Next Article in Journal
Magnetic Hydroxyapatite Composite Nanoparticles for Augmented Differentiation of MC3T3-E1 Cells for Bone Tissue Engineering
Next Article in Special Issue
Synthesis and Antimalarial Evaluation of Halogenated Analogues of Thiaplakortone A
Previous Article in Journal
Microwave-Assisted Extraction of Carrageenan from Sarcopeltis skottsbergii
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Promising Antiparasitic Natural and Synthetic Products from Marine Invertebrates and Microorganisms

1
College of Life Sciences, Nanjing Normal University, Nanjing 210046, China
2
School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
*
Author to whom correspondence should be addressed.
Mar. Drugs 2023, 21(2), 84; https://doi.org/10.3390/md21020084
Submission received: 12 January 2023 / Revised: 19 January 2023 / Accepted: 23 January 2023 / Published: 25 January 2023
(This article belongs to the Special Issue Marine Antiparasitic Agents)

Abstract

:
Parasitic diseases still threaten human health. At present, a number of parasites have developed drug resistance, and it is urgent to find new and effective antiparasitic drugs. As a rich source of biological compounds, marine natural products have been increasingly screened as candidates for developing new antiparasitic drugs. The literature related to the study of the antigenic animal activity of marine natural compounds from invertebrates and microorganisms was selected to summarize the research progress of marine compounds and the structure–activity relationship of these compounds in the past five years and to explore the possible sources of potential antiparasitic drugs for parasite treatment.

1. Introduction

Parasitic diseases common in the tropics and subtropics, including malaria, leishmaniasis, trypanosomiasis, and others, still threaten the lives and property of indigenous people [1].
Malaria, which occurs mainly in sub-Saharan Africa [2], is caused by Plasmodium. Anopheles gambiae is the principal vector of the disease in the Afrotropical Region [3]. Plasmodium enters human liver cells via infected female Anopheles and proliferates. Then, merozoites invade red blood cells and further cause disease [4], which is characterized by fever, headache, vomiting, diarrhea, chills, and muscle aches [5]. According to the World Health Organization, an estimated 240 million malaria cases were endemic in 84 countries worldwide in 2021 [6].
Leishmaniasis and trypanosomiasis are neglected tropical diseases (NTDs) that are associated with extreme poverty [7], spread in tropical and subtropical areas in 149 countries, and affect more than 2 billion poor people worldwide [8].
Leishmaniasis is affected by poor nutrition, poor sanitation, a weak immune system, and a lack of preventive measures [9]. This parasite occurs in Asia, Africa, the Americas, and the Mediterranean region. The main genera responsible for this disease are Phlebotomus and Lutzomyia [10]. Sand flies bite an infected animal host and acquires Leishmania, which multiplies in the gut. After 8 to 20 days, they become infectious and spread the disease by biting other hosts [11]. Leishmaniasis includes cutaneous leishmaniasis (CL), visceral leishmaniasis (VL), and mucocutaneous leishmaniasis (MCL). CL is the most common form, while VL is the most severe and is characterized by fever, weight loss, enlargement of the spleen and liver, and anemia [12]. Currently, the only effective treatment for leishmania is pentavalent antimony [10].
Trypanosomiasis includes sleeping sickness and Chagas disease (American trypanosomiasis); sleeping sickness is common in 36 sub-Saharan African countries [13] and is transmitted by blood-sucking tsetse flies. This parasite has two main forms: the slower-progressing form caused by Trypanosoma brucei gambiense and the faster-progressing form caused by Trypanosoma brucei rhodesiense [14]. A prominent feature of African trypanosomiasis is lethargy. T. brucei can circulate freely in the host’s blood and tissue fluids until it reaches the central nervous system, where it is usually fatal. Therefore, therapeutics at this stage must cross the blood–brain barrier [4]. American trypanosomiasis occurs in the Americas (including Mexico, Central, and South America) and is caused by Trypanosoma cruzi, which is transmitted through reduviid bugs [15,16].
Because of the widespread use of drugs, many parasites have developed resistance to treatment. For example, artemisinin-based combination therapy (ACT), which combines artemisinin and quinolines [17], is considered a first-line treatment for Plasmodium falciparum malaria globally [18]. Unfortunately, in the Greater Mekong subregion, such as Cambodia, Thailand, and Myanmar, the efficacy of artemisinin derivatives and ACT partner drugs is decreasing [19,20,21,22]. Additionally, the parasite has resistance to inexpensive drugs such as chloroquine and sulfadoxine/pyrimethamine. Similar situations were also observed with praziquantel for the treatment of schistosomiasis infection [23] and ivermectin for worms [24]. In addition to drug resistance, the efficacy and toxicity of drugs also deserve attention. Benznidazole and nifurtimox, which are used to treat Trypanosoma cruzi infection, are highly toxic to adult patients and have low efficacy [25]. Moreover, although a large number of resources have been invested, no effective vaccine against parasitic diseases has been developed thus far [26]. These reasons are forcing researchers to find new safe and effective antiparasite drugs.
The ocean covers more than 70% of the Earth’s surface area. Plants and animals, approximately 500,000 species in approximately 28 phyla, exist in this environment [27]. Compared with the terrestrial environment, the ocean has much richer biodiversity. The marine environment is more complex, and marine organisms have been in a harsh environment of high salinity, high pressure, lack of oxygen, limited food supply, and lack of photosynthesis for a long time [28]. Some organisms have evolved adaptations that allow them to synthesize toxic compounds or acquire toxic compounds from others. These toxic compounds can help protect marine life from predators [29]. Marine natural products are bioactive metabolites extracted from marine organisms, including marine animals, plants, and microorganisms [30]. Therefore, the ocean is an important source of bioactive compounds. Currently, compounds isolated from marine organisms mainly include terpenoids, alkaloids, polyketones, steroids, peptides, lactones, and so on [27,31], which have effective antibacterial, antifungal, anti-inflammatory, antiviral, antiparasitic, and other bioactivities [32,33].
We searched the Web of Science database from January 2017 to November 2022 for references with the keywords “marine-derived natural antiparasite products” and further screened the relevant research literature on invertebrates and microorganisms. We did not include meetings or review articles. In this review, we also used the following criteria to determine the activity of compounds:
  • When IC50 > 20 μM, the activity of the compounds was low or inactive; when 1 ≤ IC50 ≤ 20 μM, the compounds showed moderate activity. When IC50 < 1 μM, they showed good potent activity [34];
  • When measured in μg/mL, if IC50 > 20 μM, the activity of the compounds was low or inactive; if 3 ≤ IC50 ≤ 10 μg/mL, the compound showed moderate activity. If IC50 < 3 μg/mL, the compound showed good potent activity [35].
We screened 36 studies on the derivatives from invertebrates and microorganisms (Table 1) and six studies on their crude extracts (Table 2). We reviewed the literature on the purification of the derived compounds. Twelve invertebrate marine sponges came from 11 genera: Aplysinella, Dysidea, Fascaplysinopsis, Hyrtios, Ircinia, Pseudoceratina, Monanchora, Mycale, Tedania, and Xestospongia. Five genera, Bebryce, Macrorhynchia, Plumarella, and Sinulari, were included in the seven studies regarding cnidarians. Two genera, Amathia and Orthoscuticella, were involved in two bryozoan studies. For microorganisms, two genera, including Streptomyces and Pseudomonas, were studied in three bacterial studies. Aspergillus, Cochliobolus, Exserohilum, and Paecilomyces were involved in four fungal studies. Nine cyanobacteria studies involved Caldora, Dapis, Leptolyngbya, Okeania, Salileptolyngbya, and Moorea. Finally, we summarized the chemical structures with good potent activity (Figure 1, Figure 2, Figure 3 and Figure 4) and the possible structure–activity relationships.

2. Marine Invertebrate-Derived Antiparasitic Compounds

Invertebrates make up a large part of the literature collected on antiparasitic compounds of marine origin (58.33%). Most of these compounds are alkaloids (including bromotyrosine alkaloids, tryptophan-derived alkaloids, acyclic guanidine alkaloids, etc.), sesquiterpenoids, diterpenoids, sterols, steroids, etc. (Table 1). Invertebrate-derived compounds against P. falciparum have highly effective bioactivity (Figure 1).

2.1. Alkaloid Compounds

Bromopyrrole alkaloids are a field worth exploring for antiparasitic drugs [46]. The bromotyrosine alkaloid bisaprasin (3) extracted from marine sponges was moderately effective against T. cruzi (IC50 = 0.61 µM) [36]. Pseudoceratidine (1) (29) and its derivatives extracted from Tedania brasiliensis have moderate efficacy against P. falciparum, L. infantum, L. amazonensis, and T. cruzi (Table 1). The antiplasmodium activity of this alkaloid is related to the length of the polyamine chain containing basic nitrogen and the presence of bromine atoms on the terminal portion of pyrrole or furan. Moreover, Parra et al. [46] found that pseudoceratidine (1) (29) had additive effects when used in combination with artesunate. Consequently, pseudoceratidine (1) (29) can be used as a promising source of antiplasmodial drugs.
Campos et al. [44] extracted pentacyclic alkaloids (ptilomycalin E, ptilomycalin F, and ptilomycalins G+H (2123)) and acyclic guanidine alkaloids (crambescidin 800 (24) and fromiamycalin (25)) from Monanchora unguiculata sponges, which have extremely high activity against the chloroquine-sensitive 3D7 strain of P. falciparum (IC50 were 0.35, 0.23, 0.46, 0.52, and 0.24 µM, respectively) [82]. The antimalarial activity of pentacyclic alkaloids is related to their five-ring structure. Unguiculin A (20), which has no five-ring structure, has lower antimalarial activity (IC50 = 12.89 µM). Ceratinadin E (19), a new bromotyrosine alkaloid, was isolated from the marine sponge Pseudoceratina by Kurimoto et al. [43] and showed good potent activity against the chloroquine-resistant strain FCR3 (IC50 = 0.77 µg/mL) and multidrug-resistant strain K1 (IC50 = 1.03 µg/mL) of P. falciparum. In 2019, Campos et al. [38] isolated 8-oxo-tryptamine (7) and the mixture of (E) with (Z)-6-bromo-2′-demethyl-3′-N-methylaplysinopsin (8), which showed moderate activity against the P. falciparum 3D7 strain ((IC50 were 8.8 and 8.0 µg/mL, respectively). These two aplysinopsins with antimalarial activity have a double bond between C-8 and C-1′, suggesting that antimalarial activity may be connected to the skeleton of the compounds.
Brominated alkaloids extracted from the bryozoan Amathia lamourouxi showed effective antimalarial activity against the P. falciparum 3D7 strain. Moreover, volutamide F (60) showed a higher selectivity index for the human embryonic kidney cell line HEK293. The antimalarial activity of volutamide H (62) (IC50 = 1.6 µM) was lower than that of volutamide F (60) (IC50 = 0.61 μM) and volutamide G (61) (IC50 = 0.57 µM), indicating that the presence of tertiary amides plays an important role against Plasmodium [55]. Alkaloids (orthoscuticellines A, D, E, 1-ethyl-β-carboline (63, 65, 66, 68)) isolated from Orthoscuticella ventricosa, another bryophyte, also had moderate antimalarial activity, ranging from 12–21 μM (Table 1). Ligand efficiency calculations showed that β-carboline was partly related to the antiplasmodium activity [56].

