Next Article in Journal
The Cd/Zn Axis: Emerging Concepts in Cellular Fate and Cytotoxicity
Previous Article in Journal
Recent Progress on Circular RNAs in the Development of Skeletal Muscle and Adipose Tissues of Farm Animals
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Escin’s Multifaceted Therapeutic Profile in Treatment and Post-Treatment of Various Cancers: A Comprehensive Review

by
Sunnatullo Fazliev
1,2,
Khurshid Tursunov
3,4,
Jamoliddin Razzokov
5,6,7,8 and
Avez Sharipov
3,9,*
1
Max Planck School Matter to Life, Jahnstrasse 29, 69120 Heidelberg, Germany
2
Faculty of Engineering Sciences, Heidelberg University, Im Neuenheimer Feld 205, 69120 Heidelberg, Germany
3
Department of Inorganic, Physical and Colloidal Chemistry, Tashkent Pharmaceutical Institute, Oybek Street 45, Tashkent 100015, Uzbekistan
4
State Center for Expertise and Standardization of Medicines, Medical Devices and Medical Equipment, Agency for the Development of the Pharmaceutical Industry under the Ministry of Health of the Republic of Uzbekistan, Ozod Street 16, Tashkent 100002, Uzbekistan
5
Institute of Fundamental and Applied Research, National Research University TIIAME, Kori Niyoziy 39, Tashkent 100000, Uzbekistan
6
College of Engineering, Akfa University, Milliy Bog Street 264, Tashkent 111221, Uzbekistan
7
Department of Physics, National University of Uzbekistan, Universitet 4, Tashkent 100174, Uzbekistan
8
Laboratory of Experimental Biophysics, Centre for Advanced Technologies, Universitet 7, Tashkent 100174, Uzbekistan
9
Department of Analytical and Pharmaceutical Chemistry, Institute of Pharmaceutical Education and Research, Yunusota Street 46, Tashkent 100114, Uzbekistan
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(2), 315; https://doi.org/10.3390/biom13020315
Submission received: 4 December 2022 / Revised: 30 January 2023 / Accepted: 3 February 2023 / Published: 7 February 2023
(This article belongs to the Section Natural and Bio-inspired Molecules)

Abstract

:
Although modern medicine is advancing at an unprecedented rate, basic challenges in cancer treatment and drug resistance remain. Exploiting natural-product-based drugs is a strategy that has been proven over time to provide diverse and efficient approaches in patient care during treatment and post-treatment periods of various diseases, including cancer. Escin—a plant-derived triterpenoid saponin—is one example of natural products with a broad therapeutic scope. Initially, escin was proven to manifest potent anti-inflammatory and anti-oedematous effects. However, in the last two decades, other novel activities of escin relevant to cancer treatment have been reported. Recent studies demonstrated escin’s efficacy in compositions with other approved drugs to accomplish synergy and increased bioavailability to broaden their apoptotic, anti-metastasis, and anti-angiogenetic effects. Here, we comprehensively discuss and present an overview of escin’s chemistry and bioavailability, and highlight its biological activities against various cancer types. We conclude the review by presenting possible future directions of research involving escin for medical and pharmaceutical applications as well as for basic research.

1. Introduction

Advances in medicine and pharmacology allow us to diagnose, prevent and treat many diseases that hamper the quality of life. These achievements are partially realised thanks to developments in molecular biology, genetics and drug discovery. However, several basic challenges, like drug resistance and cancer treatment, continue to pose a predicament. Although drug design is progressing through computational, machine learning and automation methods [1,2], natural products remain a cornerstone in pharmaceutical science. Metabolites of microorganisms, plants and animals are rich sources of valuable chemicals that demonstrate potent activities against a broad range of diseases. Cancer, as a bottleneck of medicine, is a leading cause of death without differentiating between wealthy and poor countries. Worldwide, it takes the lives of approximately 10 million people per annum [3]. This disease is characterized by abnormal growth of cells and results in very serious complications, poor quality of life and shortened life expectancy. Cancer diagnosis and treatment are expensive and difficult, and not always possible. Medicinal plants offer cheap, readily available sources of active pharmaceutical compounds (APCs) against many diseases. Approximately 25% of all the newly approved FDA drugs between 1981 and 2019 are considered to be natural products or derived from natural products [4]. In this time frame, FDA approved 182 new drugs against cancer, 120 of which were natural product-based drugs [4]. It is remarkable how APCs of plant origin can be used to combat serious diseases like cancer in a variety of ways: ranging from diagnosis to treatment to post-treatment care.
One group of multifaceted APCs of plant origin is escin. These are triterpenoid saponins usually isolated from the seeds of Horse chestnut trees. As we shall show below, more than 30 escin isomers have been isolated and studied. Originally, escin was proven to be efficient against inflammation, chronic venous insufficiency and gastroprotection. But over the past two decades, numerous novel activities of escin have been published, such as anti-oxidative and anti-cancer effects. These studies demonstrated that escin has anti-proliferative and anti-metastasis effects against more than 15 different cancer types. In addition, a new trend—composite drugs of escin and other APCs—is on the rise. Therefore, escin’s broad scope of biological activities combined with various composite drugs makes it appealing for use in cancer treatment and post-treatment care.
Although there are reviews of escin beginning in the 2000s, all of these reviews focus on the most common biological activities of escin; few have presented the chemistry and bioavailability of escin. Therefore, we sought to comprehensively review the chemistry, pharmacology and pharmaceutics of escin by extensively discussing and presenting a big picture of escin’s role in the treatment of cancer, as well as providing an updated resource with recent findings.
In literature, two types of escin, α- and β-escin, are often reported and they correspond to escin Ia and escin Ib, respectively [5]. Pharmacologically relevant escin is β-escin, which is less soluble in water. There are some disparities in the nomenclatural use of the word “escin”. Generally, the terms aescin and escin are equivalent. However, there are literature sources that refer to these terms differently; some refer to it as a mixture of 2, 4 [6,7] or many saponins [8,9,10,11,12]. The European Medicines Agency defines escin as a mixture of saponins with Mr = 1131 g/mol [13]. This corresponds to a mixture of escin Ia, Ib, IVc, IVd and VIb and isoescin Ia, Ib and VIIa. These discrepancies could be the result of various isolation methods that influence the composition of the final extracted product. In the context of pharmacology and pharmaceutics, we advocate using the terms “aescin” or “escin” to refer to a mixture of many saponins (the exact number is not defined; we shall show 34 escin isomers in Table 1) whose aglycone substructure is either that of protoascigenin or barrigenol C. In the course of this review, we shall use this nomenclature.

2. Chemistry of Escin

The chemical structure of escin is made up of two parts: (i) the aglycone part as a derivative of protoascigenin and (ii) the glycone part. β-d-glucouronopyranosyl acid, β-d-glucopyranose, β-d-galactopyranose and β-d-xylopyranose constitute the glycone part of escin (Figure 1).
The aglycone substructure of escin has three or four OH groups at C21, C22, C23 and C28 and thus can produce many isomers by forming various esters with acetic, angelic and tiglic acids (Table 1). The OH group at C3 escin is connected via a glycoside bond to β-d-glucouronopyranosyl acid, which in turn is connected to a β-d-glucopyranosyl unit (1→4). These two sugars are present in all escins. The third sugar glycosylates the β-D-glucouronopyranosyl acid and, depending on the escin isomer, can be one of the following moieties: β-d-glucopyranosyl (1→2), β-d-galactopyranosyl (1→2) and β-d-xylopyranosyl (1→2) (Table 1).
Isolated escin is colourless fine crystals or white powder. Typical melting temperatures of 187–230 °C are observed. The aglycone part of escin is relatively stable; even at temperatures as high as 150 °C, the saponins do not undergo denaturation. However, under certain conditions, deacetylation might occur by producing deacetylescin (Table 1). For example, alkaline hydrolysis of escin liberates acetic, angelic and/or tiglic acids resulting in the respective deacetylescin. Further methanolysis of deacetylescin breaks the glycoside bond, and the respective aglycones without acidic moieties are formed. Escin isomers can be differentiated by determining melting points and infrared, nuclear magnetic resonance and mass spectra.

