Abstract
Three new ferrocene complexes were synthesized with 4-(1H-pyrrol-1-yl)phenol group appended to one of the Cp ring. These are: 1,1′-4-(1H-pyrrol-1-yl)phenyl ferrocenedicarboxylate, (“Fc-(CO2-Ph-4-Py)2”), 1,4-(1H-pyrrol-1-yl)phenyl, 1′-carboxyl ferrocenecarboxylate (“Fc-(CO2-Ph-4-Py)CO2H”) and 4-(1H-pyrrol-1-yl)phenyl ferroceneacetylate (“Fc-CH2CO2-Ph-4-Py”). The new species were characterized by standard analytical methods. Cyclic voltammetry experiments showed that Fc-CH2CO2-Ph-4-Py has redox potential very similar to the Fc/Fc+ redox couple whereas Fc-(CO2-Ph-4-Py)2 and Fc-(CO2-Ph-4-Py)CO2H have redox potentials of over 400 mV higher than Fc/Fc+ redox couple. The in vitro studies on Fc-(CO2-Ph-4-Py)2 and Fc-(CO2-Ph-4-Py)CO2H revealed that these two compounds have moderate anti-proliferative activity on MCF-7 breast cancer cell line. In contrast Fc-CH2CO2-Ph-4-Py which displayed low anti-proliferative activity. In the HT-29 colon cancer cell line, the new species showed low anti-proliferaive activity. Cytokinesis-block micronucleus assay (CBMN) was performed on these ferrocenes and it was determined they induce micronucleus formation on binucleated cells and moderate genotoxic effects on the MCF-7 breast cancer cell line. There is a correlation between the IC50 values of the ferrocenes and the amount of micronucleus formation activity on binucleated cells and the reactive oxygen species (ROS) production on MCF-7 cell line.
Keywords: ferrocene, anti-proliferative, cytokinesis-block micronucleus assay, MCF-7, HT-29, MCF-10A
1. Introduction
Drug resistance and toxic side effects are the major challenges encountered in chemotherapy treatments. Researchers have driven their interests into the development of metallic drugs, other than platinum compounds, to overcome some of the abovementioned problems. Among them, ferricenium and ferrocene derivatives opened a new ground in the discovery of new chemotherapeutic agents [1]. The versatility and ease of ferrocene functionalization has allowed the design and synthesis of a wide variety of ferrocene derivatives with biological activity [2–14].
Numerous pendant (functional) groups have been attached or linked to the Cp ring to tailor the anti-proliferative properties of ferrocene, many of them with great success [2–14]. Recently a new range of possible chemotherapeutic compounds have been studied using pyrrole derivatives. Those pyrrole derivatives have been demonstrated to have good anti-proliferative activity and an increase in membrane permeability allowing the compounds reaching the nucleus [15–22].
Our research group has been working for many years in the structure modification of the Cp ring with different pendant groups aimed to enhance the cytotoxic activity of the corresponding functionalized ferrocene [13,14]. One of our major accomplishment is the synthesis of ferrocenoyl 17β-hydroxy-estra-1,3,5(10)-trien-3-olate complex (“Fc-CO2-estradiol”) which exhibited enhanced anti-proliferative activity on hormone-dependent breast epithelial adenocarcinoma cell line MCF-7 [13]. Docking studies of this species into the alpha estrogen receptor (ERα) revealed that the ferrocene docks partially in the estrogen binding pocket impairing the protein function [13]. More recently, we published a series of ferrocenoyl esters which have anti-proliferative activities depending on the pendant group attached to the Cp ring [14]. Of particular interest is 4-bromophenyl ferrocenecarboxylate (Fc-CO2-Ph-4-Br) which also displayed good anti-proliferative activity on breast cancer cell line MCF-7 and low anti-proliferative activity on non-tumorigenic human breast epithelial cell line MCF-10A [14]. Also a ferrocenecarboxylate modified with a pyrrole group, 4-(1H-pyrrol-1-yl)phenyl ferrocenecarboxylate, (Fc-CO2-Ph-4-Py), has been reported showing good antiproliferative activity on MCF-7 [14]. In contrast to Fc-CO2-estradiol, Fc-CO2-Ph-4-Br anti-proliferative activity does not correspond to the anti-estrogenic properties and it is correlated to the cytotoxic effects of the ferrocene moiety.
In order to gain more insights regarding the structure-activity relationship of these metallocenes, we have prepared new ferrocene derivatives modified with pyrrole group and their anti-proliferative activities and mechanism of action have been studied and compared to Fc-CO2-estradiol complex. Herein we report these new findings.
2. Experimental section
2.1 General procedure
All reactions were performed under an atmosphere of dry nitrogen using schlenk glassware or a glovebox, unless otherwise stated. Reaction vessels were flame dried under a stream of nitrogen, and anhydrous solvents were transferred by oven-dried syringes or cannula. CH2Cl2 was dried and deoxygenated by distillation over CaH2 under nitrogen. Infrared spectra were recorded on a Perkin Elmer Spectrum ATR spectrometer in solid. The NMR spectra were obtained on a 500MHz Bruker spectrometer. Electrochemical characterizations were carried out on a BAS CV-50W voltammetric analyzer. A Bruker Daltonics Esquire 6000 instrument was used to record the mass spectral data. The electrospray positive ion was used as ionization mode during the MS experiment. Elemental analyses were obtained from Atlantic Microlab Inc. Silica gel was heated at about 200°C while a slow stream of dry nitrogen was passed through it prior to use. 4-(1H-Pyrrol-1-yl)phenol, ferroceneacetic acid and ferrocenedicarboxylic acid were purchased from Aldrich and used as received.
Hydrolytic stability studies of Fc-(CO2-Ph-4-Py)CO2H, and Fc-CH2CO2-Ph-4-Py were performed by H1NMR using a solvent mixture of 5% DMSOd6 - 95% D2O (v/v). The complex solutions were prepared first by dissolving the corresponding ferrocene in DMSO-d6 and then D2O was added to a final composition of 5% DMSOd6–95% D2O. The H1NMR spectrum of the complex solution was then recorded at 0 h, 24 h and 72 h. Attempts to study the hydrolytic stability of Fc-(CO2-Ph-4-Py)2 was hampered by the limited solubility of this compound on aqueous solution.
