Next Article in Journal
Carbon-Filled Organic Phase-Change Materials for Thermal Energy Storage: A Review
Next Article in Special Issue
Design, Synthesis and Biological Evaluation of a New Series of 1-Aryl-3-{4-[(pyridin-2-ylmethyl)thio]phenyl}urea Derivatives as Antiproliferative Agents
Previous Article in Journal
Compositional Analysis and Aroma Evaluation of Feijoa Essential Oils from New Zealand Grown Cultivars
Previous Article in Special Issue
Synthesis, Biological Evaluation, and In Silico Studies of Novel Aminated Xanthones as Potential p53-Activating Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Potent Anti-Ovarian Cancer with Inhibitor Activities on Both Topoisomerase II and V600EBRAF of Synthesized Substituted Estrone Candidates

by
Mohamed El-Naggar
1,
Abd El-Galil E. Amr
2,3,*,
Ahmed A. Fayed
3,4,
Elsayed A. Elsayed
5,6,
Mohamed A. Al-Omar
2 and
Mohamed M. Abdalla
7
1
Chemistry Department, Faculty of Sciences, University of Sharjah, Sharjah 27272, UAE
2
Drug Exploration & Development Chair (DEDC), Pharmaceutical Chemistry Department, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
3
Applied Organic Chemistry Department, National Research Center, Cairo 12622, Egypt
4
Respiratory Therapy Department, College of Medical Rehabilitation Sciences, Taibah University, Madinah Munawara 22624, Saudi Arabia
5
Zoology Department, Bioproducts Research Chair, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
6
Chemistry of Natural and Microbial Products Department, National Research Centre, Cairo 12622, Egypt
7
Atos Pharma, Elkatyba Land, Belbis 44621, El Sharkya, Egypt
*
Author to whom correspondence should be addressed.
Molecules 2019, 24(11), 2054; https://doi.org/10.3390/molecules24112054
Submission received: 25 April 2019 / Revised: 23 May 2019 / Accepted: 28 May 2019 / Published: 29 May 2019
(This article belongs to the Special Issue Anticancer Agents: Design, Synthesis and Evaluation)

Abstract

:
A series of 16-(α-alkoxyalkane)-17-hydrazino-estra-1(10),2,4-trien[17,16-c]-3-ol (3al) and estra-1(10),2,4-trien-[17,16-c]pyrazoline-3-ol derivatives (4ad) were synthesized from corresponding arylidines 2a,b which was prepared from estrone 1 as starting material. Condensation of 1 with aldehydes gave the corresponding arylidine derivatives 2a,b which were treated with hydrazine derivatives in alcohols to give the corresponding derivatives 3al, respectively. Additionally, treatment of 2a,b with methyl- or phenylhydrazine in ethanolic potassium hydroxide afforded the corresponding N-substituted pyrazoline derivatives 4ad, respectively. All these derivatives showed potent anti-ovarian cancer both in vitro and in vivo. The mechanism of anti-ovarian cancer was suggested to process via topoisomerase II and V600EBRAF inhibition.

1. Introduction

Synthetic alterations of estrone lead to discovering of compounds with diverse biological activities, for example with antitumor effect [1], as anti-breast cancer agent [2] and with antioxidant activity [3]. Estrone derivatives with antitumor activities must be devoid completely of the estrogenic activities [4,5,6]. The inversion of the configuration at C-13 lead to estrone derivatives with antitumor activities devoid from hormonal actions due to conformational change for the overall molecule resulting from the cis junction of rings C and D [4,7]. Some recent publications report on the syntheses and in vitro biological evaluation of several 13α-estrone derivatives [8,9,10,11]. These derivatives exhibited biological activities with substantial anti-proliferative or enzyme inhibitory potentials. Most literature data are mainly focusing on 13α-estrone substitution in ring D, while modified derivatives with ring A substitutions are rarely reported [12].
Cancer is considered one of the most significant causes of death worldwide. Ovarian cancer is one of the major causes for death among adult females [13]. Cancer is characterized by abnormal cell proliferation bypassing normal cell growth and death mechanisms. Recently, DNA topoisomerase II inhibitors have been researched as a potential target, which interferes with cancer growth and development [14]. Topoisomerase II inhibition leads finally to cell cycle inhibition and apoptosis in abnormal cancer cells [15]. BRAF kinase inhibition is another possible control mechanism, which was also developed to interfere with cancer cell growth [16]. BRAF kinase constitutes a major signaling process integrated in the RAS-MEK proliferation process. The activation of MEK by BRAF signals finally induces cell proliferation and survival [17]. Inhibiting BRAF activity results in inhibition of the proliferation signals, and thus inhibiting cancer cell development and growth [18].
In view of these observations and in continuation of our previous work in heterocyclic chemistry [19,20,21,22], the current work aimed at evaluating the effect or remote long cage distortion caused by introducing some moieties in both 16 and 17 locations of ring D. We screened some of synthesized estrone candidates for their anti-ovarian cancer potential both in vitro and in vivo. Furthermore, the inhibitory effects of the synthesized compounds were investigated against topoisomerase II and V600EBRAF kinase inhibitors.

2. Results and Discussion

2.1. Results

2.1.1. Chenical Synthesis

A series of 17-hydrazino-estratrienol (3a–l) and pyrazolinestratrienol (4a–d) were synthesized from corresponding arylidines 2a,b which was prepared from estrone 1 as starting material.
Treatment of aryledine derivatives 2a,b with hydrazine hydrate, in refluxing methanol or ethanol afforded the corresponding 17-hydrazino-16-α-methoxymethane estrone derivatives 3a,b and 17-hydrazino-16-α-ethoxyethane estrone derivatives 3c,d, respectively.
Also, reaction of compounds 2a,b with N-methyl hydrazine, in refluxing methanol or ethanol afford 17-N-methyl hydrazino-16-α-methoxymethane estrone derivatives 3e,f and 17-N-methyl hydrazino-16-α-ethoxyethane estrone derivatives 3g,h respectively.
Additionally, when compounds 2a,b reacted with N-phenyl hydrazine, under the same conditions afford the corresponding 17-N-phenylhydrazino- 16-α-methoxymethane estrone derivatives 3i,j and 17-N-phenylhydrazino- 16-α-ethoxyethane estrone derivatives 3k,l, respectively. Finally, condensation of 2a,b with CH3NHNH2 or PhNHNH2 in refluxing dioxane afforded the corresponding N-substituted pyrazoline derivatives 4a–d, respectively (Scheme 1).

2.1.2. Biological Screening

In Vitro Cytotoxic Activities

The cytotoxicity of some synthesized derivatives against SKOV-3 cancer cell line in vitro was performed with the MTT assay. The obtained results showed that all synthesized compounds showed potent in vitro cytotoxic activities against SKOV-3 cells at the nanomolar level. Moreover, the effect of the synthesized compounds was dose-dependent. From the obtained IC50 values (Figure 1), prepared compounds can be arranged discerningly in the following order: 3a, 3b, 3c, 3d, 3e, 3f, 3g, 3h, 3i, 3j, 3k, 3l, 4a, 4b, 4c, 4d (Figure 1). Compounds 3a–l are characterized with the presence of hydrazine group rather than the pyrazoline group present in compounds 4ad. Furthermore, the alkyl group is found to be more active than the aromatic group. Generally Cpd. 3a showed highest cytotoxic activity with IC50 value of 4.23 ± 0.12 nM, while Cpd. 4d was the least active, where its IC50 value was about 9.5% of that obtained for Cpd. 3a.
The effect of resveratrol (RES) and doxorubicin (DOX) on SKOV-3 showed IC50 of 55 and 250 nM, respectively.
The cytotoxicity of the newly synthesized compounds against fibroblast were as follow IC50 3a (1.2 µM), 3b (2.45 µM), 3c (5.67 µM), 3d (7.44 µM), 3e (8.11 µM), 3f (9.20 µM), 3g (11.18 µM), 3h(16.22 µM), 3i (15.13 µM), 3j (16.20 µM), 3k (17.78 µM), 3l (20.21 µM), 4a (23.34 µM), 4b (24.55 µM), 4c (25.55 µM), 4d (30.34 µM).

In Vivo Anti-Ovarian Cancer

The in vivo anti-ovarian cancer activities of the tested compounds were evaluated using mouse xenograft model. Figure 2 represents the percentages of decrease in tumor growth upon treatment with different synthesized compounds. The obtained results showed that all compounds potentially reduced tumor growth over a period of 40 days of treatment, in comparison to control treatments, which showed normal tumor growth development. Moreover, it can be seen that the reduction in tumor size started directly from the 5th day of treatment, where the reduction percentage ranged from 45.7 ± 0.05% to 54.71 ± 0.01%. The reduction in tumor growth increased significantly reaching its maximal by the end of treatment period (40 days). The highest reduction in tumor growth was recorded for occurred Cpd. 3a, which significantly decreased tumor growth by about 93.61 ± 0.7%. It can also be seen that Cpd. 3a, was the most potent for in vivo treatment, where its effect was more obvious starting from day15 of treatment, where the reduction percentage in tumor size due to other compounds was more or less within the same range. We also can see that the in vivo results are coincided with those obtained for in vitro results, where the reduction in tumor size followed the same descending order: 3a, 3b, 3c, 3d, 3e, 3f, 3g, 3h, 3i, 3j, 3k, 3l, 4a, 4b, 4c, and 4d.

