Next Article in Journal
Pseudane-VII Isolated from Pseudoalteromonas sp. M2 Ameliorates LPS-Induced Inflammatory Response In Vitro and In Vivo
Previous Article in Journal
Krishnolides A–D: New 2-Ketokhayanolides from the Krishna Mangrove, Xylocarpus moluccensis
Previous Article in Special Issue
Bifurcatriol, a New Antiprotozoal Acyclic Diterpene from the Brown Alga Bifurcaria bifurcata
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Marine Algae as Source of Novel Antileishmanial Drugs: A Review

by
Lauve Rachel Tchokouaha Yamthe
1,2,3,*,
Regina Appiah-Opong
4,
Patrick Valere Tsouh Fokou
3,
Nole Tsabang
1,
Fabrice Fekam Boyom
3,
Alexander Kwadwo Nyarko
4,5 and
Michael David Wilson
2
1
Institute for Medical Research and Medicinal Plants Studies, Yaoundé 13033, Cameroon
2
Department of Parasitology, Noguchi Memorial Institute for Medical Research (NMIMR), College of Health Sciences (CHS), University of Ghana, Legon LG 581, Ghana
3
Antimicrobial and Biocontrol Agents Unit, University of Yaoundé 1, Yaoundé 812, Cameroon
4
Department of Clinical Pathology, NMIMR, CHS, University of Ghana, Legon LG 581, Ghana
5
Department of Pharmacology and Toxicology, School of Pharmacy, CHS, University of Ghana, Legon LG 43, Ghana
*
Author to whom correspondence should be addressed.
Mar. Drugs 2017, 15(11), 323; https://doi.org/10.3390/md15110323
Submission received: 14 July 2017 / Revised: 9 October 2017 / Accepted: 17 October 2017 / Published: 29 October 2017
(This article belongs to the Special Issue Antiprotozoal Marine Natural Products)

Abstract

:
Leishmaniasis is a vector-borne neglected tropical disease caused by protozoan parasites of the Leishmania genus and transmitted by the female Phlebotomus and Lutzomyia sand flies. The currently prescribed therapies still rely on pentavalent antimonials, pentamidine, paromomycin, liposomal amphotericin B, and miltefosine. However, their low efficacy, long-course treatment regimen, high toxicity, adverse side effects, induction of parasite resistance and high cost require the need for better drugs given that antileishmanial vaccines may not be available in the near future. Although most drugs are still derived from terrestrial sources, the interest in marine organisms as a potential source of promising novel bioactive natural agents has increased in recent years. About 28,000 compounds of marine origin have been isolated with hundreds of new chemical entities. Recent trends in drug research from natural resources indicated the high interest of aquatic eukaryotic photosynthetic organisms, marine algae in the search for new chemical entities given their broad spectrum and high bioactivities including antileishmanial potential. This current review describes prepared extracts and compounds from marine macroalgae along with their antileishmanial activity and provides prospective insights for antileishmanial drug discovery.

Graphical Abstract

1. Introduction

Leishmaniasis is a vector-borne disease caused by protozoan kinetoplastid parasites of the genus Leishmania. The disease is transmitted through the bite of infected female phlebotomine sandflies of the genera Phlebotomus and Lutzomyia respectively in the Old World (Europe, Asia and Africa) and in the New World (America). Leishmania life cycle is dimorphic and heteroxene with an extracellular fusiform and flagellated promastigotes stage within the midgut of the sandfly, and a morphologicaly distinct intracellular amastigote stage within macrophages of a mammalian host [1,2]. It is worth noting that based on these two stages of Leishmania parasite, various models have been developed for drug susceptibility tests. The promastigote model is often used, however, this model does not necessarily reflect the physiological situation, as the disease-causing stage of the parasite, amastigote resides inside the host cells. Axenic amastigote model is used as an alternative to the labor-intensive intracellular amastigote model. However, several reports distinguish between axenic amastigotes and intracellular amastigotes both in terms of drug susceptibility and protein expression [3]. Leishmaniasis is a disease which is distributed worldwide in the tropics, subtropics, and the mediterranean basin and affect both humans and animals [3,4]. This neglected tropical disease represents a major public health problem in 98 endemic countries where it is responsible for approximately 2–4 million new cases and around 70,000 deaths per year [4]. Leishmaniasis presents two main clinical manifestations including cutaneous leishmaniasis (CL) affecting macrophages resident in the skin and visceral leishmaniasis (VL) affecting cells of the mononuclear phagocyte system of liver, spleen, bone marrow, lymph nodes and intestine [1]. VL is the most severe form of leishmaniasis with about 0.2–0.4 million cases each year in the world [1,5]. Besides, VL represents the second deadliest parasitic disease after malaria and the third-most common cause of morbidity after malaria and schistosomiasis [6]. Leishmania species, which cause VL include L. donovani, L. infantum, L. martiniquensis and L. tropica [1]. However, CL is the most common manifestation of the disease, with between 700,000 to 1.2 million new cases every year. It usually causes ulcers on the face, arms and legs. Furthermore, CL occurs in three differents forms, namely, localised cutaneous leishmaniasis, diffuse cutaneous leishmaniasis, and mucocutaneous leishmaniasis. Many Leishmania species are responsible for CL, including L. mexicana, L. amazonensis, L. braziliensis, L. panamensis, L. guyanensis, L. aethiopica, L. venezuelensis, L. lainsoni, L. shawi, L. peruviana, L. naiffi, L. lindenbergi, L. infantum, L. martiniquensis and L. waltoni in the New World and L. tropica, L. aethiopica and L. major in the Old World [1].
For the past 60 years, the first line-drugs of choice for leishmaniasis treatment have been pentavalent antimonials that include two formulations: sodium stibogluconate (pentostam) and meglumine antimoniate (glucantime) with comparable efficacy. However, emergence of resistance to these drugs added to toxic effects has been reported [7]. The second line-drug amphotericin B exhibits excellent activity against resistant parasites, but prolonged administration and adverse effects are major shortcomings to its use. The newer lipid formulations of amphotericin B are amphotericin B liquid complex, liposomal amphotericin B and amphotericin B colloidal dispersion. These new formulations allow for short-term treatment and provide excellent activity at low concentration with less toxicity. They are however extremely costly for poor patients in many affected countries [7,8,9,10,11,12]. Pentamidine, miltefosine, paromomycin, azoles and allopurinol are other drugs used against Leishmania parasites [6,13,14]. For most of these drugs, there are many drawbacks in terms of undesirable side effects, emergence of drug resistance, relapse after treatment or long-course of treatment and high cost [3,12,13,15]. In addition to the afore-mentioned limitations, the increased number of co-infection with HIV have challenged drug discovery research over the past two decades [3,4,8,9]. Ideally, the new therapeutic agents should have more innovative mechanisms of action, higher efficacy, affordability and shorter treatment duration, and less toxicity to improve compliance for the treatment and reduce the likelihood of emergence of resistance by the parasites.
Natural products are considered as rich and credible sources of compounds for drug discovery. At least one-third of the current top twenty drugs on the market are derived from a natural source, and approximately 50% of the marketed drugs are classified as naturally derived or designed on the basis of natural compounds [16,17].
Many studies worldwide have focused on the prospection of terrestrial plants for the discovery of effective compounds against the causative agents of leishmaniasis but little attention has been paid to the biological properties of marine organisms [18]. The interest in marine organisms as a potential source of promising novel bioactive natural agents has increased in recent times [19,20]. About 28,000 compounds have been isolated from marine organisms with hundreds of new chemical entities [21,22,23]. Natural products from marine algae are known for their potent and broad spectrum bioactivities including antimicrobial, antiviral, anti-helminthic, antituberculosis, antimycobacterial, antioxidative, anticoagulant, anti-inflammatory, antipyretic, analgesic, anticancer, insecticidal, antidiabetic, and antiprotozoan activities [21,24,25,26,27,28,29,30]. Moreover, the ability of marine algae to grow through mariculture and their short generation time make them sustainable sources of active ingredients. This is considered an environment-friendly strategic approach that overcomes problems associated with the overexploitation of marine resources and the use of destructive collection methods [21]. Despite this great potential, no attempt has been made to provide an overview of marine algae with leishmanicidal properties.
This review presents the antileishmanial activity of marine macroalgae and their phytochemicals and provides prospective insights for antileishmanial drug discovery.

2. Marine Algae

Marine algae are aquatic eukaryotic photosynthetic organisms, diversified in size, from microalgae to macroalgae [21,31,32].
Microalgae are a polyphyletic group of unicellular marine algae that constitute one of the major components of marine and freshwater phytoplankton. They are primary producers and a food source for other marine organisms [33]. There are at least 40,000 to 70,000 species belonging to three different groups which are diatoms, dinoflagellates and flagellates [21,34]. Microalgae are known to produce numerous useful natural products, but compared to macroalgae, they have attracted little attention in the search for novel anti-infective compounds [35] particulary against Leishmania parasite with only one report paper [30].
Marine macroalgae commonly known as seaweeds are macroscopic and multicellular. With an estimation of more than 30,000 species, they represent a considerable part of the marine environment. Seaweeds have been used as human food from 600 to 800 BC in China and other countries in Asia [36,37]. Based on their pigmentation, macroalgae are classified into three main phyla which include red seaweed (Rhodophyta), brown seaweed (Phaeophyta) and green seaweed (Chlorophyta) [38,39].

3. Current Status of Antileishmanial Drug Discovery from Marine Macroalgae

An overview of the findings reported so far on the search for Leishmania parasites inhibitors from macroalgae is presented in Table 1, including information on the macroalgae species, the type of extract, the Leishmania species and parasites forms. Algae with a determined IC50 value ≤100 µg/mL, or with a percentage of inhibition >50% were reported in Tables. Also found on Table 1 are the methods used for evaluation of the activities, and the activity parameters (IC50 -Extract concentration that inhibited the proliferation of parasites by 50%, CC50 -Extract concentration that inhibited the proliferation of normal mammalian cells by 50% or percentage of parasite inhibition).
The amount of research studies focusing on therapeutic properties of macroalgae has increased in recent years. However, only few antileishmanial secondary metabolites have been reported in 33 papers describing the antileishmanial activity of 151 marine macroalgae against different Leishmania parasites viz. L. infantum, L. donovani, L. major, L. amazonensis, L. Mexicana and L. braziliensis (Table 1 and Table 2). From the data assembled in Table 1 and Table 2, it appears that marine macroalgae could inhibit Leishmania parasites with IC50 values as low as 0.27 μg/mL [51]. This level of activity denotes the presence of highly potent natural products in these organisms. The following two sections highlight the macroalgae that were reported to have been investigated for antileishmanial activity.