3. Pharmacokinetics and Bioavailability of Escin

The bioavailability of APCs is closely related to their structure, absorption in the body, passage through barriers and metabolism. The pharmacokinetics and bioavailability of escin have been studied extensively. Due to the fact that escin was known in traditional medicine and has been used for hundreds of years, early reports of isolation, discovery and structural studies were published concurrently with pharmacokinetic studies. Scientists were eager to identify the essential pharmacological components of escin, which is a complex mixture of saponins. As mentioned in earlier sections, different isomers of escin demonstrate unique biological activities. Pharmacokinetic studies also shed light on distinct behaviours of escin isomers in terms of bioavailability and kinetics.
Wu and colleagues systematically studied the pharmacokinetics of various escin isomers in animals and were able to obtain valuable information about the relationship between the structure and pharmacokinetic properties of the isomers. It was shown that escin Ib and isoescin Ib, after intake, were converted into escin Ia and isoescin Ia, respectively [20]. Based on this phenomenon, they concluded that escin isomers with a tigeloyl moiety (present in escin Ia and isoescin Ia) are better able to permeate into the intracellular space than escin isomers with the angeloyl moiety (present in escin Ib and isoescin Ib, also see Table 1). They also found that preparations containing several escin isomers have a longer duration of action than single, pure isomers of escin [21].
Several studies demonstrated very high bioavailability (>90% compared to reference substances) of escin irrespective of the form of medicine, i.e., extracts or tablets [22,23]. Nonetheless, there are assumptions that circadian rhythms and food can affect escin’s bioavailability [22,23]. Escin reaches its maximal plasma concentration about 2 h after the first dose and circulates in blood with a half-life of t1/2 = 6–8 h. Multiple dose studies (up to a week) with escin tablets showed that its pharmacokinetics does not change over time [24]. Some soft dosage forms of escin, especially hydrophilic gels, showed enhanced bioavailability and prolonged release. For instance, acetylated, annealed starch [25] and polyacrylate cross-linked hydrophilic polymer [26] positively contributed to the prolonged release of escin from such gels. On the contrary, it has been observed that a hydrogel made of methylcellulose, which is hydrophobic polymer, released escin too quickly [25]. Another emerging drug delivery approach—encapsulating drugs into liposomes—has already been applied to escin, and the results demonstrated that such liposomes increased escin’s bioavailability several times [27].

4. Escin’s Relevant Biological Activities in Cancer Therapy

4.1. Anti-Cancer Effects

It has long been known that escin has potential anti-cancer effects. Escin’s anti-cancer mechanisms can be grouped into three categories: (i) inducing apoptosis, (ii) reducing cell proliferation and (iii) inhibiting metastasis. Over the past two decades, the results of numerous in vitro and in vivo studies revealed the efficacy of escin in suppressing and/or preventing over 15 cancer types, which are summarised in Table 2.
Regarding the molecular mechanisms of escin-induced apoptosis of cancer cells, ample evidence has been compiled to support the mitochondria-mediated (intrinsic) apoptosis pathway and reactive oxygen species (ROS)-induced DNA damage (Figure 2). Nevertheless, there are reports of death-receptor-induced (extrinsic) apoptosis induced by escin [29,48].
The intrinsic apoptosis pathway is a mitochondria-actuated event at which various stimuli decrease membrane potential and hence increase the permeability of the mitochondrial membrane [60]. Consequently, several enzymes and molecules are released from mitochondria to activate the caspase-dependent mitochondrial apoptosis. It is believed that escin disrupts several pathways leading to the mitochondrial depolarisation. This includes the inhibition of NF–κB, which results in the downregulation of anti-apoptotic Bcl family proteins [55,58,61] and ROS/p38 MAPK pathway [52] (Figure 2). Elevated caspase protein levels corroborate this mechanistic explanation (Table 2). Meanwhile, ROS can give rise to cancer cell death through DNA damage [32,37,38].
Molecular mechanisms of the anti-proliferative effects of escin mostly depend upon the inhibition of the NF–κB, JAK/STAT and ERK1/2 pathways. Here, the NF–κB pathway, which is the central target of escin’s anti-inflammatory effects, is believed to be one of the key linkages between inflammation and cancer [62]. Apart from mediating the proinflammatory [63] and anti-apoptotic [61] genes, NF–κB also targets the genes of cell proliferation regulators such as cyclins and cytokines [64] (Figure 2). Moreover, NF–κB contributes to cancer metastasis and invasion through the upregulation of adhesion molecules [64] and chemokines [47,57]. Therefore, NF–κB inhibition is the main contribution to the anti-cancer, as well as anti-inflammatory effects of escin. In addition, ERK1/2, JAK/STAT and Akt proteins and their signalling pathways were also shown to be affected by escin to inhibit cell proliferation. At a cellular level and depending on cancer type, escin-treated cancer cells were caught in cell cycle arrests at the G0/G1, G1/S and G2/M cell cycle phases (Table 2).
Anti-metastasis effects of escin, albeit being less investigated, seem to encompass a wide variety of mechanisms. Many reported the downregulation of NF–κB and downstream molecules of NF–κB signalling pathways, like vascular epidermal growth factor (VEGF), IL-8 [57], TNF [47] and Bcl family proteins [6]. Others advocated contributions from Akt and ERK1/2 [41,51]. Meanwhile, more recent studies communicated that escin inhibits metastasis by regulating the tumour microenvironment, for instance, by blocking ECM production, inhibiting hypoxia-inducible factor 1-alpha (HIF1α) targeted protein expression [54] and downregulating inducible nitric oxide synthase (iNOS) [48], RhoA and Rock proteins [49]. Escin’s regulation of the tumour microenvironment is evidenced by the downregulation of several matrix metalloproteinases (MMPs) [31,65]. Escin also displays potent anti-angiogenetic effects to prevent tumour metastasis, which will be discussed next.

4.2. Anti-Angiogenetic Effects

Escin’s anti-angiogenetic effects are also of interest, especially in the context of cancer treatment. Several in vitro and in vivo studies have indicated escin’s efficacy in the inhibition of tumour invasion, migration and metastasis (Table 2). It was demonstrated that escin inhibits proliferation [66] and migration of HUVECs [67]. The exact mechanisms by which escin alters HUVECs proliferation and migration remain elusive, but it was suggested that escin acts on HUVECs through Akt, p38/MAPK and ERK signalling pathways and by inhibition of PKC-α, EFNB2 and growth factor expression [66,67]. Escin’s anti-angiogenic effects are also facilitated by disrupting the extracellular matrix (ECM) modelling and adhesion. Reports showed escin to inhibit the secretion of vascular epidermal growth factor (VEGF) in HUVECs [31], matrix metalloproteinases (MMP-3 and MMP-9) in rats [68] and ECM production in mesothelial cells and fibroblasts [54]. Because of the anti-inflammatory, anti-oedematous and venotonic effects, scientists were quick to consider escin for the treatment of widespread chronic venous insufficiency (CVI). Escin’s efficacy against CVI has been tested by numerous in vitro, in vivo and clinical trials [69,70]. Nowadays, standardised horse-chestnut dry extract is used at various stages of CVI, varicose and other venous disorders associated with oedema [13].

4.3. Anti-Inflammatory Effects

Inflammation is a basic immune response to infection and injury that sets complex defence signalling pathways on “fire”. When out of control, inflammation can be chronic, which is also associated with tumour development [64]. Escin’s potential effect in reducing inflammation was already shown in the 1960s [71]. Extensive research on this topic proposed several key players of inflammatory networks as the targets of escin’s anti-inflammatory properties. There are numerous reports indicating that escin supresses the activation of nuclear factor kappa B (NF–κB) and inflammation processes initiated by this protein. This is substantiated by in vitro and in vivo studies where escin decreases the level of proinflammatory cytokines [72,73,74,75,76,77], tumour necrosis factor α (TNF-α), and interleukins IL-1β and IL-6, whose production is directly regulated by NF–κB as a transcription factor. Results of many studies with proinflammatory factors (TNFα, IL-1β), Toll-receptor ligands (lipopolysaccharides) and 11-β-hydroxysteroid dehydrogenase type 2 (11β-HSD2) put forward a glucocorticoid-like (GC) mechanism of escin in the regulation of inflammation (Figure 3).
Initially, it was shown that escin upregulates adrenocorticotropin and corticosterone levels [78]. Hence, anti-inflammatory effects of escin were accredited to increased levels of GCs [7]. However, following results indicated that escin’s anti-inflammatory effects are realised through upregulation of glucocorticoid receptor (GR) and induction of the conformation change in the receptor to facilitate GC binding [75,77,79,80]. This conformational change is necessary to expose GR to nucleoporins and importins that transfer the GC-bound GR into the nucleus. After, the GR can be recruited to indirect binding sites in DNA by interacting with DNA-TF complexes [81]. These interactions between GR and DNA-TF complexes directly influence gene transcription by changing DNA–TF interactions and/or by recruiting other TFs and cofactors that can result in suppression of proinflammatory factors like TNFα, IL-1β, and IL-6 (Figure 3).
Meanwhile, GC-bound GRs can inhibit the activation of NF–κB in the cytosol. Studies indicate that escin itself can inhibit the activation of NF–κB by downregulating the expression of p65 [57,75] and by inhibiting the IKK complex (comprised of the IKKα and IKKβ catalytic subunits and the IKKγ regulatory subunit) activation [47]. P65 is a necessary component of the p50/p65 dimer that is a primary target of the canonical NF–κB activation pathway [64]. There are other findings that report the inhibition of NF–κB by escin in mice [82,83], rats [75,84], human bladder cancer cells [28], human endothelial cells [85], and human pancreatic cancer cells (BxPC-3, PANC-1) [55,57]. However, the exact mechanisms of direct NF–κB inhibition by escin still remain unresolved. Decreased TNF-α, IL-1β, IL-6, and NO levels and increased antioxidant factors are ascribed as plausible explanations for escin-induced NF–κB inhibition [7,72,74,86].