2.1.3. Inhibition of Topoisomerase II Activities

In order to further investigate the possible mechanism of action of the newly synthesized derivatives, we investigated their inhibitory effects on topoisomerase II. Results obtained (Figure 3) showed that all synthesized compounds showed potential inhibitory effects against topoisomerase II. Furthermore, the inhibitory effect was also found to follow the same descending order obtained before during in vitro and in vivo investigation: 3a, 3b, 3c, 3d, 3e, 3f, 3g, 3h, 3i, 3j, 3k, 3l, 4a, 4b, 4c, and 4d. Cpd. 3a showed the most inhibitory action for topoisomerase II with IC50 value of 3.45 ± 0.13 nM, which was about 7,9% of that obtained for the lest potent compound (IC50 for Cpd. 4d: 43.56 ± 0.98 nM).

2.1.4. In Vitro Kinase Assay

The in vitro kinase assay of synthesized derivatives was investigated against both WTBRAF (BRAF kinase wild type) and V600EBRAF (mutant BRAF kinase). Results obtained in Figure 4 showed that all synthesized compounds were highly active inhibitors for V600EBRAF compared with moderate activities against WTBRAF. Again, the descending order of activities was as follow 3a, 3b, 3c, 3d, 3e, 3f, 3g, 3h, 3i, 3j, 3k, 3l, 4a, 4b, 4c, and 4d. Compound 3a recorded the most active inhibition (IC50: 0.041 ± 0.0016 and 4.23 ± 0.12 μM for mutant and wild type BRAF kinase, respectively. Furthermore, it can be seen that the inhibitory effect of Cpd. 3a was much more superior to that obtained for different positive control drugs (0.48, 3.87 and 0.97 μM for Sorafenib, Dabrafenib and Vemurafenib, respectively.

2.2. Discussion

Within the framework of the current work we synthesized new estrone derivatives from their corresponding arylidines. The newly synthesized derivatives showed potential cytotoxic activities against SKOV-3 cells. Moreover, in vivo investigations revealed that the synthesized compounds were able to potentially reduce tumor volume growth over a treatment period of 45 days.
The newly introduced 1-alkoxy benzyl moiety alone or combined with the 17- hydrazino- moities make remote destortion of the structure cage that completely elminate any hormonal activities of te estrone molceules and deviate the biological activities towards the aniticancer ones, so we investigated this property and tried to find the mechnisms of anticancer actions of these newly synthesized compounds.
In search for the possible mechanism of action of the anticancer activities of the tested compounds, we investigated the possibility of the compounds to inhibit both topoisomerase II and kinase enzymes. Results showed that the prepared compounds can potentially serve as inhibitors for these enzymes. The cytotoxic activities of the prepared estrone derivatives can be due to the inhibition of 17-hydroxysteroid dehydrogenases. We have previously reported on the anti-breast cancer activities of different estrone derivatives [23], and explained that their anticancer activities can be attributed to the inhibition of 17-hydroxysteroid dehydrogenase [24].
Concerning topoisomerase II and V600EBRAF kinase inhibition, our results suggest that the newly synthesized compounds exert their cytotoxic action against SKOV-3 cancer cells by interfering with the metabolic activity of these enzymes, thus preventing cancer cells from obtaining proliferation signaling molecules essential for their growth and survival [16,17]. Different activities of the prepared compounds may be attributed to the structure activity relationship of these compounds.
Hydrazine derivatives are generally potent than pyrazoline ones, due to their open chain structure as well as their higher electron density. Furthermore, it can be suggested that N-methyl derivatives are more active than N-phenyl ones. On the other hand, hydrazine derivatives can be divided into more active α-methoxyl derivatives and the less active α-ethoxyl, and then the least active N-phenyl derivatives. This can be explained due to the fact that phenyl group act as a withdrawing group. On the other hand, the weaker effect of α-ethoxyl derivatives than the α-methoxyl ones can be attributed to their bulky nature. This effect can be observed among different series of compound 3. As the bulky nature of the substituted groups increase, accordingly more steric hindrance can be observed, leading to decreased cytotoxic activities.

3. Materials and Methods

3.1. Chemical Synthesis

“Melting points reported are inaccurate. IR spectra were registered on Shimadzu FT-IR 8300 E (Shimadzu Corporation, Kyoto, Japan) spectrophotometer using the (KBr) disk technique. The 1H-NMR spectra were determined with bruker 600 MHz NMR spectrometer. The chemical shifts are expressed on the δ (ppm) scale using TMS as the standard reference. Mass spectra were recorded on Finnigan SSQ operating at 70 ev. Elemental analysis determined on a Perkin Elmer 240 (Microanalysis), Microanalysis Center, Cairo University, Cairo, Egypt”.

3.1.1. Synthesis of 3-Hydroxy-16-[substituted]-estra-1(10),2,4-trien-17-ones (2a,b)

“A mixture of (1) (0.54 g, 20 mmol) and formaldehyde or acetaldehyde (20 mmol) in EtOH (50 mL) and aqueous KOH (10 mL, 30%) was stirred for 24 h at room temperature. The formed solid was collected by filtration, crystallized from ethanol to give compounds 2a [25] and 2b, respectively”.

3-Hydroxy-16-[ethylene]-estra-1(10),2,4-trien-17-one (2b)

Yield 97%, mp 198–200 °C, [ α ] D 25 = + 71 (c 1, MeOH). IR (KBr, cm–1): 3340 (OH), 2944 (CH, aliphatic), 1743 (C=O), 1642 (C=C). 1H NMR: (600 MHz, CDCl3): δ ppm 0.61 (1H, m, H-8β), 0.90 (3H, s, CH3-18), 1.01 (1H, m, H-11β), 1.12 (1H, m, H-7α), 1.15 (1H, m, H-12α), 1.25 (1H, m, H-14α), 1.44 (1H, m, H-15β), 1.63 (1H, m, H-15α), 1.72 (1H, m, H-7β), 2.01 (1H, m, H-9α), 2.13 (1H, m, H-11α), 2.50 (1H, m, H-12β), 2.56 (1H, m, H-6α), 2.66 (1H, m, H-6β), 4.90 (1H, s, OH, disappeared with D2O), 5.75 (1H, dd, H-2), 6.67 (1H, d, H-4), 2.08 (3H, d, CH3, enone), 6.41 (1H, s, enone proton), 7.12 (1H, d, H-1). 13C NMR: (150 MHz, CDCl3): δ 13.52, 21.73, 25.51, 26.64, 27.48, 29.75, 36.35, 38.61, 43.81, 48.92, 50.43, 112.79, 115.53, 126.56, 128.48, 132.50, 137.02, 138.44, 153.82, 210.02 (20 C). MS (EI): m/z 296 (100%) [M+]. Anal. Calcd for C20H24O2 (296.40): Calcd C, 81.04; H, 8.16. Found C, 81.00; H, 8.10.

3.1.2. Synthesis of 16-(α-alkoxy-alkane)-17-hydrazino-estra-1(10),2,4-trien[17,16-c]-3-ol and their N-substituted derivatives (3a–l)

“A mixture of 2a,b (14 mmol) and hydrazine derivatives, namely, hydrazine hydrate, methyl hydrazine or phenyl hydrazine (8 mmol) in absolute methanol or ethanol (30 mL) was refluxed for 5–10 h. The solvent was concentrated under reduced pressure, the formed precipitate was filtered off, washed with water, dried and crystallized from ethanol to give the corresponding derivatives 3a–l, respectively”.

16-(α-Methoxy-methane)-17-hydrazino-estra-1(10),2,4-trien-[17,16-c]-3-ol (3a)

Yield 80%, mp 199–201 °C, [ α ] D 25 = + 81 (c 1, MeOH). IR (KBr, cm–1): 3444–3377 (NH, NH2), 3345 (OH), 2951 (CH, aliphatic), 1638 (C=C). 1H NMR: (600 MHz, CDCl3): δ ppm 0.61 (1H, m, H-8β), 0.90 (3H, s, CH3-18), 1.00 (1H, m, H-11β), 1.11 (1H, m, H-7α), 1.14 (1H, m, H-12α), 1.26 (1H, m, H-14 α), 1.39 (1H, m, H-15β), 1.58 (1H, m, H-15α), 1.68 (1H, m, H-7β), 2.01(1H, m, H-9α), 1.84 (1H, m, CH, 16-Hα), 2.11 (1H, m, H-11α), 2.49 (1H, m, H-12β), 2.61 (1H, m, H-6α), 2.66 (1H, m, H-6β), 2.83 (1H, d, H-17), 3.48 (3H, s, OCH3), 4.65 (2H, m, NH2, disappeared with D2O), 4.80 (2H, d, CH2-O), 4.87 (1H, s, OH, disappeared with D2O), 5.70 (1H, dd, H-2), 6.61 (1H, d, H-4), 7.10 (1H, d, H-1), 7.70 (1H, br.s, NH, disappeared with D2O). 13C NMR: (150 MHz, CDCl3): δ 13.58, 21.76, 25.57, 26.76, 28.00, 29.78, 36.38, 38.65, 43.89, 48.40, 50.45, 59.45, 69.11, 112.37, 115.56, 126.53, 132.81, 138.46, 153.84, 167.02 (20 C). MS (EI): m/z 330 (90%) [M+]. Anal. Calcd for C20H30N2O2 (330.46): Calcd C, 72.69; H, 9.15; N, 8.48. Found C, 72.60; H, 9.10; N, 8.40.

