• Nem Talált Eredményt

Synthesis and investigation of the anticancer effects of estrone-16-oxime ethers in vitro

N/A
N/A
Protected

Academic year: 2022

Ossza meg "Synthesis and investigation of the anticancer effects of estrone-16-oxime ethers in vitro"

Copied!
10
0
0

Teljes szövegt

(1)

Synthesis and investigation of the anticancer effects of estrone-16-oxime ethers in vitro

Ágnes Berényi

a

, Renáta Minorics

a

, Zoltán Iványi

b

, Imre Ocsovszki

c

, Eszter Ducza

a

, Hubert Thole

b

, Josef Messinger

b

, János Wölfling

b

, Gerg } o Mótyán

b

, Erzsébet Mernyák

b

, Éva Frank

b

, Gyula Schneider

b,

, István Zupkó

a,

aDepartment of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary

bDepartment of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary

cDepartment of Biochemistry, University of Szeged, Dóm tér 9, H-6720 Szeged, Hungary

a r t i c l e i n f o

Article history:

Received 27 June 2012

Received in revised form 28 September 2012

Accepted 10 October 2012 Available online 2 November 2012

Keywords:

Estrone-16-oxime ethers Cell viability

Apoptosis Cell cycle blockade

a b s t r a c t

An expanding body of evidence indicates the possible role of estrane derivatives as useful anticancer agents. The aim of this study was to describe the cytotoxic effects of 63 newly synthetized estrone-16- oxime ethers on human cancer cell lines (cervix carcinoma HeLa, breast carcinoma MCF7 and skin epider- moid carcinoma A431), studied by means of the MTT assay. Four of the most promising compounds were selected for participation in additional experiments in order to characterize the mechanism of action, including cell cycle analysis, morphological study and the 5-bromo-20-deoxyuridine incorporation assay.

The cancer selectivity was tested on a noncancerous fibroblast cell line (MRC-5). Since apoptosis and cell cycle disturbance were observed, caspase-3 activities were further assayed for the two most effective agents. These estrone-16-oxime analogs activated caspase-3 and changed the mRNA level expression of endogenous factors regulating the G1–S phase transition (retinoblastoma protein, CDK4 and p16).

The repression of retinoblastoma protein was reinforced at a protein level too. These experimental data lead to the conclusion that estrone-16-oxime ethers may be regarded as potential starting structures for the design of novel anticancer agents.

Ó2012 Elsevier Inc. All rights reserved.

1. Introduction

Endogenous steroidal compounds exert an outstandingly broad spectrum of physiological functions in spite of the conserved struc- ture, indicating that minor chemical changes may lead to mole- cules with substantial property differences. The steroidal skeleton is therefore the backbone for the design and synthesis of original drug candidates with a wide range of pharmacological activities. Many plant steroids are reported to possess inotropic, antihypercholesterolemic, anti-inflammatory and antioxidant properties[1–5]. The anticancer capacity is one of the most inten- sively investigated characters of natural steroid products and syn- thetic analogs. Diosgenin, one of the most widely studied saponins, exerts antiproliferative activities against several human cancer cell lines, such as osteosarcoma 1547, HER2-overexpressing breast cancer, melanoma M4Beu, leukemia K562, HEL, and colon HT-29 [6–11]. Apoptosis induction and up-regulation of tumor suppressor p53 have been reported to be crucial components of the mecha-

nism of the growth-inhibitory action of the compound in several types of malignant cells [8]. Similarly, steroidal glycoalkaloids, including

a

-tomatine and solanine, displayed an appreciable inhi- bition of human colon (HT-29) and liver (HepG2) cancer cell prolif- eration [12]. These capacities were greater than those of the aglycones tomatidine and solanidine, respectively, indicating that the steroidal backbone is not an exclusive factor determining the overall cytostatic activities of these natural products.

Besides those of the active components isolated from plants, the outstanding anticancer and cardioprotective properties of endoge- nous estrogen hormone metabolites have been described[13]. The final steps in the oxidative conversion of estradiol involve hydrox- ylation of rings A and D of the estrone skeleton. 2-Methoxyestradi- ol, an intensively investigated metabolite synthesized by catechol- O-methyltransferase, exerts a pronounced anticancer effect on a broad variety of human malignant cell lines of reproductive origin [14–16].

Introduction of an oxime function into an appropriate skeleton is a reasonable approach to the preparation of potent cytotoxic agents. A set of oxime-containing flavone and isoflavone deriva- tives have been synthetized and tested against three human adher- ent cell lines, and the effective agents were further evaluated 0039-128X/$ - see front matterÓ2012 Elsevier Inc. All rights reserved.

http://dx.doi.org/10.1016/j.steroids.2012.10.009

Corresponding authors. Tel.: +36 62 546839 (I. Zupkó).

E-mail addresses:Schneider@chem.u-szeged.hu(G. Schneider),zupko@pharm.

u-szeged.hu(I. Zupkó).

Contents lists available atSciVerse ScienceDirect

Steroids

j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / s t e r o i d s

(2)

against the full panel of 60 human tumor cell lines derived from nine cancer types. Flavon-6-yl oximes proved to exhibit the most pronounced cytostatic effects, with the lowest mean GI50 value as low as 0.08

l

M. The apoptosis induction of the most effective compound was evidenced by flow-cytometry.

The antiproliferative activity of a 5-nitrofuran-2-yl-vinyl quino- line derivative was increased when its oxo function was substi- tuted with a H-bond-donating oxime group. The derivative containing the H-bond-accepting methyloxime was less favorable against cancer cells of reproductive origin[17]. The (Z) and (E) iso- mers of cholest-4-en-6-one oxime also exhibited moderate, but cancer-selective growth-inhibitory properties[18].

The aim of the present study was the synthesis of a set of es- trone oximes and a two-step investigation of their direct action against the viability of human adherent cancer cell lines HeLa (cer- vix adenocarcinoma), MCF7 (breast adenocarcinoma) and A431 (skin epidermoid carcinoma). Compounds exhibiting noteworthy IC50 values were subjected to further experiments targeting the cancer selectivity and the mechanism of action, including cell cycle analysis, fluorescent microscopy, and determination of the cas- pase-3 activity and the expression of crucial cell cycle-regulating factors.

