Next Article in Journal
GC-MS Analysis of Defensive Secretions of a Cuban Endemic Millipede of the Genus Rhinocricus—Chemical Prospecting of Neotropical Biodiversity
Previous Article in Journal
Theoretical and Experimental Study of the Chemical Modification of Poly(epichlorohydrin) by Grafting Menthol
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Proceeding Paper

Synephrine Analogues as Glucocorticoid Receptor Agonists †

by
Eugenia Oleynik
1,*,
Ekaterina Mikhina
1,
Diana Grigoreva
2,
Varvara Maksimova
2,
Lyubov Grebenkina
1,
Andrey Valerevich Matveev
1,
Ekaterina Zhidkova
2,
Ekaterina Lesovaya
2 and
Marianna Yakubovskaya
2
1
Department of Biotechnology and Industrial Pharmacy, Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Moscow 119454, Russia
2
Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology”, Ministry of Health of the Russian Federation, Moscow 115522, Russia
*
Author to whom correspondence should be addressed.
Presented at the 27th International Electronic Conference on Synthetic Organic Chemistry (ECSOC-27), 15–30 November 2023; Available online: https://ecsoc-27.sciforum.net/.
Chem. Proc. 2023, 14(1), 58; https://doi.org/10.3390/ecsoc-27-16152
Published: 15 November 2023

Abstract

:
This work carried out the synthesis of several new synephrine analogues by universal method. Some of the synthesized compounds showed cytotoxicity on myeloid leukaemia cells K562 and lymphoma cell line Granta-519. Molecular docking using the glucocorticoid receptor (GR) model (PDB identifier 1P93) was performed to understand the possible underlying mechanism of compound action. The simulation showed the similarity of synephrine analogues’ binding to the binding of dexamethasone in the GR ligand-binding domain. The synthesized analogues exhibited cytotoxicity profiles similar to those of dexamethasone.

1. Introduction

Glucocorticoids (GC) are steroid hormones regulating many cellular and physiological functions and they are best known for their anti-inflammatory properties. Recently, more evidence has emerged that glucocorticoid regulation of inflammation affects oncogenesis [1,2]. Steroid GC are being used as immunomodulators during the application of main antitumor therapy methods, including chemotherapy and radiation therapy [3,4]. In addition, GC are widely used in the therapy of leukaemia and lymphoma as cytostatic drugs [5]. GC implement their biological effect via activation of glucocorticoid receptors which leads to the suppression of tumor cells’ growth and proliferation, inducing their apoptosis [6,7,8].
Synthetic GC dexamethasone (Dex) is a glucocorticoid receptor agonist commonly used as a direct chemotherapy agent in the case of certain malignant neoplasm types’; however, it has several shortcomings. The effect of Dex outside the zone of therapeutic interests may cause a wide range of complications, including systemic toxicity, local allergic reactions, changes in heart function etc., which makes the search and development of different less toxic agents especially important [9,10].
Nowadays it is known that the class of non-steroidal Dex analogues shows similar biological effects while being less overall toxic [11,12]. The design of such compounds may consist of replacing the Dex sterane backbone with a less rigid hydrocarbon skeleton, predominantly preserving the spatial arrangement of original functional groups. We have assumed that such analogues could be obtained based on the synephrine molecule, which mimics the Dex backbone (Figure 1).
These suggestions were examined by the molecular docking approach using the glucocorticoid receptor model. Simulation has shown selective binding of synephrine analogues in the hydrophobic pocket of the GR binding domain.

2. Results and Discussion

2.1. Chemistry Section

There are different synthetic approaches to obtaining synephrine analogues [13,14]. We have applied, in our opinion, the most expedient one which consists of the synthesis of intermediate epoxide and its following interaction with aliphatic primary and secondary amines (Figure 2).
The starting compound 2 was obtained according to known methods [15,16]. The 4-Alcoxychloroacetophenones 2 was reduced with the excess of NaBH4 in methanol followed by the treatment with KOH and the addition of a fivefold excess of amine without isolation of intermediate products. The target compounds 1fo were isolated by column chromatography on silica gel after the volatile components’ evaporation followed by excess amine extraction. The compounds 1ae were obtained by hydrogenation of 1ko on palladium on carbon (Figure 2).
For the initial study of biological properties compounds were synthesized as a mix of enantiomers. Further investigation suggests a synthesis of individual enantiomers only in the case of active compound detection. Individual enantiomers can be obtained by stereoselective reduction of corresponding ketones 2 [17].