16-(α-Methoxy-ethane)-17-hydrazino-estra-1(10),2,4-trien[17,16-c]-3-ol (3b)

Yield 57%, mp. 308–310 °C, [ α ] D 25 = + 112 (c 1, MeOH). IR (KBr, cm–1): 3448-3377 (NH, NH2), 3348 (OH), 2959 (CH, aliphatic), 1634 (C=C). 1H NMR: (600 MHz, CDCl3): δ ppm 0.59 (1H, m, H-8β), 0.91 (3H, s, CH3-18), 1.01 (1H, m, H-11β), 1.12 (1H, m, H-7α), 1.14 (1H, m, H-12 α), 1.26 (1H, m, H-14 α), 1.39 (1H, m, H -15 β), 1.58 (1H, m, H-15α), 1.42 (3H, m, CH3,C-16 ethane), 1.69 (1H, m, H-7β), 2.02(1H, m, H-9α), 1.83 (1H, m, CH, H-16α), 2.11 (1H, m, H-11α), 2.49 (1H, m, H-12β), 2.61 (1H, m, H-6α), 2.66 (1H, m, H-6β), 2.83 (1H, d, H-17), 3.48 (3H, s, OCH3), 4.65 (2H, s, NH2, disappeared with D2O), 4.84 (1H, d, CH-O), 4.99 (1H, s, OH, disappeared with D2O), 5.72 (1H, dd, H-2), 6.61 (1H, d, H-4), 7.11 (1H, d, H-1), 7.78 (1H, bs, NH, disappeared with D2O). 13C NMR: (150 MHz, CDCl3): δ 13.58, 21.77, 22.22, 25.57, 26.47, 28.00, 29.78, 36.68, 38.47, 43.89, 48.69, 50.45, 59.45, 69.11, 112.37, 115.56, 126.55, 132.48, 138.46, 153.54, 167.76 (21 C). MS (EI): m/z 344 (11%) [M+]. Anal. Calcd for C21H32N2O2 (344.49): Calcd C, 73.22; H, 9.36; N, 8.13. Found C, 73.18; H, 9.30; N, 8.10.

16-(α-Ethoxy-methane)-17-hydrazino-estra-1(10),2,4-trien-[17,16-c]-3-ol (3c)

Yield 56%, mp 219–221 °C, [ α ] D 25 = + 77 (c 1, MeOH). IR (KBr, cm–1): 3445–3376 (NH, NH2), 3346 (OH), 2951 (CH, aliphatic), 1638 (C=C). 1H NMR: (600 MHz, CDCl3): δ ppm 0.61 (1H, m, H-8β), 0.90 (3H, s, CH3-18), 1.00 (1H, m, H-11β), 1.11 (1H, m, H-7α), 1.15 (1H, m, H-12α), 1.21 (3H, m, CH3, ethoxyl), 1.25 (1H, m, H-14α), 1.36 (1H, m, H-15β), 1.57 (1H, m, H-15α), 1.68 (1H, m, H-7β), 2.01 (1H, m, H-9α), 1.84 (1H, m, CH, H-16α), 2.10 (1H, m, H-11α), 2.50 (1H, m, H-12β), 2.60 (1H, m, H-6α), 2.70 (1H, m, H-6β), 6.61 (1H, d, H-4), 2.80 (1H, d, H-17), 3.50 (2H, m, OCH2), 4.70 (2H, m, NH2, disappeared with D2O), 4.80 (2H, d, CH-O), 4.87 (1H, s, OH, disappeared with D2O), 5.70 (1H, dd, H-2), 7.10 (1H, d, H-1), 7.70 (1H, br.s, NH, disappeared with D2O). 13C NMR: (150 MHz, CDCl3): δ 13.65, 18.74, 21.57, 25.17, 26.38, 28.04, 29.64, 36.54, 38.15, 44.11, 48.44, 50.54, 68.45, 69.11, 112.44, 115.67, 126.34, 131.11, 138.46, 153.84, 161.47 (21 C). MS (EI): m/z 344 (35%) [M+]. Anal. Calcd for C21H32N2O2 (344.49): Calcd C, 73.22; H, 9.36; N, 8.13. Found C, 73.16; H, 9.30; N, 8.10.

16-(α-Ethoxy-ethane)-17-hydrazino-estra-1(10),2,4-trien-[17,16-c]-3-ol (3d)

Yield 90%, mp 223–225 °C, [ α ] D 25 = + 95 (c 1, MeOH). IR (KBr, cm–1): 3440-3370 (NH, NH2), 3350 (OH), 2951 (CH, aliphatic), 1631 (C=C). 1H NMR: (600 MHz, CDCl3): δ ppm 0.57 (1H, m, H-8β), 0.90 (3H, s, CH3-18), 1.00 (1H, m, H-11β), 1.10 (1H, m, H-7α), 1.15 (1H, m, H-12α), 1.20 (3H, m, CH3, ethoxyl), 1.24 (1H, m, H-14α), 1.36 (1H, m, H-15β), 1.45 (3H, m, CH3,C-16 ethane), 1.59 (1H, m, H-15α), 1.71 (1H, m, H-7β), 1.96 (1H, m, H-9α), 1.82 (1H, m, CH, H-16α), 2.10 (1H, m, H-11α), 2.50 (1H, m, H-12β), 2.62 (1H, m, H-6α), 2.67 (1H, m, H-6β), 2.86 (1H, d, H-17), 3.48 (2H, m, OCH2), 4.64 (2H, s, NH2, disappeared with D2O), 4.83 (1H, d, CH-O), 4.88 (1H, s, OH, disappeared with D2O), 5.74 (1H, dd, H-2), 6.65 (1H, d, H-4), 7.12 (1H, d, H-1), 7.78 (1H, br.s, NH, disappeared with D2O). 13C NMR: (150 MHz, CDCl3): δ 13.55, 18.79, 22.07, 22.28, 25.47, 26.52, 27.77, 28.09, 36.68, 37.97, 44.09, 48.11, 51.41, 68.43, 69.13, 112.56, 115.57, 126.37, 133.18, 137.87, 155.56, 162.74 (22 C). MS (EI): m/z 358 (45%) [M+]. Anal. Calcd for C22H34N2O2 (358.51): Calcd C, 73.70; H, 9.56; N, 7.81. Found C, 73.64; H, 9.52; N, 7.75.

16-(α-Methoxy-methane)-17-[N-methyl-hydrazino]estra-1(10),2,4-trien[17,16-c]-3-ol (3e)

Yield 76%, mp 222–224 °C, [ α ] D 25 = + 66 (c 1, MeOH). IR (KBr, cm–1): 3443–3371 (NH), 3344 (OH), 2953 (CH, aliphatic), 1634 (C=C). 1H NMR: (600 MHz, CDCl3): δ ppm 0.61 (1H, m, H-8β), 0.92 (3H, s, CH3-18), 1.03 (1H, m, H-11β), 1.10 (1H, m, H-7α), 1.14 (1H, m, H-12α), 1.25 (1H, m, H-14α), 1.37 (1H, m, H-15β), 1.57 (1H, m, H-15α), 1.66 (1H, m, H-7β), 2.00 (1H, m, H-9α), 1.84 (1H, m, CH, H-16α), 2.13 (1H, m, H-11α), 2.32 (3H, s, NCH3), 2.48 (1H, m, H-12β), 2.63 (1H, m, H-6α), 2.68 (1H, m, H-6β), 2.84 (1H, d, H-17), 3.49 (3H, s, OCH3), 4.86 (2H, d, CH-O), 4.87 (1H, s, OH, disappeared with D2O), 5.72 (1H, dd, H-2), 6.63 (1H, d, H-4), 7.11 (1H, d, H-1), 7.76 (1H, brs, NH, disappeared with D2O), 7.99 (1H, bs, NH, disappeared with D2O). 13C NMR: (150 MHz, CDCl3): δ 13.53, 21.44, 25.67, 26.36, 28.03, 29.23, 36.33, 38.43, 43.83, 47.47, 48.43, 50.34, 59.35, 69.31, 112.36, 115.67, 126.52, 132.32, 138.21, 153.45, 162.32 (21 C). MS (EI): m/z 344 (45%) [M+]. Anal. Calcd for C21H32N2O2 (344.50): Calcd C, 73.22; H, 9.36; N, 8.13. Found C, 73.15; H, 9.31; N, 8.09.