3.1. Macroalgae with Antileishmanial Properties

It is estimated that there are more than 30,000 macroalgae identified around the world [38] out of which only 151 have been investigated against Leishmania parasites.
Extracts from 48 species of brown macroalgae (Phaeophyceae) have been screened against Leishmania parasites. Spavieri et al. [40] assessed the antileishmanial activity of isopropyl alcohol-chloroform/methanol extracts from 21 brown macroalgae and found that all the extracts showed antileishmanial activity against axenic L. donovani amastigotes with IC50 values ranging from 6.4 to 77.4 µg/ mL (Table 1). B. bifurcata and H. siliquosa extracts were the most active with IC50 values of 6.4 and 8.6 µg/mL, respectively. However, B. bifurcata and H. siliquosa extracts showed an extent of cytotoxic effects against the mammalian skeletal myoblasts, L6 cells with CC50 values of 32.7 and 45.0 μg/mL, respectively [40]. Hydroalcoholic and ethyl acetate extracts from eight brown algae were also assessed for their inhibitory activity against axenic L. donovani amastigotes [41]. Ethyl acetate extracts of B. bifurcata, D. dichotoma and D. polypodioides showed strong activities with IC50 values in the range 3.9 to 10.8 μg/mL. Nevertheless, they also exhibited cytotoxic profile on L6 cells with a CC50 value of 6.0 μg/mL, indicating poor selectivity of the extracts for the parasites. In another study, hexane, ether and chloroform extracts from B. bifurcata, showed moderate activities against L. infantum promastigotes with IC50 values ranging from 46.83 to 63.83 μg/mL and CC50 values below 41 μg/mL on Brine Shrimp larvae of Artemia salina [42]. These results also suggest that the extracts have poor selectivities towards the parasites. The phytochemistry of B. bifurcata has been extensively investigated and compounds such as sterols, polyphenols, diterpenes have been reported. Eleganolone was described as the major oxygenated diterpene [28,62,63,64,65,66]. The compounds present in the alga may be responsible for the antileishmanial activities. According to Vonthron-Sénécheau et al. [41], the cytotoxicity of B. bifurcata could be explained by the presence of terpenoids with cytotoxic activity.
Brown macroalgae of Dictyotaceae family, viz. Canistrocarpus, Dictyota and Stypopodium genera (Table 1 and Table 2) appeared to be among the most interesting Phaeophyceae. In fact, six out of the seven species from Dictyota genus (D. caribaea, D. ciliolata, D. dichotoma, D. menstrualis, D. mertensii and D. pfaffii) as well as C. cervicornis and S. zonale exhibited potent antileishmanial activities against promastigote, axenic amastigote and intracellular amastigote of L. donovani and L. amazonensis with IC50 values ranging from 0.27 to 81.4 μg/mL [40,41,46,47,48,49,50,51]. Extracts from three species showed interesting activity profiles, with IC50 values below 10 μg/mL and selectivity indices (ratio of CC50 value to IC50 value) greater than 20. Amongst these, ethylacetate/hexane extract from S. zonale exhibited the strongest activity against promastigotes and intracellular amastigotes of L. amazonensis with IC50 values of 0.75l and 0.27 μg/mL respectively and with respective CC50 values of 29.5 and >50 μg/mL on murine peritoneal macrophages [47,51]. Also, ethylacetate and hexane extracts of D. menstrualis showed potent activity with IC50 values ranging from 0.7 to 0.75 μg/mL and 0.61 μg/mL, respectively against promastigotes of L. amazonensis and CC50 > 18.2 μg/mL on murine peritoneal macrophages. Hexane extract of D. ciliolata, showed a strong antileishmanial activity with an IC50 value of 1.15 μg/mL on L. amazonensis promastigotes with CC50 value of 23.5 μg/mL [47]. This suggests a good selectivity of the extract since selectivity index is about 20. Besides, extracts from others brown macroalgae were also found to have antileishmanial activity, including C. barbata, C. crinata, Lobophora variegata, Padina sp., S. muticum, S. natans, S. oligocystum, S. furcellata, T. turbinata and U. pinnatifida. These extracts moderately inhibited the growth of Leishmania parasites with IC50 values ranging from 10.9 to 90.9 μg/mL or percentage of inhibition values ranging from 80.9% to 98.5% at the tested concentration (Table 1). The organic extract of T. turbinata that showed IC50 value of 10.9 μg/mL and a selectivity index of 70.41 is also of interest [46] and could further be investigated for its antileishmanial metabolites.
The most screened macroalgae phylum was Rhodophyta where 80 species were investigated. Extracts from many studied species of this phylum were shown to exhibit activity against cutaneous and visceral Leishmania parasites (Table 1 and Table 2). For instance, ethanolic extracts from O. pinnatifida (IC50 = 6.25 μg/mL), M. afaqhusainii (IC50 = 32.6 μg/mL), G. corticata (IC50 = 37.5 μg/mL), S. hatei (IC50 = 14.1 μg/mL), S. indica (IC50 = 59.6 μg/mL), C. clavulatum (IC50 = 57.89 μg/mL), B. leptopoda (IC50 = 60.81 μg/mL) and water extracts from G. salicornia (IC50 = 46.0 to 74.0 μg/mL) and G. corticata (IC50 = 38.0 to 65.0 μg/mL) inhibited the growth of promastigotes of cutaneous leishmaniasis agent, L. major [25,50,55] with. O. pinnatifida extract being the most promising inhibitor. Crude extracts from B. fruticulosa, C. jubata, C. rubrum, C. virgatum, C. verticillata, C. ovatum, C. granifera, C. officinalis, C. ramosa, C. purpureum, D. pedicellata, D. incrassata, G. crinale, G. pulchellum, G. gracilis, G. verrucosa, H. incurvus, H. equisetifolius, J. rubens, F. lumbricalis, L. articulata, M. stellatus, O. hybrid, O. pinnatifida, P. cartilagineum, P. rotundus, P. linearis [45,54] and ethanol and n-butanol fractions from C. hornemanni [58] were shown to be active against the visceral leishmaniasis causative agent L. donovani with IC50 values ranging from 16.76 to 85.6 μg/mL. Apart from the extracts from C. officinalis and D. pedicellata which showed cytotoxicity with CC50 values of 88.6 and 14.7 μg/mL respectively, all the other samples presented acceptable selectivity indices with CC50 values ranging from >90 μg/mL [45,54] to >200 μg/mL [58]. Asparagopsis species showed promising activities and might serve as starting point for future search of bioactive compounds against visceral leishmaniasis. In fact, dichloromethane and hexane extracts from A. taxiformis, ethanol-hexane:ethylacetate and ethanol-ethylacetate fractions from A. taxiformis and A. armata also showed significant effect on L. donovani promastigotes with IC50 values ranging from 10 to 20 μg/mL [52]. Additionally, ethanol extract of A. taxiformis was shown to be active against promastigotes and axenic amastigotes of L. infantum with IC50 values of 25 μg/mL and 9 μg/mL, respectively and acceptable safety profile on VERO and DH82 cell lines (CC50 > 90 μg/mL) [53]. These activities of Asparagopsis species could be attributed to the presence of volatile halogenated compounds (halomethanes, haloethers, haloacetals) which have been otherwise described as responsible for the antimicrobial properties of A. armata [67].
Besides, hexane (HO) and dichloromethane (DO) fractions and subfractions from B. tenella and aqueous extract from B. triquetrum showed antileishmanial activity against L. amazonensis promastigotes with IC50 values ranging from 1.5 to 78.6 μg/mL [56,57]. Among these, subfractions HO2 (IC50 = 1.5 μg/mL), HO3 (IC50 = 2.7 μg/mL), DO1 (IC50 = 4.4 μg/mL) and DO2 (IC50 = 4.3 μg/mL) were the most active [56]. Considering the activities of hexane (IC50 = 17.4 μg/mL) and dichloromethane (IC50 > 100 μg/mL) fractions of B. tenella as compared to subsequent subfractions, it is likely that chromatography may have concentrated the active principles in those subfractions or eventually reduce antagonistic interactions among molecules. These subfractions can be worked up to purify/characterized the antileishmanial molecules. Furthermore, crude extracts from the red macroalgae Laurencia complex (L. aldingensis, L. dendroidea, L. microcladia, P. flagellifera and P. perforata) showed great potential as antileishmanial drug sources (Table 1) and must be further explored. Reported findings indicate that extracts from these macroalgae have in vitro antileishmanial activity with IC50 values ranging from 16.3 to 97.2 μg/mL against the insect-stage promastigotes of L. amazonensis and L. mexicana and 8.7 to 34.5 μg/mL against the mammalian-stage amastigotes of L. amazonensis [46,59,60]. Moreover, they were globally not cytotoxic with CC50 value ≥1000 μg/mL. Extracts of L. aldingensis, L. dendroidea and L. microcladia exhibited the most interesting activity profile with IC50 values ranging from 8.7 to 30.1 μg/mL. Thus, these extracts may contain potential therapeutic agents, which must be further investigated. The antileishmanial potency of Laurencia genus could be attributed to the presence of bioactive sesquiterpenoids. In fact, more than 700 compounds mainly sesquiterpenoids and rearranged derivatives from Laurencia species have been shown to display a vast array of biological activities (antiviral, antibacterial, antimalarial) [68,69,70].
Extracts from few Chlorophyta marine macroalgae have been reported to have activity against leishmaniasis agents (Table 1). Indeed, from the investigation of 23 green macroalga species, acetone extract of A. saldanhae was active against the promastigotes (87.9% inhibition at 50 μg/mL) and intracellular amastigote (IC50 = 23.9 μg/mL) of L. braziliensis with CC50 value of 294.2 μg/mL on J774.G8 macrophage cell line [26]. This implies that the extract has good selectivity of 12.3 and can be suggested for chemical work-up to identify its bioactive principles. Also, crude extracts of C. racemosa, C. rupestris, C. bursa, C. fragile, U. intestinalis and U. lactuca significantly inhibited the growth of axenic L. donovani amastigotes in culture, with IC50 values ranging from 5.9 to 31.76 μg/mL and acceptable selectivity toward mammalian L6 cells (CC50 > 90 μg/mL). Amongst these extracts, U. lactuca and U. intestinalis extracts showed the highest antileishmanial activities [45,48,61]. The data suggest that green macroalgae from Ulva species could be valuable sources of antileishmanial compounds. Approximately 50 marine algae species from Ulva genus have been identified. Ulva species are known for their various sulphated polysaccharide (SP) compounds with ulvan as the most important one [71]. Extracts and SP from U. lactuca have been found to have antimicrobial, anticoagulant, antiprotozoal, antioxidant, antiperoxidative, antihyperlipidemic, antiviral, anticancer and hepatoprotective activities at low concentrations [72,73], making them good starting points for drug discovery against many diseases. Aqueous extract of H. opuntia showed activity on promastigotes and intracellular amastigotes of L. amazonensis with IC50 values of 83.5 μg/mL and 70.7 μg/mL respectively and CC50 value of 526.4 μg/mL on macrophages [57]. Bioguided fractionation should allow identifying bioactive compounds from this species.
Among the different extracts tested, the ethyl acetate extracts were globally the most active. Seven macroalgae were extracted with a non-polar solvent, ethyl acetate viz. B. bifurcata, C. cervicornis, D. polypodioides, D. ciliolata, D. dichrotoma, D. menstrualis and D. carnosa (Table 1). All the extracts strongly inhibited the growth of Leishmania with IC50 values ranging from 0.7 to 10.8 μg/mL.

