Open access peer-reviewed chapter

Biological and Medicinal Importance of Sponge

Written By

Musarat Amina and Nawal M. Al Musayeib

Submitted: 04 June 2017 Reviewed: 05 January 2018 Published: 25 April 2018

DOI: 10.5772/intechopen.73529

From the Edited Volume

Biological Resources of Water

Edited by Sajal Ray

Chapter metrics overview

3,462 Chapter Downloads

View Full Metrics

Abstract

Sponges are multicellular, heterotrophic parazoan organisms, characterized by the possession of unique feeding system among the animals. They are the most primitive types of animals in existence, featuring a cell-based organization where different cells have different tasks, but do not form tissues. Sponges (Porifera) are a predominantly marine phylum living from the intertidal to the abyssal (deepest ocean) zone. There are approximately 8500 described species of sponges worldwide with a prominent role in many reef coral communities. Several ecological studies reported have shown that secondary metabolites isolated from sponges often serve defensive purposes to protect them from threats such as predator attacks, biofouling, microbial infections, and overgrowth by other sessile organisms. In the recent years, interest in marine sponges has risen considerably due to presence of high number of interesting biologically active natural products. More than 5300 different natural products are known from sponges and their associated microorganisms, and every year hundreds of new substances are discovered. In addition to the unusual nucleosides, other classes of substances such as bioactive terpenes, sterols, fatty acids, alkaloids, cyclic peptides, peroxides, and amino acid derivatives (which are frequently halogenated) have been described from sponges or from their associated microorganisms. Many of these natural products from sponges have shown a wide range of pharmacological activities such as anticancer, antifungal, antiviral, anthelmintic, antiprotozoal, anti-inflammatory, immunosuppressive, neurosuppressive, and antifouling activities. This chapter covers extensive work published regarding new compounds isolated from marine sponges and biological activities associated with them.

Keywords

  • sponges
  • anticancer
  • antibacterial
  • chemical constituents

1. Introduction

Sponges are the ancient, efficient designed multicellular parazoan organisms and show relatively little differentiation and tissue coordination. A sponge is a sessile, sedentary, filter-feeding primitive aquatic invertebrate animal which attaches itself to solid surfaces from intertidal zone to depths of 29,000 ft (85000m) or more, where they can get sufficient food to grow [1, 2]. Sponges feed on microscopic organisms (protozoa, bacteria and other small organisms in water) and organic particles [3]. There are about 10,000 known species inhabit a wide variety of marine and fresh water habitats and are found throughout deep ocean depths to rock pools, warm tropical seas to frozen arctic seas, rivers and streams [3, 4]. They are very diverse and occur in various colors, sizes and shapes such as tubular (tube-like), globular (ball-shaped), caliculate (cup-shaped), arboresecent (plant-shaped), flabellate (fan-shaped) and amorphous (shapeless). The scientific term for sponges is Porifera meaning “pore-bearing” and has bodies full of pores and channels allowing water to circulate through them, consisting of jelly-like mesohyl sandwiched between two layers of cells [5]. The shapes of their bodies are adapted for maximal efficiency of water flow through the central cavity, where it deposits the nutrients, and leaves through a hole called the osculum. Several sponges have spicules of silicon dioxide or calcium carbonate and a mesh of proteins called spongin as an internal skeleton. One of the remarkable properties of sponges is their ability to suffer damage and regenerative capacity [6, 7, 8]. Marine sponges have attracted growing attention as a source of overwhelming structurally diverse secondary metabolites with potential biological activities and were placed at the top with respect to discovery of biologically active chemical constituents [9, 10]. Although thousands of chemical compounds have been reported in the literature from these sponges, only few of them are clinically described. Many studies revealed that sponge-derived metabolites are used directly in therapy or as a prototype of bioactive leads to develop more active and less toxic analogs [11, 12]. Sponges are most primitive type of aquatic animals in existence which are dominating many benthic habitats, featuring a cell-based organization where different cells conduct all forms of bodily function, but do not form tissues [13]. They consume food and excrete waste products within cells without a body cavity [14]. Several ecological studies reported that high quantity of bioactive constituents produced by sponges often serve defensive against environmental threats such as predation, microbial infection, competition for space or overgrowth by fouling organisms [15, 16]. For this reason marine sponges are the subject of attraction for chemists due to the sheer number of metabolites produced, the novelty of structure encountered, and the therapeutic potential of these compounds in the treatment of human diseases. Scientists working in the field of natural product chemistry and research suggest that these sponges have promising potential to provide future drugs which can serve various diseases. In this chapter, we describe main isolated chemical entities from sponges and their pharmacological application.