2.2 Synthesis and characterization
2.2.1 Synthesis of 1,1′-4-(1H-pyrrol-1-yl)phenyl ferrocenedicarboxylate (“Fc-(CO2-Ph-4-Py)2”)
In a 50 mL three neck round bottom flask, 1,1′-ferrocenedicarboxylic acid (0.2741g, 0.001 mol), oxalyl chloride (174.5 μL, 0.002 mol) and pyridine (161 μL, 0.002 mol) were reacted in 20 mL of dichloromethane (DCM) under nitrogen atmosphere at room temperature. The reaction was carried out for 12 h to obtain 1,1′-ferrocenoyl dichloride. For esterification, 1,1′-ferrocenyl chloride was reacted with 4-(1H-pyrrol-1-yl)phenol (0.1592g, 0.001 mol) and pyridine (161 μL, 0.002 mol) for 12 h. After that period, three consecutive washing with 0.01 M HCl were carried out. The organic phase was then dried over sodium sulfate. From the reaction a mixture of three components were obtained; 4-(1H-pyrrol-1-yl)phenyl ferrocenecarboxylate, (“Fc-CO2-Ph-4-Py”)[14], 1,1′-4-(1H-pyrrol-1-yl)phenyl ferrocenedicarboxylate, (“Fc-(CO2-Ph-4-Py)2”), and 1-4-(1H-pyrrol-1-yl)phenyl, 1′-carboxyl ferrocenecarboxylate (“Fc-(CO2-Ph-4-Py)CO2H”). The products were separated by column chromatography using silica gel and DCM as mobile phase. The three products, Fc-CO2-Ph-4-Py, Fc-(CO2-Ph-4-Py)2, and Fc-(CO2-Ph-4-Py)CO2H were isolated in 3–9% yield, 12–15 % yield, and 45–47 % yield respectively. Characterization of 4-(1H-pyrrol-1-yl)phenyl ferrocenecarboxylate, (“Fc-CO2-Ph-4-Py”) was previously reported [14].
1,1′Ferrocenyl dichloride
1HNMR (500 MHz, CDCl3) (δppm): 4.99 (4H, s), 4.72 (4H, s).
1,1′-4-(1H-pyrrol-1-yl)phenyl ferrocenedicarboxylate (“Fc-(CO2-Ph-4-Py)2”)
1HNMR (500 MHz, CDCl3) (δppm): 7.37 (2H, d, ph; 3J = 8.8 Hz), 7.25 (2H, d, py; 3J = 2.8 Hz), 7.03 (2H, dd, ph; 3J = 1.3 Hz), 6.34 (2H, dd, py; 3J = 1.6 Hz), 5.08 (2H, overlapping doublets, AA′, Cp), 4.64(2H, overlapping doublets, BB′, Cp).
13CNMR (125 MHz, CDCl3) (δppm): 169.0 (C=O), 148.3, 138.6, 122.9, 121.5, 119.5, 110.5, 73.4, 72.4, 72.0.
ATR-IR(cm−1): 3145, 3092, 2925, 2841, 1727 (C=O), 1523, 1265, 1205, 1106, 848, 796, 727.
Anal. Calc. for C32H24O4FeN2: C, 69.05; H, 4.40 Found: C, 68.62; H, 4.46.
1-4-(1H-pyrrol-1-yl)phenyl, 1′-carboxyl ferrocenecarboxylate (“Fc-(CO2-Ph-4-Py)CO2H”)
1HNMR (500 MHz, CDCl3): 7.45 (2H, d, ph; 3J = 8.5 Hz), 7.31 (2H, d, py; 3J = 8.5 Hz), 7.08 (2H, dd, ph; 3J = 1.6 Hz), 6.37 (2H, dd, py; 3J = 1.6 Hz), 5.30 (CH2Cl2, s), 5.01 (2H, overlapping doublets, AA′, Cp), 4.95 (2H, overlapping doublets, BB′, Cp), 4.57 (2H, overlapping doublets, AA′, Cp′), 4.56 (2H, overlapping doublets, BB′ Cp′)
13CNMR (125 MHz, CDCl3) (δppm): 168.9 (C=O)′, 166.4 (C=O), 148.3, 138.5, 122.9, 121.5, 119.5, 110.5, 74.3, 73.8, 73.4, 72.5, 72.1, 71.0, 53.5 (CH2Cl2).
ATR-IR (cm−1): 3410(COOH), 3137, 3032, 2963, 1772(C=O), 1697 (C=O)′, 1515, 1250, 1068, 1015, 825, 712.
ESI-MS (positive mode), m/z (relative intensity, ≥ 4): [Fc-(CO2-Ph-4-Py)CO2H H]+ 413(5), 415(100), 416(23), 417(4). Anal. Calc. for C22H17O4NFe(CH2Cl2)0.125 : C, 62.40; H, 4.08 Found: C, 62.59; H, 4.28.
2.2.2 Synthesis of 4-(1H-pyrrol-1-yl)phenyl ferroceneacetylate (“Fc-CH2CO2-Ph-4-Py”)
In a 50 mL three neck round bottom flask, ferroceneacetic acid (0.0600g, 0.25 mmol), oxalyl chloride (21.8 μL, 0.25 mmol), pyridine (20.1 μL, 0.25 mmol) and 4-(1H-pyrrol-1yl)phenol (0.0398g, 0.25 mmol) were reacted in 10 mL of dichloromethane (DCM) under nitrogen atmosphere at room temperature. The reaction was carried out for 12 h to get 4(1H-pyrrol-1-yl)phenyl ferroceneacetylate. After that period three consecutive washing with 0.01 M HCl were carried out. The organic phase was then dried over sodium sulfate. The product was separated by column chromatography using silica gel and DCM as mobile phase. Fc-CH2CO2-Ph-4-Py was isolated in 62 % yield.
4-(1H-pyrrol-1-yl)phenyl ferroceneacetylate (“Fc-CH2CO2-Ph-4-Py”)
1HNMR (500 MHz, CDCl3): 7.44 (2H, d, ph; 3J = 8.8 Hz ), 7.18 (2H, d, py; 3J = 8.8 Hz), 7.01(2H, dd, ph; 3J = 1.9 Hz), 6.36 (2H, dd, py; 3J = 1.9 Hz), 5.30 (CH2Cl2, s), 4.32 (2H, bs, Cp), 4.19 (2H, bs, Cp′, 5H), 3.62 (2H, s, CH2, 2H).
13CNMR (125 MHz, CDCl3) (δppm): 169.5 (C=O), 148.32, 138.6, 122.6, 121.6, 119.54, 110.5, 79.7, 68.8, 68.1, 53.5 (CH2Cl2), 35.6.
ATR-IR (cm−1): 3092, 2963, 2917, 1750(C=O), 1508, 1334, 1143, 1098, 810, 727.
ESI-MS (positive mode), m/z (relative intensity, ≥ 4): [Fc-CH2CO2-Ph-4-Py]+ 384(5), 386(100), 387(22). Anal. Calc. for C22H19O2NFe(CH2Cl2)0.06: C, 67.88; H, 4.94 Found: C, 67.99; H, 5.04.