2.2. In Silico Studies

Molecular docking has shown several significant non-covalent interactions demonstrated by studied compounds in the GR ligand-binding domain. Thus, π-alkyl interactions with Met604 and Leu608 were identified, which are also characteristic of steroid ligands. In addition, important hydrogen interactions which are characteristic of Dex were also found, including interactions with Gln642 and Thr739 (Figure 3). Dex location determined in the experiment coincides with literature data reported by other authors [12].

2.3. Biology Section

In vitro, experiments were held to estimate the cytotoxicity of new compounds on the model of the hematopoietic system malignant neoplasms. Cell proliferation was evaluated using myeloid chronic leukaemia cells K562 and B cell lymphoma cell line Granta-519 (Table 1).
Dex showed cytostatic on 48 and 72 depending on the cell line. Synthesized compounds demonstrated an effect similar to that of Dex, which suggests that the mechanism of their action is similar to the mechanism of Dex. In the case of the cell line K562, the studied compounds demonstrated an inhibitory effect on cell proliferation in 48 h (1km) and 72 h (1g), which is completely similar to the action of the reference drug. For the Granta-519 cell line, such a pattern could also be traced for 1a,g,k, and m, but these cells did not maintain a long-term effect, unlike Dex.

3. Conclusions

New synephrine analogues modified at the phenolic hydroxyl were synthesized. They showed a comparable Dex effect on the myeloid leukaemia cell line K562 and lymphoma cell line Granta-519. The in silico modelling data suggests that these analogues may compete with Dex to bind to the GR active site. These results encourage further investigation of synephrine analogues as GR agonists.

4. Materials and Methods

4.1. Materials

All the chemicals were obtained from commercial sources (Merck KGaA, Darmstadt, Germany) and were used without further purification. Deuterated solvents were purchased from Cambridge Isotope Laboratories, Inc. (Tewksbury, MA, USA). Silica gel 60 (Merck KGaA, Darmstadt, Germany) was used for column chromatography. Analytical TLC was performed on Silufol UV-254 plates.
The 1H NMR spectra were recorded at 300 MHz and 13C NMR spectra were recorded at 75 MHz on Bruker DPX-300 spectrometer. High-resolution mass spectra (HRMS) were recorded using the Agilent 6224 using electron spray ionization (ESI). HPLC measurements were carried out on the Agilent 1200 Series.