Advertisement

2. Anticancer agents

In the recent years, marine natural products bioprospecting has yielded a considerable number of drug candidates, most still being in preclinical or early clinical development, with only a limited number already in the market [17]. A typical example of marine anticancer drugs is eribulinmesylate, a derivative of halichondrin B isolated from the marine sponge. Halichondria okadai has achieved success in phase III clinical trials. Literature studies have shown sponge-derived discodermolides antitumor compounds can play remarkable role in future to treat cancer. Plethora of secondary metabolites is produced by marine sponges and their symbionts. The spongothymidine and spongouridine nucleosides were the first successful sponge-derived pharmaceutical drugs isolated from Tectitethya crypta [18]. Ara-C (cytarabineor1-beta-D-Arabinofuranosylcytosine) recently used for the cure of leukemia [19, 20] and its combination with Daunoribicin and other anticancer drugs, is screened in clinical trials for the treatment of acute myeloid neoplasms [21] During the last few years several marine derived natural compounds are in the pipeline for evaluation in Phase I–III clinical trials for various cancers treatment [22]. A review in 2003 listed the most important anticancer candidate from marine natural compounds undergoing preclinical and clinical (I, II, III) trials and following compounds were from sponge origin: Isohomohalichondrin B, Halichondrin B, Laulimalide/Fijianolide,5-methoxyamphimedine(alkaloid)Discodermolide, Hemiasterlins A and B, Fascaphysins (alkaloid), modified halichondrin B, KRN-70000, Alipkinidine (alkaloid), and Variolin (alkaloid) [23]. Moreover marine sponges are the important source for vital diverse bioactive constituents including alkaloids, terpenoids, sterols and macrolides. Renieramycins, members of tetrahydroiso-quinoline family were isolated from marine sponges from genus Reniera with promising anticancer potential. The preclinical results reported that Renieramycin M, a natural constituent from sponge induced lung cancer cells apoptosis through p53-dependent pathway and may inhibit progression and metastasis of lung cancer cells [24]. A novel polycyclic guanidine alkaloid monanchocidin isolated from Monanchora pulchra marine sponge reported to induce cell death in human cervical cancer (HeLa),human monocytic leukemia (THP-1)and mouse epidermal (JB6 Cl41) cells [25]. In the early 1987, as esquiterpene aminoquinone, Smenospongine extracted from Smenospongia sp. reported to induces cytotoxic, antiproliferetive, antiangiogenic, and antimicrobial activities [26]. Spongistatin a macrocyclic lactone polyether isolated from Spongia sp. marine sponge in 1993 was shown to inhibit microtubule assembly, mitosis, and the binding of tubulin to vinblastine thereby inducing cytotoxic cell death in numerous cancer cell lines [27, 28]. Recently a very important compound named lectin has been isolated from Cinachyrella apion marine sponge was evaluated for antiproliferative, hemolytic, and cytotoxic properties, besides the ability to induce cell death in tumor cells. Results showed that the lectin induces cell death by apoptosis activation by pro-apoptotic protein Bax, promoting permeabilization of mitochondrial membrane, S phase cell cycle arrest and acting as both dependent and/or independent of caspases pathway. These results indicate the potential of lectin for treating cancer [29]. Another marine sponge component, heteronemin a sesterterpene isolated from Hyrtios sp. has attracted the interest of researchers as an antimour agent especially for its pharmacological effects on chronic myelogenous leukemia cells. Results revealed that heteronemin affected the various cellular processes such as cell cycle, nitrogen-activated protein kinases pathways, apoptosis, and nuclear factor kappa B signaling cascade. Thus the compound has shown anti-inflammatory as well as anticancer agent [30]. A collaborative program between experimental therapeutics laboratory of Henry Ford Hospital in Detroit and University of California Santa Cruz initiated in 1990 focused on the development and discovery of anticancer drugs from sponge extracts. About 2036 extracts from 683 individual sponges were examined by using novel in vitro assay led to the identification pure bioactive compounds from many sponges for treating solid tumors. The collaborative efforts and analogs led to the isolation of number of constituents with of anticancer potential [31].

Thus the possibility of development of new anticancer drugs for curing or reducing cancer is promising. Until now, in vitro antitumor activity studies of sponge-derived compounds were tested. Thus, the detailed pharmaceutical studies to investigate the mechanism of action and clinical trials are needed. Moreover, the extensive ongoing research on sponges and development of new advanced techniques have made it possible to access deep sea, new anticancer marine isolates with unprecedented carbon skeleton and inhibitory activities of human cancer cell continued to be discovered and developed, which will offer in future the new candidate for cancer therapy. The chemical constituents so far reported for anticancer activity include (Table 1).