3.2. Isolated Compounds from Marine Macroalgae Screened for Antileishmanial Properties

Table 2 below contains a summary of isolated compounds from marine macroalgae that were screened against Leishmania parasites.
In the framework of the reported investigations of marine algae for antileishmanial drug search, isolated compounds (Figure 1) were screened against Leishmania parasites.
More than 50% of antileishmanial compounds isolated from macroalgae belong to the family of diterpenes (Prenylated guaiane, dolastane, secodolastane, xeniane, dolabellane, dichotomanes and meroterpenoids). These are largely distributed among the brown seaweeds of the Dictyotaceae family as major secondary metabolites with biological activities against a vast panel of pathogens, cancer cells and cardiovascular diseases [49,78,79,80,81].
From the diterpenes isolated, (4R,9S,14S)-4α-acetoxy-9β,14α-dihydroxydolast-1(15),7-diene (1, Figure 1), a 4-acetoxydolastane diterpene obtained from Canistrocarpus cervicornis showed interesting activity with IC50 values of 2.0 μg/mL, 12.0 μg/mL, and 4.0 μg/mL for promastigote, axenic amastigote and intracellular amastigote forms of L. amazonensis, respectively. Moreover, cytotoxicity assay showed that this compound was 93 times less toxic to the J774G8 macrophages than to Leishmania parasites. Studies on the mechanism of action revealed that the activity of this molecule may be due by its interference with the mitochondrial membrane potential and lipid peroxidation in parasite cells [43]. Compared to other isolated diterpenes, this compound was the most active, suggesting that the acetoxyl group in its structure seemed to increase the activity. On the other hand, the observed antileishmanial activity could be also related to the Leishmania species and the parasite stages, which often result in different drug susceptibility [3]. A new linear diterpene, bifurcatriol (2, Figure 1) isolated from Bifurcaria bifurcata was active against L. donovani with IC50 value of 18.8 μg/mL and CC50 value of 56.6 μg/mL on L6 rat myoblast cell line [74]. However, the assay was done on axenic amastigote form of the parasite, which is not the suitable target for antileishmanial drug discovery. Two meroditerpenoids, (3R)- and (3S)-tetraprenyltoluquinol (1a/1b) (3, Figure 1) and (3R)- and (3S)-tetraprenyltoluquinone (2a/2b) (4, Figure 1) isolated from Cystoseira baccata inhibited the growth of the promastigotes of visceral leishmaniasis parasite, L. infantum moderately with IC50 values of 44.9 μM and 94.4 μM, respectively [44]. (3R)- and (3S)-tetraprenyltoluquinol (1a/1b) also showed activity against the intracellular amastigotes with IC50 value of 25.0 μM and CC50 value of 126.6 μM on murine peritoneal macrophages. They were able to induce cytoplasmic vacuolization and the presence of coiled multilamellar structures in mitochondria as well as an intense disruption of the mitochondrial membrane potential. Comparing the structures of the two meroditerpenoids, it appears that the presence of a second carbonyl group in (3S)-tetraprenyltoluquinone (2a/2b) may be responsible for the observed decrease in activity. This assumption is reinforced by the lack of the second carbonyl group in the structure of another meroditerpenoid, atomaric acid (5, Figure 1) isolated from the active lipophilic extract of Stypopodium zonale [51]. In fact, atomaric acid showed a similar IC50 value 20.2 μM against the intracellular amastigote of L. amazonensis. This compound also inhibited promastigotes growth by up to 86% at 50 μΜ with low toxicity towards host cells (CC50 = 169.5 μΜ). The results showed that leishmanicidal activity of atomaric acid was independent of nitric oxide production, but the generation of reactive oxygen species may be at least partially responsible its activity against the amastigote form [51]. A mixture of diterpene isomers pachydictyol A/isopachydictyol A (6, Figure 1) isolated from the dichloromethane extract from Dictyota menstrualis showed antileishmanial activity against L. amazonensis promastigotes with IC50 value of 23.5 μg/mL. However, cytotoxic effect was detected on murine peritoneal macrophages (CC50 = 30.0 μg/mL) [47]. A dolabellane diterpene, Dolabelladienetriol (7, Figure 1), isolated from Dictyota pfaffii repressed the intracellular amastigote of L. amazonensis replication with IC50 value of 43.9 µM and a CC50 value >100 μM on murine peritoneal macrophages. At 100 μM, dolabelladienetriol inhibited 95.5% of promastigote growth [14]. Dolabelladienetriol was also active against Leishmania/HIV-1 co-infection with 56.0% inhibition at 50.0 μM. This compound was able to modulate macrophage activity by inhibiting nitrogen oxide and cytokines TGF-β and TNF-α production, which could be responsible of the activity of the compound.
One triterpene derivative, fucosterol (8, Figure 1) has been isolated from Lessonia vadosa a brown macroalga [75]. This phytosterol, was found to be significantly more active against the intracellular amastigotes of L. amazonensis and L. infantum (IC50 values of 7.89 μM and 10.30 μM respectively) compared to the vector stage, promastigotes (IC50 values of 55.0 μM and 45.0 μΜ, respectively) [75], indicating that the antileishmanial activity of fucosterol is dependent on a macrophage function. These results justify the intracellular amastigote model as the suitable model for drug-screening. In addition, fucosterol displayed little cytotoxicity against the host macrophagic cell line with CC50 value >100 μM [75].
With the aim to identifying the compounds responsible for the strong antileishmanial activity of Laurencia dendroidea, crude extract of the red macroalga was fractionated and sesquiterpene compounds, elatol (9, Figure 1), obtusol (10, Figure 1) and silphiperfol-5-en-3-ol (11, Figure 1) were isolated [26,60]. From the antileishmanial assay against the promastigotes and intracellular amastigotes of L. amazonensis, the two chamigrene sesquiterpne compounds (elatol and obtusol) were strongly active against the promastigote form (IC50 = 9.7 μg/mL and 6.2 μg/mL, respectively). Furthermore, they were strongly active against the intracellular amastigote form (IC50 = 4.5 μg/mL and 3.9 μg/mL) with low cytotoxicity. Althoough the triquinane sesquiterpene compound, silphiperfol-5-en-3-ol was also active, it was less active against both the promastigote (IC50 = 43.8 μg/mL) and the intracellular amastigote (IC50 = 48.7 μg/mL) forms. [26,60]. These similar IC50 values of elatol and obtusol is probably related to the presence of cyclohexane ring and chloride and bromine atoms that are not found in the triquinane sesquiterpene compound. None of these three sesquiterpnes significantly activated the production of nitric oxide by infected macrophages, suggesting that their antileishmanial activity is likely to be direct on the parasites rather than through macrophage activation [60]. Elatol induced the parasite’s killing through significant changes on parasite, including pronounced swelling of the mitochondrion, appearance of concentric membrane structures inside the organelle; destabilization of the plasma membrane and formation of autophagic vacuoles [26].
Among the sulfated polysaccharides screened, fucoidan (12, Figure 1), a polyanionic sulfated polysaccharide found in many brown algae was the most interesting. This compound showed an inhibitory effect on intracellular amastigote of L. donovani with 93% inhibition at 50.6 μg/mL. In vivo, a complete elimination of liver and spleen parasite burden was achieved at a dose of 200 mg/kg/day three times daily. Fucoidan was able to induce a protective response from the host by means of the production of cytokines and significant increment in the levels of reactive oxygen species and nitric oxide in infected macrophages, which may be involved in the observed reduction of the parasite multiplication [76]. Despite this promising potential, fucoidan, as a high-molecular-weight product has high hemorrhagic risk, poor solubility and bioavailability [82].
Overall, among the 151 macroalgae that were screened, only extracts from twelve species (Botryoclada occidentalis, Canistrocarpus cervicornis, Caulerpa racemosa, Cystoseira baccata, Dictyota menstrualis, Dictyota pfaffii, Fucus vesiculosus, Gracilaria caudata, Laurencia dendroidea, Lessonia vadosa, Solieria filiformis and Stypopodium zonale) were further investigated for identification of bioactive compounds (Figure 1). This denotes a gap in knowledge that should be filled in. Therefore, an integrated approach of identification of macroalgae with antileishmanial properties followed by identification of bioactive compounds should be undertaken to speed up the research and development of marine algae as sources of druggable molecules for treatment of Leishmania diseases.

4. Approaches Used for Assessment of Antileishmanial Activity by the Authors

Different approaches viz. microscopic, green fluorescent protein, resazurin, MTT, XTT, enzymatic hydrolysis of p-nitrophenyl phosphate and models viz. promastigotes, axenic amastigotes, intracellular amastigotes, and mouse model were used to determine the antileishmanial activity of natural products from marine algae.
In the microscopic assays, Leishmania spp. axenic amastigotes, promastigotes or harboring Green Fluorescent Protein (GFP) were treated with varying concentrations of natural products. After incubation, parasite viability was measured using microscopic counting technique [43,59]. In intracellular antileishmanial assay, differentiated macrophages were incubated in complete medium containing stationary phase Leishmania spp. promastigotes. After incubation, non-internalized promastigotes were removed and the infected macrophages were treated with the natural products. Parasite inhibition was therefore assessed either by microscopic counting using a compound or fluorescent microscope for GFP assay [51,59,60,76].
The resazurin/alamar blue, MTT and XTT assays consisted of incubating Leishmania parasites with plant products in microtiter plate format followed by addition of the dyes (resazurin or MTT or XTT) and further incubation for additional 2–3 h. In the case of the resazurin assay, the blue dye resazurin is reduced to the pink-coloured resorufin in the medium by cell activity (growing parasite); while for MTT and XTT, the yellow tetrazolium salt is reduced into blue-violet and orange formazan, respectively. These assays depend on an easily recognised colour change and parasite viability can be determined visually or measured spectrophotometrically [75,76,83].
Phosphatase activity is based on enzymatic hydrolysis of p-nitrophenyl phosphate followed by measurement of the incorporation of labeled nucleosides into nucleic acids. Phosphatase activity is proportional to the number of surviving parasites [84].
The in vivo assay involved the treatment of mice infected with promastigotes with various concentrations of drugs for a given period of time. Activity against visceral infection is assessed by measuring spleen or liver parasite burdens after giemsa-stained smear observation [76]. The antileishmanial drug screening reported mainly the use of parasite promastigotes and to a lesser extent, axenic amastigotes as they are easily maintained. The marine algae studied appeared to have good antileishmanial activity in vitro against the promastigote and axenic amastigote forms of Leishmania parasites (Table 1 and Table 2). Only 20 species (Anadyomene saldanhae, Bryothamnion triquetrum, Canistrocarpus cervicornis, Caulerpa cupressoides, Ceramium nitens, Chondrococcus hornemanni, Cystoseira baccata, Dictyota mertensii, Dictyota pfaffii, Dictyota sp., Fucus vesiculosus, Halimeda opuntia, Lessonia vadosa, Laurencia aldingensis, Laurencia dendroidea, Ochtodes secundiramea, Padina sp., Palisada flagellifera, Palisada perforata and Stypopodium zonale) were reportedly tested against the intracellular amastigote form. Extracts from some species such as Dictyota sp., O. secundiramea and C. cupressoides showed activity against Leishmania promastigotes, which is the insect vector-based form of the parasite. However, these extracts were inactive on intracellular amastigotes. In fact, the promastigote may not be the appropriate target for an antileishmanial drug due to significant cellular, physiological, biochemical and molecular differences when compared to intracellular amastigotes. Similarly, axenic amastigotes model has been developed to mimic the intracellular parasite stage; however it has been shown that some promising hits against this form of the parasite were inactive on intracellular amastigotes [85,86]. This limitation is due to several differences in cellular processes, including intracellular transport, response to oxidative stress, and metabolism. Moreover, with this model, the natural niche of the parasite, the host-parasite interactions, and the accessibility of the target have not been taken into account. Also, as evidenced in the reports, some marine algae extracts that showed activity against axenic amastigotes were inactive on the intracellular amastigote. This is likely due to their inability to cross host cells membrane or to maintain stability under low pH [87]. Fucoidan (12, Figure 1) was reported to be inactive on promastigotes of L. donovani, but it otherwise showed a good activity against the intracellular amastigote of the parasite [76]. Failure to identify all active compounds and selection of numerous false-positive hits has recently been associated with the use of the insect stage promastigotes and axenic amastigotes in primary screenings. Therefore, subsequent to using promastigotes or axenic amastigotes as models for screening, an important next step in the validation process should involve testing for activity on intracellular amastigotes that represents an appropriate target for an antileishmanial drug.
Microscopy counting method has been used for assaying drugs against intracellular amastigote form of Leishmania parasites (Table 1 and Table 2). Nevertheless, microscopic quantification of parasite burdens is laborious, time-consuming and requires specific processes including staining and microscopic observation. An alternative, the Trypanothione reductase (TryR)-based assay developed by Bogaart et al. [88] is a simple and efficient assay. It is a quantitative colorimetric assay in which the activity of a native enzyme (Trypanothione reductase) of the kinetoplast-unique thiol-redox metabolism is used to assess parasite viability by monitoring its 5,5′-dithiobis 2-nitrobenzoic acid-coupled reducing activity. More recently, another promising method was developed for Leishmania disease drug discovery called ex vivo model, which uses cell explants from infected rodents. This model involves real amastigote-infected organ macrophages with the full repertoire of immune cells that are important in both the pathogenesis of leishmaniasis and healing response to the disease. With ex vivo model, the replication of the intracellular amastigote could easily be quantified by measurement of luciferase activity within a system that mimics the immunopathological environment, which is known to strongly have an impact on parasite replication, killing, and drug efficacy [89,90].
To, date, only Chondrococcus hornemanni, Fucus vesiculosus and Osmundaria obtusiloba have been evaluated for antileishmanial activity using both the in vitro and in vivo models (Table 1 and Table 2). Extracts from O. obtusiloba C. hornemanni and fucoidan (12, Figure 1) isolated from F. vesiculosus showed promising in vitro antileishmanial activities that were confirmed in vivo [48,58,76]. In vitro assays play an essential role in drug discovery process because of their advantages consisting of a simplicity, convenience and short course, as well as limited amounts of samples used. However, most identified potent hits using in vitro assays do not translate their activities when tested in in vivo. In fact, in vivo assays provide an integrated system in which the efficacy of compound can be assessed in the physiological context [91,92] and can provide the combined effect of permeability, distribution, metabolism and excretion, yielding measurable sets of pharmacokinetic parameters and toxicology endpoints [91].

