Next Article in Journal
Exploring the Roles of Liver X Receptors in Lipid Metabolism and Immunity in Atherosclerosis
Previous Article in Journal
The B22 Dilemma: Structural Basis for Conformational Differences in Proinsulin B-Chain Arg22 Mutants
Previous Article in Special Issue
A Review of the Phytochemistry, Molecular Docking, Pharmacology, Toxicology, Ethnopharmacology, Botany, and Clinical Studies of Maytenus senegalensis (Lam.) Excell
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Synthesis and Antiproliferative Effects of Grossheimin-Derived Aminoanalogues

by
Meruyert Ashimbayeva
1,
Zsolt Szakonyi
1,*,
Sergazy M. Adekenov
2,
Nikoletta Szemerédi
3,
Gabriella Spengler
3 and
Tam Minh Le
1,4,*
1
Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
2
JSC Research and Production Center “Phytochemistry”, Karaganda 100009, Kazakhstan
3
Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary
4
HUN-REN–SZTE Stereochemistry Research Group, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
*
Authors to whom correspondence should be addressed.
Biomolecules 2025, 15(4), 578; https://doi.org/10.3390/biom15040578
Submission received: 11 February 2025 / Revised: 8 April 2025 / Accepted: 10 April 2025 / Published: 14 April 2025

Abstract

:
Grossheimin, a guaiane-type sesquiterpene lactone, displayed a diverse range of biological activities, including anticancer, anti-inflammatory and antimicrobial effects. Various amino analogues of grossheimin were prepared through a Michael addition at its highly active α-methylene-γ-lactone motif. On the other hand, grossheimin was reduced to diol, which was then subjected to nucleophilic addition or acetylation to introduce heteroatoms associated with oxygen, sulfur or nitrogen functionalities. All of the synthesised Michael and acetylated adducts were evaluated for their in vitro cytotoxic action on human colon adenocarcinoma lines, including Colo205 and Colo320. The bioassay results indicated that the acetylated adducts displayed a potent cytotoxic effect compared to grossheimin, the parent molecule. A docking study was also performed to exploit the observed results.

1. Introduction

Colorectal cancer (CRC) is the third most common malignancy globally and one of the leading causes of mortality among cancer patients [1]. The global incidence of colorectal cancer in 2020 was approximately 1.9 million cases, resulting in roughly one million fatalities [2]. The current clinical treatment for CRC mainly involves surgery and chemotherapy [3]. Although 5-fluorouracil (5-FU) and oxaliplatin are used clinically for the treatment of CRC, the effects of cytotoxicity, drug resistance and adverse reactions are the main problems associated with chemotherapy [4]. Therefore, novel therapeutic strategies are constantly needed to improve the safety and effectiveness of CRC therapy.
Guaianolides, as secondary metabolites, constitute one of the largest groups of naturally occurring sesquiterpene lactones with structural complexity and a wide range of biological activities [5]. Grossheimin (1), an abundant and active guaianolide, was first isolated in Grossheimia macrocephala (Muss.-Puschk. ex Willd.) and G. ossica (C. Koch) Sosn. et Takht and also presented as a minor constituent in other asteraceous plants, such as Amberboa lipii DC., Chartolepis biebersteinii Jaub. Et Spach., Ch. glastifolia (L.) Cass., Ch. intermedia Boiss., Ch. pterocaula Trautv., Centaurea behen Linn., C. helenioides Boiss., C. lippii L., C. ornata Willd., C. ruthenica Lam., C. scabiosa L., Cynara cardunculus L., C. scolymus L., Crepis virens Vill., Venidium decurrens Less., and Youngia japonica (L.) DC [6]. Conversely, grossheimin, a major constituent from Chartolepis intermedia Boiss. (C. intermedia Boiss.) in Kazakhstan (ca. 0.1%) [7], is widely reported as a phytotoxic compound [8,9,10,11,12]. Furthermore, this compound is also receiving attention as a result of its reputed pharmacological properties beyond its function as a hypolipidemic [13] and anti-inflammatory agent [14,15]. Grossheimin has also been proven to possess anti-opisthorchiasis [16], antiobesity [17,18], antiallergic [19], antihyperlipidemic [20] and cytotoxic effects [21]. In addition, the high antitumor, antiparasitic, antioxidant and antimicrobial potential of grossheimin has been previously reported in the literature [22].
The structure–activity relationship (SAR) has indicated that the α-methylene-γ-butyrolactone moiety plays a significant role in determining the biological properties [23,24]. However, α-methylene-γ-lactone can exhibit non-selective binding as a Michael acceptor with undesired targets [25]. Therefore, a strategy employing reactions such as a Michael addition [26,27,28,29,30,31,32], Heck reaction [6,33] and others has been developed in which the reactive α, β-unsaturated enone is masked. These semi-synthetic modifications not only improve the pharmacokinetic profile but also maintain or enhance the biological activity of the parent molecule [34]. In addition to the electrophilic site, the secondary hydroxy group may serve as a nucleophilic site for halogenation [6,35] or acylation. Furthermore, Mo and co-workers recently reported that grossheimin displayed potential anticancer activity against MDA-MB-231 and colorectal cancer HCT-116 cell lines with an IC50 value of 16.86 and 30.94 μmol/L, respectively [36]. Stimulated by this result, and with the aim of finding novel biologically active grossheimin-based adducts that exert anti-cancer effects against a variety of CRC, a library of amino analogues has been prepared and studied for their cytotoxic activity on human colon adenocarcinoma lines. A docking model has also been developed for the most potent analogues.

2. Materials and Methods

2.1. Plant Materials

The air-dry aerial part (flower heads, buds and leaves) of Chartolepis intermedia Boiss. was collected in July 2023 in the vicinity of the village of Akbastau, Abay district, Karaganda region, Republic of Kazakhstan. A voucher specimen has been archived in the herbarium fund of the JSC Research and Production Center “Phytochemistry” (KG) (Karaganda, Republic of Kazakhstan).