2.3 Anti-proliferative studies
Biological activity was determined using the MTT assay originally described by Mossman [23,24] but using 10% Triton in isopropanol as a solvent for the MTT formazan crystals [23,24]. The breast adenocarcinoma cell line MCF-7 and MCF-10A were purchased from American Type Culture Collection (ATCC HTB22 and CRL-10317) and were grown under sterile conditions in a culture chamber at 37 °C and 95%Air/5%CO2. Breast adenocarcinoma cell line MCF-7 was grown in Eagles’s Minimum Essential medium supplemented with 10%(v/v) fetal bovine serum, 1%(v/v) penicillin/streptomycin , and 0.01mg/mL bovine insulin. Non-tumorigenic human breast epithelial cell line MCF-10A, was grown in Dulbecco’s modified Eagle’s medium (DMEM), 5% horse serum, 0.5 μg/mL hydrocortisone (Sigma Cat. no. H-0888), 10 μg/mL insulin (Sigma-ca. no. I1882), 20 ng/mL epidermal growth factor (Sigma Cat. no. E-9644). HT-29 cells were maintained at 37 °C and 95% Air–5% CO2 in McCoy’s 5A (ATCC) complete medium, which had been supplemented with 10% (v/v) fetal bovine serum (ATCC) and 1% (v/v) penicillin/streptomycin (Sigma). Asynchronously growing cells were seeded at 1.0 × 105 cells per well in 96-well plates containing 100 μL of complete growth medium, and allowed to recover overnight. Various concentrations of the complexes (10–1300 mM) dissolved in 5% DMSO–95% medium were added to the wells (eight wells per concentration; experiments performed in quadruplicate plates). The complex solutions were prepared first by dissolving the corresponding ferrocene in DMSO and then medium was added to a final composition of 5% DMSO–95% medium. In addition to the cells treated with the ferrocenes, two controls experiments were run: one without any addition of solvent mixture (5% DMSO–95% medium) and one adding 5% DMSO–95% medium to the cells. Both control experiments behaved identically, showing that 5% of DMSO in the medium did not render toxic to these types of cells. The cells were incubated for an additional 70 h. After this time, MTT dissolved in complete growth medium was added to each well to a final concentration of 1.0 mg mL−1 and incubated for two additional hours. After this period of time, all MTT containing medium was removed, cells were washed with cold PBS and dissolved with 200 mL of a 10% (v/v) Triton X-100 solution in isopropanol. After complete dissolution of the formazan crystals, absorbances were recorded in triplicate on a 340ATTC microplate reader (SLTLab Instruments) at 570 nm with background subtraction at 630 nm. Concentrations of compounds required to inhibit cell proliferation by 50% (IC50) were calculated by fitting data to a four-parameter logistic plot by means of Sigma Plot software from SPSS (SPSS; Chicago, IL).
2.4 Electrochemistry
Cyclic voltammetric experiments were performed in deoxygenated CH3CN solutions of ferrocene complexes with 0.1 M [NBun4]PF6 as supporting electrolyte and ferrocene complex concentration of 2 × 10−3 M. The three electrodes used were platinum disk as the working electrode, Ag/AgCl as a reference electrode, and Pt wire as an auxiliary electrode. The working electrode was polished with 0.05 μm alumina slurry for 1–2 minutes, and then rinsed with double-distilled and deionized water. This cleaning process is done before each CV experiment and a sweep between 0 and 2000 mV is performed on the electrolyte solution to detect any possible deposition of ferrocene on the electrode surface.
2.5 Cytokinesis-block micronucleus assay (CBMN)
Cytokinesis-block micronucleus assay (CBMN) was performed as a modified procedure from Fenech M. [25]. Breast adenocarcinoma cells (MCF-7) were seeded in a 75 cm2 flask with 20 mL of growth media and incubated for about 72 h. Following disaggregation with Trypsin/EDTA and re-suspension in complete medium, with concentration of 1 × 106 cells/mL, the cells were seed on a six well plate. After 24 h the medium was replaced with a solution at different concentrations of ferrocene complexes. Various concentrations (10 to 0.01 μM) of the corresponding ferrocene were set below IC50. Ferrocene solutions were prepared first by dissolving the corresponding ferrocene in DMSO and then medium was added to a final composition of 5% DMSO–95% medium. The different solutions of the desired ferrocene were added to individual wells, in addition to solutions of a negative control (cells without treatment) and a positive control (cell with methyl methane sulphonate, MMS). Methyl methane sulphonate concentration was selected according to Decordier et al. [26]. After 72 h of drug exposure, the solution of ferrocene complex was then replaced with fresh media containing only 6μg/mL of cytochalasin-β and incubated for another 24 h. The cells were washed with phosphate-buffer saline (PBS) and disaggregated with trypsin/EDTA. Following by centrifugation, the cells were re-suspended in 50μL of medium and then fixed with methanol and stained with hemacolor stain set (Harleco, cat# 65044-93). For each treatment condition, micronuclei in 500 binucleated MCF-7 cells for three experiments were scored (mean ± SD). Mitotic rate was assessed as per cent of binucleated cells (mean ± SD). Probability values (P) were estimated by Student’s t-test comparing treated cells versus control.
2.6 Measurements of reactive oxygen species (ROS)
The ROS production was determined on MCF-7 treated with the oxidation-sensitive dye, 2′7′-dichlorodihydrofluoresceindiacetate (H2DCF-DA, Sigma-Aldrich). The exposure of cells to H2DCF-DA generates a fluorescent product 2′7′-dichlorofluorescein (DCF) which has a excitation/emission maxima of ~495/529 nm. The breast cancer cells (MCF-7) were seeded in 96-well plates at a concentration of 5 × 104 cells/mL containing 100 μL of complete growth medium, and allowed to recover overnight. Various concentrations of the complexes (10–1300 mM) dissolved in 5% DMSO–95% medium were added to the wells (eight wells per concentration; experiments performed in quadruplicate plates). The complex solutions were prepared first by dissolving the corresponding ferrocene in DMSO and then medium was added to a final composition of 5% DMSO–95% medium. In addition to the cells treated with the ferrocenes, two controls experiments were run: one without any addition of solvent mixture (5% DMSO–95% medium) and one adding 5% DMSO–95% medium to the cells. Both control experiments behaved identically, showing that 5% of DMSO in the medium did not render toxic to these types of cells. The cells were incubated for an additional 70 h. After this time, the cells were treated with 10 μM of H2DCF-DA and incubated for 40 minutes at 37 °C after which time ROS production was evaluated at 605 nm.
3. Results and discussion
3.1 Synthesis and characterization
The subject ferrocenoyl esters were synthesized by an established procedure reacting the 1.1′-ferrocenoyl dichloride (Figure 1) and ferrocene acetyl chloride (Figure 2) with 4-(1H-pyrrol-1-yl)phenol. However, the first reaction (Figure 1) rendered not very efficient since in the synthesis of Fc-(CO2-Ph-4-Py)2, the desired complex was isolated in low yield (12–15% ) and it yielded two by-products: Fc-CO2-Ph-4-Py and Fc-(CO2-Ph-4-Py)CO2H. Interestingly, Fc-CO2-Ph-4-Py was synthesized previously from ferrocenecarboxylic acid but in this synthesis it is the by-product of Fc-(CO2-Ph-4-Py)CO2H decarboxylation.
Figure 1.
Synthesis of ferrocenoyl esters containing 4-(1H-pyrrol-1-yl)phenolate as pendant group. First and second complexes are the principal products of the reaction. The last product results from decarbonylation of the cyclopentadiene.
Figure 2.

Synthesis of 4(1H-pyrrol-1-yl)phenyl ferroceneacetylate.
The structural characterization was very straightforward. The IR spectra showed bands in the 1727 – 1772 cm−1 region indicating the presence of the carbonyl stretching of the ester groups on the ferrocene. Fc-(CO2-Ph-4-Py)CO2H showed a second carbonyl stretching at 1697 cm−1 and a broad band at 3410 cm−1 indicating the presence of the carboxyl group attached on the other Cp ring. The 13C NMR spectra exhibit signals about 169 ppm corresponding to the carbonyl groups. The Fc-(CO2-Ph-4-Py)CO2H showed a second carbonyl signal as expected at 166 ppm attributed to the carboxyl group. The 1H NMR spectral data for Fc-CH2CO2-Ph-4-Py showed a singlet corresponding to the unsubstituted Cp ring and two overlapping signals with AA′BB″ pattern corresponding to the monosubstituted Cp ring.