CategoriesSpeciesActive agentsAntitumor testedReferences
AlkaloidsPapuaHyrtiocarbolineH522-T1, MDA-MB- 435, U937 tumor cell lines[31]
Penares sp.HL-60, HeLa[32]
Aaptos suberitoidesAaptamineL5178Y[33]
Monanchora arbusculaNorbatzelladine
DinorbatzelladineMDA-MB-231 breast cancer[34]
Dinordehy-drobatzelladine
Dinorbatzelladine
Dihomodehy-drobatzelladineMDA-MB-231 breast cancer[34]
Clathria callaNorbatzelladine
Clathriadic acid
Xestospongia sp.Renieramycin THCT116, QC56, AsPC1[35]
T47D tumor cell lines
Smenospongia sp.6′-IodoaureolMOLT-3, HepG2 cells[36]
Hyrtios sp.Hyrtimomine AHuman epidermoid carcinoma KB, murine leukemia L1210[37]
Pseudoceratina verrucosaAplysamineHeLa, NFF cells[38]
Amphimedon sp.Pyrinodemin GP388 murine leukemia cells
Pyrinodemin H[39]
Oceanapia sp.Sagitol CPC12, L5178Y, HeLa cells[40]
Monanchora pulchraMonanchocidins B
Monanchocidins CHL-60 human leukemia cells[41]
Monanchocidins D
Monanchocidins E
Agelas sp.Hexazosceptrin
Agelestes A-BU937, PC9 human
(9S, 10R, 90S, 100R)-nakamuric acidCancer cell lines[42]
SterolsIanthella sp.Petrosterol-3,6-dioneA549 (lung), HT-29 (colon),
5α,6α-epoxy-petrosterolSK-OV-3 (ovary), MCF-7 (breast) HL-60 and U937[43]
Lissodendryx fibrosaManadosterol A-BUbc13–Uev1A complex[44]
TerpenoidsCarteriospongia sp.Homoscalarane sesterterpenesA2780, H522-T1, A2058[45]
Monanchora sp.9SesterterpenoidsA498, ACHN (renal cancer)[46]
MIA-paca, and PANC-1 (pancreatic cancer)[47]
Psammocinia sp.Scalarane sesterterpenesA498, ACHN MIA-paca,PANC-1[48]
Pseudoaxinella flavaDiterpene isonitrilePC3(prostate cancer cell line)[49]
Agelas axiferaThree axistatins (pyrimidine diterpenes)P338, BXPC-3 MCF-7, SF-268 NCI-H460, KML20L2, and DU-145 cell lines growth[50]
Thorectare ticulateMetachromins U
Metachromins V
SF-268, H460,MCF-7, HT-29, and CHOK1 (mammalian cell line)[51]
Dactylospongia elegansNakijinol B and CHO-K1SF-268, H460, MCF-7, HT-29[51]
Coscinoderma sp.Sesterterpenes coscinolactams CK562 and A549 (human cancer cells)[52]
Coscinolactams D,
Coscinolactams E
Coscinolactams F
Coscinolactams G[53]
MacrolideCinachyrella enigmaticaEnigmazole ANCI 60 human tumor cells[54]
Jaspis splendansJaspamide MMCF-7and HT-29[55]
Jaspamide N(antimicrofilament)
Jaspamide O
Jaspamide P
Mycale hentscheliPeloruside AP388 HL-60 cells[56]
Peloruside B
Pipestela candelabraPipestelide AKB cell lines[57]
Pipestelide B
PolyketonePlakortis simplexSimplextone CHeLa, K562, A-549 cell lines
Plakortoxide A[58]
Plakortis halichondrioidesEpiplakinidioic acidDU-145, A2058[59]
Plakortoxide Atumor cell lines
LithoplocamialithistoidesPolyketides PM050489HT-29, A549, MDA-MB-231
Polyketides PM060184Human tumor cell lines[60]
PeptidesHomophymia sp.Homophymines BKB, MCF7, MCF7R, HCT116
Homophymines EHCT15, HT29, OVCAR 8, OV3,
Homophymines A1-E1PC3, Vero, MRC5, HL60, HL60R, K562, PaCa, SF268, A549, MDA231, MDA435, HepG2, and EPC human tumor cells[61]
Neamphius huxleyiNeamphamide BA549,HeLa, LNCaP,
Neamphamide CPC3, NFF human tumor
Neamphamide Dcell lines[62]
Eurypon laughliniRolloamide ALNCap, PC3MM2, PC3, DU145 (Prostrate), MDA361, MCF7, MDA231 (breast), OVCAR3, SKOV3, U87MG (Glioma), (ovarian), A498 (renal)[63]
Stylissa caribicaStylissamide HHCT-116.[64]
Homophymia lamellosePipecolidepsin AA549, HT-29 MDA-MB-231
Pipecolidepsin BHuman tumor cells[65]
GlycosidesPandaros acanthifoliumAcanthifoliosides A–EL6 cell lines[66]
Rhabdastrella globostellataRhabdastin E-GHL-60[67]
QuinonesDysidea avaraDysidavarone AHeLa, A549, MDA231, QGY7703
Dysidavarone DHeLa tumor cells[68]
Dactylospongia metachromia5 Sesquiterpene aminoquinonesL5178Y mouse cancer cell lines[69]
Dactylospongia avara3 Dysideanones A–CHeLa HepG2 cancer cell lines[70]
MiscellaneousPetrosia sp.3(−) Petrosynoic acids A–DA2058, H522-T1,
H460 human tumor cell line
IMR-90 human fibroblast cells[71]
SubereamollisSubereaphenol DHeLa cell lines[72]
Mixture of Smenospongia aurea(E)-10-benzyl-5,7-dimethylun-1 deca,5,10-trien-4-olHL-60 human leukemia
Smenospongia cerebriformis
Verongula rigida[73]
Myrmekioderma dendyiMyrmekioside E-2NSCLC-N6 and A549 tumor cell lines[74]
Genus Suberea.Four novel Psammaplysin analogsCytotoxicity[75]

Table 1.

Marine sponge-derived anticancer compounds and their effects.

Advertisement

3. Antibacterial active agents

Marine sponges are among the richest sources of interesting chemicals produced by marine organisms. Exploitation of bioactive metabolites by natural product chemist from marine sources by using antimicrobial or cytotoxic assays started back in 1970s. Later, various reputed pharmaceutical companies joined hands for this effort using more advance assay systems, including enzyme inhibition assays. As a result several new promising bioactive candidates have been discovered from marine sponges [76]. Bioactive constituents are claimed for potent in vivo or in vitro activity against infectious and parasitic diseases, such as bacterial, fungal, viral and protozoan infections. Studies revealed that the crude extracts of marine sponge have shown high incidences of antibacterial activity against terrestrial pathogenic bacteria, but very low incidences of antibacterial activity against marine bacteria [77, 78]. Very few cases of sponge infection by exogenous microorganisms are known, presumably due to the accumulation/or product by the marine sponges of substances which have antimicrobial activity [1]. A number of new metabolites with antibiotic applications are discovered every year, but in marine sponges their ubiquity is remarkable. Antibacterial screening of marine sponges led to identification and characterization of wide range of active chemical constituents, including some with promising therapeutic leads [79, 80]. Around 850 antibiotic constituents are reported from marine sponges [81]. Various antibacterial substances were identified from marine sponges by continuous efforts of marine natural product community. Despite of discovery of huge number of natural product from marine sponges, none of them has yet led to antibacterial product, but currently several are under investigation. Examples of some isolated substances from marine sponges with antibacterial activity are shown in Table 2. The first discovered antibiotic from a marine sponge was manoalide, a seterterpenoid isolated from Luffariella variabilis [82]. The most promising constituents with antibacterial properties reported from marine sponges include: agelasine D, cribrostatin 3 and 6, petrosamine B, psammaplin A and alkylpyridines (haliclonacyclamine E, arenosclerins) and among these constituents, manzamine A and psammaplin A are in preclinical trials. Many of these have excellent potential for drug development, but no commercial medication has been originated from them so far.