Isolation of Grossheimin

The powdered raw material (0.2 kg) was pre-soaked for 10 min utilising a 1:1 mixture of EtOH:H2O (400 mL) followed by extraction with a laboratory ultrasonic system (HO-230.00 at an ultrasonic frequency of 22 kHz at 20–22 °C) for 90 min and a sequential extraction with CHCl3, resulting in an odorous dark green oil (3.1 g, 1.55%). The crude extract was subjected to gravity column chromatography on silica gel and eluted with a petroleum ether (PE)–EtOAc solvent system (v/v, 0:100, 100:0). Grossheimin 1 (2.4 g, 1.2%) as colourless crystals was obtained in the fraction that was purified by PE-EtOAc = 4:1. All of the physical and spectroscopic properties of grossheimin were similar to those described in the literature [6].

2.2. Synthetic Part

All of the solvents from the suppliers (Molar Chemicals Ltd., Halásztelek, Hungary; Merck Ltd., Budapest, Hungary and VWR International Ltd., Debrecen, Hungary) were dried following the standard procedures before use, while all of the reagents were obtained from commercial suppliers and used without further purification. The optical rotations were measured in MeOH at 20 °C with a Perkin-Elmer 341 polarimeter (PerkinElmer Inc., Shelton, CT, USA). The reaction progress was monitored by using analytical thin layer chromatography (TLC) on precoated silica gel GF254 plates (Macherey-Nagel GmbH & Co. KG, Düren, Germany). Column chromatography was performed using Merck silica gel 60 (0.015–0.040 mm). An HRMS flow injection analysis was performed with the Thermo Scientific Orbitrap Exploris 240 hybrid quadrupole-Orbitrap (Thermo Fischer Scientific, Waltham, MA, USA) mass spectrometer coupled to a Waters Acquity I-Class UPLC TM (Waters, Manchester, UK). The melting points were determined using a Kofler melting point apparatus (Nagema, Dresden, Germany) and are uncorrected. 1H NMR spectra were recorded at 500 MHz, while 13C NMR spectra were measured at 125 MHz at ambient temperature with a Bruker Avance NEO Ascend 500 spectrometer (Bruker Biospin, Karlsruhe, Germany). The chemical shifts are given, relative to tetramethylsilane (TMS) as an internal standard, in δ (ppm). The J values are given by Hz.
The detailed experimental process, the physical and chemical characterisation and all of the spectroscopic data (1H-, 13C-, COSY-, NOESY-, HMBC- and HSQC-NMR together with the HRMS data) of the new compounds can be found in the ESI.

2.3. Antiproliferative Assay

2.3.1. Cell Lines and Their Maintenance

The doxorubicin-sensitive Colo205 (ATCC-CCL-222) and the doxorubicin-resistant ABCB1- and LRP-expressing Colo320/MDR-LRP (ATCC-CCL-220.1) human colon adenocarcinoma cell lines were obtained from LGC Promochem in Teddington, UK. For culturing, the RPMI 1640 medium was applied and was supplemented with a 10% heat-inactivated foetal bovine serum (FBS), 2 mM L-glutamine, 1 mM Na-pyruvate and 10 mM HEPES along with nystatin and gentamicin.

2.3.2. MTT Assay

The anticancer activity of the compounds was tested in 96-well flat-bottomed microtiter plates. In this assay, the two-fold serial dilutions of the compounds (concentration range: 100–0.19 μM) were made in a 100 μL RPMI 1640 medium. Next, 1 × 104 of human colonic adenocarcinoma cells (Colo205 or Colo320) in a 100 μL RPMI 1640 medium were added to each well except for the medium control wells. The starting concentration of the solvent DMSO in the plate was 2% v/v. The samples were incubated at 37 °C for 24 h. At the end of the incubation, 20 μL of an MTT (thiazolyl blue tetrazolium bromide) solution (from a 5 mg/mL stock solution) was pipetted to each well. After incubation at 37 °C for 4 h, 100 μL of a sodium dodecyl sulfate (SDS) solution (10% SDS in 0.01 M HCl) was pipetted to each well, and the plates were kept at 37 °C overnight. The growth of the cells was determined by measuring the optical density (OD) at 540 nm (refering to 630 nm) with a Multiscan EX ELISA reader (Thermo Labsystems, Cheshire, WA, USA). The inhibition of cell growth was calculated as IC50 values, defined as the inhibitory dose that reduces the growth of the cells exposed to the tested compounds by 50%. The IC50 values and the SD of the triplicate experiments were determined using GraphPad Prism software version 5.00 for Windows with a non-linear regression curve fit (GraphPad Software, San Diego, CA, USA; www.graphpad.com). Doxorubicin (from a 2 mg/mL stock solution, Teva Pharmaceuticals, Singapore) served as a positive control. The solvent (DMSO) did not interfere with the cell growth in the tested concentrations.

2.4. Molecular Docking Method

The crystallographic structure of the protein was retrieved from the Protein Data Bank (PDB). The 2D structures of the synthesised compounds were sketched using ChemBioDraw Ultra 11.0. The docking analysis was performed by the Accelrys Discovery Studio 3.5 software.

2.4.1. Preparation of the Crystal Structures

Hydrogen atoms, added by applying several force fields (CHARMm), could lead to a steric hindrance and subsequently to a high-energy and unstable molecule. Energy minimisation—finding the most stable and lowest-energy structure and reducing H–H interactions without affecting the basic protein skeleton atoms—was performed using adopted basis minimisation. In the next step, the active site, surrounded in a 10 Å radius sphere, was determined [37].