2.2. Terpenoids, Sesquiterpenoids, and Diterpenoids Compounds

Imperatore et al. [37] obtained the natural sesquiterpenoid quinone avarone (4) and avarol (6) from Dysidea avara sponges. They obtained the semisynthetic thiazinoquinone derivative thiazoavarone (5) by condensation reaction of avarone (4) with subtaurine. Compared with the two natural products, thiazoavarone (5) showed better activity against the chloroquine-resistant strain W2 (IC50 = 0.21 μM) and drug-sensitive strain D10 (IC50 = 0.38 μM) of P. falciparum. In addition, this derivative also had bioactivity against Schistosoma mansoni (IC50 = 5.90 μM). These results suggested that the substituent of the 1,1-dioxo-1,4-thiazine ring played a vital part in bioactivity.
Among the five new furan diterpenes keikipukalides (A–E) (4650) isolated from Plumarella delicatissima, four keikipukalides (B–E) (4750) showed moderate activity against L. donovani (IC50 were 8.5, 8.8, 12, and 8.8 μM, respectively). In addition, the two known compounds pukalide (51) and norditerpenoid (52) ineleganolide that were isolated, also showed good biological activity (IC50 were 1.9 and 4.4 μM, respectively). In particular, these compounds were not toxic to human lung carcinoma cells when they were below 50 μM. [52]. The sesquiterpenoids alcyopterosin V (41) and alcyopterosin E (42) obtained from another cnidarian Alcyonium sp. also had moderate activity against L. donovani (IC50 were 7.0 and 3.1 μM, respectively) [49].

2.3. Steroids and Sterols Compounds

Chlorinated steroid (3) (53), 24-methylenecholestane-3β-5α,6β-triol-6-monoacetate (55), and dibromoditerpene compounds pinnaterpene C (54) extracted from Sinularia brassica at 50 μM showed positive effects. The inhibitory effects of L. donovaniamastigote on amastigotes were 58.7%, 54.7%, and 74.3%, respectively. In addition, the three compounds showed little toxicity to THP-1 cells at these concentrations [53].
Two sterol compounds, kaimanol (39) and saringosterol (40), were extracted from the sponge Xestospongia sp. The antimalarial activity of kaimanol (39) was lower than that of saringosterol (40), suggesting that benzoyl may reduce the activity in the sterol structure [47]. The terpenoids extracted from the sponge Hyrtios erectus and the cnidarian Bebryce grandis showed moderate or greater activity against chloroquine-resistant Dd2 strains [39,50]. It is worth noting that both compounds extracted from B. grandis act on the life cycle of Plasmodium parasites. They found that the addition of nitenin (44) before the ring transition to the early trophozoite stage inhibited the maturation of the parasites. Bebrycin A (43) prevented the parasite from maturing. Among the clinical antimalarial drugs, only artemisinin is active against the merozoite of Plasmodium [83]. Consequently, Wright et al. [50] noted that it might be possible to develop new artemisinin combination therapy partner drugs based on the properties of these two terpenoids.

2.4. Other Compounds

Sala et al. extracted several nitrile-containing polyacetylene secondary metabolites from the sponge Mycale sp.SS5; however, only albanitrile A (26) showed moderate bioactivity against Giardia duodenalis (IC50 =12 μM). The lower bioactivity of albanitrile B (27) than A 26 also suggested that the activity of antigenic animals depended on the chain length of the alkyl group [45].
Notably, isololiolide (45), which was extracted in the sponge Macrorhynchia philippina, had certain effects on T. cruzi trypomastigotes and amastigotes (IC50 = 31.9 and 40.4 μM, respectively). Lima et al. [51] studied the lethal mechanism of this compound and suggested that isololiolide (45) may cause damage to plasma membrane integrity and depolarization of mitochondrial membrane potential.

3. Marine Microorganisms-Derived Antiparasitic Compounds

3.1. Steroids and Sterols Compounds

Previous studies have shown that polyketones, alkaloids, fatty acids, terpenes, and other compounds isolated from marine bacteria have potential antibacterial, antifungal, and antiparasitic activities [74,84,85]. Salinivibrio and Streptomyces from Actinomycetes are Gram-positive bacteria [74], while Pseudomonas from Proteobacteria is Gram-negative bacteria [86]. The active compounds extracted from these bacteria mainly include alkaloids and quinoline (Table 1) (Figure 2).
Marinopyrrole A (70), an alkaloid compound found in marine Streptomyces sp., has strong antibacterial activity against methicillin-resistant Staphylococcus aureus [87]. Martens et al. [58] explored the activity of this compound against Toxoplasma gondii. In in vitro experiments, marinopyrrole A (70) showed potent inhibitory activity at 0.31 µM against Toxoplasma gondii tachyzoites. However, the anti-toxoplasma effect was inhibited when more than 20% bovine calf serum was added to the liquid medium. Based on compound (70), they obtained three analogs, RL002, RL003, and RL125 (7173), which showed 3.6- to 6.8-fold increased efficacy against toxoplasmosis (P < 0.001, Student’s paired t-test) and decreased serum sensitivity. RL003 (72), the most inhibitory analog, is highly active against cysts in vitro (IC50 = 0.245 μM). Hence, further in vivo chronic studies are needed to assess the potential antiparasitic activity of RL003 (72) in the host. Another alkaloid, staurosporine (69), isolated from Streptomyces sp. PBLC04 can kill the trophozoites of Acanthamoeba (IC50 = 0.265 µg/mL) [57]. The cysts of Acanthamoeba allow the parasite to cope with harsh environments such as a lack of nutrients, high temperatures, and high osmotic pressure, so Acanthamoeba, in this stage is highly resistant [88,89]. Notably, taurosporine also showed good potent inhibition against cysts (IC50 = 0.771 μg/mL). The protein kinase family is generally considered to be the main target of staurosporine (69) [90]. Acanthamoeba is rich in known kinase genes, which may explain the high activity of this compound against Acanthamoeba.
Martinez-Luis et al. [59] isolated five hydroxyquinoline compounds from Pseudomonas aeruginosa, among which three compounds had good antiparasitic effects: 3-heptyl-3-hydroxy-1,2,3,4-tetrahydroquinoline-2.4-dione (2), 2-heptyl-4-hydroxyquinoline (3), and 2-nonyl-4-hydroxyquinoline (4) (7476). These three compounds showed moderate and greater antimalarial activity against the chloroquine-resistant strain W2 of P. falciparum (IC50 = 3.47, 2.57, and 2.79 µg/mL, respectively). Compounds (3) (75) (IC50 = 3.66 µg/mL) and (4) (76) (IC50 = 3.99 µg/mL) also showed resistance to Trypanosoma cruzi. In addition, this study also found that the corresponding tautomers of compounds (3) (75) and (4) (76) showed strong activity against the chloroquine-sensitive D6 strain and chloroquine-resistant Plasmodium falciparum W2 strain [91], indicating that the hydroxyquinoline compounds maintained antimalarial activity independently of their tautomers [59].

3.2. Marine Fungi

Endophytes are microfungi that reside in the internal tissues of plants without causing any immediate obvious negative effects [92,93]. Marine invertebrates, algae endophytes, or fungi found in marine sediments are also rich sources of bioactive natural products [94,95,96]. In the four studies on marine fungi from 2017 to 2022, the natural products were mostly polyketones and alkaloids (Table 1).
The compound harzialactone A (117) was extracted from Paecilomyces sp.7A22, a marine fungus isolated from sea squirts. This known polyketone compound has been isolated from Trichoderma harzianum, an endophytic fungus of the sponge Halichondria okadai [97]. Braun et al. [72] investigated the antiparasitic activity of this polyketone compound.
Harzialactone A (117) had the ability to overcome the transmembrane barriers to reach the macrophage phagolysosome, where amastigotes grow, and showed moderate activity against L. amazonensis promastigotes (IC50 = 5.25 μg/mL). In addition, another polyketone isolated from Cochliobolus lunatus by Xu et al. [70] (Derivatives 103111, Acyl derivatives 6971) showed moderate antiplasmodial activity (Table 1). The structure–activity relationships showed that biphenyl substituents at C-2, acetone at C-5′ and C-6′, and triple or quadruple substitution of acyl groups increased antiplasmodium activity.
Isocoumarins (1) (112) and isocoumarins (3) (113) extracted from Exserohilum sp. (CHNSCLM-0008) fungus isolated from button coral Palythoa haddoni by Coronado et al. [71] showed moderate activity against chloroquine-sensitive HB3 strains of Plasmodium falciparum (IC50 values were 1.13 and 11.7 μM, respectively). Semisynthetic derivatives were obtained by changing the substituents of the aromatic ring and adipose chain to explore the structure–activity relationship of the compounds. The newly synthesized compounds, derivatives 114116 (Figure 3), showed good potent activity against P. falciparum (IC50 values were 0.77, 0.38, and 2.58 μM, respectively). Among them, derivative 115 was an accidental ring-opening product obtained during the demethylation process, which had a very strong antimalarial effect. Moreover, structure–activity analysis demonstrated that the configuration of methoxy groups and 3R, 4R, and 10S was necessary for antimalarial activity, and the lipid solubility of the side chain could help improve antimalarial activity. On the one hand, derivative 115 can inhibit heme polymerization and reduce mitochondrial membrane potential in the parasite; on the other hand, they can inhibit DNA gyrase enzymes and thus inhibit DNA replication. In conclusion, this study suggested that derivatives 115 may be a potential lead agent for malaria treatment.
Bunbamrung et al. [69] isolated the fungus Aspergillus terreus BCC51799 from decaying wood samples in the ocean and extracted new natural products from this fungus. Among them, the alkaloid astechrome (99) (Figure 3) showed strong antimalarial activity (IC50 = 0.94 μM) (Table 1).

4. Cyanophyta

Cyanobacteria, also known as blue-green algae because of the presence of phycocyanin and chlorophyll, are the only prokaryotes that can produce oxygen through photosynthesis [98]. Some secondary metabolites in marine cyanobacteria have good activity and are considered lead compounds for drugs [99]. Some of these compounds are antimicrobial peptides, and cyanobacterial peptides can be divided into linear peptides, depsipeptides, and cyclic peptides according to their structure [98].

4.1. Linear Peptides

Ozaki et al. [66] isolated the linear peptides mabuniamide (1) (90) and stereoisomer 2 (91), from Okeania sp., which showed moderate activity (IC50 were both 1.4 μM) against the chloroquine-sensitive 3D7 strain of P. falciparum. In 2020, Iwasaki et al. [65] isolated another linear peptide, ikoamide (1) (89) (Figure 4), from Okeania and discovered strong activity against the P. falciparum 3D7 strain. Kurisawa et al. [62] isolated three linear peptides from the cyanobacteria Dapis sp. However, only iheyamides A (86) showed moderate activity against T. b. rhodesiense (IC50 = 1.5 μM) and T. b. brucei (IC50 = 1.5 μM). Structure–activity analysis proved that the C-terminal pyrrolinone moiety was vital for antiparasitic activity. The team then isolated the C-terminal part of iheyamide A (1) to obtain iheyanone (2), which also showed some activity against T. b. rhodesiense. To further clarify the structure–activity relationship of this compound, Iswasaki et al. [61] synthesized a variety of compounds with different peptide chain lengths and found that longer lengths of the peptide chain were more effective in inhibiting the growth of Trypanosoma. Hoshinoamide C (77) (Figure 4), a natural product discovered by Iswasaki et al. [60] in Caldora penicillate, also had some effective activity against P. falciparum (IC50 = 0.96 μM) and T. b. rhodesiense (IC50 = 2.9 μM). Finally, the configuration at C-43 (Figure 4) did not affect antiparasitic activity when used to synthesize two possible isomers of hoshinoamide C (77,78). The linear peptide Kinenzoline (1) (92) isolated from Salileptolyngbya sp. showed moderate activity (IC50 = 5.0 μM) against the IL-1501 strain of T. b. rhodesiense. Kurisawa et al. [67] also identified a synthetic pathway for kinenzoline (1) (92) and showed that neither natural nor synthetic Kinenzoline (1) (92,93) was toxic to WI-38 cells.