CategoriesSpeciesActive agentsAntibacterial testedReferences
AlkaloidsAxinella sp.Axinellamines B-DH. pylori Gram-(-ve)[83]
Acanthostrongylophora sp.12,34-Oxamanzamine E,M. tuberculosis[84]
8-Hydroxymanzamine J
6-Hydroxymanzamine E
Arenosclera brasiliensisHaliclonacyclamine E,S. aureus, P. aeruginosa
Arenosclerins A-C[85]
Spongosorites sp.Deoxytopsentin, bromotopsentinS. aureus (MRSA strain)
4,5-Dihydro-6”-deoxybromotopsentin, bis(indole)[86]
Cribrochalina sp.Cribrostatin 3N. gonorrheae[87]
Cribrochalina sp.Cribrostatin 6S. pneumonia[88]
Spongosorites sp.Hamacanthin AS. aureus (MRSA strain)[86]
Oceanapia sp.Petrosamine BH. pylori[89]
Latrunculia sp.Discorhabdin RS. aureus, M. luteus
S. marcescens, E. coli
[90]
Hamacantha sp.Hamacanthin A 1C. albicans
Hamacanthin B 2C. neoformans[91]
NitrogenousPachychalina sp.Cyclostellettamines A-I,S. aureus (MRSA strain),[92]
Cyclostel K-LP. aeruginosa (antibiotic-resistant strain), M. tuberculosis[93]
Pachychalina sp.Ingenamine GS. aureus (MRSA strain)
E. coli, M. tuberculosis[92]
M. sarassinorumMelophlin CB. subtilis, S. aureus[47]
Agelas sp.Agelasine DM. tuberculosis Gram (+ve, -ve)[94]
TerpenoidsCacospongia sp.Isojaspic acid, cacospongin D, jaspaquinolS. epidermidis[95]
Myrmekiodermastyx(S)-(+)-curcuphenolM. tuberculosis[96]
MiscellaneousOceanapia sp.C14 acetylenic acidE. coli, P. aeruginosa, B. subtilis, S. aureus[97]
C. sphaeroconiaCaminosides A-DE. coli[98]
A. coralliphagaCorallidictyals A-DS. aureus[99]
C. variansCvLB. subtilis, S. aureus[100]
N. magnificaLatrunculinsS. aureus and B. cereus[101]
Discodermia sp.Polydiscamide AB. subtilis[93]
PsammaplysillaPsammaplin AS. aureus (MRSA strain)[102]

Table 2.

Marine sponge-derived antibacterial compounds and their effects.

Advertisement

4. Antiviral compounds and their efficacy

The search for new antiviral substances from marine sources led to the isolation of several promising therapeutic leads which are presented in Table 3. The literature presents a good number of reports about different biological activities of marine sponges. Several papers reports the screening results of marine organisms for antiviral activity, and a diverse range of active constituents have been isolated and characterized from them [80, 103, 104]. For some of these isolated substances important antiviral activities were reported. Perhaps the most important antiviral lead of marine origin reported thus far is the nucleoside ara-A (vidarabine) isolated from the sponge Cryptotethya crypta. Ara-A is a semisynthetic compound, based on the arabinosyl nucleosides, that inhibits viral DNA synthesis [105]. Once it was realized that biological systems would recognize the nucleoside base after modifications of the sugar moiety, chemists began to substitute the typical pentoses with acyclic entities or with substituted sugars, leading to the drug azidothymidine (zidovudine). Ara-A, ara-C (1-β-Darabinosyl cytosine, cytarabine), acyclovir, and azidothymidine are in clinical use and are all examples of products of semisynthetic modifications of the arabinosyl nucleosides [106]. Several of these substances have a great potential for drug development. Ara-A has been used for the treatment of herpes virus infections, but it is less efficient and more toxic than acyclovir [107, 108]. However, ara-A is capable of inhibiting a cyclovir-resistant HSV and VZV (varicella-zoster virus) [109]. The most promising antiviral substances from sponges appear to be 4-methylaaptamine, avarol, manzamines, mycalamide A and B. Among these substances, preclinical assessments were started for avarol and manzamine A. In general, antiviral molecules from sponges do not give protection against viruses, but they may result in drugs to treat already infected individuals. In addition, broad-based antiviral agents such as 2-5A and α-glucosidase inhibitors may be useful in cases of sudden outbreaks of (less familiar) viruses such as SARS and Ebola [80].

CategoriesSpeciesActive agentsAntiviral testsReferences
AlkaloidAaptosa aptos4-MethylaaptamineHSV-1[110]
Halicortex sp.Dragmacidin FHSV-1[111]
Indo-PacificManzamine A, 8-hydroxymanzamine A, 6-deoxymanzamine X neokauluamineHIV-1[112]
NucleosidesMycale sp.Mycalamide A-BA59 coronavirus, HSV-1[113]
Hamacantha sp.Coscinamides 60-62,
Chondriamides 63-65Anti-HIV[91]
Cyclic depsipeptidesTheonella sp.Papuamides A-DHIV-1[114]
S. microspinosaMicrospinosamideHIV-1[115]
SterolsHaplosclerid spongesHaplosamates AHV-1
Haplosamates B[116]
TerpenoidsD. avaraAvarol 6′-hydroxy avarol, 3′-hydroxy avaroneHV-1[117]
NucleosideCryptotethya cryptaAra-AHSV-1, HSV-2, VZV[105]
Mycale sp.Mycalamide A-BA59 coronavirus, HSV-1[118]
MiscellaneousDysidea avaraCallyspongymic acidHIV, hepatitis B virus[119]
2′-5′ OligoadenylatesViral replication[120]
H. tarangaensisHamigeran BHerpes, polio viruses[121]
Petrosia weinbergiWeinbersterols A-BLeukemia virus, mouse influenza virus, mouse corona virus[122]

Table 3.

Antiviral compounds from marine sponges and their effects.

Advertisement

5. Antifungal compounds

Marine sponges have been considered a gold mine for the discovery of marine natural products during the past 50 years. The need of new antifungals in clinical medicine due to various kinds of mycoses, in particular invasive mycoses have become serious health problems as their incidences has increased dramatically during last few years in relation to AIDS, transplant recipients, hematological malignancies, transplant recipients and other immunosuppressed individuals. One of the major causes of death in patients suffering from malignant disease is fungal infections and emerging resistance is also an important problem. Immunocompromised patients are mainly infected by Aspergillus, Cryptococcus, Candida, and other opportunistic fungi. Candida albicans is most often associated with serious invasive fungal infections, but other Candida species and yeast-like organisms (Blastoschizomyces, Trichosporon and Malassezia) have emerged as etiological agents of severe mycoses problem [123, 124, 125, 126]. Fungicides which are presently being used are less diverse than antimicrobials, and the usage of many of them is restricted because of their toxic effects to animals, plants and humans. Moreover the progress in this area is slow as comparison to antibacterial agents [126]. Antifungal compounds isolated from marine sponges are listed in Table 4.