2.4.2. Docking Study (CDocker)

In the docking study utilising the CDocker method, various conformations of the compound within the protein’s active site can be generated. The outcomes can subsequently be assessed based on both the CDocker energy and the interactions between the ligand and the active site. This approach necessitates the preparation of the crystal structure (as previously mentioned) and the design of the compounds using the Accelrys Discovery Studio protocol along with the application of a force field.
Prior to commencing this study, it is crucial to underscore the validity of the employed methodology. This is achieved by comparing the conformation of the reference compound to those generated through the applied docking technique, ensuring that the Root Mean Square Deviation (RMSD) does not exceed 3 Å.
The ADMET properties of compounds 2, 12 and 13 were assessed using ADMET descriptors in Accelrys Discovery Studio 3.5. A total of six quantitative models were employed to predict the key properties of these compounds, including aqueous solubility (solubility in water at 25 °C), blood–brain barrier (BBB) permeability, inhibition of cytochrome P450 (CYP450) 2D6, human intestinal absorption (HIA) and plasma protein binding (PPB). Additionally, a dedicated ADMET model was developed to evaluate the HIA and BBB penetration for the tested compounds. This model incorporates confidence ellipses at 95% and 99% levels within the ADMET_PSA_2D and ADMET_ALogP98 frameworks. The detailed in silico ADMET properties are provided in Table S2 and are available in the Supplementary Materials.

3. Results and Discussion

3.1. Chemical Protocols

Sesquiterpene lactone grossheimin 1 was isolated from C. intermedia on a gram scale. The presence of a hydroxy group in the structure allowed the preparation of its acetylated derivative, and this modification led to an improved activity of the obtained derivatives [38]. In this regard, acetylated derivative 2 was prepared under standard conditions [39]. The reduction of 2 with either NaBH4 or Zn (BH4)2 delivered 3 as a major product (d.r. = 3:1, based on NMR determination) [40]. The acetylation of 3 with the Ac2O/pyridine system resulted in diacetylated derivative 4 (Scheme 1).
In addition to the interest of the hydroxy group in the grossheimin structure, the α-methylene-γ-butyrolactone moiety provided opportunities for further functionalisation. The synthesis started with the introduction of the amino groups. The aza-Michael addition of 1 with benzylamine led to the formation of β-aminolactone 5 [34], which was reduced with NaBH4 to provide 6 with high stereoselectivity. Furthermore, the reaction of 1 with formamide in the presence of n-Bu4NBr and aqueous 2 M NaOH as the base in THF at 70 °C selectively gave compound 7 [41]. In order to obtain the thiol group at the β-position of the lactone ring of grossheimin, benzylmercaptan was used in the synthesis of the corresponding thiol 8. As an applied amine, the amount of base like Et3N was a critical factor for the completion of the reaction owing to its lower reactivity [31]. When grossheimin 1 was subjected to synthetic modification using benzylmercaptan catalysed by Et3N, the reaction was a completely and cleanly furnished thiol adduct 8 with a satisfactory yield. The stereoselective reduction of thiol 8 gave product 9. A simple method to obtain methoxy derivative 10 was previously described in the literature using MeONa in MeOH [41]. When this procedure was applied to 1, compound 10 was isolated as a single product with a moderate yield (Scheme 2).
In addition to amines, azoles like imidazole, benzimidazole and others were employed as Michael donors. When the conditions used to obtain 8 were applied, product 11 was not obtained. This was probably caused by the lower nucleophilicity of N-heterocycles. Our attempts ultimately led to the use of DBU as a base for successfully completing the reaction [42]. When the above-mentioned synthetic modification was applied using diverse azoles, the reactions successfully furnished the expected products with acceptable yields (Scheme 3, Table 1).
In order to investigate the effect of the stereochemistry of hydroxy groups on the Michael addition at the exocyclic double bond of α-methylene-γ-butyrolactone, the reduction of ketone function in 1 was attempted to have alcohol 12. However, the reduction of 1 with NaBH4 only led to the formation of 3 as a single product. Fortunately, the desired transformation was achieved by reacting 1 with Zn (BH4)2 [43,44]. The acetylation of 12 with acetic anhydride in pyridine provided diacetylated derivative 13 with a satisfactory yield. New functional groups including amino (6, 16), thiol (9), carbamoyl (14) and alkoxy (15) were introduced at the exomethylene lactone through the Michael addition according to the same synthetic methods as described above (Scheme 3).

3.2. Determination of Relative Configuration

Through the aza-Michael addition as well as reduction, new stereogenic centres were formed. The relative configuration of the new stereogenic centres was determined by NOESY experiments. As shown in Figure 1, clear NOE signals were observed between H-3 and H-4 together with H-9b as well as between H-10 and H-3a. Significant NOE signals were found between H-8 and H-6a together with CH3-9 protons. Therefore, the structure of grossheimin-derived amino analogues derived from grossheimin was determined as outlined in Figure 1.

3.3. Determination of Cytotoxic Effect

Since several sesquiterpene lactone-based aminoadducts exerted a significant cytotoxic action on different human cancer cell lines [34], the in vitro cytotoxic activities of the prepared grossheimin-based amino analogues were also tested on human colon adenocarcinoma lines, including Colo205 and Colo320 by MTT assay. Doxorubicin, a clinically applied anticancer agent, was used as a reference compound, and the results are summarised in Table 2.
The results indicated that grossheimin derivatives 2, 12 and 13 exhibit considerable inhibitory properties on cancer cells. Among them, the O-acetyl-substituted grossheimin derivatives showed the most pronounced cytotoxic activities comparable to those of the reference agent, doxorubicin. Compound 2 with the O-acetyl substituent showed potent inhibitory activity against colon adenocarcinoma cancers (IC50 value around 9 μM) with no cytotoxicity against fibroblasts (>100 μM), while di-O-acetyl adduct 13 exhibited modest inhibitory activities with a higher IC50 value on the CDD-19Lu fibroblast cells (29.07 μM) than on the malignant cell lines (around 8 μM). On the other hand, none of the prepared Michael-type amino analogues exerted relevant activity (IC50 > 100 μM). According to the literature, α-methylene-γ-butyrolactones are excellent conjugate acceptors and act as warheads, resulting in a covalent engagement with the target protein (e.g., the reaction with L-cysteinyl residues in proteins/enzymes) [45,46]. A modification in the α-methylene-γ-lactone motif led to the loss of function as Michael acceptors in reactions with thiol groups of bionucleophiles, whereas acetylation improved lipophilicity, enhancing the biological activity of the parent molecule.