4.2. Cyclic Peptides

Cyclic peptides are likely to mimic peptide substrates or ligands of endogenous proteins (such as enzymes or receptors). Therefore, they are often considered “privileged structures” of bioactivity [100,101]. Motobamide (1) (87), a cyclic decapeptide isolated from Leptolyngbya sp., inhibited the growth of T. b. rhodesiense. Almaliti et al. [68] explored the relationship between the structure and activity of several dudawalamides 9497, which are cyclic depsipeptides isolated from the cyanobacterium Moorea producens. The results indicated that the activity of Dhoya natural products was affected by the structure of the configuration and order of residues. Keller et al. [64] isolated Palstimolide A (88), a polyhydroxy macrolide compound from cyanobacteria, with an IC50 of 0.1725 μM against the Dd2 strain of P. falciparum, showing very high antiplasmodium activity. This compound also showed moderate activity against the promastigotes phase of L. donovani (IC50 = 4.67μM).

5. Conclusions

Our review of the literature published in the last five years found that sponges are still the major source of marine-derived compounds. Marine sponge-derived compounds have shown excellent activity against Plasmodium falciparum in in vitro studies. A total of 40 natural products or synthetic compounds from marine sponges were included in this study, among which 12 compounds had good potent activity. These sponges belong to Xestospongia, Dyside, Hyrtios, Pseudoceratina, and Monanchora. Approximately 17 compounds were derived from cnidarians, and one compound from Bebryce showed good potent activity. In addition, 11 compounds from bryophytes and two high bioactivity compounds were derived from Amathia. A total of 8 compounds from marine bacteria were collected, and seven compounds with effective bioactivity were extracted from Streptomyces, Salinivibrio, and Pseudomonas. Twenty compounds were identified from marine fungi, with three highly active compounds from Exserohilum and Aspergillus. Finally, 21 were derived from Cyanophyta, with 4 highly active compounds from Caldora, Okeania, and Leptolyngbya.
Naturally derived or semisynthetic molecular analogs can be developed by structure–activity relationship (SAR) analysis and tend to have higher bioactivity and less toxicity [102]. In addition, it has been shown that coupling natural products with nanomaterials may enhance the activity of compounds. Walvekar et al. used silver nanoparticles coupled with extracts of Kappaphycus alvarezii, which enhanced anti-acanthamoebic activity [103].
Although the association between the structure of some compounds and their antiparasitic activity has been explored through SAR, the molecular targets and mechanisms of some compound molecules have not been clarified [104]. At present, a large number of promising active antiparasitic compounds have been discovered, but translating them into a drug for clinical use still faces many difficulties: (1) if the purified antiparasitic product is not chemically synthesized, clinical studies and mass production of those compounds often require more biomass than discovering new compounds and (2) if the compounds can be obtained through chemical synthesis, it is also worth considering how to reduce the synthesis steps and reduce the cost of chemical synthesis.

Author Contributions

L.Z. conceived the ideas and methodology. M.Z., Q.Z. (Qunde Zhang), Q.Z. (Qinrong Zhang) and X.C. collected the data. M.Z. and L.Z. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

Financial support was provided by the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD).

Data Availability Statement

Not applicable.

Acknowledgments

We thank the team members for the help during data collection.

