. Agric. Food Chem. 2003, 51, 7884–7891. [Google Scholar] [CrossRef]
  122. Achour, M.; Saguem, S.; Sarriá, B.; Bravo, L.; Mateos, R. Bioavailability and Metabolism of Rosemary Infusion Polyphenols Using Caco-2 and HepG2 Cell Model Systems. J. Sci. Food Agric. 2018, 98, 3741–3751. [Google Scholar] [CrossRef] [Green Version]
  123. Mateos, R.; Goya, L.; Bravo, L. Uptake and Metabolism of Hydroxycinnamic Acids (Chlorogenic, Caffeic, and Ferulic Acids) by HepG2 Cells as a Model of the Human Liver. J. Agric. Food Chem. 2006, 54, 8724–8732. [Google Scholar] [CrossRef] [Green Version]
  124. Su, J.; Jia, F.; Lu, J.; Chen, W.; Sun, H.; Liu, T.; Wu, X. Characterization of the Metabolites of Rosmarinic Acid in Human Liver Microsomes Using Liquid Chromatography Combined with Electrospray Ionization Tandem Mass Spectrometry. Biomed. Chromatogr. 2020, 34, e4806. [Google Scholar] [CrossRef]
  125. Moridani, M.Y.; Scobie, H.; O’Brien, P.J. Metabolism of Caffeic Acid by Isolated Rat Hepatocytes and Subcellular Fractions. Toxicol. Lett. 2002, 133, 141–151. [Google Scholar] [CrossRef] [PubMed]
  126. Moridani, M.Y.; Scobie, H.; Jamshidzadeh, A.; Salehi, P.; O’Brien, P.J. Caffeic Acid, Chlorogenic Acid, and Dihydrocaffeic Acid Metabolism: Glutathione Conjugate Formation. Drug Metab. Dispos. 2001, 29, 1432–1439. [Google Scholar] [PubMed]
  127. Wong, C.C.; Meinl, W.; Glatt, H.R.; Barron, D.; Stalmach, A.; Steiling, H.; Crozier, A.; Williamson, G. In Vitro and in Vivo Conjugation of Dietary Hydroxycinnamic Acids by UDP-Glucuronosyltransferases and Sulfotransferases in Humans. J. Nutr. Biochem. 2010, 21, 1060–1068. [Google Scholar] [CrossRef] [PubMed]
  128. Gonthier, M.P.; Remesy, C.; Scalbert, A.; Cheynier, V.; Souquet, J.M.; Poutanen, K.; Aura, A.M. Microbial Metabolism of Caffeic Acid and Its Esters Chlorogenic and Caftaric Acids by Human Faecal Microbiota in Vitro. Biomed. Pharmacother. 2006, 60, 536–540. [Google Scholar] [CrossRef] [PubMed]
  129. Parkar, S.G.; Trower, T.M.; Stevenson, D.E. Fecal Microbial Metabolism of Polyphenols and Its Effects on Human Gut Microbiota. Anaerobe 2013, 23, 12–19. [Google Scholar] [CrossRef]
  130. Ludwig, I.A.; Paz de Peña, M.; Concepción, C.; Alan, C. Catabolism of Coffee Chlorogenic Acids by Human Colonic Microbiota. BioFactors 2013, 39, 623–632. [Google Scholar] [CrossRef]
  131. Shen, H.; Tong, X.; Yang, J.; Yu, L.; Zhou, H.; Wang, Y.; He, Y.; Wan, H.; Li, C. Biotransformation of Natural Hydroxycinnamic Acids by Gut Microbiota from Normal and Cerebral Ischemia-Reperfusion Injured Rats: A Comparative Study. Food Funct. 2020, 11, 5389–5395. [Google Scholar] [CrossRef]
  132. Bel-Rhlid, R.; Crespy, V.; Pagé-Zoerkler, N.; Nagy, K.; Raab, T.; Hansen, C.E. Hydrolysis of Rosmarinic Acid from Rosemary Extract with Esterases and Lactobacillus Johnsonii in Vitro and in a Gastrointestinal Model. J. Agric. Food Chem. 2009, 57, 7700–7705. [Google Scholar] [CrossRef]