4.2. Synthesis

Synephrine analogues 1f–o (general synthesis procedure)
To a solution of 4-alcoxychloroacetophenone 2 (1.5 mmol) in methanol, 1 eq NaBH4 by portions while cooling on the ice bath. After compound 2 consumption (controlled by TLC, system petroleum ether/diethyl ether 1:1) KOH (1.2 eq) was added in portions. Subsequently, a solution of amine (5 eq) in methanol (1:1 by volume) was added. The reaction was TLC controlled by conversion of 3 (Rf = 0.65–0.75 system petroleum ether/diethyl ether 1:1) in the reaction mixture. Then the mixture was acidified with a diluted aqueous HCl up to pH = 3, the solvent was removed using a vacuum rotary evaporator, after which the residue was dissolved in 5 mL of water and extracted with methylene chloride (2 times, 10 mL). The organic layer was dried with anhydrous calcium chloride followed by filtering off the drying agent and evaporation. The product was isolated by column chromatography on silica gel using chloroform–methanol eluent.
2-(hexylamino)-1-(4-methoxyphenyl)ethanol (1g)
In total, 200 mg (54%) of 1g was obtained. Rf (10% methanol in chloroform) = 0.44. Purity by HPLC ≥ 98%. 1H NMR (CDCl3) δ, ppm: 7.35–7.26 (m, 2H, 2 × CH); 6.94–6.78 (m, 2H, 2 × CH); 5.01–4.71 (m, 1H, CH); 3.90–3.80 (m, 2H, NH and OH); 3.76 (s, 3H, O-CH3); 3.07–2.60 (m, 4H, 2 × CH2); 1.70–1.47 (m, 2H, CH2); 1.40–1.17 (m, 6H, 3 × CH2); 0.95–0.80 (m, 3H, CH3). 13C NMR (CDCl3) δ, ppm: 159.13; 133.70; 127.06; 113.80; 70.24; 56.30; 55.24; 49.11; 31.47; 28.39; 26.67; 22.51; 13.99. HRMS: for C15H26NO2 [M + H]+ calculated: 252.1963; found: 252.1967.
1-[4-(benzyloxy)phenyl]-2-piperidin-1-ylethanol (1k)
In total, 360 mg (77%) of 1k was obtained. Rf (5% methanol in chloroform) = 0.46. Purity by HPLC ≥ 97% 1H NMR (CDCl3) δ, ppm: 7.48–7.20 (m, 7H, 7 × CH); 7.00–6.87 (m, 2H, 2 × CH); 5.04 (s, 2H, CH2-O); 4.95–4.82 (m, 1H, CH-OH); 2.98–2.38 (m, 6H, 3 × CH2); 1.87–1.37 (m, 6H, 3 × CH2). 13C NMR (CDCl3) δ, ppm: 158.31; 136.89; 133.81; 128.54; 127.93; 127.43; 127.13; 114.77; 69.96; 67.97; 66.56; 54.62; 24.93; 23.42. HRMS: for C20H26NO2 [M + H]+ calculated: 312.1963; found: 312.1970.
1-[4-(benzyloxy)phenyl]-2-[(2-hydroxyethyl)amino]ethanol (1m)
In total, 95 mg (22%) of 1m was obtained. Rf (20% methanol in chloroform) = 0.33. Purity by HPLC ≥ 98% 1H NMR (DMSO-d6) δ, ppm: 7.50–7.25 (m, 7H, 7 × CH); 7.08–6.95 (m, 2H, 2 × CH); 5.09 (s, 2H, CH2-O); 5.27 (br.s, 1H, CH-OH); 4.89 (br.s, 1H, NH); 4.13–3.95 (m, 1H, CH); 3.74–3.45 (m, 4H, 2 × CH2); 2.85–2.52 (m, 2H, CH2). 13C NMR (DMSO-d6) δ, ppm: 158.29; 136.96; 129.43; 128.38; 127.78; 127.60; 122.08; 114.74; 69.18; 63.46; 63.00; 57.65; 48.01. HRMS: for C17H22NO3 [M + H]+ calculated: 288.1599; found: 288.1604.
Synephrine analogues 1 a-e (general synthesis procedure)
To 0.5 mmol of 1ko dissolved in 5 mL of EtOH, a 10% mass excess of Pd on C was added. The suspension was mixed at room temperature in a hydrogen atmosphere. After compound 1ko consumption (controlled by TLC, system 5% methanol in chloroform) the reaction mixture was filtered through a layer of celite and evaporated.
4-(1-hydroxy-2-piperidin-1-ylethyl)phenol (1a)
In total, 91 mg (82%) of 1a was obtained. Rf (25% methanol in chloroform) = 0.30. Purity by HPLC ≥ 98% 1H NMR (DMSO-d6) δ, ppm: 7.19–7.05 (m, 2H, 2 × CH); 6.73–6.63 (m, 2H, 2 × CH); 4.60–4.52 (m, 1H, CH-OH); 2.46–2.22 (m, 6H, 3 × CH2); 1.57–1.30 (m, 6H, 3 × CH2). 13C NMR (DMSO-d6) δ, ppm: 158.19; 134.74; 127.07; 114.64; 69.08; 67.11; 54.32; 25.52; 23.94. HRMS: for C17H28NO2 [M + H]+ calculated: 222.1494; found: 222.1501.

4.3. In Silico Studies

The 3D geometry of compounds was optimized using an MM2 force field in Chem3D software (Perkin Elmer Informatics, Inc., Waltham, MA, USA).
The crystal structure of GR was obtained from the Protein Data Bank (PDB ID: 1P93). The selected structure of the complex has a resolution of 2.7 Å and does not contain gaps in the main protein chain near the ligand–binding domain. Removing solvent molecules, adding hydrogen atoms, assigning atom types, combining non-polar hydrogen atoms, and calculating partial Gasteiger charges and Kollmann charges were done using the AutoDockTools 1.5.7 software (The Scripps Research Institute, La Jolla, CA, USA).
During the docking process, all the torsion bonds of the ligands were free to rotate, while the protein remained rigid. A 40 × 40 × 40 grid was created with 1 Å spacing centred on the GR active site. The docking calculations were performed using the Autodock Vina software (The Scripps Research Institute, La Jolla, CA, USA) utilizing the Lamarckian genetic algorithm (LGA). The visualization and graphical representation of the results of ligand interaction were conducted using the Discovery Studio Visualizer software (version 21.1.0.20298, Dassault Systems Biovia Corp., San Diego, CA, USA).

4.4. Biology

Cell viability (cell proliferation): The cells were cultured in 24-well plates (50,000 cells/well) and treated with solvent (DMSO), dexamethasone (25 µM), 1a (50 µM), 1g (50 µM), 1k (100 µM), and 1m (100 µM) for 24, 48, or 72 h for observe cytostatic action. After, the incubation cells were mixed 1:1 with 0.4% trypan blue in PBS solution. The viable cells were immediately counted using the TC20 automatic cell counter (“Bio-Rad”, Hercules, California, USA).