2. Experimental 2.1. General

Melting points (mp) were determined on a Kofler block and are uncorrected. Specific rotations were measured in CHCl3 (c 1) at 20sC with a POLAMAT-A (Zeiss-Jena) polarimeter and are given in units of 101cm2g1. Elementary analysis data were deter- mined with a Perkin-Elmer CHN analyzer model 2400. The reac- tions were monitored by TLC on Kieselgel-G (Merck Si 245 F) layers (0.25 mm thick); solvent systems (ss): (A)tert-butyl methyl ether/dichloromethane (5:95, v/v), (B) tert-butyl methyl ether/

dichloromethane (10:90, v/v), (C) acetone/toluene/n-hexane (35:30:30, v/v). The spots were detected by spraying with 5% phos- phomolybdic acid in 50% aqueous phosphoric acid. TheRfvalues were determined for the spots observed by illumination at 245 and 365 nm. Flash chromatography: silica gel 60, 40–63

l

M. All solvents were distilled prior to use. NMR spectra were recorded on a Bruker DRX500 instrument at 500 (1H NMR) or 125 MHz (13C NMR). Chemical shifts are reported in ppm (dscale), and cou- pling constants (J) in Hz. For the determination of multiplicities, theJ-MOD pulse sequence was used.

2.2. Materials and methods

The search for steroids with novel biological effects led to the synthesis of 16-oximinoestrone derivatives. The starting materials of the syntheses were 3-hydroxyestra-1,3,5(10)-trien-17-one (1a), 3-methoxyestra-1,3,5(10)-trien-17-one (1b), 3-benzyloxyestra-1,3, 5(10)-trien-17-one (1c), 3-p-methoxybenzyloxyestra-1,3,5(10)-tri- en-17-one (1d), 3-sulfamoyloxyestra-1,3,5(10)-trien-17-one (1e), and their 13

a

-methyl epimers (2a–d). The different steroidal oxi- mes were synthetized as outlined inFig. 1.

Introduction of the oxime group at position 16 was achieved after the deprotonation of 1a–eand2a–dthrough the action of potassiumtert-butoxide and reaction with an excess of isoamyl ni- trite, affording oximes3a–eand4a–din yields of 85–90%.

O-Alkylation of3a–eand4a–dwith a large excess of alkyl io- dide in the presence of 4 equivalents of Ag2O in diethyl ether affor- ded the requiredO-alkyl oximes5a–d,7a–e,9a–dand11a,bin the natural (13b-methyl) series. The same methods were used in the 13-epimer (13

a

-methyl) series, yielding8a–cand10a–d. The reac-

tion was generally complete within 30–45 min.O-Benzoylation of oximes3a–eand4a–dwith benzyl bromide or substituted benzyl bromides yielded O-benzyl oximes 7f,g, 9e–g, 10e,f, 11c–e and 12a–c. The reactions at room temperature needed 6–12 h, depend- ing on the electron-donating or electron-withdrawing character of the benzyl bromide substituent. 3-Sulfamoyloxy-16-alkoximinoe- strone derivatives (6a,b) were prepared by the reaction of sulfa- moyl chloride and 16-alkoximinoestrone (5b,c) in N,N- dimethylformamide.O-Acyl oximes7h,i,8d,e,9h,i, and10g,hwere prepared with acetic anhydride or propionic anhydride in pyridine.

The crude products were purified by flash chromatography.

2.3. Synthesis of 16-oximinoestrone derivatives (3a–eand4a–d;

general method)

Estrone (1a) and its 3-substituted derivatives (1b–e) in the nat- ural series (13b-methyl) and their 13

a

-methyl analogs (2a–d) (10.0 mmol) were stirred under nitrogen in 40 ml of freshly dis- tilledtert-butanol for 10 min. Potassium tert-butoxide (55 mmol, 6.0 g) was added to the suspension, and stirring was continued for 30 min, when dissolution was complete. n-Butyl nitrite (20 mmol, 2.8 ml) was added from a syringe, and the solution turned deep-red and then orange within a few minutes. A second portion ofn-butyl nitrite (20 mmol, 2.8 ml) was added after 6 h, and stirring was continued for an additional 12 h. The reaction mixture was next quenched in 100 g of ice and acidified with gla- cial acetic acid. The precipitate that formed was filtered off, washed until neutral with water, and dried over P2O5. The crude products3a–dand4a–dwere subjected to chromatographic sepa- ration on a silica gel column withtert-butyl methyl ether/light petroleum (20:90).3ewas chromatographed with ethyl acetate/

dichloromethane (10:90).

2.4. Synthesis of 16-alkoximinoestrone derivatives (5a–d,7a–e,8a–c, 9a–d,10a–dand11a,b; general method)

The 16-oximinoestrone derivative (3a–d and 4a–d) (1.00 mmol) and 700 mg (3.00 mmol) of Ag2O were suspended in anhydrous diethyl ether (30 ml) and the suspension was stirred at room temperature for 30 min. Alkyl iodide (25 mmol) was then added dropwise to the reaction mixture, which was stirred until completion of the reaction (monitored by TLC). The excess Ag2O and silver salt were filtered off from the suspension and washed with diethyl ether (50 ml), and the organic phase was evaporated.

The crude product was purified by flash chromatography.5a–d were chromatographed withtert-butyl methyl ether/light petro- leum (20:80), and7a–e,8a–c,9a–d,10a–dand11a,bwith dichlo- romethane/petroleum ether (30:70) or dichloromethane.

2.5. Synthesis of 16-benzyloximinoestrone derivatives (7f,g,9f,g,10e,f, 11d,eand12a–c; general method)

The 16-oximinoestrone derivative (3a–d and 4a–d) (1.00 mmol) and 700 mg (3.00 mmol) of Ag2O were suspended in anhydrous diethyl ether (50 ml) and the suspension was stirred at room temperature for 30 min. Benzyl bromide or substituted benzyl bromide (4.00 mmol) was then added to the reaction mix- ture, which was stirred until completion of the reaction (monitored by TLC). The excess Ag2O and silver salt were next filtered off and washed with diethyl ether (50 ml), and the organic phase was evaporated. The residual crude product was chromatographed with dichloromethane/light petroleum (1:1) or dichloromethane.

70 Á. Berényi et al. / Steroids 78 (2013) 69–78

(3)

2.6. Synthesis of 3-sulfamoyloxy-16-alkoxyiminoestrone derivatives (6a,b)

The 16-alkoxyiminoestrone derivative (5b,5c) (1.00 mmol) was dissolved in dimethylformamide (25 ml) and sulfamoyl chloride (10 mmol) was added in small portions during cooling in ice. The reaction mixture was stirred at 0sC for 6 h, and was then poured into ice-water. The precipitate that formed was filtered off and dis- solved in dichloromethane, and the solution was chromato-

graphed on silica gel with tert-butyl methyl ether/light petroleum (20:80).

