IMR Press / JMCM / Volume 1 / Issue 4 / DOI: 10.31083/j.jmcm.2018.04.502
Open Access Research article
Phenolic peptides as antioxidant and anti-proliferative agents
Show Less
1 Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, c/Profesor García González 1, E-41012, Seville, Spain
2 Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, 09230 México, D. F., Mexico

#These authors contributed equally to the experimental part of the manuscript.

J. Mol. Clin. Med. 2018, 1(4), 237–248; https://doi.org/10.31083/j.jmcm.2018.04.502
Submitted: 20 April 2018 | Accepted: 24 June 2018 | Published: 20 December 2018
Abstract

We report an efficient synthesis of phenolic peptides starting from 3,4-dihydroxyphenylacetic acid (DOPAC) and L-configured amino acid esters (glycine, phenylalanine, valine, serine, tryptophan, and cystine) using different coupling reagents. The combination of a phenolic scaffold with an amino acid residue might modulate the bioavailability and the therapeutic properties of title derivatives. Moreover, the incorporation of a catechol group, with inherent redox activity, can contribute to alter the redox status of the cancer cells, and therefore, provide anti-proliferative properties. Their activities as antioxidants (i.e. scavenging free radicals and H2O2 as well inhibition of lipid peroxidation) and as anti-proliferative agents against three human cervical carcinoma cell lines (HeLa, ViBo, and CaSki) and normal lymphocytes were evaluated. All compounds exhibited an excellent antioxidant activity; remarkably, the peptide derived from L-cystine exhibited the best antioxidant activity, displaying a 2.5-fold increase in radical-scavenging activity when compared with the natural 2-(3’,4’-dihydroxyphenyl)ethanol (hydroxytyrosol, HT). Moreover, this compound was also the most potent antitumor agent against the three human tumor cell lines (IC50 values 108-122 µM), with a 2-7-fold increase in activity when compared with natural DOPAC and HT, used as reference compounds. Importantly, the cytotoxic activity of these phenolic peptidomimetics against normal human lymphocytes was very low, hence confirming their selectivity towards tumor cells. Moreover, a disulfide-containing peptide also exhibited negligible cell necrosis and a high selectivity against tumor cells when compared to normal lymphocytes. Such derivative incorporates two fragments characterized with redox properties, the catechol moiety, and the disulfide linker. Thus, disulfide-containing phenolic peptidomimetics emerge as good lead candidates for the development of a novel family of anti-tumor agents.

Keywords
Polyphenols
peptides
antioxidant
antiradical
anti-proliferative
cervical cancer
1. Introduction

Oxidative stress is a cellular state characterized by the disruption of the balance between pro-oxidants and natural antioxidant defences [1]. Pro-oxidants, including Reactive Oxygen Species (ROS) and Reactive Nitrogen Species (RNS), are comprised of a series of highly reactive compounds (free radicals, hydrogen peroxide, superoxide anion, alkyl peroxides, nitric oxide, etc.) that are produced either endogenously, mainly in mitochondrial oxidative phosphorylation and inflammatory processes, or, alternatively by exogenous agents including ionizing radiation, certain chemicals or pathogens [2]. When present at low concentrations, these pro-oxidants exhibit important biological activities including their acting as redox messengers in intracellular signalling pathways[3]; however, at higher concentrations, they inflict oxidative damage which can be observed in virtually all biomolecules. Oxidative stress has been demonstrated to participate in pathogenic pathways related to cell ageing[4], inflammatory processes [5], the development of cardiovascular (e.g. atherogenesis) [6] and neurodegenerative diseases (e.g. Parkinson and Alzheimer) [7], or carcinogenesis [2]. In fact, regulation of redox homeostasis has been proposed as a key point for preventing or combating such disorders [8].

In order to counteract the deleterious effect of high concentrations of pro-oxidant species, a complex machinery of detoxification has been developed in living organisms, ranging from antioxidant enzymes (e.g. glutathione peroxidase) [9], certain proteins (e.g. albumin) [10], and an heterogeneous group of low molecular weight molecules, comprised of glutathione [11], vitamins [12], carotenes [13], and polyphenols [14]. The latter compounds are ubiquitous phytochemicals especially abundant in fruits, wine, tea, cocoa and virgin olive oil [15] as secondary metabolites [14]. Numerous polyphenols, either naturally-occurring or synthetic are strong scavengers [16,17] of pro-oxidant species. Moreover, they have also shown remarkable bioactivities as antimicrobials [18], antiinflammatory [19], cardio-[20], and neuroprotective [21] agents or glycosidase inhibitors with hypoglycemic effects [22]. Polyphenols have also shown anti-cancer activity against a plethora of human tumor cells [23]. In this respect, hydroxytyrosol, an abundant polyphenol in olive tree and in extra-virgin olive oil is particularly remarkable; epidemiological studies have demonstrated cardio-and neuroprotective effects of hydroxytyrosol, enhancement of immune system, and protection against inflammatory processes [24]. Moreover,its anti-proliferative effects against a series of tumor cell lines are also widely reported [25]. In this context, Fabiani and co-workers have demonstrated the pro-apoptotic effect of hydroxytyrosol against breast, prostate, colon and human leukemia tumor cells by the extracellular production of H2O2 [26].

For all these reasons, polyphenols have emerged as promising candidates to be used as components in the fields of cosmetics, pharmaceuticals, or as additives in functional foods.

2. Results and Discussion
2.1. Chemistry

Herein we report the efficient synthesis of phenolic peptides by coupling amino acid esters and phenolic acids in the presence of a series of promoters; we then evaluated their activities as both antioxidant and anti-proliferative agents. The attachment of different amino acid residues to a phenolic structure might modulate both their bioavailability by improving uptake through the cell membrane, and their biological properties. This might therefore supply valuable information for structure-activity relationship studies.

A plethora of coupling reagents has been reported to date for the activation of the carboxylic acid moiety in peptide bond formation[27]. Among them, a carbodiimide, a phosphonium salt and a uranium salt were selected herein, giving four different synthetic pathways (Scheme 1, Methods A1, A2, B and C). Methods A1 and A2 involve the use of PyBOP (benzotriazol-1-yloxy)tripyrrolidino-phosphonium hexafluorophosphate) with different solvents and basic conditions (anhydrous CH2Cl2 and DIPEA for A1, and anhydrous DMF and Et3N for A2), whereas EDC/HOBt 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride)/1-hydroxybenzotriazole) and HATU (1-(bis(dimethylamino)methylene)-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate) were selected in methods B and C, respectively.

Scheme 1.

Peptide coupling between O-protected phenolic acid 1 and amino acid esters 2-6

In this context, the chosen phenolic templates were 3,4-dihydroxyphenylacetic acid (DOPAC) 12 and its O-protected counterpart 1. The former is a naturally-occurring phenolic compound formed by monoamine oxidase-mediated dopamine metabolism[28], exhibiting relevant biological properties; for instance, sub-stoichiometric concentrations of DOPAC have proven to inhibit the fibrillation of α-synuclein, a critical step in the etiology of Parkinson’s disease[29]. A series of derivatives bearing the 1,2-methylenebenzene scaffold, present in compound 1, have been reported to exhibit different biological properties[30]. Herein, the four methodologies indicated above were applied to compound 1 together with amino acid ester hydrochlorides 2-6, derived from glycine, phenylalanine, valine, serine and tryptophan (Scheme 1). When the uranium salt HATU was used as the coupling reagent (Scheme 1, method C), the formation of the targeted peptide 7 [30]b was observed by 1H-NMR spectroscopy, but the reaction was not complete even after 60 h, and furthermore, a significant amount of side-products was also observed; for all these reasons, this procedure was discarded.

Replacement of HATU with the phosphonium salt PyPOP in CH2Cl2 (method A1) or with the combination of EDC and HOBt as an additive (method B) allowed for the isolation of 7, 9-11 with moderate to good yields (40-74%, or 33-79%, respectively), but the reaction times were still relatively long, especially for the latter conditions (22-60 h). Remarkably, replacement of anhydrous CH2Cl2 by dimethylformamide (DMF) as a solvent, but retaining PyBOP as the coupling reagent (method A2), led to a significant increase in the yield of peptides 7-11 (77%-quantitative), and a general decrease in the reaction time (24-28 h), as depicted in Scheme 1.

The same conditions were employed for DOPAC 12 and amino acid ester derivatives 2-6 (Scheme 2).

Scheme 2.

Peptide coupling between DOPAC 12 and amino acid esters 2-6.

Regarding the formation of the peptidomimetic derived from glycine 13, its synthesis was attempted using the four synthetic methodologies indicated in Scheme 2. Analogously to the preparation of its O-protected counterpart 7, a high amount of side-products was observed in 1H-NMR when Method C was selected; the use of HATU was therefore discarded. Methods A1, A2 and C led to the formation of 13 in 43%, quantitative, and 72% yields, respectively.