3.4. Molecular Docking

A docking study was conducted to predict the potential target of the most biologically active compounds based on the antiproliferative result (compounds 2, 12 and 13). To achieve this purpose, a group of protein targets, such as Aurora A (PDB code: 4DEE, resolution: 2.30 Å), ALK (PDB code: 4ZJI, resolution: 1.99 Å), Pim-1 (PDB code: 4ALW, resolution: 1.92 Å) and tubulin (PDB code: 1SAO, resolution: 3.58 Å), were used as templates for the docking study using the CDocker method [47]. The results indicated that all of the tested compounds are likely to exhibit an affinity toward the active site of tubulin by forming strong bonds with several amino acids (Figure 2) [48]. The CDocker interaction energy of the tested compounds is shown in Table S1 in the Supplementary Materials. The docking results of the studied compounds show the key role of the hydroxy and acetyl groups in forming hydrogen bonding and ionic interactions with the key amino acids (Figure 2). The in silico ADMET study showed that all of the tested compounds might have good absorption properties with a low ability to penetrate the BBB (Table S2 and Figure S1 in the Supplementary Materials).
Tubulin plays a crucial role in cell division, intracellular transport and cytoskeletal stability. Cancer cells exploit tubulin dynamics to maintain uncontrolled proliferation and resistance to apoptosis. Tubulin inhibitors arrest mitosis, leading to tumor cell death [49]. Our results show that compounds 2 and 13 bind to the colchicine site of tubulin, suggesting a mechanism of microtubule destabilisation, which may offer a novel approach for colorectal cancer therapy. This study highlights compounds 2 and 13 as promising tubulin inhibitors, with interactions involving Thr179, Glu183 and Lys352. Their strong binding suggests the potential for further development in colorectal cancer therapy.

4. Conclusions

In conclusion, starting from isolated grossheimin, a new family of grossheimin-derived chiral amino analogues was prepared via stereoselective transformations. All of the compounds were screened for their cytotoxic effects on human colon adenocarcinoma lines. Only the acetylated compounds exhibited significant cytotoxicity within the series of derivatives as compared to the precursor grossheimin. Among these O-acetyl derivatives, compound 2 exerted outstanding activities against the malignant cells with no action on fibroblasts, indicating considerable cancer selectivity. The structure–activity relationship clearly indicated that functionalisation at the hydroxy group led to an enhanced effect against all of the studied cell lines, whereas the modification at the α-methylene-γ-butyrolactone moiety resulted in a total loss of activity.
The molecular docking studies proved that derivatives 2, 12 and 13 could foster a potent affinity by forming significant hydrogen bonding and ionic interactions with the key amino acids with tubulin (PDB code: 1SAO, resolution: 3.58 Å). Thus, these novel leads from grossheimin can be further developed into potential chemotherapeutic agents in colorectal cancer therapy.
In the next stage of our project, esterfication with heterocyclic carboxylic or halogenated acids as well as halogenation could be achieved and their cytotoxic activities on various human cancer cell lines investigated. Additionally, for the optimised compounds, a tubulin inhibitory activity and molecular dynamics study will also be performed to obtain insight into the dynamics of ligand interaction.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biom15040578/s1, Figure S1: Plot of Polar Surface Area (PSA) vs. LogP for a standard and test set; Table S1: Cytotoxic effects of grossheimin-based aminoanalogues on colon adenocarcinoma cell lines and CDD-19Lu fibroblasts.; Table S2: CDocker energy values for compounds 2, 12 and 13; Table S3: In-silico ADMET study.

Author Contributions

T.M.L., Z.S. and G.S. conceived and designed the experiments. S.M.A. collected and isolated grossheimin from the plant samples. M.A. and N.S. performed the experiments, analysed the data and wrote the experimental part. Z.S., T.M.L. and G.S. discussed the results and contributed to the writing of the paper. All authors have read and agreed to the published version of the manuscript.

Funding

The authors thank the Hungarian Research Foundation (OTKA No. K-138871), the Ministry of Human Capacities, Hungary grant, TKP-2021-EGA-32, the scientific and technical program BR21882180 “Development of a program for the conservation and development of the resource base of plants of Kazakhstan that are promising for medicine and veterinary medicine in a changing climate” financed by the Science Committee of the Ministry of Science and Higher Education of the Republic of Kazakhstan. G.S. was supported by the János Bolyai Research Scholarship (BO/00158/22/5) of the Hungarian Academy of Sciences. This research was funded by University of Szeged Open Access Fund (grant No. 7593).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Acknowledgments

The HR–MS analysis was performed by Róbert Berkecz.