16-(α-Methoxy-ethane)-17-[N-methyl-hydrazino]-estra-1(10),2,4-trien-[17,16-c]-3-ol (3f)

Yield 57%, mp 310–312 °C, [ α ] D 25 = + 112 (c 1, MeOH). IR (KBr, cm–1): 3443–3374 (NH), 3344 (OH), 2955 (CH, aliphatic), 1628 (C=C). 1H NMR: (600 MHz, CDCl3): δ ppm 0.58 (1H, m, H-8β), 0.90 (3H, s, CH3-18), 1.00 (1H, m, H-11β), 1.10 (1H, m, H-7α), 1.14 (1H, m, H-12α), 1.26 (1H, m, H-14 α), 1.39 (1H, m, H-15 β), 1.60 (1H, m, H-15α), 1.44 (3H, m, CH3, C-16 ethane), 1.70 (1H, m, H-7β), 2.00 (1H, m, H-9α), 1.82 (1H, m, CH, H-16α), 2.11 (1H, m, H-11α), 2.36 (3H, s, NCH3), 2.51 (1H, m, H-12β), 2.62 (1H, m, H-6α), 2.65 (1H, m, H-6β), 2.85 (1H, d, H-17), 3.44 (3H, s, OCH3), 4.83 (1H, d, CH-O), 4.99 (1H, s, OH, disappeared with D2O), 5.74 (1H, dd, H-2), 6.65 (1H, d, H-4), 7.13 (1H, d, H-1), 7.73 (1H, br.s, NH, disappeared with D2O), 7.99 (1H, m, NH, disappeared with D2O). 13C NMR: (150 MHz, CDCl3): δ 13.78, 21.54, 22.27, 25.97, 26.69, 28.03, 29.43, 36.45, 38.54, 43.68, 47.88, 48.57, 50.86, 59.46, 69.15, 112.54, 115.69, 126.46, 132.90, 138.75, 153.45, 163.60 (22 C). MS (EI): m/z 358 (100%) [M+]. Anal. Calcd for C22H34N2O2 (358.50): Calcd C, 73.70; H, 9.56; N, 7.81. Found C, 73.60; H, 9.49; N, 7.75.

16-(α-Ethoxy-methane)-17-[N-methyl-hydrazino]-estra-1(10),2,4-trien-[17,16-c]-3-ol (3g)

Yield 90%, mp 286–288 °C, [ α ] D 25 = + 121 (c 1, MeOH). IR (KBr, cm–1): 3441–3379 (NH), 3348 (OH), 2957 (CH, aliphatic), 1636 (C=C). 1H NMR: (600 MHz, CDCl3): δ ppm 0.61 (1H, m, H-8β), 0.91 (3H, s, CH3-18), 1.01 (1H, m, H-11β), 1.13 (1H, m, H-7α), 1.15 (1H, m, H-12α), 1.22 (3H, m, CH3, ethoxyl), 1.25 (1H, m, H-14α), 1.37 (1H, m, H-15β), 1.58 (1H, m, H-15α), 1.69 (1H, m, H-7β), 2.00 (1H, m, H-9 α), 1.81 (1H, m, CH, H-16α), 2.12 (1H, m, H-11α), 2.33 (3H, s, NCH3), 2.54 (1H, m, H-12β), 2.65 (1H, m, H-6α), 2.76 (1H, m, H-6β), 2.87 (1H, d, H-17), 3.50 (2H, m, OCH2), 4.85 (2H, d, CH2-O), 4.98 (1H, s, OH, disappeared with D2O), 5.74 (1H, dd, H-2), 6.65 (1H, d, H-4), 7.10 (1H, d, H-1), 7.76 (1H, br.s, NH, disappeared with D2O), 7.95 (1H, m, NH, disappeared with D2O). 13C NMR: (150 MHz, CDCl3): δ 13.35, 18.45, 21.43, 25.78, 26.80, 28.14, 29.88, 36.47, 38.90, 44.41, 47.37, 48.34, 50.54, 68.66, 69.67, 113.34, 115.55, 125.54, 131.53, 137.99, 154.44, 162.27 (22 C). MS (EI): m/z 358 (100%) [M+]. Anal. Calcd for C22H34N2O2 (358.50): Calcd C, 73.70; H, 9.56; N, 7.81. Found C, 73.62; H, 9.50; N, 7.74.

16-(α-Ethoxy-ethane)-17-[N-methyl-hydrazino]-estra-1(10),2,4-trien-[17,16-c]-3-ol (3h)

Yield 78%, mp 280–282 °C, [ α ] D 25 = + 135 (c 1, MeOH). IR (KBr, cm–1): 3441–3371 (NH), 3351 (OH), 2951 (CH, aliphatic), 1631 (C=C). 1H NMR: (600 MHz, CDCl3): δ ppm 0.60 (1H, m, H-8β), 0.93 (3H, s, CH3-18), 1.04 (1H, m, H-11β), 1.11 (1H, m, H-7α), 1.16 (1H, m, H-12α), 1.19 (3H, m, CH3,ethoxyl), 1.23 (1H, m, H-14α), 1.37 (1H, m, H-15β), 1.46 (3H, m, CH3,C-16 ethane), 1.61 (1H, m, H-15α), 1.72 (1H, m, H-7β), 1.82 (1H, m, CH, H-16α), 1.93 (1H, m, H-9α), 2.10 (1H, m, H-11α), 2.50 (1H, m, H-12β), 2.34 (3H, s, NCH3), 2.63 (1H, m, H-6α), 2.62 (1H, m, H-6β), 2.85 (1H, d, H-17), 3.43 (2H, m, OCH2), 4.64 (1H, s, OH, disappeared with D2O), 4.83 (1H, d, CH-O), 5.72 (1H, dd, H-2), 6.64 (1H, d, H-4), 7.10 (1H, d, H-1), 7.75 (1H, br.s, NH, disappeared with D2O), 7.88 (1H, s, NH, disappeared with D2O). 13C NMR: (150 MHz, CDCl3): δ 13.53, 18.67, 22.43, 22.90, 25.67, 26.78, 27.45, 28.29, 36.89, 37.65, 44.65, 47.35, 48.68, 51.90, 68.78, 69.13, 112.56, 115.56, 126.34, 133.55, 137.43, 155.34, 163.34 (23 C). MS (EI): m/z 372 (100%) [M+]. Anal. Calcd for C23H36N2O2 (372.54): Calcd C, 74.15; H, 9.74; N, 7.52. Found C, 74.05; H, 9.68; N, 7.48.

16-(α-Methoxy-methane)-17-[N-phenyl-hydrazino]-estra-1(10),2,4-trien-[17,16-c]-3-ol (3i)

Yield 67%, mp 312–314 °C, [ α ] D 25 = + 95 (c 1, MeOH). IR (KBr, cm–1): 3430–3367 (NH), 3345 (OH), 3068 (CH, aromatic), 2951 (CH, aliphatic), 1638 (C=C). 1H NMR: (600 MHz, CDCl3): δ ppm 0.61(1H, m, H-8β), 0.91 (3H, s, CH3-18), 1.00 (1H, m, H-11β), 1.12 (1H, m, H-7α), 1.15 (1H, m, H-12α), 1.28 (1H, m, H-14α), 1.40 (1H, m, H-15β), 1.60 (1H, m, H-15α), 1.68 (1H, m, H-7β), 2.02 (1H, m, H-9α), 1.86 (1H, m, CH, H-16α), 2.12 (1H, m, H-11α), 2.49 (1H, m, H-12 β), 2.59 (1H, m, H-6α), 2.65 (1H, m, H-6β), 2.84 (1H, d, H-17), 3.47 (3H, s, OCH3), 4.80 (2H, d, CH-O), 4.87 (1H, s, OH, disappeared with D2O), 5.70 (1H, dd, H-2), 6.61 (1H, d, H-4), 7.10 (1H, d, H-1), 7.28-7.48 (5H, m, Ar-H), 7.76 (1H, br.s, NH, disappeared with D2O), 7.99 (1H, s, NH, disappeared with D2O). 13C NMR: (150 MHz, CDCl3): δ 13.45, 21.76, 25.76, 26.67, 28.45, 29.66, 36.43, 38.65, 43.45, 48.40, 50.87, 59.45, 69.67, 112.43, 112.76, 115.66, 119.11, 126.68, 129.45, 132.67, 138.53, 152.55, 153.79, 161.02 (26 C). MS (EI): m/z 406 (77%) [M+]. Anal. Calcd for C26H34N2O2 (406.56): Calcd C, 76.81; H, 8.43; N, 6.89. Found C, 76.75; H, 8.40; N, 6.80.