CategoriesspeciesActive agentsAntifungal testsReferences
AlkaloidsA. brasiliensisArenosclerins A-CC. albicans
Haliclonacyclamine E[127]
Acanthostrongylophora sp.Manzamine AC. neoformans[112]
Leucetta cf.Naamine DChagosensis C. neoformans[128]
Pseudoceratina sp.Ceratinadins A-CC. albicans[129]
A. citrina(−)-Agelasidine F,C. albicans
(−)-Agelasidine C[130]
M. arbuscularBatzelladine LA. flavus[131]
TerpenoidsL. variabilisSecomanoalideC. glabrata, C. krusei
C. albicans[132]
M. herdmaniMicrosclerodermins A-BA. fumigatus[133]
Hyrtios sp.PuupehenonolC. neoformans, C. krusei[134]
SterolsEuryspongia sp.Eurysterols A-BC. albicans[135]
Topsentia spGeodisterol-3-O-sulfite, 29-demethylgeodisterol-3-OCl-sulfiteS. cerevisiae, C. albicans
C. albicans[136]
PeptidesDiscodermia sp.Discobahamin A-BC. albicans[137]
Jaspis sp.Jasplakinolide or jaspamideC. albicans[138]
Latrunculia sp.Callipeltins F-IC. albicans[139]
Latrunculia sp.Callipeltin J-KC. albicans[42]
T. swinhoeiTheonellamide GC. albicans[140]
Theonella sp.Theonellamide TNM-FCandida spp, Trichophyton spp, Aspergillus sp.[141]
Purine derivativesAgelas sp.Agelasines, agelasiminesC. krusei[142]
MiscellaneousP. reticulateCrambescin A2 392C. albicans
Crambescin A2 406C. neoformans var. gattii,
Crambescin A2 420C. glabrata, C. krusei
Sch 575948[143]
SpongeTheonellamidesAntifungal[144]
Melophlus sp.Aurantoside KC. albicans (wild-type)[145]
P. halichondrioidesPlakortide FC. albicans, C. neoformans, A. fumigatus[146]
H. viscosaHaliscosamineC. neoformans, C. albicans[147]
D. herbacea3,5-Dibromo-2-(3,5-dibromo-2-methoxyphenoxy) phenolAspergillus[148]
P. onkodesTwo α and β1,2-dioxolane peroxide acidsC. albicans[149]
T. laevispiruliferNematocide, onnamide FS. cerevisiae[150]
T. swinhoeiSwinhoeiamide AC. albicans, A. fumigates[151]
Family NeopeltidaeNeopeltolideC. albicans[152]
PlakinastrellaEpiplakinic acid FC. albicans[153]
H. communis(−)-Untenospongin BC. albicans, C. tropicalis, F. oxysporum[154]
H. lachneHippolachnin AC. neoformans, T. rubrum, M. gypseum[155]

Table 4.

Antifungal compounds from marine sponges and their effects.

Advertisement

6. Anti-inflammatory compounds

Marine organisms and microorganisms have provided a large proportion of the anti-inflammatory and natural antioxidants products over the last years. Reports suggest that marine invertebrates represent new marine resources for the isolation of novel agents which are active on inflammatory conditions have also been found in the literature. Herencia and coworkers [156] studied the effects of dichloromethane and methanol extracts from some Mediterranean marine invertebrates on carrageenan-induced paw edema in mice. Extracts partially decreased elastase activity and PGE2 levels measured in homogenates from inflamed paws, without affecting the levels of this prostanoid present in stomach homogenates. Within the framework of the European MAST III Project, extracts of different polarity from sponges, ascidians and cnidarians have been screened for immunomodulating activities [157]. It was demonstrated that endotoxin-free samples of marine origin possess effects on certain components of the immune system. As a result of all these investigations, bioassay-directed separation of active extracts identified many structurally diverse compounds as future leads. Anti-inflammatory compounds found in the marine environment include terpenes and steroids, alkaloids, peptides and proteins, polysaccharides and others. Examples of anti-inflammatory compounds marine sponge origin are presented in Table 5. Also includes diterpenes of (8E, 13Z, 20Z)-strobilinin and (7E, 13Z, 20Z)-felixinin from a marine sponge Psammocinia sp. [158], and novel anti-inflammatory spongian diterpenes from the New Zealand marine sponge Chelonaplysill aviolacea [159].

CategoriesspeciesActive agentsAnti-inflammatory testsReferences
TerpenoidsF. cavernosaCavernolideTNF-α, NO and PGE2 production[160]
Axinella spp.6-CycloamphilectenesNO, PGE2 and TNF-α production[161]
2-CycloamphilectenesInhibit NF-КB pathway[161]
Psammocinia spp.Chromarols A-EInhibition of 15-LOX[162]
Psammocinia spp.(8E, 13Z, 20Z)-strobilininAnti-inflammatory
(7E, 13Z, 20Z)-felixininAnti-inflammatory[158]
C. violaceaSpongianAnti-inflammatory[163]
D. avaraAvarol, avarone,Inhibition of eicosanoid release[164]
Spongiaquinone, ilimaquinoneand depression of superoxide generation[165]
Dysidea spp.Dysidotronic acidInhibited production of TNF-α, IL-1 PGE2, and LTB4[166]
Plakortis spp.Plakolide AInhibit iNOS[167]
D. elegansCymopolDNA binding of NF-КB[168]
L. variabilisManoalide, scalaradialInhibited IL-1 and TNF-α[169]
F. cavernosaCacospongiolide BInhibited PLA2[170]
Dysidea sppDysidenones A-BInhibited human synovial PLA2[171]
L. variabilisCladocorans A-BInhibition of secretory PLA2[172]
P. nigraPetrosa spongiolidesInhibitor of PLA2[173]
P. nigraPetrosa spongiolide MInhibited LTB4 levels[174]
Cacospongia spp.ScalaradialInactivate the enzyme PLA2[175]
G. sednaHomoscalaraneModerate activity to inhibit mammalian PLA2[176]
Hyrtios sp.Puupehenone, hyrtenoneA high potency against 12-human, 15-human and 15-soybean LOX[177]
C. linteiformisCyclolinteinoneiNOS and COX-2 protein expression in LPS-stimulated J774 macrophages[178]
Callyspongia spp.AkaterpinInhibitor of phosphatidylinositol-specific Phospholipase C[179]
SteroidsC. lissosderaClathriolIn vitro anti-inflammatory activity against human neutrophil and rat mast cells[180]
Euryspongia spp.Petrosterol, 3β-hydroxy-26-nor-campest-5-en-25 oic acidAgainst 6-keto-PGF1α release in a human keratinocyte cell line HaCaT[181]
AlkaloidsX. testudinariaHymenialdisineInhibitor of NF-КB and ILs production[182]
Agelas spp.Nagelamides A-HNF-КB in inflammatory diseases[183]
S. flabellateStylissadines A-BAntiinflammatory activity[184]