  133. Luo, Z.; Liu, Y.; Zhao, B.; Tang, M.; Dong, H.; Zhang, L.; Lv, B.; Wei, L. Ex Vivo and in Situ Approaches Used to Study Intestinal Absorption. J. Pharmacol. Toxicol. Methods 2013, 68, 208–216. [Google Scholar] [CrossRef]
  134. Deußer, H.; Rogoll, D.; Scheppach, W.; Volk, A.; Melcher, R.; Richling, E. Gastrointestinal Absorption and Metabolism of Apple Polyphenols Ex Vivo by the Pig Intestinal Mucosa in the Ussing Chamber. Biotechnol. J. 2013, 8, 363–370. [Google Scholar] [CrossRef]
  135. Erk, T.; Hauser, J.; Williamson, G.; Renouf, M.; Steiling, H.; Dionisi, F.; Richling, E. Structure- and Dose-Absorption Relationships of Coffee Polyphenols. BioFactors 2014, 40, 103–112. [Google Scholar] [CrossRef] [PubMed]
  136. Garrait, G.; Jarrige, J.F.; Blanquet, S.; Beyssac, E.; Cardot, J.M.; Alric, M. Gastrointestinal Absorption and Urinary Excretion of Trans-Cinnamic and p-Coumaric Acids in Rats. J. Agric. Food Chem. 2006, 54, 2944–2950. [Google Scholar] [CrossRef] [PubMed]
  137. Zhai, L.; Shi, J.; Xu, W.; Heinrich, M.; Wang, J.; Deng, W. Ex Vivo and in Situ Evaluation of “Dispelling-Wind” Chinese Medicine Herb-Drugs on Intestinal Absorption of Chlorogenic Acid. Phytother. Res. 2015, 29, 1974–1981. [Google Scholar] [CrossRef] [PubMed]
  138. Spencer, J.P.E.; Chowrimootoo, G.; Choudhury, R.; Debnam, E.S.; Srai, S.K.; Rice-Evans, C. The Small Intestine Can Both Absorb and Glucuronidate Luminal Flavonoids. FEBS Lett. 1999, 458, 224–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Kelber, O.; Wittwer, A.; Lapke, C.; Kroll, U.; Weiser, D.; Okpanyi, S.N.; Heilmann, J. Ex Vivo/in Vitro Absorption of STW 5 (Iberogast®) and Its Extract Components. Phytomedicine 2006, 13, 107–113. [Google Scholar] [CrossRef] [PubMed]
  140. Konishi, Y.; Zhao, Z.; Shimizu, M. Phenolic Acids Are Absorbed from the Rat Stomach with Different Absorption Rates. J. Agric. Food Chem. 2006, 54, 7539–7543. [Google Scholar] [CrossRef]
  141. Wang, S.J.; Zeng, J.; Yang, B.K.; Zhong, Y.M. Bioavailability of Caffeic Acid in Rats and Its Absorption Properties in the Caco-2 Cell Model. Pharm. Biol. 2014, 52, 1150–1157. [Google Scholar] [CrossRef] [PubMed]
  142. Lafay, S.; Morand, C.; Manach, C.; Besson, C.; Scalbert, A. Absorption and Metabolism of Caffeic Acid and Chlorogenic Acid in the Small Intestine of Rats. Br. J. Nutr. 2006, 96, 39–46. [Google Scholar] [CrossRef] [PubMed]
  143. Lafay, S.; Gil-Izquierdo, A.; Manach, C.; Morand, C.; Besson, C.; Scalbert, A. Chlorogenic Acid Is Absorbed in Its Intact Form in the Stomach of Rats. J. Nutr. 2006, 136, 1192–1197. [Google Scholar] [CrossRef] [Green Version]
  144. Silberberg, M.; Morand, C.; Mathevon, T.; Besson, C.; Manach, C.; Scalbert, A.; Remesy, C. The Bioavailability of Polyphenols Is Highly Governed by the Capacity of the Intestine and of the Liver to Secrete Conjugated Metabolites. Eur. J. Nutr. 2006, 45, 88–96. [Google Scholar] [CrossRef] [PubMed]