2.7. Synthesis of 16-acetoximinoestrone derivatives (7h,8d,9hand 10g) and 16-propionyloxyestrone derivatives (7i,8e,9iand10h)

The 16-oximinoestrone compound (3b,c and 4b,c) (1 mmol) was dissolved in pyridine (5 ml), and acetic anhydride (5 ml) or propionic anhydride (5 ml) was added during cooling in ice. The Fig. 1.Chemical structures of the synthetized and investigated estrone-16-oxime ethers. Reagents: (i) isoamyl nitrite, KOtBu,tBuOH; (ii) BnBr or substituted BnBr, Ag2O, Et2O.

(4)

reaction mixture was allowed to stand at room temperature for 24 h, and was then poured into ice-water. The precipitate that formed was filtered off, washed, dried and recrystallized from ace- tone/light petroleum to yield7h,8d,9hand10g, or7i,8e,9iand 10h.

2.8. Cell culturing

Human cancer cell lines (HeLa, MCF7 and A431, isolated from cervix and breast cancers and skin epidermoid carcinoma, respec- tively) and noncancerous MRC-5 human lung fibroblasts were maintained in minimal essential medium supplemented with 10% fetal bovine serum (FBS), 1% non-essential amino acids and an antibiotic–antimycotic mixture (AAM). A2780 cells (isolated from ovarian cancer) were maintained in RPMI medium supple- mented with 10% FBS, 1% AAM and 1%L-glutamine. All cell lines were purchased from the European Collection of Cell Cultures (Salisbury, UK). For pharmacological investigations, 10 mM stock solutions of the tested compounds were prepared with dimethyl sulfoxide (DMSO). The highest DMSO concentration of the medium (0.3%) did not have any substantial effect on the determined cellu- lar functions. All the chemicals, if otherwise not specified, were purchased from Sigma–Aldrich Ltd. (Budapest, Hungary).

2.9. Antiproliferative activity measured by MTT assay

The effects on the viability of malignant cells were determined in vitroon the four human cancer cell lines: HeLa, MCF7, A431 and A2780. The cells were grown in a humidified atmosphere of 5% CO2

at 37°C. Cells were seeded onto 96-well plates at a density of 5000 cells/well and allowed to stand overnight, after which the medium containing the tested compound was added. After a 72 h incubation period, viability was determined by the addition of 20

l

L of MTT ([3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoli- um bromide]) solution (5 mg/mL). The precipitated formazan crys- tals were solubilized in DMSO and the absorbance was read at 545 nm with an ELISA reader. Two independent experiments were performed with five parallel wells; cisplatin, an agent administered clinically in some gynecological malignancies, was used as positive control. Sigmoidal dose–response curves were fitted to the mea- sured points, and the IC50 values were calculated by means of GraphPad Prism 4.0 (GraphPad Software; San Diego, CA, USA)[19].

2.10. Analysis of cell cycle by flow cytometry

Flow cytometric analysis was performed in order to character- ize the cellular DNA content of treated HeLa cells. After treatment for 24 or 48 h, the cells (200,000/condition) were trypsinized (Gib- co BRL, Paisley, U.K.), washed with phosphate-buffered saline (PBS) and fixed in 1.0 mL of cold 70% ethanol for 30 min on ice. After two washing steps in cold PBS, the DNA was stained with propidium io- dide (10

l

g/mL) in the presence of RNA-ase (50

l

g/mL). The sam- ples were then analyzed by FACStar (Becton–Dickinson;

Mountain View, CA, USA). In each analysis, 20,000 events were re- corded, and the percentages of the cells in the different cell-cycle phases (subG1, G1, S and G2/M) were calculated by using win- MDI2.9. The subG1 fractions were regarded as the apoptotic cell population[20].

2.11. Hoechst 33258–propidium iodide double staining

Near-confluent HeLa cells were seeded into a 96-well plate (5000 cells/well). After incubation for 24 h with the test com- pound, Hoechst 33258 and propidium iodide were added to the culture medium to give final concentrations of 5 and 2

l

g/mL, respectively. The cells were incubated with the staining mixture

for 1 h at 37°C and were then photographed by means of a Nikon Eclipse microscope equipped with an epifluorescence attachment containing the appropriate optical blocks and a QCapture CCD camera. The staining allowed the identification of live, early-apop- totic, late-apoptotic and necrotic cells. Hoechst 33258 permeates all the cells and makes the nuclei appear blue. Apoptosis was re- vealed by nuclear changes such as chromatin condensation and nu- clear fragmentation. The necrotic and the late-apoptotic cells were identified as cells with propidium iodide uptake, which indicates loss of membrane integrity, the cell nuclei being stained red[21].

2.12. 5-Bromo-20-deoxyuridine (BrdU) incorporation assay

BrdU incorporation into the cellular DNA was determined with the BrdU Labeling and Detection Kit I (Roche Diagnostic, Mann- heim, Germany) on HeLa cells (3000/well) treated with the test compound for 24 h. The incorporation of BrdU in place of thymi- dine was monitored as a parameter for DNA synthesis. In accor- dance with the manufacturer’s instructions, cells were labeled with BrdU for 1 h, followed by fixation. The cellular DNA was par- tially digested by nuclease treatment and labeled with mouse monoclonal antibody. Finally, the fluorescein-conjugated anti- mouse antibody was added and the wells were examined by fluo- rescent microscopy with the use of an appropriate optic block (ex:

465–495 nm, em: 515–555 nm, dichromatic mirror: 505 nm). At least 400 cells were counted from four parallel wells for the expression of the BrdU-positive cells.

2.13. Caspase-3 assay

The activity of caspase-3 from treated cells was determined in triplicate by means of a commercially available colorimetric kit in accordance with the instructions of the supplier (Sigma–Aldrich, Budapest, Hungary). Briefly, HeLa cells (16 million per condition) were exposed to the test item for 48 h and then scraped, counted and resuspended in lysis buffer (10

l

L for 1 million of cells). The caspase-3 activity was measured by the addition of substrate (Ac-DEVD-pNA) and the amount of product (pNA) was measured at 405 nm after incubation for 17 h. Results on treated cells are gi- ven as fold-increase by direct comparison with the untreated con- trol results.