Methods A1 and B proceeded with moderate to good yields, but significant amounts of starting materials were observed even after 60 h of reaction. Remarkably, Method A2 has again proven to be the best synthetic choice, where reactions took place in shorter reaction times (15-38 h) giving rise to compounds 13-17 in excellent to quantitative yields (Scheme 2).

Considering the excellent pro-apoptopic properties against HL60 tumor cells observed by hydroxytyrosyl disulfide, and chalcogen-containing polyphenols, recently prepared in our research group[31], we envisioned the possibility of preparing amides derived from L-cystine, where the combination of a dimeric phenolic template, together with a disulfide tether, might reveal significant anti-proliferative activities. Thus, using the optimized conditions of Method A2 (PyBOP, Et3N, DMF), coupling between O-methylidene-protected DOPAC 1 and L-cystine methyl ester dihydrochloride 18 took place in a successful manner, with peptidomimetic 19 being obtained with a yield of 83% (Scheme 3).

Scheme 3.

Preparation of L-cystine derivative 19.

We also attempted the preparation of unprotected disulfide 21 by direct PyBOP-mediated coupling reaction between 12 and 18. Nevertheless, attempts to isolate compound 21 using this procedure were unsuccessful, due to the difficulty in the chromatographic separation of the target compound from side-products derived from the coupling reagent. In order to overcome this problem, the crude coupling reaction between 12 and 18 was conventionally acetylated to furnish tetra-O-acetyl derivative 20, in a 72% overall yield for the two steps (Scheme 4).

Scheme 4.

Preparation of L-cystine and L-cysteine derivatives 21 and 23

Deprotection of the phenolic ester motifs was a more difficult task than previously anticipated. Firstly, strongly basic conditions (NaOMe/MeOH) should be avoided, as they enhance significantly the catechol oxidation to o-quinones, and subsequently to a complex mixture of compounds[32]. Nevertheless, mild conditions (K2CO3/MeOH or NH4OAc/MeOH), or even the use of lipases from C. Antarctica and P. cepacia for the deprotection of 20 failed, as extensive decomposition or a complex mixture of compounds was observed. To our delight, treatment with CsCO3 in a 1:1 mixture of CH2Cl2-MeOH afforded unprotected dimeric disulfide 21 in a good yield (65%). To our knowledge, this is the first example of a L-cystine-containing phenolic peptidomimetic.

The transformation of compound 21 into the L-cysteine derivative 23 was accomplished in a straightforward redox pathway by treatment of 21 with (±)-dithiothreitol (Scheme 4), an inexpensive dithiol quite useful in the mild reduction of disulfide linkages[33]; derivative 23 was isolated with a 44% yield after chromatographic purification.

2.2. Biological evaluation

2.2.1. Antioxidant activity

The capacity of unprotected peptidomimetics 13-17, 21 and 23 to scavenge ROS (free radicals, H2O2 and alkyl hydroperoxides) has been evaluated; the results were compared with natural hydroxytyrosol (HT), an abundant phenolic compound in olives.[32] b This study is an important task for the preliminary evaluation of the compounds, as their inherent antioxidant capacity is responsible for many of their biological properties.

To evaluate the antiradical activity, the 2,2-diphenyl-1-picrylhydrazyl (DPPH) method was used [34]. DPPH is a commercially-available free radical widely used for the quantification of the free radical scavenging activity of an antioxidant agent. DPPH solutions exhibit a deep purple color, with a λmax at 515 nm; the presence of an H-donor acting as a free radical scavenger reduces DPPH, and hence a decrease in the absorbance is observed. Quantification of the potency of an antioxidant is carried out by calculating EC50, that is, the concentration of the antioxidant that scavenges 50% of the original DPPH. Therefore, there is an inverse relationship between EC50 values and the potency of the antioxidant. Calculated EC50 values in the DPPH assay for various compounds, along with that found for HT, are depicted in Figure 1. It can be observed that phenolic peptides 13-17 and 23 exhibit an antiradical activity similar to HT (EC50 = 11.3 ± 1.3 µM), with EC50 values ranging from 10.5-18.4 µM. The activity found for disulfide 21 is especially remarkable as it behaved as a potent antioxidant agent (EC50 = 4.5 ± 0.2 µM), with a 2.5-fold increase in activity when compared with natural HT.

Figure 1.

Evaluation of antiradical activity using the DPPH assay.

To determine H2O2 scavenging activity, we used the methodology developed by Bahorun et al.[35]; in this assay, the peroxidase-mediated oxidation of phenol red with H2O2 is followed spectrophotometrically at 610 nm. The percentage of scavenging exerted by the tested compounds at 1.0 mM concentration is depicted in Figure 2. Compounds 14-16 exhibited virtually the same scavenging activity (60.7-65.1%) as HT (68.8 ± 5.0%); derivatives 17 and 23 showed reduced activity compared to reference compound (34.5 ± 1.0, 54.0 ± 5.3%, respectively). Remarkably, glycine and L-cystine derivatives 13 and 21 behaved as more potent H2O2 scavengers (74.6 ± 2.3% and 77.7 ± 10.3%, respectively) than HT.

Figure 2.

H2O2 scavenging and inhibition of lipid peroxidation exerted by compounds 13-17, 21, 23, DOPAC and HT

The inhibition of the first stages of the lipid peroxidation was also evaluated. For this purpose, linoleic acid was used as the lipid model, and AAPH (2,2'-azobis(2-methylpropionamidine) dihydrochloride) was employed as a free radical initiator that affords alkyl peroxides by thermal degradation of linoleic acid [36]. The inhibition of the lipid peroxidation exerted by the tested compounds at 0.74 mM concentration is depicted in Figure 2. In this assay, HT behaved as a poor antioxidant agent (27.4 ± 8.6% inhibition). Remarkably, peptides derived from glycine 13 (47.7 ± 4.9%), L-phenylalanine 14 (54.8 ± 0.4%), L-serine 16 (45.2 ± 4.6%), L-tryptophan 17 (43.1 ± 11.7%) and L-cysteine 23 (53.9 ± 5.3%) turned out to be more efficient antioxidants for lipid protection than natural HT.

2.2.2. Anti-proliferative activity assay

Some natural polyphenols, like curcumin, ferulic acid, green tea polyphenols, or resveratrol, have been reported to exhibit chemopreventive and chemotherapeutic effects against cervical cancer [37], the second most common cancer worldwide [38]. This prompted us to assess the anti-proliferative effects of the synthesized peptidomimetics and to compare their activity with natural products like DOPAC and HT; in this study, three different cervical cancer cell lines were used: HeLa and CaSki, associated with the human papillomavirus (HPV), and ViBo, which is not associated with HPV.

The degree of necrosis involved in cell death, together with the selectivity against tumor cells compared to normal healthy human lymphocytes were also determined, a key property of chemotherapeutic agents.

In vitro anti-proliferative activity

Anti-proliferative activity against human cervical cancer cell lines (HeLa, CaSki, and ViBo) was determined after 24 h of incubation of the tested compounds (13-16, 20, 21, 23, HT and DOPAC) by crystal violet staining[39], and using MeOH or DMSO as solvent vehicles. Compound 17 could not be tested due to the lack of solubility under the experimental conditions. The inhibitory effect of title compounds on the proliferation of the tumor cell lines was observed to occur in a dose-dependent manner; calculated IC50 values are depicted in Table 1.

Table 1 Antiproliferative activities of tested compounds against cervical cancer (HeLa, CaSki and ViBo cell lines) (mean ± SD, 6 independent experiments)

Natural phenolic compounds HT and DOPAC behaved as moderate antitumor agents, with EC50 values ranging from 227.8-765.0 µM (Table 1). In general, synthetic peptides 13-16 exhibited reduced activity compared to HT and DOPAC. O-Acetylated peptide 20, bearing a disulfide linkage behaved as a slightly better antitumor agent (IC50 199.7-229.6 µM) than HT. Similar results were obtained for unprotected L-cysteine derivative 23. Remarkably, symmetrical fully unprotected compound 21, derived from L-cystine, was found to be the strongest anti-proliferative agent (108.2-121.8 µM), with a 2-7-fold increase in activity in comparison with natural DOPAC and HT. This result indicates that combining a dimeric phenolic template with a disulfide linkage results in a substantial enhancement of the anti-proliferative properties. A similar behavior was found for hydroxytyrosyl disulfide against HL60 tumor cells.

Evaluation of cell necrosis in tumor and non-tumor cells

Cell death can occur via two different pathways: apoptosis (a programmed cell death) or necrosis (an unordered and accidental form of cell death) [40] . As the desired cellular death in a chemotherapy treatment is apoptosis, measurement of the degree of necrosis is an important concern when developing antitumor agents. In order to determine if a necrotic process is involved in the anti-proliferative activity, the lactate dehydrogenase (LDH) assay was carried out. The amount of LDH released to the supernatant is a measure of the loss of membrane integrity, and thus, an indication that a necrotic process takes place. Triton X-100, a non-ionic polyethoxylated surfactant detergent was used as a reference compound to induce cellular lysis [41]. The three tumor cell lines were treated with Triton X-100 in three independent experiments, and released LDH was adjusted to 100%. Cervical tumor cell cultures were also stimulated with the tested compounds at their IC50 concentrations, and the release of LDH was compared to the control groups (Figure 3A-C). The same experiments were also undertaken when studying human lymphocytes instead of human tumor cell lines (Figure 3D).