5. Conclusions

Given the shortcomings of existing treatments, there is an urgent need for novel drugs to treat Leishmania diseases. Marine algae have become an important base in research to discover new chemical entities with potential to be developed into drugs. In fact, the metabolic and physiological capabilities of marine algae that allow them to survive in a complex habitat provide a tremendous potential for the production of unique metabolites, which are not found in terrestrial environment. Recent trends in drug research from natural sources have indicated that marine algae are a promising source of novel active compounds, especially those with antileishmanial activity. Indeed, this review has documented the updated list of marine macroalgae and their isolated compounds that have been tested against Leishmania parasites. Compared to terrestrial plants, only few studies have been done with marine macroalgae and only 151 marine macroalgae were tested against Leishmania parasites highlighting a gap in knowledge and stressing the need for extensive attempts to systematically scrutinize these marine raw materials for new antileishmanial drugs. Species from Dictotaceae family and Anadyomene, Laurencia complex, Ulva and Asparagopsis genera are the most interesting macroalgae for antileishmanial drugs discovery. Moreover, analysis of the reports indicates that investigating marine macroalgal compounds has the potential as promising avenue for identifying novel compounds with potent antileishmanial activity and low toxicity. Such examples include elatol, obtusol, (4R,9S,14S)-4α-acetoxy-9β,14α-dihydroxydolast-1(15),7-diene and fucosterol that could be interesting scaffolds for the development of new and effective antileishmanial drugs. However, reports indicate that most marine algae have been tested against the promastigote and axenic amastigote forms of Leishmania parasites, and this might result in many false active natural products. This situation emphasizes the importance of using additional and sensitive advanced methods for drug discovery strategies against leishmaniasis including testing against the intracellular amastigotes, the relevant stage for pathogenesis of the disease. This strategy should avoid selection of fasely active samples or lack of detection of truely active samples and the efficacy study using animal models would be of great value for validation of the results.
Overall, the results reported till date have shown promising antileishmanial extracts/compounds from marine macroalgae that support further exploration for the discovery of new leads with high therapeutical value.

Acknowledgments

The authors gratefully acknowledge the financial support from Bill and Melinda Gates Foundation through the postdoctoral fellowship training program in Infectious Diseases at Noguchi Memorial Institute for Medical Research, Ghana (Global Health Grant number OPP52155). We are also very thankful to Dorcas Osei-Safo for her assistance in drawing figure.

Author Contributions

L.R.T.Y. conceived the topic and developed the manuscript. R.A.-O., P.V.T.F., N.T., F.F.B., A.K.N., M.D.W. reviewed and contributed intellectually for the improvement of the manuscript.

Conflicts of Interest

The authors declare that there is no conflict of interests regarding the publication of this review paper. The funding sponsors had no role in the design.