  145. Adam, A.; Crespy, V.; Levrat-Verny, M.-A.; Leenhardt, F.; Leuillet, M.; Demigné, C.; Rémésy, C. The Bioavailability of Ferulic Acid Is Governed Primarily by the Food Matrix Rather than Its Metabolism in Intestine and Liver in Rats. J. Nutr. 2002, 132, 1962–1968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Camarasa, J.; Escubedo, E.; Adzet, T. Pharmacokinetics of Caffeic Acid in Rats by a High-Performance Liquid Chromatography Method. J. Pharm. Biomed. Anal. 1988, 6, 503–510. [Google Scholar] [CrossRef] [PubMed]
  147. Konishi, Y.; Hitomi, Y.; Yoshida, M.; Yoshioka, E. Pharmacokinetic Study of Caffeic and Rosmarinic Acids in Rats after Oral Administration. J. Agric. Food Chem. 2005, 53, 4740–4746. [Google Scholar] [CrossRef] [PubMed]
  148. Wang, X.; Li, W.; Ma, X.; Chu, Y.; Li, S.; Guo, J.; Jia, Y.; Zhou, S.; Zhu, Y.; Liu, C. Simultaneous Determination of Caffeic Acid and Its Major Pharmacologically Active Metabolites in Rat Plasma by LC-MS/MS and Its Application in Pharmacokinetic Study. Biomed. Chromatogr. 2015, 29, 552–559. [Google Scholar] [CrossRef]
  149. Qi, W.; Zhao, T.; Yang, W.W.; Wang, G.H.; Yu, H.; Zhao, H.X.; Yang, C.; Sun, L.X. Comparative Pharmacokinetics of Chlorogenic Acid after Oral Administration in Rats. J. Pharm. Anal. 2011, 1, 270–274. [Google Scholar] [CrossRef] [Green Version]
  150. Jung, J.W.; Kim, J.M.; Jeong, J.S.; Son, M.; Lee, H.S.; Lee, M.G.; Kang, H.E. Pharmacokinetics of Chlorogenic Acid and Corydaline in DA-9701, a New Botanical Gastroprokinetic Agent, in Rats. Xenobiotica 2014, 44, 635–643. [Google Scholar] [CrossRef]
  151. De Oliveira, D.M.; Sampaio, G.R.; Pinto, C.B.; Catharino, R.R.; Bastos, D.H.M. Bioavailability of Chlorogenic Acids in Rats after Acute Ingestion of Maté Tea (Ilex Paraguariensis) or 5-Caffeoylquinic Acid. Eur. J. Nutr. 2017, 56, 2541–2556. [Google Scholar] [CrossRef]
  152. Chen, L.; Liu, C.S.; Chen, Q.Z.; Wang, S.; Xiong, Y.A.; Jing, J.; Lv, J.J. Characterization, Pharmacokinetics and Tissue Distribution of Chlorogenic Acid-Loaded Self-Microemulsifying Drug Delivery System. Eur. J. Pharm. Sci. 2017, 100, 102–108. [Google Scholar] [CrossRef]
  153. Konishi, Y.; Hitomi, Y.; Yoshioka, E. Intestinal Absorption of P-Coumaric and Gallic Acids in Rats after Oral Administration. J. Agric. Food Chem. 2004, 52, 2527–2532. [Google Scholar] [CrossRef]
  154. Lv, H.; Zou, M.; Yu, W.; Sun, B.; Cui, Y. Simultaneous Determination of Gallic Acid and p-Coumaric Acid in Rat Plasma by UPLC-MS/MS and Its Application to a Comparative Pharmacokinetic Study after Oral Administration of Monomer Compound and Red Wine Extract. J. Polyphen. 2019, 1, 1–11. [Google Scholar]
  155. Kim, H.; Choi, Y.; An, Y.; Jung, Y.-R.; Lee, J.-Y.; Lee, H.-J.; Jeong, J.; Kim, Z.; Kim, K. Development of P-Coumaric Acid Analysis in Human Plasma and Its Clinical Application to PK/PD Study. J. Clin. Med. 2020, 10, 108. [Google Scholar] [CrossRef] [PubMed]