Figure 3.

Necrotic activity of tested compounds in the human cervical cell lines HeLa (A), CaSki (B), ViBo (C) as well as normal human lymphocytes (D). 7500 cells/well were seeded in 96-well tissue culture plates. After 24 h, the growth medium was discarded and cells were treated with tested compounds at their IC50 concentrations, or with the vehicle solvent (MeOH or DMSO), and evaluated after 24 additional hours by the amount of LDH released to the medium. Experimental data are expressed as the mean ± SD of three independent experiments performed in triplicates. *p < 0.05 vs. vehicle.

Regarding tumor cells, the compound provoking the highest degree of necrosis was the L-valine derivative 15 (37%, 22% and 37% for HeLa, CaSki and ViBo, respectively). The rest of the peptides exhibited an exceptionally low ratio of necrosis; HT, which exhibited a good IC50 value, turned out to be involved to a significant extent in necrosis when considering tumor cells death (35% and 37% for HeLa and ViBo cells, respectively). It is worth mentioning the behavior found for compounds 21 (5% and 4% of necrosis against HeLa and ViBo cells) and 23 (3%, 1% and < 0.5% of necrosis against the three cervical cultures). These data strongly suggest that when these compounds exert their cytotoxic activity against the tumor cell lines studied, they do not act via necrosis but probably via an apoptotic pathway.

Concerning human lymphocytes, the worst compound in terms of necrosis was L-phenylalanine derivative 14. Remarkably, O-unprotected Sulphur-containing peptidomimetics 21 and 23 were found to exhibit a necrotic activity that was even lower than the corresponding solvent used as a vehicle, which could indicate that they do not provoke the death of lymphocytes by necrosis, but even a moderate activation of such cells.

Evaluation of anti-proliferative activity on non-tumor cells

One of the major problems associated with conventional chemotherapeutic treatments is the lack of selectivity of the cytotoxic agents, leading to the appearance of a series of undesirable side-effects; such side effects are predominantly due to the suppression of the host immune system by affecting normal lymphocytes. It is therefore crucial to assess the selectivity of cytotoxic agents towards non-tumor cells. Herein we carried out the study of the anti-proliferative effect of the tested compounds towards peripheral blood lymphocytes. In this assay, an enriched lymphocyte population (ELP) from a normal blood donor is labelled with carboxyfluorescein succinimidyl ester (CFSE), a fluorescent dye widely used in monitoring cell division, and stimulated with phytohemagglutinin (PHA) and/or treated with the tested compounds or with the pure vehicle. After 72 h, cells were harvested and the anti-proliferative activity was determined by flow cytometry[42]; results are depicted in Figure 4. Untreated control proliferating cells were around 87% under normal conditions, and after treatment with the vehicle, roughly 83% (MeOH) and 80% (DMSO). Whereas, peptides 15, 21 and 23 displayed a remarkable selectivity against tumor cell lines, as normal human lymphocytes were practically unaffected, as shown in Figure 4 (i.e. only 2-9% loss of lymphocyte proliferation was observed). These results strongly contrast with the natural HT and DOPAC, which, although active against tumor cells, also elicited a significant decrease in lymphocyte proliferation (roughly 24% and 26%, respectively). Moreover, compound 20, the tetra-O-acetylated derivative of 21 exhibited a decreased selectivity, as it inflicted a ~ 25% decrease in lymphocyte proliferation. This observation suggests the importance of free phenolic OH’s for exhibiting a good selectivity over normal healthy cells.

Figure 4.

Effect of compounds 13-16, 20, 21, 23, HT and DOPAC on the proliferation of peripheral blood lymphocytes as determined by flow cytometry. Cells were seeded in 96-well tissue culture plates, stimulated with phytohemagglutinin (PHA, 10 µg/mL) and treated with title compounds at their IC50 concentrations; analyses were performed after 72 h of incubation, by measuring carboxyfluorescein incorporation. M1 stands for proliferating cells.

3. Materials and Methods
3.1. General procedures

General procedures concerning NMR, MS, TLC visualization and UV-Vis spectroscopy can be found in references [31,43,44].

3.2. Determination of the antioxidant activity

3.2.1. DPPH method

The antiradical activity of the phenolic peptides (DPPH method) was measured using the procedure reported by Prior et al [34].

3.2.2. H2O2 scavenging activity

The H2O2 scavenging activity of the peptides was measured using the procedure reported by Bahorun et al. [35].

3.2.3. Lipid peroxidation assay (ferric thiocyanate method, FTC)

Inhibition of the lipid perxoxidation was measured using the ferric thiocyanate method (FTC) [36].

3.3. General procedures for anti-proliferation assays

3.3.1. Cell culture

General procedures for the cell culture of HeLa, CaSki and ViBo cell lines can be found in references [45,46].

3.3.2. Cell proliferation assay

Assays were performed as reported in reference [47]. (IC50) values were determined after 24 h using crystal violet staining [39].

3.3.3. Cell necrosis study

The necrotic activity was determined as reported in reference [48].

3.3.4. Carboxyfluorescein succinimidyl ester (CSFE) labeling assay

Assays involving carboxyfluorescein were carried out as reported in reference [48].

3.3.5. Statistical analysis

For antioxidant assays, all tests were run in triplicates for each experimental condition. Values are expressed as the confidence interval, which was calculated for n=3, P = 0.95 using the Student’s t-distribution.

For anti-proliferative assays, the median and standard deviation (SD) for six measurements were calculated. Statistical analysis of differences was performed by analysis of variance (ANOVA) using SPSS 10.0 for Windows. A p-value < 0.05 (Tukey’s t-test) was considered to be significant.

3.3.6 General procedures for the preparation of peptides derived from (3,4-methylenedioxy) phenylacetic acid 1

Method A1. To a solution of 1 (50 mg, 0.28 mmol) in anhydrous CH2Cl2 (1.5 mL), PyBOP (166 mg, 0.32 mmol, 1.15 equiv.), the corresponding amino acid ester hydrochloride 2, 4 or 5 (0.28 mmol, 1.0 equiv.) and DIPEA (0.83 mL, 4.86 mmol, 17.4 equiv.) were added under N2. The corresponding mixture was stirred at room temperature for 24-48 h. After that, the solvent was removed under reduced pressure, and the residue was purified as indicated in each case.

Method A2. To a solution of 1 (50 mg, 0.28 mmol) in anhydrous DMF (4.0 mL) at 0ºC, PyBOP (144 mg, 0.28 mmol, 1.0 equiv.), the corresponding amino acid ester hydrochloride 2-6 (0.28 mmol, 1.0 equiv.) and Et3N (0.16 mL, 1.11 mmol, 4.0 equiv.) were added under N2. The corresponding mixture was stirred at room temperature for 15-38 h. Thereafter, the solvent was eliminated under reduced pressure, and the residue was purified as indicated in each case.

Method B. To a solution of 1 (50 mg, 0.28 mmol) in anhydrous CH2Cl2 (3.0 mL), HOBt (41 mg, 0.31 mmol, 1.1 equiv.), EDC (64 mg, 0.33 mmol, 1.2 equiv.), the corresponding amino acid ester hydrochloride 2, 5 or 6 (0.31 mmol, 1.1 equiv.) and Et3N (0.06 mL, 0.42 mmol, 1.5 equiv.) were added under N2. The corresponding mixture was stirred at room temperature for 60 h. Then, the solution was diluted with EtOAc (15 mL) and washed with H2O (1 x 15 mL), and the aqueous phase was further washed with EtOAc (3 x 10 mL). The combined organic fractions were washed with sat. aq. NaHCO3 (1x10 mL) and brine (1x10 mL), dried over MgSO4 and filtrated; the filtrate was concentrated to dryness and the residue was purified by column chromatography using the eluent indicated in each case.

Method C. To a solution of 1 (50 mg, 0.28 mmol) in anhydrous CH2Cl2 (2.0 mL), HATU (116 mg, 0.31 mmol, 1.1 equiv.), glycine ethyl ester hydrochloride 2 (41 mg, 0.29 mmol, 1.05 equiv.) and DIPEA (0.10 mL, 0.58 mmol, 2.1 equiv.) were added under N2. The corresponding mixture was stirred at room temperature for 72 h. Thereafter, the crude reaction was washed with 1M HCl (1 x 10 mL), saturated aqueous NaHCO3 (1 x 10 mL) and brine (1 x 10 mL). The organic phase was dried over MgSO4, filtered and the filtrate was concentrated to dryness.

Spectroscopical data of the new compounds prepared herein can be found in https://idus.us.es/xmlui/handle/11441/53604 (researchrepository of Seville University, Doctoral Thesis of Azucena Marset-Castro).