16-(α-Methoxy-ethane)-17-[N-phenyl-hydrazino]-estra-1(10),2,4-trien-[17,16-c]-3-ol (3j)

Yield 66%, mp 229–231 °C, [ α ] D 25 = + 147 (c 1, MeOH). IR (KBr, cm–1): 3433–3363 (NH), 3346 (OH), 3066 (CH, aromatic), 2955 (CH, aliphatic), 1635 (C=C). 1H NMR: (600 MHz, CDCl3): δ ppm 0.62 (1H, m, H-8β), 0.92 (3H, s, CH3-18), 1.02 (1H, m, H-11β), 1.13 (1H, m, H-7α), 1.18 (1H, m, H-12α), 1.26 (1H, m, H-14α), 1.39 (1H, m, H-15β), 1.42 (3H, m, CH3, C-16 ethane), 1.58 (1H, m, H-15α), 1.69 (1H, m, H-7β), 1.83 (1H, m, CH, H-16α), 2.03 (1H, m, H-9α), 2.10 (1H, m, H-11α), 2.52 (1H, m, H-12β), 2.62 (1H, m, H-6α), 2.65 (1H, m, H-6β), 2.85 (1H, d, H-17), 3.49 (3H, s, OCH3), 4.86 (1H, d, CH-O), 4.89 (1H, s, OH, disappeared with D2O), 5.71 (1H, dd, H-2), 6.61 (1H, d, H-4), 7.12 (1H, d, H-1), 7.27-7.49 (5H, m, Ar-H), 7.77 (1H, br.s, NH, disappeared with D2O), 7.97 (2H, s, NH, disappeared with D2O). 13C NMR: (150 MHz, CDCl3): δ 13.52, 21.78, 22.92, 25.67, 26.45, 28.10, 29.90, 36.17, 38.47, 43.80, 48.69, 50.55, 59.25, 69.11, 112.12, 112.78, 115.54, 119.17, 126.46, 129.53, 132.79, 138.78, 152.42, 153.76, 163.11 (27 C). MS (EI): m/z 420 (81%) [M+]. Anal. Calcd for C27H36N2O2 (420.58): Calcd C, 77.10; H, 8.63; N, 6.66. Found C, 77.00; H, 8.58; N, 6.60.

16-(α-Ethoxy-methane)-17-[N-phenyl-hydrazino]-estra-1(10),2,4-trien-[17,16-c]-3-ol (3k)

Yield 60%, mp 338–340 °C, [ α ] D 25 = + 155 (c 1, MeOH). IR (KBr, cm–1): 3431–3362 (NH), 3343 (OH), 3064 (CH, aromatic), 2956 (CH, aliphatic), 1637 (C=C). 1H NMR: (600 MHz, CDCl3): δ ppm 0.63 (1H, m, H-8β), 0.90 (3H, s, CH3-18), 1.01 (1H, m, H-11β), 1.11 (1H, m, H-7α), 1.16 (1H, m, H-12α), 1.21 (3H, m, CH3, ethoxyl), 1.24 (1H, m, H-14α), 1.36 (1H, m, H-15β), 1.59 (1H, m, H-15α), 1.68 (1H, m, H-7β), 2.00 (1H, m, H-9α), 1.84 (1H, m, CH, H-16α), 2.11 (1H, m, H-11α), 2.50 (1H, m, H-12β), 2.61 (1H, m, H-6α), 2.70 (1H, m, H-6β), 6. disappeared (1H, d, H-4), 2.80 (1H, d, H-17), 3.53 (2H, m, OCH2), 4.80 (2H, d, CH-O), 4.84 (1H, s, OH, exchangeable with D2O), 5.70 (1H, dd, H-2), 7.15 (1H, d, H-1), 7.26-7.47 (5H, m, Ar-H), 7.76 (1H, br.s, NH, disappeared with D2O), 7.98 (1H, m, NH, disappeared with D2O). 13C NMR: (150 MHz, CDCl3): δ 13.64, 18.74, 21.57, 25.17, 26.30, 28.05, 29.64, 36.54, 38.15, 44.18, 48.46, 50.55, 68.43, 69.13, 112.49, 112.72, 115.60, 119.11, 126.34, 129.50, 131.11, 138.49, 152.40, 153.84, 162.47 (27 C). MS (EI): m/z 420 (88%) [M+]. Anal. Calcd for C27H36N2O2 (420.58): Calcd C, 77.10; H, 8.63; N, 6.66. Found C, 77.02; H, 8.57; N, 6.61.

16-(α-Ethoxy-ethane)-17-[N-phenyl-hydrazino]-estra-1(10),2,4-trien-[17,16-c]-3-ol (3l)

Yield 96%, mp 288–300 °C, [ α ] D 25 = + 95 (c 1, MeOH). IR (KBr, cm–1): 3431–3367 (NH), 3343 (OH), 3068 (CH, aromatic), 2951 (CH, aliphatic), 1631 (C=C). 1H NMR: (600 MHz, CDCl3): δ ppm 0.58 (1H, m, H-8β), 0.91 (3H, s, CH3-18), 1.00 (1H, m, H-11β), 1.10 (1H, m, H-7α), 1.14 (1H, m, H-12α), 1.21 (3H, m, CH3, ethoxyl), 1.24 (1H, m, H-14α), 1.36 (1H, m, H-15β), 1.44 (3H, m, CH3, C-16 ethane), 1.60 (1H, m, H-15α), 1.71 (1H, m, H-7β), 1.97 (1H, m, H-9 α), 1.83 (1H, m, CH, H-16α), 2.11 (1H, m, H-11α), 2.51 (1H, m, H-12β), 2.62 (1H, m, H-6α), 2.67 (1H, m, H-6 β), 2.88 (1H, d, H-17), 3.49 (2H, m, OCH2), 4.84 (1H, d, CH-O), 4.87 (1H, s, OH, disappeared with D2O), 5.73 (1H, dd, H-2), 6.62 (1H, d, H-4), 7.13 (1H, d, H-1), 7.23-7.47 (5H, m, Ar-H), 7.77 (1H, br.s, NH, disappeared with D2O), 7.87 (1H, m, NH, disappeared with D2O). 13C NMR: (150 MHz, CDCl3): δ 13.56, 18.77, 22.27, 22.32, 25.76, 26.78, 27.34, 28.29, 36.54, 37.90, 44.21, 48.23, 51.32, 68.34, 69.56, 112.67, 112.77, 115.56, 119.10, 126.45, 129.50, 133.47, 137.23, 152.55, 155.51, 163.73 (28 C). MS (EI): m/z 434 (93%) [M+]. Anal. Calcd for C28H38N2O2 (434.61): Calcd C, 77.38; H, 8.81; N, 6.45. Found C, 77.30; H, 8.75; N, 6.40.

3.1.3. Synthesis of pyrazoline-3-ol derivatives (4a–d)

A mixture of 2a,b (4 mmol) and hydrazine derivatives, namely, methy hydrazine or phenyl hydrazine (16 mmol) in dioxane (25 mL) was refluxed for 5 h. The mixture was evaporated under vacuum, the residue formed was triturated with water, filtered off, washed with water, dried and crystallized from MeOH to give derivatives 4a–d, respectively.

1`-Methyl-1`H-estra-1(10),2,4-trien-[17,16-c]pyrazoline-3-ol (4a)

Yield 96%, mp 117–119 °C, [ α ] D 25 = + 109 (c 1, MeOH). IR (KBr, cm–1): 3351 (OH), 2946 (CH, aliphatic), 1627 (C=C), 1610 (C=N). 1H NMR: (600 MHz, CDCl3): δ ppm 0.61 (1H, m, H-8β), 0.94 (3H, s, CH3-18), 1.08 (1H, m, H-11β), 1.13 (1H, m, H-7α), 1.18 (1H, m, H-12α), 1.27 (1H, m, H-14α), 1.41 (1H, m, H-15β), 1.66 (1H, m, H-15α), 1.74 (1H, m, H-7β), 1.84 (1H, m, CH, H-16α), 2.01 (1H, m, H-9α), 2.10 (3H, s, NCH3), 2.22 (1H, m, H-11α), 2.42 (1H, m, H-12β), 2.52 (1H, m, H-6α), 2.66 (1H, m, H-6β), 2.84 (2H, d, pyrazoline-5′), 4.95 (1H, s, OH, disappeared with D2O), 5.75 (1H, dd, H-2), 6.65 (1H, d, H-4), 7.15 (1H, d, H-1). 13C NMR: (150 MHz, CDCl3): δ 14.60, 22.56, 25.35, 26.46, 28.77, 29.56, 36.68, 38.68, 43.34, 45.36, 48.21, 50.24, 64.45, 112.46, 115.57, 126.44, 132.56, 138.45, 152.67, 156.46 (20 C). MS (EI): m/z 310 (100%) [M+]. Anal. Calcd for C20H26N2O (310.43): Calcd C, 77.38; H, 8.44; N, 9.02. Found C, 77.29; H, 8.36; N, 8.98.