Likewise for Fc-(CO2-Ph-4-Py)2 and Fc-(CO2-Ph-4-Py)CO2H, the Cp signals appear as overlapping doublets in a AA′BB′ pattern. The phenyl and pyrrole groups appear in the aromatic and conjugated vinylic regions respectively.
The hydrolytic stability of Fc-CH2CO2-Ph-4-Py and Fc-(CO2-Ph-4-Py)CO2H was investigated by 1H NMR in 5% DMSOd6–95% D2O solutions. Fc-(CO2-Ph-4-Py)2 in this medium forms an emulsion and could not be investigated using this technique. At 24 h, Fc-CH2CO2-Ph-4-Py and Fc-(CO2-Ph-4-Py)CO2H showed less than 5 % of decomposition. At 72 h, Fc-CH2CO2-Ph-4-Py was decomposed by 40% and Fc-(CO2-Ph-4-Py)CO2H by 30%. This is evidence that these ferrocenes are robust under pseudo-physiological conditions.
3.2 Electrochemical characterization
Electrochemical characterization of these new species was pursued by cyclic voltammetry since one of the proposed mechanism of cytotoxic activity involves the facile formation of ferrocenium ion in the cell [27,28]. As proposed by Osella et al., this redox property is responsible for the formation of reactive oxygen species (ROS) inducing oxidative damage to DNA [27,28]. Table 1 presents the redox properties of the subject complexes. The complexes have limited solubility in aqueous solution and soluble in 5% DMSO/90 % H2O but when the [NBun4]PF6 is present the solution becomes cloudy. We selected CH3CN as solvent since it provided the best CV data.
Table 1.
Redox potentials of functionalized ferrocenes in CH3CN with 0.1 M [NBu4n]PF6 at a scan rate of 100 mVs−1, using Ag/AgCl saturated as reference electrode.
| Complexes | Epa(mV) | Epc (mV) | ΔE (mV) | E1/2 (mV) |
|---|---|---|---|---|
| Fc-(CO2-Ph-4-Py)2 | 958 | 862 | 96 | 910 |
| Fc-(CO2-Ph-4-Py)CO2H | 1027 | 901 | 126 | 964 |
| Fc-CH2CO2-Ph-4-Py | 503 | 431 | 73 | 467 |
| * Fc-CO2-estradiol | 801 | 711 | 90 | 756 |
| ** Fc-CO2-Ph-4-Br | 580 | 484 | 96 | 532 |
|
| ||||
| Fc/Fc+ | 494 | 414 | 80 | 454 |
13. Dalton Trans. 40 (2011) 9557–9565.
14. J. Organometal. Chem 749 (2014) 204–214.
In CH3CN solution, Fc-(CO2-Ph-4-Py)2 (Epa = 958 mV) and Fc-(CO2-Ph-4-Py)CO2H (Epa = 1027 mV) showed oxidation potentials higher than Fc (494 mV), while Fc-CH2CO2-Ph-4-Py showed similar oxidation potential (Epa = 503 mV) to ferrocene. This data suggests that the 4(1H-pyrrol-1-yl)phenylcarboxylate substituent is providing a place for electron delocalization away from the metal center and making them for robust to oxidation. For Fc-CH2CO2-Ph-4-Py, the CH2 group between the ferrocene and the functional group avoid electron delocalization between the ferrocene unit and the 4(1H-pyrrol-1-yl)phenylcarboxylate. In light of the proposed mechanism of action and the possible correlation with the oxidation potentials, as a point of reference, it could be worth to compare these with previously reported potentials of biologically active ferrocene, Fc-CO2-estradiol and 4-bromophenyl ferrocenecarboxylate (Fc-CO2-Ph-4-Br), in MCF-7 and HT-29 cancer cell lines [13,14].
From the electrochemical point of view, Fc-CO2-Ph-4-Br lacks of the pyrrole group and as a consequence, electron delocalization is reduced as well as the oxidation potential. Conversely, Fc-CO2-estradiol has a steroid group which apparently is able to delocalize electrons and as a result higher oxidation potential. In spite of these differences, Fc-CO2-estradiol and Fc-CO2-Ph-4-Br have identical anti-proliferative activities in MCF-7 and HT-29 cancer cell lines thereby the correlation oxidation potential and cytotoxicity cannot be made. Attempts to study the redox behavior of these new species under physiological conditions were hampered by the limited solubility of these species in buffer or aqueous solutions. Nonetheless the current data provide an idea about the redox behavior of these species and can be used qualitatively.
3.3 Anti-proliferative activity studies
In vitro anti-proliferative activities of the new ferrocene complexes and their starting materials were determined by MTT-based assays involving a 72-h drug exposure on two malignant human cell lines, MCF-7 breast cancer and HT-29 colon cancer, and a normal healthy breast cell line, MCF-10A [23,24]. Their IC50 values listed on Table 2 are compared with our two most active compounds at the present time in MCF-7 cell line, Fc-CO2-estradiol and Fc-CO2-Ph-4-Br [13,14]. In MCF-7 cell line, all derivatives showed dose dependent relationship with more anti-proliferative activity than their precursors (starting materials). Fc-(CO2-Ph-4-Py)2 (45.5(6) μM) and Fc-(CO2-Ph-4-Py)CO2H (57(7) μM) have moderate anti-proliferative activity on MCF-7 and low anti-proliferative activity non-tumorigenic human breast epithelial cell line MCF-10A. But their anti-proliferative activities on MCF-7 do not exceed those of Fc-CO2-estradiol and Fc-CO2-Ph-4-Br [13,14]. Fc-CH2CO2-Ph-4-Py has low antiproliferative activity on all cell lines, MCF-7, MCF-10A and HT-29. We should point out that none of the complexes have higher anti-proliferative activity on MCF-7 and HT-29 than Fc-CO2-estradiol, and Fc-CO2-Ph-4-Br.
Table 2.
Anti-proliferative activities of ferrocene derivatives studied on MCF-7 breast cancer, MCF-10A normal breast, and HT-29 colon cancer cell lines, as determined using MTT assay after 72 h of drug exposure. IC50 values based on quadruplicate experiments and standard deviation in parenthesis. N/A is non-active.
| Complex | IC50, MCF-7 | IC50, MCF-10A | IC50, HT-29 |
|---|---|---|---|
| Fc-(CO2-Ph-4-Py)2 | 45.5(6) | 298(5) | 189(16) |
| Fc-(CO2-Ph-4-Py)CO2H | 57(7) | 219(21) | 121(8) |
| Fc-CH2CO2-Ph-4-Py | 103(6) | 288(57) | 283(11) |
| Fc-CO2-estradiol | *9(2) | 119(8) | *24.4(6) |
| Fc-CO2-Ph-4-Br14 | **9.2(8) | **353(8) | 24(6) |
| 1-1′ferrocenedicarboxilic acid | N/A | N/A | 410(21) |
| Ferroceneacetic acid | 590(25) | 86(7) | 253(10) |
| 4-(1H-pyrrol-1yl)phenol | 249(5) | 553(76) | 211(44) |
| Ferricenium tetrafluoroborate13 | 150 | 180 |
3.4 Cytokinesis-block micronucleus assay
The genotoxic effects of Fc-CO2-estradiol, Fc-(CO2-Ph-4-Py)CO2H, Fc-CH2CO2-Ph-4-Py, and Fc-(CO2-Ph-4-Py)2 were studied on MCF-7 cells using cytokinesis-block micronucleus (CBMN) assay after 72 h of treatment with these compounds. Micronuclei were scored in 500 binucleated MCF-7 cells as micronucleated binucleated cells. Figure 3 presents the photomicrographs of the MCF-7 cells in the CBMN assay without the presence of the drugs to demonstrate the appearance of mononucleated cell, binucleated cell and binucleated cell containing one micronucleus.