Author Contributions

Conceptualization, A.V.M., E.L. and M.Y.; methodology and data curation, A.V.M., E.Z. and L.G.; investigation, E.O., E.M., D.G. and V.M.; writing—original draft preparation, A.V.M. and E.O.; writing—review and editing, E.Z., L.G., E.L. and M.Y.; visualization, E.O.; funding acquisition, E.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Russian Science Foundation grant, grant number 23-15-00321.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Acknowledgments

This work was performed using the equipment of the Shared Science and Training Center for Collective Use RTU MIREA and supported by the Ministry of Science and Higher Education of the Russian Federation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Khadka, S.; Druffner, S.R.; Duncan, B.C.; Busada, J.T. Glucocorticoid regulation of cancer development and progression. Front. Endocrinol. 2023, 14, 1161768. [Google Scholar] [CrossRef] [PubMed]
  2. Lin, K.T.; Wang, L.H. New dimension of glucocorticoids in cancer treatment. Steroids 2016, 111, 84–88. [Google Scholar] [CrossRef] [PubMed]
  3. Chari, A.; Martinez-Lopez, J.; Mateos, M.V.; Bladé, J.; Benboubker, L.; Oriol, A.; Arnulf, B.; Rodriguez-Otero, P.; Pineiro, L.; Jakubowiak, A.; et al. Daratumumab plus carfilzomib and dexamethasone in patients with relapsed or refractory multiple myeloma. Blood 2019, 134, 421–431. [Google Scholar] [CrossRef] [PubMed]
  4. Huang, X.; Zheng, Z.; Zeng, B.; Xiao, H.; Zheng, H.; Lin, Z.; Song, J.; Li, A.; Chi, P.; Yang, Y.; et al. Impact of glucocorticoids on the efficacy of neoadjuvant chemoradiotherapy and survival of patients with locally advanced rectal cancer: A retrospective study. BMC Cancer 2023, 23, 238. [Google Scholar] [CrossRef] [PubMed]
  5. Herr, I. Glucocorticoiduse in prostate cancer and other solid tumours: Implications for effectiveness of cytotoxic treatment and metastases. Lancet Oncol. 2006, 7, 425–430. [Google Scholar] [CrossRef] [PubMed]
  6. Budunova, I.V.; Kowalczyk, D.; Pérez, P.; Yao, Y.J.; Jorcano, J.L.; Slaga, T.J. Glucocorticoid receptor functions as a potent suppressor of mouse skin carcinogenesis. Oncogene 2003, 22, 3279–3287. [Google Scholar] [CrossRef] [PubMed]
  7. Mayayo-Peralta, I.; Zwart, W.; Prekovic, S. Duality of glucocorticoid action in cancer: Tumor-suppressor or oncogene? Endocr. Relat. Cancer 2021, 28, 157–171. [Google Scholar] [CrossRef] [PubMed]
  8. Schoepe, S.; Schäcke, H.; May, E.; Asadullah, K. Glucocorticoid therapy-induced skin atrophy. Exp. Dermatol. 2006, 15, 406–420. [Google Scholar] [CrossRef] [PubMed]
  9. Schlossmacher, G.; Stevens, A.; White, A. Glucocorticoid receptor-mediated apoptosis: Mechanisms of resistance in cancer cells. J. Endocrinol. 2011, 211, 17–25. [Google Scholar] [CrossRef] [PubMed]
  10. Bal, M.P.; de Vries, W.B.; van der Leij, F.R.; van Oosterhout, M.F.; Berger, R.M.; Baan, J.; van der Wall, E.E.; van Bel, F.; Steendijk, P. Neonatal glucocorticosteroid treatment causes systolic dysfunction and compensatory dilation in early life: Studies in 4-week-old prepubertal rats. Pediatr. Res. 2005, 58, 46–52. [Google Scholar] [CrossRef] [PubMed]
  11. Zheng, Y.; Ishiguro, H.; Ide, H.; Inoue, S.; Kashiwagi, E.; Kawahara, T.; Jalalizadeh, M.; Reis, L.O.; Miyamoto, H. Compound A Inhibits Bladder Cancer Growth Predominantly via Glucocorticoid Receptor Transrepression. Mol Endocrinol. 2015, 29, 1486–1497. [Google Scholar] [CrossRef] [PubMed]
