Next Article in Journal
Recent Advances in Secondary Metabolites from Marine Aspergillus
Next Article in Special Issue
Cancer Cell Cytotoxicity of Marinopyrroles, Pyrrolomycins, and Their Derivatives
Previous Article in Journal
Methodologies for Detoxifying Bivalves from Marine Paralytic Shellfish Toxins
Previous Article in Special Issue
The Marine Natural Compound Aplysinamisine I Selectively Induces Apoptosis and Exhibits Synergy with Taxol™ in Triple-Negative Breast Cancer Spheroids
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Marine-Derived Steroids for Cancer Treatment: Search for Potential Selective Glucocorticoid Receptor Agonists/Modulators (SEGRAMs)

by
Ekaterina M. Zhidkova
1,
Ekaterina D. Savina
1,
Ekaterina A. Yurchenko
2 and
Ekaterina A. Lesovaya
1,3,4,*
1
Department of Chemical Carcinogenesis, Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center for Oncology, Kashirskoe Shosse 24-15, Moscow 115478, Russia
2
G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-Letiya Vladivostoka, Vladivostok 690022, Russia
3
Institute of Medicine, Peoples’ Friendship University of Russia, Miklukho-Maklaya St. 6, Moscow 117198, Russia
4
Faculty of Oncology, I.P. Pavlov Ryazan State Medical University, Vysokovol’tnaya St. 9, Ryazan 390026, Russia
*
Author to whom correspondence should be addressed.
Mar. Drugs 2025, 23(10), 399; https://doi.org/10.3390/md23100399
Submission received: 12 September 2025 / Revised: 11 October 2025 / Accepted: 13 October 2025 / Published: 14 October 2025
(This article belongs to the Special Issue Marine Natural Products as Anticancer Agents, 5th Edition)

Abstract

Steroids, particularly glucocorticoids, are essential components of cancer treatment for both hematological malignancies and solid tumors. The adverse effects of standard steroid-based drugs have forced drug discovery research to develop chemotherapeutics with a more selective mechanism of action and an improved therapeutic index. Steroids of natural origin and their analogs are a significant source of novel molecules with a wide spectrum of biological activities. In this review, we aimed to analyze marine-derived steroids and their anti-cancer activity. Moreover, we specifically discussed molecules with not only anti-cancer but also anti-inflammatory activities that could potentially mimic the effects of glucocorticoids. We hypothesized that several of the reviewed compounds could exhibit affinity to the glucocorticoid receptor, and possess the properties of selective glucocorticoid receptor agonists/modulators with increased therapeutic activity and decreased side effects. The review is based on the literature available in the PubMed, Cochrane, and ClinicalTrials.gov databases and covers the period from 1986 to 2025. The keywords used were “steroids”, “cancer”, and “marine-derived steroids”. The second iteration of the literature search included the keywords “selective glucocorticoid receptor agonists”, “marine-derived”, and “cancer”. In silico calculations of several marine-derived compounds were performed to support the hypothesis based on the literature data.

1. Introduction

Cancer pathologies are characterized by the uncontrolled proliferation of transformed cells, replacement of normal tissues, and invasion and metastasis to adjacent and distant organs. The pleiotropic mechanisms of cell transformation, including cell cycle disruption, attenuation of apoptosis, aberrant signaling, neoangiogenesis, invasion, and changes in tumor epigenetics and microenvironment, require a multifactorial treatment strategy [1,2,3]. Targeted cancer therapy is crucially dependent on the identification of specific biomarkers, which are frequently associated with the activation of alternative signaling pathways and, consequently, the development of pharmacological resistance [4,5]. Multitargeted therapies could be a more effective alternative, using either drug combinations or a single drug with multiple targets [6]. There are several options for multitargeted drug exploration: (1) the design of novel molecules with a number of targets (i.e., low-molecular-weight multikinase inhibitors) [6,7,8,9,10,11]; (2) repurposing of registered and marketed drugs after a detailed study of the mechanism of action and revelation of novel targets (i.e., thalidomide, rapamycin) [12,13,14,15,16,17,18,19]; and (3) the use of biologically active compounds of natural origin and their secondary metabolites (i.e., polyphenols, flavonoids) with multiple targets and usually mild and reversible effects [20,21,22,23,24,25,26,27,28].
Steroids can be discussed as multitargeted molecules that can be applied and repurposed in various ways, including in cancer treatment. Structurally, steroids are hydrophobic molecules that are biosynthesized from cholesterol and can also be obtained from numerous terrestrial and marine sources [29,30,31]. Physiologically, steroid hormones are responsible for sex differentiation and reproduction (androgens, estrogens, progestogens), metabolism and immunity (glucocorticoids, GC), homeostasis, blood volume and electrolyte maintenance (mineralocorticoids), and calcium absorption (calciferols) [32,33]. In cancer therapy, steroids are used because of their binding to specific receptors overproduced in particular tissues. Ligand-dependent receptors activate transcription factors, which regulate the transcription of dependent genes, resulting in changes in cancer cell proliferation [34,35,36,37]. Moreover, competitive synthetic ligands for steroid hormone receptors can be designed as antagonists or selective agonists that completely or partially block receptor functions. For example, anti-androgens are used for prostate cancer therapy, and anti-estrogens are used for breast cancer treatment. GC represents a significant part of blood cancer therapy as well as supportive therapy in solid cancers [35,38,39,40,41,42,43,44,45,46].
Selective hormone receptor modulators may be safer alternatives to classic steroids. In particular, selective glucocorticoid receptor (GR) agonists/modulators (SEGRAMs) of natural and synthetic origin are considered anti-cancer and anti-inflammatory drugs with an improved therapeutic index [22,47,48,49,50,51,52,53,54,55,56,57,58,59,60]. Several SEGRAMs have entered clinical trials, but to date, no drug from the SEGRAM class has reached the pharmaceutical market [61,62,63,64,65,66,67,68]. In our previous studies, we considered Compound A (CpdA) or synephrine, originating from terrestrial plants, as templates for novel SEGRAM synthesis. The mechanism of SEGRAM biological activity is realized via GR binding as standard GCs but lacking GR dimerization, nuclear translocation, and GR-dependent gene transcription (transactivation, TA) associated with GC-related adverse effects. Protein–protein interactions of GR monomers with transcription factors (TFs) mediating therapeutic effects are fully implemented in the SEGRAM activity profile [51,52].
Marine life demonstrates infinite biodiversity, with biologically active low-molecular-weight molecules from the classes of polyketides, alkaloids, terpenoids, polyphenols, and steroids with antimicrobial, anti-cancer, anti-inflammatory, and wound-healing activities [69,70,71,72,73,74,75,76,77,78]. In this review, we aimed to follow marine sources of biologically active steroids and analyze the possibility of finding potential SEGRAMs for cancer treatment. The review is based on the literature available in the PubMed, Cochrane, and web resource ClinicalTrials.gov databases. The review covers the period from 1986 to 2025. The keywords used were “steroids”, “cancer”, and “marine-derived steroids”. The second iteration of the literature search included the keywords “selective glucocorticoid receptor agonists”, “marine-derived”, and “cancer”. In silico calculations of several marine-derived compounds were performed to support the hypothesis based on the literature data.