Table 5.

Anti-inflammatory compounds from marine sponges and their effects.

Advertisement

7. Marine sponge-derived compounds with enzyme inhibitory activity

Derivatives of halenaquinone and xestoquinone showed various enzyme inhibitory activities besides the phosphatidylinositol 3-kinase and topoisomerase I and II inhibitory activities mentioned above. Compound xestoquinone inhibited both Ca2+ and K+-ATPase of skeletal muscle myosin [185]. SAR Investigations showed that halenaquinone and three synthetic analogs with a quinone structure significantly inhibited Ca2+ ATPase activity. In contrast, four xestoquinone analogs in which the quinine structure was converted to quinol dimethyl ether did not inhibit the Ca2+ ATPase activity [186]. The protein tyrosine kinase (PTK) inhibitory activities of halenaquinone, halenaquinol, and 14-methoxyhalenaquinone were the most remarkable with IC50 values <10 mm. The other analogs was either less potent or inactive, and a rationalization for this SAR pattern was also reported [187]. Xestoquinone also showed significant protein kinase inhibitory activity toward Pfnek-1, a serine/threonine malarial kinase, with an IC50 value of ca. 1 mm, and moderate activity toward PfPK5, a member of the cyclin-dependent kinase (CDK) family [188]. Adociaquinone B and 3-ketoadociaquinone B were the most potent inhibitors of the Cdc25 B phosphatase inhibitory activities, and the dihydro-benzothiazine dioxide in compounds Adociaquinone A, Adociaquinone B, 3-Ketoadociaquinone A, and 3-Ketoadociaquinone B appeared to be an important structural feature for this enhanced activity. Four cyclostellettamines, cyclostellettamine A, cyclostellettamine G, dehydrocyclostellettamine D and dehydrocyclostellettamine E inhibited histone deacetylase derived from K562 human leukemia cells with IC50 values ranging from 17 to 80 mm [189]. Xestospongic acid ethyl ester (207) was found to inhibit the Na+/K+ ATPase [190]. Compounds are listed in Table 6.

CategoriesSpeciesActive agentsEnzyme-inhibitoryReferences
QuinonesX. exiguaHalenaquinoneCa2+ ATPase activity[191]
X. exiguaXestoquinoneCa2+ and K+-ATPase activity[192]
X. sapraHalenaquinolProtein tyrosine kinase activity[193]
X. cf. carbonaria14-MethoxyhalenaquinoneProtein tyrosine kinase activity[187]
Xestospongia sp.Adociaquinone BProtein tyrosine kinase activity[194]
Xestospongia sp.3-Ketoadociaquinone BCdc25B phosphatase activity[195]
Xestospongia sp.Adociaquinone ACdc25B phosphatase[194]
Xestospongia sp.3-KetoadociaquinoneCdc25B phosphatase[195]
CyclostellettaminesXestospongia sp.CyclostellettamineA histone deacetylase derived inhibition
Cyclostellettamine G
Dehydrocyclostellettamine D
Dehydrocyclostellettamine E[189]
Fatty acidsX. testudinariaXestospongic acid ethyl esterinhibit the Na+/K+ ATPase[190]

Table 6.

Marine sponge-derived compounds showing enzyme-inhibitory activities.

Advertisement

8. Sponge-derived immunosuppressive compounds and their efficacy

Recently natural constituents isolated from marine sponges were tested for immunosuppressive activities and in the end of 1980s, deep water marine sponges resulted in isolation of pure compounds with immunosuppressive properties. Two important compounds: 4a-merhyl-5a-cholest-8-en-3~-ol and 4,5-dibromo-2-pyrrolic acid discovered by American scientist from deep water sponge Agelasfla bellrform is showed significant immunosuppressive activity. Both compounds were found significantly active in suppression of the response of murine splenocytes in the two-way mixed lymphocyte reaction (MLR) with little to no demonstrable cytotoxicity at low doses ([196]. Constituents isolated from the Aurora globostellata marine sponge showed immunomodulatory potential. The immunomodulatory potential was evaluated by oral administration of ethyl acetate extract of marine sponge (200 mg/kg) to Wistar rats and the results obtained showed that extracts exhibited immunosuppressant activity and can further be studied [197]. A recent investigation on an Indian marine sponge aimed to isolate and characterize bacteria with immunomodulatory and antimicrobial activity. Callyspongia difusa (Gulf of Mannar province) a marine sponge resulted in isolation of 10 marine bacterial strains which exhibited remarkable antagonistic activity against clinical bacterial pathogens. These findings suggested that the sponge associated bacterial strain Virgibacillus sp. can contribute the search for novel antibiotics to overcome infections and also for the production of potential immunomodulators [109].