  156. Li, Y.; Liu, C.; Zhang, Y.; Mi, S.; Wang, N. Pharmacokinetics of Ferulic Acid and Potential Interactions with Honghua and Clopidogrel in Rats. J. Ethnopharmacol. 2011, 137, 562–567. [Google Scholar] [CrossRef] [PubMed]
  157. Zhang, Y.; Li, Z.; Zhang, K.; Yang, G.; Wang, Z.; Zhao, J.; Hu, R.; Feng, N. Ethyl Oleate-Containing Nanostructured Lipid Carriers Improve Oral Bioavailability of Trans-Ferulic Acid Ascompared with Conventional Solid Lipid Nanoparticles. Int. J. Pharm. 2016, 511, 57–64. [Google Scholar] [CrossRef] [PubMed]
  158. Liu, C.S.; Chen, L.; Hu, Y.N.; Dai, J.L.; Ma, B.; Tang, Q.F.; Tan, X.M. Self-Microemulsifying Drug Delivery System for Improved Oral Delivery and Hypnotic Efficacy of Ferulic Acid. Int. J. Nanomed. 2020, 15, 2059–2070. [Google Scholar] [CrossRef] [PubMed]
  159. Baba, S.; Osakabe, N.; Natsume, M.; Terao, J. Orally Administered Rosmarinic Acid Is Present as the Conjugated and/or Methylated Forms in Plasma, and Is Degraded and Metabolized to Conjugated Forms of Caffeic Acid, Ferulic Acid and m-Coumaric Acid. Life Sci. 2004, 75, 165–178. [Google Scholar] [CrossRef]
  160. Wang, J.; Li, G.; Rui, T.; Kang, A.; Li, G.; Fu, T.; Li, J.; Di, L.; Cai, B. Pharmacokinetics of Rosmarinic Acid in Rats by LC-MS/MS: Absolute Bioavailability and Dose Proportionality. RSC Adv. 2017, 7, 9057–9063. [Google Scholar] [CrossRef] [Green Version]
  161. Yang, J.-H.; Mao, K.-J.; Huang, P.; Ye, Y.-J.; Guo, H.-S.; Cai, B.-C. Effect of Piperine on the Bioavailability and Pharmacokinetics of Rosmarinic Acid in Rat Plasma Using UPLC-MS/MS. Xenobiotica 2018, 48, 178–185. [Google Scholar] [CrossRef]
  162. Wang, X.; Qian, Y.; Li, X.; Jia, X.; Yan, Z.; Han, M.; Qiao, M.; Ma, X.; Chu, Y.; Zhou, S.; et al. Rapid Determination of Rosmarinic Acid and Its Two Bioactive Metabolites in the Plasma of Rats by LC–MS/MS and Application to a Pharmacokinetics Study. Biomed. Chromatogr. 2021, 35, e4984. [Google Scholar] [CrossRef]
  163. Omar, M.H.; Mullen, W.; Stalmach, A.; Auger, C.; Rouanet, J.M.; Teissedre, P.L.; Caldwell, S.T.; Hartley, R.C.; Crozier, A. Absorption, Disposition, Metabolism, and Excretion of [3-14C]Caffeic Acid in Rats. J. Agric. Food Chem. 2012, 60, 5205–5214. [Google Scholar] [CrossRef]
  164. Chen, W.C.; Huang, P.W.; Yang, W.L.; Chen, Y.L.; Shih, Y.N.; Wang, H.J. Fundamentals of Pharmacokinetics to Assess the Correlation Between Plasma Drug Concentrations and Different Blood Sampling Methods. Pharm. Res. 2019, 36, 1–15. [Google Scholar] [CrossRef]
  165. Shargel, L.; Yu, A.B.C. Applied Biopharmaceutics and Pharmakinetics, 7th ed.; Appleton & Lange Reviews; McGraw-Hill, Medical Pub. Division: New York, NY, USA, 2016. [Google Scholar]
  166. Davies, B.; Morris, T. Physiological Parameters in Laboratory Animals and Humans. Pharm. Res. 1993, 10, 1093–1095. [Google Scholar] [CrossRef] [PubMed]
  167. Kurlbaum, M.; Högger, P. Plasma Protein Binding of Polyphenols from Maritime Pine Bark Extract (USP). J. Pharm. Biomed. Anal. 2011, 54, 127–132. [Google Scholar] [CrossRef] [PubMed]