2. Marine-Derived Steroids with Anti-Cancer Activity

To date, the development of novel molecules has demonstrated a reverse shift to molecules of natural origin, particularly from the marine environment. Owing to geographical and topographical peculiarities, the components of marine organisms are not well studied compared to terrestrial organisms; however, technological progress has made it possible to collect organisms from deep-sea water and study their biologically active components [79]. Natural marine products frequently show favorable pharmacokinetic profiles, multiple molecular targets, and a wide spectrum of high biological activities, including anti-inflammatory, antimicrobial, antiviral, wound healing, and anti-cancer effects [27,28,80,81,82,83]. Moreover, marine-derived compounds are characterized by great structural diversity and may include polyketides, terpenoids, alkaloids, steroids, peptides, and others [84,85].
Marine ecosystems, including microorganisms, algae, sea grass, echinoderms, chordates, cnidarians, sponges, and other invertebrates and vertebrates, produce many steroids with significant anti-cancer potential (Table 1). Thus, novel steroids, 5a-cholesta-24-en-3b,20b-diol-23-one (1) and 5α-cholesta-9(11)-en-3β,20β-diol (2), were isolated from Acanthaster planci (crown of thorns starfish) and characterized by anti-cancer activity on MCF-7 breast cancer cells of luminal A subtypes [86]. Steroid dendrodoristerol (3) is found in Vietnamese nudibranch Dendrodoris fumata and demonstrates cytotoxic effects on a panel of cancer cells of different origins (hepatocellular carcinoma cells HepG2, prostate cancer cells LNCaP, breast cancer cells MCF-7, lung adenocarcinoma cells SK-LU-1, epidermal carcinoma cells KB, leukemia cells HL-60) [87]. Among the compounds isolated from the cold-water starfish Ctenodiscus crispatus, the cytotoxic effects of (25S)-5α-cholestane-3β,5,6β,15α,16β,26-hexaol (4) against hepatocellular carcinoma cells HepG2 and glioblastoma cells U87MG were reported [88].
(3E)-cholest-4-en-3,6-dione-3-oxime (5) from the marine sponge Cinachyrella australiensis also exhibited cytotoxic activity against hepatocellular carcinoma cells HepG2 [89]. Other steroid compounds from marine sponges, gracilosulfates A-G (610) from Haliclona gracilis species [90], sterols (1112) from Echinoclathria gibbosa [91], and trihydroxysterols (1316) from Psammoclema sp. [92], have been demonstrated to inhibit the proliferation of prostate cancer cells 22Rv1, PC-3, and DU-145, respectively.
Two asterosaponins, archasterosides A (17) and B (18), containing 3β,6α-dihydroxysteroid aglycons with a 9(11)-double bond and sulfate group at C-3, from the starfish Archaster typicus, showed moderate anti-cancer activity against cervical cancer cells HeLa [93]. Other asterosaponins and glycosylated steroids were found in starfish from Culcita novaeguineae, Linckia laevigata, and Halityle regularis (1922) and exhibited significant cytotoxic effects against prostate cancer cells LNCaP [94]. Spiculiferosides A (23), B (24), and C (25), isolated from the starfish Henricia leviuscula spiculifera collected from the Sea of Okhotsk, exhibited weak cytotoxic effects on melanoma SK-MEL-28, breast cancer MDA-MB-231, and colorectal cancer HCT 116 cells. However, they demonstrated the possibility of inducing cell cycle arrest and suppressing colony formation via the inhibition of CDK2, CDK4, cyclins, and MAPK/ERK signaling [95]. Steroidal 3β,21- and 3β,22-disulfates (2631), isolated from the Eastern starfish Pteraster marsippus, have also been shown to inhibit colony formation of breast cancer cells [96]. Esters of polyhydroxy steroids and long-chain fatty acids (3235) from the starfish Ceramaster patagonicus have been shown to inhibit the proliferation of breast and colorectal cancer cells and suppress their migratory activity, suggesting the role of these steroids in the therapy of metastatic cancers [97]. (23R)-Methoxycholest-5,24-dien-3β-ol (36), isolated from the marine bryozoan Cryptosula pallasiana, exhibited cytotoxic effects against leukemia, liver, and gastric cancer [98].
Three ergostane-type steroid compounds from marine-derived fungus Penicillium levitum, namely cerevisterol (37), ergosterol peroxide (38), and (3b,5a,22E)-ergosta-6,8(14),22-triene-3,5-diol (39), are characterized by an antiproliferative effect in vitro, and (39) is the most potent cytotoxin against cancer cell lines Hep-G2, A549, and MCF-7, while IC50 values for (37) and (38) were not reached [99]. Steroids from marine algae Tydemania expeditionis, (E)-stigmasta-24(28)-en-3,6-dione (40), fucosterol (41), and saringosterol (42) demonstrate cytotoxic activity against prostate cancer cells LNCaP, DU-145, and PC-3, with IC50 values in the micromolar range [80,100]. Cytotoxic and pro-apoptotic effects realized via ERK1/2-MAPK signaling inactivation in prostate carcinoma PC-3 have also been demonstrated for steroidal constituents (4354) from the sea urchin Diadema savignyi Michelin [101].
Soft corals represent a distinct class of the marine biosphere and are another source of steroid compounds with potential in cancer treatment. In particular, klyflaccisteroids (5558) from the soft coral Klyxum flaccidum exhibit cytotoxicity against colon cancer HT-29 cells, lung cancer A549 cells, and murine leukemia P388 [102]. One of the 12 novel steroids found in the soft coral Sinularia conferta, ergosta-24(28)-ene-3β,5α,6β-triol-6-acetate (59), exhibited a higher cytotoxic effect in lung and cervical cancer cells compared to camptothecin and etoposide [29]. The component of the soft coral Dendronephthya species extract, dendronestadione (60), significantly inhibits the proliferation of hepatocellular, prostate, and colorectal carcinoma cells in vitro [103]. (22E)-4α,24-dimethyl-5α-cholesta-22,24(28)-dien-3β,8β-diol (61) and (22E,24R)-7β-acetoxy-24-methyl-cholesta-5,22-dien-3β,19-diol (62) exhibited strong cytotoxic effects on breast cancer cells MCF-7 [104].
In summary, it should be noted that the number of marine-derived steroids with weak or moderate anti-cancer activity in vitro has not been mentioned above. Moreover, many steroids of marine origin have been chemically characterized, but their biological properties, particularly anti-cancer activity, have not yet been tested. And vice versa, many of the tested steroids were studied in the form of total extracts with cytotoxic effects in vitro, but they were not isolated and characterized as individual chemicals [105,106,107,108,109]. To date, none of the steroidal compounds characterized in vitro have entered in vivo preclinical studies or clinical trials, providing a broad field of investigation. Interestingly, hormone-dependent cancers, such as breast and prostate neoplasms, presented as the cancer models most frequently used for cytotoxicity evaluation and confirmation. This provides a rationale for further studies on androgen and estrogen receptor signaling as potential molecular mechanisms of action of marine-derived steroids [110]. In in vitro experiments, blood cancer cells were sensitive to several steroidal compounds, assuming specific cytotoxic activity against lymphocytes and similarity to glucocorticoid effects in hematological malignancies. In the next section, we discuss the possibility of replacing GC-based therapies with marine-derived steroids/ligands of GC receptors.
Table 1. Steroids from marine ecosystems with anti-cancer activity.
Table 1. Steroids from marine ecosystems with anti-cancer activity.
No.NameStructureSourceAnti-Cancer ActivityRef.
15α-cholesta-24-en-3β,20β-diol-23-oneMarinedrugs 23 00399 i001Crown-of-thorns starfish Acanthaster planciCytotoxic activity against luminal A breast cancer cells MCF-7 in MTT assay IC50 49 ± 1.6 μg/mL[86]
25α-cholesta-9(11)-en-3β,20β-diolMarinedrugs 23 00399 i002Crown-of-thorns starfish Acanthaster planciCytotoxic activity against luminal A breast cancer cells MCF-7 in MTT assay IC50 57.5 ± 1.5 μg/mL[86]
3DendrodoristerolMarinedrugs 23 00399 i003Sea slug Dendrodoris
fumata
Cytotoxic activity against hepatocellular carcinoma cells HepG2, prostate cancer cells LNCaP, breast cancer cells MCF-7, lung adenocarcinoma cells SK-LU-1, epidermal carcinoma cells KB, leukemia cells HL-60 in SRB assay IC50 21.63 ± 2.22, 22.22 ± 1.81, 24.53 ± 2.47, 41.19 ± 3.25, 25.34 ± 3.81, and 21.59 ± 1.38 μM[87]
4(25S)-5α-cholestane-3β,5,6β,15α,16β,26-hexaolMarinedrugs 23 00399 i004Mud star Ctenodiscus crispatusShows cytotoxic activity against hepatocellular carcinoma cells HepG2 in MTT assay[88]
5(3E)-cholest-4-en-3,6-dione-3-oximeMarinedrugs 23 00399 i005Sea sponge Cinachyrella australiensisCytotoxic activity against hepatocellular carcinoma cells HepG2 in MTT assay IC50 2.91 mg/mL[89]
6Gracilosulfate AMarinedrugs 23 00399 i006Sea sponge Haliclona gracilisCytotoxic activity against prostate cancer cell line 22Rv1 in MTT assay IC50 64.4 ± 14.9 μM[90]
7Gracilosulfate BMarinedrugs 23 00399 i007Sea sponge Haliclona gracilisCytotoxic activity against prostate cancer cell line 22Rv1 in MTT assay IC50 > 100 μM[90]
8Gracilosulfate DMarinedrugs 23 00399 i008Sea sponge Haliclona gracilisCytotoxic activity against prostate cancer cell line 22Rv1 in MTT assay IC50 > 100 μM[90]
9Gracilosulfate FMarinedrugs 23 00399 i009Sea sponge Haliclona gracilisCytotoxic activity against prostate cancer cell line 22Rv1 in MTT assay IC50 > 100 μM[90]
10Gracilosulfate GMarinedrugs 23 00399 i010Sea sponge Haliclona gracilisCytotoxic activity against prostate cancer cell line 22Rv1 in MTT assay IC50 > 100 μM[90]
11β-sitosterol-3-O-(3Z)-pentacosenoateMarinedrugs 23 00399 i011Sea sponge Echinoclathria gibbosaCytotoxic activity against prostate cancer cells PC-3 in MTT assay IC50 64 μM[91]
125α-pregna-3β-acetoxy-12β,16β-diol-20-oneMarinedrugs 23 00399 i012Sea sponge Echinoclathria gibbosaCytotoxic activity against prostate cancer cells PC-3 in MTT assay IC50 > 100 μM [91]
133α,12α,16α-trihydroxy-24ξ-ethylcholest-25-eneMarinedrugs 23 00399 i013Sea sponge PsammoclemaCytotoxic activity against prostate cancer cells DU-145 in MTT assay GI50 13 ± 1 μM[92]
143α,12α,16α-trihydroxy-24R-methylcholest-22E-eneMarinedrugs 23 00399 i014Sea sponge PsammoclemaCytotoxic activity against prostate cancer cells DU-145 in MTT assay GI50 27 ± 1 μM[92]
153α,12α,16α-trihydroxy-24-methylcholest-24(28)-eneMarinedrugs 23 00399 i015Sea sponge PsammoclemaCytotoxic activity against prostate cancer cells DU-145 in MTT assay GI50 27 ± 1 μM[92]
163α,12α,16α-trihydroxycholestaneMarinedrugs 23 00399 i016Sea sponge PsammoclemaCytotoxic activity against prostate cancer cells DU-145 in MTT assay GI50 6.7 ± 0.2 μM[92]
17Archasteroside AMarinedrugs 23 00399 i017 [93]
18Archasteroside BMarinedrugs 23 00399 i018 [93]
19Halityloside A Marinedrugs 23 00399 i019Starfish Culcita novaeguineaeCytotoxic activity against prostate cancer cells LNCaP in SRB assay IC50 48.59 ± 2.30 μM[94]
20Halityloside BMarinedrugs 23 00399 i020Starfish Culcita novaeguineaeCytotoxic activity against prostate cancer cells LNCaP in SRB assay IC50 39.68 ± 2.65 μM[94]
21Culcitoside C5Marinedrugs 23 00399 i021Starfish Culcita novaeguineaeCytotoxic activity against prostate cancer cells LNCaP in SRB assay IC50 57.08 ± 1.81 μM[94]
22Halityloside D Marinedrugs 23 00399 i022Starfish Culcita novaeguineaeCytotoxic activity against prostate cancer cells LNCaP in SRB assay IC50 31.80 ± 1.59 μM[94]
23Spiculiferosides AMarinedrugs 23 00399 i023Starfish Henricia leviuscula spiculiferaInhibition of colony formation colorectal carcinoma cells HCT 116 at concentration 40 μM was 65%[95]
24Spiculiferosides BMarinedrugs 23 00399 i024Starfish Henricia leviuscula spiculiferaInhibition of colony formation colorectal carcinoma cells HCT 116 at concentration 40 μM was 81%[95]
25Spiculiferosides CMarinedrugs 23 00399 i025Starfish Henricia leviuscula spiculiferaCytotoxic activity against colorectal carcinoma cells HCT 116 in MTS assay IC50 87.6 μM
Inhibition of colony formation colorectal carcinoma cells HCT 116 at concentration 40 μM was 87%
[95]
26(20R,22E)-24-norcholesta-5,22-diene-3β,21-diol 3,21-disulfate disodium saltMarinedrugs 23 00399 i026Starfish Pteraster marsippusInhibition of colony formation breast cancer cells T-47D at concentration 50 μM was 76%[111]
27(20R,22E)-24-nor-5α-cholest-22-ene-3β,21-diol 3,21-disulfate disodium saltMarinedrugs 23 00399 i027Starfish Pteraster marsippusInhibition of colony formation breast cancer cells T-47D at concentration 50 μM was 86%[111]
28(20R)-7-oxo-24-methylcholesta-5,24(28)-diene-3β
,21-diyl disulfate disodium salt
Marinedrugs 23 00399 i028Starfish Pteraster marsippusCytotoxic activity of the mixture of 28 and 29 against human breast carcinoma cells ZR-75-1 in MTS assay IC50 90.4 μM[96]
29(20R)-7-oxo-24-methyl-5α-cholest-24(28)-ene-3
β,21-diyl disulfate disodium salt
Marinedrugs 23 00399 i029Starfish Pteraster marsippusCytotoxic activity of the mixture of 28 and 29 against human breast carcinoma cells ZR-75-1 in MTS assay IC50 90.4 μM[96]
30(20S,22R)-24-metylcholesta-5,24-diene-3β,22-diol 3,22-disulfate disodium saltMarinedrugs 23 00399 i030Starfish Pteraster marsippusInhibition of colony formation breast cancer cells T-47D at concentration 50 μM was 71%[111]
31(20S,22R)-24-metyl-5α-cholest-24-ene-2β,3α,22-triol 3,22-disulfate disodium saltMarinedrugs 23 00399 i031Starfish Pteraster marsippusInhibition of colony formation breast cancer cells T-47D at concentration 50 μM was 79%[111]
32(25S)-5α-cholestane-3β,6β,15α,16β-tetraol-26-yl 5′Z,11′Z-octadecadienoateMarinedrugs 23 00399 i032Starfish Ceramaster patagonicusInhibitory activity against migration of colorectal carcinoma cells HCT 116 was 36%[97]
33(25S)-5α-cholestane-3β,6β,15α,16β-tetraol-26-yl 11’Z-octadecenoateMarinedrugs 23 00399 i033Starfish Ceramaster patagonicusInhibitory activity against migration of colorectal carcinoma cells HCT 116 was 73%[97]
34(25S)-5α-cholestane-3β,6β,15α,16β-tetraol-26-yl 5’Z,11’Z-eicosadienoateMarinedrugs 23 00399 i034Starfish Ceramaster patagonicusInhibitory activity against migration of colorectal carcinoma cells HCT 116 was 30%[97]
35(25S)-5α-cholestane-3β,6β,15α,16β-tetraol-26-yl 7’Z-eicosenoateMarinedrugs 23 00399 i035Starfish Ceramaster patagonicusInhibitory activity against migration of colorectal carcinoma cells HCT 116 was 24%[97]
36(23R)-methoxycholest-5,24-dien-3β-olMarinedrugs 23 00399 i036Colonial bryozoan Cryptosula pallasianaCytotoxic activity against hepatocellular carcinoma cells HepG2, gastric carcinoma cells SGC-7901, and leukemia cells HL-60 in MTT assay IC50 12.34 ± 0.12, 18.37 ± 0.17, and 17.64 ± 0.32 μM[98]
37CerevisterolMarinedrugs 23 00399 i037Marine fungus Penicillium levitumCytotoxic activity against hepatocellular carcinoma cells HepG2, lung carcinoma cells A549, and breast cancer cells MCF-7 in MTT assay was not detected[99]
38Ergosterol peroxideMarinedrugs 23 00399 i038Marine fungus Penicillium levitumCytotoxic activity against hepatocellular carcinoma cells HepG2, lung carcinoma cells A549, and breast cancer cells MCF-7 in MTT assay IC50 16.22, 22.48, and 27.11 μM[99]
39(3β,5α,22E)-ergosta-6,8(14),22-triene-3,5-diolMarinedrugs 23 00399 i039Marine fungus Penicillium levitumCytotoxic activity against hepatocellular carcinoma cells HepG2, lung carcinoma cells A549, and breast cancer cells MCF-7 in MTT assay IC50 2.89, 18.51, and 16.47 μM[99]
40(24E)-stigmasta-24(28)-en-3,6-dioneMarinedrugs 23 00399 i040Green algae Tydemania expeditionisCytotoxic activity against prostate cancer cells DU-145, prostate cancer cells PC-3, and prostate cancer cells LNCaP in MTT assay IC50 31.27 ± 1.50, 40.59 ± 3.10, and 19.80 ± 3.84 μM[100]
41FucosterolMarinedrugs 23 00399 i041Green algae Tydemania expeditionisCytotoxic activity against prostate cancer cells DU-145, prostate cancer cells PC-3, and prostate cancer cells LNCaP in MTT assay IC50 12.38 ± 2.47, 2.14 ± 0.33, and 1.38 ± 0.07 μM[100]
42SaringosterolMarinedrugs 23 00399 i042Green algae Tydemania expeditionisCytotoxic activity against prostate cancer cells DU-145, prostate cancer cells PC-3, and prostate cancer cells LNCaP in MTT assay IC50 > 50, > 50, and 41.60 ± 4.26 μM[100]
43Cholest-8-ene-3β,5α,6β,7α-tetraolMarinedrugs 23 00399 i043Sea urchin Diadema savignyiCytotoxic activity against prostate cancer cells PC-3 in MTT assay IC50 40.43 ± 1.45 μM[101]
44Cholest-8(14)-ene-3β,5α,6β,7α-tetraolMarinedrugs 23 00399 i044Sea urchin Diadema savignyiCytotoxic activity against prostate cancer cells PC-3 in MTT assay IC50 5.49 ± 0.22 μM[101]
45Cholest-7-ene-3β,5α,6β-triolMarinedrugs 23 00399 i045Sea urchin Diadema savignyiCytotoxic activity against prostate cancer cells PC-3 in MTT assay IC50 74.06 ± 3.46 μM[101]
46Cholest-7-ene-3β,5α,6α,9α-tetraolMarinedrugs 23 00399 i046Sea urchin Diadema savignyiCytotoxic activity against prostate cancer cells PC-3 in MTT assay IC50 27.41 ± 0.50 μM[101]
47Cholest-7-ene-6-one-3β,5α,9α-triolMarinedrugs 23 00399 i047Sea urchin Diadema savignyiCytotoxic activity against prostate cancer cells PC-3 in MTT assay IC50 24.40 ± 0.46 μM[101]
48Cholest-5-ene-3β,7α-diolMarinedrugs 23 00399 i048Sea urchin Diadema savignyiCytotoxic activity against prostate cancer cells PC-3 in MTT assay IC50 29.22 ± 0.17 μM[101]
49Cholest-5-ene-3β,7β-diolMarinedrugs 23 00399 i049Sea urchin Diadema savignyiCytotoxic activity against prostate cancer cells PC-3 in MTT assay IC50 27.94 ± 0.63 μM[101]
50Cholest-5-ene-7β-methoxy-3β-olMarinedrugs 23 00399 i050Sea urchin Diadema savignyiCytotoxic activity against prostate cancer cells PC-3 in MTT assay IC50 9.22 ± 0.67 μM[101]
51CampesterolMarinedrugs 23 00399 i051Sea urchin Diadema savignyiCytotoxic activity against prostate cancer cells PC-3 in MTT assay IC50 22.26 ± 0.59 μM[101]
52Cholest-5-ene-3β-sulfate sodium soltMarinedrugs 23 00399 i052Sea urchin Diadema savignyiCytotoxic activity against prostate cancer cells PC-3 in MTT assay IC50 68.87 ± 6.08 μM[101]
53Cholest-6-ene-5α,8α-epidioxy-3β-olMarinedrugs 23 00399 i053Sea urchin Diadema savignyiCytotoxic activity against prostate cancer cells PC-3 in MTT assay IC50 6.99 ± 0.28 μM[101]
54Cholest-5-ene-3β-olMarinedrugs 23 00399 i054Sea urchin Diadema savignyiCytotoxic activity against prostate cancer cells PC-3 in MTT assay was not detected[101]
55Klyflaccisteroid AMarinedrugs 23 00399 i055Soft coral Klyxum flaccidumCytotoxic activity against colon cancer cells HT-29, lung cancer cells A549, and murine leukemia cells P388 in Alamar Blue assay ED50 > 20, 7.7, and >20 μg mL−1[102]
56Klyflaccisteroid FMarinedrugs 23 00399 i056Soft coral Klyxum flaccidumCytotoxic activity against colon cancer cells HT-29, lung cancer cells A549, and murine leukemia cells P388 in Alamar Blue assay ED50 > 20, 14.5, and 17.9 μg mL−1[102]
57Klyflaccisteroid CMarinedrugs 23 00399 i057Soft coral Klyxum flaccidumCytotoxic activity against colon cancer cells HT-29, lung cancer cells A549, and murine leukemia cells P388 in Alamar Blue assay ED50 8.2, 6.1, and 10.8 μg mL−1[102]
58Klyflaccisteroid EMarinedrugs 23 00399 i058Soft coral Klyxum flaccidumCytotoxic activity against colon cancer cells HT-29 and murine leukemia cells P388 in Alamar Blue assay ED50 6.9 and 3.7 μg mL−1[102]
59Ergosta-24(28)-ene-3β,5α,6β-triol-6-acetateMarinedrugs 23 00399 i059Soft coral Sinularia confertaCytotoxic activity against lung cancer cells A549, cervical adenocarcinoma cells HeLa, and pancreatic epithelioid carcinoma cells PANC-1 in MTT assay IC50 3.64 ± 0.18, 19.34 ± 0.42, and 1.78 ± 0.69 μM[112]
60DendronestadioneMarinedrugs 23 00399 i060Soft coral DendronephthyaCytotoxic activity against hepatocellular carcinoma cells HepG2, colon cancer cells HT-29, and prostate cancer cells PC-3 in MTT assay IC50 19.1 ± 1.81, 32.4 ± 2.84, and 7.8 ± 0.80 μM[113]
61(22E)-4α,24-dimethyl-5α-cholesta-22,24(28)-dien-3β,8β-diol Marinedrugs 23 00399 i061Soft coral Litophyton mollisCytotoxic activity against hepatocellular carcinoma cells HepG2, breast cancer cells MCF-7, and lung carcinoma cells NCI-H1299 in SRB assay IC50 > 50 μM in all cases[104]
62(22E,24R)-7β-acetoxy-24-methylcholesta-5,22-dien-3β,19-diol Marinedrugs 23 00399 i062Soft coral Litophyton mollisCytotoxic activity against hepatocellular carcinoma cells HepG2, breast cancer cells MCF-7, and lung carcinoma cells NCI-H1299 in SRB assay IC50 32.5, 8.4, and 15.1 μM[104]