  12. Yemelyanov, A.; Czwornog, J.; Gera, L.; Joshi, S.; Chatterton, R.T., Jr.; Budunova, I. Novel steroid receptor phyto-modulator compound a inhibits growth and survival of prostate cancer cells. Cancer Res. 2008, 68, 4763–4773. [Google Scholar] [CrossRef] [PubMed]
  13. Tilova, L.R.; Savinkova, A.V.; Bocharov, A.K.; Kuzin, K.A.; Borisova, O.I.; Zhidkova, E.M.; Kirsanov, K.I.; Belitsky, G.A.; Yakubovskaya, M.G.; Yaminova, L.V.; et al. Synthesis of novel selective glucocorticoid receptor agonist and evaluation of its anticancer activity in the model of hematologic malignancies in vitro. Mod. Probl. Sci. Educ. 2016, 6, 110. [Google Scholar]
  14. Chen, J. Synthesis and Uses of Synephrine Derivatives. China Patent 0250944, 4 May 2005. [Google Scholar]
  15. Gu, X.L.; Liu, H.B.; Jia, Q.H.; Li, J.F.; Liu, Y.L. Design and synthesis of novel miconazole-based ciprofloxacin hybrids as potential antimicrobial agents. Monatshefte Chem.-Chem. Mon. 2014, 146, 713–720. [Google Scholar] [CrossRef]
  16. Zeng, H. Preparation Method of 2-(4-benzyloxy phenyl)ethanol. China Patent 113200825, 27 April 2021. [Google Scholar]
  17. Bailey, W.F.; Bischoff, J.J. Formation of cyclic ethers in the double Baeyer-Villiger oxidation of ketals derived from cyclic ketones. J. Org. Chem. 1985, 50, 3009–3010. [Google Scholar] [CrossRef]
Figure 1. Structures of Dex, synephrine, and its synthetic analogues 1.
Figure 1. Structures of Dex, synephrine, and its synthetic analogues 1.
Chemproc 14 00058 g001
Figure 2. Synthesis of synephrine derivatives 1ao.
Figure 2. Synthesis of synephrine derivatives 1ao.
Chemproc 14 00058 g002
Figure 3. Structure of the GR active site with the analogue of synephrine 1m in comparison to Dex (shown in blue and green, respectively).
Figure 3. Structure of the GR active site with the analogue of synephrine 1m in comparison to Dex (shown in blue and green, respectively).
Chemproc 14 00058 g003
Table 1. The proportion of live cells compared to the vehicle control.
Table 1. The proportion of live cells compared to the vehicle control.
Cell LineTime, hCompound No.
1a
(C = 50 µM)
1g
(C = 50 µM)
1k
(C = 100 µM)
1m
(C = 100 µM)
Dex
(C = 25 µM)
K562240.65 ± 0.080.80 ± 0.080.75 ± 0.060.53 ± 0.050.80 ± 0.05
480.52 ± 0.060.84 ± 0.070.60 ± 0.050.28 ± 0.090.65 ± 0.04
720.37 ± 0.120.53 ± 0.080.20 ± 0.050.18 ± 0.070.44 ± 0.10
Granta-519240.61 ± 0.110.93 ± 0.090.95 ± 0.040.95 ± 0.050.90 ± 0.04
480.54 ± 0.080.73 ± 0.070.48 ± 0.080.64 ± 0.080.53 ± 0.05
720.84 ± 0.091.00–0.070.70 ± 0.120.85 ± 0.060.64 ± 0.07
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Oleynik, E.; Mikhina, E.; Grigoreva, D.; Maksimova, V.; Grebenkina, L.; Matveev, A.V.; Zhidkova, E.; Lesovaya, E.; Yakubovskaya, M. Synephrine Analogues as Glucocorticoid Receptor Agonists. Chem. Proc. 2023, 14, 58. https://doi.org/10.3390/ecsoc-27-16152

AMA Style

Oleynik E, Mikhina E, Grigoreva D, Maksimova V, Grebenkina L, Matveev AV, Zhidkova E, Lesovaya E, Yakubovskaya M. Synephrine Analogues as Glucocorticoid Receptor Agonists. Chemistry Proceedings. 2023; 14(1):58. https://doi.org/10.3390/ecsoc-27-16152

Chicago/Turabian Style

Oleynik, Eugenia, Ekaterina Mikhina, Diana Grigoreva, Varvara Maksimova, Lyubov Grebenkina, Andrey Valerevich Matveev, Ekaterina Zhidkova, Ekaterina Lesovaya, and Marianna Yakubovskaya. 2023. "Synephrine Analogues as Glucocorticoid Receptor Agonists" Chemistry Proceedings 14, no. 1: 58. https://doi.org/10.3390/ecsoc-27-16152

Article Metrics

Back to TopTop