3.3.7. Ethyl 2-[2’-(3”,4”-methylenedioxyphenyl)acetamido] acetate (7) [30]b

Method A1. Glycine ethyl ester hydrochloride 2 was used (39 mg, 0.28 mmol), and the reaction proceeded for 48 h. Then, the solvent was removed under reduced pressure, and the residue was purified by column chromatography (hexane→1:2 hexane-EtOAc) to give compound 7: 51 mg, 69%.

Method A2. Glycine ethyl ester hydrochloride 2 was used (39 mg, 0.28 mmol), and the reaction proceeded for 24 h. Thereafter, the crude reaction was washed with 1M HCl (1 x 10 mL), sat. aq. NaHCO3 (1 x 10 mL) and brine (1 x 10 mL). The organic layer was dried over MgSO4, filtered and the filtrate was concentrated to dryness to give compound 7: 65 mg, 89%.

Method B. Glycine ethyl ester hydrochloride 2 was used (43 mg, 0.31 mmol), and the reaction proceeded for 48 h. Column chromatography (hexane→1:2 hexane-EtOAc) yielded compound 7: 58 mg, 79%.

Method C. Glycine ethyl ester hydrochloride 2 was used (41 mg, 0.29 mmol), and the reaction proceeded for 72 h. After removal of the solvent, the crude product was analyzed by 1H-NMR.

RF 0.62 (1:2 hexane-EtOAc); 1H-NMR (300 MHz, CDCl3) δ 6.79-6.76 (m, 2H, H-2’’, H-5’’), 6.71 (dd, 1H, J2’’,6’’=1.7 Hz, J5’’,6’’=7.8 Hz, H-6’’), 6.03 (brs, 1H, NH), 5.94 (s, 2H, O-CH2-O), 4.17 (q, 2H, JH,H=7.2 Hz, CH2-CH3), 3.98 (d, 2H, J2,NH=5.3 Hz, H-2), 3.51 (s, 2H, H-2’), 1.25 (t, 3H, CH2-CH3); 13C-NMR (75.5 MHz, CDCl3) δ 171.3, 169.9 (C-1’, C-1), 148.2 (C-3’’), 147.1 (C-4’’), 128.1 (C-1’’), 122.8 (C-6’’), 109.9 (C-2’’), 108.7 (C-5’’), 101.2 (O-CH2-O), 61.6 (CH2-CH3), 43.1 (C-2’), 41.6 (C-2), 14.2 (CH2-CH3); CI-MS m/z 265 ([M]+, 57%); HRCI-MS calculated for C13H15NO5 ([M]+): 265.0950, found: 265.0946.

3.3.8. Ethyl (S)-2-[2’-(3”,4”-methylenedioxyphenyl)acetamido]-3-phenylpropanoate (8)

Method A2. L-Phenylalanine ethyl ester hydrochloride 3 was used (64 mg, 0.28 mmol), and the reaction proceeded for 38 h. Column chromatography (hexane→1:1 hexane-EtOAc) yielded 8: 76 mg, 77%.$[α]_{D}^{26}$-5 (c 0.94, MeOH); RF 0.48 (1:2 hexane-EtOAc 1:2); 1H-NMR (300 MHz, CD3OD) δ 7.28-7.16 (m, 3H, Ar-H), 7.15-7.10 (m, 2H, Ar-H), 6.70 (d, 1H, J5’’,6’’=7.8 Hz, H-5’’), 6.67 (d, 1H, J2’’,6’’=1.3 Hz, H-2’’), 6.63 (dd, 1H, H-6’’), 5.91 (s, 2H, O-CH2-O), 4.64 (dd, 1H, J2,3a=5.7 Hz, J2,3b=8.8 Hz, H-2), 4.13 (q, 2H, JH,H=7.2 Hz, CH2-CH3), 3.39 (s, 2H, H-2’), 3.14 (dd, 1H, J3a,3b=13.9 Hz, H-3a), 2.96 (dd, 1H, H-3b), 1.20 (t, 3H, CH2-CH3); 13C-NMR (75.5 MHz, CD3OD) δ 173.9, 173.0 (C-1’, C-1), 149.2 (C-3’’), 148.0 (C-4’’), 138.0 (Ar-C), 130.2 (x2) (2Ar-C), 130.1 (x2) (2Ar-C), 129.5 (C-1’’), 127.9 (Ar-C), 123.4 (C-6’’), 110.4 (C-2’’), 109.1 (C-5’’), 102.3 (O-CH2-O), 62.4 (CH2-CH3), 55.3 (C-2), 43.1 (C-2’), 38.3 (C-3), 13.4 (CH2-CH3); CI-MS m/z 355 ([M]+, 52%); HRCI-MS calculated for C20H21NO5 ([M]+): 355.1420, found: 355.1408.

3.3.9. Ethyl (S)-3-methyl-2-[2’-(3”,4”-methylenedioxyphenyl) acetamido] butanoate (9)

Method A1. L-Valine ethyl ester hydrochloride 4 was used (50 mg, 0.28 mmol), and the reaction proceeded for 44 h. Column chromatography (hexane→2:1 hexane-EtOAc) yielded 9: 34 mg, 40%.

Method A2. L-Valine ethyl ester hydrochloride 4 was used (50 mg, 0.28 mmol), and the reaction proceeded for 17 h. Column chromatography (hexane→1:2 hexane-EtOAc) yielded compound 9: 81 mg, 95%. $[α]_{D}^{24}$ +9 (c 0.98, CH2Cl2); RF 0.54 (1:2 hexane-EtOAc); CI-MS m/z 307 ([M]+, 64%); HRCI-MS m/z calcd for C16H21NO5 ([M]+): 307.1420, found: 307.1424.

3.3.10. Ethyl (S)-3-Hydroxy-2-[2’-(3”,4”-methylenedioxyphenyl) acetamido]propanoate (10)

Method A1. L-Serine ethyl ester hydrochloride 5 was used (47 mg, 0.28 mmol), and the reaction proceeded for 30 h. Column chromatography (hexane→1:3 hexane-EtOAc) resulted in compound 10: 61 mg, 74%.

Method A2. L-Serine ethyl ester hydrochloride was used (47 mg, 0.28 mmol), and the reaction proceeded for 19 h. Column chromatography (hexane→1:3 hexane-EtOAc) yielded compound 10: 65 mg, 80%.

Method B. L-Serine ethyl ester hydrochloride was used (52 mg, 0.31 mmol), and the reaction proceeded for 60 h. Liquid-liquid extractions, as described in the general procedure, yielded pure 10: 45 mg, 55%. $[α]_{D}^{23}$ +3 (c 1.22, MeOH); RF 0.64 (1:2 hexane-EtOAc); HRCI-MS calcd for C14H17NO6 ([M]+): 295.1056, found: 295.1049.

3.3.11. Methyl (S)-3-(1”’H-indol-3”’-yl)-2-[2’-(3”,4”-methylenedioxyphenyl) acetamido]propanoate (11)

Method A2. L-tryptophan methyl ester hydrochloride 6 was used (71 mg, 0.28 mmol), and the reaction proceeded for 15 h. Column chromatography (hexane→1:7 hexane-EtOAc) resulted in compound 11: 106 mg, quant.

Method B. L-tryptophan methyl ester hydrochloride 6 was used (78 mg, 0.31 mmol), and the reaction proceeded for 60 h. Column chromatography (hexane→1:2 hexane-EtOAc) yielded compound 11: 35 mg, 33%. $[α]_{D}^{24}$ +28 (c 1.10, (CH3)2CO); RF 0.55 (1:2 hexane-EtOAc); CI-MS 381 ([M]+, 21%); HRCI-MS calcd for C21H21N2O5 ([M+H]+): 381.1450, found: 381.1436.

3.3.12. General procedures for the preparation of peptides derived from (3,4-dihydroxyphenyl)acetic acid 12 (DOPAC)

Method A1. To a solution of DOPAC (50 mg, 0.30 mmol) in anhydrous CH2Cl2 (1.5 mL), PyBOP (178 mg, 0.34 mmol, 1.15 equiv.), glycine ethyl ester hydrochloride 2 (42 mg, 0.30 mmol, 1.0 equiv.) and DIPEA (0.89 mL, 5.20 mmol, 17.4 equiv.) were added under N2. The corresponding mixture was stirred at room temperature during 48 h. Then, the solvent was evaporated under reduced pressure, and the residue was purified by column chromatography (CH2Cl2→10:1 CH2Cl2-MeOH).

Method A2. To a solution of DOPAC (50 mg, 0.30 mmol) in anhydrous DMF (4.0 mL) at 0ºC, PyBOP (155 mg, 0.30 mmol, 1.0 equiv.), the corresponding amino acid ester hydrochlorides 2-6 (0.30 mmol, 1.0 equiv.) and Et3N (0.17 mL, 1.19 mmol, 4.0 equiv.) were added under N2. The corresponding mixture was stirred at room temperature for 16-38 h. Thereafter, the solvent was eliminated under reduced pressure, and the residue was purified by column chromatography using the eluant indicated in each case.