4.4. Antioxidant, Protective and Ameliorative Effects

In general, escin’s potent anti-inflammatory and antioxidant activities prompted its evaluation for alleviating pain and complications in several diseases and might be very useful in chemotherapy and surgical treatment of cancer diseases. This is very crucial for healthy cells to survive detrimental conditions of cancer chemotherapies and act as a scavenger of ROS whose levels are elevated in tumour microenvironments. For instance, escin increased glutathione, catalase and SOD activities, decreased MMP-9 levels and mitigated cardiac autonomic neuropathy in rats with diabetes induced by streptozotocin [87]. The same antioxidant effects of escin (increased catalase and SOD activities) appear to be useful against cyclophosphamide-induced cardiotoxicity [88]. Escin’s antioxidant mechanisms can also be realised via the AKT-Nrf2 signalling pathway, as shown in studies of H2O2-induced cytotoxicity in established retinal pigment epithelium (ARPE-19) and primary murine RPE cells [89].
Escin also displays promising effects in brain injuries and neurodegenerative diseases - a hot topic of contemporary medicine. Escin attenuated cerebral ischemia-reperfusion injury in rats by reducing the volume of cerebral infarct and water content, and by ameliorating the neurological deficit [90]. Likewise, escin was effective in inhibiting inflammation, attenuating cognitive deficit and protecting hippocampal neurons in ischemic brain injury [91]. There is a growing interest in escin’s potential role in neurodegenerative diseases. I is reported that oral treatment with escin diminished behavioural impairments, oxidative stress and inflammation in the chronic MPTP/probenecid mouse model of Parkinson’s disease (PD) by reducing the levels of TNF-α, IL-6, IL-4, IL-10, SOD and catalase [86]. Later, escin’s positive effects on mitochondrial dysfunction, oxidative stress and apoptosis in a mouse PD model was demonstrated [92]. Meanwhile, escin was proven to be an autophagy inducer that degrades mutant huntingtin protein (mHtt) and inhibits mHtt-induced apoptosis in vitro and in vivo [93]. Escin’s autophagy induction mechanisms in HT22 cells were attributed to the regulation of mTOR and ERK pathways [93].
Ameliorative effects of escin also rely on intervening inflammatory processes, mostly through the NF–κB pathway. For instance, Zhang et al. recently reported the ameliorative effects of escin on neuropathic pain in chronic constriction injury of the sciatic nerve by suppressing the NF–κB pathway and its targets: pro-inflammatory cytokines, TNF-α, glial fibrillary acidic protein and nerve growth factor [94]. Escin was found to be a useful analgesic in bone cancer pain by suppressing inflammation and microglial activation, possibly through the suppression of the p38 MAPK/c-Fos signalling pathway [95]. Moreover, escin can contribute to muscle regeneration and prevention of muscle atrophy by reducing inflammatory infiltration, fibrosis and by increasing the number of muscle fibres [96,97].

5. Conclusions and Future Perspectives

In this paper, we provided a comprehensive discussion of escin’s chemistry, structural characteristics, and its multifaceted role in cancer therapy. Escin’s chemistry and physicochemical properties were presented, including structural information on the 34 isomers. We showed that there is ample evidence to suggest the anti-cancer effects of escin to be mainly realised through the NF–κB pathway and can be summarised into three groups: apoptotic, anti-cell proliferative and anti-metastasis effects. Escin’s broad biological activity scope enables it to be used as a potential anti-cancer compound, a supplementary medicine in cancer therapy and as an adjunct compound in composite drugs. In addition, escin can be used in treatment and post-treatment periods of cancer therapy due to its anti-inflammatory, anti-oxidative, protective and ameliorative effects. We reviewed these effects of escin and concluded that escin’s anti-inflammatory effects are realised by the GC-like mechanism.
Escin has been an interesting and wide-reaching topic in the pharmaceutical industry. Hence, it is a subject of systematic research. Still, some critical research is needed to further advance the state of the art. For instance, pharmacological studies of escin were performed in vitro and in vivo; however, as technology progresses, there are now new methods and techniques that can offer more precise analysis. One such trend is single-cell studies. As the mechanisms of escin’s pharmacological activities are not yet fully realised, single-cell experiments can provide a platform to elucidate mechanisms of action. This is especially true for the biological activities of escin against cancer cells. Computational advances allow us to work with big data, and high-throughput single-cell experiments are becoming popular. Therefore, it might be of significant interest to reveal the finest mechanistic details of escin’s biological activities in the context of cancer. This should enable us to understand the nature of cancer diseases better and to increase the therapeutic efficacy of medicines, including escin against such diseases.
Another important area of research is to ensure the protection of our natural resources. The development of new medicines based on escin’s structure can contribute to “green pharmacy”. Apart from escin’s use as an APC, it can be used to develop drug carriers that are cheap, safe and with additional therapeutic effects. Modification of escin with synthetic or other biological surfactants is also an interesting direction. This could be an alternative way to design semi-synthetic surfactants with improved properties, which are crucial to producing drug delivery tools that increase the solubility, bioavailability and stability of drugs, especially those with high molecular weights. Such nanoparticles and vesicles containing escin can enhance drug action by increasing the membrane’s permeability and sensitising cancer cells to chemotherapy drugs, as well as by contributing to therapeutic activity via its anti-inflammatory activity.