2.14. Reverse transcription-polymerase chain reaction (RT-PCR) studies

The effects of the tested compounds on the mRNA expression pattern of the regulator factors retinoblastoma protein (Rb), cy- clin-dependent kinases 2 (CDK2), 4 (CDK4) and 6 (CDK6), and p16, p21, p27 and p53, which play crucial roles in the transition from the G1 to the S phase, were determined by RT-PCR in HeLa cells. After a 24 h incubation period, the media containing the var- ious test compounds were discarded and the total RNA was iso- lated from the cells (5105) by using the TRIzol Reagent in accordance with the instructions of the manufacturer (Molecular Research Center, Cincinnati, OH, USA)[22]. The pellet was resus- pended in 100

l

L of DNase- and RNase-free distilled water. The RNA concentrations of the samples were determined from their absorbances at 260 nm. The RNA (0.5

l

g) was mixed with DNase- and RNase-free distilled water and 20

l

M oligo(dT) (Invitrogen, Carlsbad, CA, USA) in a final reaction volume of 10

l

L, which was incubated at 70°C for 5 min. After the mixture had been cooled to 4°C, 20 U of RNase inhibitor, 20 U of MMLV reverse transcrip- tase (both from Promega, Madison, WI, USA), 200

l

M dNTP in 50 mM Tris–HCl, pH 8.3, 75 mM KCl and 5 mM MgCl2 in a final reaction volume of 10

l

L were added. The mixture was incubated at 37°C for 60 min. The PCR was carried out with 5

l

L of cDNA,

72 Á. Berényi et al. / Steroids 78 (2013) 69–78

(5)

25

l

L of ReadyMix Taq PCR reaction mix, 2

l

L of 20 pM sense and antisense primer of Rb, CDK2, CDK4, CDK6, p16, p21, p27 or p53 and 16

l

L of DNase- and RNase-free distilled water. The primer se- quences used to amplify Rb, CDK2, CDK4, p16, p21 and p27 were described by Gao et al. [23], that for p53 by Matsuhashi et al.

[24]and that for CDK6 by Huang et al.[25]. Human glyceraldehyde 3-phosphate dehydrogenase primers were used as internal control in all samples (Supplementary Table 1). The PCR was performed with an ESCO SWIFT MAXI thermal cycler (Esco Technologies, Phil- adelphia, PA, USA) and the products were separated on 2% agarose gels, stained with ethidium bromide and photographed under a UV transilluminator. Semiquantitative analysis was performed by den- sitometric scanning of the gel with Kodak Image Station 2000R (Eastman Kodak, Rochester, NY, USA).

2.15. Western blotting studies

HeLa cells were harvested in 60-mm dishes at a density of 2105cells/mL and treated with compounds3aand3e. Whole- cell extracts were prepared by washing the cells with PBS and sus- pending them in lysis buffer (50 mM Tris, 5 mM EDTA, 150 mM NaCl, 1% NP-40, 0.5% deoxycholic acid, 1 mM sodium orthovana- date, 100

l

g/mL PMSF and protease inhibitors)[26]. 50

l

g of pro- tein per well was subjected to electrophoresis on 4–12% NuPAGE Bis–Tris Gel in XCell SureLock Mini-Cell Units (Invitrogen, Carls- bad, CA, USA). Proteins were transferred from gels to nitrocellulose membranes, using the iBlot Gel Transfer System (Invitrogen, Carlsbad, CA, USA). Antibody binding was detected with the West- ernBreeze Chromogenic Western blot immunodetection kit (Invit- rogen, Carlsbad, CA, USA). The blots were incubated on a shaker with Rb, phosphorylated Rb (pRb) andb-actin polyclonal antibody (Santa Cruz Biotechnology, Santa Cruz, CA, USA) 1:200 in the block- ing buffer.

2.16. Statistical analysis

Statistical analysis was carried out by analysis of variance (AN- OVA), followed by the Dunnett post-tests. RT-PCR data were ana- lyzed by ANOVA, followed by the Neuman–Keuls post-tests. All statistical analyses of the data were performed with GraphPad Prism 4.0 (GraphPad Software; San Diego, CA, USA).

3. Results

3.1. Effects on viability of cancer cells (MTT assays)

The results of MTT assays with the 63 tested estrone analogs on three human adherent cancer cell lines are presented inSupple- mentary Table 2. Although a lower level of formazan production can be attributed to a decreased metabolic activity of the treated cells, the MTT assay is generally considered to be a suitable method for the determination of cell viability. Estrone (1a) exhibited pro- nounced action on HeLa cells, limited action on MCF7 cells and no relevant action on the A431 cell line. None of the estrone ethers (1b–d) proved more potent and the 13

a

epimers (2a–d) were also less effective. Introduction of an oxime function at position 16 of the estrane skeleton resulted in a general increase in cytostatic ac- tion.3aand3ewere highly effective, especially against HeLa cells.

Estrone analogs with a substituted oxime function (5a–d) were completely ineffective and their 3-sulfamoyloxy relatives (6a–c) likewise exhibited only limited actions. Estrone methyl ethers with a substituted oxime group at position 16 (7a–i and8a–e) were similarly minimally active, without any marked effect depending on the configuration of the methyl function at position 13. Sub- stantially more marked effects were detected when the aromatic

hydroxy group was etherified with benzyl or p-methoxybenzyl alcohol (9a–i,10a–j,11a–e,12a–c). Among these more effective compounds, 13b-methyl generally seemed to be preferred, though with some exceptions (9i–10h,9h–10g). In the light of these re- sults, four oximes were selected for furtherin vitroinvestigations (3a,3e,10hand11a). The viability assays were repeated with a set of dilutions of these agents (0.1–30

l

M) and extended to ovar- ian cancer cell line A2780, and IC50values were calculated (Table 1, Supplementary Fig. 1). All of these estrone oximes affected the pro- liferation of HeLa cells comparably to the reference agent cisplatin, while MCF7, A413 and A2780 cells were less sensitive. The viability of noncancerous fibroblast cell line MRC-5 was affected only by 11a, with a higher calculated IC50 value than that of cisplatin.

Agents with an unsubstituted oxime function (3aand3e) could be regarded as selective for HeLa cells, with some modest action against ovarian cancer (A2780) cell line, while10hand11adis- played a broader spectrum of activities.