3. Potential Glucocorticoid Receptor Modulators from Natural Marine Products

To discuss the role of GC in cancer therapy, the signaling of the glucocorticoid receptor (GR), a well-known transcription factor (TF) and mediator of GC biological effects in cells and tissues, should be briefly explained. Binding of GC to inactive GR in the cytoplasm leads to receptor activation, homodimerization, and translocation to the nucleus. The GR-GR homodimer binds to GC-responsive elements (GREs) in DNA, resulting in induction or inhibition of the transcription of different gene subsets. Protein–protein interactions of GR monomers with other TFs are followed by the suppression of their activity [114,115]. GR-dependent inhibition of pro-proliferative and anti-apoptotic TF activity or suppression of gene transcription (transrepression, TR) mediates the therapeutic effects of GC. Induction of GR-dependent gene transcription (transactivation, TA) is mainly associated with metabolic and atrophic GC-related complications [116,117]. Therefore, the development of GC analogs of synthetic or natural origin with an improved therapeutic index and attenuated side effects is of interest, with the compounds of the SEGRAM class as an option [50,118,119,120].
GR is a member of the nuclear receptor superfamily, which also includes the estrogen receptor (ER), progesterone receptor (PR), androgen receptor (AR), mineralocorticoid receptor (MR), vitamin D receptor (VDR), and thyroid hormone receptor (ThR). Steroid receptors have a highly conserved DNA-binding domain (DBD), which allows them to bind to the responsive elements of other family members [121,122]. It could mediate the glucocorticoid-like activity of potential ER, PR, or AR ligands, and vice versa. Thus, GR can form a heterodimer with AR, modulating its activity [123,124,125,126]. The homology of GR and PR DBDs allows the sharing of responsive elements and regulation of the expression of immunophilins, oncogenes, and TFs [127,128]. Cross-talk between GR and ER occurs via protein–protein interactions of receptor monomers, followed by binding of the heterodimer to estrogen-responsive elements (EREs) as well as direct suppression of ER activity. This interaction could explain the anti-proliferative activity of GC in ER-positive breast cancer cells [129,130,131,132]. However, it should be noted that ER-GR interactions may lead to breast cancer progression and metastasis in ER-negative cancer subtypes [133,134,135,136].
To date, little is known about the GR-dependent anti-cancer activity of marine-derived steroids. Gene expression profiles in GC anti-inflammatory and anti-cancer effects significantly intersect, allowing marine-derived steroids with anti-inflammatory properties to be considered as a potential option for cancer treatment (Table 2). Thus, klyflaccisteroids from soft corals (56, 57) with cytotoxic potential also exhibit strong anti-inflammatory effects in vitro, specifically the inhibition of superoxide anion generation and elastase release in human neutrophils [102]. The fungus Penicillium oxalicum, associated with the soft coral Sinularia gaweli, produces ergostane-type sterol ester (63). This sterol ester demonstrated anti-inflammatory activity in RAW264.7 macrophage cells by inhibiting the expression of pro-inflammatory cytokines TNF-α and INF-β1 [137]. (22E,24R)-ergosta-5,7,22-trien-3β-ol (64), isolated from the Avicennia mangrove-associated marine fungus Amorosia sp., suppressed LPS-induced NO production and pro-inflammatory factors IL-6, TNF-α, and MCP-1. However, no cytotoxic activity was observed [138]. Ergosterol (65), found in the deep-sea fungus Samsoniella hepiali, inhibits NO production in LPS-activated microglia cells [139]. Similar inhibitory effects on the inflammation markers iNOS, TNF-α, IL-6, and IL-1β, at both the mRNA and protein levels in vitro, have been described for sesquiterpenoid (66) isolated from the marine-derived fungus Eutypella sp. [140], persteroid (67) isolated from the marine-derived fungus Penicillium sp. ZYX-Z-143 [141], and ergostane-type steroid components (3739) from the marine-derived fungus Penicillium levitum with cytotoxic potential against breast, lung, and liver cancer cells [99]. Non-steroidal components of a marine-derived actinomycete strain, identified as a Streptomyces sp., taking the form of ten new nine-membered bis-lactones, splenocins A-J (6877), with anti-inflammatory activity compared to GC dexamethasone in a splenocyte cytokine assay, were also described in the literature [142].
The most intriguing case in the reviewed subsets of marine-derived steroids is fucosterol (41), which has been reported to exhibit anti-cancer activity in vitro in prostate and breast cancer cells. Molecular docking of fucosterol on LXR-β, GR, TrkB, TLR2/4, BACE1, and AChE showed that fucosterol formed several hydrophobic interactions with GR via Met604, Leu608, and Phe623. The reported molecular docking data on fucosterol’s GR-binding affinity also suggest its anti-inflammatory action [143]. Following the observation that fucosterol decreases angiotensin-converting enzyme (ACE) levels in endothelial cells by inhibiting GR synthesis involved in ACE regulation [144], the interaction mode of fucosterol with GR could be considered antagonism, but further studies are needed.
Thus, the possibility of interaction with GR has been described only for one marine steroid, despite the fact that more than 1000 marine steroids, including those isolated from marine fungi and sponges, are currently known. We calculated in silico the GR interaction with some marine sponge- and fungal-derived steroidal compounds, which were previously investigated by one of us, to propose their GR binding (Table 3). Molecular docking evaluation is described in detail in [145]. The structure of GR (PDB ID 1P93) was obtained from the RCSB Protein Data Bank (https://www.rcsb.org, accessed on 27 August 2025).
3β,15β-Dihydroxy-(22E,24R)-ergosta-5,8(14),22-trien-7-one (78) has been isolated from Vietnamese marine fungus Penicillium chermesinum 2104NT-1.3 and reported as a cardioprotective agent [110]. Molecular docking calculations showed that (78) did not interact with the ligand-binding domain (LBD) of the GR. A number of new oxygenated sterol derivatives have been isolated from the marine sponge Inflatella sp., collected from the Sea of Okhotsk [146]. 24-Methylcholesta-5,24(28)-diene-3β,4α-diol (79) was not active against 6-hydroxydopamine (6-OHDA) toxicity, and 24-nor-cholesta-5,22-diene-3β,7α-diol (80) increased the 6-OHDA-treated Neuro-2a cell viability. Moreover, (80) enhanced formazan production in the MTT assay. The results indicated that (79) can interact with Arg611, and (80) interacts with Met604, Leu566, Tyr735, Phe623, and Cys736, which form LBD-GR.
Moreover, we attempted to determine whether marine non-steroidal compounds are of interest in this regard. The library of secondary metabolites isolated at various times from the fungal strains of the Collection of Marine Microorganisms of the PIBOC FEB RAS (https://kmm644.ru, accessed on 27 August 2025) was analyzed from the point of view of structural similarity with dexamethasone using the PubChem Score Matrix Service to calculated substructure key-based 2D Tanimoto similarity (https://pubchem.ncbi.nlm.nih.gov/docs/score-matrix-service (accessed on 30 August 2025)). In total, 202 compounds were analyzed, and the highest Tanimoto index of 69 was calculated for decumbenone C (81) from Aspergillus sulphureus KMM 4640 [147] and conidiogenone F (82) from Penicillium antarcticum KMM 4670 [148]. The list of compounds and full PubChem Score Matrix calculations are presented in Supplementary File S1.
Molecular docking calculations were also performed for these compounds (Figure 1, Table 3), and they showed a good possibility of binding to LBD-GR. Decumbenone C (81) formed a complex with ΔG of -8.23 kcal/mol and interacted with Arg611, Gly567, Trp600, Met604, Met601, Met646, Phe623, and Met560. Two complexes were calculated for conidiogenone F (82). The first complex, with a ΔG of −8.46 kcal/mol, included hydrophobic interactions with Gly567, Met604, Met601, Trp600, Cys736, Tyr735, Met560, Met646, and Phe623 in LBD-GR. The second complex, with a ΔG of −7.91 kcal/mol, involved hydrogen binding with Gln642 and hydrophobic interactions with Met560, Gly567, Met604, Met646, and Cys736 in LBD-GR. Figure 1 illustrates the complexes of LBD-GR with compounds exhibiting the highest affinity.
Table 2. Anti-inflammatory marine-derived compounds of steroidal and non-steroidal structures.
Table 2. Anti-inflammatory marine-derived compounds of steroidal and non-steroidal structures.
No.NameStructureSourceAnti-Inflammatory Effects Reference
56Klyflaccisteroid FMarinedrugs 23 00399 i063Soft coral Klyxum flaccidumActivity in inhibiting the superoxide anion generation 88.26 ± 35.86% at 10 μM and activity in inhibiting elastase release 104.22 ± 6.55% at 10 μM in N-formyl-methionyl-leucyl-phenylalanine/
cytochalasin B (fMLP/CB)-induced neutrophils
[102]
57Klyflaccisteroid CMarinedrugs 23 00399 i064Soft coral Klyxum flaccidumActivity in inhibiting the superoxide anion generation 76.24 ± 5.64% at 10 μM and activity in inhibiting elastase release 88.38 ± 1.19% at 10 μM in N-formyl-methionyl-leucyl-phenylalanine/
cytochalasin B (fMLP/CB)-induced neutrophils
[102]
63(22E,24S)-9a,15a-dihydroxyergosta-4,6,8(14),22-
tetraen-3-one 15-palmitate
Marinedrugs 23 00399 i065Marine fungus Penicillium oxalicum HL-44Inhibition of expression of pro-inflammatory cytokines TNF-α and INF-β1 on DMXAA-stimulated Raw264.7 cells by 68% and 94% at 20 μM[137]
64(22E, 24R)-ergosta-5,7,22-trien-3β-olMarinedrugs 23 00399 i066Mangrove fungus Amorosia sp.Inhibition of expression of pro-inflammatory cytokines TNF-α, IL-6, and MCP-1 on LPS-activated RAW264.7 M1-type cells by 55%, 50%, and 50% at 10 μM[138]
65ErgosterolMarinedrugs 23 00399 i067Marine fungus Samsoniella hepiali W7Inhibition of NO production in LPS-activated BV-2-microglia cells by 32.9 ± 1.6% at 1 μM[139]
66ArctiolMarinedrugs 23 00399 i068Marine fungus Eutypella sp. F0219Inhibition of NO production in LPS-treated BV-2-microglia cells by 71% at 20 μM[140]
67PersteroidMarinedrugs 23 00399 i069Marine fungus Penicillium sp. ZYX-Z-143NO half-maximal inhibitory concentration on LPS-stimulated RAW 264.7 cells IC50 25.81 ± 0.92 μM[141]
37CerevisterolMarinedrugs 23 00399 i070Marine fungus Penicillium levitumNO half-maximal inhibitory concentration on LPS-stimulated RAW 264.7 cells IC50 25.45 μg/mL[99]
38Ergosterol peroxideMarinedrugs 23 00399 i071Marine fungus Penicillium levitumNO half-maximal inhibitory concentration on LPS-stimulated RAW 264.7 cells IC50 2.85 μg/mL[99]
39(3β,5α,22E)-ergosta-6,8(14),22-triene-3,5-diolMarinedrugs 23 00399 i072Marine fungus Penicillium levitumNO half-maximal inhibitory concentration on LPS-stimulated RAW 264.7 cells IC50 2.79 μg/mL[99]
68Splenocin AMarinedrugs 23 00399 i073Marine bacterium Streptomyces sp.Inhibition of expression of pro-inflammatory cytokines IL-5 on TH2 cells (helper T lymphocytes) IC50 3.1 ± 1.2 nM[142]
69Splenocin BMarinedrugs 23 00399 i074Marine bacterium Streptomyces sp.Inhibition of expression of pro-inflammatory cytokines IL-5 and IL-13 on TH2 cells (helper T lymphocytes) IC50 1.8 ± 0.2 and 1.6 ± 0.02 nM[142]
70Splenocin CMarinedrugs 23 00399 i075Marine bacterium Streptomyces sp.Inhibition of expression of pro-inflammatory cytokines IL-5 and IL-13 on TH2 cells (helper T lymphocytes) IC50 6.7 ± 0.2 and 7.3 ± 4.2 nM[142]
71Splenocin DMarinedrugs 23 00399 i076Marine bacterium Streptomyces sp.Inhibition of expression of pro-inflammatory cytokines IL-5 and IL-13 on TH2 cells (helper T lymphocytes) IC50 47.9 ± 2.9 and 43.7 ± 3.5 nM[142]
72Splenocin EMarinedrugs 23 00399 i077Marine bacterium Streptomyces sp.Inhibition of expression of pro-inflammatory cytokines IL-5 and IL-13 on TH2 cells (helper T lymphocytes) IC50 16.6 ± 1.8 and 15.9 ± 1.1 nM[142]
73Splenocin FMarinedrugs 23 00399 i078Marine bacterium Streptomyces sp.Inhibition of expression of pro-inflammatory cytokines IL-5 and IL-13 on TH2 cells (helper T lymphocytes) IC50 9.4 ± 2.8 and 6.8 ± 0.3 nM[142]
74Splenocin GMarinedrugs 23 00399 i079Marine bacterium Streptomyces sp.Inhibition of expression of pro-inflammatory cytokines IL-5 and IL-13 on TH2 cells (helper T lymphocytes) IC50 5 ± 0.4 and 5.2 ± 0.1 nM[142]
75Splenocin HMarinedrugs 23 00399 i080Marine bacterium Streptomyces sp.Inhibition of expression of pro-inflammatory cytokines IL-5 and IL-13 on TH2 cells (helper T lymphocytes) IC50 4.3 ± 0.5 and 5.1 ± 0.1 nM[142]
76Splenocin IMarinedrugs 23 00399 i081Marine bacterium Streptomyces sp.Inhibition of expression of pro-inflammatory cytokines IL-5 and IL-13 on TH2 cells (helper T lymphocytes) IC50 15.8 ± 1.0 and 15.2 ± 1.3 nM[142]
77Splenocin JMarinedrugs 23 00399 i082Marine bacterium Streptomyces sp.Inhibition of expression of pro-inflammatory cytokines IL-5 and IL-13 on TH2 cells (helper T lymphocytes) IC50 1022.7 ± 52.3 and 826.3 ± 187.6 nM[142]
41FucosterolMarinedrugs 23 00399 i083Green algae Tydemania expeditionisHydrophobic interaction with GR via Leu563, Phe623, Leu608, and Met604 (molecular docking analysis)[143]
Interestingly, decaline polyketide decumbenone C (81) exhibited potent cytotoxic activity against SK-MEL-5 human melanoma cells, with an IC50 of 0.9 µM, and inhibited colony formation at 0.25 µM [147]. Cyclopiane diterpene conidiogenone F (82) exhibited weak antimicrobial activity and was non-toxic to H9c2 cardiomyocytes [148]. However, its bioactivity has not been studied in detail because of difficulties in isolating it from fungal extracts.
Table 3. Molecular docking calculations of natural marine compounds with GR (PDB ID 1P93).
Table 3. Molecular docking calculations of natural marine compounds with GR (PDB ID 1P93).
No.CompoundChemical StructureΔG, kcal/molFF Score, kcal/molH-BindingHydrophobic Interactions
783β,15β-Dihydroxy-(22E, 24R)-ergosta-5,8(14),22-trien-7-oneMarinedrugs 23 00399 i084−7.23−1226.71-Val538, ILe539, Lys576, Ala573, Leu544, Trp577
7924-Methylcholesta-5,24(28)-diene-3β,4α-diolMarinedrugs 23 00399 i085−7.40−1204.67H29 Arg611 2.607Val543, Trp610, Tyr660
8024-nor-Cholesta-5,22-diene-3β,7α-diolMarinedrugs 23 00399 i086−6.37−1165.36-Met604, Leu566, Leu732, Asn630, Leu563, Tyr735, Phe623, Leu608, Cys736
81Decumbenone CMarinedrugs 23 00399 i087−8.23−1298.75H29 Arg611 O 2.666
H24 Asn564 1.682
Gly567, Trp600, Met604, Met601, Leu732, Met646, Phe623, Leu563, Met560
82Conidiogenone FMarinedrugs 23 00399 i088−8.46−1229.36-Gly567, Met604, Met601, Leu732, Trp600, Cys736, Tyr735, Met560, Leu563, Met646, Phe623
−7.91−1211.54H29 Gln642 2.103Met560, Leu563, Leu753, Gly567, Met604, Met646, Leu732, Cys736
DexamethasoneMarinedrugs 23 00399 i089−10.19−1206.31H29 Arg611 O 2.146
H27 Gln642 2.351
H26 Thr739 2.115
Met560, Leu566, Gly567, Trp600, Met601, Met604, Phe623, Met646, Tyr735, Cys736, Thr739, Ile747
Structural similarity analysis involves the study of the presence of substructural key elements, along with the distance between pharmacophores or functional groups in compounds; at the same time, the detailed structure and biogenetic origin are not taken into account. This allowed us to select decalin polyketide (81) and diterpene derivative (82) as compounds structurally similar to dexamethasone, and modular docking confirmed their prospects as GR ligands. Moreover, 10 marine fungal metabolites had Tanimoto indexes in the range of 52–66, and 30 compounds had Tanimoto indexes in the range of 41–49 (Supplementary File S1); these 40 compounds may also be promising GR ligands. We propose that the structural similarity of non-steroidal compounds to steroids may ensure their biological activity but reduce steroid-dependent side effects. This clearly requires additional in-depth research.