Author Contributions

Conceptualization, S.F. and A.S.; resources, A.S., K.T. and J.R.; writing—original draft preparation, S.F. and A.S.; writing—review and editing, S.F., A.S., K.T. and J.R.; funding acquisition, A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Ministry of Innovation of the Republic of Uzbekistan, Research Grant No. IL-21091385.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank Elbek Kurbanov and Nilufar Yokubova for their critical proofreading of the manuscript and fruitful discussions. A.S. and K.T. are grateful to the Ministry of Innovation of Uzbekistan for Research Grant No. IL-21091385, “Development of technology of escin substance with pharmacopeial standards from seeds of Horse chestnut”.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Augostine, C.R.; Avery, S.V. Discovery of Natural Products with Antifungal Potential Through Combinatorial Synergy. Front. Microbiol. 2022, 13, 866840. [Google Scholar] [CrossRef] [PubMed]
  2. Urbina, F.; Lentzos, F.; Invernizzi, C.; Ekins, S. Dual use of artificial-intelligence-powered drug discovery. Nat. Mach. Intell. 2022, 4, 189–191. [Google Scholar] [CrossRef] [PubMed]
  3. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef]
  4. World Heath Organisation, Cancer. Available online: https://www.who.int/news-room/fact-sheets/detail/cancer (accessed on 2 February 2023).
  5. Sharipov, A.; Tursunov, K.; Fazliev, S.; Azimova, B.; Razzokov, J. Hypoglycemic and Anti-Inflammatory Effects of Triterpene Glycoside Fractions from Aeculus hippocastanum Seeds. Molecules 2021, 26, 3784. [Google Scholar] [CrossRef] [PubMed]
  6. Zhang, X.; Zhang, S.; Yang, Y.; Wang, D.; Gao, H. Natural barrigenol-like triterpenoids: A comprehensive review of their contributions to medicinal chemistry. Phytochemistry 2019, 161, 41–74. [Google Scholar] [CrossRef] [PubMed]
  7. Yang, Y.; Wang, L.; Yuan, M.; Yu, Q.; Fu, F. Anti-Inflammatory and Gastroprotective Effects of Escin. Nat. Prod. Commun. 2020, 15, 1–10. [Google Scholar] [CrossRef]
  8. Yoshikawa, M.; Harada, E.; Murakami, T.; Matsuda, H.; Wariishi, N.; Yamahara, J.; Murakami, N.; Kitagawa, I. Escins-Ia, Ib, IIa, IIb, and IIIa, bioactive triterpene oligoglycosides from the seeds of Aesculus hippocastanum L.: Their inhibitory effects on ethanol absorption and hypoglycemic activity on glucose tolerance test. Chem. Pharm. Bull. 1994, 42, 1357–1359. [Google Scholar] [CrossRef]
  9. Yoshikawa, M.; Murakami, T.; Matsuda, H.; Yamahara, J.; Murakami, N.; Kitagawa, I. Bioactive saponins and glycosides. III. Horse chestnut. (1): The structures, inhibitory effects on ethanol absorption, and hypoglycemic activity of escins Ia, Ib, IIa, IIb, and IIIa from the seeds of Aesculus hippocastanum L. Chem. Pharm. Bull. 1996, 44, 1454–1464. [Google Scholar] [CrossRef]
  10. Matsuda, H.; Li, Y.; Murakami, T.; Ninomiya, K.; Yamahara, J.; Yoshikawa, M. Effects of escins Ia, Ib, IIa, and IIb from horse chestnut, the seeds of Aesculus hippocastanum L., on acute inflammation in animals. Biol. Pharm. Bull. 1997, 20, 1092–1095. [Google Scholar] [CrossRef]
  11. Yoshikawa, M.; Murakami, T.; Yamahara, J.; Matsuda, H. Bioactive saponins and glycosides. XII. Horse chestnut. (2): Structures of escins IIIb, IV, V, and VI and isoescins Ia, Ib, and V, acylated polyhydroxyoleanene triterpene oligoglycosides, from the seeds of horse chestnut tree (Aesculus hippocastanum L., Hippocastanaceae). Chem. Pharm. Bull. 1998, 46, 1764–1769. [Google Scholar] [CrossRef] [Green Version]
  12. Yoshikawa, M.; Murakami, T.; Otuki, K.; Yamahara, J.; Matsuda, H. Bioactive saponins and glycosides. XIII. Horse chestnut. (3): Quantitative analysis of escins Ia, Ib, IIa, and IIb by means of high performance liquid chromatography. Yakugaku Zasshi 1999, 119, 81–87. [Google Scholar] [CrossRef] [PubMed]
  13. Assessment Report on Aesculus hippocastanum L., semen; European Medicines Agency: Amsterdam, The Netherlands, 2020.
  14. Yang, X.W.; Zhao, J.; Cui, Y.X.; Liu, X.H.; Ma, C.M.; Hattori, M.; Zhang, L.H.; Zhang, L.H. Anti-HIV-1 protease triterpenoid saponins from the seeds of Aesculus chinensis. J. Nat. Prod. 1999, 11, 1510–1513. [Google Scholar] [CrossRef] [PubMed]
  15. Jing Zhao, X.-W.Y.M.H. Three New Triterpene Saponins from the Seeds of Aesculus chinensis. Chem. Pharm. Bull. 2001, 49, 626–628. [Google Scholar] [CrossRef] [PubMed]
  16. Zhao, J.; Yang, X.-W. Four new triterpene saponins from the seeds of Aesculus chinensis. J. Asian Nat. Prod. Res. 2003, 5, 197–203. [Google Scholar] [CrossRef] [PubMed]
  17. Yang, X.-W.; Zhao, J.; Hattori, M. Three new triterpenoid saponins from the seeds of Aesculus turbinata. J. Asian Nat. Prod. Res. 2008, 10, 243–247. [Google Scholar] [CrossRef]
  18. Kimura, H.; Jisaka, M.; Kimura, Y.; Katsube, T.; Yokota, K. Attenuating Effect of Saponins Isolated from Edible Horse Chestnuts (Aesculus turbinata BL.) on Increasing Blood Glucose Levels and Their Reducing Bitter Taste. J. Jpn. Soc. Food Sci. Technol.-Nippon Shokuhin Kagaku Kogaku Kaishi-J. Jpn. Soc. Food Sci. Technol. 2006, 53, 31–38. [Google Scholar] [CrossRef]
  19. Kimura, H.; Watanabe, A.; Jisaka, M.; Yamamoto, T.; Kimura, Y.; Katsube, T.; Yokota, K. Chemical Structures of Saponins from Seeds of Edible Horse Chestnuts (Aesculus turbinata) after Treatment with Wooden Ashes and Their Hypoglycemic Activity. Nippon Shokuhin Kagaku Kogaku Kaishi 2004, 51, 672–679. [Google Scholar] [CrossRef]
  20. Wu, X.J.; Zhang, M.L.; Cui, X.Y.; Gao, F.; He, Q.; Li, X.J.; Zhang, J.W.; Fawcett, J.P.; Gu, J.K. Comparative pharmacokinetics and bioavailability of escin Ia and isoescin Ia after administration of escin and of pure escin Ia and isoescin Ia in rat. J. Ethnopharmacol. 2012, 139, 201–206. [Google Scholar] [CrossRef]
  21. Wu, X.J.; Zhang, M.L.; Cui, X.Y.; Paul Fawcett, J.; Gu, J.K. Comparative pharmacokinetics and the bioavailability of escin Ib and isoescin Ib following the administration of escin, pure escin Ib and isoescin Ib in rats. J. Ethnopharmacol. 2014, 151, 839–845. [Google Scholar] [CrossRef]
  22. Oschmann, R.; Biber, A.; Lang, F.; Stumpf, H.; Kunz, K. Pharmaokinetics of beta-escin after administration of various Aesculus extract containing formulations. Pharmazie 1996, 51, 577–581. [Google Scholar]
  23. Kunz, K.; Lorkowski, G.; Petersen, G.; Samcova, E.; Schaffler, K.; Wauschkuhn, C.H. Bioavailability of escin after administration of two oral formulations containing Aesculus extract. Arzneimittel-Forschung-Drug Res. 1998, 48, 822–825. [Google Scholar]
  24. Bässler, D.; Okpanyi, S.; Schrödter, A.; Loew, D.; Schürer, M.; Schulz, H.-U. Bioavailability of β-aescin from horse chestnut seed extract: Comparative clinical studies of two galenic formulations. Adv. Ther. 2003, 20, 295–304. [Google Scholar] [CrossRef] [PubMed]