Conflicts of Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

  1. Mostafa, O.; Al-Shehri, M.; Moustafa, M. Promising antiparasitic agents from marine sponges. Saudi J. Biol. Sci. 2021, 29, 217–227. [Google Scholar] [CrossRef]
  2. Greenwood, B.; Mutabingwa, T. Malaria in 2002. Nature 2002, 415, 670. [Google Scholar] [CrossRef] [PubMed]
  3. della Torre, A.; Tu, Z.; Petrarca, V. On the distribution and genetic differentiation of Anopheles gambiae ss molecular forms. Insect Biochem. Mol. Biol. 2005, 35, 755–769. [Google Scholar] [CrossRef]
  4. Horn, D.; Duraisingh, M.T. Antiparasitic chemotherapy–from genomes to mechanisms. Annu. Rev. Pharmacol. Toxicol. 2014, 54, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Seo, E.B.; du Plessis, L.H.; Viljoen, J.M. Solidification of Self-Emulsifying Drug Delivery Systems as a Novel Approach to the Management of Uncomplicated Malaria. Pharmaceuticals 2022, 15, 120. [Google Scholar] [CrossRef] [PubMed]
  6. World Health Organization. World Malaria Report 2022; World Health Organization: Geneva, Switzerland, 2022. [Google Scholar]
  7. Hotez, P.J.; Aksoy, S.; Brindley, P.J.; Kamhawi, S. What Constitutes a Neglected Tropical Disease? Public Library of Science: San Francisco, CA USA, 2020; Volume 14, p. e0008001. [Google Scholar]
  8. Engels, D.; Zhou, X.-N. Neglected tropical diseases: An effective global response to local poverty-related disease priorities. Infect. Dis. Poverty 2020, 9, 1–9. [Google Scholar] [CrossRef] [Green Version]
  9. Pinheiro, A.C.; de Souza, M.V.N. Current leishmaniasis drug discovery. RSC Med. Chem. 2022, 13, 1029–1043. [Google Scholar] [CrossRef]
  10. Torres-Guerrero, E.; Quintanilla-Cedillo, M.R.; Ruiz-Esmenjaud, J.; Arenas, R. Leishmaniasis: A review. F1000Research 2017, 6, 22–25. [Google Scholar] [CrossRef]
  11. Desjeux, P. Leishmaniasis: Current situation and new perspectives. Comp. Immunol. Microbiol. Infect. Dis. 2004, 27, 305–318. [Google Scholar] [CrossRef]
  12. Pal, M.; Gutama, K.P.; Steinmetz, C.H.; Dave, P. Leishmaniasis: An Emerging and Re-emerging Disease of Global Public Health Concern. Am. J. Infect. Dis. 2022, 10, 22–25. [Google Scholar] [CrossRef]
  13. Shah, V.V.; Patel, V.M.; Vyas, P. Human African Trypanosomiasis–A rare case report from India. Indian J. Med. Microbiol. 2022, 40, 169–171. [Google Scholar] [CrossRef] [PubMed]
  14. Simarro, P.P.; Cecchi, G.; Paone, M.; Franco, J.R.; Diarra, A.; Ruiz, J.A.; Fèvre, E.M.; Courtin, F.; Mattioli, R.C.; Jannin, J.G. The Atlas of human African trypanosomiasis: A contribution to global mapping of neglected tropical diseases. Int. J. Health Geogr. 2010, 9, 1–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Chimelli, L.; Scaravilli, F. Trypanosomiasis. Brain Pathol. 1997, 7, 599–611. [Google Scholar] [CrossRef] [PubMed]
  16. Montgomery, S.P.; Parise, M.E.; Dotson, E.M.; Bialek, S.R. What do we know about Chagas disease in the United States? Am. J. Trop. Med. Hyg. 2016, 95, 1225. [Google Scholar] [CrossRef] [Green Version]
  17. Beteck, R.M.; Smit, F.J.; Haynes, R.K.; N’Da, D.D. Recent progress in the development of anti-malarial quinolones. Malar. J. 2014, 13, 1–10. [Google Scholar] [CrossRef] [Green Version]
  18. Ouji, M.; Augereau, J.-M.; Paloque, L.; Benoit-Vical, F. Plasmodium falciparum resistance to artemisinin-based combination therapies: A sword of Damocles in the path toward malaria elimination. Parasite 2018, 25, 24. [Google Scholar] [CrossRef] [Green Version]
  19. Thu, A.M.; Phyo, A.P.; Landier, J.; Parker, D.M.; Nosten, F.H. Combating multidrug-resistant Plasmodium falciparum malaria. FEBS J. 2017, 284, 2569–2578. [Google Scholar] [CrossRef] [Green Version]
  20. Duru, V.; Khim, N.; Leang, R.; Kim, S.; Domergue, A.; Kloeung, N.; Ke, S.; Chy, S.; Eam, R.; Khean, C. Plasmodium falciparum dihydroartemisinin-piperaquine failures in Cambodia are associated with mutant K13 parasites presenting high survival rates in novel piperaquine in vitro assays: Retrospective and prospective investigations. BMC Med. 2015, 13, 305. [Google Scholar] [CrossRef] [Green Version]
  21. Leang, R.; Taylor, W.R.; Bouth, D.M.; Song, L.; Tarning, J.; Char, M.C.; Kim, S.; Witkowski, B.; Duru, V.; Domergue, A. Evidence of Plasmodium falciparum malaria multidrug resistance to artemisinin and piperaquine in western Cambodia: Dihydroartemisinin-piperaquine open-label multicenter clinical assessment. Antimicrob. Agents Chemother. 2015, 59, 4719–4726. [Google Scholar] [CrossRef] [Green Version]
  22. AlKadi, H.O. Antimalarial drug toxicity: A review. Chemotherapy 2007, 53, 385–391. [Google Scholar] [CrossRef]
  23. da Silva, V.B.R.; Campos, B.R.K.L.; de Oliveira, J.F.; Decout, J.-L.; de Lima, M.d.C.A. Medicinal chemistry of antischistosomal drugs: Praziquantel and oxamniquine. Bioorg. Med. Chem. 2017, 25, 3259–3277. [Google Scholar] [CrossRef]
  24. Lespine, A.; Ménez, C.; Bourguinat, C.; Prichard, R.K. P-glycoproteins and other multidrug resistance transporters in the pharmacology of anthelmintics: Prospects for reversing transport-dependent anthelmintic resistance. Int. J. Parasitol. Drugs Drug Resist. 2012, 2, 58–75. [Google Scholar] [CrossRef]
  25. Bermudez, J.; Davies, C.; Simonazzi, A.; Real, J.P.; Palma, S. Current drug therapy and pharmaceutical challenges for Chagas disease. Acta Trop. 2016, 156, 1–16. [Google Scholar] [CrossRef] [PubMed]
  26. Versteeg, L.; Almutairi, M.M.; Hotez, P.J.; Pollet, J. Enlisting the mRNA vaccine platform to combat parasitic infections. Vaccines 2019, 7, 122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Lu, W.-Y.; Li, H.-J.; Li, Q.-Y.; Wu, Y.-C. Application of marine natural products in drug research. Bioorg. Med. Chem. 2021, 35, 116058. [Google Scholar] [CrossRef] [PubMed]
  28. Danovaro, R.; Corinaldesi, C.; Dell’Anno, A.; Snelgrove, P.V. The deep-sea under global change. Curr. Biol. 2017, 27, R461–R465. [Google Scholar] [CrossRef] [Green Version]
  29. Thomas, T.R.A.; Kavlekar, D.P.; LokaBharathi, P.A. Marine drugs from sponge-microbe association—A review. Mar. Drugs 2010, 8, 1417–1468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Nweze, J.A.; Mbaoji, F.N.; Li, Y.-M.; Yang, L.-Y.; Huang, S.-S.; Chigor, V.N.; Eze, E.A.; Pan, L.-X.; Zhang, T.; Yang, D.-F. Potentials of marine natural products against malaria, leishmaniasis, and trypanosomiasis parasites: A review of recent articles. Infect. Dis. Poverty 2021, 10, 1–19. [Google Scholar] [CrossRef]
  31. Lenz, K.D.; Klosterman, K.E.; Mukundan, H.; Kubicek-Sutherland, J.Z. Macrolides: From toxins to therapeutics. Toxins 2021, 13, 347. [Google Scholar] [CrossRef]
  32. Rushdi, M.I.; Abdel-Rahman, I.A.; Attia, E.Z.; Abdelraheem, W.M.; Saber, H.; Madkour, H.A.; Amin, E.; Hassan, H.M.; Abdelmohsen, U.R. A review on the diversity, chemical and pharmacological potential of the green algae genus Caulerpa. S. Afr. J. Bot. 2020, 132, 226–241. [Google Scholar] [CrossRef]
  33. Nweze, J.A.; Mbaoji, F.N.; Huang, G.; Li, Y.; Yang, L.; Zhang, Y.; Huang, S.; Pan, L.; Yang, D. Antibiotics development and the potentials of marine-derived compounds to stem the tide of multidrug-resistant pathogenic bacteria, fungi, and protozoa. Mar. Drugs 2020, 18, 145. [Google Scholar] [CrossRef] [Green Version]
  34. Murillo, T.; Schneider, D.; Fichtel, C.; Daniel, R. Dietary shifts and social interactions drive temporal fluctuations of the gut microbiome from wild redfronted lemurs. ISME Commun. 2022, 2, 1–11. [Google Scholar] [CrossRef]
  35. Ioset, J.-R.; Brun, R.; Wenzler, T.; Kaiser, M.; Yardley, V. Drug Screening for Kinetoplastid Diseases: A Training Manual for Screening in Neglected Diseases. DNDi 2009, 1–74. [Google Scholar]
  36. Oluwabusola, E.T.; Tabudravu, J.N.; Al Maqbali, K.S.; Annang, F.; Pérez-Moreno, G.; Reyes, F.; Jaspars, M. Antiparasitic activity of bromotyrosine alkaloids and new analogues isolated from the Fijian marine sponge Aplysinella rhax. Chem. Biodivers. 2020, 17, e2000335. [Google Scholar] [CrossRef] [PubMed]
  37. Imperatore, C.; Gimmelli, R.; Persico, M.; Casertano, M.; Guidi, A.; Saccoccia, F.; Ruberti, G.; Luciano, P.; Aiello, A.; Parapini, S. Investigating the antiparasitic potential of the marine sesquiterpene avarone, its reduced form avarol, and the novel semisynthetic thiazinoquinone analogue thiazoavarone. Mar. Drugs 2020, 18, 112. [Google Scholar] [CrossRef] [Green Version]
  38. Campos, P.-E.; Pichon, E.; Moriou, C.; Clerc, P.; Trepos, 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]
  39. Ju, E.; Latif, A.; Kong, C.-S.; Seo, Y.; Lee, Y.-J.; Dalal, S.R.; Cassera, M.B.; Kingston, D.G. Antimalarial activity of the isolates from the marine sponge Hyrtios erectus against the chloroquine-resistant Dd2 strain of Plasmodium falciparum. Z. Für Nat. C 2018, 73, 397–400. [Google Scholar] [CrossRef]
  40. Shady, N.H.; Fouad, M.A.; Ahmed, S.; Pimentel-Elardo, S.M.; Nodwell, J.R.; Kamel, M.S.; Abdelmohsen, U.R. A new antitrypanosomal alkaloid from the Red Sea marine sponge Hyrtios sp. J. Antibiot. 2018, 71, 1036–1039. [Google Scholar] [CrossRef]
  41. Chianese, G.; Silber, J.; Luciano, P.; Merten, C.; Erpenbeck, D.; Topaloglu, B.L.; Kaiser, M.; Tasdemir, D. Antiprotozoal linear furanosesterterpenoids from the marine sponge Ircinia oros. J. Nat. Prod. 2017, 80, 2566–2571. [Google Scholar] [CrossRef]
  42. Majer, T.; Bhattarai, K.; Straetener, J.; Pohlmann, J.; Cahill, P.; Zimmermann, M.O.; Hübner, M.P.; Kaiser, M.; Svenson, J.; Schindler, M. Discovery of Ircinianin Lactones B and C—Two New Cyclic Sesterterpenes from the Marine Sponge Ircinia wistarii. Mar. Drugs 2022, 20, 532. [Google Scholar] [CrossRef]