4. Conclusions

In conclusion, it should be mentioned that marine ecosystems represent a significant source of natural steroids with potential applications in anti-cancer and anti-inflammatory therapies. More specifically, ligands of glucocorticoid receptors and putative selective glucocorticoid receptor agonists/modulators can be found among marine-derived compounds and can be characterized by their chemical structure and biological activity. Evaluation of the affinity to the glucocorticoid receptor and the assessment of the expression of marker genes specific for glucocorticoid receptor activity could be a useful set of methods for the screening of biological activity. The previously published review paper cited in the present manuscript primarily aimed to summarize marine-derived steroids with various biological effects. The novelty of the present review lies not only in highlighting the dual anti-cancer and anti-inflammatory effects mimicking the action of the specific class of steroids, glucocorticoids, but also in the description of the glucocorticoid receptor affinity of several compounds available in the Collection of Marine Microorganisms of the PIBOC FEB RAS. Future directions in this field could involve the experimental validation of the biological effects of the compounds presented in this review and the study of the molecular mechanisms underlying their action. The lead compounds are assumed to bind to the glucocorticoid receptor, revealing anti-cancer and anti-inflammatory effects and, ideally, a “dissociated” activity profile with a shift towards GR TR. In the case of successful proof-of-concept studies, efforts could be made to develop a finished dosage form of the proposed SEGRAMs, followed by preclinical studies of the safety and efficacy of the leader compounds, with further design of first-in-human clinical trials. The results of this study could have translational potential not only in terms of cancer therapy but also for the treatment of inflammatory and autoimmune diseases, which affect millions of patients worldwide.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/md23100399/s1, Table S1. The list of metabolites isolated from fungal strains of the Collection of Marine Microorganisms PIBOC FEB RAS [149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164].

Author Contributions

Conceptualization, E.M.Z., E.A.Y. and E.A.L.; methodology, E.A.Y.; software, E.D.S. and E.A.Y.; investigation, E.A.Y. and E.A.L.; resources, E.A.Y. and E.A.L.; writing—original draft preparation, E.M.Z., E.D.S., E.A.Y. and E.A.L.; writing—review and editing, E.M.Z., E.A.Y. and E.A.L.; visualization, E.D.S. and E.A.Y.; supervision, E.A.L.; project administration, E.A.L.; funding acquisition, E.A.L. All authors have read and agreed to the published version of the manuscript.

Funding

The work was funded by the Russian Science Foundation, grant number 23-15-00321 (to E.A.L.).

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ACEAngiotensin-Converting Enzyme
AChEAcetyl Cholinesterase
ARAndrogen Receptor
BACE1Beta-Secretase 1
CDKCyclin-Dependent Kinase
CpdACompound A
DBDDNA-Binding Domain
EREstrogen Receptor
EREEstrogen Response Element
ERKExtracellular-Regulated Kinase
GCGlucocorticoid
GRGlucocorticoid Receptor
GREGlucocorticoid Response Element
ILInterleukin
INFInterferon
iNOSInducible Nitric Oxide Synthase
LPSLipopolysaccharide
LXR-βLiver X Receptor Beta
MAPKMitogen-Activated Protein Kinase
MCP-1Monocyte Chemoattractant Protein-1
MRMineralocorticoid Receptor
PRProgesterone Receptor
SEGRAMsSelective Glucocorticoid Receptor Agonists/Modulators
TATransactivation
TFTranscription Factor
ThRThyroid Hormone Receptor
TLRToll-Like Receptor
TNF-αTumor Necrosis Factor Alpha
TRTransrepression
TrkBTropomyosin Receptor Kinase B
VDRVitamin D Receptor

References

  1. Hanahan, D.; Weinberg, R.A. The Hallmarks of Cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [PubMed]
  2. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  3. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef]
  4. Mellinghoff, I.K.; Sawyers, C.L. The Emergence of Resistance to Targeted Cancer Therapeutics. Pharmacogenomics 2002, 3, 603–623. [Google Scholar] [CrossRef]
  5. Khamisipour, G.; Jadidi-Niaragh, F.; Jahromi, A.S.; Zandi, K.; Hojjat-Farsangi, M. Mechanisms of Tumor Cell Resistance to the Current Targeted-Therapy Agents. Tumor Biol. 2016, 37, 10021–10039. [Google Scholar] [CrossRef]
  6. Gentile, C.; Martorana, A.; Lauria, A.; Bonsignore, R. Kinase Inhibitors in Multitargeted Cancer Therapy. Curr. Med. Chem. 2017, 24, 1671–1686. [Google Scholar] [CrossRef]
  7. Valerio, L.; Matrone, A. Multikinase and Highly Selective Kinase Inhibitors in the Neoadjuvant Treatment of Patients with Thyroid Cancer. Explor. Target. Antitumor Ther. 2025, 6, 1002291. [Google Scholar] [CrossRef]
  8. Buzatu, I.M.; Tataranu, L.G.; Duta, C.; Stoian, I.; Alexandru, O.; Dricu, A. A Review of FDA-Approved Multi-Target Angiogenesis Drugs for Brain Tumor Therapy. Int. J. Mol. Sci. 2025, 26, 2192. [Google Scholar] [CrossRef]
  9. Díaz Vico, T.; Martínez-Amores Martínez, B.; Mihic Góngora, L.; Jiménez-Fonseca, P.; Peinado Martín, P.; Grao Torrente, I.; García Muñoz-Nájar, A.; Durán-Poveda, M. Systemic Therapeutic Options in Radioiodine-Refractory Differentiated Thyroid Cancer: Current Indications and Optimal Timing. Cancers 2025, 17, 1800. [Google Scholar] [CrossRef]
  10. Flauto, F.; Damiano, V. The Efficacy and Safety of Multi-Kinase Inhibitors in Adrenocortical Carcinoma: A Systematic Review and Single-Arm Meta-Analysis. Cancers 2025, 17, 2004. [Google Scholar] [CrossRef]
  11. Guo, C.; Gasparian, A.V.; Zhuang, Z.; Bosykh, D.A.; Komar, A.A.; Gudkov, A.V.; Gurova, K. V 9-Aminoacridine-Based Anticancer Drugs Target the PI3K/AKT/MTOR, NF-ΚB and P53 Pathways. Oncogene 2009, 28, 1151–1161. [Google Scholar] [CrossRef]
  12. Gupta, S.C.; Sung, B.; Prasad, S.; Webb, L.J.; Aggarwal, B.B. Cancer Drug Discovery by Repurposing: Teaching New Tricks to Old Dogs. Trends Pharmacol. Sci. 2013, 34, 508–517. [Google Scholar] [CrossRef]
  13. Swamidass, S.J. Mining Small-Molecule Screens to Repurpose Drugs. Brief. Bioinform. 2011, 12, 327–335. [Google Scholar] [CrossRef] [PubMed]
  14. Lesovaya, E.; Agarwal, S.; Readhead, B.; Vinokour, E.; Baida, G.; Bhalla, P.; Kirsanov, K.; Yakubovskaya, M.; Platanias, L.C.; Dudley, J.T.; et al. Rapamycin Modulates Glucocorticoid Receptor Function, Blocks Atrophogene REDD1, and Protects Skin from Steroid Atrophy. J. Investig. Dermatol. 2018, 138, 1935–1944. [Google Scholar] [CrossRef] [PubMed]
  15. Blatt, J.; Corey, S.J. Drug Repurposing in Pediatrics and Pediatric Hematology Oncology. Drug Discov. Today 2013, 18, 4–10. [Google Scholar] [CrossRef] [PubMed]
  16. McCabe, B.; Liberante, F.; Mills, K.I. Repurposing Medicinal Compounds for Blood Cancer Treatment. Ann. Hematol. 2015, 94, 1267–1276. [Google Scholar] [CrossRef]
  17. Koval, A.; Ahmed, K.; Katanaev, V.L. Inhibition of Wnt Signalling and Breast Tumour Growth by the Multi-Purpose Drug Suramin through Suppression of Heterotrimeric G Proteins and Wnt Endocytosis. Biochem. J. 2016, 473, 371–381. [Google Scholar] [CrossRef]
  18. Xi, Z.; Jie, W.; Long, Z.; Shasha, S. A Review of Thalidomide and Digestive System Related Diseases. Front. Oncol. 2025, 15, 1543757. [Google Scholar] [CrossRef]
  19. de Miranda Drummond, P.L.; de Araújo Silva, C.; de Souza, J.E.; Magno, B.A.R.; Battaglia, G.M.; Candido, R.C.F.; Menezes de Pádua, C.A.; Pereira, B.G. Efficacy and Safety of Thalidomide for Oncology-Related Uses Approved in Brazil: An Overview of Systematic Reviews. J. Oncol. Pharm. Pract. 2025; ahead of print. [Google Scholar] [CrossRef]
  20. Chen, Y.; Yu, D.; Zhu, D.; Muthusamy, S.; Deshpande, M.; Kiruthiga, N.; Theivendren, P.; Rajalakshmi, K.; Wu, S.; Zhu, C. Exploring Alkaloids and Flavonoids from Natural Sources: Emerging Natural Agents for Inhibiting Cervical Cancer Progression through Apoptosis Induction, Anti-Inflammatory Effects, and Oxidative Stress Reduction. Pathol. Res. Pr. 2025, 272, 156092. [Google Scholar] [CrossRef]
  21. Al Khalily, I.; Megantara, S.; Aulifa, D. Targeting Molecular Pathways in Breast Cancer Using Plant-Derived Bioactive Compounds: A Comprehensive Review. J. Exp. Pharmacol. 2025, 17, 375–401. [Google Scholar] [CrossRef]
  22. Dodonova, S.A.; Zhidkova, E.M.; Kryukov, A.A.; Valiev, T.T.; Kirsanov, K.I.; Kulikov, E.P.; Budunova, I.V.; Yakubovskaya, M.G.; Lesovaya, E.A. Synephrine and Its Derivative Compound A: Common and Specific Biological Effects. Int. J. Mol. Sci. 2023, 24, 17537. [Google Scholar] [CrossRef]
  23. Zhang, Y.; Zhang, Y.; Yao, S.; Chen, L.; Dong, Q.; Luo, R.; Zheng, K.; Liu, J.; Liu, Y.; Chen, Y.; et al. Inflammatory Bowel Disease Induces Colorectal Cancer: Risk Factors, Triggering Mechanisms, and Treatment with Phyto-Derivatives. Phytother. Res. 2025, 39, 3386–3418. [Google Scholar] [CrossRef]
  24. Coşkun, N.; Sarıtaş, S.; Bechelany, M.; Karav, S. Polyphenols in Foods and Their Use in the Food Industry: Enhancing the Quality and Nutritional Value of Functional Foods. Int. J. Mol. Sci. 2025, 26, 5803. [Google Scholar] [CrossRef] [PubMed]
  25. Menchinskaya, E.S.; Chingizova, E.A.; Pislyagin, E.A.; Yurchenko, E.A.; Klimovich, A.A.; Zelepuga, E.A.; Aminin, D.L.; Avilov, S.A.; Silchenko, A.S. Mechanisms of Action of Sea Cucumber Triterpene Glycosides Cucumarioside A0-1 and Djakonovioside A Against Human Triple-Negative Breast Cancer. Mar. Drugs 2024, 22, 474. [Google Scholar] [CrossRef] [PubMed]
  26. Borkunov, G.V.; Leshchenko, E.V.; Berdyshev, D.V.; Popov, R.S.; Chingizova, E.A.; Shlyk, N.P.; Gerasimenko, A.V.; Kirichuk, N.N.; Khudyakova, Y.V.; Chausova, V.E.; et al. New Piperazine Derivatives Helvamides B–C from the Marine-Derived Fungus Penicillium Velutinum ZK-14 Uncovered by OSMAC (One Strain Many Compounds) Strategy. Nat. Prod. Bioprospect 2024, 14, 32. [Google Scholar] [CrossRef]
  27. Yurchenko, A.N.; Girich, E.V.; Yurchenko, E.A. Metabolites of Marine Sediment-Derived Fungi: Actual Trends of Biological Activity Studies. Mar. Drugs 2021, 19, 88. [Google Scholar] [CrossRef]
  28. Yurchenko, E.A.; Yurchenko, A.N.; Van Minh, C.; Aminin, D.L. Achievements in the Study of Marine Low-Molecular Weight Biologically Active Metabolites from the Vietnamese Territorial Waters as a Result of Expeditions Aboard the Research Vessel ‘Akademik Oparin’ (2004–2017). Chem. Biodivers. 2019, 16, e1800654. [Google Scholar] [CrossRef]
  29. Hajdaś, G.; Koenig, H.; Pospieszny, T. Recent Advances in Steroid Discovery: Structural Diversity and Bioactivity of Marine and Terrestrial Steroids. Int. J. Mol. Sci. 2025, 26, 3203. [Google Scholar] [CrossRef]
  30. Hu, J.; Zhang, Z.; Shen, W.-J.; Azhar, S. Cellular Cholesterol Delivery, Intracellular Processing and Utilization for Biosynthesis of Steroid Hormones. Nutr. Metab. 2010, 7, 47. [Google Scholar] [CrossRef]
  31. Singh, R.; Bansal, R. Revisiting the Role of Steroidal Therapeutics in the 21st Century: An Update on FDA Approved Steroidal Drugs (2000–2024). RSC Med. Chem. 2025, 16, 2902–2918. [Google Scholar] [CrossRef]
  32. Samuel, S.; Nguyen, T.; Choi, H.A. Pharmacologic Characteristics of Corticosteroids. J. Neurocrit. Care 2017, 10, 53–59. [Google Scholar] [CrossRef]
  33. DeLuca, H.F. Overview of General Physiologic Features and Functions of Vitamin D. Am. J. Clin. Nutr. 2004, 80, 1689S–1696S. [Google Scholar] [CrossRef]
  34. Fuller, P.J.; Yang, J.; Young, M.J.; Cole, T.J. Mechanisms of Ligand-Mediated Modulation of Mineralocorticoid Receptor Signaling. Mol. Cell. Endocrinol. 2025, 600, 112504. [Google Scholar] [CrossRef] [PubMed]
  35. Martinez, G.J.; Appleton, M.; Kipp, Z.A.; Loria, A.S.; Min, B.; Hinds, T.D. Glucocorticoids, Their Uses, Sexual Dimorphisms, and Diseases: New Concepts, Mechanisms, and Discoveries. Physiol. Rev. 2024, 104, 473–532. [Google Scholar] [CrossRef] [PubMed]
  36. Mauvais-Jarvis, F.; Lange, C.A.; Levin, E.R. Membrane-Initiated Estrogen, Androgen, and Progesterone Receptor Signaling in Health and Disease. Endocr. Rev. 2022, 43, 720–742. [Google Scholar] [CrossRef] [PubMed]
  37. Agnoletto, A.; Brisken, C. Hormone Signaling in Breast Development and Cancer. Adv. Exp. Med. Biol. 2025, 1464, 279–307. [Google Scholar]
  38. Quistini, A.; Chierigo, F.; Fallara, G.; Depalma, M.; Tozzi, M.; Maggi, M.; Jannello, L.M.I.; Pellegrino, F.; Mantica, G.; Terracciano, D.; et al. Androgen Receptor Signalling in Prostate Cancer: Mechanisms of Resistance to Endocrine Therapies. Res. Rep. Urol. 2025, 17, 211–223. [Google Scholar] [CrossRef]
  39. Chakrabarti, D.; Albertsen, P.; Adkins, A.; Kishan, A.; Murthy, V.; Parker, C.; Pathmanathan, A.; Reid, A.; Sartor, O.; Van As, N.; et al. The Contemporary Management of Prostate Cancer. CA Cancer J. Clin. 2025. Early View. [Google Scholar] [CrossRef]
  40. Mansour, R.; Abunasser, M.; Sharaf, B.; Abdel-Razeq, H. Update in the Clinical Utilization of Chemoprevention for Breast Cancer: A Narrative Review. Front. Oncol. 2025, 15, 1435253. [Google Scholar] [CrossRef]
  41. Zulkipli, N.N.; Zakaria, R.; Wan Taib, W.R. Bibliometric Analysis of The Global Research Trends on The Application of Tamoxifen in The Treatment of Breast Cancer Over The Past 50 Years. Malays. J. Med. Sci. 2025, 32, 35–55. [Google Scholar] [CrossRef]
  42. Hiltunen, J.; Helminen, L.; Paakinaho, V. Glucocorticoid Receptor Action in Prostate Cancer: The Role of Transcription Factor Crosstalk. Front. Endocrinol. 2024, 15, 1437179. [Google Scholar] [CrossRef] [PubMed]
  43. Posani, S.H.; Gillis, N.E.; Lange, C.A. Glucocorticoid Receptors Orchestrate a Convergence of Host and Cellular Stress Signals in Triple Negative Breast Cancer. J. Steroid Biochem. Mol. Biol. 2024, 243, 106575. [Google Scholar] [CrossRef] [PubMed]
  44. Clark, A.B.; Conzen, S.D. Glucocorticoid Receptor-Mediated Oncogenic Activity Is Dependent on Breast Cancer Subtype. J. Steroid Biochem. Mol. Biol. 2024, 243, 106518. [Google Scholar] [CrossRef] [PubMed]
  45. Zhidkova, E.M.; Lylova, E.S.; Grigoreva, D.D.; Kirsanov, K.I.; Osipova, A.V.; Kulikov, E.P.; Mertsalov, S.A.; Belitsky, G.A.; Budunova, I.; Yakubovskaya, M.G.; et al. Nutritional Sensor REDD1 in Cancer and Inflammation: Friend or Foe? Int. J. Mol. Sci. 2022, 23, 9686. [Google Scholar] [CrossRef]
  46. Faggiano, A.; Mazzilli, R.; Natalicchio, A.; Adinolfi, V.; Argentiero, A.; Danesi, R.; D’Oronzo, S.; Fogli, S.; Gallo, M.; Giuffrida, D.; et al. Corticosteroids in Oncology: Use, Overuse, Indications, Contraindications. An Italian Association of Medical Oncology (AIOM)/Italian Association of Medical Diabetologists (AMD)/Italian Society of Endocrinology (SIE)/Italian Society of Pharmacology (SIF) Multidisciplinary Consensus Position Paper. Crit. Rev. Oncol. Hematol. 2022, 180, 103826. [Google Scholar] [CrossRef]
  47. Lesovaya, E.A.; Yemelyanov, A.Y.; Kirsanov, K.I.; Yakubovskaya, M.G.; Budunova, I.V. Antitumor Effect of Non-Steroid Glucocorticoid Receptor Ligand CpdA on Leukemia Cell Lines CEM and K562. Biochemistry 2011, 76, 1242–1252. [Google Scholar] [CrossRef]
  48. Lesovaya, E.; Yemelyanov, A.; Kirsanov, K.; Popa, A.; Belitsky, G.; Yakubovskaya, M.; Gordon, L.I.; Rosen, S.T.; Budunova, I. Combination of a Selective Activator of the Glucocorticoid Receptor Compound A with a Proteasome Inhibitor as a Novel Strategy for Chemotherapy of Hematologic Malignancies. Cell Cycle 2013, 12, 133–144. [Google Scholar] [CrossRef]
  49. Lesovaya, E.; Yemelyanov, A.; Swart, A.C.; Swart, P.; Haegeman, G.; Budunova, I. Discovery of Compound A—A Selective Activator of the Glucocorticoid Receptor with Anti-Inflammatory and Anti-Cancer Activity. Oncotarget 2015, 6, 30730–30744. [Google Scholar] [CrossRef]
  50. Lesovaya, E.A.; Chudakova, D.; Baida, G.; Zhidkova, E.M.; Kirsanov, K.I.; Yakubovskaya, M.G.; Budunova, I.V. The Long Winding Road to the Safer Glucocorticoid Receptor (GR) Targeting Therapies. Oncotarget 2022, 13, 408–424. [Google Scholar] [CrossRef]
  51. Zhidkova, E.M.; Tilova, L.R.; Fetisov, T.I.; Kirsanov, K.I.; Kulikov, E.P.; Enikeev, A.D.; Budunova, I.V.; Badun, G.A.; Chernysheva, M.G.; Shirinian, V.Z.; et al. Synthesis and Anti-Cancer Activity of the Novel Selective Glucocorticoid Receptor Agonists of the Phenylethanolamine Series. Int. J. Mol. Sci. 2024, 25, 8904. [Google Scholar] [CrossRef]
  52. Zhidkova, E.M.; Oleynik, E.S.; Mikhina, E.A.; Stepanycheva, D.V.; Grigoreva, D.D.; Grebenkina, L.E.; Gordeev, K.V.; Savina, E.D.; Matveev, A.V.; Yakubovskaya, M.G.; et al. Synthesis and Anti-Cancer Activity In Vitro of Synephrine Derivatives. Biomolecules 2024, 15, 2. [Google Scholar] [CrossRef]
  53. Clarisse, D.; Van Moortel, L.; Van Leene, C.; Gevaert, K.; De Bosscher, K. Glucocorticoid Receptor Signaling: Intricacies and Therapeutic Opportunities. Trends Biochem. Sci. 2024, 49, 431–444. [Google Scholar] [CrossRef]
  54. Van Moortel, L.; Gevaert, K.; De Bosscher, K. Improved Glucocorticoid Receptor Ligands: Fantastic Beasts, but How to Find Them? Front. Endocrinol. 2020, 11, 559673. [Google Scholar] [CrossRef]
  55. Hsu, S.-J.; He, M.; Salomé-Abarca, L.F.; Choi, Y.H.; Wang, M. Uncovering Anti-Inflammatory Activity of Ginsenoside Rg1 in a Wound-Inured Zebrafish Model by GC-MS-Based Chemical Profiling. Planta Med. 2025, 91, 609–620. [Google Scholar] [CrossRef] [PubMed]
  56. Jakob, F.; Hennen, S.; Gautrois, M.; Khalil, F.; Lockhart, A. Novel Selective Glucocorticoid Receptor Modulator GRM-01 Demonstrates Dissociation of Anti-Inflammatory Effects from Adverse Effects on Glucose and Bone Metabolism. Front. Pharmacol. 2025, 16, 1542351. [Google Scholar] [CrossRef] [PubMed]
  57. Zhidkova, E.M.; Lylova, E.S.; Savinkova, A.V.; Mertsalov, S.A.; Kirsanov, K.I.; Belitsky, G.A.; Yakubovskaya, M.G.; Lesovaya, E.A. A Brief Overview of the Paradoxical Role of Glucocorticoids in Breast Cancer. Breast Cancer 2020, 14, 1178223420974667. [Google Scholar] [CrossRef] [PubMed]
  58. He, M.; Halima, M.; Xie, Y.; Schaaf, M.J.M.; Meijer, A.H.; Wang, M. Ginsenoside Rg1 Acts as a Selective Glucocorticoid Receptor Agonist with Anti-Inflammatory Action without Affecting Tissue Regeneration in Zebrafish Larvae. Cells 2020, 9, 1107. [Google Scholar] [CrossRef]
  59. Karra, A.G.; Tziortziou, M.; Kylindri, P.; Georgatza, D.; Gorgogietas, V.A.; Makiou, A.; Krokida, A.; Tsialtas, I.; Kalousi, F.D.; Papadopoulos, G.E.; et al. Boswellic Acids and Their Derivatives as Potent Regulators of Glucocorticoid Receptor Actions. Arch. Biochem. Biophys. 2020, 695, 108656. [Google Scholar] [CrossRef]
  60. Morsy, M.A.; Patel, S.S.; El-Sheikh, A.A.K.; Savjani, J.K.; Nair, A.B.; Shah, J.N.; Venugopala, K.N. Computational and Biological Comparisons of Plant Steroids as Modulators of Inflammation through Interacting with Glucocorticoid Receptor. Mediat. Inflamm. 2019, 2019, 3041438. [Google Scholar] [CrossRef]
  61. Brown, M.N.; Fuhr, R.; Beier, J.; Su, H.-L.; Chen, Y.; Forsman, H.; Hamrén, U.W.; Jackson, H.; Aggarwal, A. Efficacy and Safety of AZD7594, an Inhaled Non-Steroidal Selective Glucocorticoid Receptor Modulator, in Patients with Asthma: A Phase 2a Randomized, Double Blind, Placebo-Controlled Crossover Trial. Respir. Res. 2019, 20, 37. [Google Scholar] [CrossRef]
  62. Cheng, F.; Shen, T.; Zhang, F.; Lei, C.; Zhu, Y.; Luo, G.; Xiao, D. Bioequivalence Study of Fluticasone Propionate Nebuliser Suspensions in Healthy Chinese Subjects. Front. Pharmacol. 2025, 15, 1452596. [Google Scholar] [CrossRef]
  63. Hunt, H.; Donaldson, K.; Strem, M.; Zann, V.; Leung, P.; Sweet, S.; Connor, A.; Combs, D.; Belanoff, J. Assessment of Safety, Tolerability, Pharmacokinetics, and Pharmacological Effect of Orally Administered CORT125134: An Adaptive, Double-Blind, Randomized, Placebo-Controlled Phase 1 Clinical Study. Clin. Pharmacol. Drug Dev. 2018, 7, 408–421. [Google Scholar] [CrossRef]
  64. Kuna, P.; Aurivillius, M.; Jorup, C.; Prothon, S.; Taib, Z.; Edsbäcker, S. Efficacy and Tolerability of an Inhaled Selective Glucocorticoid Receptor Modulator—AZD5423—In Chronic Obstructive Pulmonary Disease Patients: Phase II Study Results. Basic. Clin. Pharmacol. Toxicol. 2017, 121, 279–289. [Google Scholar] [CrossRef]
  65. Werkström, V.; Prothon, S.; Ekholm, E.; Jorup, C.; Edsbäcker, S. Safety, Pharmacokinetics and Pharmacodynamics of the Selective Glucocorticoid Receptor Modulator AZD5423 after Inhalation in Healthy Volunteers. Basic. Clin. Pharmacol. Toxicol. 2016, 119, 574–581. [Google Scholar] [CrossRef] [PubMed]
  66. Conrado, D.J.; Krishnaswami, S.; Shoji, S.; Kolluri, S.; Hey-Hadavi, J.; McCabe, D.; Rojo, R.; Tammara, B.K. Predicting the Probability of Successful Efficacy of a Dissociated Agonist of the Glucocorticoid Receptor from Dose–Response Analysis. J. Pharmacokinet. Pharmacodyn. 2016, 43, 325–341. [Google Scholar] [CrossRef] [PubMed]
  67. Bareille, P.; Hardes, K.; Donald, A.C. Efficacy and Safety of Once-Daily GW870086 a Novel Selective Glucocorticoid in Mild-Moderate Asthmatics: A Randomised, Two-Way Crossover, Controlled Clinical Trial. J. Asthma 2013, 50, 1077–1082. [Google Scholar] [CrossRef] [PubMed]
  68. Harcken, C.; Scholl, P.; Nabozny, G.; Thomson, D.; Bianchi, D. Clinical Profile of the Functionally Selective Glucocorticoid Receptor Agonist BI 653048 in Healthy Male Subjects. Expert Opin. Investig. Drugs 2019, 28, 489–496. [Google Scholar] [CrossRef]
  69. Yurchenko, E.A.; Chingizova, E.A.; Aminin, D.L.; Yurchenko, A.N. Marine Fungi: In Search of New Antibacterial Drugs. Mol. Biol. 2025, 59, 43–59. [Google Scholar] [CrossRef]
  70. Borkunov, G.V.; Kirichuk, N.N.; Chausova, V.E.; Popov, R.S.; Zhuravleva, O.I.; Chingizova, E.A.; Yurchenko, E.A.; Isaeva, M.P.; Yurchenko, A.N. Differences in Metabolite Profiles and Bioactivities of Intra-Strain Variants of Marine Fungus Penicillium Antarcticum KMM 4668. Metabolites 2025, 15, 77. [Google Scholar] [CrossRef]
  71. Chingizova, E.A.; Yurchenko, E.A.; Chingizov, A.R.; Klimovich, A.A.; Pislyagin, E.A.; Menchinskaya, E.S.; Kuzmich, A.S.; Trinh, P.T.H.; Ngoc, N.T.D.; Van, T.T.T.; et al. The Effects of Marine Fungal Asterripeptides A–C on In Vitro and In Vivo Staphylococcus Aureus Skin Infection. Pharmaceuticals 2024, 17, 1345. [Google Scholar] [CrossRef]
  72. Belousova, E.B.; Zhuravleva, O.I.; Yurchenko, E.A.; Oleynikova, G.K.; Antonov, A.S.; Kirichuk, N.N.; Chausova, V.E.; Khudyakova, Y.V.; Menshov, A.S.; Popov, R.S.; et al. New Anti-Hypoxic Metabolites from Co-Culture of Marine-Derived Fungi Aspergillus Carneus KMM 4638 and Amphichorda sp. KMM 4639. Biomolecules 2023, 13, 741. [Google Scholar] [CrossRef]
  73. Zhuravleva, O.I.; Oleinikova, G.K.; Antonov, A.S.; Kirichuk, N.N.; Pelageev, D.N.; Rasin, A.B.; Menshov, A.S.; Popov, R.S.; Kim, N.Y.; Chingizova, E.A.; et al. New Antibacterial Chloro-Containing Polyketides from the Alga-Derived Fungus Asteromyces Cruciatus KMM 4696. J. Fungi 2022, 8, 454. [Google Scholar] [CrossRef]
  74. Leshchenko, E.V.; Chingizova, E.A.; Antonov, A.S.; Shlyk, N.P.; Borkunov, G.V.; Berdyshev, D.V.; Chausova, V.E.; Kirichuk, N.N.; Khudyakova, Y.V.; Chingizov, A.R.; et al. New Zosteropenillines and Pallidopenillines from the Seagrass-Derived Fungus Penicillium Yezoense KMM 4679. Mar. Drugs 2024, 22, 317. [Google Scholar] [CrossRef] [PubMed]
  75. Zhuravleva, O.I.; Belousova, E.B.; Oleinikova, G.K.; Antonov, A.S.; Khudyakova, Y.V.; Rasin, A.B.; Popov, R.S.; Menchinskaya, E.S.; Trinh, P.T.H.; Yurchenko, A.N.; et al. Cytotoxic Drimane-Type Sesquiterpenes from Co-Culture of the Marine-Derived Fungi Aspergillus Carneus KMM 4638 and Beauveria Felina (=Isaria Felina) KMM 4639. Mar. Drugs 2022, 20, 584. [Google Scholar] [CrossRef] [PubMed]
  76. Krautforst, K.; Kulbacka, J.; Fornasier, M.; Mocci, R.; Porcheddu, A.; Pusceddu, A.; Moccia, D.; Murgia, S.; Bazylińska, U. Caulerpin Delivery via Pluronic-Free Cubosomes: Unlocking the Therapeutic Potential of a Pigment from an Invasive Marine Algae. Mol. Pharm. 2025, 22, 4747–4761. [Google Scholar] [CrossRef] [PubMed]
  77. Raksat, A.; Atanu, M.S.H.; McDermid, K.J.; Wall, M.M.; Chang, B.L.; Wongwiwatthananukit, S.; Chang, L.C. Bioactive Constituents from the Edible Seaweed Halymenia Hawaiiana (Rhodophyta). Pharm. Biol. 2025, 63, 447–459. [Google Scholar] [CrossRef]
  78. Asoudeh-Fard, A.; Soltanmohammadi, F.; Kashkouie Jahromi, M.; Javanmardi, F.; Roosta, A.; Zareian Jahromi, M.; Parsaei, A. Potential Anticancer Properties of Tetraselmis Suecica Extract against Oral and Colorectal Cancer Cells. Med. Oncol. 2025, 42, 282. [Google Scholar] [CrossRef]
  79. Lindequist, U. Marine-Derived Pharmaceuticals—Challenges and Opportunities. Biomol. Ther. 2016, 24, 561–571. [Google Scholar] [CrossRef]
  80. Hussain, A.; Sohail, A.; Akash, M.S.H.; Iqbal, S.; Rehman, K.; Imran, M.; Khan, S.; Ayub, M.A.; Wang, D.; Ahmed, D.; et al. Marine Life as a Source of Anti-Prostate Cancer Agents: An Updated Overview (2003–2023). Naunyn Schmiedebergs Arch. Pharmacol. 2025, 398, 7971–8074. [Google Scholar] [CrossRef]
  81. Shikov, A.N.; Flisyuk, E.V.; Obluchinskaya, E.D.; Pozharitskaya, O.N. Pharmacokinetics of Marine-Derived Drugs. Mar. Drugs 2020, 18, 557. [Google Scholar] [CrossRef]
  82. Li, S.; Xiao, Y.; Li, Q.; Su, M.; Guo, Y.; Jin, X. Recent Advances in Natural Products Derived from Marine Echinoderms and Endophytic Microbes: Chemical Insights and Therapeutic Potential. Mar. Drugs 2025, 23, 33. [Google Scholar] [CrossRef] [PubMed]
  83. Qiu, Y.; Chen, S.; Yu, M.; Shi, J.; Liu, J.; Li, X.; Chen, J.; Sun, X.; Huang, G.; Zheng, C. Natural Products from Marine-Derived Fungi with Anti-Inflammatory Activity. Mar. Drugs 2024, 22, 433. [Google Scholar] [CrossRef]
  84. Islam, F.; Mitra, S.; Emran, T.B.; Khan, Z.; Nath, N.; Das, R.; Sharma, R.; Al Awadh, A.A.; Park, M.N.; Kim, B. Natural Small Molecules in Gastrointestinal Tract and Associated Cancers: Molecular Insights and Targeted Therapies. Molecules 2022, 27, 5686. [Google Scholar] [CrossRef] [PubMed]
  85. Carroll, A.R.; Copp, B.R.; Davis, R.A.; Keyzers, R.A.; Prinsep, M.R. Marine Natural Products. Nat. Prod. Rep. 2021, 38, 362–413. [Google Scholar] [CrossRef]
  86. Abd El Hafez, M.S.M.; Aziz Okbah, M.A.E.; Ibrahim, H.A.H.; Hussein, A.A.E.R.; El Moneim, N.A.A.; Ata, A. First Report of Steroid Derivatives Isolated from Starfish Acanthaster Planci with Anti-Bacterial, Anti-Cancer and Anti-Diabetic Activities. Nat. Prod. Res. 2022, 36, 5545–5552. [Google Scholar] [CrossRef]
  87. Huong, P.T.M.; Phong, N.V.; Thao, N.P.; Binh, P.T.; Thao, D.T.; Van Thanh, N.; Cuong, N.X.; Nam, N.H.; Thung, D.C.; Minh, C. Van Dendrodoristerol, a Cytotoxic C20 Steroid from the Vietnamese Nudibranch Mollusk Dendrodoris Fumata. J. Asian Nat. Prod. Res. 2020, 22, 193–200. [Google Scholar] [CrossRef]
  88. Quang, T.H.; Lee, D.; Han, S.J.; Kim, I.C.; Yim, J.H.; Kim, Y.; Oh, H. ChemInform Abstract: Steroids from the Cold Water Starfish Ctenodiscus Crispatus with Cytotoxic and Apoptotic Effects on Human Hepatocellular Carcinoma and Glioblastoma Cells. ChemInform 2015, 46. [Google Scholar] [CrossRef]
  89. Xiao, D.-J.; Peng, X.-D.; Deng, S.-Z.; Ma, W.-J.; Wu, H.-M. Structure Elucidation of (3E)-Cholest-4-En-3,6-Dione-3-Oxime in Marine Sponge Cinachyrella Australiensis from the South China Sea. Chin. J. Org. Chem. 2005, 25, 1606–1609. [Google Scholar]
  90. Shubina, L.K.; Makarieva, T.N.; Denisenko, V.A.; Popov, R.S.; Dyshlovoy, S.A.; Grebnev, B.B.; Dmitrenok, P.S.; von Amsberg, G.; Stonik, V.A. Gracilosulfates A–G, Monosulfated Polyoxygenated Steroids from the Marine Sponge Haliclona Gracilis. Mar. Drugs 2020, 18, 454. [Google Scholar] [CrossRef]
  91. Mohamed, G.A.; Abd-Elrazek, A.E.E.; Hassanean, H.A.; Youssef, D.T.A.; van Soest, R. New Compounds from the Red Sea Marine Sponge Echinoclathria Gibbosa. Phytochem. Lett. 2014, 9, 51–58. [Google Scholar] [CrossRef]
  92. Holland, I.P.; McCluskey, A.; Sakoff, J.A.; Gilbert, J.; Chau, N.; Robinson, P.J.; Motti, C.A.; Wright, A.D.; van Altena, I.A. Steroids from an Australian Sponge Psammoclema sp. J. Nat. Prod. 2009, 72, 102–106. [Google Scholar] [CrossRef] [PubMed]
  93. Kicha, A.A.; Ivanchina, N.V.; Huong, T.T.T.; Kalinovsky, A.I.; Dmitrenok, P.S.; Fedorov, S.N.; Dyshlovoy, S.A.; Long, P.Q.; Stonik, V.A. Two New Asterosaponins, Archasterosides A and B, from the Vietnamese Starfish Archaster Typicus and Their Anticancer Properties. Bioorg. Med. Chem. Lett. 2010, 20, 3826–3830. [Google Scholar] [CrossRef] [PubMed]
  94. Ngoan, B.T.; Hanh, T.T.H.; Vien, L.T.; Diep, C.N.; Thao, N.P.; Thao, D.T.; Van Thanh, N.; Cuong, N.X.; Nam, N.H.; Thung, D.C.; et al. Asterosaponins and Glycosylated Polyhydroxysteroids from the Starfish Culcita Novaeguineae and Their Cytotoxic Activities. J. Asian Nat. Prod. Res. 2015, 17, 1010–1017. [Google Scholar] [CrossRef] [PubMed]
  95. Kicha, A.A.; Tolkanov, D.K.; Malyarenko, T.V.; Malyarenko, O.S.; Kuzmich, A.S.; Kalinovsky, A.I.; Popov, R.S.; Stonik, V.A.; Ivanchina, N.V.; Dmitrenok, P.S. Sulfated Polyhydroxysteroid Glycosides from the Sea of Okhotsk Starfish Henricia Leviuscula Spiculifera and Potential Mechanisms for Their Observed Anti-Cancer Activity against Several Types of Human Cancer Cells. Mar. Drugs 2024, 22, 294. [Google Scholar] [CrossRef]
  96. Kicha, A.A.; Kalinovsky, A.I.; Malyarenko, T.V.; Malyarenko, O.S.; Ermakova, S.P.; Popov, R.S.; Stonik, V.A.; Ivanchina, N.V. Disulfated Ophiuroid Type Steroids from the Far Eastern Starfish Pteraster Marsippus and Their Cytotoxic Activity on the Models of 2D and 3D Cultures. Mar. Drugs 2022, 20, 164. [Google Scholar] [CrossRef]
  97. Malyarenko, T.V.; Kicha, A.A.; Malyarenko, O.S.; Zakharenko, V.M.; Kotlyarov, I.P.; Kalinovsky, A.I.; Popov, R.S.; Svetashev, V.I.; Ivanchina, N.V. New Conjugates of Polyhydroxysteroids with Long-Chain Fatty Acids from the Deep-Water Far Eastern Starfish Ceramaster Patagonicus and Their Anticancer Activity. Mar. Drugs 2020, 18, 260. [Google Scholar] [CrossRef]
  98. Tian, X.-R.; Gao, Y.-Q.; Tian, X.-L.; Li, J.; Tang, H.-F.; Li, Y.-S.; Lin, H.-W.; Ma, Z.-Q. New Cytotoxic Secondary Metabolites from Marine Bryozoan Cryptosula Pallasiana. Mar. Drugs 2017, 15, 120. [Google Scholar] [CrossRef]
  99. Hoang, C.K.; Le, C.H.; Nguyen, D.T.; Tran, H.T.N.; Luu, C.V.; Le, H.M.; Tran, H.T.H. Steroid Components of Marine-Derived Fungal Strain Penicillium Levitum N33.2 and Their Biological Activities. Mycobiology 2023, 51, 246–255. [Google Scholar] [CrossRef]
  100. Zhang, J.-L.; Tian, H.-Y.; Li, J.; Jin, L.; Luo, C.; Ye, W.-C.; Jiang, R.-W. Steroids with Inhibitory Activity against the Prostate Cancer Cells and Chemical Diversity of Marine Alga Tydemania Expeditionis. Fitoterapia 2012, 83, 973–978. [Google Scholar] [CrossRef]
  101. Thao, N.P.; Luyen, B.T.T.; Kim, E.J.; Kang, J.I.; Kang, H.K.; Cuong, N.X.; Nam, N.H.; Van Kiem, P.; Van Minh, C.; Kim, Y.H. Steroidal Constituents from the Edible Sea Urchin Diadema Savignyi Michelin Induce Apoptosis in Human Cancer Cells. J. Med. Food 2015, 18, 45–53. [Google Scholar] [CrossRef]
  102. Tsai, C.-R.; Huang, C.-Y.; Chen, B.-W.; Tsai, Y.-Y.; Shih, S.-P.; Hwang, T.-L.; Dai, C.-F.; Wang, S.-Y.; Sheu, J.-H. New Bioactive Steroids from the Soft Coral Klyxum Flaccidum. RSC Adv. 2015, 5, 12546–12554. [Google Scholar] [CrossRef]
  103. Abdel-Lateff, A.; Alarif, W.M.; Asfour, H.Z.; Ayyad, S.-E.N.; Khedr, A.; Badria, F.A.; Al-lihaibi, S.S. Cytotoxic Effects of Three New Metabolites from Red Sea Marine Sponge, Petrosia sp. Env. Toxicol. Pharmacol. 2014, 37, 928–935. [Google Scholar] [CrossRef]
  104. Eissa, A.H.; Abdel-Tawab, A.M.; Hamed, E.S.A.E.; El-Ablack, F.Z.N.; Ayyad, S.-E. Cytotoxic Evaluation of New Polyhydroxylated Steroids from the Red Sea Soft Coral Litophyton Mollis (Macfadyen, 1936). Nat. Prod. Res. 2024, 38, 4390–4398. [Google Scholar] [CrossRef] [PubMed]
  105. Ma, X.-Y.; Wang, H.-N.; Sun, L.-X.; Sun, J.; Jin, S.-H.; Dai, F.-X.; Sai, C.-M.; Zhang, Z. Bioactive Steroids from Marine-Derived Fungi: A Review (2015–2023). J. Asian Nat. Prod. Res. 2025, 27, 1236–1262. [Google Scholar] [CrossRef]
  106. Khaledi, M.; Moradipoodeh, B.; Moradi, R.; Baghbadorani, M.A.; Mahdavinia, M. Antiproliferative and Proapoptotic Activities of Sea Cucumber H. Leucospilota Extract on Breast Carcinoma Cell Line (SK-BR-3). Mol. Biol. Rep. 2022, 49, 1191–1200. [Google Scholar] [CrossRef]
  107. Heidary Jamebozorgi, F.; Yousefzadi, M.; Firuzi, O.; Nazemi, M.; Zare, S.; Chandran, J.N.; Schneider, B.; Baldwin, I.T.; Jassbi, A.R. Cytotoxic Furanosesquiterpenoids and Steroids from Ircinia Mutans Sponges. Pharm. Biol. 2021, 59, 573–581. [Google Scholar] [CrossRef]
  108. Hadisaputri, Y.E.; Andika, R.; Sopyan, I.; Zuhrotun, A.; Maharani, R.; Rachmat, R.; Abdulah, R. Caspase Cascade Activation During Apoptotic Cell Death of Human Lung Carcinoma Cells A549 Induced by Marine Sponge Callyspongia Aerizusa. Drug Des. Dev. Ther. 2021, 15, 1357–1368. [Google Scholar] [CrossRef]
  109. Ruiz-Torres, V.; Rodríguez-Pérez, C.; Herranz-López, M.; Martín-García, B.; Gómez-Caravaca, A.-M.; Arráez-Román, D.; Segura-Carretero, A.; Barrajón-Catalán, E.; Micol, V. Marine Invertebrate Extracts Induce Colon Cancer Cell Death via ROS-Mediated DNA Oxidative Damage and Mitochondrial Impairment. Biomolecules 2019, 9, 771. [Google Scholar] [CrossRef]
  110. Thi Duy Ngoc, N.; Yurchenko, E.A.; Thi Hoai Trinh, P.; Menchinskaya, E.S.; Thi Dieu, T.V.; Savagina, A.D.; Minin, A.; Thinh, P.D.; Khanh, H.H.N.; Thi Thanh Van, T.; et al. Secondary Metabolites of Vietnamese Marine Fungus Penicillium Chermesinum 2104NT-1.3 and Their Cardioprotective Activity. Reg. Stud. Mar. Sci. 2025, 81, 104003. [Google Scholar] [CrossRef]
  111. Kicha, A.A.; Malyarenko, T.V.; Kuzmich, A.S.; Malyarenko, O.S.; Kalinovsky, A.I.; Popov, R.S.; Tolkanov, D.K.; Ivanchina, N.V. Rare Ophiuroid-Type Steroid 3β,21-, 3β,22-, and 3α,22-Disulfates from the Slime Sea Star Pteraster Marsippus and Their Colony-Inhibiting Effects against Human Breast Cancer Cells. Mar. Drugs 2024, 22, 43. [Google Scholar] [CrossRef]
  112. Ngoc, N.T.; Huong, P.T.M.; Van Thanh, N.; Chi, N.T.P.; Dang, N.H.; Cuong, N.X.; Nam, N.H.; Thung, D.C.; Van Kiem, P.; Minh, C. Van Cytotoxic Steroids from the Vietnamese Soft Coral Sinularia Conferta. Chem. Pharm. Bull. 2017, 65, 300–305. [Google Scholar] [CrossRef] [PubMed]
  113. Ghandourah, M.A. Cytotoxic Ketosteroids from the Red Sea Soft Coral Dendronephthya sp. Open Chem 2023, 21. [Google Scholar] [CrossRef]
  114. Adcock, I.M.; Mumby, S. Glucocorticoids. In Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2016; pp. 171–196. [Google Scholar]
  115. Tan, C.K.; Wahli, W. A Trilogy of Glucocorticoid Receptor Actions. Proc. Natl. Acad. Sci. USA 2016, 113, 1115–1117. [Google Scholar] [CrossRef]
  116. Cain, D.W.; Cidlowski, J.A. Specificity and Sensitivity of Glucocorticoid Signaling in Health and Disease. Best. Pr. Res. Clin. Endocrinol. Metab. 2015, 29, 545–556. [Google Scholar] [CrossRef]
  117. Oakley, R.H.; Cidlowski, J.A. The Biology of the Glucocorticoid Receptor: New Signaling Mechanisms in Health and Disease. J. Allergy Clin. Immunol. 2013, 132, 1033–1044. [Google Scholar] [CrossRef]
  118. Pofi, R.; Caratti, G.; Ray, D.W.; Tomlinson, J.W. Treating the Side Effects of Exogenous Glucocorticoids; Can We Separate the Good From the Bad? Endocr. Rev. 2023, 44, 975–1011. [Google Scholar] [CrossRef]
  119. De Bosscher, K.; Beck, I.M.; Ratman, D.; Berghe, W.V.; Libert, C. Activation of the Glucocorticoid Receptor in Acute Inflammation: The SEDIGRAM Concept. Trends Pharmacol. Sci. 2016, 37, 4–16. [Google Scholar] [CrossRef]
  120. Sundahl, N.; Bridelance, J.; Libert, C.; De Bosscher, K.; Beck, I.M. Selective Glucocorticoid Receptor Modulation: New Directions with Non-Steroidal Scaffolds. Pharmacol. Ther. 2015, 152, 28–41. [Google Scholar] [CrossRef]
  121. Gronemeyer, H.; Gustafsson, J.-Å.; Laudet, V. Principles for Modulation of the Nuclear Receptor Superfamily. Nat. Rev. Drug Discov. 2004, 3, 950–964. [Google Scholar] [CrossRef]
  122. Hudson, W.H.; de Vera, I.M.S.; Nwachukwu, J.C.; Weikum, E.R.; Herbst, A.G.; Yang, Q.; Bain, D.L.; Nettles, K.W.; Kojetin, D.J.; Ortlund, E.A. Cryptic Glucocorticoid Receptor-Binding Sites Pervade Genomic NF-ΚB Response Elements. Nat. Commun. 2018, 9, 1337. [Google Scholar] [CrossRef] [PubMed]
  123. Hudson, W.H.; Kossmann, B.R.; de Vera, I.M.S.; Chuo, S.-W.; Weikum, E.R.; Eick, G.N.; Thornton, J.W.; Ivanov, I.N.; Kojetin, D.J.; Ortlund, E.A. Distal Substitutions Drive Divergent DNA Specificity among Paralogous Transcription Factors through Subdivision of Conformational Space. Proc. Natl. Acad. Sci. USA 2016, 113, 326–331. [Google Scholar] [CrossRef]
  124. Stechschulte, L.A.; Wuescher, L.; Marino, J.S.; Hill, J.W.; Eng, C.; Hinds, T.D. Glucocorticoid Receptor β Stimulates Akt1 Growth Pathway by Attenuation of PTEN. J. Biol. Chem. 2014, 289, 17885–17894. [Google Scholar] [CrossRef] [PubMed]
  125. Kwack, M.H.; Ben Hamida, O.; Kim, M.K.; Kim, J.C.; Sung, Y.K. Dexamethasone, a Synthetic Glucocorticoid, Induces the Activity of Androgen Receptor in Human Dermal Papilla Cells. Skin Pharmacol. Physiol. 2022, 35, 299–304. [Google Scholar] [CrossRef] [PubMed]
  126. Burnstein, K.L.; Maiorino, C.A.; Dai, J.L.; Cameron, D.J. Androgen and Glucocorticoid Regulation of Androgen Receptor CDNA Expression. Mol. Cell. Endocrinol. 1995, 115, 177–186. [Google Scholar] [CrossRef]
  127. Pecci, A.; Ogara, M.F.; Sanz, R.T.; Vicent, G.P. Choosing the Right Partner in Hormone-Dependent Gene Regulation: Glucocorticoid and Progesterone Receptors Crosstalk in Breast Cancer Cells. Front. Endocrinol. 2022, 13, 1037177. [Google Scholar] [CrossRef]
  128. Wan, Y.; Nordeen, S.K. Overlapping but Distinct Gene Regulation Profiles by Glucocorticoids and Progestins in Human Breast Cancer Cells. Mol. Endocrinol. 2002, 16, 1204–1214. [Google Scholar] [CrossRef]
  129. Hundertmark, S.; Buhler, H.; Rudolf, M.; Weitzel, H.; Ragosch, V. Inhibition of 11 Beta-Hydroxysteroid Dehydrogenase Activity Enhances the Antiproliferative Effect of Glucocorticosteroids on MCF-7 and ZR-75-1 Breast Cancer Cells. J. Endocrinol. 1997, 155, 171–180. [Google Scholar] [CrossRef]
  130. Meyer, M.-E.; Gronemeyer, H.; Turcotte, B.; Bocquel, M.-T.; Tasset, D.; Chambon, P. Steroid Hormone Receptors Compete for Factors That Mediate Their Enhancer Function. Cell 1989, 57, 433–442. [Google Scholar] [CrossRef]
  131. Pan, D.; Kocherginsky, M.; Conzen, S.D. Activation of the Glucocorticoid Receptor Is Associated with Poor Prognosis in Estrogen Receptor-Negative Breast Cancer. Cancer Res. 2011, 71, 6360–6370. [Google Scholar] [CrossRef]
  132. West, D.C.; Pan, D.; Tonsing-Carter, E.Y.; Hernandez, K.M.; Pierce, C.F.; Styke, S.C.; Bowie, K.R.; Garcia, T.I.; Kocherginsky, M.; Conzen, S.D. GR and ER Coactivation Alters the Expression of Differentiation Genes and Associates with Improved ER+ Breast Cancer Outcome. Mol. Cancer Res. 2016, 14, 707–719. [Google Scholar] [CrossRef]
  133. Karmakar, S.; Jin, Y.; Nagaich, A.K. Interaction of Glucocorticoid Receptor (GR) with Estrogen Receptor (ER) α and Activator Protein 1 (AP1) in Dexamethasone-Mediated Interference of ERα Activity. J. Biol. Chem. 2013, 288, 24020–24034. [Google Scholar] [CrossRef]
  134. Miranda, T.B.; Voss, T.C.; Sung, M.-H.; Baek, S.; John, S.; Hawkins, M.; Grøntved, L.; Schiltz, R.L.; Hager, G.L. Reprogramming the Chromatin Landscape: Interplay of the Estrogen and Glucocorticoid Receptors at the Genomic Level. Cancer Res. 2013, 73, 5130–5139. [Google Scholar] [CrossRef]
  135. Voss, T.C.; Schiltz, R.L.; Sung, M.-H.; Yen, P.M.; Stamatoyannopoulos, J.A.; Biddie, S.C.; Johnson, T.A.; Miranda, T.B.; John, S.; Hager, G.L. Dynamic Exchange at Regulatory Elements during Chromatin Remodeling Underlies Assisted Loading Mechanism. Cell 2011, 146, 544–554. [Google Scholar] [CrossRef]
  136. Volden, P.A.; Conzen, S.D. The Influence of Glucocorticoid Signaling on Tumor Progression. Brain Behav. Immun. 2013, 30, S26–S31. [Google Scholar] [CrossRef] [PubMed]
  137. Pang, C.; Chen, Y.-H.; Bian, H.-H.; Zhang, J.-P.; Su, L.; Han, H.; Zhang, W. Anti-Inflammatory Ergosteroid Derivatives from the Coral-Associated Fungi Penicillium Oxalicum HL-44. Molecules 2023, 28, 7784. [Google Scholar] [CrossRef] [PubMed]
  138. Ren, X.; Chen, C.; Ye, Y.; Xu, Z.; Zhao, Q.; Luo, X.; Liu, Y.; Guo, P. Anti-Inflammatory Compounds from the Mangrove Endophytic Fungus Amorosia sp. SCSIO 41026. Front. Microbiol. 2022, 13, 976399. [Google Scholar] [CrossRef] [PubMed]
  139. Zou, Z.-B.; Wu, T.-Z.; Yang, L.-H.; He, X.-W.; Liu, W.-Y.; Zhang, K.; Xie, C.-L.; Xie, M.-M.; Zhang, Y.; Yang, X.-W.; et al. Hepialiamides A–C: Aminated Fusaric Acid Derivatives and Related Metabolites with Anti-Inflammatory Activity from the Deep-Sea-Derived Fungus Samsoniella Hepiali W7. Mar. Drugs 2023, 21, 596. [Google Scholar] [CrossRef]
  140. Jiang, Z.-P.; Su, R.; Chen, M.-T.; Li, J.-Y.; Chen, H.-Y.; Yang, L.; Liu, F.-F.; Liu, J.; Xu, C.-J.; Li, W.-S.; et al. Ent-Eudesmane Sesquiterpenoids with Anti-Neuroinflammatory Activity from the Marine-Derived Fungus Eutypella sp. F0219. Phytochemistry 2024, 223, 114121. [Google Scholar] [CrossRef]
  141. Dai, L.-T.; Yang, L.; Wang, Z.-P.; Guo, J.-C.; Ma, Q.-Y.; Xie, Q.-Y.; Dai, H.-F.; Yu, Z.-F.; Zhao, Y.-X. Persteroid, a New Steroid from the Marine-Derived Fungus Penicillium sp. ZYX-Z-143. Nat. Prod. Res. 2024, 1–8. [Google Scholar] [CrossRef]
  142. Strangman, W.K.; Kwon, H.C.; Broide, D.; Jensen, P.R.; Fenical, W. Potent Inhibitors of Pro-Inflammatory Cytokine Production Produced by a Marine-Derived Bacterium. J. Med. Chem. 2009, 52, 2317–2327. [Google Scholar] [CrossRef]
  143. Hannan, M.A.; Dash, R.; Sohag, A.A.M.; Moon, I.S. Deciphering Molecular Mechanism of the Neuropharmacological Action of Fucosterol through Integrated System Pharmacology and In Silico Analysis. Mar. Drugs 2019, 17, 639. [Google Scholar] [CrossRef]
  144. Hagiwara, H.; Wakita, K.; Inada, Y.; Hirose, S. Fucosterol Decreases Angiotensin Converting Enzyme Levels with Reduction of Glucocorticoid Receptors in Endothelial Cells. Biochem. Biophys. Res. Commun. 1986, 139, 348–352. [Google Scholar] [CrossRef] [PubMed]
  145. Yurchenko, E.A.; Khmel, O.O.; Nesterenko, L.E.; Aminin, D.L. The Kelch/Nrf2 Antioxidant System as a Target for Some Marine Fungal Metabolites. Oxygen 2023, 3, 374–385. [Google Scholar] [CrossRef]
  146. Kolesnikova, S.A.; Lyakhova, E.G.; Kalinovsky, A.I.; Popov, R.S.; Yurchenko, E.A.; Stonik, V.A. Oxysterols from a Marine Sponge Inflatella sp. and Their Action in 6-Hydroxydopamine-Induced Cell Model of Parkinson’s Disease. Mar. Drugs 2018, 16, 458. [Google Scholar] [CrossRef] [PubMed]
  147. Zhuravleva, O.I.; Afiyatullov, S.S.; Vishchuk, O.S.; Denisenko, V.A.; Slinkina, N.N.; Smetanina, O.F. Decumbenone C, a New Cytotoxic Decaline Derivative from the Marine Fungus Aspergillus Sulphureus KMM 4640. Arch. Pharm. Res. 2012, 35, 1757–1762. [Google Scholar] [CrossRef]
  148. Yurchenko, A.N.; Zhuravleva, O.I.; Khmel, O.O.; Oleynikova, G.K.; Antonov, A.S.; Kirichuk, N.N.; Chausova, V.E.; Kalinovsky, A.I.; Berdyshev, D.V.; Kim, N.Y.; et al. New Cyclopiane Diterpenes and Polyketide Derivatives from Marine Sediment-Derived Fungus Penicillium Antarcticum KMM 4670 and Their Biological Activities. Mar. Drugs 2023, 21, 584. [Google Scholar] [CrossRef]
  149. Zhuravleva, O.I.; Afiyatullov, S.S.; Denisenko, V.A.; Ermakova, S.P.; Slinkina, N.N.; Dmitrenok, P.S.; Kim, N.Y. Secondary Metabolites from a Marine-Derived Fungus Aspergillus Carneus Blochwitz. Phytochemistry 2012, 80, 123–131. [Google Scholar] [CrossRef]
  150. Afiyatullov, S.S.; Zhuravleva, O.I.; Antonov, A.S.; Leshchenko, E.V.; Pivkin, M.V.; Khudyakova, Y.V.; Denisenko, V.A.; Pislyagin, E.A.; Kim, N.Y.; Berdyshev, D.V.; et al. Piltunines A–F from the Marine-Derived Fungus Penicillium Piltunense KMM 4668. Mar. Drugs 2019, 17, 647. [Google Scholar] [CrossRef]
  151. Yurchenko, A.N.; Smetanina, O.F.; Kalinovsky, A.I.; Pushilin, M.A.; Glazunov, V.P.; Khudyakova, Y.V.; Kirichuk, N.N.; Ermakova, S.P.; Dyshlovoy, S.A.; Yurchenko, E.A.; et al. Oxirapentyns F–K from the Marine-Sediment-Derived Fungus Isaria Felina KMM 4639. J. Nat. Prod. 2014, 77, 1321–1328. [Google Scholar] [CrossRef]
  152. Zhuravleva, O.I.; Chingizova, E.A.; Oleinikova, G.K.; Starnovskaya, S.S.; Antonov, A.S.; Kirichuk, N.N.; Menshov, A.S.; Popov, R.S.; Kim, N.Y.; Berdyshev, D.V.; et al. Anthraquinone Derivatives and Other Aromatic Compounds from Marine Fungus Asteromyces Cruciatus KMM 4696 and Their Effects against Staphylococcus Aureus. Mar. Drugs 2023, 21, 431. [Google Scholar] [CrossRef]
  153. Dalinova, A.; Smirnov, S.; Dubovik, V.; Senderskiy, I.; Stepanycheva, E.; Kovalenko, N.; Berestetskiy, A. Production and Toxicological Characterization of Secondary Metabolites of Pyrenophora Tritici-Repentis. J. Plant Dis. Prot. 2025, 132, 149. [Google Scholar] [CrossRef]
  154. Afiyatullov, S.S.; Leshchenko, E.V.; Sobolevskaya, M.P.; Antonov, A.S.; Denisenko, V.A.; Popov, R.S.; Khudyakova, Y.V.; Kirichuk, N.N.; Kuz’mich, A.S.; Pislyagin, E.A.; et al. New Thomimarine E from Marine Isolate of the Fungus Penicillium Thomii. Chem. Nat. Compd. 2017, 53, 290–294. [Google Scholar] [CrossRef]
  155. Smetanina, O.F.; Yurchenko, A.N.; Ivanets, E.V.; Kirichuk, N.N.; Khudyakova, Y.V.; Yurchenko, E.A.; Afiyatullov, S.S. Metabolites of the Marine Fungus Penicillium Citrinum Associated with a Brown Alga Padina sp. Chem. Nat. Compd. 2016, 52, 111–112. [Google Scholar] [CrossRef]
  156. Girich, E.V.; Yurchenko, A.N.; Smetanina, O.F.; Trinh, P.T.H.; Ngoc, N.T.D.; Pivkin, M.V.; Popov, R.S.; Pislyagin, E.A.; Menchinskaya, E.S.; Chingizova, E.A.; et al. Neuroprotective Metabolites from Vietnamese Marine Derived Fungi of Aspergillus and Penicillium Genera. Mar. Drugs 2020, 18, 608. [Google Scholar] [CrossRef]
  157. Yurchenko, A.; Trinh, P.; Girich (Ivanets), E.; Smetanina, O.; Rasin, A.; Popov, R.; Dyshlovoy, S.; von Amsberg, G.; Menchinskaya, E.; Thanh Van, T.; et al. Biologically Active Metabolites from the Marine Sediment-Derived Fungus Aspergillus Flocculosus. Mar. Drugs 2019, 17, 579. [Google Scholar] [CrossRef]
  158. Smetanina, O.F.; Yurchenko, A.N.; Kalinovsky, A.I.; Pushilin, M.A.; Slinkina, N.N.; Yurchenko, E.A.; Afiyatullov, S.S. 4-Methoxy-3-Methylgoniothalamin from Marine-Derived Fungi of the Genus Penicillium. Russ. Chem. Bull. 2011, 60, 760–763. [Google Scholar] [CrossRef]
  159. Yurchenko, A.N.; Smetanina, O.F.; Ivanets, E.V.; Phan, T.T.H.; Ngo, N.T.D.; Zhuravleva, O.I.; Rasin, A.B.; Dyshlovoy, S.A.; Menchinskaya, E.S.; Pislyagin, E.A.; et al. Auroglaucin-Related Neuroprotective Compounds from Vietnamese Marine Sediment-Derived Fungus Aspergillus Niveoglaucus. Nat. Prod. Res. 2020, 34, 2589–2594. [Google Scholar] [CrossRef]
  160. Antonov, A.S.; Leshchenko, E.V.; Zhuravleva, O.I.; Dyshlovoy, S.A.; von Amsberg, G.; Popov, R.S.; Denisenko, V.A.; Kirichuk, N.N.; Afiyatullov, S.S. Naphto-Γ-Pyrones from the Marine-Derived Fungus Aspergillus Foetidus. Nat. Prod. Res. 2021, 35, 131–134. [Google Scholar] [CrossRef]
  161. Afiyatullov, S.S.; Leshchenko, E.V.; Sobolevskaya, M.P.; Gerasimenko, A.V.; Khudyakova, Y.V.; Kirichuk, N.N.; Mikhailov, V.V. New 3-[2′(R)-Hydroxybutyl]-7-Hydroxyphthalide from Marine Isolate of the Fungus Penicillium Claviforme. Chem. Nat. Compd. 2015, 51, 111–115. [Google Scholar] [CrossRef]
  162. Yurchenko, A.N.; Ivanets, E.V.; Smetanina, O.F.; Pivkin, M.V.; Dyshlovoi, S.A.; von Amsberg, G.; Afiyatullov, S.S. Metabolites of the Marine Fungus Aspergillus Candidus KMM 4676 Associated with a Kuril Colonial Ascidian. Chem. Nat. Compd. 2017, 53, 747–749. [Google Scholar] [CrossRef]
  163. Leshchenko, E.V.; Berdyshev, D.V.; Yurchenko, E.A.; Antonov, A.S.; Borkunov, G.V.; Kirichuk, N.N.; Chausova, V.E.; Kalinovskiy, A.I.; Popov, R.S.; Khudyakova, Y.V.; et al. Bioactive Polyketides from the Natural Complex of the Sea Urchin-Associated Fungi Penicillium Sajarovii KMM 4718 and Aspergillus Protuberus KMM 4747. Int. J. Mol. Sci. 2023, 24, 16568. [Google Scholar] [CrossRef]
  164. Zhuravleva, O.I.; Kirichuk, N.N.; Denisenko, V.A.; Dmitrenok, P.S.; Yurchenko, E.A.; Min′ko, E.M.; Ivanets, E.V.; Afiyatullov, S.S. New Diorcinol J Produced by Co-Cultivation of Marine Fungi Aspergillus Sulphureus and Isaria Felina. Chem. Nat. Compd. 2016, 52, 227–230. [Google Scholar] [CrossRef]
Figure 1. Complexes of LBD-GR with compounds (PDB ID 1P93). Green lines indicate hydrogenic bonds, and yellow lines indicate hydrophobic interactions.
Figure 1. Complexes of LBD-GR with compounds (PDB ID 1P93). Green lines indicate hydrogenic bonds, and yellow lines indicate hydrophobic interactions.
Marinedrugs 23 00399 g001
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

Zhidkova, E.M.; Savina, E.D.; Yurchenko, E.A.; Lesovaya, E.A. Marine-Derived Steroids for Cancer Treatment: Search for Potential Selective Glucocorticoid Receptor Agonists/Modulators (SEGRAMs). Mar. Drugs 2025, 23, 399. https://doi.org/10.3390/md23100399

AMA Style

Zhidkova EM, Savina ED, Yurchenko EA, Lesovaya EA. Marine-Derived Steroids for Cancer Treatment: Search for Potential Selective Glucocorticoid Receptor Agonists/Modulators (SEGRAMs). Marine Drugs. 2025; 23(10):399. https://doi.org/10.3390/md23100399

Chicago/Turabian Style

Zhidkova, Ekaterina M., Ekaterina D. Savina, Ekaterina A. Yurchenko, and Ekaterina A. Lesovaya. 2025. "Marine-Derived Steroids for Cancer Treatment: Search for Potential Selective Glucocorticoid Receptor Agonists/Modulators (SEGRAMs)" Marine Drugs 23, no. 10: 399. https://doi.org/10.3390/md23100399

APA Style

Zhidkova, E. M., Savina, E. D., Yurchenko, E. A., & Lesovaya, E. A. (2025). Marine-Derived Steroids for Cancer Treatment: Search for Potential Selective Glucocorticoid Receptor Agonists/Modulators (SEGRAMs). Marine Drugs, 23(10), 399. https://doi.org/10.3390/md23100399

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