  25. Sirtori, C.R. Aescin: Pharmacology, pharmacokinetics and therapeutic profile. Pharm. Res 2001, 44, 183–193. [Google Scholar] [CrossRef] [PubMed]
  26. Kobryn, J.; Zieba, T.; Sowa, S.K.; Musial, W. Influence of Acetylated Annealed Starch on the Release of beta-Escin from the Anionic and Non-Ionic Hydrophilic Gels. Pharmaceutics 2020, 12, 84. [Google Scholar] [CrossRef] [PubMed]
  27. Kobryń, J.; Sowa, S.; Gasztych, M.; Dryś, A.; Musiał, W. Influence of Hydrophilic Polymers on theβFactor in Weibull Equation Applied to the Release Kinetics of a Biologically Active Complex ofAesculus hippocastanum. Int. J. Polym. Sci. 2017, 2017, 3486384. [Google Scholar] [CrossRef]
  28. Huang, S.; Wang, X.; Liu, M.; Lin, Z.; Gu, W.; Zhao, H.; Zhang, Y.; Ding, B.; Liu, J.; Wu, X.; et al. Modification of sodium aescinate into a safer, more stable and effective water-soluble drug by liposome-encapsulation: An in vitro and in vivo study. Drug Deliv. 2022, 29, 1132–1141. [Google Scholar] [CrossRef]
  29. Cheng, C.L.; Chao, W.T.; Li, Y.H.; Ou, Y.C.; Wang, S.S.; Chiu, K.Y.; Yuan, S.Y. Escin induces apoptosis in human bladder cancer cells: An in vitro and in vivo study. Eur. J Pharm. 2018, 840, 79–88. [Google Scholar] [CrossRef]
  30. Wang, Y.; Xu, X.; Zhao, P.; Tong, B.; Wei, Z.; Dai, Y. Escin Ia suppresses the metastasis of triple-negative breast cancer by inhibiting epithelial-mesenchymal transition via down-regulating LOXL2 expression. Oncotarget 2016, 7, 23684–23699. [Google Scholar] [CrossRef]
  31. Akar, S.; Donmez-Altuntas, H.; Hamurcu, Z. β-Escin reduces cancer progression in aggressive MDA-MB-231 cells by inhibiting glutamine metabolism through downregulation of c-myc oncogene. Mol. Biol. Rep. 2022, 49, 7409–7415. [Google Scholar] [CrossRef]
  32. Mojzisova, G.; Mojzis, J.; Pilatova, M.; Varinska, L.; Ivanova, L.; Strojny, L.; Richnavsky, J. Antiproliferative and antiangiogenic properties of horse chestnut extract. Phytother. Res. 2013, 27, 159–165. [Google Scholar] [CrossRef]
  33. Mojzisova, G.; Kello, M.; Pilatova, M.; Tomeckova, V.; Vaskova, J.; Vasko, L.; Bernatova, S.; Mirossay, L.; Mojzis, J. Antiproliferative effect of beta-escin—An in vitro study. Acta Biochim. Pol. 2016, 63, 79–87. [Google Scholar] [CrossRef] [PubMed]
  34. Mazrouei, R.; Raeisi, E.; Lemoigne, Y.; Heidarian, E. Activation of p53 Gene Expression and Synergistic Antiproliferative Effects of 5-Fluorouracil and beta-escin on MCF7 Cells. J. Med. Signals Sens. 2019, 9, 196–203. [Google Scholar] [CrossRef] [PubMed]
  35. Yang, Y.; Long, L.; Zhang, X.; Song, K.; Wang, D.; Xiong, X.; Gao, H.; Sha, L. 16-Tigloyl linked barrigenol-like triterpenoid from Semen Aesculi and its anti-tumor activity in vivo and in vitro. RSC Adv. 2019, 9, 31758–31772. [Google Scholar] [CrossRef] [PubMed]
  36. Patlolla, J.M.; Raju, J.; Swamy, M.V.; Rao, C.V. Beta-escin inhibits colonic aberrant crypt foci formation in rats and regulates the cell cycle growth by inducing p21(waf1/cip1) in colon cancer cells. Mol. Cancer 2006, 5, 1459–1466. [Google Scholar] [CrossRef]
  37. Seweryn, E.; Glehsk, M.; Środa-Pomianek, K.; Ceremuga, I.; Włodarczyk, M.; Gamian, A. Cytotoxic Effects of Four Aescin Types on Human Colon Adenocarcinoma Cell Lines. Nat. Prod. Commun. 2014, 9, 387–390. [Google Scholar] [CrossRef]
  38. Wang, Z.; Chen, Q.; Li, B.; Xie, J.-M.; Yang, X.-D.; Zhao, K.; Wu, Y.; Ye, Z.-Y.; Chen, Z.-R.; Qin, Z.-H.; et al. Escin-induced DNA damage promotes escin-induced apoptosis in human colorectal cancer cells via p62 regulation of the ATM/γH2AX pathway. Acta Pharmacol. Sin. 2018, 39, 1645–1660. [Google Scholar] [CrossRef]
  39. Li, B.; Wang, Z.; Xie, J.M.; Wang, G.; Qian, L.Q.; Guan, X.M.; Shen, X.P.; Qin, Z.H.; Shen, G.H.; Li, X.Q.; et al. TIGAR knockdown enhanced the anticancer effect of aescin via regulating autophagy and apoptosis in colorectal cancer cells. Acta Pharm. Sin. 2019, 40, 111–121. [Google Scholar] [CrossRef]
  40. Shen, D.Y.; Kang, J.H.; Song, W.; Zhang, W.Q.; Li, W.G.; Zhao, Y.; Chen, Q.X. Apoptosis of human cholangiocarcinoma cell lines induced by beta-escin through mitochondrial caspase-dependent pathway. Phytother. Res. 2011, 25, 1519–1526. [Google Scholar] [CrossRef]
  41. Huang, G.L.; Shen, D.Y.; Cai, C.F.; Zhang, Q.Y.; Ren, H.Y.; Chen, Q.X. beta-escin reverses multidrug resistance through inhibition of the GSK3beta/beta-catenin pathway in cholangiocarcinoma. World J. Gastroenterol. 2015, 21, 1148–1157. [Google Scholar] [CrossRef]
  42. Lee, H.S.; Hong, J.E.; Kim, E.J.; Kim, S.H. Escin suppresses migration and invasion involving the alteration of CXCL16/CXCR6 axis in human gastric adenocarcinoma AGS cells. Nutr. Cancer 2014, 66, 938–945. [Google Scholar] [CrossRef]
  43. Harford-Wright, E.; Bidère, N.; Gavard, J. β-escin selectively targets the glioblastoma-initiating cell population and reduces cell viability. Oncotarget 2016, 7, 66865–66879. [Google Scholar] [CrossRef] [PubMed]
  44. Ming, Z.J.; Hu, Y.; Qiu, Y.H.; Cao, L.; Zhang, X.G. Synergistic effects of β-aescin and 5-fluorouracil in human hepatocellular carcinoma SMMC-7721 cells. Phytomedicine 2010, 17, 575–580. [Google Scholar] [CrossRef] [PubMed]
  45. Zhou, X.Y.; Fu, F.H.; Li, Z.; Dong, Q.J.; He, J.; Wang, C.H. Escin, a natural mixture of triterpene saponins, exhibits antitumor activity against hepatocellular carcinoma. Planta Med. 2009, 75, 1580–1585. [Google Scholar] [CrossRef] [PubMed]
  46. Tan, S.M.-L.; Li, F.; Rajendran, P.; Kumar, A.P.; Hui, K.M.; Sethi, G. Identification of β-Escin as a Novel Inhibitor of Signal Transducer and Activator of Transcription 3/Janus-Activated Kinase 2 Signaling Pathway that Suppresses Proliferation and Induces Apoptosis in Human Hepatocellular Carcinoma Cells. J. Pharmacol. Exp. Ther. 2010, 334, 285. [Google Scholar] [CrossRef]
  47. Zhang, Z.; Gao, J.; Cai, X.; Zhao, Y.; Wang, Y.; Lu, W.; Gu, Z.; Zhang, S.; Cao, P. Escin sodium induces apoptosis of human acute leukemia Jurkat T cells. Phytother. Res. 2011, 25, 1747–1755. [Google Scholar] [CrossRef] [PubMed]
  48. Harikumar, K.B.; Sung, B.; Pandey, M.K.; Guha, S.; Krishnan, S.; Aggarwal, B.B. Escin, a Pentacyclic Triterpene, Chemosensitizes Human Tumor Cells through Inhibition of Nuclear Factor-κB Signaling Pathway. Mol. Pharmacol. 2010, 77, 818. [Google Scholar] [CrossRef]
  49. Ji, D.B.; Xu, B.; Liu, J.T.; Ran, F.X.; Cui, J.R. beta-Escin sodium inhibits inducible nitric oxide synthase expression via downregulation of the JAK/STAT pathway in A549 cells. Mol. Carcinog. 2011, 50, 945–960. [Google Scholar] [CrossRef]