  168. Kang, Y.J.; Lee, C.H.; Park, S.J.; Lee, H.S.; Choi, M.K.; Song, I.S. Involvement of Organic Anion Transporters in the Pharmacokinetics and Drug Interaction of Rosmarinic Acid. Pharmaceutics 2021, 13, 83. [Google Scholar] [CrossRef] [PubMed]
  169. Azuma, K.; Ippoushi, K.; Nakayama, M.; Ito, H.; Higashio, H.; Terao, J. Absorption of Chlorogenic Acid and Caffeic Acid in Rats after Oral Administration. J. Agric. Food Chem. 2000, 48, 5496–5500. [Google Scholar] [CrossRef]
  170. Yan, N.; Tang, Z.; Xu, Y.; Li, X.; Wang, Q. Pharmacokinetic Study of Ferulic Acid Following Transdermal or Intragastric Administration in Rats. AAPS PharmSciTech 2020, 21, 1–7. [Google Scholar] [CrossRef]
  171. Kishida, K.; Matsumoto, H. Urinary Excretion Rate and Bioavailability of Chlorogenic Acid, Caffeic Acid, p-Coumaric Acid, and Ferulic Acid in Non-Fasted Rats Maintained under Physiological Conditions. Heliyon 2019, 5, e02708. [Google Scholar] [CrossRef] [Green Version]
  172. Choudhury, R.; Srai, S.K.; Debnam, E.; Rice-Evans, C.A. Urinary Excretion of Hydroxycinnamates and Flavonoids after Oral and Intravenous Administration. Free Radic. Biol. Med. 1999, 27, 278–286. [Google Scholar] [CrossRef]
  173. Stompor-Goracy, M.; Machaczka, M. Recent advances in biological activity, new formulations and prodrugs of ferulic acid. Int. J. Mol. Sci. 2021, 22, 12889. [Google Scholar] [CrossRef] [PubMed]
  174. Veras, K.S.; Fachel, F.N.S.; Teixeira, H.F.; Koester, L.S. Technological strategies applied for rosmarinic acid delivery through different routes—A review. J. Drug Deliv. Sci. Technol. 2022, 69, 103054. [Google Scholar] [CrossRef]
  175. Shukla, D.; Nandi, N.K.; Singh, B.; Singh, A.; Kumar, B.; Narang, R.K.; Singh, C. Ferulic acid-loaded drug delivery systems for biomedical applications. J. Drug Deliv. Sci. Technol. 2022, 75, 103621. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of hydroxycinnamic acids (HCAs).
Figure 1. Chemical structure of hydroxycinnamic acids (HCAs).
Pharmaceutics 14 02663 g001
Figure 2. Number of violations for HCAs in each prediction rule.
Figure 2. Number of violations for HCAs in each prediction rule.
Pharmaceutics 14 02663 g002
Figure 3. Scheme of processes involved in the oral absorption of rosmarinic acid (RA) and its remaining fraction after gastrointestinal stability (Fgi), intestinal absorption (FCaco-2), hepatic metabolism (FHepG2) and total oral bioavailability (Fabs), evaluated by in vitro methods.
Figure 3. Scheme of processes involved in the oral absorption of rosmarinic acid (RA) and its remaining fraction after gastrointestinal stability (Fgi), intestinal absorption (FCaco-2), hepatic metabolism (FHepG2) and total oral bioavailability (Fabs), evaluated by in vitro methods.
Pharmaceutics 14 02663 g003
Table 2. Pharmacokinetic parameters of orally administered HCAs.
Table 2. Pharmacokinetic parameters of orally administered HCAs.