Conflicts of Interest

Author Sergazy M. Adekenov was employed by the company JSC Research and Production Center “Phytochemistry”. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Mármol, I.; Sánchez-de-Diego, C.; Pradilla Dieste, A.; Cerrada, E.; Rodriguez Yoldi, M. Colorectal Carcinoma: A General Overview and Future Perspectives in Colorectal Cancer. Int. J. Mol. Sci. 2017, 18, 197. [Google Scholar] [CrossRef] [PubMed]
  2. Xi, Y.; Xu, P. Global Colorectal Cancer Burden in 2020 and Projections to 2040. Transl. Oncol. 2021, 14, 101174. [Google Scholar] [CrossRef] [PubMed]
  3. Huang, X.; Yang, Z.; Xie, Q.; Zhang, Z.; Zhang, H.; Ma, J. Natural Products for Treating Colorectal Cancer: A Mechanistic Review. Biomed. Pharmacother. 2019, 117, 109142. [Google Scholar] [CrossRef]
  4. Vodenkova, S.; Buchler, T.; Cervena, K.; Veskrnova, V.; Vodicka, P.; Vymetalkova, V. 5-Fluorouracil and Other Fluoropyrimidines in Colorectal Cancer: Past, Present and Future. Pharmacol. Therapeut. 2020, 206, 107447. [Google Scholar] [CrossRef] [PubMed]
  5. Yang, H.; Qiao, X.; Li, F.; Ma, H.; Xie, L.; Xu, X. Diastereoselective Total Synthesis of 8-Epigrosheimin. Tetrahedron Lett. 2009, 50, 1110–1112. [Google Scholar] [CrossRef]
  6. Adekenov, S.M.; Shaimerdenova, Z.R.; Adekenova, K.S.; Kishkentayeva, A.S. Synthesis and Biological Evaluation of New Derivatives of Grossheimin. Fitoterapia 2022, 158, 105154. [Google Scholar] [CrossRef]
  7. Kubentayev, S.A.; Alibekov, D.T.; Perezhogin, Y.V.; Lazkov, G.A.; Kupriyanov, A.N.; Ebel, A.L.; Izbastina, K.S.; Borodulina, O.V.; Kubentayeva, B.B. Revised Checklist of Endemic Vascular Plants of Kazakhstan. PhytoKeys 2024, 238, 241–279. [Google Scholar] [CrossRef]
  8. Mejías, F.J.R.; Fernández, I.P.; Rial, C.; Varela, R.M.; Molinillo, J.M.G.; Calvino, J.J.; Trasobares, S.; Macías, F.A. Encapsulation of Cynara Cardunculus Guaiane-Type Lactones in Fully Organic Nanotubes Enhances Their Phytotoxic Properties. J. Agric. Food Chem. 2022, 70, 3644–3653. [Google Scholar] [CrossRef]
  9. Scavo, A.; Rial, C.; Molinillo, J.M.G.; Varela, R.M.; Mauromicale, G.; Macías, F.A. Effect of Shading on the Sesquiterpene Lactone Content and Phytotoxicity of Cultivated Cardoon Leaf Extracts. J. Agric. Food Chem. 2020, 68, 11946–11953. [Google Scholar] [CrossRef]
  10. Scavo, A.; Rial, C.; Varela, R.M.; Molinillo, J.M.G.; Mauromicale, G.; Macias, F.A. Influence of Genotype and Harvest Time on the Cynara cardunculus L. Sesquiterpene Lactone Profile. J. Agric. Food Chem. 2019, 67, 6487–6496. [Google Scholar] [CrossRef]
  11. Rial, C.; García, B.F.; Varela, R.M.; Torres, A.; Molinillo, J.M.G.; Macías, F.A. The Joint Action of Sesquiterpene Lactones from Leaves as an Explanation for the Activity of Cynara cardunculus. J. Agric. Food Chem. 2016, 64, 6416–6424. [Google Scholar] [CrossRef] [PubMed]
  12. Rial, C.; Novaes, P.; Varela, R.M.; Molinillo, J.M.G.; Macias, F.A. Phytotoxicity of Cardoon (Cynara cardunculus) Allelochemicals on Standard Target Species and Weeds. J. Agric. Food Chem. 2014, 62, 6699–6706. [Google Scholar] [CrossRef] [PubMed]
  13. Kasenova, S.B.; Atazhanova, G.A.; Sagintaeva, Z.I.; Kasenov, B.K.; Kishkentaeva, A.S.; Adekenov, S.M. Thermodynamic Properties of Sesquiterpene Lactone Grossheimin. Russ. J. Phys. Chem. A 2016, 90, 1521–1524. [Google Scholar] [CrossRef]
  14. Matsumoto, T.; Nakashima, S.; Nakamura, S.; Hattori, Y.; Ando, T.; Matsuda, H. Inhibitory Effects of Cynaropicrin and Related Sesquiterpene Lactones from Leaves of Artichoke (Cynara scolymus L.) on Induction of iNOS in RAW264.7 Cells and Its High-Affinity Proteins. J. Nat. Med. 2021, 75, 381–392. [Google Scholar] [CrossRef]
  15. Schepetkin, I.A.; Kirpotina, L.N.; Mitchell, P.T.; Kishkentaeva, A.S.; Shaimerdenova, Z.R.; Atazhanova, G.A.; Adekenov, S.M.; Quinn, M.T. The Natural Sesquiterpene Lactones Arglabin, Grosheimin, Agracin, Parthenolide, and Estafiatin Inhibit T Cell Receptor (TCR) Activation. Phytochemistry 2018, 146, 36–46. [Google Scholar] [CrossRef]
  16. Adekenov, S.M. Study of Antiopisthorchiasis Activity of Sesquiterpene Lactones and Their Derivatives. Fitoterapia 2019, 133, 200–211. [Google Scholar] [CrossRef]
  17. Adekenova, A.S.; Sakenova, P.Y.; Ivasenko, S.A.; Khabarov, I.A.; Adekenov, S.M.; Berthod, A. Gram-Scale Purification of Two Sesquiterpene Lactones from Chartolepsis Intermedia Boiss. Chromatographia 2016, 79, 37–43. [Google Scholar] [CrossRef]
  18. Grothe, T.; Roemer, E.; Wabnitz, P. Use of Tricyclic Sesquiterpene Lactones in the Treatment of Obesity and Related Diseases and Non-Therapeutic Treatable Conditions 2020. U.S. Patent Application No. 13/522,110, 21 July 2011. [Google Scholar]
  19. Yae, E.; Yahara, S.; El-Aasr, M.; Ikeda, T.; Yoshimitsu, H.; Masuoka, C.; Ono, M.; Hide, I.; Nakata, Y.; Nohara, T. Studies on the Constituents of Whole Plants of Youngia japonica. Chem. Pharm. Bull. 2009, 57, 719–723. [Google Scholar] [CrossRef]
  20. Shimoda, H.; Ninomiya, K.; Nishida, N.; Yoshino, T.; Morikawa, T.; Matsuda, H.; Yoshikawa, M. Anti-Hyperlipidemic Sesquiterpenes and New Sesquiterpene Glycosides from the Leaves of Artichoke (Cynara scolymus L.): Structure Requirement and Mode of Action. Bioorg. Med. Chem. Lett. 2003, 13, 223–228. [Google Scholar] [CrossRef]