  50. Patlolla, J.M.; Qian, L.; Biddick, L.; Zhang, Y.; Desai, D.; Amin, S.; Lightfoot, S.; Rao, C.V. beta-Escin inhibits NNK-induced lung adenocarcinoma and ALDH1A1 and RhoA/Rock expression in A/J mice and growth of H460 human lung cancer cells. Cancer Prev. Res. 2013, 6, 1140–1149. [Google Scholar] [CrossRef]
  51. Ciftci, G.A.; Iscan, A.; Kutlu, M. Escin reduces cell proliferation and induces apoptosis on glioma and lung adenocarcinoma cell lines. Cytotechnology 2015, 67, 893–904. [Google Scholar] [CrossRef]
  52. Kwak, H.; An, H.; Alam, M.B.; Choi, W.-S.; Lee, S.Y.; Lee, S.-H. Inhibition of Migration and Invasion in Melanoma Cells by β-Escin via the ERK/NF-κB Signaling Pathway. Biol. Pharm. Bull. 2018, 41, 1606–1610. [Google Scholar] [CrossRef]
  53. Zhu, J.; Yu, W.; Liu, B.; Wang, Y.; Shao, J.; Wang, J.; Xia, K.; Liang, C.; Fang, W.; Zhou, C.; et al. Escin induces caspase-dependent apoptosis and autophagy through the ROS/p38 MAPK signalling pathway in human osteosarcoma cells in vitro and in vivo. Cell Death Dis. 2017, 8, e3113. [Google Scholar] [CrossRef] [PubMed]
  54. Zhu, M.; Ying, J.; Lin, C.; Wang, Y.; Huang, K.; Zhou, Y.; Teng, H. beta-Escin inhibits the proliferation of osteosarcoma cells via blocking the PI3K/Akt pathway. RSC Adv. 2018, 8, 29637–29644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Kenny, H.A.; Hart, P.C.; Kordylewicz, K.; Lal, M.; Shen, M.; Kara, B.; Chen, Y.J.; Grassl, N.; Alharbi, Y.; Pattnaik, B.R.; et al. The Natural Product beta-Escin Targets Cancer and Stromal Cells of the Tumor Microenvironment to Inhibit Ovarian Cancer Metastasis. Cancers 2021, 13, 3931. [Google Scholar] [CrossRef] [PubMed]
  56. Wang, Y.W.; Wang, S.J.; Zhou, Y.N.; Pan, S.H.; Sun, B. Escin augments the efficacy of gemcitabine through down-regulation of nuclear factor-kappaB and nuclear factor-kappaB-regulated gene products in pancreatic cancer both in vitro and in vivo. J Cancer Res. Clin. Oncol. 2012, 138, 785–797. [Google Scholar] [CrossRef]
  57. Rimmon, A.; Vexler, A.; Berkovich, L.; Earon, G.; Ron, I.; Lev-Ari, S. Escin Chemosensitizes Human Pancreatic Cancer Cells and Inhibits the Nuclear Factor-kappaB Signaling Pathway. Biochem. Res. Int. 2013, 2013, 251752. [Google Scholar] [CrossRef]
  58. Omi, K.; Matsuo, Y.; Ueda, G.; Aoyama, Y.; Kato, T.; Hayashi, Y.; Imafuji, H.; Saito, K.; Tsuboi, K.; Morimoto, M.; et al. Escin inhibits angiogenesis by suppressing interleukin8 and vascular endothelial growth factor production by blocking nuclear factorkappaB activation in pancreatic cancer cell lines. Oncol. Rep. 2021, 45, 55. [Google Scholar] [CrossRef]
  59. Piao, S.; Kang, M.; Lee, Y.J.; Choi, W.S.; Chun, Y.S.; Kwak, C.; Kim, H.H. Cytotoxic effects of escin on human castration-resistant prostate cancer cells through the induction of apoptosis and G2/M cell cycle arrest. Urology 2014, 84, 982.e1–982.e7. [Google Scholar] [CrossRef]
  60. Yuan, S.Y.; Cheng, C.L.; Wang, S.S.; Ho, H.C.; Chiu, K.Y.; Chen, C.S.; Chen, C.C.; Shiau, M.Y.; Ou, Y.C. Escin induces apoptosis in human renal cancer cells through G2/M arrest and reactive oxygen species-modulated mitochondrial pathways. Oncol. Rep. 2017, 37, 1002–1010. [Google Scholar] [CrossRef]
  61. Elmore, S. Apoptosis: A Review of Programmed Cell Death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef]
  62. Luo, J.-L.; Kamata, H.; Karin, M. IKK/NF-κB signaling: Balancing life and death—A new approach to cancer therapy. J. Clin. Investig. 2005, 115, 2625–2632. [Google Scholar] [CrossRef]
  63. Taniguchi, K.; Karin, M. NF-kappaB, inflammation, immunity and cancer: Coming of age. Nat. Rev. Immunol. 2018, 18, 309–324. [Google Scholar] [CrossRef] [PubMed]
  64. Li, Q.; Verma, I.M. NF-kappaB regulation in the immune system. Nat. Rev. Immunol. 2002, 2, 725–734. [Google Scholar] [CrossRef] [PubMed]
  65. Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-kappaB signaling in inflammation. Signal Transduct. Target 2017, 2, 17023. [Google Scholar] [CrossRef] [PubMed]
  66. Paneerselvam, C.; Ganapasam, S. beta-Escin alleviates cobalt chloride-induced hypoxia-mediated apoptotic resistance and invasion via ROS-dependent HIF-1alpha/TGF-beta/MMPs in A549 cells. Toxicol. Res. 2020, 9, 191–201. [Google Scholar] [CrossRef] [PubMed]
  67. Varinska, L.; Faber, L.; Kello, M.; Petrovova, E.; Balazova, L.; Solar, P.; Coma, M.; Urdzik, P.; Mojzis, J.; Svajdlenka, E.; et al. beta-Escin Effectively Modulates HUVECS Proliferation and Tube Formation. Molecules 2018, 23, 197. [Google Scholar] [CrossRef] [PubMed]
  68. Wang, X.H.; Xu, B.; Liu, J.T.; Cui, J.R. Effect of beta-escin sodium on endothelial cells proliferation, migration and apoptosis. Vasc. Pharm. 2008, 49, 158–165. [Google Scholar] [CrossRef]
  69. Cheng, P.; Kuang, F.; Ju, G. Aescin reduces oxidative stress and provides neuroprotection in experimental traumatic spinal cord injury. Free Radic. Biol. Med. 2016, 99, 405–417. [Google Scholar] [CrossRef]
  70. Diehm, C.; Trampisch, H.J.; Lange, S.; Schmidt, C. Comparison of leg compression stocking and oral horse-chestnut seed extract therapy in patients with chronic venous insufficiency. Lancet 1996, 347, 292–294. [Google Scholar] [CrossRef]
  71. Dudek-Makuch, M.; Studzińska-Sroka, E. Horse chestnut—Efficacy and safety in chronic venous insufficiency: An overview. Rev. Bras. Farmacogn. 2015, 25, 533–541. [Google Scholar] [CrossRef]
  72. Vogel, G.; Marek, M.L.; Stoeckert, I. Weitere Untersuchengen zun Wirkungsmechanismus des Rosskastanien-Saponins Aescin. Arzneim. Forsch. 1963, 13, 59–64. [Google Scholar]
  73. Xin, W.; Zhang, L.; Fan, H.; Jiang, N.; Wang, T.; Fu, F. Escin attenuates acute lung injury induced by endotoxin in mice. Eur. J. Pharm. Sci. 2011, 42, 73–80. [Google Scholar] [CrossRef]
  74. Xin, W.; Zhang, L.; Sun, F.; Jiang, N.; Fan, H.; Wang, T.; Li, Z.; He, J.; Fu, F. Escin exerts synergistic anti-inflammatory effects with low doses of glucocorticoids in vivo and in vitro. Phytomedicine 2011, 18, 272–277. [Google Scholar] [CrossRef]
  75. Liu, S.; Wang, H.; Qiu, C.; Zhang, J.; Zhang, T.; Zhou, W.; Lu, Z.; Rausch-Fan, X.; Liu, Z. Escin inhibits lipopolysaccharide-induced inflammation in human periodontal ligament cells. Mol. Med. Rep. 2012, 6, 1150–1154. [Google Scholar] [CrossRef] [Green Version]
  76. Wang, H.; Zhang, L.; Jiang, N.; Wang, Z.; Chong, Y.; Fu, F. Anti-inflammatory effects of escin are correlated with the glucocorticoid receptor/NF-kappaB signaling pathway, but not the COX/PGF2alpha signaling pathway. Exp. Med. 2013, 6, 419–422. [Google Scholar] [CrossRef]
  77. Gupta, S.C.; Tyagi, A.K.; Deshmukh-Taskar, P.; Hinojosa, M.; Prasad, S.; Aggarwal, B.B. Downregulation of tumor necrosis factor and other proinflammatory biomarkers by polyphenols. Arch Biochem Biophys 2014, 559, 91–99. [Google Scholar] [CrossRef]