Figure 3.

Photomicrographs of the MCF-7 cells scored in the CBMN assay taken with an electronic microscope Olympus BX60. A) Mononucleated cell; B) binucleated cell; C) binucleated cell containing one micronucleus. All cells were treated with 6μg/mL Cytochalasin-β for 24 hours.
Three concentrations of each ferrocene complex were selected below IC50 values of the corresponding ferrocene. For Fc-CO2-estradiol with an IC50 of 9(2) μM the concentrations selected were 1μM, 0.1μM, and 0.01μM. For Fc-(CO2-Ph-4-Py)2, Fc-(CO2-Ph-4-Py)CO2H, and Fc-acPy with IC50 values between 45 to 103 μM, the concentrations were 10 μM, 1μM, and 0.1μM. The induction of micronucleus formation was characterized by an increase in single micronucleus in binucleated cells relative to a negative control that consists of untreated cells. Methyl methane sulphonate (MMS) was selected as positive control due to its well-known genotoxic activity [27]. Mononucleated, binucleated and micronucleated binucleated scored cells after 24 h of cytochalasin-β addition are represented Figure 3. Micronucleus formation in binucleated cells is associated to the genotoxic effect of the complex under study.
Figure 4 shows the micronuclei-forming activity of the ferrocene complexes as total micronucleated binucleated MCF-7 cells in 500 scored cells. Fc-CO2-estradiol at concentrations of 1μM (mean 29, P < 0.0001) and 0.1μM (mean 25, P < 0.01) and Fc-(CO2-Ph-4-Py)CO2H at a concentration of 10 μM (mean 25.5, P < 0.01) showed a 4-fold increase in micronucleus forming activity compared to the control experiment (untreated cells). At lower concentrations of both ferrocene complexes showed a decrease in micronucleus forming activity. At a concentration of 0.01μM (mean 24, P < 0.0004) Fc-CO2-estradiol has 3-fold increase in micronucleus forming activity while Fc-(CO2-Ph-4-Py)CO2H shows similar micronuclei-forming activity for the two lower concentrations (1μM (mean 20, P < 0.01), and 0.1μM (mean 18.5, P < 0.03)). Fc-(CO2-Ph-4-Py)2 exhibits higher micronucleus forming activity than Fc-CO2-estradiol and Fc-carbPy at a concentration of 10 μM (up to 5-fold, mean 33.5, P < 0.05) meanwhile, at 0.1μM Fc-(CO2-Ph-4-Py)2 has no significant (mean 10, P < 0.1) increase in micronucleus forming activity compared to our control experiment. Fc-CH2CO2-Ph-4-Py has no significant (P < 1) micronucleus forming activity at any concentrations. Induction of single micronucleus formation in binucelated cells was found for Fc-CO2-estradiol, Fc-(CO2-Ph-4-Py)CO2H, and Fc-(CO2-Ph-4-Py)2. Binucleated cells with more than one micronucleus were not significantly formed. Important differences in micronucleus forming activity depending on concentrations for Fc-CO2-Estradiol, Fc-(CO2-Ph-4-Py)CO2H, and Fc-(CO2-Ph-4-Py)2 can be observed. For instance, a considerable increase in the number of total micronucleated binucleated cells at a concentration close to IC50 was observed for Fc-(CO2-Ph-4-Py)2. Likewise, Fc-CO2-estradiol and Fc-(CO2-Ph-4-Py)CO2H showed similar genotoxic effect on MCF-7 cells at concentrations close to their IC50. Fc-CH2CO2-Ph-4-Py with the lowest cytotoxicity appears to have no significant genotoxic effect on MCF-7 cells. For Fc-CO2-estradiol, Fc-(CO2-Ph-4-Py)CO2H, and Fc-(CO2-Ph-4-Py)2, the micronucleus forming activity decrease as concentration decreases suggesting a dose dependent relation. The micronucleus forming activities in Fc-CO2-estradiol, Fc-(CO2-Ph-4-Py)CO2H, Fc-CH2CO2-Ph-4-Py, and Fc-(CO2-Ph-4-Py)2 are lower than the activity for positive control (MMS) at all concentrations.
Figure 4.
Micronucleus forming activity of Fc-CO2-estradiol, Fc-(CO2-Ph-4-Py)CO2H , Fc-CH2CO2-Ph-4-Py and Fc-(CO2-Ph-4-Py)2 on MCF7 cells (mean ± SD). Cells were seeded at a concentration of 1 × 106 cells per well on a six well plate, followed by 72 hours of drug exposure at different concentrations of the corresponding ferrocene. After treatment the cells were blocked at cytokinesis by addition of fresh medium containing 6μg/mL cytochalasin-β. Cells were culture for a further 24 h prior to fixation and staining with methanol and Hemacolor Staining Set. Micronucleus formation was scored on 500 binucleated cells and compared to untreated cells as negative control and MMS as positive control.
Mitotic rate effects on MCF-7 cells after 72 h treatment are presented on Table 3 as percent of binucleated cells following cytokinesis block. Statistical correlation of the percent of binucleated cells and untreated cells was estimated by Student’s t-test. A decrease in the mitotic rate is observed upon a treatment of MCF-7 cells with all four ferrocene complexes and the positive control (MMS), in comparison with untreated cells. However, those results reflect no significant differences (P < 1).
Table 3.
Micronucleus forming activity values of control (untreated cells), MMS, Fc-CO2-estradiol, Fc-(CO2-Ph-4-Py)CO2H, Fc-CH2CO2-Ph-4-Py, and Fc-(CO2-Ph-4-Py)2 on MCF-7 cells (mean ± SD) after 72 h treatment. Mitotic rate values represented as per cent binucleated MCF-7 cells (mean ± SD).
| Ferrocene Complex | Concentrations (μM) | Micronucleus (MNi) (mean ± SD) | % Binucleated cells (Mitotic rate) |
|---|---|---|---|
| Control (untreated cells) | 7 (4) | 40 (15) | |
| (MMS) | 94.4 | 62 (6)*** | 28 (3)* |
| Fc-CO2-estradiol | 1.0 | 29 (4)**** | 34 (9)* |
| 0.1 | 25 (10)*** | 39 (9)* | |
| 0.01 | 23 (4)**** | 33 (2)* | |
| Fc-(CO2-Ph-4-Py)2 | 10.0 | 34 (6)*** | 25 (6)* |
| 1.0 | 15 (1)** | 33 (13)* | |
| 0.1 | 10 (1)** | 18 (7)* | |
| Fc-(CO2-Ph-4-Py)CO2H | 10.0 | 26 (4)*** | 12 (2)* |
| 1.0 | 20 (1)*** | 16 (6)* | |
| 0.1 | 19 (5)*** | 13 (3)* | |
| Fc-CH2CO2-Ph-4-Py | 10.0 | 11 (7)* | 38 (12)* |
| 1.0 | 8 (2)* | 41 (15)* | |
| 0.1 | 7 (4)* | 33 (21)* |
P < 1,
P < 0.1,
P < 0.05,
P < 0.0005 (treatment versus control) as determined by Student’s t-test.