  43. Kurimoto, S.-i.; Ohno, T.; Hokari, R.; Ishiyama, A.; Iwatsuki, M.; Ōmura, S.; Kobayashi, J.I.; Kubota, T. Ceratinadins E and F, new bromotyrosine alkaloids from an Okinawan marine sponge Pseudoceratina sp. Mar. Drugs 2018, 16, 463. [Google Scholar] [CrossRef] [Green Version]
  44. 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, 1404–1410. [Google Scholar] [CrossRef] [PubMed]
  45. Sala, S.; Fromont, J.; Gomez, O.; Vuong, D.; Lacey, E.; Flematti, G.R. Albanitriles A–G: Antiprotozoal Polyacetylene Nitriles from a Mycale Marine Sponge. J. Nat. Prod. 2019, 82, 3450–3455. [Google Scholar] [CrossRef] [PubMed]
  46. Parra, L.L.; Bertonha, A.F.; Severo, I.R.; Aguiar, A.C.; de Souza, G.E.; Oliva, G.; Guido, R.V.; Grazzia, N.; Costa, T.R.; Miguel, D.C. Isolation, derivative synthesis, and structure–activity relationships of antiparasitic bromopyrrole alkaloids from the marine sponge Tedania brasiliensis. J. Nat. Prod. 2018, 81, 188–202. [Google Scholar] [CrossRef] [PubMed]
  47. Murtihapsari, M.; Salam, S.; Kurnia, D.; Darwati, D.; Kadarusman, K.; Abdullah, F.F.; Herlina, T.; Husna, M.H.; Awang, K.; Shiono, Y. A new antiplasmodial sterol from Indonesian marine sponge, Xestospongia sp. Nat. Prod. Res. 2021, 35, 937–944. [Google Scholar] [CrossRef]
  48. Fröhlich, T.; Çapcı Karagöz, A.; Reiter, C.; Tsogoeva, S.B. Artemisinin-derived dimers: Potent antimalarial and anticancer agents. J. Med. Chem. 2016, 59, 7360–7388. [Google Scholar] [CrossRef] [PubMed]
  49. Limon, A.-C.D.; Patabendige, H.M.; Azhari, A.; Sun, X.; Kyle, D.E.; Wilson, N.G.; Baker, B.J. Chemistry and Bioactivity of the Deep-Water Antarctic Octocoral Alcyonium sp. Mar. Drugs 2022, 20, 576. [Google Scholar] [CrossRef]
  50. Wright, A.E.; Collins, J.E.; Roberts, B.; Roberts, J.C.; Winder, P.L.; Reed, J.K.; Diaz, M.C.; Pomponi, S.A.; Chakrabarti, D. Antiplasmodial compounds from deep-water marine invertebrates. Mar. Drugs 2021, 19, 179. [Google Scholar] [CrossRef]
  51. Lima, M.L.; Romanelli, M.M.; Borborema, S.E.; Johns, D.M.; Migotto, A.E.; Lago, J.H.G.; Tempone, A.G. Antitrypanosomal activity of isololiolide isolated from the marine hydroid Macrorhynchia philippina (Cnidaria, Hydrozoa). Bioorg. Chem. 2019, 89, 103002. [Google Scholar] [CrossRef]
  52. Thomas, S.A.; Von Salm, J.L.; Clark, S.; Ferlita, S.; Nemani, P.; Azhari, A.; Rice, C.A.; Wilson, N.G.; Kyle, D.E.; Baker, B.J. Keikipukalides, furanocembrane diterpenes from the Antarctic deep sea octocoral Plumarella delicatissima. J. Nat. Prod. 2018, 81, 117–123. [Google Scholar] [CrossRef] [Green Version]
  53. Pham, G.N.; Kang, D.Y.; Kim, M.J.; Han, S.J.; Lee, J.H.; Na, M. Isolation of sesquiterpenoids and steroids from the soft coral sinularia brassica and determination of their absolute configuration. Mar. Drugs 2021, 19, 523. [Google Scholar] [CrossRef]
  54. Qin, G.-F.; Tang, X.-L.; Sun, Y.-T.; Luo, X.-C.; Zhang, J.; Van Ofwegen, L.; Sung, P.-J.; Li, P.-L.; Li, G.-Q. Terpenoids from the soft coral Sinularia sp. collected in Yongxing Island. Mar. Drugs 2018, 16, 127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Kleks, G.; Holland, D.C.; Kennedy, E.K.; Avery, V.M.; Carroll, A.R. Antiplasmodial alkaloids from the Australian bryozoan Amathia lamourouxi. J. Nat. Prod. 2020, 83, 3435–3444. [Google Scholar] [CrossRef] [PubMed]
  56. Kleks, G.; Duffy, S.; Lucantoni, L.; Avery, V.M.; Carroll, A.R. Orthoscuticellines A–E, β-Carboline Alkaloids from the Bryozoan Orthoscuticella ventricosa Collected in Australia. J. Nat. Prod. 2020, 83, 422–428. [Google Scholar] [CrossRef]
  57. Cartuche, L.; Sifaoui, I.; Cruz, D.; Reyes-Batlle, M.; López-Arencibia, A.; Javier Fernández, J.; Díaz-Marrero, A.R.; Piñero, J.E.; Lorenzo-Morales, J. Staurosporine from Streptomyces sanyensis activates programmed cell death in Acanthamoeba via the mitochondrial pathway and presents low in vitro cytotoxicity levels in a macrophage cell line. Sci. Rep. 2019, 9, 11651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Martens, M.C.; Liu, Y.; Sanford, A.G.; Wallick, A.I.; Warner, R.C.; Li, R.; Davis, P.H. Analogs of Marinopyrrole A Show Enhancement to Observed In Vitro Potency against Acute Toxoplasma gondii Infection. Antimicrob. Agents Chemother. 2022, 66, e00794-21. [Google Scholar] [CrossRef]
  59. Martínez-Luis, S.; Cherigo, L.; Spadafora, C.; Gutiérrez, M. Antiparasitic Compounds from the Panamanian Marine Bacterium Pseudomonas aeruginosa. Nat. Prod. Commun. 2019, 14, 1934578X1901400109. [Google Scholar] [CrossRef]
  60. Iwasaki, A.; Ohtomo, K.; Kurisawa, N.; Shiota, I.; Rahmawati, Y.; Jeelani, G.; Nozaki, T.; Suenaga, K. Isolation, structure determination, and total synthesis of hoshinoamide c, an antiparasitic lipopeptide from the marine cyanobacterium Caldora penicillata. J. Nat. Prod. 2020, 84, 126–135. [Google Scholar] [CrossRef]
  61. Iwasaki, A.; Teranuma, K.; Kurisawa, N.; Rahmawati, Y.; Jeelani, G.; Nozaki, T.; Gerwick, W.H.; Suenaga, K. First Total Synthesis and Structure–Activity Relationship of Iheyamide A, an Antitrypanosomal Linear Peptide Isolated from a Dapis sp. Marine Cyanobacterium. J. Nat. Prod. 2021, 84, 2587–2593. [Google Scholar] [CrossRef]
  62. Kurisawa, N.; Iwasaki, A.; Jeelani, G.; Nozaki, T.; Suenaga, K. Iheyamides A–C, antitrypanosomal linear peptides isolated from a marine Dapis sp. cyanobacterium. J. Nat. Prod. 2020, 83, 1684–1690. [Google Scholar] [CrossRef]
  63. Takahashi, H.; Iwasaki, A.; Kurisawa, N.; Suzuki, R.; Jeelani, G.; Matsubara, T.; Sato, T.; Nozaki, T.; Suenaga, K. Motobamide, an antitrypanosomal cyclic peptide from a Leptolyngbya sp. marine cyanobacterium. J. Nat. Prod. 2021, 84, 1649–1655. [Google Scholar] [CrossRef]
  64. Keller, L.; Siqueira-Neto, J.L.; Souza, J.M.; Eribez, K.; LaMonte, G.M.; Smith, J.E.; Gerwick, W.H. Palstimolide A: A complex polyhydroxy macrolide with antiparasitic activity. Molecules 2020, 25, 1604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Iwasaki, K.; Iwasaki, A.; Sumimoto, S.; Matsubara, T.; Sato, T.; Nozaki, T.; Saito-Nakano, Y.; Suenaga, K. Ikoamide, an antimalarial lipopeptide from an Okeania sp. marine cyanobacterium. J. Nat. Prod. 2020, 83, 481–488. [Google Scholar] [CrossRef] [PubMed]
  66. Ozaki, K.; Iwasaki, A.; Sezawa, D.; Fujimura, H.; Nozaki, T.; Saito-Nakano, Y.; Suenaga, K.; Teruya, T. Isolation and total synthesis of mabuniamide, a lipopeptide from an Okeania sp. marine cyanobacterium. J. Nat. Prod. 2019, 82, 2907–2915. [Google Scholar] [CrossRef]
  67. Kurisawa, N.; Otomo, K.; Iwasaki, A.; Jeelani, G.; Nozaki, T.; Suenaga, K. Isolation and Total Synthesis of Kinenzoline, an Antitrypanosomal Linear Depsipeptide Isolated from a Marine Salileptolyngbya sp. Cyanobacterium. J. Org. Chem. 2021, 86, 12528–12536. [Google Scholar] [CrossRef] [PubMed]
  68. Almaliti, J.; Malloy, K.L.; Glukhov, E.; Spadafora, C.; Gutiérrez, M.; Gerwick, W.H. Dudawalamides A–D, antiparasitic cyclic depsipeptides from the marine cyanobacterium Moorea producens. J. Nat. Prod. 2017, 80, 1827–1836. [Google Scholar] [CrossRef]
  69. Bunbamrung, N.; Intaraudom, C.; Dramae, A.; Komwijit, S.; Laorob, T.; Khamsaeng, S.; Pittayakhajonwut, P. Antimicrobial, antimalarial and anticholinesterase substances from the marine-derived fungus Aspergillus terreus BCC51799. Tetrahedron 2020, 76, 131496. [Google Scholar] [CrossRef]
  70. Xu, W.-F.; Wu, N.-N.; Wu, Y.-W.; Qi, Y.-X.; Wei, M.-Y.; Pineda, L.M.; Ng, M.G.; Spadafora, C.; Zheng, J.-Y.; Lu, L. Structure modification, antialgal, antiplasmodial, and toxic evaluations of a series of new marine-derived 14-membered resorcylic acid lactone derivatives. Mar. Life Sci. Technol. 2022, 4, 88–97. [Google Scholar] [CrossRef]
  71. Coronado, L.; Zhang, X.-Q.; Dorta, D.; Escala, N.; Pineda, L.M.; Ng, M.G.; Del Olmo, E.; Wang, C.-Y.; Gu, Y.-C.; Shao, C.-L. Semisynthesis, antiplasmodial activity, and mechanism of action studies of isocoumarin derivatives. J. Nat. Prod. 2021, 84, 1434–1441. [Google Scholar] [CrossRef]
  72. Braun, G.H.; Ramos, H.P.; Candido, A.C.; Pedroso, R.C.; Siqueira, K.A.; Soares, M.A.; Dias, G.M.; Magalhães, L.G.; Ambrósio, S.R.; Januário, A.H. Evaluation of antileishmanial activity of harzialactone a isolated from the marine-derived fungus Paecilomyces sp. Nat. Prod. Res. 2021, 35, 1644–1647. [Google Scholar] [CrossRef]
  73. Morales-Landa, J.L.; Lazcano-Pérez, F.; Cedillo-Rivera, R.; Sánchez-Rodríguez, J. Ultrastructure and molecular toxicological effects of the Coronate Scyphomedusa Linuche unguiculata Venom on Giardia duodenalis. Biologia 2021, 76, 1033–1039. [Google Scholar] [CrossRef]
  74. Shamikh, Y.I.; El Shamy, A.A.; Gaber, Y.; Abdelmohsen, U.R.; Madkour, H.A.; Horn, H.; Hassan, H.M.; Elmaidomy, A.H.; Alkhalifah, D.H.M.; Hozzein, W.N. Actinomycetes from the Red Sea sponge Coscinoderma mathewsi: Isolation, diversity, and potential for bioactive compounds discovery. Microorganisms 2020, 8, 783. [Google Scholar] [CrossRef] [PubMed]
  75. McCarthy, P.J.; Roberts, B.F.; Carbonell, A.; Roberts, J.; Wright, A.E.; Chakrabarti, D. Marine microbiome as a source of antimalarials. Trop. Med. Infect. Dis. 2019, 4, 103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Quintero, M.; Blandón, L.M.; Vidal, O.M.; Guzman, J.D.; Gómez-Marín, J.E.; Patiño, A.D.; Molina, D.A.; Puerto-Castro, G.M.; Gómez-León, J. In vitro biological activity of extracts from marine bacteria cultures against Toxoplasma gondii and Mycobacterium tuberculosis. J. Appl. Microbiol. 2022, 132, 2705–2720. [Google Scholar] [CrossRef] [PubMed]