Advertisement

9. Hypocholesterolemic compounds

In the last decade studies reported that marine sponges could have been a source of hypocholesterolemic compounds. For example, lysophosphatidylcholines and lyso-PAF analogs derived from Spirastrella abata are reported as successful inhibitors of cholesterol biosynthesis in vitro study [198, 199]. Zhao et al. [200] extracted novel lysophosphatidylcholines from marine sponges with hypocholesterolemic properties and thereby aroused an interest of compounds from marine sponge due to short lifespan of conventional lysophosphatidylcholines in vivo.

Advertisement

10. Sponge–derived antibiotics

Also, over the years marine sponges are considered as a rich source of natural products and metabolites for antibiotics possessing strong inhibitory against bacteria, fungi and microbes. Several studies revealed that many natural bioactive components isolated from various marine sponges can be useful for the production of new antibiotics and antimicrobial drugs. In the recent years many scientific studies provided evidences for marine sponge metabolites with efficient antibiotic, antibacterials and antimicrobial properties. Purpuroines A-J, halogenated alkaloids isolated from Lotrochota purpurea marine sponge showed promising inhibitory activities against bacteria and fungi related diseases [201]. Haliclona sp. sponge from Korea resulted in isolation of novel cyclic bis-1,3-dialkylpyridiniums and cyclostellettamines, which showed moderate cytotoxic and antibacterial activities against A549 cell-line and Gram-positive strains, respectively [202]. A number of new alkaloids were isolated from the marine sponge Agelas mauritiana: (+)-2-oxo-agela-sidine C, (−)-8′-oxo-agelasine D,4-bromo-N-(butoxymethyl)-1H-pyrrole-2-carboxamide, ageloxime B, and (−)-ageloxime D and some of these isolated components exhibited antifungal activity against Cryptococcus neoformans, antileishmanial activity in vitro and antibacterial activity against S. aureus and methicillin-resistant S. aureus in vitro [203]. Extracts prepared from the sponge’s species Petromica citrina, Haliclona sp. and Cinachyrella sp. exhibited antibacterial activity against 61% of the coagulase-negative staphylococci (CNS) strains, including strains resistant to conventional antibiotics. P. citrina extracts showed the largest spectrum of inhibitory activity. This current study according scientist shows potential of marine sponges to become new sources of antibiotics and disinfectants for the control of CNS involved in bovine mastitis in future [204]. Isolation of isonitriles ditepene from Cymbastela hooperi, tropical marine sponge and the axisonitrile-3 sesquiterpene isolated Acanthella kletra, from the tropical marine sponge were tested for series of bioassays antibacterial, antiphotosynthetic, antifouling, antialgal, antifouling, antialgal, antiphotosynthetic, antifungal, and antitubercular. The results showed majority of the tested compounds were active against at least two of the applied test systems [152]. Recently, sponge-derived actinomycetes and sediments isolated from marine sponge were tested for bioactive constituents with antifungal and antimicrobial activity. Out of 15 prepared active extract nine were found active against Enterococcus fascism (vancomycin-resistant) and Candida albicans multidrug-resistant [132], including strains resistant to conventional antibiotics. Thus the bacterial actinomycetes from marine sponges and other marine organisms have been proved prolific producers of pharmacologically active compounds. Literature studies revealed that 70% of naturally derived antibiotics which are currently in clinical use have been derived from actinomycetes. In the recent study, Streptomyces sp. strains from Mediterranean sponges and secondary metabolite namely, cyclic depsipeptide valinomycin, indolocarbazole alkaloid staurosporine and butenolide, were screened for anti-infective activities. All the isolated compounds along with Streptomyces sp. exhibited antiparasitic activities. Researchers also claim the anti-infective potential of marine actinomycetes is very promising.

11. Marine sponges-derived antifouling and antibiofilm compounds

Bacterial biofilms are surface-attached microorganism’s communities that are protected by an extracellular matrix of biomolecules. Continuous use of chemical antifoulants resulted in increased tributyltin concentration and created extensive pollution problems in marine organisms. Natural antifouling molecules from marine have been recently reviewed and researches hope that will provide more specific and less toxic antifouling activity in future. Antifouling compounds derived from sponges were found to be very effective, environmentally friendly biocides and less toxic [205]. In the last few years several studies were directed to find the most promising alternative technologies to antifouling in marine organisms, especially from sponges. In a recent study structurally different compounds containing 3-alkylpyridine moiety were evaluated for antifouling potential. The compounds, namely haminols, saraine and 3-alkylpyridinium salts extracted from Reniera sarai, Haliclona sp. and the mollusk Haminoea fusar is obtained by synthesis, showed very good antifouling potential larvae of the barnacle Amphibalanus amphitrite. Bromopyrrole or diterpene alkaloids derivatives isolated from Agelas linnaei and Ageles nakamurai Indonesian marine sponges exhibited cytotoxic activity. Moreover, agelasine derivatives inhibited settling of larvae of Balanus improvisus in an antifouling bioassay as well as the growth of planktonic forms of biofilm forming bacteria S. epidermidis [206].

12. Conclusion

Marine invertebrates (Porifera, Cnidaria, Mollusca, Arthropoda, Echinodermata, etc.) are considered as one of the major groups of biological organisms which gave huge number of natural products and secondary metabolites with interesting pharmacological properties and led in the formation of novel drugs. Among marine invertebrates, marine sponges (phylum: Porifera) is the most dominant responsible group for discovering significant number of natural components, which has been used as template to develop therapeutic drugs. These natural products possesses vast range of therapeutic application, including antimicrobial, antihypertensive, antioxidant, anticancer, anticoagulant, anti-inflammatory, immune modulator, and wound healing and other medicinal effects. Therefore, marine sponges are considered a rich source of chemical diversity and health benefits for developing drug candidates, nutritional supplements, cosmetics, and molecular probes that can be supported to increase the healthy life span of humans. In this chapter we included the most important and biologically active marine sponge-derived compounds and presented selected studies of most important bioactive and promising natural products and secondary metabolites from marine sponges.

Conflict of interest

The authors declare that they have no conflict of interest.