References

  1. Steverdind, D. The history of Leishmaniasis. Parasit. Vectors 2017, 10, 82. [Google Scholar] [CrossRef] [PubMed]
  2. Alcolea, P.D.; Alonso, A.; Larraga, V. Genome-wide gene expression profile induced by exposure to cadmium acetate in Leishmania infantum promastigotes. Int. Microbiol. 2011, 14, 1–11. [Google Scholar] [PubMed]
  3. Khraiwesh, M.; Leed, S.; Roncal, N.; Johnson, J.; Sciotti, R.; Smith, P.; Rrad, L.; Paris, R.; Hudson, T.; Hickman, M.; et al. Antileishmanial activity of compounds derived from the Medicines for Malaria Venture Open Access Box against intracellular Leishmania major amastigotes. Am. J. Trop. Med. Hyg. 2016, 94, 340–347. [Google Scholar] [CrossRef] [PubMed]
  4. Armeli Minicante, S.; Michelet, S.; Bruno, F.; Castelli, G.; Vitale, F.; Sfriso, A.; Morabito, M.; Genovese, G. Bioactivity of phycocolloids against the mediterranean protozoan Leishmania infantum: An inceptive study. Sustainability 2016, 8, 1131. [Google Scholar] [CrossRef]
  5. Alvar, J.; Vélez, I.D.; Bern, C.; Herrero, M.; Desjeux, P.; Cano, J.; Jannin, J.; den Boer, M. Leishmaniasis Worldwide and Global Estimates of Its Incidence. PLoS ONE 2012, 5, e35671. [Google Scholar] [CrossRef] [PubMed]
  6. Savoia, D. Recent updates and perspectives on leishmaniasis. J. Infect. Dev. Ctries 2015, 6, 588–596. [Google Scholar] [CrossRef] [PubMed]
  7. Haldar, A.K.; Sen, P.; Roy, S. Use of Antimony in the Treatment of Leishmaniasis: Current Status and Future Directions. Mol. Biol. Int. 2011. [Google Scholar] [CrossRef] [PubMed]
  8. World Health Organization (WHO). Control of the Leishmaniasis: Report of the Meeting of the WHO Expert Committee on the Control of Leishmaniases; WHO Technical Report Series; WHO: Geneva, Switzerland, 2010; pp. 22–26. [Google Scholar]
  9. Singh, O.P.; Singh, B.; Chakravarty, J.; Sundar, S. Current challenges in treatment options for visceral leishmaniasis in India: A public health perspective. Infect. Dis. Poverty 2016, 5, 19. [Google Scholar] [CrossRef] [PubMed]
  10. Moore, E.M.; Lockwood, D.N. Treatment of visceral leishmaniasis. J. Glob. Infect. Dis. 2010, 2, 151–158. [Google Scholar] [PubMed]
  11. Mishra, J.; Dey, A.; Singh, N.; Somvanshi, R.; Singh, S. Evaluation of toxicity and therapeutic efficscy of a new liposomal formulation of amphotericin B in a mouse model. Indian J. Med. Sci. 2013, 137, 767–776. [Google Scholar]
  12. Lakshmi, V.; Khare, P.; Misra, P.; Dube, A. Antileishmanial potential of a marine sponge Haliclona oculata against experimental visceral Leishmaniasis. J. Coast. Life Med. 2015, 3, 187–192. [Google Scholar]
  13. Meena, A.K.; Kandale, A.; Rao, M.M.; Panda, P.; Kaur, K.P. A Review on Leishmaniasis and treatment with natural drugs. Available online: https://www.researchgate.net/publication/234002706 (accessed on 20 October 2017).
  14. Soares, D.C.; Calegari-Silva, T.C.; Lopes, U.G.; Teixeira, V.L.; de Palmer Paixão, I.C.N.; Cirne-Santos, C.; Bou-Habib, D.C.; Saraiva, E.M. Dolabelladienetriol, a compound from Dictyota pfaffii algae, inhibits the infection by Leishmania amazonensis. PLoS Negl. Trop. Dis. 2012, 6, e1787. [Google Scholar] [CrossRef] [PubMed]
  15. Dalimi, A.; Delavari, M.; Ghaffarifar, F.; Sadraei, J. In vitro and in vivo antileishmanial effects of aloe-emodin on Leishmania major. J. Tradit. Complement. Med. 2015, 5, 96–99. [Google Scholar] [CrossRef] [PubMed]
  16. Harvey, L.A.; Edrada-Ebel, R.; Quinn, R.J. The re-emergence of natural products for drug discovery in the genomics era. Nat. Rev. Drug Discov. 2015, 14, 111–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Ruiz-Torres, V.; Encinar, J.A.; Herranz-López, M.; Pérez-Sánchez, A.; Galiano, V.; Barrajón-Catalán, E.; Micol, V. An updated review on marine anticancer compounds: The use of virtual screening for the discovery of small-molecule cancer drugs. Molecules 2017, 22, 1037. [Google Scholar] [CrossRef] [PubMed]
  18. Mayer, A.M.; Rodriguez, A.D.; Berlinck, R.G.S.; Fusetani, N. Marine pharmacology in 2007–8: Marine compounds with antibacterial, anticoagulant, antifungal, antiinflammatory, antimalarial, antiprotozoal, antituberculosis, and antiviral activities: Affecting the immune and nervous system, and other miscellaneous mechanisms of action. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2011, 153, 191–222. [Google Scholar] [PubMed]
  19. Newman, D.J.; Cragg, G.M. Marine natural products and related compounds in clinical and advanced preclinical trials. J. Exp. Mar. Biol. Ecol. 2004, 67, 1216–1238. [Google Scholar] [CrossRef] [PubMed]
  20. Molinski, T.F.; Dalisay, D.S.; Lievens, S.L.; Saludes, J.P. Drug development from marine natural products. Nat. Rev. Drug Discov. 2009, 8, 69–85. [Google Scholar] [CrossRef] [PubMed]
  21. Lauritano, C.; Aenderson, H.J.; Hansen, E.; Albrigtsen, M.; Escarlera, L.; Esposito, F.; Helland, K.; Hansen, Ø.K.; Romano, G.; Ianora, A. Bioactivity screening of microalgae for antioxidant, anti-inflammatory, anticancer, anti-Diabetes, and antibacterial activities. Front. Mar. Sci. 2016, 3, 68. [Google Scholar] [CrossRef]
  22. Bajpai, V.K. Antimicrobial bioactive compounds from marine algae: A mini review. Indian J. Geomar. Sci. 2016, 45, 1076–1085. [Google Scholar]
  23. Singh, R.D.; Mody, S.K.; Patel, H.B.; Devi, S.; Sarvaiya, V.N.; Patel, H.A.; Patel, B.R. Antimicrobial drug discovery: Evident shifting from terrestrial to marine micro-organisms. Int. J. Curr. Microbiol. App. Sci. 2017, 6, 2322–2327. [Google Scholar] [CrossRef]
  24. Rovirosa, J.; Sepulveda, M.; Quezada, E.; San-Martin, A. Isoepitaondiol, a diterpenoid of Stypopodium flabelliforme and the insecticidal activity of stypotriol Epitaondiol and derivatives. Phytochemistry 1992, 31, 2679–2681. [Google Scholar] [CrossRef]
  25. Sabina, H.; Tasneem, S.; Kausar, Y.; Choudhary, M.I.; Aliya, R. Antileishmanial activity in the crude extract of various seaweed from the coast of Karachi, Pakistan. Pakistan J. Bot. 2005, 37, 163–168. [Google Scholar]
  26. dos Santos, A.O.; Veiga-Santos, P.; Ueda-Nakamura, T.; Filho, B.P.D.; Sudatti, B.D.; Bianco, M.É.; Pereira, C.R.; Nakamura, V.C. Effect of Elatol, isolated from red seaweed Laurencia dendroidea, on Leishmania amazonensis. Mar. Drugs 2010, 8, 2733–2743. [Google Scholar] [CrossRef] [PubMed]
  27. Torres, F.A.E.; Passalacqua, T.G.; Velásquez, A.M.A.; de Souza, R.A.; Colepicolo, P.; Graminha, M.A.S. New drugs with antiprotozoal activity from marine algae: A review. Rev. Bras. Farmacogn. 2014, 24, 265–276. [Google Scholar] [CrossRef]
  28. Gallé, J.B.; Attioua, B.; Kaiser, M.; Rusig, A.M.; Lobstein, A.; Vonthron-Sénécheau, C. Eleganolone, a diterpene from the french marine alga Bifurcaria bifurcata inhibits growth of the human pathogens Trypanosoma brucei and Plasmodium falciparum. Mar. Drugs 2013, 11, 599–610. [Google Scholar] [CrossRef] [PubMed]
  29. Bianco, E.M.; de Oliveira, S.Q.; Rigotto, C.; Tonini, M.L.; Guimaraes, T.R.; Bittencourt, F.; Gouvea, L.P.; Aresi, C.; de Almeida, R.M.T.; Moritz, G.M.I.; et al. Anti-infective potential of marine invertebrates and seaweeds from the brazilian coast. Molecules 2013, 18, 5761–5778. [Google Scholar] [CrossRef] [PubMed]
  30. Pereira, H.; Custódio, L.; Rodrigues, M.J.; de Sousa, C.B.; Oliveira, M.; Barreira, L.; Neng, D.N.; Nogueira, J.M.F.; Alrokayan, S.A.; Mouffouk, F.; et al. Biological activities and chemical composition of methanolic extracts of selected autochthonous microalgae strains from the red sea. Mar. Drugs 2015, 13, 3531–3549. [Google Scholar] [CrossRef] [PubMed]
  31. Milledge, J.J.; Smith, B.; Dyer, P.W.; Harvey, P. Macroalgae-Derived Biofuel: A Review of methods of energy extraction from seaweed biomass. Energies 2014, 7, 7194–7222. [Google Scholar] [CrossRef] [Green Version]
  32. Suganya, T.; Varman, M.; Masjuki, H.H.; Renganathan, S. Macroalgae and microalgae as a potential source for commercial applications along with biofuels production: A biorefinery approach. Renew. Sustain. Energy Rev. 2016, 55, 909–941. [Google Scholar] [CrossRef]
  33. Mimouni, V.; Ulmann, L.; Haimeur, A.; Guéno, F.; Meskini, N.; Tremblin, G. Marine microalgae used as food supplements and their implication in preventing cardiovascular diseases. OCL 2012, 22, 4. [Google Scholar] [CrossRef]
  34. El Gamal, A.A. Biological importance of marine algae. Saudi Pharm. J. 2010, 18, 1–25. [Google Scholar] [CrossRef] [PubMed]
  35. Kyoung, H.K.; Seo, H.H.; Park, Y. Marine peptides and their anti-infective activities. Mar. Drugs 2015, 13, 618–654. [Google Scholar] [CrossRef] [PubMed]
  36. Noel, V.T.; Se-Kwon, K. Beneficial effects of marine algal compounds in cosmeceuticals. Mar. Drugs 2013, 11, 146–164. [Google Scholar]
  37. Anand, N.; Rachel, D.; Thangaraju, N.; Anantharaman, P. Potential of marine algae (seaweeds) as source of medicinally important compounds. Plant Genet. Resour. 2016, 14, 303–313. [Google Scholar] [CrossRef]
  38. Plouguerné, E.; da Gama, B.A.P.; Pereira, R.C.; Barreto-Bergter, E. Glycolipids from seaweeds and their potential biotechnological applications. Front. Cell. Infect. Microbiol. 2014, 4, 174. [Google Scholar] [CrossRef] [PubMed]
  39. Sahayaraj, K. Biological values and conservation of marine algae: An overview. In Proceedings of the Conservation and Sustainable Utilization of Marine Resources, National Conference on Conservation and Sustainable Utilization of Marine Resources, Tamil Nadu, India, 22–23 January 2015; pp. 21–24. [Google Scholar]
  40. Spavieri, J.; Allmendinger, A.; Kaiser, M.; Casey, R.; Hingley-Wilson, S.; Lalvani, A.; Guiry, M.D.; Gerald, B.; Tasdemir, D. Antimycobacterial, antiprotozoal and cytotoxic potential of twenty-one brown algae (Phaeophyceae) from british and irish waters. Phytother. Res. 2010, 24, 1724–1729. [Google Scholar] [CrossRef] [PubMed]
  41. Vonthron-Sénécheau, C.; Kaiser, M.; Devambez, I.; Vastel, A.; Mussio, I.; Rusig, A.M. Antiprotozoal activities of organic extracts from French marine seaweeds. Mar. Drugs 2011, 9, 922–933. [Google Scholar] [CrossRef] [PubMed]
  42. Ainane, T.; Abourriche, A.; Bennamara, A.; Charrouf, M.; Lemrani, M. Anti-leishmanial activity of extracts from a brown seaweed Bifurcaria bifurcata the Atlantic coast of Morocco. Phytothérapie 2015, 1–6. [Google Scholar] [CrossRef]
  43. Dos Santos, A.O.; Britta, E.A.; Bianco, E.M.; Ueda-Nakamura, T.; Filho, B.P.D.; Pereira, R.C.; Nakamura, C.V. 4-Acetoxydolastane diterpene from the Brazilian brown alga Canistrocarpus cervicornis as antileishmanial agent. Mar. Drugs 2011, 9, 2369–2383. [Google Scholar] [CrossRef] [PubMed]
  44. De Souza, C.B.; Katkam, G.N.; Morais, T.R.; Conserva, G.A.A.; Vizetto-Duarte, C.; Pereira, H.; Laurenti, M.D.; Campino, L.; Levy, D.; Uemi, M.; et al. Antileishmanial activity of meroditerpenoids from the macroalgae Cystoseira baccata. Exp. Parasitol. 2017, 174, 1–9. [Google Scholar] [CrossRef] [PubMed]
  45. Süzgeç-Selçuk, S.; Mericli, A.H.; Guven, K.C.; Kaiser, M.; Casey, R.; Hingley-Wilson, S.; Lalvani, A.; Tasdemir, D. Evaluation of Turkish seaweeds for antiprotozoal, antimycobacterial and cytotoxic activities. Phytother. Res. 2011, 25, 778–783. [Google Scholar] [CrossRef] [PubMed]
  46. Freile-Pelegrin, Y.; Robledo, D.; Chan-Bacab, M.J.; Ortega-Morales, B.O. Antileishmanial properties of tropical marine algae extracts. Fitoterapia 2008, 79, 374–377. [Google Scholar] [CrossRef] [PubMed]
  47. Lira, M.L.F.; Lopes, R.; Gomes, A.P.; Barcellos, G.; Verícimo, M.; Osako, K.; Ortiz-Ramirez, F.A.; Ramos, C.J.B.; Cavalcanti, D.N.; Teixeira, V.L.; et al. Anti-leishmanial activity of Brazilian green, brown, and red algae. J. Appl. Phycol. 2015, 28, 591–598. [Google Scholar] [CrossRef]