HCA Animal Species Dose
(mg/kg)
Cmax
(mg/L)
Tmax
(h)
t1/2
(h)
MRT0→∞
(h)
AUC0→∞
(mg h/L)
Vd
(L/kg)
Cl
(L/h kg)
AUC0→∞/Dose F
(%)
References
CA Sprague-Dawley rats 120.0 - - absorption: 0.08
α: 0.14
β: 3.14
- 278.3 0.518 0.115 2.32 26.54 [146]
CA Wistar rats 18.0 2.023 a
0.409 b
0.17 a
0.17 b
0.58 a
0.57 b
-
-
1.755 a,c
0.329 b,c
-
-
-
-
0.0975 a,c
0.0183 b,c
-
-
[147]
CA Sprague-Dawley rats 20.0 7.871 0.33 1.25 1.72 14.03 - 0.702 3.4 [148]
CA Sprague–Dawley rats 10.0 0.25 0.33 β: 2.13 2.96 0.355 2.41 3.35 0.0355 14.7 [141]
CGA Wistar rats 50.0 0.55 0.48 1.7 - 1.61 97.5 39 0.003 - [149]
CGA Sprague–Dawley rats 1.0
2.0
4.0
8.0
0.00245
0.00912
0.019
0.021
0.25
0.5
1.5
0.75
-
-
-
-
-
-
-
-
0.0078 c
0.017 c
0.041 c
0.065 c
-
-
-
-
 
0.618–0.726
 
0.0078
0.0085
0.0103
0.0081
0.478
0.522
0.718
0.569
[150]
CGA Wistar rats 240.0 1.855 0.5 - - - - - - - [151]
CGA Kunming mices 1200.0 82.6 0.17 - - 51.388 c - - 0.0428 - [152]
p-COA Sprague-Dawley rats 16.4 27.17 a
16.29 b
0.17 a
0.17 b
0.27 a
-
-
-
8.176 a,c
-
-
-
-
-
0.499 a
-
-
-
[153]
p-COA Wistar rats 2.35 3.15 0.17 1.28 - 2.32 - - 0.987 51.8 [75]
p-COA Wistar rats 7.38 4.29 0.18 0.77 - 2.502 - - 0.339 - [154]
p-COA Human 258 * 0.02195 0.5 0.9 - 0.2082 - - 0.00008 - [155]
FA Sprague-Dawley rats 10.0 8.175 0.03 absorption: 0.07
α: 0.16
β: 1.77
- 2.962 - - 0.296 - [156]
FA Sprague-Dawley rats 80.0 9.98 1.33 2.14 3.33 34.75 - - 0.434 - [157]
FA Wistar rats 40.0 73.2 0.58 1.39 1.40 d 82.86 c 1.22 × 106 2.5 × 105 2.072 - [158]
RA Sprague-Dawley rats 50.0 1.667 0.5 - - - - - - - [159]
RA Wistar rats 36.0 0.489 a
0.166 b
0.17 a
0.08 b
0.95 a
1.1 b
-
-
0.3624 a,c
0.0996 b,c
-
-
-
-
0.01 a,c
0.003 a,c
-
-
[147]
RA Sprague-Dawley rats 50.0 0.327 0.33 6.77 - 1.395 - - 0.028 - [105]
RA Sprague-Dawley rats 12.5
25.0
50.0
0.215
0.362
0.791
0.14
0.18
0.31
5.54
5.24
4.92
7.32
7.25
6.11
0.867
1.310
1.867
111.83
141.81
197.65
15
19.2
27.6
0.069
0.053
0.037
1.69
1.28
0.91
[160]
RA Sprague-Dawley rats 50.0 0.416 0.15 5.02 - 1.246 - - 0.025 - [161]
RA Sprague-Dawley rats 50.0 1.088 0.19 1.34 1.57 0.972 - - 0.019 4.13 [162]
Cmax: maximum concentration. Tmax: maximum time. t1/2: half-life time. MRT: mean residence time. AUC: area under the curve. Vd: volume of distribution. Cl: clearance. F: absolute bioavailability. a Sample collected from portal vein. b Sample collected from abdominal artery. c AUC0→t (mg h/L). d MRT0→t (h). * Only mg.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Veras, K.S.; Fachel, F.N.S.; de Araújo, B.V.; Teixeira, H.F.; Koester, L.S. Oral Pharmacokinetics of Hydroxycinnamic Acids: An Updated Review. Pharmaceutics 2022, 14, 2663. https://doi.org/10.3390/pharmaceutics14122663

AMA Style

Veras KS, Fachel FNS, de Araújo BV, Teixeira HF, Koester LS. Oral Pharmacokinetics of Hydroxycinnamic Acids: An Updated Review. Pharmaceutics. 2022; 14(12):2663. https://doi.org/10.3390/pharmaceutics14122663

Chicago/Turabian Style

Veras, Kleyton Santos, Flávia Nathiely Silveira Fachel, Bibiana Verlindo de Araújo, Helder Ferreira Teixeira, and Letícia Scherer Koester. 2022. "Oral Pharmacokinetics of Hydroxycinnamic Acids: An Updated Review" Pharmaceutics 14, no. 12: 2663. https://doi.org/10.3390/pharmaceutics14122663

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