  21. Zhao, Q.-S.; Li, X.; Qian, P.; Liu, Z.; Zhao, Y.; Xu, G.; Tao, D.; Sun, H. The Sesquiterpenoids from Cynara scolymus. Heterocycles 2005, 65, 287–291. [Google Scholar] [CrossRef]
  22. Mukhametzhanova, G.; Asanova, G.; Adekenova, G.S.; Medeubayeva, B.; Kishkentayeva, A.; Adekenov, S. Chartolepis intermedia Boiss. and Centaurea ruthenica Lam.—New Medicina Plants Containing Pharmacologically Active Compounds. Open Access Maced. J. Med. Sci. 2022, 10, 56–64. [Google Scholar] [CrossRef]
  23. Moujir, L.; Callies, O.; Sousa, P.M.C.; Sharopov, F.; Seca, A.M.L. Applications of Sesquiterpene Lactones: A Review of Some Potential Success Cases. Appl. Sci. 2020, 10, 3001. [Google Scholar] [CrossRef]
  24. Fernandes, R.A.; Moharana, S.; Khatun, G.N. Recent Advances in the Syntheses of Guaianolides. Org. Biomol. Chem. 2023, 21, 6652–6670. [Google Scholar] [CrossRef]
  25. Ghantous, A.; Gali-Muhtasib, H.; Vuorela, H.; Saliba, N.A.; Darwiche, N. What Made Sesquiterpene Lactones Reach Cancer Clinical Trials? Drug Discov. Today 2010, 15, 668–678. [Google Scholar] [CrossRef] [PubMed]
  26. Adekenov, S.M. Natural Sesquiterpene Lactones as Renewable Chemical Materials for New Medicinal Products. Eurasian Chem.-Technol. J. 2013, 15, 163–174. [Google Scholar] [CrossRef]
  27. Adekenov, S.M.; Kishkentaeva, A.S.; Shaimerdenova, Z.R.; Atazhanova, G.A. Bimolecular Compounds Based on Natural Metabolites. Chem. Nat. Compd. 2018, 54, 464–470. [Google Scholar] [CrossRef]
  28. Kulyyasov, A.T.; Adekenov, S.M.; Seitembetov, T.S.; Rakhimov, K.D. Chemical Modification of Grosshemin in the Lactone Ring. Chem. Nat. Compd. 1995, 31, 192–195. [Google Scholar] [CrossRef]
  29. Rybalko, K.S.; Evstratova, R.I.; Sheichenko, V.I. Production of a Pyridine Derivative of Grosshemin. Chem. Nat. Compd. 1976, 12, 272–274. [Google Scholar] [CrossRef]
  30. Jalmakhanbetova, R.I.; Elkaeed, E.B.; Eissa, I.H.; Metwaly, A.M.; Suleimen, Y.M. Synthesis and Molecular Docking of Some Grossgemin Amino Derivatives as Tubulin Inhibitors Targeting Colchicine Binding Site. J. Chem. 2021, 2021, 5586515. [Google Scholar] [CrossRef]
  31. Fardella, G.; Barbetti, P.; Grandolini, G.; Chiappini, I.; Ambrogi, V.; Scarcia, V.; Furlani Candiani, A. Phenylthio-Derivatives of α-Methylene-γ-Lactones as pro-Drugs of Cytotoxic Agents§. Eur. J. Med. Chem. 1999, 34, 515–523. [Google Scholar] [CrossRef]
  32. Barbetti, P.; Fardella, G.; Chiappini, I.; Scarcia, V.; Furlani Candiani, A. New Cytotoxic Selenoderivatives of Guaianolides. Eur. J. Med. Chem. 1989, 24, 299–305. [Google Scholar] [CrossRef]
  33. Adekenov, S.M.; Kishkentayeva, A.S.; Khasenova, A.B.; Shul’ts, E.E.; Gatilov, Y.V.; Bagryanskaya, I.Y. New Arylhalo-Derivatives of Grosshemin. Chem. Nat. Compd. 2021, 57, 685–690. [Google Scholar] [CrossRef]
  34. Woods, J.R.; Mo, H.; Bieberich, A.A.; Alavanja, T.; Colby, D.A. Amino-Derivatives of the Sesquiterpene Lactone Class of Natural Products as Prodrugs. Med. Chem. Commun. 2013, 4, 27–33. [Google Scholar] [CrossRef]
  35. Ivasenko, S.A.; Kulyyasov, A.T.; Adekenov, S.M. Synthesis of a New Chloroderivative of Grosshemin Guaianolide. Chem. Nat. Compd. 2003, 39, 601–602. [Google Scholar] [CrossRef]
  36. Liya, M.; Weiling, L.; Jialu, Q.; Qian, P.; Xiansheng, Y.; Binlian, S.; Kanghong, H. Chemical Composition of Youngia japonica and Its Antitumor Activity. Biot. Resour. 2023, 45, 267–275. [Google Scholar] [CrossRef]
  37. Rampogu, S.; Baek, A.; Son, M.; Park, C.; Yoon, S.; Parate, S.; Lee, K.W. Discovery of Lonafarnib-Like Compounds: Pharmacophore Modeling and Molecular Dynamics Studies. ACS Omega 2020, 5, 1773–1781. [Google Scholar] [CrossRef]
  38. Khlebnikov, A.I.; Schepetkin, I.A.; Kishkentaeva, A.S.; Shaimerdenova, Z.R.; Atazhanova, G.A.; Adekenov, S.M.; Kirpotina, L.N.; Quinn, M.T. Inhibition of T Cell Receptor Activation by Semi-Synthetic Sesquiterpene Lactone Derivatives and Molecular Modeling of Their Interaction with Glutathione and Tyrosine Kinase ZAP-70. Molecules 2019, 24, 350. [Google Scholar] [CrossRef]
  39. Yayli, N.; Baltaci, C.; Gök, Y.; Aydin, E.; Üçüncü, O. Sesquiterpene Lactones from Centaurea helenioides Boiss. Turk. J. Chem. 2006, 30, 229–233. [Google Scholar]
  40. Scotti, L.; Scotti, M.; Ishiki, H.; Ferreira, M.; Emerenciano, V.; Desmenezes, C.; Ferreira, E. Quantitative Elucidation of the Structure–Bitterness Relationship of Cynaropicrin and Grosheimin Derivatives. Food Chem. 2007, 105, 77–83. [Google Scholar] [CrossRef]
  41. Cala, A.; Zorrilla, J.G.; Rial, C.; Molinillo, J.M.G.; Varela, R.M.; Macías, F.A. Easy Access to Alkoxy, Amino, Carbamoyl, Hydroxy, and Thiol Derivatives of Sesquiterpene Lactones and Evaluation of Their Bioactivity on Parasitic Weeds. J. Agric. Food Chem. 2019, 67, 10764–10773. [Google Scholar] [CrossRef]
  42. Lone, S.H.; Bhat, K.A.; Shakeel-u-Rehman; Majeed, R.; Hamid, A.; Khuroo, M.A. Synthesis and Biological Evaluation of Amino Analogs of Ludartin: Potent and Selective Cytotoxic Agents. Bioorg. Med. Chem. Lett. 2013, 23, 4931–4934. [Google Scholar] [CrossRef] [PubMed]