3.2. Morphological studies and cell cycle distribution

HeLa cells were incubated with compounds3aand3eat 3, 10 and 30

l

M and with10hand11aat 10 and 30

l

M for 24 h. The presence of apoptosis or necrosis was determined according to the cell morphology and membrane integrity. Separate photos were recorded, in which Hoechst 33258 and propidium iodide fluo- rescence served as morphological markers (Fig. 2). Concentration- dependent increases in nuclear condensation and fragmentation and in membrane permeability were generally observed. The most markedly perturbed membrane integrity was seen in the case of 3e, while treatment with10hresulted in pronounced nuclear con- densation with poor staining, even at 30

l

M, indicating apoptotic cell death.

Treatments with these selected agents resulted in significant changes in the cell cycle distribution of HeLa cells (Fig. 3). A 24 h incubation with unsubstituted oximes (3aand3e) caused a pro- nounced decrease in the synthetic (S) phase and an increase in the G1 phase. At the highest concentration applied (30

l

M), an in- crease in the subdiploid (subG1) population was detected.

Although 11acaused a decrease in the G1 phase, the actions of agents with substituted oximes were less obvious. After a longer incubation (48 h),3aand3eresulted in a concentration-dependent increase in the subG1 cells, but the S population was reduced. This induction in subdiploid ratio was characteristic for 10h too, whereas the action of11awas limited to a slight S phase decrease at 10

l

M.

3.3. BrdU incorporation

The amount of synthetic thymidine analog BrdU incorporated was used to detect the overall activity of DNA synthesis. HeLa cells Table 1

Calculated antiproliferative IC50values of the tested estrone-16-oxime ethers.

Compounds IC50values (lM)a HeLa

cells

MCF7 cells

A431 cells

A2780 cells

MRC-5 cells

3a 4.41 >30 >30 18.28 >30

3e 4.04 >30 >30 11.96 >30

10h 3.52 4.13 >30 4.61 >30

11a 5.63 >30 13.25 25.05 6.94

Cisplatin 5.66 7.99 8.81 0.86 4.13

aMean value from two independent determinations with five parallel wells, standard deviation less than 15%.

(6)

were treated with two or three concentrations of the investigated compounds (3, 10 and 30

l

M) for 24 h. The incorporation of BrdU into the DNA was substantially and statistically significantly inhib- ited by all of the selected agents (Fig. 4). Compound3ewas the most potent in inhibiting BrdU incorporation and, similarly to3a, exhibited a clear concentration dependence; their actions were comparable to that of cisplatin.

3.4. Caspase-3 activity

The results detailed above led to two of the investigated com- pounds (3a and 3e) being selected for additional experiments, including determinations of the activity of caspase-3. The activity of this apoptosis-executing key enzyme was increased substan- tially and statistically significantly by3a and 3e(Fig. 5). While Fig. 2.Fluorescence microscopy images of Hoechst 33258–propidium iodide double staining. Two separate pictures from the same field were taken for the two markers. HeLa cells were treated with the vehicle (control), or with3a,3e,10hor11aat the given concentrations. The blue fluorescence (left panels) indicates Hoechst 33258, and the red coloration (right panels) is a result of cellular propidium iodide accumulation. The bar in the Hoechst 33258 control picture denotes 100lM.

Fig. 3.Effects of compounds3a,3e,10hand11aon the HeLa cell cycle distribution after incubation for 24 (panel A) or 48 h (panel B).,andindicatep< 0.05,p< 0.01 andp< 0.001, respectively, as compared with the control cells.

74 Á. Berényi et al. / Steroids 78 (2013) 69–78

(7)

3eresulted in a clear concentration–effect relationship,3acaused slightly lower activity at 30

l

M than at 10

l

M.

3.5. RT-PCR studies

The expressions of four cell cycle-regulating factors (CDK4, CDK6, p16 and Rb) that play key roles in the early G1–S transition were additionally determined by means of a semiquantitative RT- PCR technique (Fig. 6A). On the basis of the results of the cell cycle analyses and the BrdU incorporation assays, two compounds (3a and3e) were included at two concentrations (3 and 10

l

M), with exposure for 24 h. The expression of tumor suppressor gene p16 was substantially and statistically significantly increased at the mRNA level under all tested conditions. Treatment with these se- lected agents resulted in a concentration-dependent repression of CDK4, but not of CDK6. Retinoblastoma protein was significantly repressed by3aand3eat the above concentrations. 3eseemed more potent than3a in this respect. Further assayed factors in- volved in the regulation of the cell cycle (CDK2, p21, p53 and p27) did not exhibit statistically significant differences as com- pared with the control values (Fig. 6C).

3.6. Western blotting

Western blot analysis was performed to determine the expres- sion of Rb and postsynthetically phosphorylated Rb at a protein le- vel. Treatment with 3a and 3e at 3 and 10

l

M reduced the expression of both forms of Rb relative to the untreated cells (Fig. 6B). The relative expression of pRb, expressed as the density ratio Rb/pRb, was concentration-dependently decreased by the

two tested steroids. This ratio in the control cells was 1.53, which was decreased to 0.59 and 0.46 by3a, and to 1.06 and 0.80 by3e, in concentrations of 3 and 10

l

M, respectively. The expressions of this regulating factor at mRNA and protein levels were in good agreement.

4. Discussion

A substantial amount of evidence has become available con- cerning the antiproliferative properties of natural steroidal com- pounds and synthetic analogs. Digitalis glycosides have long been used in the treatment of congestive heart failure, and convincing observational data bear witness to their anticancer action [27,28]. The early reports have been reinforced and the mechanism of this action has been postulated, leading to the generally ac- cepted belief that the indications of cardiac glycosides may be ex- tended to some cancers in the near future [29]. Diosgenin and many related steroidal alkaloids characteristic of the Solanum and Dioscoreagenera exhibit potent anticancer effects, and may suggest starting structures for novel synthetic antiproliferative drugs[12,30].

Cytotoxic steroids containing an oxime function at position 6 have been isolated from marine sponges and promising synthetic analogs were recently designed which exhibit activities similar to those of the present agents [31]. (Z)- and (E)-cholest-4-en-7-one oximes and their lactam derivatives were reported to exert anti- proliferative action against HeLa and chronic myelogenous leuke- mia (K-562) cells, and the apoptosis-inducing capacity was additionally evidenced by means of morphological and biochemi- cal approaches[18]. Huang et al. have reported the synthesis and screening of a set of ring A-modified cholestanes bearing an oxime function at position 6. Some of these compounds exhibited consid- erable activities against HeLa, human liver (SMMC 7404) and hu- man gastric carcinoma (MGC 7901) cells [32]. These oximes are stated to exhibit anticancer properties similar to or lower than those of the currently investigated 16-oximes.