  77. Alkhalifah, D.H.M. Sponge-associated sp. RM66 metabolome induction with N-acetylglucosamine: Antibacterial, antifungal and anti-trypanosomal activities. Saudi J. Biol. Sci. 2021, 28, 4691–4698. [Google Scholar] [CrossRef] [PubMed]
  78. Santos, J.D.; Vitorino, I.; de la Cruz, M.; Díaz, C.; Cautain, B.; Annang, F.; Pérez-Moreno, G.; Gonzalez, I.; Tormo, J.R.; Martin, J. Diketopiperazines and other bioactive compounds from bacterial symbionts of marine sponges. Antonie Van Leeuwenhoek 2020, 113, 875–887. [Google Scholar] [CrossRef] [PubMed]
  79. Ghania, A.; Nabila, B.-B.; Larbi, B.; Elisabeth, M.; Philippe, G.; Mariem, B.; Khadidja, K.-K.; Wacila, B.-R.; Fawzia, A.-B. Antimicrobial and antiparasitic activities of three algae from the northwest coast of Algeria. Nat. Prod. Res. 2019, 33, 742–745. [Google Scholar] [CrossRef]
  80. Chiboub, O.; Ktari, L.; Sifaoui, I.; López-Arencibia, A.; Reyes-Batlle, M.; Mejri, M.; Valladares, B.; Abderrabba, M.; Piñero, J.E.; Lorenzo-Morales, J. In vitro amoebicidal and antioxidant activities of some Tunisian seaweeds. Exp. Parasitol. 2017, 183, 76–80. [Google Scholar] [CrossRef]
  81. Stein, E.M.; Tajú, S.G.; Miyasato, P.A.; de Freitas, R.P.; Tallarico, L.d.F.; Dos Santos, G.S.; Luiz, G.L.; Rofatto, H.K.; da Silva, F.N.; Colepicolo, P. The prospective use of Brazilian marine macroalgae in schistosomiasis control. Mar. Drugs 2021, 19, 234. [Google Scholar] [CrossRef]
  82. Saini, A.; Kumar, S.; Raj, R.; Chowdhary, S.; Gendrot, M.; Mosnier, J.; Fonta, I.; Pradines, B.; Kumar, V. Synthesis and antiplasmodial evaluation of 1H-1, 2, 3-triazole grafted 4-aminoquinoline-benzoxaborole hybrids and benzoxaborole analogues. Bioorg. Chem. 2021, 109, 104733. [Google Scholar] [CrossRef]
  83. Wilson, D.W.; Langer, C.; Goodman, C.D.; McFadden, G.I.; Beeson, J.G. Defining the timing of action of antimalarial drugs against Plasmodium falciparum. Antimicrob. Agents Chemother. 2013, 57, 1455–1467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Dalisay, D.S.; Williams, D.E.; Wang, X.L.; Centko, R.; Chen, J.; Andersen, R.J. Marine sediment-derived Streptomyces bacteria from British Columbia, Canada are a promising microbiota resource for the discovery of antimicrobial natural products. PLoS ONE 2013, 8, e77078. [Google Scholar] [CrossRef]
  85. Subramani, R.; Aalbersberg, W. Marine actinomycetes: An ongoing source of novel bioactive metabolites. Microbiol. Res. 2012, 167, 571–580. [Google Scholar] [CrossRef] [PubMed]
  86. Hancock, R.E. Resistance mechanisms in Pseudomonas aeruginosa and other nonfermentative gram-negative bacteria. Clin. Infect. Dis. 1998, 27 (Suppl. 1), S93–S99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Haste, N.M.; Hughes, C.C.; Tran, D.N.; Fenical, W.; Jensen, P.R.; Nizet, V.; Hensler, M.E. Pharmacological properties of the marine natural product marinopyrrole A against methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2011, 55, 3305–3312. [Google Scholar] [CrossRef] [Green Version]
  88. Khan, N.A. Pathogenesis of Acanthamoeba infections. Microb. Pathog. 2003, 34, 277–285. [Google Scholar] [CrossRef]
  89. Lorenzo-Morales, J.; Martín-Navarro, C.M.; López-Arencibia, A.; Arnalich-Montiel, F.; Piñero, J.E.; Valladares, B. Acanthamoeba keratitis: An emerging disease gathering importance worldwide? Trends Parasitol. 2013, 29, 181–187. [Google Scholar] [CrossRef]
  90. Antonsson, A.; Persson, J.L. Induction of apoptosis by staurosporine involves the inhibition of expression of the major cell cycle proteins at the G2/M checkpoint accompanied by alterations in Erk and Akt kinase activities. Anticancer Res. 2009, 29, 2893–2898. [Google Scholar]
  91. Wang, B.; Waters, A.L.; Sims, J.W.; Fullmer, A.; Ellison, S.; Hamann, M.T. Complex marine natural products as potential epigenetic and production regulators of antibiotics from a marine Pseudomonas aeruginosa. Microb. Ecol. 2013, 65, 1068–1075. [Google Scholar] [CrossRef] [Green Version]
  92. Aly, A.H.; Debbab, A.; Proksch, P. Fungal endophytes: Unique plant inhabitants with great promises. Appl. Microbiol. Biotechnol. 2011, 90, 1829–1845. [Google Scholar] [CrossRef]
  93. Hyde, K.; Soytong, K. The fungal endophyte dilemma. Fungal Divers 2008, 33, e173. [Google Scholar]
  94. Bhadury, P.; Mohammad, B.T.; Wright, P.C. The current status of natural products from marine fungi and their potential as anti-infective agents. J. Ind. Microbiol. Biotechnol. 2006, 33, 325. [Google Scholar] [CrossRef] [PubMed]
  95. Debbab, A.; Aly, A.H.; Proksch, P. Bioactive secondary metabolites from endophytes and associated marine derived fungi. Fungal Divers. 2011, 49, 1–12. [Google Scholar] [CrossRef]
  96. Singh, R.P.; Kumari, P.; Reddy, C. Antimicrobial compounds from seaweeds-associated bacteria and fungi. Appl. Microbiol. Biotechnol. 2015, 99, 1571–1586. [Google Scholar] [CrossRef] [PubMed]
  97. Amagata, T.; Usami, Y.; Minoura, K.; Ito, T.; NUMATA, A. Cytotoxic substances produced by a fungal strain from a sponge: Physico-chemical properties and structures. J. Antibiot. 1998, 51, 33–40. [Google Scholar] [CrossRef] [Green Version]
  98. Rivas, L.; Rojas, V. Cyanobacterial peptides as a tour de force in the chemical space of antiparasitic agents. Arch. Biochem. Biophys. 2019, 664, 24–39. [Google Scholar] [CrossRef]
  99. Demay, J.; Bernard, C.; Reinhardt, A.; Marie, B. Natural products from cyanobacteria: Focus on beneficial activities. Mar. Drugs 2019, 17, 320. [Google Scholar] [CrossRef] [Green Version]
  100. Niedermeyer, T.H.J. Anti-infective natural products from cyanobacteria. Planta Med. 2015, 81, 1309–1325. [Google Scholar] [CrossRef] [Green Version]
  101. Horton, D.A.; Bourne, G.T.; Smythe, M.L. Exploring privileged structures: The combinatorial synthesis of cyclic peptides. J. Comput. -Aided Mol. Des. 2002, 16, 415–431. [Google Scholar] [CrossRef]
  102. Sawadogo, W.R.; Boly, R.; Cerella, C.; Teiten, M.H.; Dicato, M.; Diederich, M. A survey of marine natural compounds and their derivatives with anti-cancer activity reported in 2012. Molecules 2015, 20, 7097–7142. [Google Scholar] [CrossRef]
  103. Walvekar, S.; Anwar, A.; Anwar, A.; Lai, N.J.Y.; Yow, Y.-Y.; Khalid, M.; Siddiqui, R.; Khan, N.A. Conjugation with Silver Nanoparticles Enhances Anti-Acanthamoebic Activity of Kappaphycus alvarezii. J. Parasitol. 2021, 107, 537–546. [Google Scholar] [CrossRef] [PubMed]
  104. Mayer, A.M.; Hamann, M.T. Marine pharmacology in 2001–2002: Marine compounds with anthelmintic, antibacterial, anticoagulant, antidiabetic, antifungal, anti-inflammatory, antimalarial, antiplatelet, antiprotozoal, antituberculosis, and antiviral activities; affecting the cardiovascular, immune and nervous systems and other miscellaneous mechanisms of action. Comp. Biochem. Physiol. Part C: Toxicol. Pharmacol. 2005, 140, 265–286. [Google Scholar]
Figure 1. The structure of compounds with effective antiparasitic activity in invertebrates.
Figure 1. The structure of compounds with effective antiparasitic activity in invertebrates.
Marinedrugs 21 00084 g001
Figure 2. The structure of compounds with effective antiparasitic activity in marine bacteria.
Figure 2. The structure of compounds with effective antiparasitic activity in marine bacteria.
Marinedrugs 21 00084 g002
Figure 3. The structure of compounds with effective antiparasitic activity in marine fungi.
Figure 3. The structure of compounds with effective antiparasitic activity in marine fungi.
Marinedrugs 21 00084 g003
Figure 4. The structure of compounds with effective antiparasitic activity in Cyanophyta.
Figure 4. The structure of compounds with effective antiparasitic activity in Cyanophyta.
Marinedrugs 21 00084 g004
Table 1. Natural products or derivatives from marine invertebrates and microorganisms.
Table 1. Natural products or derivatives from marine invertebrates and microorganisms.
CategorySpeciesCompoundsChemistryTarget ParasiteStage/StrainIC50CytotoxicitySiteReference
Type of CellsIC50
InvertebratespongesAplysinella rhax1 Psammaplin ABromotyrosine AlkaloidsT. cruziC2C430 μMNTNTFiji Islands[36]
P. falciparum3D760 μM
2 Psammaplin DT. cruziC2C443 μM
P. falciparum3D767 μM
3 BisaprasinT. cruziC2C419 μM
P. falciparum3D729 μM
Benznidazole *-T. cruziC2C42.6 μM---
Chloroquine *-P. falciparum3D70.017 μM
Dysidea avara4 AvaroneSesquiterpene Quinone AvaroneP. falciparumD102.74 μMHuman microvascular endothelial cells, HMEC-162.19 μMBay of Izmir, Turkey[37]
W22.09 μM
3D7 elo1-pfs16-CBG9915.53 μM
L. infantumpromastigote28.21 μMHuman acute monocytic leukemia cells, THP-1>100 μM
L. tropicapromastigote20.28 μM
L. infantumamastigotes7.64 μM
S. mansonischistosomula42.77 μM
5 ThiazoavaroneP. falciparumD100.38 μMHuman microvascular endothelial cells, HMEC-13.31 μM
W20.21 μM
3D7 elo1-pfs16-CBG9915.01 μM
L. infantumpromastigote 8.78 μMHuman acute monocytic leukemia cells, THP-17.41 μM
L. tropicapromastigote9.52 μM
L. infantumamastigotes 4.99 μM
S. mansonischistosomula5.90 μM
6 AvarolP. falciparumD100.96 μMHuman microvascular endothelial cells, HMEC-136.85 μM
W2 1.10 μM
3D7 elo1-pfs16-CBG999.30 μM
L. infantumpromastigote7.42 μMHuman acute monocytic leukemia cells, THP-131.75 μM
L. tropicapromastigote7.08 μM
L. infantumamastigotes3.19 μM
S. mansonischistosomula33.97 μM
Chloroquine *-P. falciparumD100.04 μM---
W20.54 μM
Methylene blue *-3D7 elo1-pfs16-CBG990.155 μM