1`-Methyl -1`H-5`-methyl-estra-1(10),2,4-trien-[17,16-c]pyrazoline-3-ol (4b)

Yield 88%, mp 216–218 °C, [ α ] D 25 = + 111 (c 1, MeOH). IR (KBr, cm–1): 3351 (OH), 2946 (CH, aliphatic), 1627 (C=C), 1617 (C=N). 1H NMR: (600 MHz, CDCl3): δ ppm 0.62 (1H, m, H-8β), 0.92 (3H, s, CH3-18), 1.02 (1H, m, H-11β), 1.05 (3H, s, CH3-5′-methyl), 1.10 (1H, m, H-7α), 1.14 (1H, m, H-12α), 1.25 (1H, m, H-14α), 1.40 (1H, m, H-15β), 1.66 (1H, m, H -15α), 1.77 (1H, m, H-7β), 1.88 (1H, m, CH, H-16α), 2.00 (1H, m, H-9α), 2.17 (3H, s, N-CH3), 2.20 (1H, m, H-11α), 2.47 (1H, m, H-12β), 2.57 (1H, m, H-6α), 2.66 (1H, m, H-6β), 2.84 (1H, m, pyrazoline-5′), 4.88 (1H, s, OH, disappeared with D2O), 5.79 (1H, dd, H-2), 6.68 (1H, d, H-4), 7.19 (1H, d, H-1). 13C NMR: (150 MHz, CDCl3): δ 13.35, 21.80, 24.27, 25.30, 26.07, 28.20, 29.77, 36.76, 38.70, 43.67, 45.67, 48.50, 50.70, 64.45, 112.37, 115.54, 126.46, 132.40, 138.77, 153.44, 155.42 (21 C). MS (EI): m/z 324 (100%) [M+]. Anal. Calcd for C21H28N2O (324.45): Calcd C, 77.74; H, 8.70; N, 8.63. Found C, 77.66; H, 8.64; N, 8.58.

1`-Phenyl-1`H-estra-1(10),2,4-trien-[17,16-c]pyrazoline-3-ol (4c)

Yield 70%, mp 255–257 °C, [ α ] D 25 = + 128 (c 1, MeOH). IR (KBr, cm–1): 3347 (OH), 3071 and 3061 (CH, aromatic), 2931 (CH, aliphatic), 1633 (C=C), 1614 (C=N). 1H NMR: (600 MHz, CDCl3): δ ppm 0.60 (1H, m, H-8β), 0.90 (3H, s, CH3-18), 1.00 (1H, m, H-11β), 1.10 (1H, m, H-7α), 1.20 (1H, m, H-12α), 1.28 (1H, m, H-14α), 1.49 (1H, m, H-15β), 1.68 (1H, m, H-15α), 1.77 (1H, m, H-7β), 1.86 (1H, m, CH, H-16α), 2.04 (1H, m, H-9α), 2.26 (1H, m, H-11α), 2.48 (1H, m, H -12β), 2.54 (1H, m, H-6α), 2.64 (1H, m, H-6β), 2.84 (2H, d, pyrazoline-5`), 5.00 (1H, s, OH, disappeared with D2O), 5.71 (1H, dd, H-2), 6.70 (1H, d, H-4), 7.10 (1H, d, H-1), 7.28-7.40 (5H, m, Ar-H). 13C NMR: (150 MHz, CDCl3): δ 13.49, 21.24, 25.55, 26.46, 28.51, 29.67, 36.64, 38.45, 43.35, 50.73, 58.26, 64.45, 112.10, 112.48, 115.46, 119.31, 126.10, 129.20, 132.52, 138.79, 152.20, 153.13, 163.48 (25 C). MS (EI): m/z 372 (81%) [M+]. Anal. Calcd for C25H28N2O (372.50): Calcd C, 80.61; H, 7.58; N, 7.52. Found C, 80.52; H, 7.50; N, 7.48.

1`-Phenyl-1`H-5`-methyl-estra-1(10),2,4-trien-[17,16-c]pyrazoline-3-ol (4d)

Yield 75%, mp 214–216 °C, [ α ] D 25 = + 127 (c 1, MeOH). IR (KBr, cm–1): 3345 (OH), 3068 (CH, aromatic), 2949 (CH, aliphatic), 1625 (C=C), 1614 (C=N). 1H NMR: (600 MHz, CDCl3): δ ppm 0.62 (1H, m, H-8β), 0.93 (3H, s, CH3-18), 1.02 (1H, m, H-11β), 1.05 (3H, s, CH3-5`-methyl), 1.12 (1H, m, H-7α), 1.17 (1H, m, H-12α), 1.27 (1H, m, H-14α), 1.41 (1H, m, H-15β), 1.71 (1H, m, H -15α), 1.77 (1H, m, H-7β), 1.87 (1H, m, CH, H-16α), 2.02 (1H, m, H-9α), 2.22 (1H, m, H-11α), 2.46 (1H, m, H -12β), 2.54 (1H, m, H-6α), 2.65 (1H, m, H-6β), 3.83 (1H, s, pyrazoline-5`), 4.91 (1H, s, OH, disappeared with D2O), 5.81 (1H, dd, H-2), 6.73 (1H, d, H-4), 7.21 (1H, d, H-1), 7.28-7.48 (5H, m, Ar-H). 13C NMR: (150 MHz, CDCl3): δ 13.81, 21.89, 24.25, 25.80, 26.35, 28.34, 29.23, 36.56, 38.67, 43.34, 48.55, 50.67, 64.44, 112.70, 115.34, 119.10, 126.76, 129.50, 132.45, 138.56, 152.40, 154.36, 163.56, 164.55 (26 C). MS (EI): m/z 386 (23%) [M+]. Anal. Calcd for C26H30N2O (386.52): Calcd C, 80.79; H, 7.82; N, 7.25. Found C, 80.70; H, 7.76; N, 7.20.

3.2. Biological Screening

3.2.1. In Vitro Cytotoxic Activities

The cytotoxic activities of the synthesized derivatives against ovarian cancer cell lines (SKOV-3) were evaluated using standard MTT assay [26,27]. SKOV-3 cells cultivated on DMEM medium supplemented with 10% FBS, 100×, 1% antibiotic/antimycotic solution and 3.6 g/L NaHCO3 were used. Cells were incubated under standard laboratory conditions [28,29]. Before testing compounds, cells were trypsinized, centrifuged and prepared as per our developed protocol [30,31]. MTT assay relies on the enzymatic conversion of the MTT substrate into purple formazan by living cells mitochondrial enzymes. Shortly, 96 well plates were seeded (10,000 cells/100 µL/well), then incubated for 24 h at standard conditions. Afterwards, cells were exposed to different concentrations from prepared compounds and incubated for another 24 h. Thereafter, MTT (10 µL, 5 mg/mL, PBS) were added to each well, and plates were incubated for 4 h. Supernatants were then replaced discarded with 200 µL of DMSO to dissolve the precipitated formazan crystals. Formazan absorbance, proportional to living cell number, was read at 550 nm using a microplate reader. Viability percentages were calculated and the corresponding IC50 values were obtained from the linear regression of the calibration curve. For comparison, doxorubicin (DOX) and resveratrol (RES) were used as positive controls. Furthermore, the effect of the newly synthesized derivatives was tested against normal fibroblast cell line to assess their toxicities.

3.2.2. In Vitro Anti-Ovarian Xenograft Model

Anti-ovarian cancer xenograft animal model was developed in nude mouse using SKOV-3 cells according to McCauley et al. [32]. Treatment protocol was approved by the University of South Dakota, Institutional Animal Care and Use Committee (study protocol 50-01-05-08B). Immunodeficient (athymic nude-Foxn1nu) female mice were inoculated (1 × 106 cells/site). Upon tumor development, (av. volume ~50 mm3), 10-mice groups were implanted s.c. with constant release pellets containing tested compound (1 µM/gm) or placebo. Animal and tumor development were monitored measured every 5 days after pellet implantation, using standardized Caliper in two perpendicular diameters of the implant according to our previous work [2]. Due to large tumor volumes, (5-fold difference between largest and smallest tumor), relative tumor volumes were used for comparison. Relative tumor volume = Vtvolume at time t/V0initial volume.

3.2.3. Topoisomerase II Inhibition

The inhibitory effects of the synthesized compounds against topoisomerase II were investigated using relaxation assay according to Goyeneche, et al. [33]. Topoisomerase II was purified from P388 cells, where enzyme unit corresponds to the activity required complete relaxation of 0.125 g of supercoiled pBR-322 DNA at 30 °C for 1 h. The assay was performed as per protocol and samples were subjected to electrophoresis (0.7% agarose gels, TBE buffer. Ethidium bromide-stained DNA was examined under UV light.

3.2.4. In Vitro Kinase Inhibition

Protein Expression and Purification

BRAF wild-type and V600E mutant kinase domains were expressed and purified according to Nakamura et al. [34]. Shortly, Sf 9 cells infected with BRAF kinase domain containing baculovirus were resuspended, sonicated, and then cleared using ultracentrifugation. Afterwards, the cleared lysate with mixed with equilibrated Talon resin, and the resin then washed with 10 column volumes of wash buffer and then eluted with buffer (25 mM Tris, pH 7.0, 250 mM NaCl, 160 mM imidazole, 10% glycerol). The eluant was then diluted, concentrated using Superdex 200 gel filtration column, purified as previously described and finally stored at –80 °C until use.