Estrogens may be responsible for carrying a pharmacophore moiety to estrogen receptor-expressing cells, and therefore deter- mining the molecular targeting of the agent[33,34]. We earlier re- ported on some estrane-based antiproliferative compounds with presumably hormone-independent actions [35,36]. The most intensively investigated estrane is an endogenous estradiol metab- olite, 2-methoxyestradiol, which does not exhibit hormonal activ- ity, but which seems to be highly effective against a broad range of cancer cell linesin vitro, and some limited but promisingin vivo data are also available.[13].

The aim of the present study was the design, synthesis and pharmacological investigation of novel estrone-16-oximes. We have found no previous report describing anticancer effects of compounds from this class. The screening of the 63 newly synthe- tized compounds against three human cancer cell lines pointed to some structure–activity relationships. As a general rule, though with a few exceptions, theborientation of the 13-methyl group is preferred. 3-Benzyl substitution typically favors the

a

position of the 13-methyl function. An unsubstituted oxime is generally preferred over an alkyl-substituted one, but an aromatic group (e.g. benzyl) may be considered. The hydroxy group on ring A may be unsubstituted, sulfamoyloxylated or substituted with an aromatic group (benzyl orp-methoxybenzyl). From the data relat- ing to the anticancer efficacy, four compounds were selected for further investigations. Two of them (3aand3e) may be regarded as HeLa-selective, with limited action on A2780 cells; the mole- cules containing substituted oxime groups (10hand11a) exerted substantial action on MCF7 and A431 cells too. Tumor selectivity is one of the most critical challenges in the development of a novel Fig. 4.Incorporation of 5-bromo-20-deoxyuridine into HeLa cells after incubation

for 24 h., andindicatep< 0.05,p< 0.01 andp< 0.001, respectively, as compared with the control cells.

Fig. 5.Induction of caspase-3 activity after incubation with compounds3aand3e for 48 h. The activity of untreated cells was taken as one unit.indicatesp< 0.001 as compared with the control cells.

(8)

anticancer agent. Although the MTT assay on intact human fibro- blasts cannot be viewed as a thorough toxicological evaluation, it is undoubtedly advantageous that three of the four selected com- pounds did not exert substantial action on MRC-5 cells.

A set of additional in vitro experiments was devoted to an experimental approach to the possible mechanism of action of these agents. Most of the currently available drugs used in antican- cer treatment have the capacity to induce programmed cell death Fig. 6.Expression of Rb, CDK4, CDK6 and p16 at the mRNA level after incubation with compounds3a, and3efor 24 h.,andindicatep< 0.05,p< 0.01 andp< 0.001, respectively, as compared with the control condition (panel A). Expressions of Rb and phosphorylated Rb at the protein level after incubation with compounds3aand3efor 24 h (panel B). Expression of CDK2, p21, p53 and p27 at the mRNA level after incubation with compounds3aand3efor 24 h (panel C).

76 Á. Berényi et al. / Steroids 78 (2013) 69–78

(9)

either by fortifying apoptotic signaling or by inhibiting antiapopto- tic signaling [37]. Treatment-dependent morphological changes were recorded and qualitatively evaluated by means of Hoechst 33258–propidium iodide fluorescent staining after incubation for 24 h. Typical apoptotic markers, such as cellular shrinkage, nuclear condensation and increased membrane permeability, were de- tected, especially at the highest concentrations of3aand3e. Corre- spondingly, flow cytometric cell cycle analysis was performed for a quantitative determination of the DNA content of HeLa cells as a function of the treatment. After a short exposure,3aand3ere- sulted in a marked suppression of the synthetic phase, and the sub- diploid population increased only at high concentration (30

l

M).

The same compounds elicited more pronounced cell cycle pertur- bation, including the accumulation of subG1 cells, after a longer incubation. It is therefore concluded that a period of 24 h is suffi- cient for the development of the morphological hallmarks of apop- tosis, but not for the complete activation of the self-decomposing enzymatic procedure, which may explain why the appearance of cells with subdiploid DNA requires 48 h. Besides apoptosis induc- tion, a substantial contribution of necrosis cannot be excluded, especially at higher concentration.

Compounds 3a and 3e significantly increased the caspase-3 activity, confirming the induction of programmed cell death.

Although caspase-independent cell death can manifest apoptotic morphology, and crucial caspases, including caspase-3, may be in- volved in non-lethal intracellular signaling, the assessment of exe- cutioner caspase activity remains an important part of apoptosis detection[38].

Since3aand3eexerted a substantial inhibition of DNA synthe- sis, comparable to that of the reference agent cisplatin, a further set of experiments was devoted to the determination of cycle-regulat- ing factors at the mRNA level by means of a RT-PCR technique.

The transition from the G1 to the S phase is tightly regulated by the expression of Rb, CDK4 and CDK6, and p16 factors, the latter regarded as crucially important[39]. Entry into the S phase, and hence cell proliferation, is inhibited as long as Rb remains unphos- phorylated by a complex containing cyclin D, CDK4 and CDK6.

Phosphorylated Rb dissociates from a heterodimeric complex of E2F, allowing the transcription of S-phase specific genes[40]. The importance of this cyclin D–CDK4–CDK6–p16–Rb–E2F pathway is indicated by the fact that it has been found to be altered in virtu- ally all human tumors[41]. An innovative agent intervening in this pathway may therefore be considered especially advantageous.

In view of our results, it seems conceivable that treatments with the two best compounds lead to the up-regulation of p16 (also re- ferred to as CDK4 inhibitor). The hypofunction of this protein has been associated with several malignancies and its expression cor- relates with the chemotherapy response in patients with solid tu- mors [42]. Since Rb is typically regulated postsynthetically by phosphorylation, its overall activity cannot be fully characterized by determining its expression at the mRNA level [40]. Our RT- PCR data were therefore supplemented with Western blot analy- ses, which indicated that the two most effective compounds have the capacity to decrease the expressions of Rb and pRb at a protein levels, and also the proportion of pRb. Since the expression of sig- nificant members of a pathway parallel to that detailed above, including CDK2, p21, p27 and p53, did not exhibit any appreciable treatment-dependent differences, the signal mechanism via pro- teins p16 and CDK4 governing the phosphorylation of Rb may be suggested as a predominant mechanism of action of the tested es- trone analogs.