  78. Zhao, S.-Q.; Xu, S.-Q.; Cheng, J.; Cao, X.-L.; Zhang, Y.; Zhou, W.-P.; Huang, Y.-J.; Wang, J.; Hu, X.-M. Anti-inflammatory effect of external use of escin on cutaneous inflammation: Possible involvement of glucocorticoids receptor. Chin. J. Nat. Med. 2018, 16, 105–112. [Google Scholar] [CrossRef]
  79. Hiai, S.; Yokoyama, H.; Oura, H. Effect of escin on adrenocorticotropin and corticosterone levels in rat plasma. Chem. Pharm. Bull. 1981, 29, 490–494. [Google Scholar] [CrossRef]
  80. Zhang, L.; Wang, H.; Fan, H.; Wang, T.; Jiang, N.; Yu, P.; Fu, F. The potent anti-inflammatory agent escin does not increase corticosterone secretion and immune cell apoptosis in mice. Fitoterapia 2011, 82, 861–867. [Google Scholar] [CrossRef]
  81. Zhang, F.; Li, Y.; Zhang, L.; Mu, G. Synergistic protective effects of escin and lowdose glucocorticoids on bloodretinal barrier breakdown in a rat model of retinal ischemia. Mol. Med. Rep. 2013, 7, 1511–1515. [Google Scholar] [CrossRef]
  82. Timmermans, S.; Souffriau, J.; Libert, C. A General Introduction to Glucocorticoid Biology. Front. Immunol. 2019, 10, 1545. [Google Scholar] [CrossRef]
  83. Cheng, Y.; Wang, H.; Mao, M.; Liang, C.; Zhang, Y.; Yang, D.; Wei, Z.; Gao, S.; Hu, B.; Wang, L.; et al. Escin Increases the Survival Rate of LPS-Induced Septic Mice Through Inhibition of HMGB1 Release from Macrophages. Cell Physiol. Biochem. 2015, 36, 1577–1586. [Google Scholar] [CrossRef] [PubMed]
  84. Ding, Y.X.; Eerduna, G.W.; Duan, S.J.; Li, T.; Liu, R.X.; Zhang, L.M.; Wang, T.; Fu, F.H. Escin ameliorates the impairments of neurological function and blood brain barrier by inhibiting systemic inflammation in intracerebral hemorrhagic mice. Exp. Neurol. 2021, 337, 113554. [Google Scholar] [CrossRef] [PubMed]
  85. Ali, F.E.M.; Ahmed, S.F.; Eltrawy, A.H.; Yousef, R.S.; Ali, H.S.; Mahmoud, A.R.; Abd-Elhamid, T.H. Pretreatment with Coenzyme Q10 Combined with Aescin Protects against Sepsis-Induced Acute Lung Injury. Cells Tissues Organs 2021, 210, 195–217. [Google Scholar] [CrossRef]
  86. Domanski, D.; Zegrocka-Stendel, O.; Perzanowska, A.; Dutkiewicz, M.; Kowalewska, M.; Grabowska, I.; Maciejko, D.; Fogtman, A.; Dadlez, M.; Koziak, K. Molecular Mechanism for Cellular Response to beta-Escin and Its Therapeutic Implications. PLoS ONE 2016, 11, e0164365. [Google Scholar] [CrossRef] [PubMed]
  87. Selvakumar, G.P.; Janakiraman, U.; Essa, M.M.; Justin Thenmozhi, A.; Manivasagam, T. Escin attenuates behavioral impairments, oxidative stress and inflammation in a chronic MPTP/probenecid mouse model of Parkinson’s disease. Brain Res. 2014, 1585, 23–36. [Google Scholar] [CrossRef]
  88. Suryavanshi, S.V.; Kulkarni, Y.A. Attenuation of Cardiac Autonomic Neuropathy by Escin in Diabetic Rats. Pharmacology 2021, 106, 211–217. [Google Scholar] [CrossRef]
  89. Gur, F.; Cengiz, M.; Kutlu, H.M.; Cengiz, B.P.; Ayhanci, A. Molecular docking analyses of Escin as regards cyclophosphamide-induced cardiotoxicity: In vivo and in Silico studies. Toxicol. Appl. Pharm. 2021, 411, 115386. [Google Scholar] [CrossRef] [PubMed]
  90. Wang, K.; Jiang, Y.; Wang, W.; Ma, J.; Chen, M. Escin activates AKT-Nrf2 signaling to protect retinal pigment epithelium cells from oxidative stress. Biochem. Biophys. Res. Commun. 2015, 468, 541–547. [Google Scholar] [CrossRef]
  91. Hu, X.M.; Zeng, F.D. Protective effects of sodium beta-aescin on ischemia-reperfusion injury in rat brain. Yao Xue Xue Bao 2004, 39, 419–423. [Google Scholar]
  92. Zhang, L.; Fu, F.; Zhang, X.; Zhu, M.; Wang, T.; Fan, H. Escin attenuates cognitive deficits and hippocampal injury after transient global cerebral ischemia in mice via regulating certain inflammatory genes. Neurochem. Int. 2010, 57, 119–127. [Google Scholar] [CrossRef]
  93. Selvakumar, G.P.; Manivasagam, T.; Rekha, K.R.; Jayaraj, R.L.; Elangovan, N. Escin, a novel triterpene, mitigates chronic MPTP/p-induced dopaminergic toxicity by attenuating mitochondrial dysfunction, oxidative stress, and apoptosis. J. Mol. Neurosci. 2015, 55, 184–197. [Google Scholar] [CrossRef] [PubMed]
  94. Sun, Y.; Jiang, X.; Pan, R.; Zhou, X.; Qin, D.; Xiong, R.; Wang, Y.; Qiu, W.; Wu, A.; Wu, J. Escins Isolated from Aesculus chinensis Bge. Promote the Autophagic Degradation of Mutant Huntingtin and Inhibit its Induced Apoptosis in HT22 cells. Front. Pharm. 2020, 11, 116. [Google Scholar] [CrossRef] [PubMed]
  95. Zhang, L.; Chen, X.; Wu, L.; Li, Y.; Wang, L.; Zhao, X.; Zhao, T.; Zhang, L.; Yan, Z.; Wei, G. Ameliorative effects of escin on neuropathic pain induced by chronic constriction injury of sciatic nerve. J. Ethnopharmacol. 2021, 267, 113503. [Google Scholar] [CrossRef] [PubMed]
  96. Yang, G.; Li, J.; Xu, Q.; Xie, H.; Wang, L.; Zhang, M. Sodium aescinate alleviates bone cancer pain in rats by suppressing microglial activation via p38 MAPK/c-Fos signaling. Mol. Cell. Toxicol. 2022, 18, 605–614. [Google Scholar] [CrossRef]
  97. Sikorska, M.; Dutkiewicz, M.; Zegrocka-Stendel, O.; Kowalewska, M.; Grabowska, I.; Koziak, K. Beneficial effects of beta-escin on muscle regeneration in rat model of skeletal muscle injury. Phytomedicine 2021, 93, 153791. [Google Scholar] [CrossRef]
Figure 1. General chemical structure of escin (A), structures of various acid moieties (B) and sugar units (C) found in escin.
Figure 1. General chemical structure of escin (A), structures of various acid moieties (B) and sugar units (C) found in escin.
Biomolecules 13 00315 g001
Figure 2. Possible molecular targets of escin’s anti-cancer mechanisms. Abbreviations: BS—binding site, NF-κB—Nuclear factor kappa B, ERK1/2—extracellular signal-regulated kinase 1/2, Bcl-2—B-cell lymphoma 2 proteins, BAX—BCL2-associated X, Bcl-xL—X-linked inhibitor of apoptosis protein (xIAP), VEGF—vascular epidermal growth factor, MAPKs—p38 mitogen-activated protein kinases, IκBα—cytoplasmic inhibitory protein of NF-κB, STAT3—signal transducer and activator of transcription protein 3.
Figure 2. Possible molecular targets of escin’s anti-cancer mechanisms. Abbreviations: BS—binding site, NF-κB—Nuclear factor kappa B, ERK1/2—extracellular signal-regulated kinase 1/2, Bcl-2—B-cell lymphoma 2 proteins, BAX—BCL2-associated X, Bcl-xL—X-linked inhibitor of apoptosis protein (xIAP), VEGF—vascular epidermal growth factor, MAPKs—p38 mitogen-activated protein kinases, IκBα—cytoplasmic inhibitory protein of NF-κB, STAT3—signal transducer and activator of transcription protein 3.
Biomolecules 13 00315 g002
Figure 3. Overview of anti-inflammatory mechanisms of escin at the molecular level.