Method B. To a solution of DOPAC (50 mg, 0.30 mmol) in anhydrous CH2Cl2 (3.0 mL), HOBt (44 mg, 0.33 mmol, 1.1 equiv.), EDC (68 mg, 0.36 mmol, 1.2 equiv.), glycine ethyl ester hydrochloride 2 (46mg, 0.33 mmol, 1.1 equiv.) and Et3N (0.06 mL, 0.42 mmol, 1.5 equiv.) were added under N2. The corresponding mixture was stirred at room temperature for 60 h. Then, the solution was diluted with EtOAc (15 mL) and washed with H2O (1 x 15 mL), and the aqueous phase was further washed with EtOAc (3 x 10 mL). The combined organic fractions were washed with 1M HCl (1 x 10 mL), sat. aq. NaHCO3 (1 x 10 mL) and brine (1x10 mL), dried over MgSO4 and filtered; the filtrate was then concentrated to dryness and the residue was purified by column chromatography (CH2Cl2→10:1 CH2Cl2-MeOH).

Method C. To a solution of DOPAC (50 mg, 0.30 mmol) in anhydrous CH2Cl2 (2.0 mL), HATU (124 mg, 0.33 mmol, 1.1 equiv.), glycine ethyl ester hydrochloride 2 (44 mg, 0.31 mmol, 1.05 equiv.) and DIPEA (0.11 mL, 0.62 mmol, 2.1 equiv.) were added under N2. The corresponding mixture was stirred at room temperature for 72 h. Thereafter, the crude reaction was washed with 1M HCl (1x10 mL), sat. aq. NaHCO3 (1x10 mL) and brine (1x10 mL). The organic phase was dried over MgSO4, filtered and the filtrate was concentrated to dryness.

3.3.13. Ethyl 2-[2’-(3”,4”-Dihydroxyphenyl)acetamido]acetate (13)

Method A1. Glycine ethyl ester hydrochloride 2 was used (42 mg, 0.30 mmol). Chromatographic purification afforded 13: 32 mg, 43%.

Method A2. Glycine ethyl ester hydrochloride 2 was used (42 mg, 0.30 mmol), and the reaction proceeded for 22 h. Column chromatography (CH2Cl2→10:1 CH2Cl2-MeOH) gave 13: 76 mg, quant.

Method B. Glycine ethyl ester hydrochloride 2 was used (46 mg, 0.33 mmol). Column chromatography gave 13: 54 mg, 72%.

Method C. Glycine ethyl ester hydrochloride 2 was used (44 mg, 0.31 mmol). After removal of the solvent, the crude product was analyzed by 1H-NMR.

Rf 0.81 (40:1 CH2Cl2-MeOH); HRLSI-MS calcd for C12H15NNaO5 ([M + Na]+: 276.0848, found: 276.0842.

3.3.14. Ethyl (S)-2-[2’-(3”,4”-dihydroxyphenyl)acetamido]-3-phenylpropanoate (14)

Method A2. L-Phenylalanine ethyl ester hydrochloride 3 was used (68 mg, 0.30 mmol), and the reaction proceeded for 38 h. Column chromatography (hexane→1:4 hexane-EtOAc) afforded 14: 102 mg, quant. $[α]_{D}^{21}$-4 (c 0.95, MeOH); RF 0.40 (10:1 CH2Cl2-MeOH ); HRCI-MS calcd for C19H22NO5 ([M+H]+): 344.1498, found: 344.1503.

3.3.15. Ethyl (S)-2-[2’-(3”,4”-dihydroxyphenyl)acetamido]-3-methylbutanoate (15)

Method A2. L-Valine ethyl ester hydrochloride 4 was used (54 mg, 0.30 mmol), and the reaction proceeded for 16 h. Column chromatography (hexane→1:2 hexane-EtOAc) yielded 15. Yield: 84 mg, 96%. $[α]_{D}^{25}$-6 (c 0.71, DMSO); RF 0.38 (10:1 CH2Cl2-MeOH); HRCI-MS calcd for C15H21NO5 ([M]+): 295.1420, found: 295.1428; calcd for C15H22NO5 ([M + H]+): 296.1498, found: 296.1488.

3.3.16. Ethyl (S)-2-[2’-(3”,4”-Dihydroxyphenyl)acetamido]-3-hydroxypropanoate (16)

Method A2. L-Serine ethyl ester hydrochloride 5 was used (50 mg, 0.30 mmol), and the reaction proceeded for 38 h. Column chromatography (hexane→1:2 hexane-EtOAc) afforded 16: 84 mg, quant.; $[α]_{D}^{25}$-6 (c 1.02, MeOH); RF 0.39 (1:2 hexane-EtOAc); HRCI-MS calcd for C13H17NO6 ([M]+): 283.1056, found: 283.1054.

3.3.17. Methyl (S)-2-[2’-(3”,4”-dihydroxyphenyl)acetamido]-3-(1”’ H-indol-3”’-yl)propanoate (17)

Method A2. L-Tryptophan methyl ester hydrochloride 6 was used (76 mg, 0.30 mmol), and the reaction proceeded for 38 h. Column chromatography (hexane→1:7 hexane-EtOAc) yielded 17: 110 mg, quant. $[α]_{D}^{23}$+4 (c 1.15, DMF); RF 0.33 (1:2 hexane-EtOAc); HRLSI-MS calcd for C20H20N2NaO5 ([M + Na]+): 391.1270, found: 391.1276.

3.3.18. Methyl (R,R)-3,3’-Dithiobis{2-[2”-(3”’,4”’-methylenedioxyphenyl) acetamido]propanoate} (19)

To a suspension of L-cystine dimethyl ester dihydrochloride 18 (102 mg, 0.03 mmol) in DMF (4.0 mL) at 0ºC, Et3N (0.33 mL, 2.40 mmol), (3,4-methylenedioxy)phenylacetic acid 1 (108 mg, 0.60 mmol) and PyBOP (312 mg, 0.60 mmol) were added under N2. The corresponding mixture was kept under stirring at room temperature and in the dark for 7 h. Then, the reaction was concentrated to dryness and the residue was purified by column chromatography (CH2Cl2→5:1 CH2Cl2-MeOH) to give 19 as a white solid. Yield: 147 mg, 83%; $[α]_{D}^{28}$ +77 (c 1.0, CH2Cl2); RF 0.44 (20:1 CH2Cl2-MeOH); m.p. 133-135 ºC; CI-MS m/z 593 ([M + H]+, 4%); HRCI-MS calcd for C26H29N2O10S2 ([M + H]+): 593.1264, found: 593.1245.

3.3.19. Methyl (R,R)-3,3’-Dithiobis{2-[2’’-(3’’’,4’’’-diacetoxyphenyl)acetamido]propanoate} (20)

To a suspension of L-cystine dimethyl ester dihydrochloride 18 (102 mg, 0.30 mmol) in DMF (4.0 mL), Et3N (0.33 mL, 2.40 mmol), DOPAC (101 mg, 0.60 mmol) and PyBOP (312 mg, 0.60 mmol) were added under N2. The corresponding mixture was stirred at room temperature in the dark for 12 h. Then, the solvent was removed under reduced pressure and the residue was conventionally acetylated with a 1:1 Ac2O-Py mixture (2.0 mL) for 6.5 h. Then, the excess of Ac2O was hydrolyzed with crushed ice, and concentrated to dryness; the residue was purified by column chromatography (CH2Cl2→60:1 CH2Cl2-MeOH) to give 20. Yield: 158 mg, 72%; $[α]_{D}^{25}$ +1 (c 1.0, CH2Cl2); RF 0.64 (10:1 CH2Cl2-MeOH); LSI-MS m/z 759 ([M + Na]+, 41%); HRLSI-MS calcd for C32H36N2NaO14S2 ([M + Na]+): 759.1506, found: 759.1502.

3.3.20. Methyl (R,R)-3,3’-Dithiobis{2-[2’’-(3’’’,4’’’-dihydroxyphenyl)acetamido]propanoate} (21)

To a solution of 20 (96 mg, 0.13 mmol) in a 1:1 CH2Cl2-MeOH mixture (2.0 mL) CsCO3 was added (10 mg, 0.031 mmol), and the corresponding mixture was stirred at room temperature in the dark under inert atmosphere for 30 min. Then, the crude reaction was neutralized using Amberlite IR-120(H+) resin, filtered, washed with MeOH, and the filtrate was concentrated to dryness. The residue was purified by column chromatography (CH2Cl2→20:1 CH2Cl2-MeOH) to give 21 as a yellowish syrup. Yield: 48 mg, 65%; $[α]_{D}^{26}$ +28 (c 1.0, MeOH); RF 0.38 (10:1 CH2Cl2-MeOH); LSI-MS m/z 591 ([M + Na]+, 11%); HRLSI-MS calcd for C24H28N2NaO10S2 ([M + Na]+): 591.1083, found: 591.1058.