In conclusion, our current results provide the first evidence that substituted estrone oximes may selectively suppress cancer cell proliferation by promoting apoptotic cell death and modulate the cell cycle progression. Although relatively high concentrations are needed to exert substantial activity, their cancer selectivity

seems to be more beneficial than that of the reference agent cis- platin. Accordingly, the estrone oxime skeleton is suggested as an appropriate scaffold for the design and development of novel antiproliferatve agents.

Acknowledgments

This publication is supported by the European Union and co- funded by the European Social Fund. Project number: TÁMOP- 4.2.2/B-10/1-2010-0012. The authors thank the Hungarian Scien- tific Research Fund (OTKA K 101659) for financial support.

Appendix A. Supplementary data

Supplementary data associated with this article can be found, in the online version, at http://dx.doi.org/10.1016/j.steroids.2012.

10.009.

References

[1] Ahlem CN, Page TM, Auci DL, Kennedy MR, Mangano K, Nicoletti F, et al. Novel components of the human metabolome: the identification, characterization and anti-inflammatory activity of two 5-androstene tetrols. Steroids 2011;76:145–55.

[2] Aperia A. New roles for an old enzyme: Na, K-ATPase emerges as an interesting drug target. J Intern Med 2007;261:44–52.

[3] Minorics R, Szekeres T, Krupitza G, Saiko P, Giessrigl B, Wölfling J, et al.

Antiproliferative effects of some novel synthetic solanidine analogs on HL-60 human leukemia cellsin vitro. Steroids 2011;76:156–62.

[4] Prokai-Tatrai K, Perjesi P, Rivera-Portalatin NM, Simpkins JW, Prokai L.

Mechanistic investigations on the antioxidant action of a neuroprotective estrogen derivative. Steroids 2008;73:280–8.

[5] Rocha M, Banuls C, Bellod L, Jover A, Victor VM, Hernandez-Mijares A. A review on the role of phytosterols: new insights into cardiovascular risk. Curr Pharm Des 2011;17:4061–75.

[6] Chiang CT, Way TD, Tsai SJ, Lin JK. Diosgenin, a naturally occurring steroid, suppresses fatty acid synthase expression in HER2-overexpressing breast cancer cells through modulating Akt, mTOR and JNK phosphorylation. FEBS Lett 2007;581:5735–42.

[7] Corbiere C, Battu S, Liagre B, Cardot PJ, Beneytout JL. SdFFF monitoring of cellular apoptosis induction by diosgenin and different inducers in the human 1547 osteosarcoma cell line. J Chromatogr B Analyt Technol Biomed Life Sci 2004;808:255–62.

[8] Corbiere C, Liagre B, Terro F, Beneytout JL. Induction of antiproliferative effect by diosgenin through activation of p53, release of apoptosis-inducing factor (AIF) and modulation of caspase-3 activity in different human cancer cells. Cell Res 2004;14:188–96.

[9] Leger DY, Liagre B, Corbiere C, Cook-Moreau J, Beneytout JL. Diosgenin induces cell cycle arrest and apoptosis in HEL cells with increase in intracellular calcium level, activation of cPLA2 and COX-2 overexpression. Int J Oncol 2004;25:555–62.

[10] Liu MJ, Yue PY, Wang Z, Wong RN. Methyl protodioscin induces G2/M arrest and apoptosis in K562 cells with the hyperpolarization of mitochondria.

Cancer Lett 2005;224:229–41.

[11] Raju J, Patlolla JM, Swamy MV, Rao CV. Diosgenin, a steroid saponin of Trigonella foenum graecum (Fenugreek), inhibits azoxymethane-induced aberrant crypt foci formation in F344 rats and induces apoptosis in HT-29 human colon cancer cells. Cancer Epidemiol Biomarkers Prev 2004;13:1392–8.

[12] Lee KR, Kozukue N, Han JS, Park JH, Chang EY, Baek EJ, et al. Glycoalkaloids and metabolites inhibit the growth of human colon (HT29) and liver (HepG2) cancer cells. J Agric Food Chem 2004;52:2832–9.

[13] Mueck AO, Seeger H. 2-Methoxyestradiol – Biology and mechanism of action.

Steroids 2010;75:625–31.

[14] Dahut WL, Lakhani NJ, Gulley JL, Arlen PM, Kohn EC, Kotz H, et al. Phase I clinical trial of oral 2-methoxyestradiol, an antiangiogenic and apoptotic agent, in patients with solid tumors. Cancer Biol Ther 2006;5:22–7.

[15] Li L, Bu S, Backstrom T, Landstrom M, Ulmsten U, Fu X. Induction of apoptosis and G2/M arrest by 2-methoxyestradiol in human cervical cancer HeLaS3 cells.

Anticancer Res 2004;24:873–80.

[16] Sweeney C, Liu G, Yiannoutsos C, Kolesar J, Horvath D, Staab MJ, et al. A phase II multicenter, randomized, double-blind, safety trial assessing the pharmacokinetics, pharmacodynamics, and efficacy of oral 2- methoxyestradiol capsules in hormone-refractory prostate cancer. Clin Cancer Res 2005;11:6625–33.

[17] Chang FS, Chen W, Wang C, Tzeng CC, Chen YL. Synthesis and antiproliferative evaluations of certain 2-phenylvinylquinoline (2-styrylquinoline) and 2- furanylvinylquinoline derivatives. Bioorg Med Chem 2010;18:124–33.

[18] Krstic NM, Bjelakovic MS, Zizak Z, Pavlovic MD, Juranic ZD, Pavlovic VD.

Synthesis of some steroidal oximes, lactams, thiolactams and their antitumor activities. Steroids 2007;72:406–14.

(10)

[19] Mosmann T. Rapid colorimetric assay for cellular growth and survival:

application to proliferation and cytotoxicity assays. J Immunol Methods 1983;65:55–63.

[20] Vermes I, Haanen C, Reutelingsperger C. Flow cytometry of apoptotic cell death. J Immunol Methods 2000;243:167–90.

[21] Ribble D, Goldstein NB, Norris DA, Shellman YG. A simple technique for quantifying apoptosis in 96-well plates. BMC Biotechnol 2005;5:12.

[22] Chomczynski P, Sacchi N. Single-step method of RNA isolation by acid guanidinium thiocyanate–phenol–chloroform extraction. Anal Biochem 1987;162:156–9.

[23] Gao Q, Tan J, Ma P, Ge J, Liu Y, Sun X, et al. PKC alpha affects cell cycle progression and proliferation in human RPE cells through the downregulation of p27kip1. Mol Vis 2009;15:2683–95.