References

  1. 1. Hentschel U, Schmid M, Wagner M, Fieseler L, Gernert C, Hacker J. Isolation and phylogenetic analysis of bacteria with antimicrobial activities from the Mediterranean sponges Aplysina aerophoba and Aplysina cavernicola. FEMS Microbiology Ecology. 2001;35:305-312
  2. 2. Radjasa OK, Sabdono A, Junaidi, Zocchi E. Richness of secondary metabolite-producing marine bacteria associated with sponge Haliclona sp. International Journal of Pharmaceutics. 2007;3:275-279
  3. 3. Thomas TR, Kavlekar DP, LokaBharathi PA. Marine drugs from sponge-microbe association—A review. Marine Drugs. 2010;8:1417-1468
  4. 4. Perdicaris S, Vlachogianni T, Valavanidis A. Bioactive natural substances from marine sponges: New developments and prospects for future pharmaceuticals. Natural Products Chemistry and Research. 2013;1:1-8
  5. 5. Mehbub MF, Lei J, Franco C, Zhang W. Marine sponge derived natural products between 2001 and 2010: Trends and opportunities for discovery of bioactive. Marine Drugs. 2014;12:4539-4577
  6. 6. Taylor MW, Radax R, Steger D, Wagner M. Sponge-associated microorganisms: Evolution, ecology, and biotechnological potential. Microbiology and Molecular Biology Reviews. 2007;71:295-347
  7. 7. Stowe SD, Richards JJ, Tucker AT, Thompson R, Melander C, et al. Anti-biofilm compounds derived from marine sponges. Marine Drugs. 2011;9:2010-2035
  8. 8. Thompson JE, Walker RP, Faulkner DJ. Exudation of biologically-active metabolites in the sponge Aplysina fistularis. I. Biological evidence. Marine Biology. 1985;88:11-21
  9. 9. Becker S, Terlau H. Toxins from cone snails: Properties, applications and biotechnological production. Applied Microbiology and Biotechnology. 2008;79:1-9
  10. 10. Fenical W, Jensen P, Palladino M, Lam K, Lloyd G, Potts B. Discovery and development of the anticancer agent salinosporamide A (NPI-0052). Bioorganic & Medicinal Chemistry. 2009;17:2175-2180
  11. 11. Gerwick WH, Moore BS. Lessons from the past and charting the future of marine natural products drug discovery and chemical biology. Chemistry & Biology. 2012;19:85-98
  12. 12. Huyck TK, Gradishar W, Manuguid F, Kirkpatrick P. Fresh from pipeline: Eribulin mesylate. Nature Reviews. Drug Discovery. 2011;10:173-174
  13. 13. Johns WE, Schott F. Meandering and transport variations of the Florida current. Journal of Physical Oceanography. 1987;17:1128-1147
  14. 14. Hooper JN, editor. Sponguide. Guide to Sponge Collection and Identification. Queensland, Australia: Queensland Museum; 2000
  15. 15. Paul VJ, Puglisi MP. Chemical mediation of interactions among marine organisms. Natural Product Reports. 2004;21:189-209
  16. 16. Paul VJ, Puglisi MP, Ritson-Williams R. Marine chemical ecology. Natural Product Reports. 2006;23:153-180
  17. 17. Beedessee G, Ramanjooloo A, Aubert G, Eloy L, et al. Cytotoxic activities of hexane, ethyl acetate and butanol extracts of marine sponges from Mauritian Waters on human cancer cell lines. Environmental Toxicology and Pharmacology. 2012;34:397-408
  18. 18. Bergmann W, Feeney RJ. The isolation of a new thymine pentoside from sponges. The Journal of the American Chemical Society. 1950;72:2809-2810
  19. 19. Proksch P, Edrada RA, Ebel R. Drugs from the seas—Current status and microbiological implications. Applied Microbiology and Biotechnology. 2002;59:125-134
  20. 20. Schwartsmann G. Marine organisms as a source of new anticancer agents. Annals of Oncology. 2000;11(3):235-243
  21. 21. Feldman EJ, Lancet JE, Kolitz JE, Ritchie EK, et al. First-in-man study of CPX-351: A liposomal carrier containing cytarabine and daunorubicin in a fixed 5:1 molar ratio for the treatment of relapsed and refractory acute myeloid leukemia. Journal of Clinical Oncology. 2011;29:979-985
  22. 22. Mayer AM, Glaser KB, Cuevas C, Jacobs RS, et al. The odyssey of marine pharmaceuticals: A current pipeline perspective. Trends in Pharmacological Sciences. 2010;31:255-265
  23. 23. Crews P, Gewick WH, Schmitz FJ, France D, Bair KW, et al. Molecular approaches to discover marine natural product anticancer leads—An update from a drug discovery group collaboration. Pharmaceutical Biology. 2003;141:39-52
  24. 24. Halim R, Gladman B, Danquah MK, Webley PA. Oil extraction from microalgae for biodiesel production. Bioresource Technology. 2011;102(1):178-185
  25. 25. Guzii AG, Makarieva TN, Denisenko VA, Dmitrenok PS, et al. Monanchocidin: A new apoptosis-inducing polycyclic guanidine alkaloid from the marine sponge Monanchora pulchra. Organic Letters. 2010;12(19):4292-4295
  26. 26. Kondracki ML, Guyot M. Smenospongine: A cytotoxic and antimicrobial aminoquinone isolated from Smenospongia sp. Tetrahedron Letters. 1987;27:5815-5818
  27. 27. Kong D, Yamori T, Kobayashi M, Duan H. Antiproliferative and antiangiogenic activities of smenospongine, a marine sponge sesquiterpene aminoquinone. Marine Drugs. 2011;9:154-161
  28. 28. Valeriote FA, Tenney K, Media J, Pietraszkiewicz H, et al. Discovery and development of anticancer agents from marine sponges: perspectives based on a chemistry-experimental therapeutics collaborative program. Journal of Experimental Therapeutics & Oncology. 2012;10:119-134
  29. 29. Rabelo L, