Next Article in Journal
Halocins and C50 Carotenoids from Haloarchaea: Potential Natural Tools against Cancer
Previous Article in Journal
Unraveling the Omega-3 Puzzle: Navigating Challenges and Innovations for Bone Health and Healthy Aging
Previous Article in Special Issue
Stonikacidin A, an Antimicrobial 4-Bromopyrrole Alkaloid Containing L-Idonic Acid Core from the Northwestern Pacific Marine Sponge Lissodendoryx papillosa
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review of Sponge-Derived Diterpenes: 2009–2022

1
Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China
2
Department of Pharmacy, NO. 971 Hospital of the People’s Liberation Army Navy, Qingdao 266000, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Mar. Drugs 2024, 22(10), 447; https://doi.org/10.3390/md22100447 (registering DOI)
Submission received: 5 September 2024 / Revised: 16 September 2024 / Accepted: 26 September 2024 / Published: 28 September 2024
(This article belongs to the Special Issue Bio-Active Components from Marine Sponges)

Abstract

:
Sponges are a vital source of pharmaceutically active secondary metabolites, of which the main structural types are alkaloids and terpenoids. Many of these compounds exhibit biological activities. Focusing specifically on diterpenoids, this article reviews the structures and biological activities of 228 diterpenes isolated from more than 33 genera of sponges from 2009 to 2022. The Spongia sponges produce the most diterpenoid molecules among all genera, accounting for 27%. Of the 228 molecules, 110 exhibit cytotoxic, antibacterial, antifungal, antiparasitic, anti-inflammatory, and antifouling activities, among others. The most prevalent activity is cytotoxicity, present in 54 molecules, which represent 24% of the diterpenes reported. These structurally and biologically diverse diterpenoids highlight the vast, yet largely untapped, potential of marine sponges in the discovery of new bioactive molecules for medicinal use.

1. Introduction

Sponges are important multicellular animals, exhibiting a vast diversity and widespread distribution. More than a decade ago, there were already about 15,000 species of sponges that had been described [1]. The number of natural products derived from sponges is substantial, with over 200 new molecules discovered each year [2,3,4]. Compounds derived from sponges exhibit a rich diversity, encompassing terpenoids [5], sterols, alkaloids [6], ceramides, macrolides, peptides, and others. Among these, alkaloids and terpenoids are the most abundant, accounting for 50% of the total [7]. Diterpenoids represent a class of structurally diverse terpenes with various biological activities.
There have been some reviews on the separation, structural elucidation, and biological activity evaluation of diterpenoids. From 1984 to 2009, Hanson James R. published a series of reviews on newly discovered diterpenes every year [8,9,10,11]. Since 2011, Hanson began publishing reviews on diterpenoids from terrestrial sources [12]. Reviews on diterpenoids from marine sources are relatively fewer.
We have consistently focused on the diterpene compounds derived from marine sources. In our review of marine-derived diterpenes, we found that fungi and sponges are primary sources of these compounds. Previously, we summarized the diterpenes from marine fungi [13]. Here, we provide another review focusing on the diterpenes from marine sponges.
There are some review articles on sponge-derived natural products. Two articles summarize the chemical and biological diversity of compounds from sponges over a decade, with the timespans being 2009–2018 [7] and 2011–2020 [14], respectively. Some reviews focus on specific types of sponges, such as molecules from the order Dictyoceratida [15], the families Hymedesmiidae [16] and Latrunculiidae [17], and the genera Amphimedon [18], Aaptos [19], Callyspongia [20], Haliclona [21], Petrosia [22], Phorbas [23], Reniera [24], Stelletta [25], and Suberea [26]. Other reviews focus on compounds with specific activities, like sponge-derived molecules with antimalarial/antiprotozoal activity [27] and natural products from Red Sea sponges with cytotoxic activity [28]. A review critically analyzed drugs/drug candidates inspired by natural sponge products [29]. There are also reviews focusing on specific types of compounds, such as alkaloids from sponges [6]. Diterpenes sourced from sponges have not been reviewed yet.
In this article, the structures and bioactivities of 228 newly discovered diterpenoids from more than 33 genera of sponges reported by 73 publications covering 14 years, from 2009 to 2022, are summarized.

2. The Characteristics of Diterpenes from Sponges

From 2009 to 2022, there were 73 reports on 228 new diterpenoids from sponges. Among these compounds, 62 were from the genus Spongia, accounting for 27% of the total, while 33, 17, 16, 13, 13, and 11 were from the genera Agelas, Acanthella, Hamigera, Darwinella, Dysidea, and Dendrilla, respectively (Figure 1A). In addition, 26 other genera of sponges have also yielded diterpenoids, with each contributing fewer than 10 molecules (Figure 1B). Among these, 12 genera have each produced a single diterpenoid. Furthermore, there are five diterpenoids that originate from sponges that have not yet been identified at the genus level.
Among these diterpenes, 110 have demonstrated a variety of bioactivities, constituting 48% of the overall number of reported compounds. The literature has reported 138 bioactivity data entries for these compounds, with 17, 4, and 1 compounds exhibiting two, three, and four different types of bioactivities, respectively. Cytotoxic activities against various tumor cell lines are considered to be a single type of activity.
The most frequently reported activity was cytotoxicity against tumor cells, with 54 molecules showing this activity, accounting for 39% of the activity entries and 24% of all reported molecules (Figure 2). The number of compounds with antibacterial activity ranked second, with 22 molecules, among which 6 had antituberculosis activity. The numbers of molecules with anti-inflammatory, antiparasitic, antifouling, antifungal, and osteoclast-inhibitory activities were 13, 12, 11, 7, and 6, respectively. Among the 12 molecules with antiparasitic activity, 9 were active against Plasmodium species, whereas 3 were active against the leishmaniasis parasite. The activities of 13 molecules were categorized as other types, including radiation sensitization activity, antioxidant activity, inhibitory activity against specific target proteins such as Casitas B-lineage lymphoma proto-oncogene-b (Cbl-b), antiviral activity, and so on.

3. Isolation, Structure, and Bioactivities of Diterpenes from Sponges

3.1. Acanthella

A total of 17 diterpenes were obtained from Acanthella sponges. Seven formamido-diterpenes—cavernenes A–D, kalihinenes E–F, and kalihipyran C (17, Figure 3)—were isolated from the South China Sea sponge Acanthella cavernosa [30]. The cytotoxicity of these compounds was evaluated using five human cancer cell lines (human colon cancer cell line HCT-116, human lung epithelial cell line A549, human cervical carcinoma cell line HeLa, human hepatocellular carcinoma cell line QGY-7701, and human mammary cancer cell line MDA-MB-231).
Compounds 1 and 2 showed cytotoxic activities against HCT-116 cells, with IC50 (50% inhibitory concentration) values of 6.31 and 8.99 µM, respectively (Table 1). Compound 5 showed cytotoxic activity against HCT-116, HeLa, QGY-7701, and MDA-MB-231 cells, with IC50 values of 14.36, 13.36, 17.78, and 12.84 µM, respectively.
A nitrogen-containing kalihinane-type diterpenoid, bisformamidokalihinol A (8, Figure 3), was obtained from the Acanthella cavernosa sponge [31]. Eight diterpenoids, kalihinols M–T (916, Figure 3), were isolated from the South China Sea sponge Acanthella cavernosa [32]. Kalihinols M–N, with a formamide functionality at C-4, extended the structural breadth of this diterpenoid family. Kalihinols O–R showed cytotoxic activity against HCT-116 cells, with IC50 values of 5.97, 10.68, 20.55, and 13.44 µM, respectively. Kalihinol P displayed cytotoxic activity against H1299 cells, with an IC50 value of 26.21 µM. Kalihinols O–T displayed significant antifouling activity against Balanus amphitrite larvae, with EC50 values of 1.43, 0.72, 1.48, 1.16, 0.53, and 0.74 µM, respectively. A kalihinol diterpene, 10-epi-kalihinol X (17, Figure 3), was obtained from the Hainan sponge Acanthella sp. [33]. Compound 17 exhibited in vitro cytotoxicity against the human lung adenocarcinoma cell line A549, with an IC50 value of 9.30 µg/mL.
Table 1. Sponge-derived diterpenes with various bioactivities.
Table 1. Sponge-derived diterpenes with various bioactivities.
Compound NumberCompound NameProducing SpongeActivityReference
Cytotoxicity to cancer cell lines
12Cavernenes A–BAcanthella cavernosaCytotoxicity against the HCT-116 cell line, with IC50 values of 6.31 and 8.99 µM, respectively[30]
5Kalihinene EAcanthella cavernosaCytotoxicity against the HCT-116, HeLa, QGY-7701, and MDA-MB-231 cell lines, with IC50 values of 14.36, 13.36, 17.78, and 12.84 µM, respectively[30]
11Kalihinol OAcanthella cavernosaCytotoxicity against HCT-116 cells, with an IC50 value of 5.97 µM[32]
12Kalihinol PAcanthella cavernosaCytotoxicity against HCT-116 and H1299 cells, with IC50 values of 10.68 and 26.21 µM, respectively[32]
1314Kalihinols Q–RAcanthella cavernosaCytotoxicity against HCT-116 cells, with IC50 values of 20.55 and 13.44 µM, respectively[32]
1710-Epi-kalihinol XAcanthella sp.Cytotoxicity against the A549 cell line, with an IC50 value of 9.30 µg/mL[33]
20Axistatin 1Agelas axifera HentschelCytotoxicity against P388, BXPC-3, MCF-7, SF-268, NCI-H460, KM20L2, and DU-145 cells, with GI50 (50% growth inhibition) values of 19.8, 4.8, 5.7, 3.6, 4.6, 4.1, and 4.8 µM, respectively[34]
21Axistatin 2Agelas axifera HentschelCytotoxicity against P388, BXPC-3, MCF-7, SF-268, NCI-H460, KM20L2, and DU-145 cells, with GI50 values of 22.8, 5.5, 6.8, 3.9, 4.3, 4.1, and 5.0 µM, respectively[34]
22Axistatin 3Agelas axifera HentschelCytotoxicity against P388, BXPC-3, MCF-7, SF-268, NCI-H460, KM20L2, and DU-145 cells, with GI50 values of 8.9, 6.0, 5.8, 3.5, 5.4, 6.9, and 7.5 µM, respectively[34]
27Nemoechine GAgelas aff. nemoechinataCytotoxicity against Jurkat cell lines, with an IC50 of 17.1 µM[35]
28Nemoechine DAgelas aff. nemoechinataCytotoxicity against the human promyelocytic leukemia HL-60 cell line, with an IC50 value of 9.9 µM[36]
36(+)-Agelasine BAgelas mauritianaCytotoxicity toward the cancer cell lines PC9, A549, HepG2, MCF-7, and U937, with IC50 values of 5.08, 14.07, 9.76, 7.64, and 4.49 µM, respectively[37]
40Iso-agelasine CAgelas nakamuraiCytotoxicity against HL-60, K562, and HCT-116 cell lines, with IC50 values of 25.3, 28.9, and 38.8 µM, respectively[38]
41Iso-agelasidine BAgelas nakamuraiCytotoxicity against HL-60 and K562 cell lines, with IC50 values of 33.0 and 39.2 µM, respectively[38]
42(−)-Agelasine DAgelas nakamuraiCytotoxicity against L5178Y mouse lymphoma cells, with an IC50 value of 4.03 µM[39]
43(−)-Agelamide DAgelas nakamuraiCytotoxicity against L5178Y mouse lymphoma cells, with an IC50 value of 12.5 µM[39,40,41]
42(−)-Agelasine DAgelas nakamuraiCytotoxic to Hep3B cells, with a GI50 of 9.9 µM[41]
43(−)-Agelamide DAgelas nakamuraiCytotoxic to Hep3B cells, with a GI50 of 12.0 µM[41]
99NN *Dysidea cf. arenariaCytotoxicity against NBT-T2 rat bladder epithelial cells, with an IC50 value of 1.9 µg/mL[42]
103104NNDysidea cf. arenariaCytotoxicity against NBT-T2 rat bladder epithelial cells, with IC50 values of 1.8 and 4.2 µg/mL, respectively[42]
105108NNDysidea cf. arenariaCytotoxicity against the NBT-T2 cell line, with IC50 values of 3.1, 1.9, 8.4, and 3.1 µM, respectively[43]
1122,5-Dihydroxy-homoverrucos-(3)-eneHalichondria sp.Cytotoxicity against the human multiple myeloma cell line RPMI-8266, with an IC50 of 49.0 µM[44]
1132-Hydroxy-5-oxo-homoverrucos-(3)-eneHalichondria sp.Cytotoxicity against RPMI-8266 cells, with an IC50 of 65.8 µM[44]
1145,18-Dihydroxy-homoverrucosaneHalichondria sp.Cytotoxicity against RPMI-8266 cells, with an IC50 of 32.7 µM[44]
1155-Hydroxy-18-aldehyde-homoverrucosaneHalichondria sp.Cytotoxicity against RPMI-8266 cells, with an IC50 of 49.3 µM[44]
116HalioxepineHaliclona sp.Cytotoxicity against NBT-T2 cells, with an IC50 of 4.8 µg/mL[45]
124125, 127128,130Hamigerans M–QHamigera tarangaensisCytotoxicity against the HL-60 cell line, with IC50 values of 6.9, 19.5, 14.7, 21.3, and 33.3 µM, respectively[46]
12618-Epi-hamigeran NHamigera tarangaensisCytotoxicity against the HL-60 cell line, with an IC50 value of 14.1 µM[46]
12918-Epi-hamigeran PHamigera tarangaensisCytotoxicity against the HL-60 cell line, with an IC50 value of 11.6 µM[46]
1363β-Hydroxyspongia-13(16),14-diene-2-oneHyattella aff. intestinalisCytotoxicity against NBT-T2 cells, with an IC50 value of 24.1 µM[47]
1436,10,18-Triacetoxy-2E,7E-dolabelladienLuffariella variabilisCytotoxicity against the MDA-MB-231 cell line, with an IC50 value of 11.57 µM[48]
145NNPseudoaxinella flavaCytotoxicity against the PC3 cell line, with an IC50 value of 7 µM[49]
1723β-Hydroxyspongia-13(16),14-dien-2-oneSpongia tubuliferaActivity against A549, human skin melanoma A2058, hepatocyte carcinoma HepG2, and pancreas carcinoma MiaPaca-2 cell lines, with IC50 values of 88.1, 71.4, 91.3, and 90.0 µM, respectively[50]
191Epoxynorspongian ESpongia sp.Activity against the PC3 and PBL-2H3 cell lines, with IC50 values of 24.8 and 27.2 µM, respectively[51]
1992β,3α,19-Triacetoxy-17-hydroxyspongia-13(16),14-dieneSpongia officinalis Linnaeus, 1759Cytotoxicity against the K562 cell line, with an IC50 value of 7.3 µM[52]
206Ceylonamide GSpongia sp.Inhibited the growth of DU145 cells in two-dimensional monolayer culture, with an IC50 of 6.9 µM; also effective on spheroids of a three-dimensional DU145 cell culture model with a minimum effective concentration of 10 µM[53]
209211Gracilins J–LSpongionella sp.Cytotoxic activity against K562 cells and normal human peripheral blood mononuclear cells (PBMCs), with IC50 values of 15 and 30, 8.5 and 9, and 2.65 and 3 µM, respectively[54]
2123′-NorspongiolactoneSpongionella sp.Cytotoxic activity against K562 cells and normal PBMCs, with IC50 values of 12 and 30 µM, respectively[54]
213Spongionellol ASpongionella sp.Activity in the cell lines PC3, PC3-DR, DU145, DU145-DR, 22Rv1, VCaP, and LNCaP, with IC50 values of 0.96, 1.23, 0.94, 1.53, 2.64, 1.30, and 1.02 µM, respectively[55]
224Luakuliide AUnidentifiedActivity against HL-60 cells, with an IC50 value of 21.7 µM[56]
227Chromodorolide DUnidentifiedCytotoxicity against the NBT-T2 cell line, with an IC50 value of 5.6 µg/mL[57]
228NNUnidentifiedCytotoxicity against the NBT-T2 cell line, with an IC50 value of 12 µg/mL[57]
Antibacterial activity
2510-Hydro-9-hydroxyagelasine FAgelas nakamuraiInhibited the growth of Mycobacterium smegmatis, with inhibition zones of 10 mm at 20 µg/disc[58]
33(+)-10-Epiagelasine BAgelas citrinaActive against Staphylococcus aureus ATCC 29213, S. aureus USA300LAC, Streptococcus pneumoniae ATCC 49619, S. pneumoniae 549 CHUAC, Enterococcus faecalis ATCC 29212, E. faecalis 256 CHUAC, and E. faecium 214 CHUAC, with MIC (minimum inhibitory concentration) values of 1, 2, 4, 8, 4, 4, and 4 µg/mL, respectively[59]
36(+)-Agelasine BAgelas mauritianaActive against a panel of methicillin-resistant S. aureus (MRSA) clinical strains 2010-260, 2010-210, 2010-292, and 2010-300, as well as a methicillin-susceptible S. aureus strain H608, with MIC90 values of 2, 1, 2, 1, and 2 µg/mL, respectively[37]
40Iso-agelasine CAgelas nakamuraiAntibacterial activities against Proteusbacillus vulgaris, with an MIC value of 18.75 µg/mL[38]
4649Agelasines O–RAgelas sp.Inhibited the growth of S. aureus and Bacillus subtilis, with MIC values of 16 and 16, 32 and 32, 8 and 8, and 8 and 8 µg/mL, respectively[60]
51Agelasine TAgelas sp.Inhibited the growth of S. aureus and B. subtilis, with MIC values of 16 and 16 µg/mL, respectively[60]
66Eleganstone ADactylospongia elegansAntibacterial activity against Escherichia coli, B. subtilis, and S. aureus, with an MIC value of 64 µg/mL[61]
67(1R*,2E,4R*,8E,10S*, 11S*,12R*)-10,18-diacetoxydolabella-2,8-dien-6-oneDactylospongia elegansAntibacterial activity against E. coli, B. subtilis, and S. aureus, with an MIC value of 64 µg/mL[61]
85Dendrillin BDendrilla antarcticaAchieved 90% eradication at 100 µg/mL in the MRSA biofilm assay[62]
88DarwinolideDendrilla membranosaCytotoxicity against MRSA, with an MIC of 132.9 μM, and activity against the biofilm formation of the same MRSA strain, with an IC50 value of 33.2 µM[63]
138Monamphilectine AHymeniacidon sp.Showed 43% and 38% of the bactericidal strength of the β-lactam antibiotics carbenicillin and amphicillin, respectively, against E. coli at a concentration of 150 nM[64]
203Spongenolactone ASpongia sp.Exhibited 46%, 47%, and 93% inhibition against S. aureus at 50, 100, and 200 µM, respectively[65]
204Spongenolactone BSpongia sp.Displayed 24%, 42%, and 40% inhibition against S. aureus at 50, 100, and 200 µM, respectively[65]
Antituberculosis activity
5759Macfarlandins F–HChelonaplysilla sp.Inhibited Mycobacterium tuberculosis, with MIC values of >20, 49, and >20 µg/mL, respectively[66]
138Monamphilectine AHymeniacidon sp.Activity against M. tuberculosis H37Rv, with an MIC value of 15.3 µg/mL[64]
2197-Methylaminoisoneoamphilecta-1(14),15-dieneSvenzea flavaActivity against M. tuberculosis H37Rv, with an MIC value of 15 µg/mL[67]
2207-Formamidoisoneoamphilecta-1(14),15-dieneSvenzea flavaActivity against M. tuberculosis H37Rv, with an MIC value of 32 µg/mL[67]
Antifungal activity
40Iso-agelasine CAgelas nakamuraiActivity against Candida albicans, with an MIC value of 4.69 µg/mL[38]
41Iso-agelasidine BAgelas nakamuraiActivity against C. albicans, with an MIC value of 2.34 µg/mL[38]
46Agelasine OAgelas sp.Inhibited the growth of Trichophyton mentagrophytes and Cryptococcus neoformans, with IC50 values of 32 and 16 µg/mL, respectively[60]
47Agelasine PAgelas sp.Inhibited the growth of C. neoformans, with an IC50 value of 32 µg/mL[60]
4849Agelasines Q–RAgelas sp.Inhibited the growth of Aspergillus niger, T. mentagrophytes, C. albicans, and C. neoformans, both with IC50 values of 16, 16, 16, and 8 µg/mL, respectively[60]
51Agelasine TAgelas sp.Inhibited the growth of C. neoformans, with an IC50 value of 16 µg/mL[60]
Antiparasitic activity
608-Isocyanoamphilecta-11(20),15-dieneCiocalapata sp.Activity against Plasmodium falciparum K1, with an IC50 value of 0.98 µM[68]
61(1S,3S,4R,7S,8S,11S,12S,13S,15R,20R)-7-Formamido-20-isocy-anoisocycloamphilectaneCymbastela hooperiInhibitory effects on three strains of P. falciparum (FCR3F86, W2, and D6), with an average IC50 value of 0.5 µg/mL[69]
62(1S,3S,4R,7S,8S,11S,12S,13S,15R,20R)-7,20-Diformamidoisocy-cloamphilectaneCymbastela hooperiInhibitory effects on P. falciparum FCR3F86, with an IC50 value of 14.8 µg/mL[69]
83Membranoid BDendrilla antarcticaActivity against Leishmania donovani (IC50 0.8 µM), with no discernible cytotoxicity against uninfected J774A.1 cells[70]
84Membranoid DDendrilla antarcticaActivity against L. donovani (IC50 1.4 µM), with no discernible cytotoxicity against uninfected J774A.1 cells[70]
85Dendrillin BDendrilla antarcticaActivity against the leishmaniasis parasite, with an IC50 value of 3.5 µM[62]
92Diacarperoxide HDiacarnus megaspinorhabdosaActivity against P. falciparum (W2 clones) in vitro, with an IC50 value of 12.9 µM[71]
93Diacarperoxide IDiacarnus megaspinorhabdosaActivity against P. falciparum (W2 and D6 clones) in vitro, with IC50 values of 4.8 and 7.9 µM, respectively[71]
94Diacarperoxide JDiacarnus megaspinorhabdosaActivity against P. falciparum (W2 and D6 clones) in vitro, with IC50 values of 1.8 and 1.6 µM, respectively[71]
138Monamphilectine AHymeniacidon sp.Activity against a chloroquine-resistant (CQ-R) P. falciparum W2 strain, with an IC50 value of 0.60 µM[64]
217218Monamphilectines B–CSvenzea flavaActivity against P. falciparum, with IC50 values of 44.5 and 43.3 nM, respectively[72]
Anti-inflammatory activity
133134Hipposponlachnins A–BHippospongia lachneInhibitory activity on the release of β-hexosaminidase in DNP-IgE-stimulated RBL-2H3 cells, with IC50 values of 49.37 and 23.91 µM, respectively[73]
141Erectcyanthin BHyrtios erectusInhibited 5-LOX, COX-2, and COX-1, with IC50 values of 0.88, 0.98, and 1.09 mM, respectively[74]
193SponalactoneSpongia officinalisInhibitory activity on the lipopolysaccharide (LPS)-induced nitric oxide (NO) production in RAW264.7 macrophages, with an IC50 value of 32 µM[75]
19417-O-acetylepispongiatriolSpongia officinalisInhibitory activity on the LPS-induced NO production in RAW264.7 macrophages, with an IC50 value of 15 µM[75]
19517-O-acetylspongiatriolSpongia officinalisInhibitory activity on the LPS-induced NO production in RAW264.7 macrophages, with an IC50 value of 12 µM[75]
19615α,16α-Dimethoxy-15,16-dihydroepispongiatrioSpongia officinalisInhibitory activity on the LPS-induced NO production in RAW264.7 macrophages, with an IC50 value of 22 µM[75]
19715α-Ethoxyepispongiatriol-16(15H)-oneSpongia officinalisInhibitory activity on the LPS-induced NO production in RAW264.7 macrophages, with an IC50 value of 12 µM[75]
19817-DehydroxysponalactoneSpongia sp.Inhibited superoxide anion generation (91%) and elastase release (90%) at 10 µM, with IC50 values of 3.37 and 4.07 µM, respectively[76]
203205Spongenolactones A–CSpongia sp.Inhibitory activity against superoxide anion generation in fMLF/CB-stimulated human neutrophils, with IC50 values of 16.5, 13.1, and 17.4 µM, respectively[65]
221TedanolTedania ignisShowed anti-inflammatory activity through the inhibition of carrageenan-induced paw edema in mice[77]
Antifouling activity
1116Kalihinols O–TAcanthella cavernosaActivity against Balanus amphitrite larvae, with EC50 (50% effective concentration) values of 1.43, 0.72, 1.48, 1.16, 0.53, and 0.74 µM, respectively[32]
42(−)-Agelasine DAgelas nakamuraiInhibited the growth of planktonic forms of the biofilm-forming bacteria Staphylococcus epidermidis (MIC < 0.0877 µM), but did not inhibit biofilm formation[39]
43(−)-Agelamide DAgelas nakamuraiInhibited only the biofilm formation but not the growth of S. epidermidis[39]
819,11-Dihydrogracilin ADendrilla antarcticaReduced the area covered by the fouling organisms[78]
829,11-Dihydrogracillinone ADendrilla antarcticaReduced the area covered by the fouling organisms[78]
139Hymerhabdrin AHymerhabdia sp.Toxic against larvae of the barnacle Balanus amphitrite, with an IC50 of 3.6 µg/mL[79]
Inhibition of osteoclasts
153154Ceylonamides A–BSpongia ceylonensisInhibitory effects on RANKL-induced osteoclastogenesis in RAW264 macrophages, with IC50 values of 13 and 18 µM, respectively[80]
160Ceylonin ASpongia ceylonensisInhibited the RANKL-induced formation of multinuclear osteoclasts in RAW264 cells by 70% (50 µM), in a dose-dependent manner, without cytotoxicity[81]
163165Ceylonins D–FSpongia ceylonensisInhibited the RANKL-induced formation of multinuclear osteoclasts by 28%, 47%, and 31%, respectively[81]
Others
43(−)-Agelamide DAgelas nakamuraiEnhanced the radiation sensitivity of Hep3B cells; enhanced the efficacy of radiotherapy in a hepatocellular carcinoma (HCC) xenograft mouse model[41]
5355Agelasines W–YAstrosclera willeyanaInhibited the Cbl-b protein that negatively regulates T-cell activation, with IC50 values of 57, 72, and 66 µM, respectively[82]
116HalioxepineHaliclona sp.Activity against 1,1-diphenyl-2-picrylhydrazyl (DPPH), with an IC50 of 3.2 µg/mL[45]
141Erectcyanthin BHyrtios erectusScavenged DPPH and ABTS+, with IC50 values of 0.45 and 0.40 mM, respectively[74]
141Erectcyanthin BHyrtios erectusActivity against 3-hydroxy-3-methylglutaryl-coenzyme A reductase, with an IC50 value of 0.07 mM[74]
144Niphateolide ANiphates olemdaInhibited the p53-Hdm2 (human Mdm2) interaction, with an IC50 value of 16 µM[83]
146RaspadieneRaspailia bouryesnaultaeInhibited the replication of HSV-1 (KOS and 29R strains) at 100 µg/mL by 83% and 74%, respectively[84]
15118-Nor-3,5,17-trihydroxyspongia-3,13(16),14-trien-2-oneSpongia sp.Inhibited aromatase in a dose-dependent manner, with an IC50 value of 34.4 µM[85]
15118-Nor-3,5,17-trihydroxyspongia-3,13(16),14-trien-2-oneSpongia sp.Doubled the quinone reductase 1 (QR1) activity in cultured Hepa 1c1c7 cells at 11.2 µM[85]
209Gracilin JSpongionella sp.Restored mitochondrial activity of neurons to control levels of 98.9% ± 4.7% (p < 0.001) at 0.1 µM, reversing the 28.6% ± 3.4% decrease caused by 200 µM H2O2 treatment[86]
21426-O-etfhylstrongylophorine-14Strongylophora strongilataInhibiting protein tyrosine phosphatase 1B (PTP1B), associated with type 2 diabetes, with an IC50 value of 8.7 µM[87]
* Not named.

3.2. Acanthodendrilla

Only two diterpenes were reported to be generated by the Acanthodendrilla sponges. These two molecules are spongian diterpenes, named 3β-acetoxy-15-hydroxyspongia-12-en and 3-methylspongia-3,12-dien-16-one (1819, Figure 4). They were isolated from the marine sponge Acanthodendrilla sp., collected in Pulau-Pulau [88]. Compounds 18 and 19 represent new chemical entities of the known spongian diterpene family. Compound 18 is a new 3-acetoxyspongia, while compound 19 represents an unreported rearranged 3-methylspongia-3-en. This is the first report of spongian diterpenes from the Acanthodendrilla genus. The cytotoxic potential of compounds 18 and 19 against A549, MDA-MB-231, human colorectal carcinoma cells (HT-29), and human pancreatic adenocarcinoma cells (PSN1) was evaluated. Both compounds proved to be inactive in the tested cancer cell lines. Their GC50 values (drug concentration causing a 50% reduction in the net protein increase) were over 27.5 and 33.3 µM, respectively, for all cell lines.

3.3. Agelas

Agelas sponges are prevalent in tropical and subtropical marine environments and are a significant source of bioactive natural products. The sponges of the genus Agelas have yielded a diverse array of metabolites, with 355 new ones reported between 1971 and 2021 [89]. Among over 19 species of Agelas sponges, A. mauritiana and A. oroides are the most prolific, yielding 45 and 36 metabolites respectively [90]. Specifically, regarding diterpenes, from 2009 to 2022, a total of 33 molecules were isolated from Agelas sponges, a number second only to those isolated from the genus Spongia.
Three pyrimidine diterpenes, named axistatins 1–3 (2022, Figure 4), were isolated from the Palauan marine sponge Agelas axifera Hentschel [34]. All of the isolated compounds were found to be inhibitors of cancer cell growth. Axistatins 1–3 showed inhibition against the murine lymphocytic leukemia P388, pancreatic adenocarcinoma BXPC-3, breast adenocarcinoma MCF-7, CNS glioblastoma SF-268, lung large-cell carcinoma NCI-H460, colon adenocarcinoma KM20L2, and prostate carcinoma DU-145 cell lines, with GI50 values of 19.8, 22.8, and 8.9; 4.8, 5.5, and 6.0; 5.7, 6.8, and 5.8; 3.6, 3.9, and 3.5; 4.6, 4.3, and 5.4; 4.1, 4.1, and 6.9; and 4.8, 5.0, and 7.5 µM, respectively. Axistatins 1–3 also exhibited antimicrobial activity. A brief review has introduced promising anti-CNS-tumor active substances derived from marine sponges that were published between 1994 and 2014 [91]. Axistatins 1–3 are among the eight compounds featured in this review.
Three N-methyladenine-containing diterpenes, named 2-oxoagelasines A and F (2324, Figure 4) and 10-hydro-9-hydroxyagelasine F (25), were isolated from the Okinawan marine sponge Agelas nakamurai Hoshino. Antibacterial experiments were performed on Mycobacterium smegmatis NBRC 3207 using the paper disc method. Compound 25 inhibited the growth of M. smegmatis, with an inhibition zone of 10 mm at 20 µg/disc [58].
Two N-methyladenine-containing diterpenes, nemoechines F–G (2627, Figure 4), were isolated from the South China Sea sponge Agelas aff. nemoechinata. Compound 27 exhibited cytotoxicity against Jurkat cell lines, with an IC50 value of 17.1 µM [35].
Diterpenoid alkaloids are complex natural compounds that are primarily derived from specific plant genera and marine organisms, exhibiting a range of biological activities, from medicinal applications to potent neurotoxicity [92]. Marine sponges have also been reported to produce different diterpenoid alkaloids. From the South China Sea sponge Agelas aff. nemoechinata, a diterpene-adenine alkaloid called nemoechine D (28, Figure 4) was obtained [36]. It showed cytotoxicity against the HL-60 cell line, with an IC50 value of 9.9 µM.
Three diterpene alkaloids, agelasidines E–F and agelasine N (2931, Figure 5), were isolated from the Caribbean sponge Agelas citrina [93]. This represents the first report of natural products from the sponge A. citrina. Three diterpene alkaloids, (+)-8-epiagelasine T, (+)-10-epiagelasine B, and (+)-12-hydroxyagelasidine C (3234, Figure 5), were also obtained from A. citrina [59]. The evaluation of antimicrobial activity against the Gram-positive pathogens S. aureus, Streptococcus pneumoniae, and Enterococcus faecalis showed that compound 33 was active against all of the tested strains, with MIC values in the range of 1–8 µg/mL.
Five diterpene alkaloids, (−)-8′-oxo-agelasine B, (+)-agelasine B, (+)-8′-oxo-agelasine C, agelasine V, and (+)-8′-oxo-agelasine E (3539, Figure 5), were isolated from the sponge Agelas mauritiana [37]. Compounds 35 and 3739 are the second examples of 8′-oxo-agelasine analogs. Compound 36 not only exhibited cytotoxicity toward the cancer cell lines PC9, A549, HepG2, MCF-7, and U937, with IC50 values of 4.49–14.07 µM, but also showed antibacterial activities against a panel of clinical MRSA isolates, with MIC values of 1–2 µg/mL (Table 1).
Two diterpene alkaloids, iso-agelasine C and iso-agelasidine B (4041, Figure 5), were isolated from the South China Sea sponge Agelas nakamurai [38]. Compound 40 showed cytotoxicity against the HL-60, K562, and HCT-116 cell lines, with IC50 values of 25.3, 28.9, and 38.8 µM, respectively. Compound 41 showed cytotoxicity against the HL-60 and K562 cell lines, with IC50 values of 33.0 and 39.2 µM, respectively. Compound 40 also exhibited antibacterial activities against Proteusbacillus vulgaris, with an MIC value of 18.75 µg/mL. Moreover, compounds 40 and 41 showed antifungal activities against Candida albicans, with MIC values of 4.69 and 2.34 µg/mL, respectively. Another two diterpene alkaloids, (−)-agelasine D and (−)-ageloxime D (4243, Figure 6), were also obtained from the Agelas nakamurai sponge [39]. They exhibited cytotoxicity against L5178Y mouse lymphoma cells, with IC50 values of 4.03 and 12.5 µM, respectively. Moreover, compound 42 inhibited the growth of planktonic forms of the biofilm-forming bacteria Staphylococcus epidermidis (MIC < 0.0877 µM), but it did not inhibit biofilm formation, whereas compound 43 showed the opposite activity profile and inhibited only biofilm formation but not bacterial growth.
It is worth mentioning that the reported compound 43, named (−)-ageloxime D, was revised as (+)-N-[4-amino-6-(methylamino)pyrimidin-5-yl]-N-copalylformamide, which was produced via hydrolysis of agelasine D [40]. It was later renamed (−)-agelamide D [41]. (−)-Agelasine D was more cytotoxic to Hep3B cells than (−)-agelamide D, with their GI50 values being 9.9 and 12.0 µM, respectively [41]. It was found that (−)-agelamide D enhanced the radiation sensitivity of Hep3B cells, reducing their ability to form colonies and boosting the rate of apoptosis [41]. It also upregulated the expression of protein kinase RNA-like endoplasmic reticulum kinase/inositol-requiring enzyme 1α/activating transcription factor 4 (PERK/eIF2α/ATF4), a pivotal pathway in the unfolded protein response (UPR) across various HCC cell lines, thereby intensifying the UPR signaling triggered by radiation. In vivo xenograft studies validated that (−)-agelamide D amplified the suppressive effect of radiation on tumor growth, without causing systemic toxicity. Immunohistochemistry confirmed that (−)-agelamide D elevated ATF4 expression and the incidence of apoptosis induced by radiation, aligning with the in vitro observations.
Diterpene alkaloids were also found from an Okinawan marine sponge (Agelas sp.) [94]. Agelamasine A (44, Figure 6) is the first diterpene alkaloid with a rearranged (4→2)-abeo-clerodane skeleton from a marine source, while agelamasine B (45, Figure 6) is a clerodane diterpene alkaloid. Agelasines O–U (4652, Figure 6) are also diterpene alkaloids isolated from the Okinawan marine sponge Agelas sp. [60]. Agelasines O–R were the third examples of diterpene alkaloids with a 9-N-methyladenine and a pyrrole unit. Agelasine O has a halimane skeleton, while agelasines P–R have a clerodane skeleton. Agelasines S–U are new diterpene alkaloids with a 9-N-methyladenine unit consisting of a halimane skeleton, a labdane skeleton, and a clerodane skeleton, respectively. Agelasines O–R and T showed antimicrobial activities against several bacterial and fungal strains (Table 1).

3.4. Astrosclera

The Astrosclera sponges yielded three diterpenes in total. The three N-methyladenine-containing diterpenes, agelasines W–Y (5355, Figure 7), were isolated from a specimen of Astrosclera willeyana collected in 1997 and frozen ever since. Agelasines W–Y have bicyclic terpenoid skeletons with a prenyl side chain that terminates with an N-methyladenine subunit. In terms of activity, these three compounds can inhibit the Cbl-b protein, with IC50 values of 57, 72, and 66 µM, respectively [82]. Cbl-b negatively regulates T-cell activation and, thus, lowers the immune system’s reaction to cancer cells. Agelasines W–Y could be promising immunotherapy agents for enhancing antitumor immunity by inhibiting the Cbl-b protein.

3.5. Cacospongia

Two unusual C17 γ-lactone norditerpenoids (a pair of inseparable enantiomers, 56a and 56b, Figure 7) were obtained from the marine sponge Cacospongia sp. as a mixture [95]. They bore the 4S,5S and 4R,5R absolute configurations, respectively. It should be noted that since these two compounds exist in the form of a mixture, they have been counted as one when tallying the number of diterpenes from different genera of sponges.

3.6. Chelonaplysilla

The Chelonaplysilla sponges yielded three diterpenes in total. These three diterpenes, macfarlandins F–H (5759, Figure 7), were obtained from a sample of the marine sponge Chelonaplysilla sp. collected in Samoa [66]. Structurally, macfarlandins F and H are the first members of this family to have oxygenation at C-2, and macfarlandins G and H are the first to have a monocyclic δ-lactone heterocycle attached to their decalin ring system. In addition, macfarlandin G exhibited activity against Mycobacterium tuberculosis, with an MIC of 49 µg/mL, whereas macfarlandins F and H both exhibited MICs > 20 µg/mL.

3.7. Ciocalapata

Only one diterpene was reported to be produced by the Ciocalapata sponge. This molecule is a rare isonitrile diterpene named 8-isocyanoamphilecta-11(20),15-diene (60, Figure 7), which was isolated from a cryopreserved sample of a Ciocalapata sp. sponge [68]. The antimalarial activity of isonitrile terpenoids has long been reported. Coincidentally, compound 60 possesses strong activity against Plasmodium falciparum K1, with an IC50 value of 0.98 µM.

3.8. Cymbastela

The total number of diterpenes reported from the Cymbastela sponges is five. These five diterpene formamides (6165, Figure 7) were obtained from the tropical marine sponge Cymbastela hooperi [69]. Compound 61 ((1S,3S,4R,7S,8S,11S,12S,13S,15R,20R)-7-formamido-20-isocyanoisocycloamphilectane) contains both formamide and isonitrile functionalities, which is not usual for natural products. Through in vitro antiplasmodial bioassays, compound 61 was found to have better activity (IC50 0.5 µg/mL) than compound 62 ((1S,3S,4R,7S,8S,11S,12S,13S,15R,20R)-7,20-diformamidoisocycloamphilectane) (IC50 14.8 µg/mL), whereas compounds 6365 were inactive.

3.9. Dactylospongia

The Dactylospongia sponges yielded two diterpenes in total. These two diterpenes were isolated from the marine sponge Dactylospongia elegans. Eleganstone A (66, Figure 8) is a rare diterpene with a 5/6/4/5 fused tetracyclic ring skeleton, and compound 67 belongs to the dolabellane diterpenes, which had not been discovered from the genus Dactylospongia previously [61]. The antibacterial activity of these two compounds was evaluated, and their MIC values were 64 µg/mL against E. coli, B. subtilis, and S. aureus.

3.10. Darwinella

The Darwinella sponges yielded 13 diterpenes in total. Nine nitrogenous spongian diterpenes—oxeatamide A (68, Figure 8), iso-oxeatamide A (69), oxeatamides B-G (7075), and oxeatamide A 23-methyl ester (76)—were isolated from the sponge Darwinella oxeata [96]. They were tested for cytotoxicity in a 48 h MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay against HL-60 cells, and they all showed IC50 values >10 µM.
Four rearranged diterpenoids, oxeatine (77, Figure 8) and oxeatamides H–J (7880), were isolated from the sponge Darwinella cf. oxeata [97]. Oxeatine has a new heterocyclic skeleton, and oxeatamide J has an N-methyl urea group included in a γ-lactam moiety. The 4,8-dimethyl-5-(1,3,3-trimethylcyclohexyl)-octahydro-1H-2λ2-isoquinoline heterocyclic skeleton found in oxeatine, which features a δ-lactam with the nitrogen atom bridging C-6 and C-15 of a rearranged spongian carbon framework, was unknown in nature or from synthesis.

3.11. Dendrilla

A total of 11 diterpenes were obtained from Dendrilla sponges. Two norditerpenes, named 9,11-dihydrogracilin A (81, Figure 9) and 9,11-dihydrogracillinone A (82), were isolated from the Antarctic sponge Dendrilla antarctica [78].
Their antifouling ability was tested using soluble-matrix paints, and both compounds showed activity against a variety of colonizing organisms. Compound 82 demonstrated a more pronounced antifouling effect, with a smaller area covered by the solitary colonial ascidian (Botrylloides sp.) compared to compound 81.
Two diterpenes bearing one or more methyl acetal functionalities, membranoids B and D (8384, Figure 9), were also obtained from the Dendrilla antarctica sponge [70]. They displayed low micromolar activity (IC50 values of 0.8 and 1.4 µM, respectively) against Leishmani donovani, with no discernible cytotoxicity against uninfected J774A.1 cells. Membranoids B and D are considered to be artifacts and can be obtained by treating aplysulfurin with methanol. Dendrillins B–D (8587, Figure 9) were also produced by the D. antarctica sponge [62]. Dendrillin B showed an IC50 of 3.5 µM against the leishmaniasis parasite. Moreover, it achieved 90% eradication at 100 µg/mL in the MRSA biofilm assay.
A rearranged spongian diterpene, darwinolide (88, Figure 9), has been isolated from the Antarctic Dendroceratid sponge Dendrilla membranosa [63]. A broth dilution assay determined darwinolide’s MIC against MRSA to be 132.9 μM. It was also found that darwinolide was cytotoxic, rather than cytostatic, toward S. aureus. Further experiments revealed an IC50 value of 33.2 µM against the biofilm formation of the same MRSA strain. Hence, the compound darwinolide displays 4-fold selectivity against the biofilm phase of MRSA compared to the planktonic phase.
Three spongian diterpenes, named aplyroseols 20–22 (8991, Figure 9), were isolated from two specimens of the Australian marine sponge Dendrilla rosea [98]. The 3-hydroxybutyrate present in aplyroseol 22 was reported in the spongian diterpenes for the first time. The compounds were screened for activity against Staphylococcus aureus, but they were inactive at 64 µg/mL.