3.3.21. Methyl (R)-2-[2’-(3’’,4’’-dihydroxyphenyl)acetamido]-3-mercaptopropanoate (23) [49]

To a solution of 21 (43 mg, 0.076 mmol) in an anhydrous 1:1 CH2Cl2-MeOH mixture (2.0 mL) (±)-dithiothreitol was added (12 mg, 0.076 mmol). The corresponding mixture was stirred at room temperature in the dark under inert atmosphere for 2 h. Then it was concentrated to dryness and the residue was purified by column chromatography (hexane→1:10 hexane-EtOAc) to give 23 as a syrup. Yield: 19 mg, 44%; $[α]_{D}^{25}$-15 (c 1.2, MeOH); RF 0.48 (1:5 1:10 hexane-EtOAc); HRLSI-MS calcd for C12H15NNaO5S ([M + Na]+): 308.0569, found: 308.0569.

4. Conclusions

A one-step and almost quantitative transformation of O-unprotected DOPAC and L-amino acid esters (glycine, phenylalanine, valine, serine, tryptophan and cystine) into phenolic peptidomimetics has been achieved using PyBOP as the coupling reagent. A peptide derived from L-cysteine was also been prepared by dithiothreitol-mediated reduction of the disulfide linkage of the L-cystine derivative.

Title compounds have been evaluated as scavengers of ROS (antiradical and H2O2 scavenging and inhibition of lipid peroxidation) as well as anti-proliferative agents against three human cervical cancer cell lines (HeLa, CaSki and ViBo); the degree of cell necrosis, and the selectivity of the various compounds against tumor cells over normal human lymphocytes has been evaluated. Remarkably, disulfide-containing peptide 21 was found to be the most active compound, with exceptional antioxidant properties, stronger than natural HT and a substantial anti-proliferative activity (IC50 108.2-121.8 µM), with a 2-7-fold increase in this cytotoxic activity when compared with HT and DOPAC; moreover, its mode of action involved almost no cell necrosis, and essentially did not affect normal lymphocytes. Therefore, compound 21 might constitute a good lead candidate for the development of a novel family of anti-proliferative agents.

Acknowledgments

We thank the Dirección General de Investigaci ón of Spain (CTQ2016-78703-P), Junta de Andalucía (FQM134), FEDER (501100008530), and Mexican SEP-CONACYT (255881, 258156, DGAPA, PAPIIT IN216718 and IN220916) for financial support. A.M.-C. also thanks the Junta de Andalucía for the award of a fellowship.

Electronic Supplementary Information (ESI) available: 1H-and 13C-NMR spectra of compounds 7-11, 13-17, 20, 21, 23.

Conflict of interest

We have no conflict of interest to declare.