The micronucleus assay (CBMN) is a well-established technique used to measure the DNA damage on human or mammalian cells measuring the quantity of binucleated cells with micronuclei (MNi) [25,29]. Micronuclei (MNi) are whole chromosomes or chromosome fragments that result from DNA damage during anaphase nuclear division [29–34]. In the CBMN assay, once-divided, cells are the one that can express MNi and are recognized by their binucleated appearance [29–32].
Different mechanism of DNA damage may induce micronuclei-forming activity. Estrogens genotoxic mechanism may involve DNA adduct formation by estrogen metabolites, free radical by redox cycling of estrogens through microsomal, mitochondrial or nuclear processes [35–38].
For ferrocene derivatives, DNA damage may be caused by oxygen free radical species, inducing oxidative damage [12–14,27,28,39]. Those oxidative species cause DNA fragmentation resulting in micronucleated cells. In the present study four ferrocene derivatives were analyzed by CBMN assay to identify the amount of DNA damage and there is clear evidence that these species induce genotoxic effects.
The mechanistic route for Fc-CO2-estradiol was recently elucidated. Fc-CO2-estradiol binds to the alpha estrogen receptor, ERα, causing a conformational change acting as an antagonist [13]. Additionally, studies with confocal microscopy revealed that Fc-CO2-estradiol migrate through the cell and reach the nucleus after 2 h of drug exposure [12]. Once Fc-CO2-estradiol reaches the nucleus it generates oxygen free radical species resulting in DNA fragmentation.
The results for the micronucleus assay show an increase in micronucleus-forming activity up to 4-fold at a concentration near IC50. These results are evidence of a moderate genotoxic effect of the Fc-CO2-estradiol on MCF-7 cells. Meanwhile, studies reported by Yared et al. using β-estradiol on MCF-7 cells shows a 3-fold increase in the micronucleus-forming activity [35]. The differences in activity of Fc-CO2-estradiol and β-estradiol can be attributed to the ferrocenoyl group.
The three ferrocene derivatives modified with 4-(1H-pyrrol-1-yl)phenol as pendant groups showed important differences in the anti-proliferative activity and genotoxicity. Fc-(CO2-Ph-4-Py)CO2H with a pyrrole group and a carboxylic group have an with IC50 of 57 μM while Fc-(CO2-Ph-4-Py)2 modified with 4-(1H-pyrrol-1-yl)phenol on each Cp ring have an with IC50 of 45.5 μM. Both Fc-(CO2-Ph-4-Py)CO2H and Fc-(CO2-Ph-4-Py)2 are the one with higher oxidation potentials than ferrocene. Fc-(CO2-Ph-4-Py)CO2H present up to 4-fold increase in micronucleus-forming activity close to IC50 while Fc-(CO2-Ph-4-Py)2 shows up to 5-fold increase. The difference in micronucleus activity of these two ferrocene complexes may be attributed to the number of pyrrole groups attached to the Cp rings. Fc-(CO2-Ph-4-Py)2 contains two pyrrole groups while Fc-(CO2-Ph-4-Py)CO2H contains one. This pendant group, 4-(1H-pyrrol-1-yl)phenol, exhibit a oxidation potential close to 1.6 V providing an additional redox center which could induce redox processes (oxidative stress) in the cell and enhancing the cytotoxic activity. In contrast, Fc-CH2CO2-Ph-4-Py which possess a CH2 between the Cp ring and the pendant group and unsubstituted Cp ring exhibit the lower cytotoxicity, and lower genotoxic effect. As a result, it exhibits a mild reduction on mitotic rate compared to the untreated cells due to its low cytotoxicity.
A series of ferrocene derivatives using phenol pendant groups with various substituents on the 4-position (Fc-CO2-Ph-4-X, X = F, Cl, Br, I, pyrrole) have been reported by our group [14]. In vitro studies on MCF-7 combined with docking studies on the ERα revealed that their anti-proliferative activities are not associated to the anti-estrogenic properties of the ferrocene but rather to the cytotxic activity of the metallocene unit [14]. Noteworthy, the binding affinity of Fc-CO2-Ph-4-Py in the ERα is lower than for the Fc-CO2-Ph-4-X ( X = F, Cl, Br, I) complexes [14]. Additionally, Fc-CO2-Ph-4-Py has high anti-proliferative activity on hormone-dependent breast cancer cell line MCF-7 but it has also high anti-proliferative activity on non-tumorigenic human breast epithelial cell line MCF-10A. Thus, ERα is not the responsible for the anti-proliferative activity of Fc-CO2-Ph-4-Py.
3.5 Measurements of reactive oxygenated species (ROS)
ROS induction assay was performed on MCF-7 to determine the relationship of the cytotoxic and genotoxic effect of the ferrocenoyl derivatives Fc-CO2-estradiol, Fc-(CO2-Ph-4-Py)CO2H, Fc-(CO2-Ph-4-Py)2, and Fc-CH2CO2-Ph-4-Py, Figure 5.
Figure 5.
ROS production induced by (A)Fc-CO2-estradiol, (B) Fc-(CO2-Ph-4-Py)CO2H, (C) Fc-(CO2-Ph-4-Py)2, and (D) Fc-CH2CO2-Ph-4-Py generated on MCF-7 at 72 h.
Fc-CO2-estradiol and Fc-(CO2-Ph-4-Py)2 which exhibit the higher micronucleus forming activity, also shows significant increase in ROS production. There is a direct correlation with ROS formation with concentration and a significant increase around 1000 μM. The amount of ROS formation by Fc-CO2-estradiol and Fc-(CO2-Ph-4-Py)2 are similar to FeCp2 but lower than [FeCp2]BF4 as reported by C.Y. Acevedo et al. [39]. Although these results suggest that the genotoxic activities of Fc-CO2-estradiol and Fc-(CO2-Ph-4-Py)2 are likely correlated to the ROS formation, other possible mechanisms cannot be ruled out. Along this line, the ROS formation of Fc-(CO2-Ph-4-Py)CO2H is the lowest but the IC50 is comparable to Fc-(CO2-Ph-4-Py)2. Meanwhile, the ROS production for Fc-CH2CO2-Ph-4-Py, is low in comparison with FcCp2 [39] and it has no relationship to concentrations. However, this last result was expected considering the low cytotoxic effect and micronucleus forming activity of Fc-CH2CO2-Ph-4-Py. This result suggests that genotoxic effect and ROS production of Fc-CH2CO2-Ph-4-Py is related to its poor cytotoxic activity.