3.12. Diacarnus

The Diacarnus sponges yielded six diterpenes in total. These included five norditerpene endoperoxides, diacarperoxides H–L (9296, Figure 10), together with a norditerpene diol, diacardiol B (97).
These were isolated from the South China Sea sponge Diacarnus megaspinorhabdosa [71]. Diacarperoxides H–J showed antimalarial activity against Plasmodium falciparum (W2 clones) in vitro, with IC50 values of 12.9, 4.8, and 1.8 µM, respectively, while diacarperoxides H–I exhibited activity against P. falciparum (D6 clones), with IC50 values of 7.9 and 1.6 µM, respectively. The IC50 values of the control drug artemisinin were 0.14 µM (W2 clones) and 0.071 (D6 clones).

3.13. Dysidea

A total of 13 diterpenes were obtained from Dysidea sponges. Seven spongian-class diterpenes (98104, Figure 11) were isolated from the sponge Dysidea cf. arenaria collected in Okinawa [42]. Compounds 99, 103, and 104 showed cytotoxicity against NBT-T2 rat bladder epithelial cells, with IC50 values of 1.9, 1.8, and 4.2 µg/mL, respectively. Four diterpenes, compounds 105108 (Figure 11), were isolated from a sample of the Dysidea cf. arenaria sponge [43]. They were found to have inhibitory effects on NBT-T2 cells, with IC50 values of 3.1, 1.9, 8.4, and 3.1 µM, respectively. Chromodorolides D–E (109110, Figure 11) were also generated by the Dysidea sp. sponge [99].

3.14. Fascaplysinopsis

Only one diterpene was reported to be produced by the Fascaplysinopsis sponge. The dolabellane diterpenoid, named clavirolide H (111, Figure 11), was isolated from the Xisha sponge Fascaplysinopsis reticulata [100]. Clavirolide H was evaluated for cytotoxic activities against human leukemia K562, HL-60, HeLa, HCT-116, A549, normal human hepatocytes L-02, and human hepatocellular carcinoma BEL-7402 cell lines, but no activity was observed.

3.15. Halichondria

A total of four diterpenes were obtained from Halichondria sponges. These four homoverrucosane-type diterpenes (112115, Figure 11), common in land plants but rare in sponges, were isolated from a sample of the marine sponge Halichondria sp. [44]. This was the first report of homoverrucosanes isolated from the marine sponge Halichondria sp. Compounds 112115 were evaluated for their activities against the RPMI-8266 cell line, and their IC50 values were all greater than 10 µM.

3.16. Haliclona

Only one diterpene was reported from the Haliclona sponges. The marine sponge genus Haliclona contains over 600 species, but only a small number of them have been classified and chemically investigated [21]. The diterpenoid halioxepine (116, Figure 12) is a meroditerpene produced by Haliclona sp. [45]. It showed cytotoxicity against NBT-T2 cells (RIKEN), with an IC50 value of 4.8 µg/mL. It also showed antioxidant activity against DPPH, with an IC50 of 3.2 µg/mL.

3.17. Hamigera

The Hamigera sponges yielded 16 diterpenes in total. Hamigera sponges, belonging to the Hymedesmiidae family, are second only to the genus Phorbas in the same family in terms of the richness of obtained secondary metabolites [16].
The 16 hamigeran diterpenoids were isolated from the New Zealand marine sponge Hamigera tarangaensis (117132, Figure 12) [46,101]. The compound hamigeran R (117) was the first benzonitrile-based marine natural product, while the compound hamigeran S (118) was the first dimeric structure in the series. The formation of hamigerans R and S is thought to occur via the reaction of hamigeran G with a nitrogen source, where the nitrile carbon of hamigeran R is derived from the terpenoid skeleton.
The compound hamigeran M (124) represents the first instance of a non-benzo-fused, oxazole-containing terpenoid isolated from the marine environment. Compounds 124130 were tested for their cytotoxicity against the HL-60 cell line using the MTT method, and their IC50 values were 6.9, 19.5, 14.1, 14.7, 21.3, 11.6, and 33.3 µM, respectively.

3.18. Hippospongia

Two diterpenes were produced by Hippospongia sponges. These two diterpenes, hipposponlachnins A and B (133134, Figure 13), with antiallergic activity, were obtained from the South China Sea marine sponge Hippospongia lachne [73]. They possess an unprecedented tetracyclo [9.3.0.02,8.03,7] tetradecane ring system. Hipposponlachnins A and B did not cause significant cytotoxicity in rat basophilic leukemia (RBL-2H3) cells after 24 h of treatment. They showed inhibitory activity on the release of β-hexosaminidase in DNP-IgE-stimulated RBL-2H3 cells, with IC50 values of 49.37 and 23.91 µM, respectively, higher than that of the market-available anti-asthmatic drug ketotifen fumarate (IC50 = 63.88 µM). In addition, hipposponlachnins A and B also suppressed IL-4 production in a dose-dependent manner and significantly inhibited LTB4 release in activated RBL-2H3 cells compared with untreated controls. The results indicate that these are promising antiallergic lead compounds.

3.19. Hyattella

The Hyattella sponges yielded three diterpenes in total. These three diterpenes, named 2α-hydroxyspongia-13(16),14-diene-3-one (135, Figure 13), 3β-hydroxyspongia-13(16),14-diene-2-one (136), and 2α,3α-diacetoxy-17,19-dihydroxyspongia-13(16),14-diene (137), were extracted from the sponge Hyattella aff. intestinalis [47]. They were tested against adenovirus (AdV), and all showed IC50 values > 20 µg/mL. Their cytotoxicity was also examined against NBT-T2 cells using the MTT assay, and compound 136 showed an IC50 value of 24.1 µM.

3.20. Hymeniacidon

Only one diterpene was produced by Hymeniacidon sponges. The molecule monamphilectine A (138, Figure 13) is a diterpenoid β-lactam alkaloid generated by Hymeniacidon sp. [64]. When tested against a CQ-R Plasmodium falciparum W2 strain, it showed an IC50 value of 0.60 µM. In vitro antituberculosis screening against Mycobacterium tuberculosis H37Rv revealed an MIC value of 15.3 µg/mL. Preliminary KB assays against E. coli revealed that, at a concentration of 150 nM, monamphilectine A possesses 43% and 38% of the bactericidal strength of the β-lactam antibiotics carbenicillin and amphicillin, respectively.

3.21. Hymerhabdia

Similarly, only one diterpene was reported from the Hymerhabdia sponges. The unusual diterpene hymerhabdrin A (139, Figure 13) was isolated from an intertidal marine sponge Hymerhabdia sp. [79]. It possessed a novel 6/6/5 fused-ring skeleton. Hymerhabdrin A exhibited antifouling activity against Balanus amphitrite larvae, with an LC50 (lethal concentration 50) value of 3.6 µg/mL.

3.22. Hyrtios

The Hyrtios sponges yielded three diterpenes in total. These three cyanthiwigin-type diterpenes, named erectcyanthins A–C (140142, Figure 13), were isolated from the marine sponge Hyrtios erectus [74]. The compound erectcyanthin B not only exhibited anti-dyslipidemia activity but also possessed antioxidant and anti-inflammatory activities. In the anti-dyslipidemia experiment, erectcyanthin B exhibited activity against 3-hydroxy-3-methylglutaryl-coenzyme A reductase, with an IC50 value of 0.07 mM, compared to 0.08 mM for the drug atorvastatin. The antioxidant activities of erectcyanthin B, assessed using the stable DPPH and ABTS+ scavenging tests, were indicated by IC50 values of 0.45 and 0.40 mM, compared to 1.51 and 1.70 mM of the standard α-tocopherol, respectively. The anti-inflammatory activity of erectcyanthin B (IC50 0.88–1.09 mM, Table 1), assessed using 5-LOX and isoforms of COX-2/1, was superior to that of other erectcyanthin analogs.

3.23. Luffariella

Only one diterpene was generated by Luffariella sponges. The dolabellane diterpene was obtained from the South China Sea sponge Luffariella variabilis and was named 6,10,18-triacetoxy-2E,7E-dolabelladien (143, Figure 13) [48]. This is the first dolabellane-type diterpenoid from the genus Luffariella. Compound 143 showed cytotoxicity against the MDA-MB-231 cell line, with an IC50 value of 11.57 µM.

3.24. Niphates

Similarly, only one diterpene was reported from the Niphates sponges. This compound, named niphateolide A (144, Figure 13), was isolated from the marine sponge Niphates olemda [83]. The inhibitory effect of compound 144 on the p53–Hdm2 interaction was examined using ELISA. It inhibited the interaction, with an IC50 value of 16 µM. Genetic mutations within the p53 tumor suppressor pathway are a common occurrence in human tumors. Mdm2/Hdm2 functions as an E3 ubiquitin ligase for p53 within the ubiquitin–proteasome system. The activity of niphateolide A in inhibiting the p53–Hdm2 interaction enables it to serve as a substance for reactivating p53, thereby giving it potential for further research and development in the field of oncology.

3.25. Pseudoaxinella

Pseudoaxinella is another genus of sponge that has produced only one diterpene. The isonitrile diterpene (145, Figure 13), with anticancer activity, was isolated from the Caribbean sponge Pseudoaxinella flava [49]. Compound 145 showed growth-inhibitory activity against human PC3 prostate cancer cells, with an IC50 value of 7 µM.

3.26. Raspailia

Raspailia sponges also produced one diterpene in total. The clerodane diterpene, named raspadiene (146, Figure 13), was isolated from the marine sponge Raspailia bouryesnaultae collected in South Brazil [84]. The evaluation of potential anti-herpes activity against herpes simplex virus type 1 (HSV-1) showed that the compound raspadiene at 100 µg/mL inhibited the replication of HSV-1 (KOS and 29R strains, sensitive and resistant to acyclovir, respectively) by 83% and 74%, respectively.

3.27. Spongia

The Spongia genus produced the highest number of diterpenoids among all sponge genera. A total of 62 diterpenoids have been derived from Spongia sponges, accounting for 27% of all sponge-derived diterpenoids reported from 2009 to 2022. Many of these molecules exhibit strong biological activity, highlighting the potential and significance of Spongia sponges as a valuable resource for marine drug development.
Three unreported furanoditerpenoids (147149, Figure 14) were isolated from the marine sponge Spongia sp. [102]. Compound 147 is a spongian diterpene with a modified oxidation pattern, while compounds 148 and 149 represent two new ring-A-contracted spongians, displaying a novel and unprecedented norspongian carbon skeleton.
Three compounds, 18-nor-3,17-dihydroxyspongia-3,13(16),14-trien-2-one (150, Figure 14), 18-nor-3,5,17-trihydroxyspongia-3,13(16),14-trien-2-one (151), and spongiapyridine (152), were isolated from an Indonesian sponge of the genus Spongia [85]. Structurally, the D-ring possessed by compound 152 belongs to the pyridyl ring system, which is different from ordinary spongians possessing standard δ-lactone. In the in vitro biological activity experiment, compound 151 exhibited aromatase-inhibitory activity, with an IC50 value of 34.4 µM, and it also induced quinone reductase 1 (QR1) activity in cultured Hepa 1c1c7 cells, with a CD value (the concentration needed to double the QR1 activity) of 11.2 µM. Aromatase is an essential cytochrome P450 enzyme that facilitates the conversion of androgens like testosterone and androstenedione into estrogens such as estradiol and estrone. Inhibiting aromatase reduces the available estrogen and demonstrates considerable effectiveness in preventing certain types of breast cancer. Quinone reductase 1 is a protective enzyme that helps prevent cancer by blocking intracellular semiquinone radicals and producing α-tocopherolhydroquinone, which acts as a chemopreventive agent. Compound 151 holds promise as a potential therapeutic agent for both breast cancer prevention and chemoprevention, due to its dual action as an aromatase inhibitor and an inducer of quinone reductase 1 activity.
Seven spongian diterpenes, ceylonamides A–F (153158, Figure 14) and 15α,16-dimethoxyspongi-13-en-19-oic acid (159), were isolated from the Indonesian marine sponge Spongia ceylonensis [80]. Compounds 153158 are nitrogenous spongian diterpenes. Ceylonamides A and B exhibited inhibitory effects on RANKL-induced osteoclastogenesis in RAW264 macrophages, with IC50 values of 13 and 18 µM, respectively. In a follow-up study of the structure–activity relationship, the authors found that the carbonyl position of the γ-lactam ring and the volume of the substituent on the nitrogen atom had a great influence on the inhibitory effect.
Another nine spongian diterpene derivatives, ceylonins A–F (160165, Figure 14) [81,103] and ceylonins G–I (166168, Figure 15) [81,103], were also isolated from the Spongia ceylonensis sponge. Ceylonins A–F contain three additional carbons in ring D to supply an ether-bridged bicyclic ring system. Ceylonins A and D–F (50 µM) inhibited the RANKL-induced formation of multinuclear osteoclasts in RAW264 cells by 70%, 28%, 47%, and 31%, respectively. The inhibitory activity of ceylonins G–I against the cancer therapeutic drug target ubiquitin-specific protease 7 (USP7) was tested, but their IC50 values were all over 50 µM.
3-Nor-spongiolide A (169, Figure 15), having the rare 3-nor-spongian carbon skeleton, and spongiolides A and B (170171), possessing a γ-butenolide ring instead of the usual furan ring for ring D, were isolated from South China Sea sponge Spongia officinalis [104]. They were evaluated for their cytotoxic activity against HL-60 cells, but none of them exhibited potent cytotoxic activity.
Two furanoditerpenes, 3β-hydroxyspongia-13(16),14-dien-2-one (172, Figure 15) and 19-dehydroxy-spongian diterpene 17 (173), were isolated from the sponge Spongia tubulifera, collected in the Mexican Caribbean [50]. Compound 172 showed activity against the A549, A2058, HepG2, and MiaPaca-2 cell lines, with IC50 values of 88.1, 71.4, 91.3, and 90.0 µM, respectively.
Spongiains A–G (174180, Figure 15) were isolated from the marine sponge Spongia sp. [105]. Spongiains A–C were the first examples of spongian diterpenes bearing a pentacyclic skeleton composed of a fused 5/5/6/6/5 ring system through ring-A rearrangement. The cytotoxic activities of spongiains A–G were evaluated, and none of them exhibited antiproliferative effects on several cancer cell lines.
Twelve norspongian diterpenes, dinorspongians A–F (181186, Figure 15) and epoxynorspongians A-F (187192), were isolated from the marine sponge Spongia sp. [51]. Among them, dinorspongians A–F were the first examples of dinorspongian diterpenes bearing the unprecedented 3,4-seco-3,19-dinorspongian diterpene skeleton. Epoxynorspongian E exhibited inhibitory activity against the PC3 and PBL-2H3 cell lines, with IC50 values of 24.8 and 27.2 µM, respectively.
Five diterpenes—including a 5,5,6,6,5-pentacyclic diterpene, sponalactone (193, Figure 16); two spongian diterpenes, 17-O-acetylepispongiatriol (194) and 17-O-acetylspongiatriol (195); and two spongian diterpene artifacts, 15α,16α-dimethoxy-15,16-dihydroepispongiatriol (196) and 15α-ethoxyepispongiatriol-16(15H)-one (197)—were isolated from the marine sponge Spongia officinalis collected from the South China Sea [75]. The in vitro anti-inflammatory activities of these compounds were tested by inhibition of LPS-induced NO production in RAW264.7 macrophages, with IC50 values of 32, 15, 12, 22, and 12 µM, respectively.
A rare A-ring contracted diterpene, 17-dehydroxysponalactone (198, Figure 16), was isolated from the Red Sea marine sponge Spongia sp. [76]. The in vitro anti-inflammatory activity of the compound was tested. Compound 198 significantly reduced the superoxide anion generation and elastase release, with inhibition rates of 91% and 90%, respectively, at a concentration of 10 µM, and the IC50 values were 3.37 and 4.07 µM, respectively.
A novel acetoxy diterpenoid, 2β,3α,19-triacetoxy-17-hydroxyspongia-13(16),14-diene (199, Figure 16), and 18-nor-2,17-hydroxyspongia-1,4,13(16),14-quaien-3-one (200), belonging to the rare 18-norspongian carbon skeleton, were isolated from the aquaculture Spongia officinalis Linnaeus, 1759 [52]. The in vitro biological activity evaluation showed that compound 199 had cytotoxic activity against the K562 cell line, with an IC50 value of 7.3 µM. A bicyclic diterpene, jellynolide A (201), with a penta-substituted carbon skeleton, was also isolated from this sponge [106]. The compound jellynolide A biogenically implies an irregular non-head-to-tail linkage between GPP (geranyl diphosphate) and isoprene units, as well as a novel cyclization position.
Dinorspongiapyridine (202, Figure 16) is a dinorspongian diterpene produced by the marine sponge Spongia sp. [107]. It was the first instance of a 3,4-seco-3,19-dinorspongian diterpene bearing a rare pyridyl D-ring system.
Spongenolactones A–C (203205, Figure 16) were obtained from a Red Sea sponge Spongia sp. [65]. They are all pentacyclic spongian diterpenes, featuring 5,5,6,6,5, 5,5,6,6,6, and 5,5,6,6,7 ring systems, respectively. They were found to exert inhibitory activity against superoxide anion generation in fMLF/CB-stimulated human neutrophils, with IC50 values of 16.5, 13.1, and 17.4 µM, respectively. Furthermore, spongenolactone A showed higher inhibitory activity against the growth of S. aureus in comparison to B (Table 1).
Ceylonamides G–I (206208, Figure 16) are diterpene alkaloids from an Indonesian marine sponge of Spongia sp. [53]. Ceylonamide G inhibited the growth of DU145 human prostate cancer cells in a two-dimensional monolayer culture, with an IC50 of 6.9 µM. It was also effective (minimum effective concentration of 10 µM) on spheroids of a three-dimensional cell culture model, which was prepared from DU145 cells.

3.28. Spongionella

The Spongionella sponges yielded five diterpenes in total. Four diterpenes, gracilins J–L and 3′-norspongiolactone (209212, Figure 16), were isolated from the extracts of the marine Spongionella sp. sponges [54]. Gracilins J–L belong to the rare classes trisnorditerpenes, bisnorditerpenes, and norditerpenes, respectively. The in vitro cytotoxicity of these compounds was determined using K562 and normal human peripheral blood mononuclear cells (PBMCs). Compounds 209212 showed cytotoxic activity against the K562 cell line, with IC50 values ranging from 2.65 to 15 µM (Table 1). They showed similar or slightly less toxicity against the normal PBMCs, with IC50 values ranging from 3 to 30 µM (Table 1). The activity of gracilins J–L was also evaluated using an oxidative in vitro stress model [86]. The compound gracilin J presented neuroprotection effects at the mitochondrial function level. The neurons’ mitochondrial activity decreased by 28.6 ± 3.4% (p < 0.001) after treatment with 200 µM H2O2. Gracilin J at 0.1 µM reduced this effect, restoring the activity to control levels (98.9 ± 4.7%, p < 0.001).
A spongian diterpene, spongionellol A (213, Figure 16), was obtained from the marine sponge Spongionella sp. [55]. It exhibited activity and selectivity in a panel of seven human prostate cancer cells. The panel included AR-negative PC3 and DU145 cells, which are known to be resistant to various hormonal and standard chemotherapeutics, docetaxel-resistant PC3-DR and DU145-DR cells (derived from PC3 cells and DU145 cells, respectively), AR-FL- (androgen receptor full-length) and AR-V7-positive (androgen receptor splice variant V7) hormone-resistant 22Rv1 and VCaP cells, and AR-FL-positive hormone-sensitive LNCaP cells (Table 1).

3.29. Strongylophora

Only one diterpene was reported to be produced by the Strongylophora sponge. This meroditerpene, 26-O-ethylstrongylophorine-14 (214, Figure 16), was isolated from the Caribbean marine sponge Strongylophora strongilata [87]. It was found to inhibit PTP1B associated with type 2 diabetes, with an IC50 value of 8.7 µM, compared with 0.7 µM for the positive control oleanolic acid. This is the first report of meroditerpenes inhibiting PTP1B activity.

3.30. Stylissa

The Stylissa sponges yielded two diterpenes in total. These two amphilectane-type diterpenes, 8-isocyanato-15-formamidoamphilect-11(20)-ene and 8-isothiocyanato-15-formamidoamphilect-11(20)-ene (215216, Figure 17), were isolated from the sponge Stylissa cf. massa [108]. Their antimalarial activities were evaluated, and both compounds were inactive.

3.31. Svenzea

A total of four diterpenes were obtained from Svenzea sponges. Two isocyanide amphilectane-type diterpenes, named monamphilectines B and C (217218, Figure 17), were isolated from the Caribbean sponge Svenzea flava [72]. Monamphilectines B and C exhibited activities against the human malaria parasite Plasmodium falciparum (the non-resistant (wild-type standard) 3D7 strain), with IC50 values of 44.5 and 43.3 nM, respectively.
Two rare isoneoamphilectane-based diterpenes, 7-methylaminoisoneoamphilecta-1(14),15-diene and 7-formamidoisoneoamphilecta-1(14),15-diene (219220, Figure 17), were also isolated from the Caribbean marine sponge Svenzea flava [67]. Their MIC values against the strain Mycobacterium tuberculosis H37Rv were 15 and 32 µg/mL, respectively.

3.32. Tedania

Only one diterpene was reported from Tedania sponges. The molecule, tedanol (221, Figure 17), was a brominated and sulfated pimarane diterpene isolated from the Caribbean sponge Tedania ignis [77]. Tedanol, which showed good solubility in water, significantly reduced both the acute (4 h) and subchronic (48 h) phases of carrageenan-induced paw edema in mice, which was coupled with a strong inhibition of COX-2 expression, cellular infiltration measured as myeloperoxidase (MPO) levels, and iNOS expression.

3.33. Theonella

The Theonella sponges yielded two diterpenes in total. The two nitrogenous prenylbisabolane diterpenes, named amitorines A and B (222223, Figure 17), were isolated from Theonella swinhoei [109]. No activity data have been reported for them.

3.34. Others

A total of five diterpenes were isolated from other sponges, specifically those whose genera have not been determined.
The Kingdom of Tonga is an archipelago in the central Indo-Pacific Ocean; many novel marine natural products with bioactivities have been reported from organisms collected within Tongan territorial waters [110]. Coincidentally, three labdane diterpenes, luakuliides A–C (224226, Figure 17), were isolated from a Tongan dictyoceratid marine sponge [56]. They share a trans-fused [4.4.0]-bicyclodecane system with a hemi-acetal functional group bridging C-8 and C-10. In terms of activity, luakuliide A has inhibitory activity against HL-60 cells, with an IC50 value of 21.7 µM.
Two diterpenes (227228, Figure 17) were isolated from an Okinawan marine sponge [57]. Compound 227, named chromodorolide D, is an example of a diterpenoid with a highly rearranged chromodorane carbon skeleton, while compound 228 retains the open side chains. Compounds 227 and 228 showed cytotoxicity against NBT-T2 cells, with IC50 values of 5.6 and 12 µg/mL, respectively.

4. Conclusions

This review summarizes the structures and bioactivities of 228 diterpenes reported in 73 research papers, originating from more than 33 different genera of sponges. Notably, the genera Spongia, Agelas, and Acanthella reported the highest number of diterpenes, with counts of 62, 33, and 17, respectively, accounting for 27%, 14%, and 7% of the total reported molecules, respectively. In contrast, in more than 26 genera of sponges, fewer than 10 diterpenoids have been reported per genus, with 12 of these genera producing just one diterpene each.
Most of the reported diterpenes have been evaluated for their bioactivity, with 110 molecules exhibiting one or more types of bioactivity. Among them, the highest numbers were seen in those with cytotoxic, antibacterial, and anti-inflammatory properties, amounting to 54, 22, and 13 molecules, respectively. These correspond to 24%, 10%, and 6% of the reported 228 molecules, respectively.
Some compounds exhibit cytotoxic activity against a variety of tumor cells. For example, the compounds axistatins 1–3 (2022) have shown inhibitory activity against seven different tumor cell lines. Axistatins 1–2 have GI50 values ≤ 5 µM against five and four cell lines, respectively. Axistatin 3 has GI50 values ranging from 3.5 to 8.9 µM against seven cell lines. It is worth noting that, in addition to cytotoxic activity against tumor cells, the potential of some diterpenes as antitumor active molecules is reflected in their inhibitory activity against specific target proteins like Cbl-b, aromatase, etc. The production of cytotoxic metabolites by sponges may be beneficial for their self-protection, such as against predators and spatial competition [28].
Reports have documented the antibacterial, antifungal, antiviral, and antiparasitic activities of diterpenoids derived from various sources, such as sponges, fungi, and plants [111]. In our review of the literature on sponge-derived diterpenoids, we noticed that some molecules exhibit multiple activities. For instance, the compound (−)-agelamide D (43) exhibited cytotoxic activity against L5178Y mouse lymphoma cells and, in another activity evaluation model, demonstrated cytotoxic activity against Hep3B cells. It has also been proven to enhance the radiosensitivity of Hep3B cells. Furthermore, (−)-agelamide D also inhibits the biofilm formation of S. epidermidis. The compound iso-agelasine C (40) showed cytotoxicity against the HL-60, K562, and HCT-116 cell lines, exhibited antibacterial activity against Proteusbacillus vulgaris, and demonstrated antifungal activity against Candida albicans.
Some compounds have very low active concentrations. For example, monamphilectines B–C (217218) exhibited antiparasitic activity against P. falciparum, with IC50 values of 44.5 and 43.3 nM, respectively. Compound (−)-agelasine D (42) inhibited the growth of planktonic forms of the biofilm-forming bacterium Staphylococcus epidermidis, with an MIC < 0.0877 µM. The compound spongionellol A (213) showed cytotoxicity against seven cancer cell lines, with IC50 values ranging from 0.94 to 2.64 µM. Kalihinols O–T (1116) showed antifouling activity against Balanus amphitrite larvae, with EC50 values ranging from 0.53 to 1.48 µM.
The diversity in chemical structure and biological activity exhibited by these sponge-derived diterpenoids demonstrates their great potential in the development of marine drugs. However, the variety of activity evaluation models, while providing opportunities to discover different activities of these molecules, makes it challenging to compare the activities among different diterpenoids, and the structure–activity relationships are difficult to define. Activity concentration units such as µM, mM, and µg/mL further complicate the comparison of different studies. Moreover, there is no unified standard for defining the presence and strength of activity. Balancing standardization with diversity is a challenge. Additionally, most activity evaluations only characterize inhibition rates or IC50 values; further research to identify active targets and elucidate mechanisms of action will better facilitate the application and development of these molecules.

Author Contributions

Conceptualization, J.X., W.W. and Z.S.; formal analysis, J.X., X.C. and P.Q.; data curation, J.X.; writing—original draft preparation, J.X., X.C., G.L. and P.Q.; writing—review and editing, J.X., W.W. and Z.S.; visualization, X.C. and J.X.; supervision, J.X., W.W. and Z.S.; funding acquisition, J.X., G.L., W.W. and Z.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the National Key R&D Program of China (2022YFC2804100), the COMRA program (DY135-B2-01), the Scientific Research Foundation of the Third Institute of Oceanography, MNR (2022007, 2019021), the Xiamen Southern Oceanographic Center Project (22GYY007HJ07), and the Open Funding Project of the Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University (20220502, 20220501).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Van Soest, R.W.M.; Boury-Esnault, N.; Vacelet, J.; Dohrmann, M.; Erpenbeck, D.; De Voogd, N.J.; Santodomingo, N.; Vanhoorne, B.; Kelly, M.; Hooper, J.N.A. Global diversity of sponges (Porifera). PLoS ONE 2012, 7, e35105. [Google Scholar] [CrossRef] [PubMed]
  2. Carroll, A.R.; Copp, B.R.; Davis, R.A.; Keyzers, R.A.; Prinsep, M.R. Marine natural products. Nat. Prod. Rep. 2022, 39, 1122–1171. [Google Scholar] [CrossRef] [PubMed]
  3. Carroll, A.R.; Copp, B.R.; Davis, R.A.; Keyzers, R.A.; Prinsep, M.R. Marine natural products. Nat. Prod. Rep. 2023, 40, 275–325. [Google Scholar] [CrossRef] [PubMed]
  4. Carroll, A.R.; Copp, B.R.; Grkovic, T.; Keyzers, R.A.; Prinsep, M.R. Marine natural products. Nat. Prod. Rep. 2024, 41, 162–207. [Google Scholar] [CrossRef]
  5. Ivanchina, N.V.; Kalinin, V.I. Triterpene and steroid glycosides from marine sponges (Porifera, Demospongiae): Structures, taxonomical distribution, biological activities. Molecules 2023, 28, 2503. [Google Scholar] [CrossRef]
  6. Bian, C.; Wang, J.; Zhou, X.; Wu, W.; Guo, R. Recent advances on marine alkaloids from sponges. Chem. Biodivers. 2020, 17, e2000186. [Google Scholar] [CrossRef]
  7. Hong, L.-L.; Ding, Y.-F.; Zhang, W.; Lin, H.-W. Chemical and biological diversity of new natural products from marine sponges: A review (2009–2018). Mar. Life Sci. Technol. 2022, 4, 356–372. [Google Scholar] [CrossRef]
  8. Hanson, J.R. Diterpenoids. Nat. Prod. Rep. 1984, 1, 171–180. [Google Scholar] [CrossRef]
  9. Hanson, J.R. Diterpenoids. Nat. Prod. Rep. 1984, 1, 339–348. [Google Scholar] [CrossRef]
  10. Hanson, J.R. Diterpenoids. Nat. Prod. Rep. 1984, 1, 533–544. [Google Scholar] [CrossRef]
  11. Hanson, J.R. Diterpenoids. Nat. Prod. Rep. 2009, 26, 1156–1171. [Google Scholar] [CrossRef] [PubMed]
  12. Hanson, J.R. Diterpenoids of terrestrial origin. Nat. Prod. Rep. 2011, 28, 1755–1772. [Google Scholar] [CrossRef] [PubMed]
  13. Qiu, P.; Xia, J.; Zhang, H.; Lin, D.; Shao, Z. A review of diterpenes from marine-derived fungi: 2009–2021. Molecules 2022, 27, 8303. [Google Scholar] [CrossRef] [PubMed]
  14. Mehbub, M.F.; Yang, Q.; Cheng, Y.; Franco, C.M.M.; Zhang, W. Marine sponge-derived natural products: Trends and opportunities for the decade of 2011–2020. Front. Mar. Sci. 2024, 11, 1462825. [Google Scholar] [CrossRef]
  15. Mehbub, M.F.; Perkins, M.V.; Zhang, W.; Franco, C.M.M. New marine natural products from sponges (Porifera) of the order Dictyoceratida (2001 to 2012); a promising source for drug discovery, exploration and future prospects. Biotechnol. Adv. 2016, 34, 473–491. [Google Scholar] [CrossRef]
  16. Elgoud Said, A.A.; Mahmoud, B.K.; Attia, E.Z.; Abdelmohsen, U.R.; Fouad, M.A. Bioactive natural products from marine sponges belonging to family Hymedesmiidae. RSC Adv. 2021, 11, 16179–16191. [Google Scholar] [CrossRef]
  17. Li, F.; Kelly, M.; Tasdemir, D. Chemistry, chemotaxonomy and biological activity of the Latrunculid sponges (order Poecilosclerida, family Latrunculiidae). Mar. Drugs 2021, 19, 27. [Google Scholar] [CrossRef]
  18. Shady, N.H.; Fouad, M.A.; Kamel, M.S.; Schirmeister, T.; Abdelmohsen, U.R. Natural product repertoire of the genus Amphimedon. Mar. Drugs 2019, 17, 19. [Google Scholar] [CrossRef]
  19. He, Q.; Miao, S.; Ni, N.; Man, Y.; Gong, K. A review of the secondary metabolites from the marine sponges of the genus Aaptos. Nat. Prod. Commun. 2020, 15, 1934578X2095143. [Google Scholar] [CrossRef]
  20. de Sousa, L.H.N.; de Araujo, R.D.; Sousa-Fontoura, D.; Menezes, F.G.; Araujo, R.M. Metabolities from marine sponges of the genus Callyspongia: Occurrence, biological activity, and NMR data. Mar. Drugs 2021, 19, 663. [Google Scholar] [CrossRef]
  21. Zhu, J.; Liu, Y.; Liu, Z.; Wang, H.; Zhang, H. Bioactive nitrogenous secondary metabolites from the marine sponge genus Haliclona. Mar. Drugs 2019, 17, 682. [Google Scholar] [CrossRef] [PubMed]
  22. Lee, Y.-J.; Cho, Y.; Huynh Nguyen Khanh, T. Secondary metabolites from the marine sponges of the genus Petrosia: A literature review of 43 years of research. Mar. Drugs 2021, 19, 122. [Google Scholar] [CrossRef] [PubMed]
  23. Caso, A.; da Silva, F.B.; Esposito, G.; Teta, R.; Sala, G.D.; Cavalcanti, L.P.A.N.; Valverde, A.L.; Martins, R.C.C.; Costantino, V. Exploring chemical diversity of Phorbas sponges as a source of novel lead compounds in drug discovery. Mar. Drugs 2021, 19, 667. [Google Scholar] [CrossRef]
  24. Bai, X.; Liu, Y.; Wang, H.; Zhang, H. Natural products from the marine sponge subgenus Reniera. Molecules 2021, 26, 1097. [Google Scholar] [CrossRef]
  25. Wu, Q.; Nay, B.; Yang, M.; Ni, Y.; Wang, H.; Yao, L.; Li, X. Marine sponges of the genus Stelletta as promising drug sources: Chemical and biological aspects. Acta Pharm. Sin. B 2019, 9, 237–257. [Google Scholar] [CrossRef]
  26. El-Demerdash, A.; Atanasov, A.G.; Horbanczuk, O.K.; Tammam, M.A.; Abdel-Mogib, M.; Hooper, J.N.A.; Sekeroglu, N.; Al-Mourabit, A.; Kijjoa, A. Chemical diversity and biological activities of marine sponges of the genus Suberea: A systematic review. Mar. Drugs 2019, 17, 115. [Google Scholar] [CrossRef]
  27. Campos Aguiar, A.C.; Parisi, J.R.; Granito, R.N.; Freitas de Sousa, L.R.; Muniz Renno, A.C.; Gazarini, M.L. Metabolites from marine sponges and their potential to treat malarial protozoan parasites infection: A systematic review. Mar. Drugs 2021, 19, 134. [Google Scholar] [CrossRef]
  28. Khan, S.; Al-Fadhli, A.A.; Tilvi, S. Discovery of cytotoxic natural products from Red Sea sponges: Structure and synthesis. Eur. J. Med. Chem. 2021, 220, 113491. [Google Scholar] [CrossRef] [PubMed]
  29. Ramanjooloo, A.; Andersen, R.J.; Bhaw-Luximon, A. Marine sponge-derived/inspired drugs and their applications in drug delivery systems. Futur. Med. Chem. 2021, 13, 487–504. [Google Scholar] [CrossRef]
  30. Xu, Y.; Lang, J.H.; Jiao, W.H.; Wang, R.P.; Peng, Y.; Song, S.J.; Zhang, B.H.; Lin, H.W. Formamido-diterpenes from the South China Sea sponge Acanthella cavernosa. Mar. Drugs 2012, 10, 1445–1458. [Google Scholar] [CrossRef]
  31. Wu, Q.; Chen, W.T.; Li, S.W.; Ye, J.Y.; Huan, X.J.; Gavagnin, M.; Yao, L.G.; Wang, H.; Miao, Z.H.; Li, X.W.; et al. Cytotoxic nitrogenous terpenoids from two South China Sea nudibranchs Phyllidiella pustulosa, Phyllidia coelestis, and their sponge-prey Acanthella cavernosa. Mar. Drugs 2019, 17, 56. [Google Scholar] [CrossRef] [PubMed]
  32. Xu, Y.; Li, N.; Jiao, W.-H.; Wang, R.-P.; Peng, Y.; Qi, S.-H.; Song, S.-J.; Chen, W.-S.; Lin, H.-W. Antifouling and cytotoxic constituents from the South China Sea sponge Acanthella cavernosa. Tetrahedron 2012, 68, 2876–2883. [Google Scholar] [CrossRef]
  33. Sun, J.-Z.; Chen, K.-S.; Yao, L.-G.; Liu, H.-L.; Guo, Y.-W. A new kalihinol diterpene from the Hainan sponge Acanthella sp. Arch. Pharmacal Res. 2009, 32, 1581–1584. [Google Scholar] [CrossRef]
  34. Pettit, G.R.; Tang, Y.; Zhang, Q.; Bourne, G.T.; Arm, C.A.; Leet, J.E.; Knight, J.C.; Pettit, R.K.; Chapuis, J.C.; Doubek, D.L.; et al. Isolation and structures of axistatins 1–3 from the Republic of Palau marine sponge Agelas axifera Hentschel. J. Nat. Prod. 2013, 76, 420–424. [Google Scholar] [CrossRef]
  35. Li, T.; Wang, B.; de Voogd, N.J.; Tang, X.-L.; Wang, Q.; Chu, M.-J.; Li, P.-L.; Li, G.-Q. Two new diterpene alkaloids from the South China Sea Sponge Agelas aff. nemoechinata. Chin. Chem. Lett. 2016, 27, 1048–1051. [Google Scholar] [CrossRef]
  36. An, L.; Song, W.; Tang, X.; de Voogd, N.J.; Wang, Q.; Chu, M.; Li, P.; Li, G. Alkaloids and polyketides from the South China Sea sponge Agelas aff. nemoechinata. RSC Adv. 2017, 7, 14323–14329. [Google Scholar] [CrossRef]
  37. Hong, L.-L.; Sun, J.-B.; Yang, F.; Liu, M.; Tang, J.; Sun, F.; Jiao, W.-H.; Wang, S.-P.; Zhang, W.; Lin, H.-W. New diterpene alkaloids from the marine sponge Agelas mauritiana. RSC Adv. 2017, 7, 23970–23976. [Google Scholar] [CrossRef]
  38. Chu, M.J.; Tang, X.L.; Qin, G.F.; Sun, Y.T.; Li, L.; de Voogd, N.J.; Li, P.L.; Li, G.Q. Pyrrole derivatives and diterpene alkaloids from the South China Sea Sponge Agelas nakamurai. Chem. Biodivers. 2017, 14, e1600446. [Google Scholar] [CrossRef]
  39. Hertiani, T.; Edrada-Ebel, R.; Ortlepp, S.; van Soest, R.W.M.; de Voogd, N.J.; Wray, V.; Hentschel, U.; Kozytska, S.; Mueller, W.E.G.; Proksch, P. From anti-fouling to biofilm inhibition: New cytotoxic secondary metabolites from two Indonesian Agelas sponges. Bioorg. Med. Chem. 2010, 18, 1297–1311. [Google Scholar] [CrossRef]
  40. Paulsen, B.; Fredriksen, K.A.; Petersen, D.; Maes, L.; Matheeussen, A.; Naemi, A.O.; Scheie, A.A.; Simm, R.; Ma, R.; Wan, B.J.; et al. Synthesis and antimicrobial activities of N6-hydroxyagelasine analogs and revision of the structure of ageloximes. Bioorg. Med. Chem. 2019, 27, 620–629. [Google Scholar] [CrossRef]
  41. Choi, C.; Cho, Y.; Son, A.; Shin, S.W.; Lee, Y.J.; Park, H.C. Therapeutic potential of (−)-agelamide D, a diterpene alkaloid from the marine sponge Agelas sp., as a natural radiosensitizer in hepatocellular carcinoma models. Mar. Drugs 2020, 18, 500. [Google Scholar] [CrossRef] [PubMed]
  42. Agena, M.; Tanaka, C.; Hanif, N.; Yasumoto-Hirose, M.; Tanaka, J. New cytotoxic spongian diterpenes from the sponge Dysideai cf. arenaria. Tetrahedron 2009, 65, 1495–1499. [Google Scholar] [CrossRef]
  43. Shingaki, M.; Wauke, T.; Ahmadi, P.; Tanaka, J. Four cytotoxic spongian diterpenes from the sponge Dysidea cf. arenaria. Chem. Pharm. Bull. 2016, 64, 272–275. [Google Scholar] [CrossRef] [PubMed]
  44. Tian, Y.-Q.; Gu, B.-B.; Jiao, W.-H.; Lin, H.-W. Four homoverrucosane-type diterpenes from the marine sponge Halichondria sp. Tetrahedron 2020, 76, 131697. [Google Scholar] [CrossRef]
  45. Trianto, A.; Hermawan, I.; de Voogd, N.J.; Tanaka, J. Halioxepine, a new meroditerpene from an Indonesian sponge Haliclona sp. Chem. Pharm. Bull. 2011, 59, 1311–1313. [Google Scholar] [CrossRef] [PubMed]
  46. Dattelbaum, J.D.; Singh, A.J.; Field, J.J.; Miller, J.H.; Northcote, P.T. The nitrogenous hamigerans: Unusual amino acid-derivatized aromatic diterpenoid metabolites from the New Zealand marine sponge Hamigera tarangaensis. J. Org. Chem. 2015, 80, 304–312. [Google Scholar] [CrossRef]
  47. Ahmadi, P.; Haruyama, T.; Kobayashi, N.; de Voogd, N.J.; Tanaka, J. Spongian diterpenes from the sponge Hyattella aff. intestinalis. Chem. Pharm. Bull. 2017, 65, 874–877. [Google Scholar] [CrossRef]
  48. Luo, X.-C.; Wang, Q.; Tang, X.-L.; Li, P.-L.; Li, G.-Q. One cytotoxic steroid and other two new metabolites from the South China Sea sponge Luffariella variabilis. Tetrahedron Lett. 2021, 65, 152762. [Google Scholar] [CrossRef]
  49. Lamoral-Theys, D.; Fattorusso, E.; Mangoni, A.; Perinu, C.; Kiss, R.; Costantino, V. Evaluation of the antiproliferative activity of diterpene isonitriles from the sponge Pseudoaxinella flava in apoptosis-sensitive and apoptosis-resistant cancer cell lines. J. Nat. Prod. 2011, 74, 2299–2303. [Google Scholar] [CrossRef]
  50. Pech-Puch, D.; Rodríguez, J.; Cautain, B.; Sandoval-Castro, C.A.; Jimenez, C. Cytotoxic furanoditerpenes from the sponge Spongia tubulifera collected in the Mexican Caribbean. Mar. Drugs 2019, 17, 416. [Google Scholar] [CrossRef]
  51. Liang, Y.-Q.; Liao, X.-J.; Zhao, B.-X.; Xu, S.-H. Novel 3,4-seco-3,19-dinorspongian and 5,17-epoxy-19-norspongian diterpenes from the marine sponge Spongia sp. Org. Chem. Front. 2020, 7, 3253–3261. [Google Scholar] [CrossRef]
  52. Jin, T.; Li, P.; Wang, C.; Tang, X.; Yv, X.; Li, K.; Luo, L.; Ou, H.; Li, G. Two new spongian diterpene derivatives from the aquaculture sponge Spongia officinalis Linnaeus, 1759. Nat. Prod. Res. 2021, 37, 216–226. [Google Scholar] [CrossRef] [PubMed]
  53. Jomori, T.; Setiawan, A.; Sasaoka, M.; Arai, M. Cytotoxicity of new diterpene alkaloids, ceylonamides G-I, isolated from Indonesian marine sponge of Spongia sp. Nat. Prod. Commun. 2019, 14, 1934578X1985729. [Google Scholar] [CrossRef]
  54. Rateb, M.E.; Houssen, W.E.; Schumacher, M.; Harrison, W.T.; Diederich, M.; Ebel, R.; Jaspars, M. Bioactive diterpene derivatives from the marine sponge Spongionella sp. J. Nat. Prod. 2009, 72, 1471–1476. [Google Scholar] [CrossRef] [PubMed]
  55. Dyshlovoy, S.A.; Shubina, L.K.; Makarieva, T.N.; Hauschild, J.; Strewinsky, N.; Guzii, A.G.; Menshov, A.S.; Popov, R.S.; Grebnev, B.B.; Busenbender, T.; et al. New diterpenes from the marine sponge Spongionella sp. overcome drug resistance in prostate cancer by inhibition of P-glycoprotein. Sci. Rep. 2022, 12, 13570. [Google Scholar] [CrossRef]
  56. Barber, J.M.; Leahy, D.C.; Miller, J.H.; Northcote, P.T. Luakuliides A–C, cytotoxic labdane diterpenes from a Tongan dictyoceratid sponge. Tetrahedron Lett. 2015, 56, 6314–6318. [Google Scholar] [CrossRef]
  57. Uddin, M.H.; Hossain, M.K.; Nigar, M.; Roy, M.C.; Tanaka, J. New cytotoxic spongian-class rearranged diterpenes from a marine sponge. Chem. Nat. Compd. 2012, 48, 412–415. [Google Scholar] [CrossRef]
  58. Abdjul, D.B.; Yamazaki, H.; Kanno, S.; Takahashi, O.; Kirikoshi, R.; Ukai, K.; Namikoshi, M. Structures and biological evaluations of agelasines isolated from the Okinawan marine sponge Agelas nakamurai. J. Nat. Prod. 2015, 78, 1428–1433. [Google Scholar] [CrossRef]
  59. Pech-Puch, D.; Forero, A.M.; Carlos Fuentes-Monteverde, J.; Lasarte-Monterrubio, C.; Martinez-Guitian, M.; Gonzalez-Salas, C.; Guillen-Hernandez, S.; Villegas-Hernandez, H.; Beceiro, A.; Griesinger, C.; et al. Antimicrobial diterpene alkaloids from an Agelas citrina sponge collected in the Yucatán Peninsula. Mar. Drugs 2022, 20, 298. [Google Scholar] [CrossRef]
  60. Kubota, T.; Iwai, T.; Takahashi-Nakaguchi, A.; Fromont, J.; Gonoi, T.; Kobayashi, J.I. Agelasines O–U, new diterpene alkaloids with a 9-N-methyladenine unit from a marine sponge Agelas sp. Tetrahedron 2012, 68, 9738–9744. [Google Scholar] [CrossRef]
  61. Yu, H.-B.; Gu, B.-B.; Wang, S.-P.; Cheng, C.-W.; Yang, F.; Lin, H.-W. New diterpenoids from the marine sponge Dactylospongia elegans. Tetrahedron 2017, 73, 6657–6661. [Google Scholar] [CrossRef]
  62. Bory, A.; Shilling, A.J.; Allen, J.; Azhari, A.; Roth, A.; Shaw, L.N.; Kyle, D.E.; Adams, J.H.; Amsler, C.D.; McClintock, J.B.; et al. Bioactivity of spongian diterpenoid scaffolds from the Antarctic sponge Dendrilla antarctica. Mar. Drugs 2020, 18, 327. [Google Scholar] [CrossRef] [PubMed]
  63. von Salm, J.L.; Witowski, C.G.; Fleeman, R.M.; McClintock, J.B.; Amsler, C.D.; Shaw, L.N.; Baker, B.J. Darwinolide, a new diterpene scaffold that inhibits methicillin-resistant Staphylococcus aureus biofilm from the Antarctic sponge Dendrilla membranosa. Org. Lett. 2016, 18, 2596–2599. [Google Scholar] [CrossRef]
  64. Avilés, E.; Rodríguez, A.D. Monamphilectine A, a potent antimalarial β-lactam from marine sponge Hymeniacidon sp.: Isolation, structure, semisynthesis, and bioactivity. Org. Lett. 2010, 12, 5290–5293. [Google Scholar] [CrossRef] [PubMed]
  65. Tai, C.-J.; Ahmed, A.F.; Chao, C.-H.; Yen, C.-H.; Hwang, T.-L.; Chang, F.-R.; Huang, Y.M.; Sheu, J.-H. Spongenolactones A–C, bioactive 5,5,6,6,5-pentacyclic spongian diterpenes from the Red Sea sponge Spongia sp. Mar. Drugs 2022, 20, 498. [Google Scholar] [CrossRef]
  66. de Oliveira, J.A.M.; Williams, D.E.; Bonnett, S.; Johnson, J.; Parish, T.; Andersen, R.J. Diterpenoids isolated from the Samoan marine sponge Chelonaplysilla sp. inhibit Mycobacterium tuberculosis growth. J. Antibiot. 2020, 73, 568–573. [Google Scholar] [CrossRef] [PubMed]
  67. Avilés, E.; Rodríguez, A.D.; Vicente, J. Two rare-class tricyclic diterpenes with antitubercular activity from the Caribbean sponge Svenzea flava. Application of vibrational circular dichroism spectroscopy for determining absolute configuration. J. Org. Chem. 2013, 78, 11294–11301. [Google Scholar] [CrossRef]
  68. Wattanapiromsakul, C.; Chanthathamrongsiri, N.; Bussarawit, S.; Yuenyongsawad, S.; Plubrukarn, A.; Suwanborirux, K. 8-Isocyanoamphilecta-11(20),15-diene, a new antimalarial isonitrile diterpene from the sponge Ciocalapata sp. Can. J. Chem. 2009, 87, 612–618. [Google Scholar] [CrossRef]
  69. Wright, A.D.; Lang-Unnasch, N. Diterpene formamides from the tropical marine sponge Cymbastela hooperi and their antimalarial activity in vitro. J. Nat. Prod. 2009, 72, 492–495. [Google Scholar] [CrossRef]
  70. Shilling, A.J.; Witowski, C.G.; Maschek, J.A.; Azhari, A.; Vesely, B.A.; Kyle, D.E.; Amsler, C.D.; McClintock, J.B.; Baker, B.J. Spongian diterpenoids derived from the Antarctic sponge Dendrilla antarcticai are potent inhibitors of the Leishmania Parasite. J. Nat. Prod. 2020, 83, 1553–1562. [Google Scholar] [CrossRef]
  71. Yang, F.; Zou, Y.; Wang, R.P.; Hamann, M.T.; Zhang, H.J.; Jiao, W.H.; Han, B.N.; Song, S.J.; Lin, H.W. Relative and absolute stereochemistry of diacarperoxides: Antimalarial norditerpene endoperoxides from marine sponge Diacarnus megaspinorhabdosa. Mar. Drugs 2014, 12, 4399–4416. [Google Scholar] [CrossRef] [PubMed]
  72. Avilés, E.; Prudhomme, J.; Le Roch, K.G.; Rodríguez, A.D. Structures, semisyntheses, and absolute configurations of the antiplasmodial alpha-substituted beta-lactam monamphilectines B and C from the sponge Svenzea flava. Tetrahedron 2015, 71, 487–494. [Google Scholar] [CrossRef]
  73. Hong, L.L.; Yu, H.B.; Wang, J.; Jiao, W.H.; Cheng, B.H.; Yang, F.; Zhou, Y.J.; Gu, B.B.; Song, S.J.; Lin, H.W. Unusual anti-allergic diterpenoids from the marine sponge Hippospongia lachne. Sci. Rep. 2017, 7, 43138. [Google Scholar] [CrossRef]
  74. Chakraborty, K.; Francis, P. Erectcyanthins A–C from marine sponge Hyrtios erectus: Anti-dyslipidemic agents attenuate hydroxymethylglutaryl coenzyme A reductase. Nat. Prod. Res. 2021, 36, 5676–5687. [Google Scholar] [CrossRef]
  75. Chen, Q.; Mao, Q.; Bao, M.; Mou, Y.; Fang, C.; Zhao, M.; Jiang, W.; Yu, X.; Wang, C.; Dai, L.; et al. Spongian diterpenes including one with a rearranged skeleton from the marine sponge Spongia officinalis. J. Nat. Prod. 2019, 82, 1714–1718. [Google Scholar] [CrossRef] [PubMed]
  76. Tai, C.J.; Huang, C.Y.; Ahmed, A.F.; Orfali, R.S.; Alarif, W.M.; Huang, Y.M.; Wang, Y.H.; Hwang, T.L.; Sheu, J.H. An anti-inflammatory 2,4-cyclized-3,4-secospongian diterpenoid and furanoterpene-related metabolites of a marine sponge Spongia sp. from the Red Sea. Mar. Drugs 2021, 19, 38. [Google Scholar] [CrossRef]
  77. Costantino, V.; Fattorusso, E.; Mangoni, A.; Perinu, C.; Cirino, G.; De Gruttola, L.; Roviezzo, F. Tedanol: A potent anti-inflammatory ent-pimarane diterpene from the Caribbean Sponge Tedania ignis. Bioorg. Med. Chem. 2009, 17, 7542–7547. [Google Scholar] [CrossRef] [PubMed]
  78. Prieto, I.M.; Paola, A.; Perez, M.; Garcia, M.; Blustein, G.; Schejter, L.; Palermo, J.A. Antifouling diterpenoids from the sponge Dendrilla antarctica. Chem. Biodivers. 2022, 19, e202100618. [Google Scholar] [CrossRef]
  79. Fang, S.T.; Yan, B.F.; Yang, C.Y.; Miao, F.P.; Ji, N.Y. Hymerhabdrin A, a novel diterpenoid with antifouling activity from the intertidal sponge Hymerhabdia sp. J. Antibiot. 2017, 70, 1043–1046. [Google Scholar] [CrossRef]
  80. El-Desoky, A.H.; Kato, H.; Angkouw, E.D.; Mangindaan, R.E.; de Voogd, N.J.; Tsukamoto, S. Ceylonamides A-F, nitrogenous spongian diterpenes that inhibit RANKL-induced osteoclastogenesis, from the marine sponge Spongia ceylonensis. J. Nat. Prod. 2016, 79, 1922–1928. [Google Scholar] [CrossRef]
  81. El-Desoky, A.H.; Kato, H.; Kagiyama, I.; Hitora, Y.; Losung, F.; Mangindaan, R.E.; de Voogd, N.J.; Tsukamoto, S. Ceylonins A-F, spongian diterpene derivatives that inhibit RANKL-induced formation of multinuclear osteoclasts, from the marine sponge Spongia ceylonensis. J. Nat. Prod. 2017, 80, 90–95. [Google Scholar] [CrossRef] [PubMed]
  82. Jiang, W.; Wang, D.; Wilson, B.A.P.; Kang, U.; Bokesch, H.R.; Smith, E.A.; Wamiru, A.; Goncharova, E.I.; Voeller, D.; Lipkowitz, S.; et al. Agelasine diterpenoids and Cbl-b inhibitory ageliferins from the coralline demosponge Astrosclera willeyana. Mar. Drugs 2021, 19, 361. [Google Scholar] [CrossRef] [PubMed]
  83. Kato, H.; Nehira, T.; Matsuo, K.; Kawabata, T.; Kobashigawa, Y.; Morioka, H.; Losung, F.; Mangindaan, R.E.P.; de Voogd, N.J.; Yokosawa, H.; et al. Niphateolide A: Isolation from the marine sponge Niphates olemda and determination of its absolute configuration by an ECD analysis. Tetrahedron 2015, 71, 6956–6960. [Google Scholar] [CrossRef]
  84. Lhullier, C.; de Oliveira Tabalipa, E.; Nienkotter Sarda, F.; Sandjo, L.P.; Zanchett Schneider, N.F.; Carraro, J.L.; Oliveira Simoes, C.M.; Schenkel, E.P. Clerodane diterpenes from the marine sponge Raspailia bouryesnaultae collected in South Brazil. Mar. Drugs 2019, 17, 57. [Google Scholar] [CrossRef]
  85. Parrish, S.M.; Yoshida, W.Y.; Kondratyuk, T.P.; Park, E.J.; Pezzuto, J.M.; Kelly, M.; Williams, P.G. Spongiapyridine and related spongians isolated from an Indonesian Spongia sp. J. Nat. Prod. 2014, 77, 1644–1649. [Google Scholar] [CrossRef]
  86. Leiros, M.; Sanchez, J.A.; Alonso, E.; Rateb, M.E.; Houssen, W.E.; Ebel, R.; Jaspars, M.; Alfonso, A.; Botana, L.M. Spongionella secondary metabolites protect mitochondrial function in cortical neurons against oxidative stress. Mar. Drugs 2014, 12, 700–718. [Google Scholar] [CrossRef] [PubMed]
  87. Lee, J.S.; Abdjul, D.B.; Yamazaki, H.; Takahashi, O.; Kirikoshi, R.; Ukai, K.; Namikoshi, M. Strongylophorines, new protein tyrosine phosphatase 1B inhibitors, from the marine sponge Strongylophora strongilata collected at Iriomote Island. Bioorg. Med. Chem. Lett. 2015, 25, 3900–3902. [Google Scholar] [CrossRef]
  88. Costa, M.; Fernandez, R.; Perez, M.; Thorsteinsdottir, M. Two new spongian diterpene analogues isolated from the marine sponge Acanthodendrilla sp. Nat. Prod. Res. 2020, 34, 1053–1060. [Google Scholar] [CrossRef]
  89. Chu, M.J.; Li, M.; Ma, H.; Li, P.L.; Li, G.Q. Secondary metabolites from marine sponges of the genus Agelas: A comprehensive update insight on structural diversity and bioactivity. RSC Adv. 2022, 12, 7789–7820. [Google Scholar] [CrossRef]
  90. Zhang, H.; Dong, M.; Chen, J.; Wang, H.; Tenney, K.; Crews, P. Bioactive secondary metabolites from the marine sponge genus Agelas. Mar. Drugs 2017, 15, 351. [Google Scholar] [CrossRef]
  91. Pejin, B.; Glumac, M. A brief review of potent anti-CNS tumourics from marine sponges: Covering the period from 1994 to 2014. Nat. Prod. Res. 2018, 32, 375–384. [Google Scholar] [CrossRef] [PubMed]
  92. Thawabteh, A.M.; Thawabteh, A.; Lelario, F.; Bufo, S.A.; Scrano, L. Classification, toxicity and bioactivity of natural diterpenoid alkaloids. Molecules 2021, 26, 4103. [Google Scholar] [CrossRef]
  93. Stout, E.P.; Yu, L.C.; Molinski, T.F. Antifungal diterpene alkaloids from the Caribbean sponge Agelas citrina: Unified configurational assignments of agelasidines and agelasines. Eur. J. Org. Chem. 2012, 2012, 5131–5135. [Google Scholar] [CrossRef] [PubMed]
  94. Lee, S.; Tanaka, N.; Kobayashi, J.; Kashiwada, Y. Agelamasines A and B, diterpene alkaloids from an Okinawan marine sponge Agelas sp. J. Nat. Med. 2018, 72, 364–368. [Google Scholar] [CrossRef]
  95. Zhang, X.; Li, P.L.; Qin, G.F.; Li, S.; de Voogd, N.J.; Tang, X.L.; Li, G.Q. Isolation and absolute configurations of diversiform C17, C21 and C25 terpenoids from the marine sponge Cacospongia sp. Mar. Drugs 2018, 17, 14. [Google Scholar] [CrossRef] [PubMed]
  96. Wojnar, J.M.; Dowle, K.O.; Northcote, P.T. The Oxeatamides: Nitrogenous spongian diterpenes from the New Zealand marine sponge Darwinella oxeata. J. Nat. Prod. 2014, 77, 2288–2295. [Google Scholar] [CrossRef]
  97. MC, A.R.; Williams, D.E.; Gubiani, J.R.; Parra, L.L.; Santos, M.F.; Ferreira, D.D.; Mesquita, J.T.; Tempone, A.G.; Ferreira, A.G.; Padula, V.; et al. Rearranged terpenoids from the marine sponge Darwinella cf. oxeata and its predator, the nudibranch Felimida grahami. J. Nat. Prod. 2017, 80, 720–725. [Google Scholar] [CrossRef]
  98. Hayton, J.B.; Grant, G.D.; Carroll, A.R. Three new spongian diterpenes from the marine sponge Dendrilla rosea. Aust. J. Chem. 2019, 72, 964–968. [Google Scholar] [CrossRef]
  99. Katavic, P.L.; Jumaryatno, P.; Hooper, J.N.A.; Blanchfield, J.T.; Garson, M.J. Oxygenated terpenoids from the Australian sponges Coscinoderma matthewsi and Dysidea sp., and the nudibranch Chromodoris albopunctata. Aust. J. Chem. 2012, 65, 531–538. [Google Scholar] [CrossRef]
  100. Qin, G.F.; Tang, X.L.; de Voogd, N.J.; Li, P.L.; Li, G.Q. Cytotoxic components from the Xisha sponge Fascaplysinopsis reticulata. Nat. Prod. Res. 2020, 34, 790–796. [Google Scholar] [CrossRef]
  101. Woolly, E.F.; Singh, A.J.; Russell, E.R.; Miller, J.H.; Northcote, P.T. Hamigerans R and S: Nitrogenous diterpenoids from the New Zealand marine sponge Hamigera tarangaensis. J. Nat. Prod. 2018, 81, 387–393. [Google Scholar] [CrossRef] [PubMed]
  102. Gross, H.; Wright, A.D.; Reinscheid, U.; König, G.M. Three new spongian diterpenes from the Fijian marine sponge Spongia sp. Nat. Prod. Commun. 2009, 4, 315–322. [Google Scholar] [CrossRef] [PubMed]
  103. El-Desoky, A.H.; Kato, H.; Tsukamoto, S. Ceylonins G-I: Spongian diterpenes from the marine sponge Spongia ceylonensis. J. Nat. Med. 2017, 71, 765–769. [Google Scholar] [CrossRef] [PubMed]
  104. Han, G.Y.; Sun, D.Y.; Liang, L.F.; Yao, L.G.; Chen, K.X.; Guo, Y.W. Spongian diterpenes from Chinese marine sponge Spongia officinalis. Fitoterapia 2018, 127, 159–165. [Google Scholar] [CrossRef]
  105. Liang, Y.-Q.; Liao, X.-J.; Lin, J.-L.; Xu, W.; Chen, G.-D.; Zhao, B.-X.; Xu, S.-H. Spongiains A–C: Three new spongian diterpenes with ring A rearrangement from the marine sponge Spongia sp. Tetrahedron 2019, 75, 3802–3808. [Google Scholar] [CrossRef]
  106. Jin, T.; Li, P.; Wang, C.; Tang, X.; Yu, X.; Sun, F.; Luo, L.; Ou, H.; Li, G. Jellynolide A, pokepola esters, and sponalisolides from the aquaculture sponge Spongia officinalis L. Phytochemistry 2022, 194, 113006. [Google Scholar] [CrossRef]
  107. Liang, Y.; Liao, X.; Ling, L.; Yang, Y.; Zhao, B.; Xu, S. A new dinorspongian diterpene with pyridyl D-ring from the marine sponge Spongia sp. Chin. J. Org. Chem. 2022, 42, 901–904. [Google Scholar] [CrossRef]
  108. Chanthathamrongsiri, N.; Yuenyongsawad, S.; Wattanapiromsakul, C.; Plubrukarn, A. Bifunctionalized amphilectane diterpenes from the sponge Stylissa cf. massa. J. Nat. Prod. 2012, 75, 789–792. [Google Scholar] [CrossRef]
  109. Ota, K.; Hamamoto, Y.; Eda, W.; Tamura, K.; Sawada, A.; Hoshino, A.; Mitome, H.; Kamaike, K.; Miyaoka, H. Amitorines A and B, nitrogenous diterpene metabolites of Theonella swinhoei: Isolation, structure elucidation, and asymmetric synthesis. J. Nat. Prod. 2016, 79, 996–1004. [Google Scholar] [CrossRef]
  110. Taufa, T.; Subramani, R.; Northcote, P.T.; Keyzers, R.A. Natural products from Tongan marine organisms. Molecules 2021, 26, 4534. [Google Scholar] [CrossRef]
  111. Saha, P.; Rahman, F.I.; Hussain, F.; Rahman, S.M.A.; Rahman, M.M. Antimicrobial diterpenes: Recent development from natural sources. Front. Pharmacol. 2022, 12, 820312. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Number of new diterpenoids isolated from each genus of sponges from 2009 to 2022. (A) Overview of diterpenoid distribution across all genera, with those having fewer than 10 compounds grouped. (B) Close-up of genera with fewer than 10 diterpenoids each.
Figure 1. Number of new diterpenoids isolated from each genus of sponges from 2009 to 2022. (A) Overview of diterpenoid distribution across all genera, with those having fewer than 10 compounds grouped. (B) Close-up of genera with fewer than 10 diterpenoids each.
Marinedrugs 22 00447 g001
Figure 2. Bioactivity distribution of sponge-derived diterpenoids from 2009 to 2022.
Figure 2. Bioactivity distribution of sponge-derived diterpenoids from 2009 to 2022.
Marinedrugs 22 00447 g002
Figure 3. Chemical structures of diterpenes from Acanthella sponges (117).
Figure 3. Chemical structures of diterpenes from Acanthella sponges (117).
Marinedrugs 22 00447 g003
Figure 4. Chemical structures of diterpenes from Acanthodendrilla (1819) and Agelas (2028) sponges.
Figure 4. Chemical structures of diterpenes from Acanthodendrilla (1819) and Agelas (2028) sponges.
Marinedrugs 22 00447 g004
Figure 5. Chemical structures of diterpenes from Agelas sponges (2941).
Figure 5. Chemical structures of diterpenes from Agelas sponges (2941).
Marinedrugs 22 00447 g005
Figure 6. Chemical structures of diterpenes from Agelas sponges (4252).
Figure 6. Chemical structures of diterpenes from Agelas sponges (4252).
Marinedrugs 22 00447 g006
Figure 7. Chemical structures of diterpenes from Astrosclera (5355), Cacospongia (56), Chelonaplysilla (5759), Ciocalapata (60), and Cymbastela (6165) sponges.
Figure 7. Chemical structures of diterpenes from Astrosclera (5355), Cacospongia (56), Chelonaplysilla (5759), Ciocalapata (60), and Cymbastela (6165) sponges.
Marinedrugs 22 00447 g007
Figure 8. Chemical structures of diterpenes from Dactylospongia elegans (66, 67) and Darwinella (6880) sponges.
Figure 8. Chemical structures of diterpenes from Dactylospongia elegans (66, 67) and Darwinella (6880) sponges.
Marinedrugs 22 00447 g008
Figure 9. Chemical structures of diterpenes from Dendrilla sponges (8191).
Figure 9. Chemical structures of diterpenes from Dendrilla sponges (8191).
Marinedrugs 22 00447 g009
Figure 10. Chemical structures of diterpenes from the sponge Diacarnus megaspinorhabdosa (9297).
Figure 10. Chemical structures of diterpenes from the sponge Diacarnus megaspinorhabdosa (9297).
Marinedrugs 22 00447 g010
Figure 11. Chemical structures of diterpenes from Dysidea (98110), Fascaplysinopsis (111), and Halichondria (112115) sponges.
Figure 11. Chemical structures of diterpenes from Dysidea (98110), Fascaplysinopsis (111), and Halichondria (112115) sponges.
Marinedrugs 22 00447 g011
Figure 12. Chemical structures of diterpenes from Haliclona (116) and Hamigera (117132) sponges.
Figure 12. Chemical structures of diterpenes from Haliclona (116) and Hamigera (117132) sponges.
Marinedrugs 22 00447 g012
Figure 13. Chemical structures of diterpenes from Hippospongia (133134), Hyattella (135137), Hymeniacidon (138), Hymerhabdia (139), Hyrtios (140142), Luffariella (143), Niphates (144), Pseudoaxinella (145), and Raspailia (146) sponges.
Figure 13. Chemical structures of diterpenes from Hippospongia (133134), Hyattella (135137), Hymeniacidon (138), Hymerhabdia (139), Hyrtios (140142), Luffariella (143), Niphates (144), Pseudoaxinella (145), and Raspailia (146) sponges.
Marinedrugs 22 00447 g013
Figure 14. Chemical structures of diterpenes from Spongia sponges (147165).
Figure 14. Chemical structures of diterpenes from Spongia sponges (147165).
Marinedrugs 22 00447 g014
Figure 15. Chemical structures of diterpenes from Spongia sponges (166192).
Figure 15. Chemical structures of diterpenes from Spongia sponges (166192).
Marinedrugs 22 00447 g015
Figure 16. Chemical structures of diterpenes from Spongia (193208), Spongionella (209213), and Strongylophora (214) sponges.
Figure 16. Chemical structures of diterpenes from Spongia (193208), Spongionella (209213), and Strongylophora (214) sponges.
Marinedrugs 22 00447 g016
Figure 17. Chemical structures of diterpenes from Stylissa (215216), Svenzea (217220), Tedania (221), Theonella (222223), and unidentified (224228) sponges.
Figure 17. Chemical structures of diterpenes from Stylissa (215216), Svenzea (217220), Tedania (221), Theonella (222223), and unidentified (224228) sponges.
Marinedrugs 22 00447 g017
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

Xia, J.; Chen, X.; Li, G.; Qiu, P.; Wang, W.; Shao, Z. A Review of Sponge-Derived Diterpenes: 2009–2022. Mar. Drugs 2024, 22, 447. https://doi.org/10.3390/md22100447

AMA Style

Xia J, Chen X, Li G, Qiu P, Wang W, Shao Z. A Review of Sponge-Derived Diterpenes: 2009–2022. Marine Drugs. 2024; 22(10):447. https://doi.org/10.3390/md22100447

Chicago/Turabian Style

Xia, Jinmei, Xiangwei Chen, Guangyu Li, Peng Qiu, Weiyi Wang, and Zongze Shao. 2024. "A Review of Sponge-Derived Diterpenes: 2009–2022" Marine Drugs 22, no. 10: 447. https://doi.org/10.3390/md22100447

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