References
[1]
Kohen R, Nyska A. Oxidation of biological systems: oxidative stress phenomena, antioxidants, redox reactions, and methods for their quantification. Toxicol Pathol, 2002; 30: 620-50. 10.1080/01926230290166724http://journals.sagepub.com/doi/10.1080/01926230290166724
[2]
Klaunig JE, Kamendulis LM, Hocevar BA. Oxidative stress and oxidative damage in carcinogenesis. Toxicol Pathol, 2010; 38(1): 96-109. 10.1177/01926233093564532001935663747f9d66fd83781f86b2c3e449c2e0http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F20019356http://journals.sagepub.com/doi/10.1177/0192623309356453
[3]
D’Autréaux B, Toledano MB. ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat Rev Mol Cell Biol, 2007; 8(10): 813-824.
[4]
Sohal RS, Orr WC. The redox stress hypothesis of aging. Free Radic Biol Med, 2012; 52(3): 539-555. 10.1016/j.freeradbiomed.2011.10.44522080087e47e2d6b86a7a0e324e53c934655a7e3http%3A%2F%2Feuropepmc.org%2Farticles%2FPMC3267846https://linkinghub.elsevier.com/retrieve/pii/S0891584911011099
[5]
Mittal M, Siddiqui MR, Tran K, Reddy SP, Malik AB. Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal, 2014; 20(7): 1126-1167. 10.1089/ars.2012.514939290101a3bf829310937550ad83b369c6121behttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F23991888http://www.liebertpub.com/doi/10.1089/ars.2012.5149
[6]
Costa VM, Carvalho F, Duarte JA, Bastos ML, Remião F. The heart as a target for xenobiotic toxicity: the cardiac susceptibility to oxidative stress. Chem Res Toxicol, 2013; 26(9): 1285-1311. 10.1021/tx400130v23902227de55ea32dcddafd4631ef57b3f55a7b1http%3A%2F%2Fpubs.acs.org%2Fdoi%2Fpdf%2F10.1021%2Ftx400130vhttp://pubs.acs.org/doi/10.1021/tx400130v
[7]
Rosini M, Simoni E, Milelli A, Minarini A, Melchiorre C. Oxidative stress in Alzheimer’s disease: are we connecting the dots? J Med Chem, 2014; 57(7): 2821-2831. 10.1021/jm400970mhttp://pubs.acs.org/doi/10.1021/jm400970m
[8]
Gorrini C, Harris IS, Mak TW. Modulation of oxidative stress as an anticancer strategy. Nat Rev Drug Discov, 2013; 12(12): 931-947. 10.1038/nrd40022428778114f7c26bd53fcd33533748a8e5ace489http%3A%2F%2Fwww.nature.com%2Fnrd%2Fjournal%2Fv12%2Fn12%2Ffig_tab%2Fnrd4002_F3.htmlhttp://www.nature.com/nrd/journal/v12/n12/fig_tab/nrd4002_F3.html
[9]
Matés JM , Effects of antioxidant enzymes in the molecular control of reactive oxygen species toxicology. Toxicology, 2000; 153(1-3): 83-104. 10.1016/S0300-483X(00)00306-11109094910fd73c2aece17a22849327459381ffahttp%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS0300483X00003061https://linkinghub.elsevier.com/retrieve/pii/S0300483X00003061
[10]
Roche M, Rondeau P, Singh NR, Tarnus E, Bourdon E. The antioxidant properties of serum albumin. FEBS Lett, 2008; 582(13): 1783-1787. 10.1016/j.febslet.2008.04.0575870b77ee3905addb97fb05a81156bedhttp%3A%2F%2Feurheartjsupp.oxfordjournals.org%2Flookup%2Fexternal-ref%3Faccess_num%3D18474236%26amp%3Blink_type%3DMED%26amp%3Batom%3D%252Fehjsupp%252F17%252Fsuppl_C%252FC34.atomhttp://doi.wiley.com/10.1016/j.febslet.2008.04.057
[11]
Ferreira B, Mendes F, Osorio N, Caseiro A, Gabriel A. Valado A. Glutathione in multiple sclerosis. Br J Biomed Sci, 2013; 70(2): 75-79. 10.1080/09674845.2013.11669939https://www.tandfonline.com/doi/full/10.1080/09674845.2013.11669939
[12]
Pallauf K, Bendall JK, Scheiermann C, Watschinger K, Hoffmann J, Roeder T , et al. Vitamin C and lifespan in model organisms. Food Chem Toxicol, 2013; 58: 255-263. 10.1016/j.fct.2013.04.046236437001801a203de20660007c7a1f1fb70cc7ahttp%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F23643700https://linkinghub.elsevier.com/retrieve/pii/S0278691513002895
[13]
Stahl W, Sies H. Antioxidant activity of carotenoids. Mol Aspects Med, 2003; 24(6): 345-351. 10.1016/S0098-2997(03)00030-Xhttps://linkinghub.elsevier.com/retrieve/pii/S009829970300030X
[14]
Quideau S, Deffieux D, Douat-Casassus C, Pouységu L. Plant polyphenols: chemical properties, biological activities, and synthesis. Angew Chem Int Ed Eng, 2011; 50(3): 586-621. 10.1002/anie.201000044212261377b0a98f932f845854c7230895e78913dhttp%3A%2F%2Fonlinelibrary.wiley.com%2Fdoi%2F10.1002%2Fanie.201000044%2Fpdfhttp://doi.wiley.com/10.1002/anie.v50.3
[15]
Soto-Vaca A, Gutiérrez A, Losso JN, Xu Z, Finley JW. Finley, Evolution of phenolic compounds from color and flavor problems to health benefits. J Agric Food Chem, 2012; 60(27): 6658-6677. 10.1021/jf300861c386f1abc8dae4424ac55bd33a2b23670http%3A%2F%2Fwww.tandfonline.com%2Fservlet%2Flinkout%3Fsuffix%3Dcit0119%26amp%3Bdbid%3D8%26amp%3Bdoi%3D10.1080%252F10408398.2016.1235546%26amp%3Bkey%3D22568556http://pubs.acs.org/doi/10.1021/jf300861c
[16]
Ishimoto H, Tai A, Yoshimura M, Amakura Y, Yoshida T, Hatano T, Hito H. Antioxidative properties of functional polyphenols and their metabolites assessed by an ORAC assay. Biosci Biotechnol Biochem, 2012; 76(2): 395-359. 10.1271/bbb.11071722313776f8b9a1751c72245b18b362f59a9b860fhttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F22313776%2Fhttp://www.tandfonline.com/doi/full/10.1271/bbb.110717
[17]
Franco N, Galeano-Diaz T, López Ó, Fernández-Bolaños JG, Sánchez J, de Miguel C, et al. Phenolic compounds and antioxidant capacity of virgin olive oil. Food Chem, 2014; 163: 289-298. 10.1016/j.foodchem.2014.04.09124912728b356057cc250d0f1ab45d4beb219c6e6http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F24912728https://linkinghub.elsevier.com/retrieve/pii/S030881461400661X
[18]
a) Banerjee DR, Dutta D, Saha B, Bhattacharyya S, Senapati K, Das AK , et al. Design, synthesis and characterization of novel inhibitors against mycobacterial β-ketoacyl CoA reductase FabG4. Org Biomol Chem, 2014; 12(1): 73-85. b) Park KD, Cho SJ. Synthesis and antimicrobial activities of 3-O-alkyl analogues of (+)-catechin: improvement of stability and proposed action mechanism. Eur J Med Chem, 2010; 45(3):1028-1033. 10.1039/c3ob41676cc78df6da968f4a76ff8904319dde8d93http%3A%2F%2Fpubs.rsc.org%2Fen%2Fcontent%2Farticlepdf%2F2013%2Fob%2Fc3ob41676chttp://xlink.rsc.org/?DOI=C3OB41676C
[19]
a) Pu H-L, Chiang W-L, Maiti B, Liao Z-X, Ho Y-C, Shim MS , et al. Nanoparticles with dual responses to oxidative stress and reduced pH for drug release and anti-inflammatory applications. ACS Nano, 2014; 8(2): 1213-1221. b) Lu D-L, Ding D-J, YanW-J, Li R-R, Dai F,Wang Q, et al. Influence of glucuronidation and reduction modifications of resveratrol on its biological activities. ChemBioChem, 2013; 14(9):1094-1104. 10.1021/nn4058787b02530437d076bf894c746ed39300de2http%3A%2F%2Fwww.tandfonline.com%2Fservlet%2Flinkout%3Fsuffix%3DCIT0031%26amp%3Bdbid%3D8%26amp%3Bdoi%3D10.1080%252F09205063.2018.1433108%26amp%3Bkey%3D24386907http://pubs.acs.org/doi/10.1021/nn4058787
[20]
Andriantsitohaina R, Auger C, Chataigneau T, Étienne-Selloum N, Li H, Martínez MC , et al. Molecular mechanisms of the cardiovascular protective effects of polyphenols. Br J Nutr, 2012; 108(9): 1532-1549. 10.1017/s0007114512003406229351437819d3b7ebb22b03bae9dfbcb1cc384chttp%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F22935143https://www.cambridge.org/core/product/identifier/S0007114512003406/type/journal_article
[21]
Narita K, Hisamoto M, Okuda T, Takeda S. Differential neuroprotective activity of two different grape seed extracts. PLOS ONE, 2011; 6(1): e14575. 10.1371/journal.pone.001457521283677d33be9c7ef92e52dd47c045d180f16dbhttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F21283677https://dx.plos.org/10.1371/journal.pone.0014575
[22]
T. Tsujita, T. Shintani and H. Sato. α-Amylase inhibitory activity from nut seed skin polyphenols. 1. Purification and characterization of almond seed skin polyphenols. J Agric Food Chem, 2013; 61(19): 4570-4576. 10.1021/jf400691q23614772bb1b5308b9c10634e7c21c6e3b0a6adahttp%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F23614772http://pubs.acs.org/doi/10.1021/jf400691q
[23]
Korkina LG, Pastore S, Dellambra E, De Luca C. polyphenols. Curr Med Chem, 2013; 20(7): 852-868.
[24]
Hu T, He X-W, Jiang J-G, Xu X-L. Hydroxytyrosol and its potential therapeutic effects. J Agric Food Chem, 2014; 62(7): 1449-1455. 10.1021/jf405820v24479643bfd67c9968343f478adfd8e01603c8b6http%3A%2F%2Fpubs.acs.org%2Fdoi%2Fabs%2F10.1021%2Fjf405820vhttp://pubs.acs.org/doi/10.1021/jf405820v
[25]
Bernini R, Merendino N, Romani A, Velotti F. Naturally occurring hydroxytyrosol: synthesis and anticancer potential. Curr Med Chem, 2013; 20(5): 655-670. 10.2174/092986713804999367<![CDATA[http://www.eurekaselect.com/openurl/content.php?genre=article&issn=0929-8673&volume=20&issue=5&spage=655]]>
[26]
a) Fabiani R, Fuccelli R, Pieravanti F, De Bartolomeo A, Morozzi G. Production of hydrogen peroxide is responsible for the induction of apoptosis by hydroxytyrosol on HL60 cells. Mol Nutr Food Res, 2009; 53(7): 887-896. b) Fabiani R, Sepporta MV, Rosignoli P, De Bartolomeo A, Crescimanno M, Morozzi G. Anti-proliferative and proapoptotic activities of hydroxytyrosol on different tumour cells: the role of extracellular production of hydrogen peroxide. Eur J Nutr, 2012; 51(4):455-464. 10.1002/mnfr.20080037619536871ffceb5bf6174929d790d16cc31379b8bhttp%3A%2F%2Fonlinelibrary.wiley.com%2Fdoi%2F10.1002%2Fmnfr.200800376%2Ffullhttp://doi.wiley.com/10.1002/mnfr.v53%3A7
[27]
a) Valeur E, Bradley M. Amide bond formation: beyond the myth of coupling reagents. Chem Soc Rev, 2009; 38(2): 606-631. b) Han S-Y, Kim Y-A. Recent development of peptide coupling reagents in organic synthesis. Tetrahedron, 2004; 60(11):2447-2467. 10.1039/B701677Hhttp://xlink.rsc.org/?DOI=B701677H
[28]