  43. Elsebai, M.F. Sesquiterpene Lactones as Potent and Broad Spectrum Antiviral Compounds Against All Genotypes of Hepatitis c Virus (Hcv). Patent WO-2016169573-A1, 20 April 2015. Available online: https://pubchem.ncbi.nlm.nih.gov/patent/WO-2016169573-A1 (accessed on 5 October 2024).
  44. Hegazy, M.-E.; Ibrahim, A.; Mohamed, T.; Shahat, A.; El Halawany, A.; Abdel-Azim, N.; Alsaid, M.; Paré, P. Sesquiterpene Lactones from Cynara cornigera: Acetyl Cholinesterase Inhibition and In Silico Ligand Docking. Planta Med. 2015, 82, 138–146. [Google Scholar] [CrossRef] [PubMed]
  45. Kitson, R.R.A.; Millemaggi, A.; Taylor, R.J.K. The Renaissance of α-Methylene-γ-butyrolactones: New Synthetic Approaches. Angew. Chem. Int. Ed. 2009, 48, 9426–9451. [Google Scholar] [CrossRef] [PubMed]
  46. Liu, W.; Winssinger, N. Synthesis of α-Exo-Methylene-γ-Butyrolactones: Recent Developments and Applications in Natural Product Synthesis. Synthesis 2021, 53, 3977–3990. [Google Scholar] [CrossRef]
  47. Khdar, Z.A.; Le, T.M.; Schelz, Z.; Zupkó, I.; Szakonyi, Z. Stereoselective Synthesis and Application of Gibberellic Acid-Derived Aminodiols. Int. J. Mol. Sci. 2022, 23, 10366. [Google Scholar] [CrossRef]
  48. Ravelli, R.B.G.; Gigant, B.; Curmi, P.A.; Jourdain, I.; Lachkar, S.; Sobel, A.; Knossow, M. Insight into Tubulin Regulation from a Complex with Colchicine and a Stathmin-like Domain. Nature 2004, 428, 198–202. [Google Scholar] [CrossRef]
  49. Jordan, M.A.; Wilson, L. Microtubules as a Target for Anticancer Drugs. Nat. Rev. Cancer 2004, 4, 253–265. [Google Scholar] [CrossRef]
Figure 1. Determination of relative configuration of grossheimin-based new aminoanalogues.
Figure 1. Determination of relative configuration of grossheimin-based new aminoanalogues.
Biomolecules 15 00578 g001
Figure 2. A 2D diagram of the interactions between compound 2 (A), compound 12 (B) and compound 13 (C) with the tubulin binding site and the crystal structure of tubulin (D).
Figure 2. A 2D diagram of the interactions between compound 2 (A), compound 12 (B) and compound 13 (C) with the tubulin binding site and the crystal structure of tubulin (D).
Biomolecules 15 00578 g002
Scheme 1. Grossheimin-based acetylated derivatives. Conditions: (i) Ac2O (20 eq.), dry pyridine, 25 °C, 24 h, 76% (2), 38% (4); (ii) NaBH4 (3 eq.), DCM:MeOH (1:1), 0 °C, 1 h, 56%.
Scheme 1. Grossheimin-based acetylated derivatives. Conditions: (i) Ac2O (20 eq.), dry pyridine, 25 °C, 24 h, 76% (2), 38% (4); (ii) NaBH4 (3 eq.), DCM:MeOH (1:1), 0 °C, 1 h, 56%.
Biomolecules 15 00578 sch001
Scheme 2. Grosshemin-based amino/thio/carbamoyl/alkoxy adducts. Conditions: (i) BnNH2 (2 eq.), dry EtOH, 25 °C, 24 h, 68%; (ii) NaBH4 (3 eq.), DCM:MeOH (1:1), 0 °C, 1 h, 43% (6), 47% (9); (iii) formamide (46 eq.), n-Bu4NBr (0.4 eq.), THF, 2 M NaOH, 70 °C, 24 h, 28%; (iv) BnSH (2 eq.), Et3N (1 eq.), dry EtOH, 25 °C, 24 h, 66%; (v) MeONa (3 eq.), dry MeOH, 25 °C, 48 h, 53%; (vi) azoles (2 eq.), DBU (1 eq.), dry ACN, 25 °C, 24 h, 34–67%.
Scheme 2. Grosshemin-based amino/thio/carbamoyl/alkoxy adducts. Conditions: (i) BnNH2 (2 eq.), dry EtOH, 25 °C, 24 h, 68%; (ii) NaBH4 (3 eq.), DCM:MeOH (1:1), 0 °C, 1 h, 43% (6), 47% (9); (iii) formamide (46 eq.), n-Bu4NBr (0.4 eq.), THF, 2 M NaOH, 70 °C, 24 h, 28%; (iv) BnSH (2 eq.), Et3N (1 eq.), dry EtOH, 25 °C, 24 h, 66%; (v) MeONa (3 eq.), dry MeOH, 25 °C, 48 h, 53%; (vi) azoles (2 eq.), DBU (1 eq.), dry ACN, 25 °C, 24 h, 34–67%.
Biomolecules 15 00578 sch002
Scheme 3. Grossheimin-derived amino/thiol/carbamoyl/alkoxy analogues. Conditions: (i) Zn(BH4)2 (2 eq.), dry ACN, 25 °C, 6 h, 81%; (ii) Ac2O (20 eq.), dry pyridine, 25 °C, 24 h, 49%; (iii) BnNH2 (2 eq.), dry EtOH, 25 °C, 24 h, 68%; (iv) formamide (46 eq.), n-Bu4NBr (0.4 eq.), THF, 2 M NaOH, 70 °C, 24 h, 31%; (v) BnSH (2 eq.), Et3N (1 eq.), dry EtOH, 25 °C, 24 h, 66%; (vi) MeONa (3 eq.), dry MeOH, 25 °C, 48 h, 33%; (vii) azoles (2 eq.), DBU (1 eq.), dry ACN, 25 °C, 24 h, 32–62%.
Scheme 3. Grossheimin-derived amino/thiol/carbamoyl/alkoxy analogues. Conditions: (i) Zn(BH4)2 (2 eq.), dry ACN, 25 °C, 6 h, 81%; (ii) Ac2O (20 eq.), dry pyridine, 25 °C, 24 h, 49%; (iii) BnNH2 (2 eq.), dry EtOH, 25 °C, 24 h, 68%; (iv) formamide (46 eq.), n-Bu4NBr (0.4 eq.), THF, 2 M NaOH, 70 °C, 24 h, 31%; (v) BnSH (2 eq.), Et3N (1 eq.), dry EtOH, 25 °C, 24 h, 66%; (vi) MeONa (3 eq.), dry MeOH, 25 °C, 48 h, 33%; (vii) azoles (2 eq.), DBU (1 eq.), dry ACN, 25 °C, 24 h, 32–62%.
Biomolecules 15 00578 sch003
Table 1. Grossheimin-based azoles 11a–h.
Table 1. Grossheimin-based azoles 11a–h.
EntryCompoundRYield (%)
111aImidazole53
211bPyrazole42
311c1,2,4-Triazole67
411d1,2,3-Triazole66
511eIndole35
611fBenzimidazole50
711gIndazole64
811hBenzotriazole34
Table 2. The cytotoxic effects of grossheimin-based aminoanalogues on colon adenocarcinoma cell lines and CDD-19Lu fibroblasts. Doxorubicin (DOX) was applied as a positive control.
Table 2. The cytotoxic effects of grossheimin-based aminoanalogues on colon adenocarcinoma cell lines and CDD-19Lu fibroblasts. Doxorubicin (DOX) was applied as a positive control.
CompoundIC50 (μM)
Colo205Colo320CDD-19Lu
123.33 ± 2.0527.89 ± 2.00>100
28.21 ± 0.519.44 ± 0.41>100
1263.79 ± 1.3253.65 ± 1.60>100
138.21 ± 0.516.98 ± 0.2929.07 ± 0.46
DOX3.21 ± 0.034.36 ± 0.08>8.62
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