Figure 3. Overview of anti-inflammatory mechanisms of escin at the molecular level.
Biomolecules 13 00315 g003
Table 1. Chemical structures of escin. (See also Figure 1).
Table 1. Chemical structures of escin. (See also Figure 1).
CompoundR1R2R3R4R5R6Ref.
Escin IaOHHHAcetylTigeloylβ-d-glucopyranosyl[8,12]
Escin IbOHHHAcetylAngeloylβ-d-glucopyranosyl[8,12]
Escin IIaOHHHAcetylTigeloylβ-d-xylopyranosyl[8]
Escin IIbOHHHAcetylAngeloylβ-d-xylopyranosyl[8]
Escin IIIaHHHAcetylTigeloylβ-d-galactopyranosyl[8,9]
Escin IIIbHHHAcetylAngeloylβ-d-galactopyranosyl[11]
Escin IVOHHHAcetylAcetylβ-d-glucopyranosyl[11]
Escin IVcOHHAcetylTigeloylHβ-d-glucopyranosyl[14]
Escin IVdOHHAcetylAngeloylHβ-d-glucopyranosyl[14]
Escin IVeOHHTigeloylHHβ-d-glucopyranosyl[14]
Escin IVfOHHAngeloylHHβ-d-glucopyranosyl[14]
Escin IVgOHHHTigeloylHβ-d-glucopyranosyl[15]
Escin IVhOHHHAngeloylHβ-d-glucopyranosyl[15]
Escin VOHHHAcetyl2-methylpropanoylβ-d-glucopyranosyl[11]
Escin VIOHHHAcetyl2-methylbutanoylβ-d-glucopyranosyl[11]
Escin VIbOHAngeloylHHAcetylβ-d-glucopyranosyl[15]
Isoescin IaOHHAcetylHTigeloylβ-d-glucopyranosyl[11]
Isoescin IbOHHAcetylHAngeloylβ-d-glucopyranosyl[11]
Isoescin IIaOHHAcetylHTigeloylβ-d-xylopyranosyl[16]
Isoescin IIbOHHAcetylHAngeloylβ-d-xylopyranosyl[16]
Isoescin IIIaHHAcetylHTigeloylβ-d-galactopyranosyl[16]
Isoescin IIIbHHAcetylHAngeloylβ-d-galactopyranosyl[16]
Isoescin VOHHAcetylH2-methylpropanoylβ-d-glucopyranosyl[11]
Isoescin VIaOHHAcetylH2-methylbutanoylβ-d-glucopyranosyl[17]
Isoescin VIIaOHHAcetylHTigeloylβ-d-galactopyranosyl[17]
Isoescin VIIIaHHAcetylHAngeloylβ-d-glucopyranosyl[17]
Deacylescin IOHHHHHβ-d-glucopyranosyl[8,18]
Deacylescin IaOHHHHTigeloylβ-d-glucopyranosyl[19]
Deacylescin IbOHHHHAngeloylβ-d-glucopyranosyl[19]
Deacylescin IIOHHHHHβ-d-xylopyranosyl[10,18]
Deacylescin IIaOHHHHTigeloylβ-d-xylopyranosyl[19]
Deacylescin IIbOHHHHAngeloylβ-d-xylopyranosyl[19]
Deacylescin IIIHHHHHβ-d-galactopyranosyl[8]
Table 2. Anti-cancer effects of escin revealed by in vitro or in vivo studies.
Table 2. Anti-cancer effects of escin revealed by in vitro or in vivo studies.
Cancer
Type
Model
System
Anti-Cancer Effects and Mechanisms of Escin byRef.
Inducing Cell ApoptosisDecreasing Cell ProliferationInhibiting
Metastasis and Invasion
Bladder
cancer
T24, J82,
TCCSUP, and RT-4 cell lines
Through death receptor and mitochondria mediated pathways↓ NF–κB p65, partially affects STAT3
expression
[28]
Breast
cancer
MDA-MB-231 cell line ↓ LOXL2, ↓ c-Myc,
↓ GSL1, ↑ASCT2
[29,30]
Mice and MCF-7 cell lineYesYes↑ p53, ↓ Bcl-2[31,32,33,34]
Colon
cancer
HT-29 cell line Cell cycle arrest at G1/S phase mediated by induction of p21WAF1/CIP1 [35]
LoVo cell line Yes [36]
Colorectal
cancer
Mice and HCT116, HCT8 cell linesThrough DNA damageYes [37]
Mice and HCT116, HCT8 cell linesThrough DNA damage
↑ TIGAR
↑ ROS
Yes [38]
CholangiocarcinomaQBC939, MZ-ChA-1 Sk-ChA-1
cell lines
↑ caspase-3,
↓ Bcl-2
Cell cycle arrest at G1 and G2/M phases [39]
QBC939 cell lineSensitised to 5-FU and VCR↓ P-glycoprotein,
↓ GSK3b/ catenin
[40]
Gastric cancerAGS cell lines Through Akt signalling pathway [41]
Glioblastoma multiformeClassical and mesenchymal
glioblastoma-initiating cells
Through mitochondria-mediated pathwayYes [42]
Hepatocellular carcinomaSMMC-7721
cell line
↑ caspases 3, 8, 9;
↓ Bcl-2 with 5-FU
Cell cycle arrest at G0/G1 with 5-FU [43]
HepG2 cell line↑ PARP, AIF, BAX, and Bcl-2Cell cycle arrest at G1/S phase [44]
HepG2 cell line ↓ Akt/JAK/STAT,
↓ cyclin D1, ↓ Bcl-2,
↓ Bcl-xL, ↓ survivin,
↓ Mcl-1, ↓ VEGF; sensitised to Dx, PTX
[45]
LeukaemiaJurkat T-cell line↑ Caspases-3, 8, 9,
↓ PARP, ↓ Bcl-2,
↑ ROS
Yes [32,46]
CEM cell line Yes [32]
KBM-5 cell lineTNF-induced apoptosis↓ TNF
↓ NF–κB
↓ TNF
↓ NF–κB
[47]
Lung
cancer
A549 cell line Through JAK/STAT signalling pathway↓ iNOS[48]
Mice and H460 cell line ↓ ALDH1A1
↓ p-Akt, ↑ p21
↓ RhoA and Rock[49]
A549 cell line↑ BAX,
↑ caspase-3
Cell cycle arrest at G0/G1phase [50]
MelanomaSK-MEL5 and B16F10 cell lines ↓ NF–κB,
↓ IκB
Through ERK1/2 signalling[51]
OsteosarcomaMice and MNNG/HOS, Saos-2, MG-63, U-2OS, HUVEC cell linesThrough
ROS/p38 MAPK signalling pathway
[52]
Mice and
MG-63, OS732 cell lines
↑ Caspases-3, 8, 9↓ PI3K/Akt pathway [53]
Ovarian cancerHeyA8, SNU-119, Kuramochi, Ovcar4, and Ovcar5 cell lines ↓ Autophagy-dependent CSC differentiation,
↓ Stromal ECM production driven by HIF1α
[54]
Pancreatic cancerMice and
BxPC-3, PANC-1 cell lines
↓ NF–κB, ↓ c-Myc, ↓ COX-2, ↓ cyclin D1, ↓ survivin, ↓ Bcl-xL, ↓ Bcl-2, ↑ caspase-3 [55]
COLO357, MIA-Paca, Panc-1, cell linesYes↓ NF–κB,
↓ cyclin D, sensitised cells to cisplatin
[56]
BxPC-3, AsPC-1, SW1990 cell lines ↓ NF-κB↓ IL-8, ↓ VEGF[57]
Prostate
cancer
Mice and CRPC, PC-3, DU-145 cell lines↑ c-caspase-3,
↑ BAX, ↓ Bcl-2,
↓ cIAP-1, ↓ cIAP-2
↓ xIAP, ↑ PARP
Cell cycle arrest at G2/M-phase [58]
Renal
cancer
786-O and Caki-1 cell lines↓ Bcl-2,
↑ ROS
Cell cycle arrest at G2/M arrest [59]
Abbreviations: Dx—doxorubicin, IL—interleukin, PTX—paclitaxel, VCR—Vincristine, LOXL2—lysyl oxidase-like 2, EMT—epithelial-mesenchymal transition, 5-FU—5-Fluorouracil, CSC—cancer stem cell, GLS1—kidney type glutaminase, ASCT2—alanine-serine-cysteine 2 protein, MAPKs—p38 mitogen-activated protein kinases, NF-κB—Nuclear factor kappa B, ROS—reactive oxygen species, TIGAR—TP53-induced glycolysis and apoptosis regulator, CXCL16—chemokine (C-X-C motif) ligand, iNOS—inducible nitric oxide synthase, ERK1/2—extracellular signal-regulated kinase 1/2, PARP (c)—poly (adenosine diphosphateribose) polymerase, Bcl-2—B-cell lymphoma 2 proteins, BAX—BCL2-associated X, Bcl-xL—X-linked inhibitor of apoptosis protein (xIAP), ALDH—aldehyde dehydrogenase, p-Akt—phospho-Akt, HUVECs—human umbilical vein endothelial cells, VEGF—vascular epidermal growth factor, ECM—extracellular matrix, and HIF1α—Hypoxia-inducible factor 1-alpha.
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

Fazliev, S.; Tursunov, K.; Razzokov, J.; Sharipov, A. Escin’s Multifaceted Therapeutic Profile in Treatment and Post-Treatment of Various Cancers: A Comprehensive Review. Biomolecules 2023, 13, 315. https://doi.org/10.3390/biom13020315

AMA Style

Fazliev S, Tursunov K, Razzokov J, Sharipov A. Escin’s Multifaceted Therapeutic Profile in Treatment and Post-Treatment of Various Cancers: A Comprehensive Review. Biomolecules. 2023; 13(2):315. https://doi.org/10.3390/biom13020315

Chicago/Turabian Style

Fazliev, Sunnatullo, Khurshid Tursunov, Jamoliddin Razzokov, and Avez Sharipov. 2023. "Escin’s Multifaceted Therapeutic Profile in Treatment and Post-Treatment of Various Cancers: A Comprehensive Review" Biomolecules 13, no. 2: 315. https://doi.org/10.3390/biom13020315

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