Gesi M, Santinami A, Ruffoli R, Conti G, Fornai F. Novel aspects of dopamine oxidative metabolism (confounding outcomes take place of certainties). Pharmacol Toxicol, 2001; 89(5): 217-224. 10.1111/j.1600-0773.2001.890501.x1188197432e6ee1d21e1baa025c02baab0e3aa80http%3A%2F%2Fonlinelibrary.wiley.com%2Fdoi%2F10.1111%2Fj.1600-0773.2001.890501.x%2Ffullhttp://doi.wiley.com/10.1034/j.1600-0773.2001.d01-151.x
[29]
Zhou W, Gallagher A, Hong D-P, Long C, Fink AL, Uversky VN. At low concentrations, 3,4-dihydroxyphenylacetic acid (DOPAC) binds non-covalently to alpha-synuclein and prevents its fibrillation. J Mol Biol, 2009; 388(3): 597-610. 10.1016/j.jmb.2009.03.0532719782410d33e42dae6a9acca9cc981dbc588ahttp%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F19328209https://linkinghub.elsevier.com/retrieve/pii/S0022283609003398
[30]
a) Maia RC, Silva LL, Mazzeu EF, Fumian MM, de Rezende CM, Doriguetto AC. Synthesis and analgesic profile of conformationally constrained N-acylhydrazone analogues: discovery of novel N-arylideneamino quinazolin-4(3H)-one compounds derived from natural safrole. Bioorg Med Chem, 2009; 17(18): 6517-6525. b) de Magalhães Moreira DR, Lima Leite AC, Pinheiro Ferreira PM, da Costa PM, Costa Lotufo LV, de Moraes MO, et al. Synthesis and antitumour evaluation of peptidyl-like derivatives containing the 1,3-benzodioxole system. Eur J Med Chem, 2007; 42(3): 351-357. c) Ishizuka N, Matsumura K-i, Sakai K, Fujimoto M, Mihara S-I, Yamamori T. Structure-activity relationships of a novel class of endothelin-A receptor antagonists and discovery of potent and selective receptor antagonist, 2-( benzo[1,3]dioxol-5-yl)-6-isopropyloxy-4-, 2002; 45(10):2041-2055.
[31]
a) Begines P, Oliete A, López Ó, Maya I, Plata GB, Padrón JM , et al. Chalcogen-containing phenolics as antiproliferative agents. Future Med Chem, 2018; 10(3): 319-334. b) Marset A, Begines P, López Ó, Maya I, García-Aranda N, Schwartz Jr S, et al. Design of chalcogen-containing norepinephrines: efficient GPx mimics and strong cytotoxic agents against HeLa cells. Future Med Chem, 2016; 8(18): 2185-2195. c) Calcatierra V, López Ó, Fernández-Bolaños JG, Plata GB, Padrón JM. Phenolic thio-and selenosemicarbazones as multi-target drugs. Eur J Med Chem, 2015; 94: 63-72. d) Sepporta MV, López-García MÁ, Fabiani R, Maya I, Fernández-Bolaños JG. Enhanced chemopreventive activity of hydroxytyrosol on HL60 and HL60R cells by chemical conversion into thio derivatives. Eur J Pharm Sci, 2013; 48(4-5):790-798. 10.4155/fmc-2017-00532940008731e28e95c48ebcea8def2ae6402852cdhttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F29400087http://www.ncbi.nlm.nih.gov/pubmed/29400087
[32]
a) Tuominen A, Sundman T. Stability and oxidation products of hydrolysable tannins in basic conditions detected by HPLC/DAD-ESI/QTOF/MS. Phytochem Anal, 2013; 24(5): 424-35. b) Fernández-Bolaños JG, López Ó, Fernández-Bolaños J, Rodríguez-Gutiérrez G. Hydroxytyrosol and derivatives: isolation, synthesis, and biological properties. Curr Org Chem, 2008; 12(6):442-463. 10.1002/pca.2456237985443add7f86d9dc5508320babe07119d368http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F23798544http://europepmc.org/abstract/med/23798544
[33]
Lamoureux GV, Whitesides GM. Synthesis of dithiols as reducing agents for disulfides in neutral aqueous solution and comparison of reduction potentials, J Org Chem, 1993; 58(3): 633-641. 10.1021/jo00055a0159f031cf5a0ab05c437cf71f5153bd6fdhttp%3A%2F%2Fpubs.acs.org%2Fdoi%2F10.1021%2Fjo00055a015http://pubs.acs.org/doi/abs/10.1021/jo00055a015
[34]
Prior RL, Wu X, Schaich K. Standardized methods for the determination of antioxidant capacity and phenolics in foods and dietary supplements, J Agric Food Chem, 2005; 53(10): 4290-4302. 10.1021/jf0502698b7a4be66fce1641d1067ded91bd50d6ahttp%3A%2F%2Fcogentoa.tandfonline.com%2Fservlet%2Flinkout%3Fsuffix%3DCIT0033%26amp%3Bdbid%3D16%26amp%3Bdoi%3D10.1080%252F23311932.2015.1081728%26amp%3Bkey%3D10.1021%252Fjf0502698https://pubs.acs.org/doi/10.1021/jf0502698
[35]
Bahorun T, Gressier B, Trotin F, Brunet C, Dine T, Luyckx M , et al. Oxygen species scavenging activity of phenolic extracts from hawthorn fresh plant organs and pharmaceutical preparations, Arzneim Forsch, 1996; 46(11): 1086-109. 10.1002/ardp.1996329110789558701955c0c4b90772526c1d1c3ba8ce1b33http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F8955870http://europepmc.org/abstract/MED/8955870
[36]
Olszewska MA, Presler A, Michel P. Profiling of phenolic compounds and antioxidant activity of dry extracts from the selected Sorbus species, Molecules, 2012; 17(3): 3093-3113. 10.3390/molecules1703309322410419ea4d5164070cce8d22556722482f71c4http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F22410419http://www.mdpi.com/1420-3049/17/3/3093
[37]
Di Domenico F, Foppoli C, Coccia R, Perluigi M. Antioxidants in cervical cancer: Chemopreventive and chemotherapeutic effects of polyphenols, Biochim Biophys Acta, 2012; 1822(5): 737-47. 10.1016/j.bbadis.2011.10.00522019724888ca8290eef97df44a0bc90e6723be2http%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS0925443911002286https://linkinghub.elsevier.com/retrieve/pii/S0925443911002286
[38]
Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002, CA-Cancer[J] Clin, 2005; 55(2): 74-108.
[39]
Kueng W, Silber E, Eppenberger U. Quantification of cells cultured on 96-well plates, Anal Biochem, 1989; 182(1): 16-9. 10.1016/0003-2697(89)90710-02604040b9bfa9e0ae7006f0b434bf9088c60a57http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F2604040https://linkinghub.elsevier.com/retrieve/pii/0003269789907100
[40]
Kanduc D, Mittelman A, Serpico R, Sinigaglia E, Sinha AA, Natale C , et al. Cell death: apoptosis versus necrosis, Int J Oncol, 2002; 21(1): 165-70.
[41]
a) Abdel-Latif L, Murray BK, Renberg RL, O'Neill KL, Porter H, Jensen JB , et al. Cell death in bovine parvovirus-infected embryonic bovine tracheal cells is mediated by necrosis rather than apoptosis . J Gen Virol, 2006; 87(Pt 9): 2539-48. b) Legrand C, Bour JM, Jacob C, Capiaumont J, Martial A, Marc A, et al. Lactate dehydrogenase (LDH) activity of the cultured eukaryotic cells as marker of the number of dead cells in the medium. J Biotechnol, 1992; 25(3):231-243. 10.1099/vir.0.81915-01689419239afa7fbfd0b34d67a9312d638e602abhttp%3A%2F%2Fnew.med.wanfangdata.com.cn%2FPaper%2FDetail%3Fid%3DPeriodicalPaper_JJ027590010http://new.med.wanfangdata.com.cn/Paper/Detail?id=PeriodicalPaper_JJ027590010
[42]
Lyons AB, Hasbold J, Hodgkin PD. Flow cytometric analysis of cell division history using dilution of carboxyfluorescein diacetate succinimidyl ester, a stably integrated fluorescent probe, Methods Cell Biol, 2001; 63: 375-398. 10.1016/S0091-679X(01)63021-8https://linkinghub.elsevier.com/retrieve/pii/S0091679X01630218
[43]
Romero-Hernández LL, Merino-Montiel P, Montiel-Smith S, Meza-Reyes S, Vega-Báez JL, Abasolo I , et al. Diosgenin-based thio(seleno)ureas and triazolyl glycoconjugates as hybrid drugs. Antioxidant and antiproliferative profile. Eur J Med Chem, 2015; 99: 67-81. 10.1016/j.ejmech.2015.05.018https://linkinghub.elsevier.com/retrieve/pii/S0223523415300453
[44]
Merino-Montiel P, López Ó, Álvarez E, Fernández-Bolaños JG. Synthesis of conformationally-constrained thio(seleno)hydantoins and α-triazolyl lactones from D-arabinose as potential glycosidase inhibitors. Tetrahedron, 2012; 68(24): 4888-4898. 10.1016/j.tet.2012.03.0879a81ac708a7ab10e356c7b902c391891http%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS0040402012004280https://linkinghub.elsevier.com/retrieve/pii/S0040402012004280
[45]
Camacho‐Camacho C, Rojas‐Oviedo I, Garza‐Ortiz A, Toscano RA, Sánchez‐Sánchez L, Cardenas J , et al. Tributyltin(IV) Schiff base complexes with amino acid derivatives: synthesis, characterization and biological activity. Appl Organomet Chem, 2016; 30(4): 199-207. 10.1002/aoc.3417ac733c90f16cd8c95b71c6686bf0b9c8http%3A%2F%2Fonlinelibrary.wiley.com%2Fdoi%2F10.1002%2Faoc.3417%2Ffullhttp://doi.wiley.com/10.1002/aoc.v30.4
[46]
Fernández-Herrera MA, Mohan S, López-Muñoz H, Hernández-Vázquez JM, Pérez-Cervantes E, Escobar-Sánchez ML , et al. Synthesis of the steroidal glycoside (25R)-3β,16β-diacetoxy-12,22-dioxo-5α-cholestan-26-yl β-D-glucopyranoside and its anti-cancer properties on cervicouterine HeLa, CaSki, and ViBo cells. Eur J Med Chem, 2010; 45(11): 4827-4837 10.1016/j.ejmech.2010.07.051https://linkinghub.elsevier.com/retrieve/pii/S0223523410005477
[47]
Alvarado-Sansininea JJ, Sánchez-Sánchez L, López-Muñoz H, Escobar ML, Flores-Guzmán F, Tavera-Hernández R et al. Quercetagetin and patuletin: antiproliferative, necrotic and apoptotic activity in tumor cell lines. Molecules, 2018; 23(10): E2579. 10.3390/molecules23102579http://www.mdpi.com/1420-3049/23/10/2579
[48]
Fernández-Herrera MA, López-Muñoz H, Hernández-Vázquez JM, Sánchez-Sánchez L, Escobar-Sánchez ML, Pinto BM et al. Synthesis and selective anticancer activity of steroidal glycoconjugates. Eur J Med Chem, 2012; 54: 721-727. 10.1016/j.ejmech.2012.06.02722770605f7c6ef7c00749a2ab9d12ae48b45f735http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F22770605https://linkinghub.elsevier.com/retrieve/pii/S0223523412003790
[49]
Silvia V, Baldisserotto A, Scalambra E, Malisardi G, Durini E, Manfredini S. Novel molecular combination deriving from natural aminoacids and polyphenols: Design, synthesis and free-radical scavenging activities. Eur J Med Chem, 2012; 50: 383-392. 10.1016/j.ejmech.2012.02.0182238567557f4d8305bf0d41d5af00edb682fbadchttp%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS0223523412001080https://linkinghub.elsevier.com/retrieve/pii/S0223523412001080
Share
Back to top