Amphotericin B *-L. infantumpromastigote0.2 μM
L. tropicapromastigote0.17 μM
L. infantumamastigotes0.189 μM
Fascaplysinopsis reticulata7 8-oxo-tryptamine Tryptophan-Derived AlkaloidsP. falciparum3D78.8 µg/mLNTNTMayotte[38]
8 The mixture of the known (E) and (Z)-6-bromo-2′-demethyl-3′-N-methylaplysinopsin 8.0 µg/mL
Artemisinin *-0.006 μg/mL---
Hyrtios erectus9 Smenotronic acidSesquiterpenoidsP. falciparumDd23.51 μMNTNTSesquiterpenoids[39]
10 Ilimaquinone2.11 μM
11 Pelorol0.80 μM
Hyrtios sp.12 Hyrtiodoline AAlkaloidT. brucei
brucei
-48 h: 15.26 μMJ774.1 macrophages>200 μMRed Sea at Sharm el-Sheikh, Egypt[40]
72 h: 7.48 μM
Ircinia oros13 Ircinin-1Linear FuranosesterterpenoidsT. b. rhodesiense-97 μML6 rat myoblast cells150 μMGökçeada, Northern Aegean Sea, Turkey[41]
T. cruzi120 μM
L. donovani31 μM
P. falciparum58 μM
14 Ircinin-2T. b. rhodesiense65 μM140 μM
T. cruzi110 μM
L. donovani28 μM
P. falciparum56 μM
15 Ircinialactam ET. b. rhodesiense130 μM>200 μM
P. falciparum95 μM
16 Ircinialactam FT. b. rhodesiense130 μM>200 μM
L. donovani95 μM
Melarsoprol *-T. b. rhodesiense-0.015 μM---
Benznidazole *T. cruzi3.07 μM
Miltefosine *L. donovani0.51 μM
Chloroquine *P. falciparum0.009 μM
Podophyllotoxin *---L6 rat myoblast cells0.010 μM
Ircinia wistarii17 Ircinianin SesterterpenesP. falciparumNF5425.4 μMHeLa>64 μg/mLWistari Reef, Great Barrier Reef, Australia[42]
T. brucei rhodesienseSTIB90082.8 μM
T. cruziC2C4190.9 μML659.5 μg/mL
L. donovaniMHOM/ET/67/L8216.6 μM
Chloroquine *-P. falciparumNF540.006 μM---
Melarsoprol *T. brucei rhodesienseSTIB9000.020 μM
Benznidazole *T. cruziC2C43.36 μM
Miltefosine *L. donovaniMHOM/ET/67/L820.486 μM
Pseudoceratina sp.18 Psammaplysin FBromotyrosine AlkaloidP. falciparumK13.77 µg/mLMRC-512.65 µg/mLOkinawa, Japan[43]
FCR32.45 µg/mL
19 Ceratinadin EK11.03 µg/mL15.99 µg/mL
FCR30.77 µg/mL
Chloroquine *-K10.34 µg/mL>25.80 µg/mL-
FCR30.035 µg/mL
Artemisinin *K10.010 µg/mL>14.12 µg/mL
FCR30.0088 µg/mL
Monanchora unguiculata20 Unguiculin AAcyclic Guanidine AlkaloidP. falciparum3D712.89 μMKB Cells7.66 μMMitsio Islands, Madagascar[44]
21 Ptilomycalin EPentacyclic Alkaloids0.35 μM0.85 μM
22 Ptilomycalin F0.23 μM1.61 μM
23 Ptilomycalins G+H0.46 μM0.92 μM
24 Crambescidin 800Acyclic Guanidine Alkaloid0.52 μM1.31 μM
25 Fromiamycalin0.24 μM1.17 μM
Artemisinin *-0.004 μM----
Mycale sp. SS526 Albanitrile ANitrile-Bearing PolyacetylenesGiardia duodenalis71312 μMMammalian myeloma cell line NS-150 μMNear Albany[45]
Normal nontumor NFF cells100 μM
27 Albanitrile B25 μMMammalian myeloma cell line NS-150 μM
Normal nontumor NFF cells100 μM
28 Albanitrile C90 μMMammalian myeloma cell line NS-1180 μM
Normal nontumor NFF cells90 μM
Metronidazole * 2.9 μM---
Sparsomycin *----Mammalian myeloma cell line NS-10.55 μM
Normal nontumor NFF cells1.7 μM
Tedania brasiliensis29 PseudoceratidineBromopyrrole AlkaloidsP. falciparum3D7EC50 = 1 μMBone marrow-derived macrophagesNTCabo Frio, Rio de Janeiro state, Brazil[46]
1.1 μM
K11.1 μM
30 Pseudoceratidine derivativeP. falciparum3D7EC50 = 6 μM
31 Pseudoceratidine derivativeEC50 = 4 μM
32 Pseudoceratidine derivativeL. infantumpromastigotesEC50 = 24 μM52 μM
L. amazonensispromastigotesEC50 = 19 μM
T. cruziepimastigotesEC50 = 7 μM
33 Pseudoceratidine derivativeL. infantumpromastigotesEC50 = 19 μM>100 μM
L. amazonensispromastigotesEC50 = 7 μM
P. falciparum3D7EC50 = 19 μM
34 Pseudoceratidine derivativeP. falciparum3D7EC50 = 44 μMNT
35 Pseudoceratidine derivativeL. infantumpromastigotesEC50 = 2 μM66 μM
L. amazonensispromastigotesEC50 = 3 μM
T. cruziepimastigotesEC50 = 24 μM
36 Pseudoceratidine derivativeP. falciparum3D7EC50 = 7 μMNT
37 Pseudoceratidine derivativeL. infantumpromastigotesEC50 = 20 μM>100 μM
L. amazonensispromastigotesEC50 = 76 μM
38 Pseudoceratidine derivativeL. infantumpromastigotesEC50 = 23 μM82 μM
L. amazonensispromastigotesEC50 = 18 μM
P. falciparum3D7EC50 = 3 μMNT
Chloroquine *-P. falciparum3D70.013 μM---
K10.167 μM
Pyrimethamine *-P. falciparum3D70.03 μM---
K13.9 μM
Cycloguanil *-P. falciparum3D70.010 μM---
K10.54 μM
Artesunate *-P. falciparum3D70.004 μM---
K10.003 μM
Xestospongia sp.39 KaimanolSterolP. falciparum3D70.359 μMNTNTIndonesia[47]
40 Saringosterol0.00025 μM
Artemisinin *-5.207 × 10−3 nM---[48]
CnidariaAlcyonium sp.41 Alcyopterosin VIlludalane SesquiterpenesL. donovani-7.0 μMJ774.A1 macrophages110 μMScotia Arc of Antarctica[49]
Host cell lines HEK293T220 μM
Host cell lines HepG2288 μM
42 Alcyopterosin E3.1 μMJ774.A1 macrophages62 μM
Host cell lines HEK293T570 μM
Host cell lines HepG2331 μM
Miltefosine *-6.2 μM---
Bebryce grandis43 Bebrycin ADiterpeneP. falciparumDd2EC50 = 1.08 μMHepG2 human hepatocyte carcinoma cell lineEC50 =21.8 μMSoutheast coast of Curacao, East of Fuikbaai[50]
44 NiteninC21 Degraded TerpeneEC50 = 0.29 μMEC50 =18.3 μM
Macrorhynchia philippina45 IsololiolideCarotenoid IsololiolideT. cruzitrypomastigotes31.9 μMBMM cells >200 μMSão Sebastião Channel, Brazil[51]
amastigotes40.4 μM
Benznidazole *-trypomastigotes16.2 μM >200 μM-
amastigotes5.3 μM
Plumarella delicatissima46 Keikipukalide AFuranocembranoid DiterpenesL. donovaniamastigotes> 28 μMHuman lung carcinoma, cells, A549 cytotoxicity>50 μMStanley, Falkland Islands (Islas Malvinas), in the Southern Ocean[52]
47 Keikipukalide B8.5 μM>50 μM
48 Keikipukalide C8.8 μM>50 μM
49 Keikipukalide D12 μM>50 μM
50 Keikipukalide E8.8 μM>50 μM
51 Pukalide aldehyde1.9 μM>50 μM
52 Norditerpenoid ineleganolide4.4 μM>50 μM
Miltefosine *-6.2 μM---
Sinularia brassica53 Chlorinated steroidSteroidL. donovaniamastigoteamastigoteInhibition of a growth of L. donovani at 50 μM = 58.7%THP-1 cells at 50 μM88.8%Van Phong bay, Khanh Hoa province, Vietnam and Institute of Oceanography, Nha Trang, Vietnam[53]
54 Pinnaterpene CDibromoditerpeneInhibition of a growth of L. donovani at 50 μM = 74.3%106.2%
55 24-methylenecholestane-3β-5α,6β-triol-6-monoacetateSteroidInhibition of a growth of L. donovani at 50μM = 54.7%96.1%
56 Cholestane-3β-5α,6β-triol-6-monoacetateInhibition of growth of L. donovani at 50μM = 39.0%92.7%
Sinularia sp.57 SinuketalSesquiterpenoidsP. falciparum3D780 μMJurkat24.9 μMYongxing Island (16°50′ N, 112°20′ E) of Xisha Islands in the South China Sea[54]
MDA-MB-23132.3 μM
U2OS41.7 μM
Dihydroartemisinine *-10 nM---
BryozoaAmathia lamourouxi58 Convolutamines KBrominated AlkaloidsP. falciparum3D71.7 μMHuman embryonic kidney cell line, HEK29317.01 μMRock pools of Woolgoolga, New South Wales, Australia[55]
59 Convolutamines L3D711 μMIA at 40 μM
60 Volutamides F3D70.61 μMIA at 40 μM
Dd20.75 μM
61 Volutamides G3D70.57 μM11 μM
Dd20.85 μM
62 Volutamides H3D71.6 μMIA at 40 μM
Dd21.9 μM
Chloroquine *-3D70.025 μM67% at 4 μM-
Dd20.18 μM
Dihydroartemisinin *-3D70.0020 μMIA at 0.1 μM-
Dd20.0020 μM
Puromycin *-3D70.11 μM0.81 μM-
Dd20.068 μM
Orthoscuticella ventricosa63 Orthoscuticellines AAlkaloidsP. falciparum3D710 μMHuman embryonic kidney cell line, HEK29310 μMNorthern NSW, Australia[56]
64 Orthoscuticellines B> 40 μM>40 μM
65 Orthoscuticellines D14 μM>40 μM
66 Orthoscuticellines E12 μM>40 μM
67 1-ethyl-4-methylsulfone-β-carboline21 μM>40 μM
68 1-ethyl-β-carboline18 μM>40 μM
Chloroquine *-0.007 μM>40 μM
Artesunate *-0.0003 μM---
MicroorganismsActinomy-cetesStreptomyces sp. PBLC0469 StaurosporineAlkaloidAcanthamoeba castellaniiTrophozoites0.265 µg/mLMurine macrophage J774.A1 cell line4.076 μMJambelí mangrove, Ecuador[57]
Cysts0.771 µg/mL
Streptomyces sp.70 Marinopyrrole AAlkaloidsT. gondiiTachyzoites/Type I RH0.31 μMHuman foreskin fibroblast (HFF)>50 μMMarinopyrrole A was obtained from Sigma-Aldrich[58]
Human hepatocarcinoma (HepG2)5.3 μM
71 RL0020.17 μMHuman foreskin fibroblast (HFF)>50 μM
Human hepatocarcinoma (HepG2)29.0 μM
72 RL0030.09 μMHuman foreskin fibroblast (HFF)>50 μM
Human hepatocarcinoma (HepG2)49.7 μM
73 RL1250.16 μMHuman foreskin fibroblast (HFF)>50 μM
Human hepatocarcinoma (HepG2)46.5 μM
Pyrimethamine *-0.61 μM---
ProteobacteriaPseudomonas aeruginosa74 3-heptyl-3-hydroxy-1,2,3,4-tetrahydroquinoline-2.4-dione HydroxyquinolineP. falciparumIndochina W23.47 µg/mLNTNTPacific of Panama[59]
75 2-heptyl-4-hydroxyquinolineP. falciparumIndochina W22.57 µg/mL
T. cruziC43.66 µg/mL
76 2-nonyl-4-hydroxyquinoline P. falciparumIndochina W22.79 µg/mL
T. cruziC43.99 µg/mL
Chloroquine *-P. falciparumIndochina W20.03 µg/mL---
Nifurtimox *-T. cruziC41.6 µg/mL---
CyanophytaCaldora penicillata77 Hoshinoamide C (natural)LipopeptideP. falciparum3D70.96 μMHuman cancer cells, HeLa and HL60No cytotoxicity at 10 μMIkei Island, Okinawa, Japan[60]
T. brucei rhodesienseIL-15012.9 μM
78 Hoshinoamide C(synthetic)P. falciparum3D73.2 μM
T. brucei rhodesienseIL-15013.7 μM
79 43-epi-hoshinoamide C(synthetic)P. falciparum3D70.87 μM
T. brucei rhodesienseIL-15014.4 μM
Atovaquone *-P. falciparum3D70.00096 μM---
Pentamidine *-T. brucei rhodesienseIL-15010.001 μM---
Dapis sp.80 Iheyanone Linear PeptidesT. brucei rhodesienseIL-150135 μMWI-38 cells>50 μMNoho Island, Okinawa, Japan[61]
81 Peptides 33 μM>50 μM
82 Peptides24 μM>50 μM
83 Peptides 15 μM>50 μM
84 Peptides 17 μM>50 μM
85 Peptides 6.2 μM>50 μM
Pentamidine *-T. brucei rhodesienseIL-15010.05 μM----
Dapis sp.86 Iheyamides ALinear PeptidesT. b. rhodesienseIL-15011.5 μMNormal human fibroblasts, WI-38 cells18 μMNoho Island, Okinawa, Japan[62]
T. b. brucei2211.5 μM
T. b. rhodesienseIL-1501> 20 μM>20 μM
T. b. brucei221> 20 μM
T. b. rhodesienseIL-1501> 20 μM>20 μM
T. b. brucei221> 20 μM
Pentamide *-T. b. rhodesienseIL-15010.005 μM---
T. b. brucei2210.001 μM