  48. Orhan, I.; Sener, B.; Atici, T.; Brun, R.; Perozzo, R.; Tasdemir, D. Turkish freshwater and marine macrophyte extracts show in vitro antiprotozoal activity and inhibit FabI, a key enzyme of Plasmodium falciparum fatty acid biosynthesis. Phytomedicine 2006, 13, 388–393. [Google Scholar] [CrossRef] [PubMed]
  49. Dos Santos, A.A.S.; dos Anjos, K.F.; de Vasconcelos, R.T.N.; Higino, T.M.; Brelaz-de-Castro, M.C.; Bianco, E.M.; de Figueiredo, R.C. The in vitro biological activity of the Brazilian brown seaweed Dictyota mertensii against Leishmania amazonensis. Molecules 2014, 19, 14052–14065. [Google Scholar]
  50. Fouladvand, M.; Barazesh, A.; Farokhzad, F.; Malekizadeh, H.; Sartavi, K. Evaluation of in vitro anti-leishmanial activity of some brown, green and red algae from the Persian Gulf. Eur. Rev. Med. Pharmacol. Sci. 2011, 15, 597–600. [Google Scholar] [PubMed]
  51. Soares, D.C.; Szlachta, M.M.; Teixeira, V.L.; Soares, A.R.; Saraiva, E.M. The brown alga Stypopodium zonale (Dictyotaceae): A potential source of Anti-Leishmania drugs. Mar. Drugs 2016, 14, 163. [Google Scholar] [CrossRef] [PubMed]
  52. Genovese, G.; Tedone, L.; Hamann, M.; Morabito, M. The Mediterranean Red Alga Asparagopsis: A Source of Compounds against Leishmania. Mar. Drugs 2009, 7, 361–366. [Google Scholar] [CrossRef] [PubMed]
  53. Vitale, F.; Genovese, G.; Bruno, F.; Castelli, G.; Piazza, M.; Migliazzo, A.; Morabito, M. Effectiveness of the red alga Asparagopsis taxiformis extracts against Leishmania infantum. Open Life Sci. 2015, 10, 490–496. [Google Scholar] [CrossRef]
  54. Allmendinger, A.; Spavieri, J.; Kaiser, M.; Casey, R.; Hingley-Wilson, S.; Lalvani, A.; Guiry, M.; Blunden, G.; Tasdemir, D. Antiprotozoal, antimycobacterial and cytotoxic potential of twenty-three british and irish red algae. Phytother. Res. 2010, 24, 1099–1103. [Google Scholar] [CrossRef] [PubMed]
  55. Sabina, H.; Aliya, R. Bioactive assessment of selected marine red algae against Leishmania major and chemical constituents of Osmundea pinnatifida. Pak. J. Bot. 2011, 43, 3053–3056. [Google Scholar]
  56. De Felício, R.; de Albuquerque, S.; Young, M.C.M.; Yokoya, N.S.; Debonsia, H.M. Trypanocidal, leishmanicidal and antifungal potential from marine red alga Bostrychia tenella J. Agardh (Rhodomelaceae, Ceramiales). J. Pharm. Biomed. Anal. 2010, 52, 763–769. [Google Scholar] [CrossRef] [PubMed]
  57. Parra, M.G.; Fidalgo, L.M.; Pasarón, O.C.; Delgado, N.G.; Hernández, A.P. Antileishmanial activity of six extracts from marine organisms. Rev. Cuba. Med. Trop. 2012, 64, 61–64. [Google Scholar]
  58. Lakshmi, V.; Khare, P.; Misra, P.; Srivastava, M.N.; Dube, A. Antileishmanial potential of Chondrococcus hornemanni against experimental visceral leishmaniasis. J. Mar. Biol. Oceanogr. 2014, 3, 4. [Google Scholar]
  59. Da Silva Machado, F.L.; Pacienza-Lima, W.; Duarte, H.M.; Rossi-Bergmann, B.; Gestinari, L.M.; Fujii, M.T.; Kaiser, C.R.; Soares, A.R. Chemical diversity and antileishmanial activity of crude extracts of Laurencia complex (Ceramiales, Rhodophyta) from Brazil. Rev. Bras. Farmacogn. 2014, 24, 635–643. [Google Scholar] [CrossRef]
  60. Da Silva Machado, F.L.; Pacienza-Limac, W.; Rossi-Bergmann, B.; de Souza Gestinari, L.M.; Fujii, M.T.; de Paula, J.C.; Costa, S.S.; Lopes, N.P.; Kaiser, C.R.; Soares, A.R. Antileishmanial sesquiterpenes from the Brazilian red alga Laurencia dendroidea. Planta Med. 2011, 77, 733–735. [Google Scholar] [CrossRef] [PubMed]
  61. Spavieri, J.; Kaiser, M.; Casey, R.; Hingley-Wilson, S.; Lalvani, A.; Blunden, G.; Tasdemir, D. Antiprotozoal, antimycobacterial and cytotoxic potential of some british green algae. Phytother. Res. 2010, 24, 1095–1098. [Google Scholar] [CrossRef] [PubMed]
  62. Glombitza, K.W.; Roesener, H.U.; Koch, M.L. Antibiotics from algae. Part 16. Polyhydroxyoligophenyls and phenyl ether from Bifurcaria bifurcata. Phytochemistry 1976, 15, 1279–1281. [Google Scholar] [CrossRef]
  63. Valls, R.; Piovetti, L.; Deffo, P. Analysis of sterols and diterpenoids of brown algae (Cystoseiraceae). Oceanis 1991, 17, 305–307. [Google Scholar]
  64. Culioli, G.; Ortalo-Magne, A.; Daoudi, M.; Thomas-Guyon, H.; Valls, R.; Piovetti, L. Trihydroxylated linear diterpenes from the brown alga Bifurcaria bifurcata. Phytochemistry 2004, 65, 2063–2069. [Google Scholar] [CrossRef] [PubMed]
  65. Ortalo-Magne, A.; Culioli, G.; Valls, R.; Pucci, B.; Piovetti, L. Polar acyclic diterpenoids from Bifurcaria bifurcata (Fucales, Phaeophyta). Phytochemistry 2005, 66, 2316–2323. [Google Scholar] [CrossRef] [PubMed]
  66. Göthel, Q.; Lichte, E.; Köck, M. Further eleganolone-derived diterpenes from the brown alga Bifurcaria bifurcata. Tetrahedron Lett. 2012, 53, 1873–1877. [Google Scholar] [CrossRef]
  67. Meena, A.K.; Kandale, A.; Nigam, S.; Panda, P.; Singh, B.; Rao, M. Review on Marine organisms with antileishmanial activity. J. Pharm. Res. 2010, 3, 818–821. [Google Scholar]
  68. Suzuki, M.; Vairappan, C.S. Halogenated secondary metabolites from Japanese species of the red algal genus Laurencia (Rhodomelaceae, Ceramiales). Curr. Top. Phytochem. 2005, 7, 1–34. [Google Scholar]
  69. Alarif, W.M.; Al-Lihaibi, S.S.; Abdel-Lateff, A.; Ayyad, S.E. New antifungal cholestane and aldehyde derivatives from the red alga Laurencia papillosa. Nat. Prod. Commun. 2011, 6, 1821–1824. [Google Scholar] [PubMed]
  70. Al-Massarani, S.M. Phytochemical and biological properties of sesquiterpene constituents from the marine red seaweed Laurencia: A review. Nat. Prod. Chem. Res. 2014, 2, 147. [Google Scholar]
  71. Aguilar-Briseño, J.A.; Cruz-Suarez, L.E.; Sassi, J.F.; Ricque-Marie, D.; Zapata-Benavides, P.; Mendoza-Gamboa, E.; Rodríguez-Padilla, C.; Trejo-Avila, L.M. Sulphated Polysaccharides from Ulva clathrata and Cladosiphon okamuranus Seaweeds both Inhibit Viral Attachment/Entry and Cell-Cell Fusion, in NDV Infection. Mar. Drugs 2015, 13, 697–712. [Google Scholar] [CrossRef] [PubMed]
  72. Kosanić, M.; Ranković, B.; Stanojković, T. Biological activities of two macroalgae from Adriatic coast of Montenegro. Saudi J. Biol. Sci. 2015, 22, 390–397. [Google Scholar] [CrossRef] [PubMed]
  73. Yung-Qing, T.; Mahmood, K.; Shehzadi, R.; Ashraf, M.F. Ulva lactuca and its polysaccharides: Food and biomedical aspects. J. Biol. Agric. Healthc. 2016, 6, 2224–3208. [Google Scholar]
  74. Smyrniotopoulos, V.; Merten, C.; Kaiser, M.; Tasdemir, D. Bifurcatriol, a new antiprotozoal acyclic diterpene from the brown alga Bifurcaria bifurcata. Mar. Drugs 2017, 15, 245. [Google Scholar] [CrossRef] [PubMed]
  75. Becerra, M.; Boutefnouchet, S.; Córdoba, O.; Vitorino, G.P.; Brehu, L.; Lamour, I.; Laimay, F.; Efstathiou, A.; Smirlis, D.; Michel, S.; et al. Antileishmanial activity of fucosterol recovered from Lessonia vadosa Searles (Lessoniaceae) by SFE, PSE and CPC. Phytochem. Lett. 2015, 11, 418–423. [Google Scholar] [CrossRef]
  76. Kar, S.; Sharma, G.; Das, P.K. Fucoidan cures infection with both antimony-susceptible and -resistant strains of Leishmania donovani through Th1 response and macrophage-derived oxidants. J. Antimicrob. Chemother. 2011, 66, 618–625. [Google Scholar] [CrossRef] [PubMed]
  77. Lehnhardt Pires, C.; Rodrigues, S.D.; Bristot, D.; Gaeta, H.H.; Toyama, D.O.; Farias, W.R.L.; Toyama, M.H. Evaluation of macroalgae sulfated polysaccharides on the Leishmania (L.) amazonensis Promastigote. Mar. Drugs 2013, 11, 934–943. [Google Scholar] [CrossRef] [PubMed]
  78. Vallim, M.A.; Paula, J.C.D.; Pereira, R.C.; Teixeira, V.L. The diterpenes from Dictyotacean marine brown algae in the Tropical Atlantic American region. Biochem. Syst. Ecol. 2015, 33, 1–16. [Google Scholar] [CrossRef]
  79. Caamal-Fuentes, E.; Moo-Puc, R.; Freile-Pelegrín, Y.; Robledo, D. Cytotoxic and antiproliferative constituents from Dictyota ciliolata, Padina sanctae-crucis and Turbinaria tricostata. Pharm. Biol. 2014, 52, 1–5. [Google Scholar] [CrossRef] [PubMed]
  80. De Andrade Moura, L.; Almeida, A.C.M.; Domingos, T.F.; Ortiz-Ramirez, F.; Cavalcanti, D.N.; Teixeira, V.L.; Fuly, A.L. Antiplatelet and anticoagulant effects of diterpenes isolated from the marine alga, Dictyota menstrualis. Mar. Drugs 2014, 12, 2471–2484. [Google Scholar] [CrossRef] [PubMed]
  81. Soares, A.R.; Duarte, H.M.; Tinnoco, L.W.; Pereira, R.C.; Teixeira, V.L. Intraspecific variation of meroditerpenoids in the brown alga Stypopodium zonale guiding the isolation of new compounds. Rev. Bras. Farmacogn. 2015, 25, 627–633. [Google Scholar] [CrossRef]
  82. Zhao, X.; Guo, F.; Jing, H.J.; Zhang, L.; Xue, C.; Zhang, Z.; Li, B. Antithrombotic activity of oral administered low molecular weight fucoidan from Laminaria japonica. Thromb. Res. 2016, 144, 46–52. [Google Scholar] [CrossRef] [PubMed]
  83. Bafghi, A.F.; Samimi, H.; Jamshidi, H.R.; Dehghani, A.; Zarezadeh, R. Antileishmanial activity of Yazd Spirogyra spp extracts against Leishmania (L.) major [MRHO/IR/75/ER] promastigotes: An in vitro study. J. Chem. Pharm. Res. 2017, 9, 112–117. [Google Scholar]
  84. Bodley, A.L.; McGarry, M.W.; Shapiro, T.A. Drug cytotoxicity assay for African Trypanosomes and Leishmania Species. J. Infect. Dis. 1995, 172, 1157–1159. [Google Scholar] [CrossRef] [PubMed]
  85. De Muylder, G.; Ang, K.K.H.; Chen, S.; Arkin, M.R.; Engel, C.J.; Mckerrow, J.H. A screen against Leishmania intracellular amastigotes: Comparaison to a promastigote screen and indentification of a host cell-specific hit. PLoS Negl. Trop. Dis. 2011, 5, e1253. [Google Scholar] [CrossRef] [PubMed]
  86. De Rycker, M.; Hallyburton, I.; Thomas, J.; Campbell, L.; Wyllie, S.; Joshi, D.; Cameron, S.; Gilbert, H.I.; Watt, G.P.; Frearson, A.J.; et al. Comparison of a high-throughput high-content intracellular Leishmania donovani assay with an axenic amastigote assay. Antimicrob. Agents Chemother. 2013, 57, 2913–2922. [Google Scholar] [CrossRef] [PubMed]
  87. França, P.H.B.; da Silva-Júnior, E.F.; Santos, B.V.O.; Alexandre-Moreira, M.S.; Quintans-Júnior, L.J.; de Aquino, T.M.; de Araújo-Júnior, J.X. Antileishmanial marine compounds: A review. Rec. Nat. Prod. 2017, 11, 92–113. [Google Scholar]
  88. Van den Bogaart, E.; Schoone, G.J.; England, P.; Faber, D.; Orrling, K.M.; Dujardin, J.C.; Sundar, S.; Schallig, H.D.; Adams, E.R. Simple colorimetric trypanothione reductase-based assay for high-throughput screening of drugs against Leishmania intracellular amastigotes. Antimicrob. Agents Chemother. 2014, 58, 527–535. [Google Scholar] [CrossRef] [PubMed]
  89. Peniche, A.G.; Osorio, Y.; Renslo, A.R.; Frantz, D.E.; Melby, P.C.; Travi, B.L. Development of an ex vivo lymph node explant model for identification of novel molecules active against Leishmania major. Antimicrob. Agents Chemother. 2014, 58, 78–87. [Google Scholar] [CrossRef] [PubMed]
  90. Peniche, A.G.; Renslo, A.R.; Melby, P.C.; Travi, B.L. Antileishmanial activity of disulfiram and thiuram disulfide analogs in an ex vivo model system is selectively enhanced by the addition of divalent metal ions. Antimicrob. Agents Chemother. 2015, 59, 6463–6470. [Google Scholar] [CrossRef] [PubMed]
  91. Jiménez-Díaz, M.B.; Viera, S.; Ibáñez, J.; Mulet, T.; Magán-Marchal, N.; Garuti, H.; Gómez, V.; Cortés-Gil, L.; Martínez, A.; Ferrer, S.; et al. A new in vivo screening paradigm to accelerate antimalarial drug discovery. PLoS ONE 2013, 8, e66967. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, D.; Luo, G.; Ding, X.; Lu, C. Preclinical experimental models of drug metabolism and disposition in drug discovery and development. Acta Pharm. Sin. B 2012, 2, 549–561. [Google Scholar] [CrossRef]
Figure 1. Structures of isolated compounds from marine macroalgae.
Figure 1. Structures of isolated compounds from marine macroalgae.
Marinedrugs 15 00323 g001aMarinedrugs 15 00323 g001bMarinedrugs 15 00323 g001c