4. Concluding remarks
In the present work, we have addressed the influence of the redox active pendant groups on ferrocene with regard to their anti-proliferative activity, genotoxic effects and ROS formation on breast cancer cell line. We utilized a combined approach using cancer cell lines and normal cell line to elucidate the real potential use of these species as chemotherapeutics and the cytokinesis-block micronucleus assay and ROS induction assay to measure the amount of DNA damage (genotoxic effects). We also performed cyclic voltammetry experiments to study the redox behavior of these new ferrocenes. First, in comparison to Fc-CO2-estradiol and Fc-CO2-Ph-4-Br which both have IC50 values of 9.2 μM but different redox potentials (756 and 532 mV respectively), we observe similar trends among the three new ferrocene-pyrrole complexes. Given that the electrochemical studies were performed on CH3CN, a non-physiological media, the redox data must be taken with caution and cannot explain the antiproliferative activities among these species. Second, Fc-CO2-estradiol has the higher micronucleus-forming activity and on MCF-7 cell line followed by Fc-(CO2-Ph-4-Py)2 > Fc-(CO2-Ph-4-Py)CO2H > Fc-CH2CO2-Ph-4-Py. This trend is correlated to their IC50 values. The ROS production assay confirmed the genotoxic activity of Fc-CO2-estradiol and Fc-(CO2-Ph-4-Py)2 due to reactive oxygen species formation. Thus, this study provides evidences that the pendant groups have influence in the anti-proliferative activity and in less degree to the DNA damage on MCF-7 breast cancer cell line. The genotoxic effects of these new functionalized ferrocene complexes come mainly from the ferrocene moiety and not from the anti-estrogenic effects.
Acknowledgments
EM thanks the financial support of NIH-RISE 2 Best program (NIH-R25GM088023) for the research assistantships of Yarelys Soto (undergraduate student) and Wanda I. Pérez (graduate student). JM acknowledge the support of the NCI Center to Reduce Cancer Health Disparities (CRCHD), Diversity Training Center to Reduce Health Disparities and NIH-MBRS Program grants #: S06 GM008239-20, MBRS-SC1 grants #: SC1 ISA157250-01 and 019SC1CA182846-04 to the Ponce School of Medicine and Health Sciences (PSHMS) and the Molecular and Genomics Core (NIMHD Grant MD007579). The authors also acknowledge the support of the PSMHS-Moffitt Cancer Center U54 Cancer Partnership through Grant 1 U54 CA163068-01A1.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Köpf-Maier P, Köpf H. A New Age for Iron: Antitumoral Ferrocenes. Cancer Res Clin Oncol. 1984;108:336–340. doi: 10.1007/BF00390468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Braga SS, Silva AMS. Ferricenium complexes: A new type of water-soluble antitumor agent. Organometallics. 2013;32:5626–5639. [Google Scholar]
- 3.Gasser G, Ott I, Metzler-Nolte N. Organometallic Anticancer Compounds. J Med Chem. 2011;54:3–25. doi: 10.1021/jm100020w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Jaouen G, Metzler-Nolte N. Medicinal Organometallic Chemistry. Springer-Verlag; Berlin, Heidelberg, Dordrecht, London, NY: 2010. pp. 81–117. [Google Scholar]
- 5.Fouda MFR, Abd-Elzaher MM, Abdelsamaia RA, Labib AA. On the medicinal chemistry of ferrocene. App Organometal Chem. 2007;21:613–625. [Google Scholar]
- 6.Jaouen G. Bioorganometallics. Wiley-VCH; Weinheim, Germany: 2006. pp. 65–95. [Google Scholar]
- 7.van Staveren DR, Metzler-Nolte N. Bioorganometallic Chemistry of Ferrocene. Chem Rev. 2004;104:5931–5985. doi: 10.1021/cr0101510. [DOI] [PubMed] [Google Scholar]
- 8.Nguyen A, Top S, Pigeon P, Vessières A, Hillard EA, Plamont MA, Huché M, Rigamonti C, Jaouen G. Synthesis and Structure-Activity Relationships of Ferrocenyl Tamoxifen Derivatives with Modified Side Chains. Chem Eur J. 2009;15:684–696. doi: 10.1002/chem.200801108. [DOI] [PubMed] [Google Scholar]
- 9.Top S, Vessières A, Leclercq G, Quivy J, Tang J, Vaissermann J, Huché M, Jaouen G. Synthesis, Biochemical Properties and Molecular Modelling Studies of Organometallic Specific Estrogen Receptor Modulators (SERMs), the Ferrocifens and Hydroxyferrocifens: Evidence for an Antiproliferative Effect of Hydroxyferrocifens on both Hormone-Dependent and Hormone-Independent Breast Cancer Cell Lines. Chem Eur J. 2003;9:5223–5236. doi: 10.1002/chem.200305024. [DOI] [PubMed] [Google Scholar]
- 10.Vessières A, Top S, Beck W, Hillard E, Jaouen G. Metal complex SERMs (selective oestrogen receptor modulators). The influence of different metal units on breast cancer cell antiproliferative effects. Dalton Trans. 2006:529–541. doi: 10.1039/b509984f. [DOI] [PubMed] [Google Scholar]
- 11.Vessières A, Top S, Pigeon P, Hillard E, Boubeker L, Spera D, Jaouen G. Modification of the Estrogenic Properties of Diphenols by the Incorporation of Ferrocene. Generation of Antiproliferative Effects in Vitro. J Med Chem. 2005;48:3937–3940. doi: 10.1021/jm050251o. [DOI] [PubMed] [Google Scholar]
- 12.Meléndez E. Metallocenes as target specific drugs for cancer treatment. Inorg Chim Acta. 2012;393:36–52. doi: 10.1016/j.ica.2012.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Vera JL, Gao LM, Santana A, Matta J, Meléndez E. Vectorized ferrocenes with estrogens and vitamin D2: synthesis, cytotoxic activity and docking studies. Dalton Trans. 2011;40:9557–9565. doi: 10.1039/c1dt10995b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Vera JL, Rullán J, Santos N, Jiménez J, Rivera J, Santana A, Briggs J, Rhenigold AL, Matta J, Meléndez E. Functionalized ferrocenes: The role of the para substituent on the phenoxy pendant group. J Organometal Chem. 2014;749:204–214. doi: 10.1016/j.jorganchem.2013.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ghorab MM, Alsaid MS, Ceruso M, Nissan YM, Supuran CT. Carbonic Anhydrase Inhibitors: Synthesis, Molecular Docking, Cytotoxic and Inhibition of the Human Carbonic Anhydrase Isoforms I, II, IX, XII with Novel Benzenesulfonamides Incorporating Pyrrole, Pyrrolopyrimidine and Fused Pyrrolopyrimidine Moieties. Bioorg Med Chem. 2014;22:3684–3695. doi: 10.1016/j.bmc.2014.05.009. [DOI] [PubMed] [Google Scholar]