Leptolyngbya sp.87 Motobamide Cyclic PeptideT. b. rhodesienseIL-15012.3 μMWI-38 cells55 μMBise, Okinawa Island, Okinawa Prefecture, Japan[63]
HeLa or HL60 cellsIA at 10 μM
Leptolyngbya sp.88 Palstimolide APolyhydroxy MacrolideP. falciparumDd20.1725 μMHepG2 human liver cell line5.04 μMPalmyra Atoll[64]
L. donovanipromastigotes4.67 μMB10R murine macrophages
(L. donovani host cell toxicity)
>10 μM
Okeania sp.89 IkoamideLipopeptideP. falciparum3D70.14 μMHeLa cells or HL60 cellsNo cytotoxicity at 10 μMIko-pier, Kuroshima Island, Okinawa, Japan[65]
Chloroquine *-6.9 nM---
doxorubicin----HeLa cells0.24 μM-
HL60 cells46 nM-
Okeania sp.90 Mabuniamide LipopeptideP. falciparum3D71.4 μML6 myotubesNo cytotoxicity at 10–40 μMThe coast of Odo, Okinawa, Japan[66]
91 Stereoisomer 21.4 μM
Chloroquine *-7.6 nM---
Salileptolyngbya sp.92 Kinenzoline (natural)Linear DepsipeptideT. b. rhodesienseIL-15015.0 μMWI-38 cells>20 μMKinenhama beach, Kagoshima, Japan[67]
93 Kinenzoline (synthetic)4.5 μM>100 μM
Pentamide *-0.001 μM---
Adriamycin *----WI-38 cells0.73 μM-
Moorea producens94 Dudawalamide ACyclic DepsipeptidesP. falciparumW23.6 μMH-460 human lung cancer cell lineLittle to no cytotoxicityPapua New Guinea[68]
T. cruziTransgenic β-galactosidase-expressing strain12% GI (Percentage growth inhibition) at 10 μg/mL
L. donovaniWR2810> 10 μM
95 Dudawalamide BP. falciparumW28.0 μM
T. cruziTransgenic β-galactosidase-expressing strain7% GI at 10 μg/mL
L. donovaniWR2810> 10 μM
96 Dudawalamide CP. falciparumW210 μM
97 Dudawalamide DP. falciparumW23.5 μM
T. cruziTransgenic β-galactosidase-expressing strain60% GI at 10 μg/mL
L. donovaniWR28102.6 μM
AscomycetesAspergillus terreus BCC5179998 AstepyrazinoxideAlkaloidP. falciparumK-124.82 μMMCF-734.70 μMThe marine fungus was isolated from a decayed wood sample at Hat Bang Pu, Khao Sam Roi Yot National Park, Prachuap Khiri Khan Province[69]
NCI–H1875.98 μM
Vero15.61 μM
99 Astechrome0.94 μMMCF-7IA
NCI–H187IA
Vero7.9 μM
Dihydroartemisinin *-2.12 × 10−3 μM---
Mefloquine *-0.422 μM---
Ellipticine *----NCI–H1879.87 μM-
Vero5.32 μM
Doxorubicin *----MCF-710.97 μM-
NCI–H1870.16 μM
Tamoxifen *----MCF-732.95 μM-
Cochliobolus lunatus TA26-46100 Derivatives14-Membered Resorcylic Acid Lactone DerivativesP. falciparumHB312.59 μmol/LHUVECNTMarine-derived[70]
101 Derivatives12.39 μmol/LNT
102 Derivatives11.55 μmol/LNT
103 Derivatives8.06 μmol/L>100 μmol/L
104 Derivatives6.69 μmol/L>100 μmol/L
105 Derivatives7.82 μmol/L>100 μmol/L
106 Derivatives9.72 μmol/L>100 μmol/L
107 Derivatives7.82 μmol/L>100 μmol/L
108 Derivatives7.25 μmol/L>100 μmol/L
109 Acyl derivatives9.18 μmol/LNT
110 Acyl derivatives6.91 μmol/L>100 μmol/L
111 Acyl derivatives3.54 μmol/L>100 μmol/L
Chloroquine *-32.9 nmol/L---
Exserohilum sp.112 Isocoumarins PolyketideP. falciparumHB31.13 μMVero cells87.5 μMZoanthid Palythoa haddoni[71]
113 Isocoumarins11.7 μM124.2 μM
114 Derivatives0.77 μM258.0 μM
115 Derivatives0.38 μM106.3 μM
116 Derivatives2.58 μM262.5 μM
Paecilomyces sp. 7A22117 Harzialactone APolyketoneL. amazonensispromastigotes5.25 μg/mLPeritoneal macrophages35.21 μg/mLAscidian Aplidiopsis sp. collected from São Sebastião Channel in Brazil[72]
amastigotes18.18 μg/mL
Amphotericin B *-L. amazonensispromastigotes0.119 μg/mL22.41 μg/mL-
amastigotes0.095 μg/mL
* Positive control; NT indicates not text; IA indicates inactive.
Table 2. Crude extracts of marine invertebrates and microorganisms.
Table 2. Crude extracts of marine invertebrates and microorganisms.
CategorySpeciesExtract TypeTarget ParasiteStage/StrainIC50SiteReferences
CnidariaLinuche unguiculataDistilled waterGiardia duodenalisTrophozoites, IMSS 0989:1 strain63 µg/mLPuerto Morelos Reef Lagoon, Mexico[73]
ActinomycetesNocardia sp. UA 23ISP2 mediumTrypanosoma bruceiTC 221MIC, 72 h = 7.2 µg/mLCoscinoderma mathewsi was collected from Ahia Reefs[74]
Micromonospora sp. W305Resin, MeOHAntiplasmodial ActivitiesDd20.42 µg/mLThe microbial population associated with deep-water invertebrates[75]
Nocardiopsis sp. V671ASE, MeOHAntiplasmodial ActivitiesDd20.88 µg/mLThe microbial population associated with deep-water invertebrates[75]
Streptomyces tendae V324Resin, MeOH/CH2Cl2Antiplasmodial ActivitiesDd20.35 µg/mLThe microbial population associated with deep-water invertebrates[75]
Streptomyces sp. INV ACT2Ethyl acetateT. gondiiGFP-RH tachyzoitesInhibition ≥ 80% at 120 μg/mLCaño Aguas Negras[76]
Streptomyces sp. RM66On ISP2, solid media with GlcNAcTrypanosoma bruceiTC 221MIC, 72 h = 4.7 µg/mLHurghada (Egypt)[77]
Streptomyces sp. V881Resin, CH2Cl2Antiplasmodial ActivitiesDd20.062 µg/mLThe microbial population associated with deep-water invertebrates[75]
Streptomyces sp. E677Resin, MeOH/CH2Cl2Antiplasmodial ActivitiesDd20.037 µg/mLThe microbial population associated with deep-water invertebrates[75]
Unidentified actinomycete V663ASE, heptaneAntiplasmodial ActivitiesDd20.89 µg/mLThe microbial population associated with deep-water invertebrates[75]
BacteroidesAlcanivorax sp. V174 (G-)Resin, MeOH/CH2Cl2Antiplasmodial ActivitiesDd20.969 µg/mLThe microbial population associated with deep-water invertebrates[75]
Alcanivorax sp. V193 (G-)Resin, MeOH/CH2Cl2Antiplasmodial ActivitiesDd21.079 µg/mLThe microbial population associated with deep-water invertebrates[75]
Endozoicomonas numazuensis H402 (G-)Resin, MeOH/CH2Cl2Antiplasmodial ActivitiesDd20.978 µg/mLThe microbial population associated with deep-water invertebrates[75]
Marinobacter sp. V184 (G-)Resin, MeOH/CH2Cl2Antiplasmodial ActivitiesDd21.008 µg/mLThe microbial population associated with deep-water invertebrates[75]
Marinobacter sp. V201 (G-)Resin, MeOH/CH2Cl2Antiplasmodial ActivitiesDd21.091 µg/mLThe microbial population associated with deep-water invertebrates[75]
Marinobacter sp. V208 (G-)Resin, MeOH/CH2Cl2Antiplasmodial ActivitiesDd21.091 µg/mLThe microbial population associated with deep-water invertebrates[75]
FirmicutesBacillus sp. INV FIR35Ethyl acetateT. gondiiGFP-RH tachyzoitesInhibition ≥ 80% at 48 μg/mLPunta Betín[76]
Bacillus sp. INV FIR48Ethyl acetateT. gondiiGFP-RH tachyzoitesInhibition ≥ 80% at 120 μg/mLCaño Grande[76]
Fictibacillus sp. INV FIR149Ethyl acetateT. gondiiGFP-RH tachyzoitesInhibition ≥ 80% at 1080 μg/mLCaño Grande[76]
Paenibacillus sp. #91_7 (IN-CRY)Waters™ Oasis® HLB extraction plates, with the sorbent Oasis® HLB, was equilibrated using methanol and HPLC grade waterT. cruziTulahuen C497%Isolated from marine sponges of the Erylus genus, collected in Portuguese waters[78]
Penicillium citrinum V170Resin, MeOH/CH2Cl2Antiplasmodial ActivitiesDd21.069 µg/mLThe microbial population associated with deep-water invertebrates[75]
Penicillium sp. N161Resin, MeOH/CH2Cl2Antiplasmodial ActivitiesDd20.266 µg/mLThe microbial population associated with deep-water invertebrates[75]
Penicillium sp. Z691Resin, CH2Cl2Antiplasmodial ActivitiesDd20.049 µg/mLThe microbial population associated with deep-water invertebrates[75]
Talaromyces rotundus S920Resin, MeOH/CH2Cl2Antiplasmodial ActivitiesDd20.677 µg/mLThe microbial population associated with deep-water invertebrates[75]
Tritirachium sp. V199Resin, MeOH/CH2Cl2Antiplasmodial ActivitiesDd20.339 µg/mLThe microbial population associated with deep-water invertebrates[75]
ɣ-ProteobacteriaEnterococcus faecalis #118_3 (IN-CRY)EPA vials: Sepabeads® SP207ss resin, HPLC-grade water and acetone; medium IN-CRYT. cruziTulahuen C4Percentage of growth inhibition = 81%Isolated from marine sponges of the Erylus genus, collected in Portuguese waters[78]
Enterococcus faecalis #118_3 (IN-CRY)Duetz extraction: Waters™ Oasis® HLB extraction plates, with the sorbent Oasis® HLB, was equilibrated using methanol and HPLC grade water; medium IN-CRYT. cruziTulahuen C4Percentage of growth inhibition = 102%Isolated from marine sponges of the Erylus genus, collected in Portuguese waters[78]
Enterococcus faecalis #118_4 (IN-CRY)Duetz extraction: Waters™ Oasis® HLB extraction plates, with the sorbent Oasis® HLB, was equilibrated using methanol and HPLC grade water; medium IN-CRYT. cruziTulahuen C4Percentage of growth inhibition = 103%Isolated from marine sponges of the Erylus genus, collected in Portuguese waters[78]
Pseudoalteromonas sp. INV PRT33Ethyl acetateT. gondiiGFP-RH tachyzoitesInhibition ≥ 80% at 48 μg/mLCaño Grande[76]
PhaeophytaCladostephus hirsutusEthyl acetateT. brucei brucei-27.2 μg/mLNorth-west coast of Algeria[79]
Cystoseira sedoidesHexaneAcanthamoeba castellaniiTrophozoite/Neff1009 μg/mLTunisian coasts, Tabarka[80]
Ethyl acetate860 μg/mL
Methanol836 μg/mL
Dictyota ciliolataHexaneSchistosoma mansoniDeath Ratio = 100%Espírito Santo State, Southeastern Brazil[81]
ChloroformDeath Ratio = 100%
Supercritical fluidDeath Ratio = 100%
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

Zhang, M.; Zhang, Q.; Zhang, Q.; Cui, X.; Zhu, L. Promising Antiparasitic Natural and Synthetic Products from Marine Invertebrates and Microorganisms. Mar. Drugs 2023, 21, 84. https://doi.org/10.3390/md21020084

AMA Style

Zhang M, Zhang Q, Zhang Q, Cui X, Zhu L. Promising Antiparasitic Natural and Synthetic Products from Marine Invertebrates and Microorganisms. Marine Drugs. 2023; 21(2):84. https://doi.org/10.3390/md21020084

Chicago/Turabian Style

Zhang, Mingyue, Qinrong Zhang, Qunde Zhang, Xinyuan Cui, and Lifeng Zhu. 2023. "Promising Antiparasitic Natural and Synthetic Products from Marine Invertebrates and Microorganisms" Marine Drugs 21, no. 2: 84. https://doi.org/10.3390/md21020084

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