Table 1. Antileishmanial activity of extracts from marine macroalgae.
Table 1. Antileishmanial activity of extracts from marine macroalgae.
Species of Marine AlgaeExtraction SolventLeishmania SpeciesParasites StageMethod of Activity EvaluationActivity (IC50 or % of Inhibition); Cytotoxicity (CC50)References
Phaeophyceae
Ascophyllum nodosum (Linnaeus) Le JolisIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay66.3 μg/mL(>90.0 μg/mL)[40]
Bifurcaria bifurcata R. RossIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay6.4 μg/mL (32.7 μg/mL)[40]
Ethyl acetate extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay3.8 μg/mL (6.0 μg/mL)[41]
Hexane extractL. infantumPromastigoteIn vitro: MTT assay46.83 μg/mL (21.8 μg/mL)[42]
Ether extractL. infantumPromastigoteIn vitro: MTT assay51.64 μg/mL (40.46 μg/mL)[42]
Chloroform extractL. infantumPromastigoteIn vitro: MTT assay63.83 μg/mL (37.0 μg/mL)[42]
Canistrocarpus cervicornis (Kützing) De Paula and De ClerckAcetate extractL. amazonensisPromastigoteIn vitro: Microscopic counting50.0 μg/mL (50.0 μg/mL)[43]
Methanol extractL. amazonensisPromastigoteIn vitro: Microscopic counting100.0 μg/mL (51.0 μg/mL)[43]
Dichloromethane extractL. amazonensisPromastigoteIn vitro: Microscopic counting20.0 μg/mL (46.0 μg/mL)[43]
Ethyl acetate fractionL. amazonensisPromastigoteIn vitro: Microscopic counting8.0 μg/mL (47.0 μg/mL)[43]
Acetone extractL. braziliensisPromastigoteIn vitro: Microscopic counting85.8% inhibition at 50.0 μg/mL (>50.0 μg/mL)[29]
Chorda filum (Linnaeus) StackhouseIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay21.1 μg/mL (>90.0 μg/mL)[40]
Colpomenia peregrina SauvageauIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay29.1 μg/mL (>90.0 μg/mL)[40]
Cystoseira baccata (S.G. Gmelin) P.C.Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay15.7 μg/mL (>90.0 μg/mL)[40]
Hexane extractL. infantumPromastigoteIn vitro: MTT assay74.0% inhibition at 250.0 μg/mL (ND)[44]
Cystoseira barbata (Stackhouse) C. AgardhMethanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay23.46–69.98 μg/mL (>90.0 μg/mL)[45]
Cystoseira crinata (Desf.) DubyMethanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay28.21 μg/mL (>90.0 μg/mL)[45]
Cystoseira tamariscifolia (Hudson) Papenf.Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay19.6 μg/mL (62.5 μg/mL)[40]
Dictyopteris polypodioides (A.P. de Candolle) J.V. LamourouxEthyl acetate extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay10.8 μg/mL (87.0 μg/mL)[41]
Dictyota caribaea Hörnig and SchnetterOrganic extractL. mexicanaPromastigoteIn vitro: Microscopic counting24.4 μg/mL (≥1000.0 μg/mL)[46]
Dictyota ciliolata Sonder ex KützingHexane extractL. amazonensisPromastigoteIn vitro: MTT assay1.15 μg/mL (23.5 μg/mL)[47]
Ethyl acetate extractL. amazonensisPromastigoteIn vitro: MTT assay4.22 μg/mL (19.0 μg/mL)[47]
Dictyota dichotoma (Hudson) J.V. LamourouxEthanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay52.0 μg/mL (>90.0 μg/mL)[48]
Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay42.4 μg/mL (>90.0 μg/mL)[40]
Ethyl acetate extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay8.8 μg/mL (27.0 μg/mL)[41]
Dictyota menstrualis (Hoyt) Schnetter, Hörning and Weber-PeukerEthyl acetate extractL. amazonensisPromastigoteIn vitro: MTT assay0.7–0.75 μg/mL (18.3–24.7 μg/mL)[47]
Hexane extractL. amazonensisPromastigoteIn vitro: MTT assay0.61 μg/mL (18.2 μg/mL)[47]
Dictyota mertensii (Martius) KützingDichloromethane/methanol extractL. amazonensisPromastigoteIn vitro: Microscopic counting71.60 μg/mL (233.10 μg/mL)[49]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting81.4 μg/mL (233.10 μg/mL)[49]
Dictyota sp.Dichloromethane/methanol extractL. braziliensisPromastigoteIn vitro: MTT assay93.3% inhibition at 50.0 μg/mL (>50.0 μg/mL)[29]
Fucus ceranoides (Linnaeus)Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay25.3 μg/mL (>90.0 μg/mL)[40]
Fucus serratus (Linnaeus)Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay34.1 μg/mL (>90.0 μg/mL)[40]
Fucus spiralis (Linnaeus)Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay34.3 μg/mL (>90.0 μg/mL)[40]
Fucus vesiculosus (Linnaeus)Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay33.0 μg/mL (>90.0 μg/mL)[40]
Halidrys siliquosa (Linnaeus) Lyngb.Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay8.6 μg/mL (45.0 μg/mL)[40]
Himanthalia elongata (Linnaeus) S.F. GrayIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay64.7 μg/mL (>90.0 μg/mL)[40]
Laminaria digitata (Hudson) J.V. LamourouxIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay34.5 μg/mL (>90.0 μg/mL)[40]
Leathesia difformis (Linnaeus) Aresch.Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay77.4 μg/mL (>90.0 μg/mL)[40]
Lobophora variegata (J.V. Lamouroux) Womersley ex E.C. OliveiraOrganic extractsL. mexicanaPromastigoteIn vitro: Microscopic counting49.9 μg/mL (≥1000.0 μg/mL)[46]
Padina sp.Acetone extractL. braziliensisPromastigoteIn vitro: MTT assay80.9% inhibition at 50.0 μg/mL (300.4 μg/mL)[29]
L. braziliensisIntracellular amastigoteIn vitro: Microscopic counting40.2 μg/mL (300.4 μg/mL)[29]
Pelvetia canaliculata (Linnaeus) Decaisne and ThuretIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay35.7 μg/mL (>90.0 μg/mL)[40]
Pylaiella littoralis (Linnaeus) Kjellm.Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay47.1 μg/mL (>90.0 μg/mL)[40]
Saccorhiza polyschides (Lightf.) Batt.Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay31.8 μg/mL (>90.0 μg/mL)[40]
Sargassum muticum (Yendo) FensholtPolysaccharide extractsL. infantumPromastigoteIn vitro: MTT assay~98.5% inhibition at 160.0 μg/mL (ND)[4]
Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay34.7 μg/mL (>90.0 μg/mL)[40]
Sargassum natans (Linnaeus) Gaill.Ethanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay90.9 μg/mL (>90.0 μg/mL)[48]
Sargassum oligocystum MontagneHot water extractL. majorPromastigoteIn vitro: MTT assay78.0 μg/mL (ND)[50]
Scinaia furcellata (Turn.) J. AgardhEthanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay64.4 μg/mL (>90.0 μg/mL)[48]
Scytosiphon lomentaria (Lyngb.) LinkIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay34.3 μg/mL (>90.0 μg/mL)[40]
Stypocaulon scoparium (L.) KützingIsopropyl alcohol- Trichloromethane/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay30.4 μg/mL (>90.0 μg/mL)[40]
Stypopodium zonale (J.V. Lamouroux) PapenfussDichloromethane extractL. amazonensisPromastigoteIn vitro: Microscopic counting100.0% inhibition at 10.0 μg/mL (>50.0 μg/mL)[51]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting0.27 μg/mL (>50.0 μg/mL)[51]
Ethyl acetate-hexane extractL. amazonensisPromastigoteIn vitro: MTT assay0.75 μg/mL (29.5 μg/mL)[47]
Turbinaria turbinata (Linnaeus) KuntzeOrganic extractsL. mexicanaPromastigoteIn vitro: Microscopic counting10.9 μg/mL (≥1000.0 μg/mL)[46]
Undaria pinnatifida (Harvey) SuringarPolysaccharide extractsL. infantumPromastigoteIn vitro: MTT assay~97.5% inhibition at 160.0 μg/mL (ND)[4]
Rhodophyta
Asparagopsis armata HarveyEthanol-hexane:ethyl acetate fractionL. donovaniPromastigoteIn vitro: Resazurin assay10.0 μg/mL (ND)[52]
Ethanol-ethyl acetate fractionL. donovaniPromastigoteIn vitro: Resazurin assay19.0 μg/mL (ND)[52]
Asparagopsis taxiformis (Delile) TrevisanHexane extractL. donovaniPromastigoteIn vitro: Resazurin assay17.0 μg/mL (ND)[52]
Dichloromethane extractL. donovaniPromastigoteIn vitro: Resazurin assay16.0 μg/mL (ND)[52]
Ethanol-hexane:ethyl acetate fractionL. donovaniPromastigoteIn vitro: Resazurin assay14.0 μg/mL (ND)[52]
Ethanol-ethyl acetate fractionL. donovaniPromastigoteIn vitro: Resazurin assay20.0 μg/mL (ND)[52]
Ethanol extractL. infantumPromastigotesIn vitro: Microscopic counting25.0 μg/mL (>90.0 μg/mL)[53]
L. infantumAxenic amastigotesIn vitro: Microscopic counting9.0 μg/mL (>90.0 μg/mL)[53]
Boergeseniella fruticulosa (Wulfen) KylinIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay26.6 μg/mL (>90.0 μg/mL)[54]
Botryocladia leptopoda (J.Agardh) KylinEthanol extractL. majorPromastigoteIn vitro: Microscopic counting60.81 μg/mL (ND)[25,55]
Bostrychia tenella (J.V. Lamouroux) J. AgardhHexane fractionsL. amazonensisPromastigoteIn vitro: MTT assay17.4 μg/mL (ND)[56]
Hexane subfractions (HO1; HO2; HO3; HO5; HO6)L. amazonensisPromastigoteIn vitro: MTT assay22.2; 1.5; 2.7; 31.7; 66.2 μg/mL (ND)[56]
Dichloromethane subfractions (DO1-DO2)L. amazonensisPromastigoteIn vitro: MTT assay4.4–4.3 μg/mL (ND)[56]
Bryothamnion triquetrum (S.G. Gmelin) M. HoweAqueous extractL. amazonensisPromastigoteIn vitro: Phosphatase assay78.6 μg/mL (258.1 μg/mL)[57]
Calliblepharis jubata (Goodenough and woodward) KützingIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay49.8 μg/mL (>90.0 μg/mL)[54]
Centroceras clavulatum (C. Agardh) MontagneEthanol extractL. majorPromastigoteIn vitro: Microscopic counting57.89–57.85 μg/mL (ND)[25,55]
Ceramium rubrum (Hudson) C. AgardhMethanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay16.76 μg/mL (>90.0 μg/mL)[46]
Ceramium virgatum RothIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay25.6 μg/mL (>90.0 μg/mL)[54]
Chylocladia verticillata (Lightf.) BlidingIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay47.3 μg/mL (>90.0 μg/mL)[54]
Chondrococcus hornemanni (Mert.) SchmitzEthanol extractL. donovaniPromastigoteIn vitro: Green fluorescent protein assay29.5 μg/mL (>200.0 μg/mL)[58]
L. donovaniIntracellular amastigoteIn vitro: Microscopic counting40.6 μg/mL (>200.0 μg/mL)[58]
L. donovani In vivo75.38% suppression at 250.0 μg/mL[58]
n-butanol soluble fraction of ethanol extractL. donovaniPromastigoteIn vitro: Green fluorescent protein assay54.2 μg/mL (>200.0 μg/mL)[58]
L. donovaniIntracellular amastigoteIn vitro: Microscopic counting61.0 μg/mL (>200.0 μg/mL)[58]
Chromatographic fraction of n-butanol soluble fraction (F011)L. donovaniPromastigoteIn vitro: Green fluorescent protein assay53.7 μg/mL (>200.0 μg/mL)[58]
L. donovaniIntracellular amastigoteIn vitro: Microscopic counting57.4 μg/mL (>200.0 μg/mL)[58]
L. donovani In vivo76.2% suppression at 100.0 μg/mL[58]
Chromatographic fraction of n-butanol soluble fraction (F012)L. donovaniPromastigoteIn vitro: Green fluorescent protein assay54.5 μg/mL (>200.0 μg/mL)[58]
L. donovaniIntracellular amastigoteIn vitro: Microscopic counting51.7 μg/mL (>200.0 μg/mL)[58]
L. donovani In vivo56.7% suppression at 100.0 μg/mL[58]
Chondrus crispus StackhouseEthanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay95.0% inhibition at 9.7 μg/mL (84.0 μg/mL)[41]
Claviclonium ovatum (J.V. Lamouroux) Kraft and Min-TheinIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay61.2 μg/mL (>90.0 μg/mL)[54]
Corallina officinalis LinnaeusIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay22.7 μg/mL (88.6 μg/mL)[54]
Corallina granifera Ell. et Sol.Methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay35.02 μg/mL (>90.0 μg/mL)[45]
Cryptopleura ramosa (Hudson) L. NewtonIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay85.6 μg/mL (>90.0 μg/mL)[54]
Cystoclonium purpureum (Hudson) Batt.Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay67.3 μg/mL (>90.0 μg/mL)[54]
Dasya pedicellata (C. Agardh) C. AgardhMethanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay23.04 μg/mL (14.7 μg/mL)[45]
Dilsea carnosa (Schmidel) KuntzeEthyl acetate extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay9.5 μg/mL (74.0 μg/mL)[41]
Dumontia incrassata (O.F. Müll.) J.V. LamourouxIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay68.6 μg/mL (>90.0 μg/mL)[54]
Furcellaria lumbricalis (Hudson) J.V. LamourouxIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay43.3 μg/mL (>90.0 μg/mL)[54]
Gelidium crinale (Hare ex Turner)Methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay19.95 μg/mL (>90.0 μg/mL)[45]