In Vitro ELISA-Based Kinase Assay

“This method was adopted from Nakamura et al. [34] and Qin et al. [35]. The previously prepared GST MEK-His protein was diluted in TTBS buffer (100 μL, 50 μg/mL), and then bound to 96-plate wells coated with glutathione (Pierce Biotechnology). Two DMSO-dilutions of each compound (1 µL) were added to 50 μL (50mM HEPES, pH7.0, 0.7 pmol of BRAFV600E kinase). Tubes were incubated at ambient temperature for 1 h, then added to GST-MEK-His-plate wells. Afterwards, 50 μL of phosphorylation buffer were added to start kinase reaction (37 °C/30 min/shaking). Reaction was stopped, then substrate was added, and the signal was recorded. The high throughput inhibitor screening was carried out according to standard assay conditions at different concentrations, to generate a sigmoidal dose response curve, a four-parameter logistic model using GraphPad Prism, for BRAF proteins, which was used to obtain corresponding IC50 values”.

3.2.5. Statistical Analysis

“Measurements were carried out in thrice and data are represented as means ± SEM. Significance was calculated by Student’s t-test using SPSS software (SPSS Inc., Paris, France). * p <0.05, ** p < 0.01, *** p < 0.001. IC50 values were calculated using GraphPad Prism software, GraphPad Software, San Diego, CA, USA [36]”.

4. Conclusions

During the current investigation, we synthesized a series of 17-hydrazino- and 17,16-c-pyrazoline estrone derivatives. Prepared compounds exhibited potential in vitro and in vivo anti-ovarian cancer activity against SKOV-3 cells. Compound 3a was found to be the most potent with an IC50 value of 4.23 ± 0.12 nM. Additionally, in vivo xenograft ovarian cancer model showed that Cpd. 3a was able to reduce tumor growth by 93.61 ± 0.7% after 40 days of treatment. Furthermore, the newly synthesized compounds were able to interfere with tumor proliferation through inhibiting the activity of topoisomerase II and V600EBRAF, where the obtained IC50 values for both enzymes were 3.45 ± 0.13 nM and 0.041 ± 0.0016 μM, respectively.

Author Contributions

M.E.-N., A.E.-G.E.A., A.A.F. performed most of the experiments; A.E.-G.E.A., M.A.A.-O., A.A.F. analyzed the data; E.A.E. and M.M.A. the contributed to the anticancer activity assays; All authors read and approved the final manuscript.

Funding

The authors are grateful to the Deanship of Scientific Research, king Saud University for funding through Vice Deanship of Scientific Research Chairs.