- 16.Abou El Ella DA, Ghorab MM, Noaman E, Heiba HI, Khalil AI. Molecular Modeling Study and Synthesis of Novel pyrrolo[2,3-D]pyrimidines and Pyrrolotriazolopyrimidines of Expected Antitumor and Radioprotective Activities. Bioorg Med Chem. 2008;16:2391–2402. doi: 10.1016/j.bmc.2007.11.072. [DOI] [PubMed] [Google Scholar]
- 17.Chatzopoulou M, Patsilinakos A, Vallianatou T, Prnova MS, Žakelj S, Ragno R, Stefek M, Kristl A, Tsantili-Kakoulidou A, Demopoulos VJ. Decreasing Acidity in a Series of Aldose Reductase Inhibitors: 2-Fluoro–4-(1H-Pyrrol-1-Yl)phenol as a Scaffold for Improved Membrane Permeation. Bioorg Med Chem. 2014;22:2194–2207. doi: 10.1016/j.bmc.2014.02.016. [DOI] [PubMed] [Google Scholar]
- 18.Mohamed MS, Kamel R, Abd El-hameed RH. Evaluation of the Anti-Inflammatory Activity of Some pyrrolo[2,3-D]pyrimidine Derivatives. Med Chem Res. 2013;22:2244–2252. [Google Scholar]
- 19.Hassan SM, El-Maghraby AA, Abdel Aal MM, Bashandy MS. Heteroaromatization with Sulfonamido Phenyl Ethanone, Part I: Synthesis of Novel Pyrrolo[2,3-D]Pyrimidine and Pyrrolo[3,2-E][1,2,4]Triazolo[1,5-C]Pyrimidine Derivatives Containing Dimethylsulfonamide Moiety. Phosphorus Sulfur Silicon Relat Elem. 2009;184:291–308. [Google Scholar]
- 20.Esteves MA, Ortet O, Capelo A, Supuran CT, Marques SM, Santos MA. New Hydroxypyrimidinone-Containing Sulfonamides as Carbonic Anhydrase Inhibitors Also Acting as MMP Inhibitors. Bioorg Med Chem Lett. 2010;20:3623–3627. doi: 10.1016/j.bmcl.2010.04.109. [DOI] [PubMed] [Google Scholar]
- 21.Clark MP, George KM, Bookland RG, Chen J, Laughlin SK, Thakur KD, Lee W, Davis JR, Cabrera EJ, Brugel TA, et al. Development of New Pyrrolopyrimidine-Based Inhibitors of Janus Kinase 3 (JAK3) Bioorg Med Chem Lett. 2007;17:1250–1253. doi: 10.1016/j.bmcl.2006.12.018. [DOI] [PubMed] [Google Scholar]
- 22.Merighi S, Mirandola P, Varani K, Gessi S, Leung E, Baraldi PG, Tabrizi MA, Borea PA. A Glance at Adenosine Receptors: Novel Target for Antitumor Therapy. Pharmacol Ther. 2003;100:31–48. doi: 10.1016/s0163-7258(03)00084-6. [DOI] [PubMed] [Google Scholar]
- 23.Mossman T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983;65:55–63. doi: 10.1016/0022-1759(83)90303-4. [DOI] [PubMed] [Google Scholar]
- 24.Denizot F, Lang R. Rapid colorimetric assay for cell growth and survival. J Immunol Methods. 1986;89:271–277. doi: 10.1016/0022-1759(86)90368-6. [DOI] [PubMed] [Google Scholar]
- 25.Fenech M. Cytokinesis-block micronucleus cytome assay. Nature Protocols. 2007;2:1084–1104. doi: 10.1038/nprot.2007.77. [DOI] [PubMed] [Google Scholar]
- 26.Decordier I. Influence of caspase activity on micronuclei detection: a possible role for caspase-3 in micronucleation. Mutagenesis. 2005;20:173–179. doi: 10.1093/mutage/gei025. [DOI] [PubMed] [Google Scholar]
- 27.Tabbi G, Cassino C, Cavigiolio G, Colangelo D, Ghiglia A, Viano I, Osella D. Water stability and cytotoxic activity relationship of a series of ferrocenium derivatives. ESR insights on the radical production during the degradation process. J Med Chem. 2002;45:5786–5796. doi: 10.1021/jm021003k. [DOI] [PubMed] [Google Scholar]
- 28.Osella D, Mahboobi H, Colangelo D, Cavigiolio G, Vessières A, Jaouen G. FACS analysis of oxidative stress induced on tumour cells by SERMs. Inorg Chim Acta. 2005;358:1993–1998. [Google Scholar]
- 29.Fenech M, Morley AA. Cytokinesis-block micronucleus method in human lymphocytes: effect of in vivo ageing and low dose X-irradiation. Mutat Res. 1986;161:193–198. doi: 10.1016/0027-5107(86)90010-2. [DOI] [PubMed] [Google Scholar]
- 30.Schmid W. The micronucleus test. Mutat Res. 1975;31:9–15. doi: 10.1016/0165-1161(75)90058-8. [DOI] [PubMed] [Google Scholar]
- 31.Heddle J. A rapid in vivo test for chromosomal damage. Mutat Res Mol Mech Mutagen. 1973;18:187–190. doi: 10.1016/0027-5107(73)90035-3. [DOI] [PubMed] [Google Scholar]
- 32.Fenech M. The in vitro micronucleus technique. Mutat Res. 2000;455:81–95. doi: 10.1016/s0027-5107(00)00065-8. [DOI] [PubMed] [Google Scholar]
- 33.Fenech M, Morley AA. Measurement of micronuclei in lymphocytes. Cytobios. 1985;43:233–246. doi: 10.1016/0165-1161(85)90015-9. [DOI] [PubMed] [Google Scholar]
- 34.Fenech M, Morley AA. Solutions to the kinetic problem in the micronucleus assay. Mutat Res. 1985;147:29–36. [PubMed] [Google Scholar]
- 35.Yared E, McMillan TJ, Martin FL. Genotoxic effects of oestrogens in breast cells detected by the micronucleus assay and the Comet assay. Mutagenesis. 2002;17:345–352. doi: 10.1093/mutage/17.4.345. [DOI] [PubMed] [Google Scholar]
- 36.Fischer WH, Keiwan A, Schmitt E, Stopper H. Increased formation of micronuclei after hormonal stimulation of cell proliferation in human breast cancer cells. Mutagenesis. 2001;16:209–212. doi: 10.1093/mutage/16.3.209. [DOI] [PubMed] [Google Scholar]
- 37.Stopper H, Schmitt E, Gregor C, Mueller SO, Fischer WH. Increased cell proliferation is associated with genomic instability: elevated micronuclei frequencies in estradiol-treated human ovarian cancer cells. Mutagenesis. 2003;18:243–247. doi: 10.1093/mutage/18.3.243. [DOI] [PubMed] [Google Scholar]
- 38.Fleck SC, Hildebrand AA, Pfeiffer E, Metzler M. Catechol metabolites of zeranol and 17β-estradiol: A comparative in vitro study on the induction of oxidative DNA damage and methylation by catechol-O-methyltransferase. Toxicol Lett. 2012;210:9–14. doi: 10.1016/j.toxlet.2012.01.010. [DOI] [PubMed] [Google Scholar]
- 39.Acevedo CY, Meléndez E, Singh SP, Ramirez-Vick JE. Cytotoxicity and Reactive Oxygen Species Generated by Ferrocenium and Ferrocene on MCF-7 and MCF-10A Cell Lines. J Cancer Sci Ther. 2012:04. [Google Scholar]