[24] Matsuhashi N, Saio M, Matsuo A, Sugiyama Y, Saji S. Expression of p53 protein as a predictor of the response to 5-fluorouracil and cisplatin chemotherapy in human gastrointestinal cancer cell lines evaluated with apoptosis by use of thin layer collagen gel. Int J Oncol 2004;24:807–13.

[25] Huang Q, Cheung AP, Zhang Y, Huang HF, Auersperg N, Leung PC. Effects of growth differentiation factor 9 on cell cycle regulators and ERK42/44 in human granulosa cell proliferation. Am J Physiol Endocrinol Metab 2009;296:

E1344–53.

[26] Lee CH, Lim H, Moon S, Shin C, Kim S, Kim BJ, et al. Novel anticancer agent, benzyldihydroxyoctenone, isolated fromStreptomycessp. causes G1cell cycle arrest and induces apoptosis of HeLa cells. Cancer Sci 2007;98:795–802.

[27] Shiratori O. Growth inhibitory effect of cardiac glycosides and aglycones on neoplastic cells:in vitroandin vivostudies. Gann 1967;58:521–8.

[28] Stenkvist B. Is digitalis a therapy for breast carcinoma? Oncol Rep 1999;6:

493–6.

[29] Mijatovic T, Van Quaquebeke E, Delest B, Debeir O, Darro F, Kiss R. Cardiotonic steroids on the road to anti-cancer therapy. Biochim Biophys Acta 2007;1776:

32–57.

[30] Raju J, Bird RP. Diosgenin, a naturally occurring furostanol saponin suppresses 3-hydroxy-3-methylglutaryl CoA reductase expression and induces apoptosis in HCT-116 human colon carcinoma cells. Cancer Lett 2007;255:194–204.

[31] Poza J, Rega M, Paz V, Alonso B, Rodríguez J, Salvador N, et al. Synthesis and evaluation of new 6-hydroximinosteroid analogs as cytotoxic agents. Bioorg Med Chem 2007;15:4722–40.

[32] Huang Y, Cui J, Chen S, Gan C, Zhou A. Synthesis and antiproliferative activity of some steroidal lactams. Steroids 2011;76:1346–50.

[33] Gupta A, Saha P, Descoteaux C, Leblanc V, Asselin E, Berube G. Design, synthesis and biological evaluation of estradiol-chlorambucil hybrids as anticancer agents. Bioorg Med Chem Lett 2010;20:1614–8.

[34] Roy S, Reddy BS, Sudhakar G, Kumar JM, Banerjee R. 17b-estradiol-linked nitro-L-arginine as simultaneous inducer of apoptosis in melanoma and tumor-angiogenic vascular endothelial cells. Mol Pharm 2011;8:350–9.

[35] Frank É, Molnár J, Zupkó I, Kádár Z, Wölfling J. Synthesis of novel steroidal 17a- triazolyl derivatives via Cu(I)-catalyzed azide–alkyne cycloaddition, and an evaluation of their cytotoxic activityin vitro. Steroids 2011;76:1141–8.

[36] Saloranta T, Zupkó I, Rahkila J, Schneider G, Wölfling J, Leino R. Increasing the amphiphilicity of an estradiol based steroid structure by Barbier-allylation – ring-closing metathesis – dihydroxylation sequence. Steroids 2012;77:110–7.

[37] Tolomeo M, Simoni D. Drug resistance and apoptosis in cancer treatment:

development of new apoptosis-inducing agents active in drug resistant malignancies. Curr Med Chem Anticancer Agents 2002;2:387–401.

[38] Kepp O, Galluzzi L, Lipinski M, Yuan J, Kroemer G. Cell death assays for drug discovery. Nat Rev Drug Discovery 2011;10:221–37.

[39] Katsumi Y, Iehara T, Miyachi M, Yagyu S, Tsubai-Shimizu S, Kikuchi K, et al.

Sensitivity of malignant rhabdoid tumor cell lines to PD 0332991 is inversely correlated with p16 expression. Biochem Biophys Res Commun 2011;413:

62–8.

[40] Sunley K, Butler M. Strategies for the enhancement of recombinant protein production from mammalian cells by growth arrest. Biotechnol Adv 2010;28:385–94.

[41] Nevins JR. The Rb/E2F pathway and cancer. Hum Mol Genet 2001;10:699–703.

[42] Borys D, Canter RJ, Hoch B, Martinez SR, Tamurian RM, Murphy B, et al. P16 expression predicts necrotic response among patients with osteosarcoma receiving neoadjuvant chemotherapy. Hum Pathol.http://dx.doi.org/10.1016/

j.humpath.2012.02.003.

78 Á. Berényi et al. / Steroids 78 (2013) 69–78

Ábra

Fig. 3. Effects of compounds 3a, 3e, 10h and 11a on the HeLa cell cycle distribution after incubation for 24 (panel A) or 48 h (panel B)
Fig. 5. Induction of caspase-3 activity after incubation with compounds 3a and 3e for 48 h

Hivatkozások

KAPCSOLÓDÓ DOKUMENTUMOK

The antiproliferative activities were determined in vitro by means of MTT assays against human adherent cervical (SiHa, HeLa) and breast cancer (MCF-7 and

Antiproliferative activity of naringenin (1) and the prepared oximes (2, 3) and oxime ethers (4–8) was determined in vitro against five human cancer cell lines, including

Antiproliferative activity of naringenin (1) and the prepared oximes (2, 3) and oxime ethers (4–8) was determined in vitro against five human cancer cell lines, including

Synthesis and evaluation of fluorescently labelled GnRH analogues were aimed to the prediction of GnRH-R targeted therapeutic efficiency on cancer cell models in

Antitumor effects and impact on RAS-related signalization of zoledronic acid (ZA) and a lipophilic bisphosphonate (BPH1222) were investigated on 7 human colorectal cancer cell lines

The in vitro cell viability inhibition was tested on Detroit-562 human pharyngeal carcinoma cells that express GnRH-R in high levels, and the results showed that all conjugates

All of the synthesized compounds were primarily screened in vitro for their antiproliferative activity on five different cancer cell lines, namely DU 145, PC-3, HeLa, MCF-7

Compounds 1–6, the isolated flavonoids and scopoletin were evaluated for their antiproliferative activities on human gynecological cancer cell lines (SiHa, HeLa, and MDA-MB-231