Acknowledgments

The authors are grateful to the Deanship of Scientific Research, king Saud University for funding through Vice Deanship of Scientific Research Chairs.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gupta, A.; Kumar, B.S.; Negi, A.S. Current status on development of steroids as anticancer agents. J. Steroid Biochem. Mol. Boil. 2013, 137, 242–270. [Google Scholar] [CrossRef] [PubMed]
  2. Amr, A.E.-G.E.; El-Naggar, M.; Al-Omar, M.A.; Elsayed, E.A.; Abdalla, M.M. In vitro and in vivo anti-breast cancer activities of some synthesized pyrazolinyl-estran-17-one candidates. Molecules 2018, 23, 1572. [Google Scholar] [CrossRef] [PubMed]
  3. Afzal, M.; Al-Awadi, S.; Oommen, S. Antioxidant activity of biotransformed sex hormones facilitated by Bacillus stearothermophilus. In Advanced Protocols in Oxidative Stress II, Methods in Molecular Biology; Armstrong, D., Ed.; Springer: Basel, Switzerland, 2010. [Google Scholar]
  4. Schönecker, B.; Lange, C.; Kötteritzsch, M.; Günther, W.; Weston, J.; Anders, E.; Görls, H. Conformational design for 13α-Steroids. J. Org. Chem. 2000, 65, 5487–5497. [Google Scholar] [CrossRef]
  5. Jovanovic-Santa, S.; Petrović, J.; Andrić, S.; Kovačević, R.; Ðurendić, E.; Sakač, M.; Lazar, D.; Stanković, S. Synthesis, structure, and screening of estrogenic and antiestrogenic activity of new 3,17-substituted- 16,17-seco-estratriene derivatives. Bioorg. Chem. 2003, 31, 475–484. [Google Scholar] [CrossRef]
  6. Minorics, R.; Bózsity, N.; Wölfling, J.; Mernyák, E.; Schneider, G.; Márki, A.; Falkay, G.; Ocsovszki, I.; Zupkó, I. Antiproliferative effect of normal and 13-epi-D-homoestrone and their 3-methyl ethers on human reproductive cancer cell lines. J. Steroid Biochem. Mol. Biol. 2013, 132, 168–175. [Google Scholar] [CrossRef] [PubMed]
  7. Ayan, D.P.; Maltais, R.; Roy, J.; Poirier, D. Impact of estradiol structural modifications (18-methyl and/or 17-hydroxy inversion of configuration) on the in vitro and in vivo estrogenic activity. In Proceedings of the Endocrine Society’s 93rd Annual Meeting & Expo, Boston, MA, USA, 4–7 June 2011; Volume 127. [Google Scholar]
  8. Szabó, J.; Bacsa, I.; Wölfling, J.; Schneider, G.; Zupkó, I.; Varga, M.; Herman, B.E.; Kalmár, L.; Szécsi, M.; Mernyák, E. Synthesis and in vitro pharmacological evaluation of N-[(1-benzyl-1,2,3-triazol-4-yl)methyl]- carboxamides on D-secoestrone scaffolds. J. Enzyme Inhib. Med. Chem. 2016, 31, 574–579. [Google Scholar] [CrossRef] [PubMed]
  9. Szabó, J.; Jerkovics, N.; Schneider, G.; Wölfling, J.; Bózsity, N.; Minorics, R.; Zupkó, I.; Mernyák, E.; De Sousa, M.E. Synthesis and in vitro antiproliferative evaluation of c-13 epimers of triazolyl-d-secoestrone alcohols: the first potent 13α-d-secoestrone derivative. Molecules 2016, 21, 611. [Google Scholar] [CrossRef] [PubMed]
  10. Szabó, J.; Pataki, Z.; Wölfling, J.; Schneider, G.; Bózsity, N.; Minorics, R.; Zupkó, I.; Mernyák, E. Synthesis and biological evaluation of 13α-estrone derivatives as potential antiproliferative agents. Steroids 2016, 113, 14–21. [Google Scholar] [CrossRef]
  11. Mernyák, E.; Kovács, I.; Minorics, R.; Sere, P.; Czégány, D.; Sinka, I.; Wölfling, J.; Schneider, G.; Újfaludi, Z.; Boros, I.; et al. Synthesis of trans-16-triazolyl-13α-methyl-17-estradiol diastereomers and the effects of structural modifications on their in vitro antiproliferative activities. J. Steroid Biochem. Mol. Boil. 2015, 150, 123–134. [Google Scholar] [CrossRef]
  12. Rao, P.N.; Cessac, J.W.; Tinley, T.L.; Mooberry, S.L. Synthesis and antimitotic activity of novel 2-methoxyestradiol analogs. Steroids 2002, 67, 1079–1089. [Google Scholar] [CrossRef]
  13. Sundov, D.; Čarić, A.; Mrklić, I.; Gugic, D.; Čapkun, V.; Hofman, I.D.; Mise, B.P.; Tomic, S. P53, MAPK, topoisomerase II alpha and Ki67 immunohistochemical expression and KRAS/BRAF mutation in ovarian serous carcinomas. Diagn. Pathol. 2013, 8, 21. [Google Scholar] [CrossRef]
  14. Kaur, P.V.; Kaur, S. DNA Topoisomerase II: promising target for anticancer drugs. In Multi-Targeted Approach to Treatment of Cancer; Gandhi, V., Ed.; Springer: Basel, Switzerland, 2015; pp. 323–338. [Google Scholar]
  15. Nijenhuis, C.; Lucas, L.; Rosing, H.; Huitema, A.; Mergui-Roelvink, M.; Jamieson, G.C.; Fox, J.; Mould, D.; Schellens, J.; Beijnen, J. Metabolism and disposition of the anticancer quinolone derivative vosaroxin, a novel inhibitor of topoisomerase II. Investig. New Drugs 2017, 35, 478–490. [Google Scholar] [CrossRef]
  16. Rahman, M.A.; Salajegheh, A.; Smith, R.A.; Lam, A.K.-Y.; Ariana, A. Inhibition of BRAF kinase suppresses cellular proliferation, but not enough for complete growth arrest in BRAF V600E mutated papillary and undifferentiated thyroid carcinomas. Endocrine 2016, 54, 129–138. [Google Scholar] [CrossRef]
  17. Rahman, M.A.; Salajegheh, A.; Smith, R.A.; Lam, A.K.-Y.; Ariana, A. Multiple proliferation-survival signalling pathways are simultaneously active in BRAF V600E mutated thyroid carcinomas. Exp. Mol. Pathol. 2015, 99, 492–497. [Google Scholar] [CrossRef]
  18. McCubrey, J.A.; Steelman, L.S.; Abrams, S.L.; Lee, J.T.; Chang, F.; Bertrand, F.E.; Navolanic, P.M.; Terrian, D.M.; Franklin, R.A.; D’Assoro, A.B.; et al. Roles of the RAF/MEK/ERK and PI3K/PTEN/AKT pathways in malignant transformation and drug resistance. Adv. Enzym. Regul. 2006, 46, 249–279. [Google Scholar] [CrossRef]
  19. Ouf, N.H.; Amr, A.E. Synthesis and anti-inflammatory activity of some pyrimidines and thieno- pyrimidines using 1-(2-benzo[d][1,3]dioxol-5-yl)vinyl)-4-mercapto-6-methyl-pyrimidine-5-yl)-ethan-2- one as a starting material. Monatsh. fur Chem. 2008, 139, 579–585. [Google Scholar] [CrossRef]
  20. Amr, A.E.; Abdulla, M.M. Synthesis and anti-inflammatory activities of new cynopyrane derivatives fused with steroidal nuclei. Archiv. der Pharm. 2008, 339, 88–95. [Google Scholar] [CrossRef]
  21. Khalifa, N.M.; Al-Omar, M.A.; Amr, A.E.; Haiba, M.E. Antiviral activity of some new polycyclic nucleoside pyrene candidate against HIV-1 and HSV-1 virus. Int. J. Biol. Macromol. 2013, 54, 51. [Google Scholar] [CrossRef]
  22. Abdel-Wahab, B.F.; Mohamed, S.F.; Amr, A.E.-G.E.; Abdalla, M.M. Synthesis and reactions of thiosemicarbazides, triazoles, and Schiff bases as antihypertensive α-blocking agents. Monatsh. Chem. 2008, 139, 1083–1090. [Google Scholar] [CrossRef]
  23. Amr, A.E.-G.E.; Elsayed, E.A.; Al-Omar, M.A.; Eldin, H.O.B.; Nossier, E.S.; Abdallah, M.M.; Eldin, H.B. Design, synthesis, anticancer evaluation and molecular modeling of novel estrogen derivatives. Molecules 2019, 24, 416. [Google Scholar] [CrossRef]
  24. Day, J.M.; Foster, P.A.; Tutill, H.J.; Schmidlin, F.; Sharland, C.M.; Hargrave, J.D.; Vicker, N.; Potter, B.V.L.; Reed, M.J.; Purohit, A. STX2171, a 17-hydroxysteroid dehydrogenase type 3 inhibitor, is efficacious in vivo in a novel hormone-dependent prostate cancer model. Endocr. Relat. Cancer 2013, 20, 53–64. [Google Scholar] [CrossRef]
  25. Donald, P.; Ho-Jin, C.; Arezki, A.; Roch, P.B.; Sheng-Xiang, L. Estrone and estradiol C-16 derivatives as inhibitors of type 1 17β-hydroxysteroid dehydrogenase. Mol. Cell. Endocrinol. 2006, 248, 236–238. [Google Scholar]
  26. Elsayed, E.A.; Sharaf-Eldin, M.; Wadaan, M. In vitro evaluation of cytotoxic activities of essential oil from moringa oleifera seeds on HeLa, HepG2, MCF-7, CACO-2 and L929 Cell Lines. Asian Pac. J. Cancer Prev. 2015, 16, 4671–4675. [Google Scholar] [CrossRef]
  27. Elsayed, E.A.; Farooq, M.; Dailin, D.; El-Enshasy, H.A.; Othman, N.Z.; Malek, R.; Danial, E.; Wadaan, M. In vitro and in vivo biological screening of kefiran polysaccharide produced by Lactobacillus kefiranofaciens. Biomed. Res. 2017, 28, 594–600. [Google Scholar]
  28. Al-Salahi, R.; Elsayed, E.A.; El Dib, R.A.; Wadaan, M.; Ezzeldin, E.; Marzouk, M. Synthesis, characterization and cytotoxicity evaluation of 5-hydrazono-[1,2,4]triazolo[1,5-a]quinazolines (Part I). Lat. Am. J. Pharm. 2016, 35, 58–65. [Google Scholar]
  29. Al-Salahi, R.; Elsayed, E.A.; El Dib, R.A.; Wadaan, M.; Ezzeldin, E.; Marzouk, M. Cytotoxicity of new 5-hydrazono-[1,2,4]triazolo[1,5-a]quinazolines (Part II). Lat. Am. J. Pharm. 2016, 35, 66–73. [Google Scholar]
  30. Elsayed, E.A.; Sharaf-Eldin, M.A.; El-Enshasy, H.A.; Wadaan, M. In vitro assessment of anticancer properties of Moringa peregrina essential seed oil on different cell lines. Pak. J. Zool. 2016, 48, 853–859. [Google Scholar]
  31. Farooq, M.; Abu Taha, N.; Butorac, R.R.; Evans, D.A.; Elzatahry, A.A.; Elsayed, E.A.; Wadaan, M.A.M.; Al-Deyab, S.S.; Cowley, A.H.; Li, J. Biological screening of newly synthesized BIAN N-heterocyclic gold carbene complexes in zebrafish embryos. Int. J. Mol. Sci. 2015, 16, 24718–24731. [Google Scholar] [CrossRef]
  32. McCauley, J.; Zivanovic, A.; Skropeta, D. Bioassays for anticancer activities. Methods Mol. Biol. 2013, 1055, 191–205. [Google Scholar]
  33. Goyeneche, A.A.; Carón, R.W.; Telleria, C.M. Mifepristone inhibits ovarian cancer cancer cell growth in vitro and in vivo. Clin. Cancer Res. 2007, 13, 3370–3379. [Google Scholar] [CrossRef]
  34. Nakamura, K.; Sugumi, H.; Yamaguchi, A.; Uenaka, T.; Kotake, Y.; Okada, T.; Kamata, J.; Niijima, J.; Nagasu, T.; Koyanagi, N.; et al. Antitumor activity of ER-37328, a novel carbazole topoisomerase II inhibitor. Mol. Cancer Ther. 2002, 1, 169–75. [Google Scholar]
  35. Qin, J.; Xie, P.; Ventocilla, C.; Zhou, G.; Vultur, A.; Chen, Q.; Liu, Q.; Herlyn, M.; Winkler, J.; Marmorstein, R. Identification of a novel family of BRAF(V600E) inhibitors. J. Med. Chem. 2012, 55, 5220–5230. [Google Scholar] [CrossRef]
  36. Stones, C.J.; Kim, J.E.; Joseph, W.R.; Leung, E.; Marshall, E.S.; Finlay, G.J.; Shelling, A.N.; Baguley, B.C. Comparison of responses of human melanoma cell lines to MEK and BRAF inhibitors. Front. Genet. 2013, 4, 66. [Google Scholar] [CrossRef] [Green Version]
Sample Availability: Samples of the compounds are available from the authors.
Scheme 1. Synthetic route for compounds 24.
Scheme 1. Synthetic route for compounds 24.
Molecules 24 02054 sch001
Figure 1. In vitro Cytotoxicity activities of the tested compounds against SKOV-3 cells.
Figure 1. In vitro Cytotoxicity activities of the tested compounds against SKOV-3 cells.
Molecules 24 02054 g001
Figure 2. Percentages of decrease in tumor volume in nude mice in response to different synthesized derivatives.
Figure 2. Percentages of decrease in tumor volume in nude mice in response to different synthesized derivatives.
Molecules 24 02054 g002
Figure 3. IC50 of Topoisomerase II Inhibitor activities of the tested compounds.
Figure 3. IC50 of Topoisomerase II Inhibitor activities of the tested compounds.
Molecules 24 02054 g003
Figure 4. IC50 values of synthesized for both WTBRAF and V600EBRAF.
Figure 4. IC50 values of synthesized for both WTBRAF and V600EBRAF.
Molecules 24 02054 g004

Share and Cite

MDPI and ACS Style

El-Naggar, M.; E. Amr, A.E.-G.; Fayed, A.A.; A. Elsayed, E.; A. Al-Omar, M.; Abdalla, M.M. Potent Anti-Ovarian Cancer with Inhibitor Activities on Both Topoisomerase II and V600EBRAF of Synthesized Substituted Estrone Candidates. Molecules 2019, 24, 2054. https://doi.org/10.3390/molecules24112054

AMA Style

El-Naggar M, E. Amr AE-G, Fayed AA, A. Elsayed E, A. Al-Omar M, Abdalla MM. Potent Anti-Ovarian Cancer with Inhibitor Activities on Both Topoisomerase II and V600EBRAF of Synthesized Substituted Estrone Candidates. Molecules. 2019; 24(11):2054. https://doi.org/10.3390/molecules24112054

Chicago/Turabian Style

El-Naggar, Mohamed, Abd El-Galil E. Amr, Ahmed A. Fayed, Elsayed A. Elsayed, Mohamed A. Al-Omar, and Mohamed M. Abdalla. 2019. "Potent Anti-Ovarian Cancer with Inhibitor Activities on Both Topoisomerase II and V600EBRAF of Synthesized Substituted Estrone Candidates" Molecules 24, no. 11: 2054. https://doi.org/10.3390/molecules24112054

Article Metrics

Back to TopTop