Gelidium pulchellum (Turner) KützingIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay32.5 μg/mL (>90.0 μg/mL)[54]
Gracilaria bursa-pastoris (S.G. Gmelin) P.C. SilvaPolysaccharide extractsL. infantumPromastigoteIn vitro: MTT assay~87.5.0% inhibition at 160.0 μg/mL (ND)[4]
Gracilaria corticata (J. Agardh) J. AgardhEthanol extractL. majorPromastigoteIn vitro: Microscopic counting37.5 μg/mL (ND)[25,55]
Cold water extractL. majorPromastigoteIn vitro: MTT assay65.0 μg/mL (ND)[50]
Hot water extractL. majorPromastigoteIn vitro: MTT assay38.0 μg/mL (ND)[50]
Gracilaria gracilis (Stackhouse) Steentoft, L.M. Irvine and FarnhamIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay53.3 μg/mL (>90.0 μg/mL)[54]
Gracilaria salicornia (C. Agardh) E.Y. DawsonCold water extractL. majorPromastigoteIn vitro: MTT assay74.0 μg/mL (ND)[50]
Hot water extractL. majorPromastigoteIn vitro: MTT assay46.0 μg/mL (ND)[50]
Gracilaria verrucosa (Hudson) PapenfussMethanol extractL. donovaniAxenic amastigotesIn vitro: Resazurin assay36.02 μg/mL (>90.0 μg/mL)[45]
Gracilaria viridis Sfriso, Wolf, Sciuto, Morabito, Andreoli and MoroPolysaccharide extractsL. infantumPromastigoteIn vitro: MTT assay~82.8% inhibition at 160.0 μg/mL (ND)[4]
Halopitys incurvus (Hudson) Batt.Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay16.5 μg/mL (>90.0 μg/mL)[54]
Halurus equisetifolius (Lightf.) KützingIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay69.2 μg/mL (>90.0 μg/mL)[54]
Jania rubens (Linnaeus) J.V. LamourouxIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay60.7 μg/mL (>90.0 μg/mL)[54]
Methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay28.0 μg/mL (>90.0 μg/mL)[45]
Laurencia aldingensis Saito and WomersleyDichloromethane/methanol extractL. amazonensisPromastigoteIn vitro: Green fluorescent protein assay24.5 μg/mL (259.8 μg/mL)[59]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting12.5 μg/mL (259.8 μg/mL)[59]
Laurencia dendroidea (Hudson) J.V. LamourouxLipophilic extractsL. amazonensisPromastigoteIn vitro: Green fluorescent protein assay17.9–34.2 μg/mL (106.2–131.7 μg/mL)[60]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting8.7–10.8 μg/mL (106.2–131.7 μg/mL)[60]
Dichloromethane/methanol extractL. amazonensisPromastigoteIn vitro: Green fluorescent protein assay30.1–97.2 μg/mL (187.0–240.0 μg/mL)[60]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting16.8–22.4 μg/mL (187.0–240.0 μg/mL)[60]
Laurencia microcladia KützingOrganic extractsL. mexicanaPromastigoteIn vitro: Microscopic counting16.3 μg/mL (119.8 μg/mL)[46]
Lomentaria articulata (Hudson) LyngbIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay60.0 μg/mL (>90.0 μg/mL)[54]
Mastocarpus stellatus (Stackhouse) GuiryIsopropyl alcohol- Trichloromethane/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay44.1 μg/mL (>90.0 μg/mL)[54]
Melanothamnus afaqhusainii M. ShameelEthanol extractL. majorPromastigoteIn vitro: Microscopic counting32.6–32.5 μg/mL (ND)[25,55]
Ochtodes secundiramea (Montagne) M. HoweAcetone extractL. braziliensisPromastigoteIn vitro: MTT assay99.7% inhibition at 50.0 μg/mL (>50.0 μg/mL)[29]
Osmundaria obtusiloba (C. Agardh) R.E. NorrisEthanol extractL. amazonensisPromastigoteIn vitro: MTT assay24.5 μg/mL (240.0 μg/mL)[47]
L. amazonensis In vivoActive at 5.0 and 20.0 mg/kg[47]
Ethyl acetate-Hexane extractL. amazonensisPromastigoteIn vitro: MTT assay22.0 μg/mL (198.0 μg/mL)[47]
Osmundea hybrida (A.P. de Candolle) K.W. NamIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay49.2 μg/mL (>90.0 μg/mL)[54]
Osmundea pinnatifida (Hudson) StackhouseEthanol extractL. majorPromastigoteIn vitro: Microscopic counting6.25 μg/mL (ND)[55]
Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay32.7 μg/mL (>90.0 μg/mL)[54]
Palisada flagellifera (J. Agardh) K.W. NamDichloromethane/methanol extractL. amazonensisPromastigoteIn vitro: Green fluorescent protein assay30.7 μg/mL (198.0 μg/mL)[59]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting34.5 μg/mL (198.0 μg/mL)[59]
Palisada perforata (Bory) K.W. NamDichloromethane/methanol extractL. amazonensisPromastigoteIn vitro: Green fluorescent protein assay36.1–46.7 μg/mL (267.0 μg/mL)[59]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting29.7–34.5 μg/mL (267.0 μg/mL)[59]
Plocamium cartilagineum (Linnaeus) P.S. DixonIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay21.2 μg/mL (>90.0 μg/mL)[54]
Polyides rotundus (Hudson) GaillonIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay57.3 μg/mL (>90.0 μg/mL)[54]
Porphyra linearis GrevIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay55.5 μg/mL (>90.0 μg/mL)[54]
Scinaia Fascicularis (Børgesen) HuismanEthanol extractL. majorPromastigoteIn vitro: Microscopic counting59.6 μg/mL (ND)[55]
Scinaia hatei BørgesenEthanol extractL. majorPromastigoteIn vitro: Microscopic counting14.1 μg/mL (ND)[25,55]
Scinaia indica BørgesenEthanol extractL. majorPromastigoteIn vitro: Microscopic counting59.6 μg/mL (ND)[25]
Chlorophyceae
Anadyomene saldanhae A.B. Joly and E.C. OliveiraAcetone extractL. braziliensisPromastigoteIn vitro: MTT assay87.9% inhibition at 50.0 μg/mL (294.2 μg/mL)[29]
L. braziliensisIntracellular amastigoteIn vitro: Microscopic counting23.9 μg/mL (294.2 μg/mL)[29]
Caulerpa racemosa (Forsskål) J. AgardhMethanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay22.66 μg/mL (>90.0 μg/mL)[45]
Ethyl acetate-Hexane extractL. amazonensisPromastigoteIn vitro: MTT assay47.5–154 μg/mL (48.0–115.0 μg/mL)[47]
Caulerpa cupressoides (Vahl) C. AgardhAcetone extractL. braziliensisPromastigoteIn vitro: MTT assay51.7% inhibition at 50.0 μg/mL (>50.0 μg/mL)[29]
Caulerpa sertularioides (S.G. Gmelin) M. HoweHot water extractL. majorPromastigoteIn vitro: MTT assay85.0 μg/mL (ND)[50]
Cladophora rupestris (L.) KützingIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay20.2 μg/mL (>90.0 μg/mL)[61]
Codium bursa (L.) C. AgardhMethanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay31.71 μg/mL (>90.0 μg/mL)[45]
Codium fragile (Sur.) Hariot ssp. Tomentosoides (van Goor) SilvaIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay16.6 μg/mL (>90.0 μg/mL)[61]
Halimeda opuntia (L.) LamourouxAqueousL. amazonensisPromastigoteIn vitro: Phosphatase assay83.5 μg/mL (526.4 μg/mL)[57]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting70.7 μg/mL (526.4 μg/mL)[57]
Ulva intestinalis LinnaeusIsopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay14.9 μg/mL (>90.0 μg/mL)[61]
Ulva lactuca LinnaeusEthanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay5.9 μg/mL (>90.0 μg/mL)[48]
Isopropyl alcohol- Chloroform/methanol extractL. donovaniAxenic amastigoteIn vitro: Resazurin assay12.0 μg/mL (>90.0 μg/mL)[61]
Table 2. Overview of the current status on natural products from marine macroalgae with antileishmanial potency.
Table 2. Overview of the current status on natural products from marine macroalgae with antileishmanial potency.
Chemical TypeChemical ClasseIsolated CompoundsSpecies of Marine Algae; PhylumLeishmania SpeciesParasites StageMethod of Activity EvaluationActivity (IC50 or % of Inhibition) Cytotoxicity (CC50)References
DiterpeneDolastane(4R,9S,14S)-4α-acetoxy-9β, 14α-dihydroxydolast-1(15), 7-dieneCanistrocarpus cervicornis (Kützing) De Paula and De Clerck; PhaeophyceaeL. amazonensisPromastigoteIn vitro: Microscopic counting2.0 μg/mL (186.0 μg/mL)[43]
L. amazonensisAxenic amastigoteIn vitro: Microscopic counting12.0 μg/mL (186.0 μg/mL)[43]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting4.0 μg/mL (186.0 μg/mL)[43]
Meroditerpenoid(3R)- and (3S)-tetraprenyltoluquinol (1a/1b) (3R)- and (3S)-tetraprenyltoluquinone (2a/2b)Cystoseira baccata (S.G. Gmelin) P.C.; PhaeophyceaeL. infantumPromastigoteIn vitro: MTT assay44.9 μM (126.6 μM)[44]
L. infantumIntracellular amastigoteIn vitro: MTT assay25.0 μM (126.6 μM)[44]
L. infantumPromastigoteIn vitro: MTT assay94.4 μM (84.5 μM)[44]
Atomaric acidStypopodium zonale (J.V. Lamouroux) Papenfuss; PhaeophyceaeL. amazonensisPromastigoteIn vitro: Microscopic counting86.0% inhibition at 50.0 μM (169.5 μM)[51]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting20.2 μM (169.5 μM)[51]
HydroazuleneMixture of isomers pachydictyol A/isopachydictyol ADictyota menstrualis (Hoyt) Schnetter, Hörning and Weber-Peuker; PhaeophyceaeL. amazonensisPromastigoteIn vitro: MTT assay23.5 μg/mL (30.0 μg/mL)[47]
DolabellaneDolabelladienetriolDictyota pfaffii Schnetter; PhaeophyceaeL. amazonensisPromastigoteIn vitro: Microscopic counting95.5% Inhibition at 100.0 μM (>100.0 μM)[14]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting43.9 μM (>100.0 μM)[14]
Leishmania/HIV-1 co-infectionIntracellular amastigote/HIV-1In vitro: Microscopic counting56.0% inhibition at 50.0 μM (>100.0 μM)[14]
Linear diterpeneBifurcatriolBifurcaria bifurcata R.Ross; PhaeophyceaeL. donovaniAxenic amastigoteIn vitro: Resazurin assay18.8 μg/mL (56.6 μg/mL)[74]
TriterpenePhytosterolFucosterolLessonia vadosa Searles; PhaeophyceaeL. infantumPromastigoteIn vitro: Resazurin assay45.0 μM (>100.0 μM)[75]
L. infantumIntracellular amastigoteIn vitro: Resazurin assay10.30 μΜ (>100.0 μM)[75]
L. amazonensisPromastigoteIn vitro: Resazurin assay55.0 μM (>100.0 μM)[75]
L. amazonensisIntracellular amastigoteIn vitro: Resazurin assay7.89 μM (>100.0 μM)[75]
SesquiterpeneChamigreneElatolLaurencia dendroidea (Hudson) J.V. Lamouroux; RhodophytaL. amazonensisPromastigoteIn vitro: Microscopic counting and Green fluorescent protein assay4.0 μM; 9.7 μg/mL (1.4 μM; 112.9–120.0 μg/mL)[26,60]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting0.45 μM; 4.5 μg/mL (1.4 μM; 112.9–120.0 μg/mL)[26,60]
ObtusolL. amazonensisPromastigoteIn vitro: Green fluorescent protein assay6.2 μg/mL (133.5–139.3 μg/mL)[60]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting3.9 μg/mL (133.5–139.3 μg/mL)[60]
TriquinaneSilphiperfol-5-en-3-olL. amazonensisPromastigoteIn vitro: Green fluorescent protein assay43.8 μg/mL (160.2–172.8 μg/mL)[60]
L. amazonensisIntracellular amastigoteIn vitro: Microscopic counting48.7 μg/mL (160.2–172.8 μg/mL)[60]
Sulfated polysaccharideFucanFucoidanFucus vesiculosus (Linnaeus); PhaeophyceaeL. donovaniIntracellular amastigoteIn vitro: Microscopic counting93.0% inhibition at 50.0 μg/mL[76]
L. donovani In vivo100.0% supression at 200.0 mg/kg/day (ND)[76]
NDSulfated polysaccharide (NI)Botryoclada occidentalis (Børgesen) Kylin; RhodophytaL. amazonensisPromastigoteIn vitro: MTT assay63.7 μg/mL (27.3 μg/mL)[77]
Caulerpa racemosa (Forsskål) J. Agardh; ChlorophytaL. amazonensisPromastigoteIn vitro: MTT assay34.5 μg/mL (49.3 μg/mL)[77]
IC50: Compound concentration that inhibited the proliferation of parasites by 50%; (CC50): Compound concentration that inhibited the proliferation of normal mammalian cells by 50%; ND: not determined; NI: Not identified.

Share and Cite

MDPI and ACS Style

Tchokouaha Yamthe, L.R.; Appiah-Opong, R.; Tsouh Fokou, P.V.; Tsabang, N.; Fekam Boyom, F.; Nyarko, A.K.; Wilson, M.D. Marine Algae as Source of Novel Antileishmanial Drugs: A Review. Mar. Drugs 2017, 15, 323. https://doi.org/10.3390/md15110323

AMA Style

Tchokouaha Yamthe LR, Appiah-Opong R, Tsouh Fokou PV, Tsabang N, Fekam Boyom F, Nyarko AK, Wilson MD. Marine Algae as Source of Novel Antileishmanial Drugs: A Review. Marine Drugs. 2017; 15(11):323. https://doi.org/10.3390/md15110323

Chicago/Turabian Style

Tchokouaha Yamthe, Lauve Rachel, Regina Appiah-Opong, Patrick Valere Tsouh Fokou, Nole Tsabang, Fabrice Fekam Boyom, Alexander Kwadwo Nyarko, and Michael David Wilson. 2017. "Marine Algae as Source of Novel Antileishmanial Drugs: A Review" Marine Drugs 15, no. 11: 323. https://doi.org/10.3390/md15110323

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