Ashimbayeva, M.; Szakonyi, Z.; Adekenov, S.M.; Szemerédi, N.; Spengler, G.; Le, T.M. Synthesis and Antiproliferative Effects of Grossheimin-Derived Aminoanalogues. Biomolecules 2025, 15, 578. https://doi.org/10.3390/biom15040578

AMA Style

Ashimbayeva M, Szakonyi Z, Adekenov SM, Szemerédi N, Spengler G, Le TM. Synthesis and Antiproliferative Effects of Grossheimin-Derived Aminoanalogues. Biomolecules. 2025; 15(4):578. https://doi.org/10.3390/biom15040578

Chicago/Turabian Style

Ashimbayeva, Meruyert, Zsolt Szakonyi, Sergazy M. Adekenov, Nikoletta Szemerédi, Gabriella Spengler, and Tam Minh Le. 2025. "Synthesis and Antiproliferative Effects of Grossheimin-Derived Aminoanalogues" Biomolecules 15, no. 4: 578. https://doi.org/10.3390/biom15040578

APA Style

Ashimbayeva, M., Szakonyi, Z., Adekenov, S. M., Szemerédi, N., Spengler, G., & Le, T. M. (2025). Synthesis and Antiproliferative Effects of Grossheimin-Derived Aminoanalogues. Biomolecules, 15(4), 578. https://doi.org/10.3390/biom15040578

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop