Next Article in Journal
Unravelling the Dermatological Potential of the Brown Seaweed Carpomitra costata
Next Article in Special Issue
The Prospective Use of Brazilian Marine Macroalgae in Schistosomiasis Control
Previous Article in Journal
Exopolysaccharide from Porphyridium cruentum (purpureum) is Not Toxic and Stimulates Immune Response against Vibriosis: The Assessment Using Zebrafish and White Shrimp Litopenaeus vannamei
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Metabolites from Marine Sponges and Their Potential to Treat Malarial Protozoan Parasites Infection: A Systematic Review

by
Anna Caroline Campos Aguiar
1,
Julia Risso Parisi
1,
Renata Neves Granito
1,
Lorena Ramos Freitas de Sousa
2,
Ana Cláudia Muniz Renno
1 and
Marcos Leoni Gazarini
1,*
1
Department of Biosciences, Federal University of São Paulo (UNIFESP), Rua Silva Jardim 136, Santos 11015-020, SP, Brazil
2
Special Academic Unit of Chemistry, Federal University of Goiás (UFG/UFCAT), Catalão Regional, Catalão 75704-020, GO, Brazil
*
Author to whom correspondence should be addressed.
Submission received: 28 January 2021 / Revised: 23 February 2021 / Accepted: 24 February 2021 / Published: 28 February 2021
(This article belongs to the Special Issue Marine Natural Products and Neglected Tropical Diseases)

Abstract

:
Malaria is an infectious disease caused by protozoan parasites of the Plasmodium genus through the bite of female Anopheles mosquitoes, affecting 228 million people and causing 415 thousand deaths in 2018. Artemisinin-based combination therapies (ACTs) are the most recommended treatment for malaria; however, the emergence of multidrug resistance has unfortunately limited their effects and challenged the field. In this context, the ocean and its rich biodiversity have emerged as a very promising resource of bioactive compounds and secondary metabolites from different marine organisms. This systematic review of the literature focuses on the advances achieved in the search for new antimalarials from marine sponges, which are ancient organisms that developed defense mechanisms in a hostile environment. The principal inclusion criterion for analysis was articles with compounds with IC50 below 10 µM or 10 µg/mL against P. falciparum culture. The secondary metabolites identified include alkaloids, terpenoids, polyketides endoperoxides and glycosphingolipids. The structural features of active compounds selected in this review may be an interesting scaffold to inspire synthetic development of new antimalarials for selectively targeting parasite cell metabolism.

1. Introduction

Human malaria is an infectious disease caused by single-celled protozoan parasites of the Plasmodium genus (P. falciparum, P. vivax, P. ovale, P. malariae, and P. knowlesi) through the bite of female Anopheles mosquitoes [1]. It affected 228 million people in 2018, and nearly half of the world’s population is still at risk for this disease [2]. Symptoms can range from being mild to very severe, causing chronic illness, physical disability, death and a huge health burden, especially to the most vulnerable populations.
Antimalarials based in quinolines scaffolds (i.e., chloroquine, mefloquine, amodiaquine, and piperaquine) possess a complex mechanism of action. One well-studied mechanism involves compromising the detoxification of hemoglobin degradation with heme polymerization for hemozoin crystal formation in digestive vacuole by protonated forms of quinolones [3]. It was noted that some strains of P. falciparum triggered resistance to protonated drugs due to a genetic mutation in the transporter (PfCRT) and could lead to antimalarial drug extrusion from the organelle [3].
Artemisinin-based combination therapies (ACTs) are the most recommended treatment for uncomplicated P. falciparum malaria, while artesunate is considered the most effective antimalarial drug for severe cases [4], with several biochemical processes reported as targets in parasite cells [3,5]. Despite the safety and efficiency that have been proven for the use of these drugs, the emergence of multidrug resistance has unfortunately limited their effects and challenged the field [6]. The resistance to ACTs is already spreading from Southeast Asia, as reported in 2008 [7], giving rise to a danger alert to other high-poverty regions in the world, and the identified resistance phenotype is associated with mutation of kelch domain protein gene (k13), which is postulated to be involved in protein trafficking organelles in the parasite during intraerythrocytic cycle [8], [3].
In this context, the ocean, with its rich biodiversity, has been emerging as a very promising resource of bioactive compounds and secondary metabolites from different marine organisms (bacteria, fungi, micro-algae, mollusks and other invertebrates) with multiple pharmacological properties [9,10,11]. Among them, the phylum Porifera (sponges) is the most promising for providing raw material for the development of biotechnological products for multiple human health problems [12,13,14]. Marine sponges are very primitive sessile animals with origins dated at least from the late Proterozoic over 580 million years ago [15]. Being considered representatives of the first multicellular animals, these filter-feeding organisms evolutionarily developed morphological and chemical defense mechanisms constituted mainly by secondary metabolites, compounds with a wide range of effects such as antitumor, antiviral, anti-inflammatory and antibiotic effects, which have been investigated for the treatment of human health problems [15,16]. Additionally, some authors have demonstrated the antimalarial effects of the secondary metabolites of marine sponges and have shown that these components present inhibitory activity against the malaria parasite Plasmodium falciparum [6,17].
Many studies have investigated the structural diversity of marine natural products from sponges worldwide showing strong evidence of their antimalarial effects; however, there is still limited understanding of their biological effects. To explore the complete therapeutic potential of marine-sponges-derived compounds, more inputs are required, especially from the comparison of the antiplasmodial potential of all of these biocompounds. Previous reviews have contributed discussion of potential antimalarial compounds from marine sources and have helped to cover the growing number of new compounds studied every year and parasite resistance to currently used antimalarials [3,18,19]. In this context, the purpose of this study was to perform a systematic review updated of the literature to examine the multiple studies reporting the in vitro antiplasmodial activity of extracts and molecules from species of marine sponges, exploring the molecules scaffold and differential target mechanisms in cell physiology.

2. Results and Discussion

2.1. Study Selection and Analysis

The flow diagram (Figure 1) demonstrated the search strategy (identification, inclusion and exclusion) used in the present study. A total of 77 articles were retrieved from the databases (PubMed, Web of Science and Scopus). Then, the duplicated records were excluded (n = 14). Thus, 66 full-text articles were assessed for eligibility, and 30 studies were excluded for different reasons, such as the following: some studies reported only the extraction of compounds and did not report the antiplasmodial activity; others described only the mechanism of the compounds; some studies were only computational. Finally, 36 studies were included and analyzed in this systematic review (Figure 1).
A summary of the studies is presented in Table 1. The articles analyzed were published from 1992 to 2019 in different countries. The antimalarial activity was assessed in vitro using Plasmodium falciparum culture [20] for quantification of cell viability over 24-96 h. For the in vitro assays, different lab strains were used (such as 3D7, W2, DD2, NF54), and a wide variety of methods were used for assessing P. falciparum viability ([3H] hypoxanthine, LDH, Microscopy, SYBR Green) presenting as IC50 values instead of the option of XC50. The Demospongiae sponge class was the most explored, where 30 studies evaluated their antiplasmodial activity. Among the genera in Table 1, most belong to the Demospongiae class except for Plakortis (Plakortis simplexs, Plakortis lita, Plakortis halichondrioides), which is from the Homoscleromorpha class. In addition, a great geographical variety was observed, which shows that sponges from different regions of the globe have this potential antiplasmodial activity. The inhibitory concentration for 50% of the parasites (IC50) varied from low micromolar to low nanomolar range, and the species Xestospongia sp showed the best bioactive potential, from which the compound Saringosterol was extracted, which had an IC50 of 0.25 nM. The individual IC50 for each extracted compound is reported in Table 1. The IC50 value units in µg/mL and ng/mL were converted to µM and nM for data comparison, and then some of compounds in Table 1, which IC50 was below 10 µg/mL became higher than 10 µM (see Section 3.2.2, exclusion criteria), as were compounds 10 and 11 [21], 52 [22], 2 and 3 [23], 99 [24].
To assess the study quality, we used the GRADE method [55]. The 36 studies analyzed were categorized as moderate quality (17) because (i) there were no controls in the experiments; (ii) the toxicity of the compounds was not assessed in parallel, which made it impossible to determine the selectivity of compounds; (iii) all compounds analyzed presented a high cytotoxicity, which demonstrates the unspecified use against P. falciparum; (iv) the methods used to measure the antiplasmodial activity were not described. A total of 19 studies were classified as being of high quality (Table S1).
After this detailed review of the articles reporting the activity of compounds from marine sponges, we made a brief survey of data in the literature to compare the number of articles published reporting the activity of marine organisms with the number of articles published reporting the activity of extracts from plants. To do so, the following combinations of keywords were used: "new antimalarials and plants" or "new antimalarials and marine" and selected the works published in the last 10 years. Figure 2 represents the number of studies reporting antiplasmodial activity of new compounds found. The search for new compounds from marine sources is still uncommon compared to the search for natural products from plants. Other recent reviews have also reported this comparison, which reinforces the importance of seeking new products from marine sources, especially considering that the diverse nature of metabolites produced by these alternative sources presents a compelling case for intensive exploration [56].

2.2. Classes of Compounds Found in Marine Sponge Extracts

The compounds isolated from marine sponges presented in the articles analyzed with antiplasmodial effect belong to alkaloids, terpenes and polyketides class of secondary metabolites. Most of the compounds with potential activity against Plasmodium sp. are alkaloids (69% of 259), followed by terpenoids (17%) and polyketides endoperoxides (13%) (Figure 3). There are also reports of glycosphingolipids (GSL) from sponges able to inhibit the malaria parasite as well. The structures with their potency are described, and some of them present the known mechanism of action, which are discussed below.

2.2.1. Alkaloids

Alkaloids from marine sponges have shown potential against infectious diseases, particularly against malaria. The natural alkaloids identified are grouped as pyrrole-imidazole [23,27], indole-imidazole [33], indole [21], manzamine [45,57], ingamine alkaloids [39], bromotyrosine [43], guanidine [28,30,32] phloeodictynes [51], pentacyclic quinones [50], pyrroloiminoquinone [38], thiazine alkaloids [34], and diterpene alkaloids [48,58].
Pyrrole-imidazole-related alkaloids (13) were verified in Demospongiae class (Porifera, horny sponges) from Agelas oroides (Agelasidae family) [23]. Moreover, a bromopyrrole alkaloid known as pseudoceratidine (4) with antiplasmodial potential (IC50 of 1.1 µM) was isolated from Tedania brasiliensis (Tedaniidae, Poecilosclerida) and Pseudoceratina purpurea (Pseudoceratinidae, Verongida) [27]. The (E)-oroidin (1) was a potent alkaloid against P. falciparum strains in vitro (IC50 of 10 µM), being revealed as a PfFabI inhibitor (IC50 of 0.77 µM) with uncompetitive behavior (Figure 4) [23]. The P. falciparum enoyl-ACP reductase (PfFabI) is an essential enzyme responsible for the catalyzes of the last step of the fatty acid pathways [59].
Indole alkaloids (59) from Spongosorites genus (Halichondriidae family) [33] and (E)-6-bromo-2’-demethyl-3’-N-methylaplysinopsin (10) and (Z)-6-bromo-2’-demethyl-3’-N-methylaplysinopsin (11) from Fascaplysinopsis reticulata (Thorectidae family) (Table 1)[21], were also shown to be inhibitors of P. falciparum, with nortopsentin A (5) as the most potent and selective compound (IC50 = 0.46 µM and SI 14.3). In addition, nortopsentin blocked trophozoite development, suggesting the inhibition of DNA synthesis in the early trophozoite stage [33].
A bioguided fractionation of Pacific marine sponge Acanthostrongylophora ingens (Petrosiidae family) using in vitro assay with P. falciparum yielded the isolation of manzamine alkaloids (1215) (IC50 values between 0.010 and 0.060 µM) [45]. Manzamine A (13) and 8-hydroxymanzamine A (12) are highlighted for transcending the observed potential of antimalarial drugs in vivo on P. berghei-infected mice compared to chloroquine and artemisinin but with high cytotoxicity [57]. Alkaloids from Hyrtios Cf. erecta sponge containing β-carboline ring (16 and 17) but lacking polycyclic moiety were also active on P. falciparum in vitro [53]. Unlike manzamine A, polycyclic alkaloids without the β-carboline ring exhibited high selectivity index and maintained antimalarial effectiveness, as observed in gamine alkaloids (18 and 19) from Petrosid Ng5 Sp5 [39].
Bromotyrosine alkaloids containing spiroisoxazoline scaffold (2026) identified in the Hyatella (Spongiidae family), Aplysinella strongylata (Aplysinellidae family), Pseudoceratina (Pseudoceratinidae family) and Verongula genus (Aplysinidae family), have been reported as inhibitors of malaria parasite as well [40,41,43]. Among them, psammaplysin H (20) showed the best IC50 potency against 3D7 line of P. falciparum at 0.41 µM and the best selectivity (SI > 97) [43].
Guanidine alkaloids are representative antimalarial NPs [28,30,32,60], including netamines G–S from Madagascar sponge Biemna laboutei (Biemnidae family, Poecilosclerida) (2734) [28,30,32] and phloeodictynes mixtures (3542) from Oceanapia fistulosa [51] (Table 1). Compounds containing guanidine moiety with pentacyclic skeleton (2934) were demonstrated to be more potent, particularly ptilomycalin F (30) and fromiamycalin (34) (IC50 of 0.23 and 0.24 µM, respectively) [28].
Pentacyclic quinone alkaloids from Xestospongia sp revealed moderate inhibitory activity on P. falciparum protein kinases (PfPK5 and Pfnek-1) (Figure 4), enzymes involved in cell division of parasite, but xestoquinone (43) was able to slightly inhibit the parasite in vivo [50]. From Australian Marine sponge Zyzzya sp. (Acarnidae), a new compound, tsitsikammamine C (44), was revealed together with six known pyrroloiminoquinone alkaloids [38]. Of the seven, four were potent in vitro against resistant strains of P. falciparum (3D7 and Dd2, IC50 < 100 nM) highlighting compound 44 with high potency and lower toxicity (SI 200), which was able to act on both blood stages of parasite, ring and trophozoite [38]. Later, Davis and co-workers [34] isolated tricyclic alkaloid from Plakortis lita with thiazine-fused quinone, thiaplakortones A–D (4548). Once more, alkaloids with quinone core revealed antimalarial potential in the nanomolar range (IC50 < 651 nM) with moderate toxicity.
Diterpene alkaloids from Agelas cf. mauritiana (49 and 50) exhibited slight antimalarial potential [48], besides [58] reported a diterpene alkaloid, monamphilectine A (51) (Hymeniacidon sp.) containing a distinct β-lactam core with high potential (Table 1).

2.2.2. Terpenes

Terpenes from sponges with antiplasmodial activity belong to the class of norterpene endoperoxides [17,25], sterols [25,26] meroterpenes [49], diterpenes [37,47,58], and sesquiterpenes [54,61]. Norterpene with cyclic endoperoxides scaffold is very common in Diacarnus genus (family Podospongiidae, order Poecilosclerida) of the marine sponges. Several norditerpene and norsesterterpene peroxide metabolites (5262) with antimalarial potential were isolated from Diacarnus megaspinorhabdosa and Diacarnus erythraeanus species, whose peroxide moiety may be related to their activities [17,22,29]. The presence of endoperoxide in sterols from Coscinoderma sp., such as (24S)-5α,8α-epidioxy-24-methylcholesta-6-en-3β-ol (63) and 5α,8α-epidioxy-24-methylcholesta-6,9(11), 24(28)-trien-3β-ol (64), revealed activity against a resistant strain of P. falciparum (Dd2) as well (Table 1) [25]. Endoperoxide bridge is a pharmacophore that is well known in artemisinin drug, whose cleavage generates reactive oxygen species (ROS) inducing parasite death [62]. However, sterols from Xestospongia sp. (Petrosiidae family) lacking peroxide (kaimanol (65) and a saringosterol (66) were able to reduce parasite development expressively (IC50 values of 359 and 0.250 nM) [26].
Meroterpenes (6770) from a new Caledonian sponge with antiplasmodial effect showed inhibitory potential against plasmodial kinase Pfnek-1 and a farnesyl transferase (Figure 5) [49]. As we described in the section above, xestoquinone (43), a quinone alkaloid from Xestospongia sp., is also a protein kinase inhibitor (PfPK5 and Pfnek-1), and it was suggested by Desoubzdanne and colleagues [49] that quinone/phenolic scaffold in the meroterpenes may be related to Pfnek-1 inhibition [50].
Diterpenes and sesquiterpenes containing isonitrile moiety with antimalarial potential have been isolated from sponges such as Stylissa cf. massa (7173) [37], Hymeniacidon sp. (74) [58], Cymbastela hooperi (75) [47] and Acanthella klethra (7680) [54] (Table 1). The isonitrile scaffold has been suggested as important for the effect of these compounds against P. falciparum; besides, there are sesquiterpenes lacking isonitrile moiety, as well as compounds smenotronic acid (81), ilimaquinone (82) and pelorol (83) from Hyrtios erectus with antimalarial potential (IC50 values ranging from 0.8 to 3.51 µM) [61].

2.2.3. Polyketides

Polyketides are common secondary metabolites identified in marine sponges with vast structural diversity. Trisoxazole macrolides (8490) are large macrocyclic polyketides from Pachastrissa nux (Calthropellidae family) [36,42]. The macrolides and polyketides with skeletons containing endoperoxides (six- or five-membered 1,2-dioxygenated rings), mostly found in the Plakinastrella and Plakortis genus (Plakinidae family), have been revealed to have antimalarial potential [24,31,44,52,63].
A series of polyketides with endoperoxides with potential against P. falciparum strains were isolated from Plakortis simplex (9197), a Caribbean sponge (IC50 values ranging from 0.39 to 6.18 µM) [31,52], and from Plakortis sp. (98, 99) and Plakortis halichondrioides (100105), whose compounds 103 and 105 are endoperoxides derivatives (lactones) (IC50 values ranging from 0.756 to 15.1 µM) (Table 1) [24,44]. The endoperoxide moiety has been described as a pharmacophore and by computational study was suggested to be a mechanism similar to the artemisinin drug, involving radical reactions as a result of the ROS [31].
Derivatives of plakortin named gracilioetheres A–C from Agelas gracilis were isolated from a bioassay-guided approach from an active extract using P. falciparum assay in vitro, highlighting gracilioether B (106) with a IC50 value of 1.41 µM and moderate cytoxicity [46].

2.2.4. Glycosphingolipids

Glycosphingolipids (GSL) are glycolipids with sugar moiety well known for the immunomodulating activity, and they have been identified in marine sponges from Agelas and Axinyssa genus [35,64]. Although there are few reports of GSL from marine sponges with antimalarial potential, Farokhi and co-workers [35] isolated a GSL with antiplasmodial activity in the low micromolar range (IC50 of 0.53 µM) and with low cytotoxic effect. The active mixture of GSL consists of different carbon chain lengths named axidjiferoside-A, -B and -C (107) from Axinyssa djiferi (Dictyonellidae family).

2.3. Mechanisms of Action of the New Compounds Found in Marine Sponge Extracts

We explore the mechanism of action of each class in the literature among other cell models to present a possible mechanism involved in the inhibition of Plasmodium development (Figure 6) because of the absence of this information in many articles described in Table 1.
The alkaloids are the largest group of compounds mentioned in this review; however, they contain a significant number of molecules (17%) with unknown mechanisms. Some alkaloid compounds can be related with inhibition of signaling pathways, and induction of apoptosis and changes in gene expression are also indicated (14–37%) [23,65,66,67,68,69,70,71,72,73,74,75,76,77]. Alkaloids could present oxidant and antioxidant effects depending on the biosynthetic precursor. For example, bromothyrosine derivatives can induce apoptosis by the formation of reactive oxygen species or selective inhibition of histone deacetylases in eukaryotic cell lines [65]. This effect can be also observed with a marine metabolite (Psammaplin A) and analogues, resulting in disruption of the epigenetic cell control and compromising the gene expression and cell survival [73,78].
Quinoline analogs have been extensively studied concerning their role as the cell targets in cancer, bacteria, virus, fungi and parasites. Some of its described mechanisms are related to key cellular processes (replication, transcription, protein metabolism, etc.) because of the interaction of quinolines compounds with DNA and inhibition of topoisomerase enzymes [79,80]. Endoplasmic reticulum stress, autophagy, and cell signaling with inhibition of several enzymes (i.e., N-acetyltransferase, cyclin dependent kinase, telomerase, caspase proteases) have also been observed [77]. The impairment of cell signaling and ionic homeostasis can be observed with the antagonist effect of voltage-dependent calcium channel by guanidine derivatives alkaloids [68] and Na+ homeostasis by selective inhibition of Plasmodium falciparum P-type ATPase with indole-based natural alkaloids in a low micro-molar range [81,82]. Another important cell target is cytoskeleton filaments, which are essential for transport, cell division and organization. Some marine sponge compounds (trisoxazole-containing macrolides) can bind to F-actin subdomains by mimicking the interaction of actin-capping gelsolin family proteins, compromising the filament dynamics and leading to cell death [70,76]. The fatty acids biosynthesis is another important process for eukaryotic cells and is responsible for building membrane structures and energy metabolism. Pyrrole-imidazole alkaloids from marine sponge Agelas oroides present an inhibition effect at low micromolar range in Plasmodium falciparum enoyl-ACP reductase assay [23], which belongs to type II fatty acid pathway (FAS-II).
The second representative group is terpenes (43), which possess action related to oxidative stress and signaling pathways (30–34%) [49,50,83,84], as reported in normal and cancer cells lines, where ROS production was increased after a norterpene endoperoxide compound treatment [85]. A third group corresponds with the polyketides compounds (34), which have been shown to interact with Fe(II)heme, compromising the cell survival [86].
The available antimalarials (i.e., artemisinin) belong to the sesquiterpene group, and to some degree, the action mechanism of related sponge metabolites in Plasmodium was found to be consistent with that observed with artemisinin affecting the cell oxidative stress state and hemoglobin metabolism [3,81,87]. Hemoglobin metabolism as the principal parasite amino acid source in the host cell leads to the formation of toxic metabolites (reactive oxygen species-ROS and ferriprotoporphyrin IX). The unbalanced detoxification of these metabolites in parasite cytosol promoted by artemisinin or analogs affects many aspects of the cell physiology [81,87] as oxidative damage in different cell molecules. Some covalent protein interactions were identified with artemisinin in P. falciparum, indicating a broad action in cell metabolism, such as ornithine aminotransferase, pyruvate kinase, L-lactate dehydrogenase, spermidine synthase and S-adenosylmethionine synthetase [81]. In the same class of the endoperoxides, plakortin-related compounds from the sponge genus Plakortis bind to Fe(II) resulting in the formation of oxygen radicals and creates a cell-damaging environment for the parasite [86].
The current scenario of the development of new antimalarial drugs shows a promising molecule source from marine organisms such as sponges. However, these organisms have some weaknesses in discovering and developing antimalarial drugs: (i) the large amount of sponges’ weight needed for each compound’s identification and isolation; (ii) sponges are organisms’ symbionts with sponge-specific microbiota (unicellular eukaryotes, bacteria, fungi, virus) [88], which increases the variability from each specimen and makes it very difficult to reproduce in laboratory cultivation for identifying the source of active compounds. However, due to the ancient relationship with the hostile environment, these organisms can present a large molecule library against pathogens, which would be useful for the development of synthetic derivatives and analogs with selective inhibition of human pathogens. The cost-accessible molecular strategies available in center facilities (i.e., high-throughput genome sequencing and mass spectrometry, molecular docking) could surpass these limitations to the identification of compounds from complex organisms. An upscaling number of articles on marine source compounds every year presenting molecules reveals its importance with different action mechanisms in eukaryotic cell physiology, as mentioned in this review.

3. Methodology

3.1. Review Protocol

A systematic review of the literature was performed according to the SYRCLE guideline [89]. The following databases were consulted for this research: PubMed, Web of Science and Scopus. The search was carried out according to the orientations of PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analysis). To start the review, some descriptors of the MeSH (Medical Subject Headings) were defined: “Plasmodium falciparum”, “P. falciparum”, “antimalarial” and “sponge”. In addition, two independent reviewers (J.R.P., A.C.C.A.) searched the databases, analyzing title and summary of the results, and identified them from the inclusion and exclusion criteria, and the selected studies were further reviewed during the full-text screening.

3.2. Eligibility Criteria

3.2.1. Inclusion Criteria

1. Studies that report the antiplasmodial activity (IC50) of extracts and molecules from marine sponges against any strain of P. falciparum in vitro;
2. Any method for determining the IC50 was included (SYBR Green, Hypoxanthine, Microscopy, ELISA);

3.2.2. Exclusion Criteria

1. Animal experiments, clinical trials, reviews, case reports;
2. Studies that reported an IC50 value above 10 µM or 10 µg/mL;
3. Studies of chemical synthesis of new derivatives that were previously extracted from marine sponges;
4. Computational studies that did not report in vitro biological activity.

3.3. Data Extraction

The analyzed data included the IC50 value, which refers to the 50% growth inhibition of the parasite in vitro after incubation with different natural products extracted from marine sponges, according to the method applied to measure the antimalarial activity with the particular Plasmodium lab strain used. In addition, the sponge species, class and extraction location were also included in the analysis.

3.4. Types of Reported Results

Due to the heterogeneity of the primary studies, it was not possible to perform a meta-analysis. In order to compare the effect size (ES) of both techniques, we calculated the normalized average difference considering the values before and after the intervention. They were further classified as small (<0.20), moderate (about 0.50) or large (>0.80), according to Cohen criteria.

4. Conclusions

In conclusion, marine sponge extracts represent a large arsenal of bioactive products with antimalarial potential. Different substances, such as alkaloids, endoperoxides (terpenes and polyketides), terpenoids and glycosphingolipids, have been isolated and identified in the extracts of different sponges around the globe. The structural features of active compounds can be an interesting core for synthetic development of new antimalarials for selectively targeting parasite cell metabolism. However, studies that aim to elucidate the mechanism of action of these new compounds are still scarce in the literature.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/1660-3397/19/3/134/s1. Table S1: GRADE analysis of included articles.

Author Contributions

Conceptualization, A.C.C.A., R.N.G., A.C.M.R. and M.L.G.; methodology A.C.C.A., J.R.P., L.R.F.d.S., R.N.G., A.C.M.R. and M.L.G.; software A.C.C.A., J.R.P.; formal analysis, A.C.C.A., J.R.P., L.R.F.d.S., R.N.G., A.C.M.R. and M.L.G.; investigation A.C.C.A., J.R.P., L.R.F.d.S., R.N.G., A.C.M.R. and M.L.G.; data curation, A.C.C.A., J.R.P., L.R.F.d.S., R.N.G., A.C.M.R. and M.L.G.; writing—original draft preparation, A.C.C.A., J.R.P., L.R.F.d.S., R.N.G., A.C.M.R. and M.L.G. All authors have read and agreed to the published version of the manuscript

Funding

This work was supported by the FAPESP (Fundação de Amparo à Pesquisa do Estado de São Paulo). M.L.G (2021/01638-5), A.C.C.A. (2019/19708-0).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Crompton, P.D.; Moebius, J.; Portugal, S.; Waisberg, M.; Hart, G.; Garver, L.S.; Miller, L.H.; Barillas, C.; Pierce, S.K. Malaria immunity in man and mosquito: Insights into unsolved mysteries of a deadly infectious disease. Annu. Rev. Immunol. 2014, 32, 157–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. World Health Organization. WHO Malaria Report 2017; World Health Organization: Geneva, Switzerland, 2017. [Google Scholar]
  3. Wicht, K.J.; Mok, S.; Fidock, D.A. Molecular Mechanisms of Drug Resistance in Plasmodium falciparum Malaria. Annu. Rev. Microbiol. 2020, 74, 431–454. [Google Scholar] [CrossRef]
  4. Roussel, C.; Caumes, E.; Thellier, M.; Ndour, P.A.; Buffet, P.A.; Jauréguiberry, S. Artesunate to treat severe malaria in travellers: Review of efficacy and safety and practical implications. J. Travel Med. 2017, 24, taw093. [Google Scholar] [CrossRef] [Green Version]
  5. Bridgford, J.L.; Xie, S.C.; Cobbold, S.A.; Pasaje, C.F.A.; Herrmann, S.; Yang, T.; Gillett, D.L.; Dick, L.R.; Ralph, S.A.; Dogovski, C.; et al. Artemisinin kills malaria parasites by damaging proteins and inhibiting the proteasome. Nat. Commun. 2018, 9, 1–9. [Google Scholar] [CrossRef] [Green Version]
  6. Nieves, K.; Prudhomme, J.; Le Roch, K.G.; Franzblau, S.G.; Rodríguez, A.D. Natural product-based synthesis of novel anti-infective isothiocyanate- and isoselenocyanate-functionalized amphilectane diterpenes. Bioorganic Med. Chem. Lett. 2016, 26. [Google Scholar] [CrossRef] [Green Version]
  7. Noedl, H.; Se, Y.; Schaecher, K.; Smith, B.L.; Socheat, D.; Fukuda, M.M. Evidence of Artemisinin-Resistant Malaria in Western Cambodia. N. Engl. J. Med. 2008, 359, 2619–2620. [Google Scholar] [CrossRef]
  8. Ariey, F.; Witkowski, B.; Amaratunga, C.; Beghain, J.; Langlois, A.C.; Khim, N.; Kim, S.; Duru, V.; Bouchier, C.; Ma, L.; et al. A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature 2014, 505, 50–55. [Google Scholar] [CrossRef]
  9. Voultsiadou Eleni, E. Therapeutic properties and uses of marine invertebrates in the ancient Greek world and early Byzantium. J. Ethnopharmacol. 2010, 130. [Google Scholar] [CrossRef] [PubMed]
  10. Mayer, A.M.S.; Avilés, E.; Rodríguez, A.D. Marine sponge Hymeniacidon sp. amphilectane metabolites potently inhibit rat brain microglia thromboxane B 2 generation. Bioorganic Med. Chem. 2012, 20, 279–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. De Alencar, D.B.; Da Silva, S.R.; Pires-Cavalcante, K.M.S.; De Lima, R.L.; Pereira, F.N.; De Sousa, M.B.; Viana, F.A.; Nagano, C.S.; Do Nascimento, K.S.; Cavada, B.S.; et al. Antioxidant potential and cytotoxic activity of two red seaweed species, amansia multifida and meristiella echinocarpa, from the coast of Northeastern Brazil. An. Acad. Bras. Cienc. 2014, 86, 251–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Leys, S.P.; Hill, A. The Physiology and Molecular Biology of Sponge Tissues. Adv. Marine Biol. 2012, 62, 1–56. [Google Scholar]
  13. Mehbub, M.F.; Lei, J.; Franco, C.; Zhang, W. Marine sponge derived natural products between 2001 and 2010: Trends and opportunities for discovery of bioactives. Mar. Drugs 2014, 12, 4539–4577. [Google Scholar] [CrossRef] [Green Version]
  14. Amina, M.; Musayeib, N.M. Al Biological and Medicinal Importance of Sponge. In Biological Resources of Water; IntechOpen: London, UK, 2018. [Google Scholar]
  15. Feuda, R.; Dohrmann, M.; Pett, W.; Philippe, H.; Rota-Stabelli, O.; Lartillot, N.; Wörheide, G.; Pisani, D. Improved Modeling of Compositional Heterogeneity Supports Sponges as Sister to All Other Animals. Curr. Biol. 2017, 27, 3864–3870. [Google Scholar] [CrossRef] [Green Version]
  16. Wang, B.; Dong, J.; Zhou, X.; Huang, R.; Zhang, S.; Liu, Y.; Lee, K.J. Nucleosides from the Marine Sponge Haliclona sp. Z. Fur Nat.-Sect. C J. Biosci. 2009, 64. [Google Scholar] [CrossRef] [PubMed]
  17. 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] [Green Version]
  18. Muniswamy, K.; Thamodaran, P. Genomic imprinting: A general overview. Biotechnol. Mol. Biol. Rev. 2013, 8. [Google Scholar] [CrossRef]
  19. Fattorusso, E.; Taglialatela-Scafati, O. Marine antimalarials. Mar. Drugs 2009, 7, 130–152. [Google Scholar] [CrossRef] [Green Version]
  20. Trager, W.; Jensen, J.B. Human malaria parasites in continuous culture. Science 1976, 193, 673–675. [Google Scholar] [CrossRef]
  21. Campos, P.E.; Pichon, E.; Moriou, C.; Clerc, P.; Trépos, R.; Frederich, M.; De Voogd, N.; Hellio, C.; Gauvin-Bialecki, A.; Al-Mourabit, A. New antimalarial and antimicrobial tryptamine derivatives from the marine sponge fascaplysinopsis reticulata. Mar. Drugs 2019, 17, 167. [Google Scholar] [CrossRef] [Green Version]
  22. El Sayed, K.A.; Hamann, M.T.; Hashish, N.E.; Shier, W.T.; Kelly, M.; Khan, A.A. Antimalarial, antiviral, and antitoxoplasmosis norsesterterpene peroxide acids from the red sea sponge Diacarnus erythraeanus. J. Nat. Prod. 2001, 64, 522–524. [Google Scholar] [CrossRef] [PubMed]
  23. Tasdemir, D.; Topaloglu, B.; Perozzo, R.; Brun, R.; O’Neill, R.; Carballeira, N.M.; Zhang, X.; Tonge, P.J.; Linden, A.; Rüedi, P. Marine natural products from the Turkish sponge Agelas oroides that inhibit the enoyl reductases from Plasmodium falciparum, Mycobacterium tuberculosis and Escherichia coli. Bioorganic Med. Chem. 2007, 15, 6834–6845. [Google Scholar] [CrossRef] [PubMed]
  24. Gochfeld, D.J.; Hamann, M.T. Isolation and biological evaluation of filiformin, plakortide F, and plakortone G from the Caribbean sponge Plakortis sp. J. Nat. Prod. 2001, 64. [Google Scholar] [CrossRef]
  25. Jeong, H.; Latif, A.; Kong, C.S.; Seo, Y.; Lee, Y.J.; Dalal, S.R.; Cassera, M.B.; Kingston, D.G.I. Isolation and characterization of antiplasmodial constituents from the marine sponge Coscinoderma sp. Z. Fur Nat.-Sect. C J. Biosci. 2019. [Google Scholar] [CrossRef]
  26. Murtihapsari, M.; Salam, S.; Kurnia, D.; Darwati, D.; Kadarusman, K.; Abdullah, F.F.; Herlina, T.; Husna, M.H.; Awang, K.; Shiono, Y.; et al. A new antiplasmodial sterol from Indonesian marine sponge, Xestospongia sp. Nat. Prod. Res. 2019. [Google Scholar] [CrossRef] [PubMed]
  27. Parra, L.L.L.; Bertonha, A.F.; Severo, I.R.M.; Aguiar, A.C.C.; De Souza, G.E.; Oliva, G.; Guido, R.V.C.; Grazzia, N.; Costa, T.R.; Miguel, D.C.; et al. Isolation, Derivative Synthesis, and Structure-Activity Relationships of Antiparasitic Bromopyrrole Alkaloids from the Marine Sponge Tedania brasiliensis. J. Nat. Prod. 2018, 81. [Google Scholar] [CrossRef]
  28. Campos, P.E.; Wolfender, J.L.; Queiroz, E.F.; Marcourt, L.; Al-Mourabit, A.; Frederich, M.; Bordignon, A.; De Voogd, N.; Illien, B.; Gauvin-Bialecki, A. Unguiculin A and Ptilomycalins E-H, Antimalarial Guanidine Alkaloids from the Marine Sponge Monanchora unguiculata. J. Nat. Prod. 2017, 80. [Google Scholar] [CrossRef]
  29. Yang, F.; Wang, R.P.; Xu, B.; Yu, H.B.; Ma, G.Y.; Wang, G.F.; Dai, S.W.; Zhang, W.; Jiao, W.H.; Song, S.J.; et al. New antimalarial norterpene cyclic peroxides from Xisha Islands sponge Diacarnus megaspinorhabdosa. Bioorganic Med. Chem. Lett. 2016, 26, 2084–2087. [Google Scholar] [CrossRef] [PubMed]
  30. Gros, E.; Martin, M.T.; Sorres, J.; Moriou, C.; Vacelet, J.; Frederich, M.; Aknin, M.; Kashman, Y.; Gauvin-Bialecki, A.; Al-Mourabit, A. Netamines O-S, Five New Tricyclic Guanidine Alkaloids from the Madagascar Sponge Biemna laboutei, and Their Antimalarial Activities. Chem. Biodivers. 2015, 12. [Google Scholar] [CrossRef]
  31. Chianese, G.; Persico, M.; Yang, F.; Lin, H.W.; Guo, Y.W.; Basilico, N.; Parapini, S.; Taramelli, D.; Taglialatela-Scafati, O.; Fattorusso, C. Endoperoxide polyketides from a Chinese Plakortis simplex: Further evidence of the impact of stereochemistry on antimalarial activity of simple 1,2-dioxanes. Bioorganic Med. Chem. 2014, 22. [Google Scholar] [CrossRef]
  32. Gros, E.; Al-Mourabit, A.; Martin, M.T.; Sorres, J.; Vacelet, J.; Frederich, M.; Aknin, M.; Kashman, Y.; Gauvin-Bialecki, A. Netamines H-N, tricyclic alkaloids from the marine sponge biemna laboutei and their antimalarial activity. J. Nat. Prod. 2014, 77. [Google Scholar] [CrossRef]
  33. Alvarado, S.; Roberts, B.F.; Wright, A.E.; Chakrabarti, D. The bis(Indolyl)imidazole alkaloid nortopsentin a exhibits antiplasmodial activity. Antimicrob. Agents Chemother. 2013, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Davis, R.A.; Duffy, S.; Fletcher, S.; Avery, V.M.; Quinn, R.J. Thiaplakortones A-D: Antimalarial thiazine alkaloids from the Australian marine sponge plakortis lita. J. Org. Chem. 2013, 78. [Google Scholar] [CrossRef] [PubMed]
  35. Farokhi, F.; Grellier, P.; Clément, M.; Roussakis, C.; Loiseau, P.M.; Genin-Seward, E.; Kornprobst, J.M.; Barnathan, G.; Wielgosz-Collin, G. Antimalarial activity of axidjiferosides, new β-galactosylceramides from the African sponge Axinyssa djiferi. Mar. Drugs 2013, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Sirirak, T.; Brecker, L.; Plubrukarn, A. Kabiramide L, a new antiplasmodial trisoxazole macrolide from the sponge Pachastrissa nux. Nat. Prod. Res. 2013, 27. [Google Scholar] [CrossRef]
  37. Chanthathamrongsiri, N.; Yuenyongsawad, S.; Wattanapiromsakul, C.; Plubrukarn, A. Bifunctionalized amphilectane diterpenes from the sponge Stylissa cf. massa. J. Nat. Prod. 2012, 75. [Google Scholar] [CrossRef]
  38. Davis, R.A.; Buchanan, M.S.; Duffy, S.; Avery, V.M.; Charman, S.A.; Charman, W.N.; White, K.L.; Shackleford, D.M.; Edstein, M.D.; Andrews, K.T.; et al. Antimalarial activity of pyrroloiminoquinones from the Australian marine sponge zyzzya sp. J. Med. Chem. 2012, 55. [Google Scholar] [CrossRef] [Green Version]
  39. Ilias, M.; Ibrahim, M.A.; Khan, S.I.; Jacob, M.R.; Tekwani, B.L.; Walker, L.A.; Samoylenko, V. Pentacyclic ingamine alkaloids, a new antiplasmodial pharmacophore from the marine sponge Petrosid Ng5 Sp5. Planta Med. 2012, 78. [Google Scholar] [CrossRef]
  40. Mudianta, I.W.; Skinner-Adams, T.; Andrews, K.T.; Davis, R.A.; Hadi, T.A.; Hayes, P.Y.; Garson, M.J. Psammaplysin derivatives from the balinese marine sponge Aplysinella strongylata. J. Nat. Prod. 2012, 75. [Google Scholar] [CrossRef]
  41. Galeano, E.; Thomas, O.P.; Robledo, S.; Munoz, D.; Martinez, A. Antiparasitic Bromotyrosine derivatives from the marine sponge Verongula rigida. Mar. Drugs 2011, 9. [Google Scholar] [CrossRef]
  42. Sirirak, T.; Kittiwisut, S.; Janma, C.; Yuenyongsawad, S.; Suwanborirux, K.; Plubrukarn, A. Kabiramides J and K, trisoxazole macrolides from the sponge Pachastrissa nux. J. Nat. Prod. 2011, 74. [Google Scholar] [CrossRef]
  43. Xu, M.; Andrews, K.T.; Birrell, G.W.; Tran, T.L.; Camp, D.; Davis, R.A.; Quinn, R.J. Psammaplysin H, a new antimalarial bromotyrosine alkaloid from a marine sponge of the genus Pseudoceratina. Bioorganic Med. Chem. Lett. 2011, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Jiménez-Romero, C.; Ortiz, I.; Vicente, J.; Vera, B.; Rodríguez, A.D.; Nam, S.; Jove, R. Bioactive cycloperoxides isolated from the Puerto Rican sponge Plakortis halichondrioides. J. Nat. Prod. 2010, 73. [Google Scholar] [CrossRef] [Green Version]
  45. Samoylenko, V.; Khan, S.I.; Jacob, M.R.; Tekwani, B.L.; Walker, L.A.; Hufford, C.D.; Muhammad, I. Bioactive (+)-manzamine A and (+)-8-hydroxymanzamine A tertiary bases and salts from Acanthostrongylophora ingens and their preparations. Nat. Prod. Commun. 2009, 4. [Google Scholar] [CrossRef] [Green Version]
  46. Ueoka, R.; Nakao, Y.; Kawatsu, S.; Yaegashi, J.; Matsumoto, Y.; Matsunaga, S.; Furihata, K.; Van Soest, R.W.M.; Fusetani, N. Gracilioethers A-C, antimalarial metabolites from the marine sponge Agelas gracilis. J. Org. Chem. 2009, 74. [Google Scholar] [CrossRef]
  47. 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. [Google Scholar] [CrossRef]
  48. Appenzeller, J.; Mihci, G.; Martin, M.T.; Gallard, J.F.; Menou, J.L.; Boury-Esnault, N.; Hooper, J.; Petek, S.; Chevalley, S.; Valentin, A.; et al. Agelasines J, K, and L from the Solomon Islands marine sponge Agelas cf. mauritiana. J. Nat. Prod. 2008, 71. [Google Scholar] [CrossRef]
  49. Desoubzdanne, D.; Marcourt, L.; Raux, R.; Chevalley, S.; Dorin, D.; Doerig, C.; Valentin, A.; Ausseil, F.; Debitus, C. Alisiaquinones and Alisiaquinol, dual inhibitors of Plasmodium falciparum enzyme targets from a new caledonian deep water sponge. J. Nat. Prod. 2008, 71. [Google Scholar] [CrossRef]
  50. Laurent, D.; Jullian, V.; Parenty, A.; Knibiehler, M.; Dorin, D.; Schmitt, S.; Lozach, O.; Lebouvier, N.; Frostin, M.; Alby, F.; et al. Antimalarial potential of xestoquinone, a protein kinase inhibitor isolated from a Vanuatu marine sponge Xestospongia sp. Bioorganic Med. Chem. 2006, 14. [Google Scholar] [CrossRef]
  51. Mancini, I.; Guella, G.; Sauvain, M.; Debitus, C.; Duigou, A.G.; Ausseil, F.; Menou, J.L.; Pietra, F. New 1,2,3,4-tetrahydropyrrolo [1,2-a]pyrimidinium alkaloids (phloeodictynes) from the New Caledonian shallow-water haplosclerid sponge Oceanapia fistulosa. Structural elucidation from mainly LC-tandem-MS-soft-ionization techniques and discovery of antiplasmodial activity. Org. Biomol. Chem. 2004, 2, 783–787. [Google Scholar] [CrossRef]
  52. Fattorusso, E.; Parapini, S.; Campagnuolo, C.; Basilico, N.; Taglialatela-Scafati, O.; Taramelli, D. Activity against Plasmodium falciparum of cycloperoxide compounds obtained from the sponge Plakortis simplex. J. Antimicrob. Chemother. 2002, 50, 883–888. [Google Scholar] [CrossRef] [Green Version]
  53. Kirsch, G.; Köng, G.M.; Wright, A.D.; Kaminsky, R. A new bioactive sesterterpene and antiplasmodial alkaloids from the marine sponge Hyrtios cf. erecta. J. Nat. Prod. 2000, 63. [Google Scholar] [CrossRef]
  54. Angerhofer, C.K.; Pezzuto, J.M.; König, G.M.; Wright, A.D.; Sticher, O. Antimalarial activity of sesquiterpenes from the marine sponge acanthella klethra. J. Nat. Prod. 1992, 55. [Google Scholar] [CrossRef]
  55. Guyatt, G.; Oxman, A.D.; Akl, E.A.; Kunz, R.; Vist, G.; Brozek, J.; Norris, S.; Falck-Ytter, Y.; Glasziou, P.; Debeer, H.; et al. GRADE guidelines: 1. Introduction-GRADE evidence profiles and summary of findings tables. J. Clin. Epidemiol. 2011, 64. [Google Scholar] [CrossRef] [PubMed]
  56. Tajuddeen, N.; Van Heerden, F.R. Antiplasmodial natural products: An update. Malar. J. 2019, 18, 1–62. [Google Scholar] [CrossRef] [Green Version]
  57. Ang, K.K.H.; Holmes, M.J.; Higa, T.; Hamann, M.T.; Kara, U.A.K. In vivo antimalarial activity of the beta-carboline alkaloid manzamine A. Antimicrob. Agents Chemother. 2000, 44. [Google Scholar] [CrossRef] [Green Version]
  58. 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. [Google Scholar] [CrossRef] [Green Version]
  59. Waller, R.F.; Keeling, P.J.; Donald, R.G.K.; Striepen, B.; Handman, E.; Lang-Unnasch, N.; Cowman, A.F.; Besra, G.S.; Roos, D.S.; Mcfadden, G.I. Nuclear-encoded proteins target to the plastid in Toxoplasma gondii and Plasmodium falciparum. Proc. Natl. Acad. Sci. USA 1998, 95. [Google Scholar] [CrossRef] [Green Version]
  60. Hua, H.M.; Peng, J.; Fronczek, F.R.; Kelly, M.; Hamann, M.T. Crystallographic and NMR studies of antiinfective tricyclic guanidine alkaloids from the sponge Monanchora unguifera. Bioorganic Med. Chem. 2004, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Ju, E.; Latif, A.; Kong, C.S.; Seo, Y.; Lee, Y.J.; Dalal, S.R.; Cassera, M.B.; Kingston, D.G.I. Antimalarial activity of the isolates from the marine sponge Hyrtios erectus against the chloroquine-resistant Dd2 strain of Plasmodium falciparum. Z. Fur Nat.-Sect. C J. Biosci. 2018, 73. [Google Scholar] [CrossRef] [PubMed]
  62. Gunjan, S.; Sharma, T.; Yadav, K.; Chauhan, B.S.; Singh, S.K.; Siddiqi, M.I.; Tripathi, R. Artemisinin Derivatives and Synthetic Trioxane Trigger Apoptotic Cell Death in Asexual Stages of Plasmodium. Front. Cell. Infect. Microbiol. 2018, 8. [Google Scholar] [CrossRef]
  63. Higgs, M.D.; Faulkner, D.J. Plakortin, an Antibiotic from Plakortis halichondrioides. J. Org. Chem. 1978, 43. [Google Scholar] [CrossRef]
  64. Pettit, G.R.; Xu, J.P.; Gingrich, D.E.; Williams, M.D.; Doubek, D.L.; Chapuis, J.C.; Schmidt, J.M. Antineoplastic agents, Part 395. Isolation and structure of agelagalastatin from the Papua New Guinea marine sponge Agelas sp. Chem. Commun. 1999, 10, 915–916. [Google Scholar] [CrossRef]
  65. Tarazona, G.; Santamaría, G.; Cruz, P.G.; Fernández, R.; Pérez, M.; Martínez-Leal, J.F.; Rodríguez, J.; Jiménez, C.; Cuevas, C. Cytotoxic Anomoian B and Aplyzanzine B, New Bromotyrosine Alkaloids from Indonesian Sponges. ACS Omega 2017, 2. [Google Scholar] [CrossRef] [Green Version]
  66. Goey, A.K.L.; Chau, C.H.; Sissung, T.M.; Cook, K.M.; Venzon, D.J.; Castro, A.; Ransom, T.R.; Henrich, C.J.; McKee, T.C.; McMahon, J.B.; et al. Screening and Biological Effects of Marine Pyrroloiminoquinone Alkaloids: Potential Inhibitors of the HIF-1α/p300 Interaction. J. Nat. Prod. 2016, 79. [Google Scholar] [CrossRef]
  67. Zhou, X.; Sun, J.; Ma, W.; Fang, W.; Chen, Z.; Yang, B.; Liu, Y. Bioactivities of six sterols isolated from marine invertebrates. Pharm. Biol. 2014, 52, 187–190. [Google Scholar] [CrossRef] [Green Version]
  68. Grkovic, T.; Blees, J.S.; Bayer, M.M.; Colburn, N.H.; Thomas, C.L.; Henrich, C.J.; Peach, M.L.; McMahon, J.B.; Schmid, T.; Gustafson, K.R. Tricyclic guanidine alkaloids from the marine sponge Acanthella cavernosa that stabilize the tumor suppressor PDCD4. Mar. Drugs 2014, 12, 4593–4601. [Google Scholar] [CrossRef] [Green Version]
  69. Kimura, J.; Ishizuka, E.; Nakao, Y.; Yoshida, W.Y.; Scheuer, P.J.; Kelly-Borges, M. Isolation of 1-methylherbipoline salts of halisulfate-1 and of suvanine as serine protease inhibitors from a marine sponge, Coscinoderma mathewsi. J. Nat. Prod. 1998, 61. [Google Scholar] [CrossRef] [Green Version]
  70. Klenchin, V.A.; Allingham, J.S.; King, R.; Tanaka, J.; Marriott, G.; Rayment, I. Trisoxazole macrolide toxins mimic the binding of actin-capping proteins to actin. Nat. Struct. Biol. 2003, 10. [Google Scholar] [CrossRef]
  71. Manikandan, S.; Ganesapandian, S.; Singh, M.; Kumaraguru, A.K. Anti-tumour activity of bromopyrrole alkaloids against human breast tumour (MCF-7) through apoptosis induction. Int. J. Pharm. Pharm. Sci. 2019. [Google Scholar] [CrossRef] [Green Version]
  72. Olkkonen, V.M.; Béaslas, O.; Nissilä, E. Oxysterols and their cellular effectors. Biomolecules 2012, 2, 76–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Baud, M.G.J.; Leiser, T.; Haus, P.; Samlal, S.; Wong, A.C.; Wood, R.J.; Petrucci, V.; Gunaratnam, M.; Hughes, S.M.; Buluwela, L.; et al. Defining the mechanism of action and enzymatic selectivity of psammaplin A against its epigenetic targets. J. Med. Chem. 2012, 55. [Google Scholar] [CrossRef]
  74. Chung, S.C.; Lee, S.H.; Jang, K.H.; Park, W.; Jeon, J.E.; Oh, H.; Shin, J.; Oh, K.B. Actin depolymerizing effect of trisoxazole-containing macrolides. Bioorganic Med. Chem. Lett. 2011, 21. [Google Scholar] [CrossRef]
  75. Furuta, A.; Salam, K.A.; Hermawan, I.; Akimitsu, N.; Tanaka, J.; Tani, H.; Yamashita, A.; Moriishi, K.; Nakakoshi, M.; Tsubuki, M.; et al. Identification and biochemical characterization of halisulfate 3 and suvanine as novel inhibitors of hepatitis C virus NS3 helicase from a marine sponge. Mar. Drugs 2014, 12, 462–476. [Google Scholar] [CrossRef]
  76. Berlinck, R.G.S.; Braekman, J.C.; Daloze, D.; Bruno, I.; Riccio, R.; Ferri, S.; Spampinato, S.; Speroni, E. Polycyclic guanidine alkaloids from the marine sponge crambe crambe and Ca++ channel blocker activity of crambescidin 816. J. Nat. Prod. 1993, 56. [Google Scholar] [CrossRef] [PubMed]
  77. Thawabteh, A.; Juma, S.; Bader, M.; Karaman, D.; Scrano, L.; Bufo, S.A.; Karaman, R. The biological activity of natural alkaloids against herbivores, cancerous cells and pathogens. Toxins 2019, 11, 656. [Google Scholar] [CrossRef] [Green Version]
  78. Pereira, R.; Benedetti, R.; Pérez-Rodríguez, S.; Nebbioso, A.; García-Rodríguez, J.; Carafa, V.; Stuhldreier, M.; Conte, M.; Rodríguez-Barrios, F.; Stunnenberg, H.G.; et al. Indole-derived psammaplin a analogues as epigenetic modulators with multiple inhibitory activities. J. Med. Chem. 2012, 55. [Google Scholar] [CrossRef]
  79. Senerovic, L.; Opsenica, D.; Moric, I.; Aleksic, I.; Spasić, M.; Vasiljevic, B. Quinolines and quinolones as antibacterial, antifungal, anti-virulence, antiviral and anti-parasitic agents. In Advances in Experimental Medicine and Biology; Springer: Cham, Switzerland, 2020. [Google Scholar]
  80. Byler, K.G.; Wang, C.; Setzer, W.N. Quinoline alkaloids as intercalative topoisomerase inhibitors. J. Mol. Model. 2009, 15. [Google Scholar] [CrossRef]
  81. Wang, J.; Zhang, C.J.; Chia, W.N.; Loh, C.C.Y.; Li, Z.; Lee, Y.M.; He, Y.; Yuan, L.X.; Lim, T.K.; Liu, M.; et al. Haem-activated promiscuous targeting of artemisinin in Plasmodium falciparum. Nat. Commun. 2015, 6. [Google Scholar] [CrossRef] [PubMed]
  82. Dangi, P.; Jain, R.; Mamidala, R.; Sharma, V.; Agarwal, S.; Bathula, C.; Thirumalachary, M.; Sen, S.; Singh, S. Natural Product Inspired Novel Indole based Chiral Scaffold Kills Human Malaria Parasites via Ionic Imbalance Mediated Cell Death. Sci. Rep. 2019, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Cheenpracha, S.; Park, E.J.; Rostama, B.; Pezzuto, J.M.; Chang, L.C. Inhibition of nitric oxide (NO) production in lipopolysaccharide (LPS)-activated murine macrophage RAW 264.7 cells by the norsesterterpene peroxide, epimuqubilin A. Mar. Drugs 2010, 8, 429–437. [Google Scholar] [CrossRef] [PubMed]
  84. Huang, C.Y.; Tseng, Y.J.; Chokkalingam, U.; Hwang, T.L.; Hsu, C.H.; Dai, C.F.; Sung, P.J.; Sheu, J.H. Bioactive Isoprenoid-Derived Natural Products from a Dongsha Atoll Soft Coral Sinularia erecta. J. Nat. Prod. 2016, 79. [Google Scholar] [CrossRef] [PubMed]
  85. Lefranc, F.; Nuzzo, G.; Hamdy, N.A.; Fakhr, I.; Moreno, Y.; Banuls, L.; Van Goietsenoven, G.; Villani, G.; Mathieu, V.; Van Soest, R.; et al. In vitro pharmacological and toxicological effects of norterpene peroxides isolated from the red sea sponge diacarnus erythraeanus on normal and cancer cells. J. Nat. Prod. 2013, 76. [Google Scholar] [CrossRef] [PubMed]
  86. Taglialatela-Scafati, O.; Fattorusso, E.; Romano, A.; Scala, F.; Barone, V.; Cimino, P.; Stendardo, E.; Catalanotti, B.; Persico, M.; Fattorusso, C. Insight into the mechanism of action of plakortins, simple 1,2-dioxane antimalarials. Org. Biomol. Chem. 2010, 8, 846–856. [Google Scholar] [CrossRef] [PubMed]
  87. Kavishe, R.A.; Koenderink, J.B.; Alifrangis, M. Oxidative stress in malaria and artemisinin combination therapy: Pros and Cons. FEBS J. 2017, 284, 2579–2591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Webster, N.S.; Taylor, M.W. Marine sponges and their microbial symbionts: Love and other relationships. Environ. Microbiol. 2012, 14, 335–346. [Google Scholar] [CrossRef]
  89. de Vries, R.B.M.; Hooijmans, C.R.; Langendam, M.W.; van Luijk, J.; Leenaars, M.; Ritskes-Hoitinga, M.; Wever, K.E. A protocol format for the preparation, registration and publication of systematic reviews of animal intervention studies. Evidence-Based Preclin. Med. 2015, 2, e00007. [Google Scholar] [CrossRef]
Figure 1. Flow diagram of literature search and selection criteria used in the present review adapted from PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analysis).
Figure 1. Flow diagram of literature search and selection criteria used in the present review adapted from PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analysis).
Marinedrugs 19 00134 g001
Figure 2. Number of published papers reporting the antimalarial activity of new compounds from marine sources or plants in the past 10 years.
Figure 2. Number of published papers reporting the antimalarial activity of new compounds from marine sources or plants in the past 10 years.
Marinedrugs 19 00134 g002
Figure 3. Chemical class of compounds (259) identified in the reviewed articles (37).
Figure 3. Chemical class of compounds (259) identified in the reviewed articles (37).
Marinedrugs 19 00134 g003
Figure 4. Alkaloids from marine sponges with antimalarial effect revealed moderate inhibitory activity on P. falciparum protein kinases (PfPK5 and Pfnek-1) and P. falciparum enoyl-ACP reductase (PfFabI).
Figure 4. Alkaloids from marine sponges with antimalarial effect revealed moderate inhibitory activity on P. falciparum protein kinases (PfPK5 and Pfnek-1) and P. falciparum enoyl-ACP reductase (PfFabI).
Marinedrugs 19 00134 g004
Figure 5. Meroterpenes from a marine sponge with antimalarial effect revealed inhibitory activity on P. falciparum protein kinase (Pfnek-1) and P. falciparum farnesyl transferase.
Figure 5. Meroterpenes from a marine sponge with antimalarial effect revealed inhibitory activity on P. falciparum protein kinase (Pfnek-1) and P. falciparum farnesyl transferase.
Marinedrugs 19 00134 g005
Figure 6. Histogram of related mechanisms of action for each chemical compound class in different cell models indicated by the literature.
Figure 6. Histogram of related mechanisms of action for each chemical compound class in different cell models indicated by the literature.
Marinedrugs 19 00134 g006
Table 1. Summary of descriptions of characteristics of included articles.
Table 1. Summary of descriptions of characteristics of included articles.
AuthorSponge Genus Material Collection LocationExtracted Material (P. falciparum Strain and IC50 Value)
Campos et al., (2019) [21]Fascaplysinopsis reticulataMayotte (Indian Ocean) Marinedrugs 19 00134 i001
Jeong., et al. (2019) [25]Coscinoderma sp.Chuuk Island, Federated States of Micronesia Marinedrugs 19 00134 i002
Ju et al., (2019) [25]Hyrtios erectusChuuk Island, Federated States of Micronesia Marinedrugs 19 00134 i003
Murtihapsari. et al., (2019) [26]Xestospongia spKaimana, West Papua, Indonesia Marinedrugs 19 00134 i004
Parra et al., (2018) [27]Tedania BrasiliensisBrazil Marinedrugs 19 00134 i005
Campos et al., (2017) [28]Monanchora unguiculataMitsio islands, Madagascar Marinedrugs 19 00134 i006
Yang et al.,
(2016) [29]
Diacarnus megaspinorhabdosaSouthChina Sea Sponge Marinedrugs 19 00134 i007
Gros et al.,
(2015) [30]
Biemna labouteiMadagascar Marinedrugs 19 00134 i008
Chianese et al., (2014) [31]Plakortis simplexsSouth China Sea Marinedrugs 19 00134 i009
Gros et al., (2014) [32]Biemna labouteiMadagascar at Salary Ba Marinedrugs 19 00134 i010
Yang et al., (2014) [17] Diacarnus megaspinorhabdosaSouth China Sea Marinedrugs 19 00134 i011
Alvarado et al.,
(2013) [33]
Spongosorites spNot reported Marinedrugs 19 00134 i012
Davis et al.,
(2013) [34]
Plakortis litaNot reported Marinedrugs 19 00134 i013
Farokhi et al.,
(2013) [35]
Axinyssa djiferiSenegalese coasts Marinedrugs 19 00134 i014
Sirirak et al.,
(2013) [36]
Pachastrissa nuxsThailand Marinedrugs 19 00134 i015
Chanthathamrongsiri et al.,
(2012) [37]
Stylissacf. massaNot reported Marinedrugs 19 00134 i016
Davis et al.,
(2012) [38]
Zyzzya spNot reported Marinedrugs 19 00134 i017
Ilias et al.,
(2012) [39]
PetrosiaEastern Fields north of Australia Marinedrugs 19 00134 i018
Mudianta et al.,
(2012) [40]
Aplysinella strongylataTulamben, Bali, Indonesia Marinedrugs 19 00134 i019
El Sayed et al., (2011) [22]Diacarnus erythraeanusRed Sea Marinedrugs 19 00134 i020
Galeano et al.,
(2011) [41]
Verongula rigidaUrabá Gulf is located in the Southwestern Caribbean Marinedrugs 19 00134 i021
Sirirak et al.,
(2011) [42]
Pachastrissa nuxKoh-Tao, Surat-Thani ProvinceChumphon IslandsNational Park, Chumphon Province, Marinedrugs 19 00134 i022
Xu et al.,
(2011) [43]
Pseudoceratina spAustralian biota Marinedrugs 19 00134 i023
Jiménez-Romero et al.,
(2010) [44]
Plakortis halichondrioidesPuerto Rico Marinedrugs 19 00134 i024
Samoylenko et al.,
(2009) [45]
Acanthostrongylophora ingensPacific Marinedrugs 19 00134 i025
Ueoka et al.,
(2009) [46]
Agelas gracilissouthern Japan Marinedrugs 19 00134 i026
Wright et al.,
(2009) [47]
Cymbastela hooperiNot reported Marinedrugs 19 00134 i027
Appenzeller et al.,
(2008) [48]
Agelas cf. mauritianaSolomon Islands Marinedrugs 19 00134 i028
Desoubzdanne et al.,
(2008) [49]
New CaledonianNorfolk Rise (New Caledonia) Marinedrugs 19 00134 i029
Tasdemir et al., (2007) [23]Agelas oroidesNorthern Aegean Sea, Turkey- fractions: fatty acid mixtures FAME (3.4 μg/mL) and FAMF (8.7 μg/mL)
Marinedrugs 19 00134 i030
Laurent et al., (2006) [50] XestospongiaVanuatu Marinedrugs 19 00134 i031
Mancini et al., (2004) [51]Oceanapia fistulosaNew Caledonia Main Island-crude mixture (0.98 μM)    -N-methyl derivatives from the crude mixture (8 μM)
Marinedrugs 19 00134 i032
Fattorusso et al., (2002) [52]Plakortis simplexBerry Island (Bahamas) Marinedrugs 19 00134 i033
Gochfeld et al., (2001) [24]Plakortis sp.Jamaica Marinedrugs 19 00134 i034
Kirsch et al., (2000) [53]Hyrtios cf. erectaFiji Marinedrugs 19 00134 i035
Angerhofer et al., (1992) [54]Acanthella klethraAustralia Marinedrugs 19 00134 i036
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Aguiar, A.C.C.; Parisi, J.R.; Granito, R.N.; de Sousa, L.R.F.; Renno, A.C.M.; Gazarini, M.L. Metabolites from Marine Sponges and Their Potential to Treat Malarial Protozoan Parasites Infection: A Systematic Review. Mar. Drugs 2021, 19, 134. https://0-doi-org.brum.beds.ac.uk/10.3390/md19030134

AMA Style

Aguiar ACC, Parisi JR, Granito RN, de Sousa LRF, Renno ACM, Gazarini ML. Metabolites from Marine Sponges and Their Potential to Treat Malarial Protozoan Parasites Infection: A Systematic Review. Marine Drugs. 2021; 19(3):134. https://0-doi-org.brum.beds.ac.uk/10.3390/md19030134

Chicago/Turabian Style

Aguiar, Anna Caroline Campos, Julia Risso Parisi, Renata Neves Granito, Lorena Ramos Freitas de Sousa, Ana Cláudia Muniz Renno, and Marcos Leoni Gazarini. 2021. "Metabolites from Marine Sponges and Their Potential to Treat Malarial Protozoan Parasites Infection: A Systematic Review" Marine Drugs 19, no. 3: 134. https://0-doi-org.brum.beds.ac.uk/10.3390/md19030134

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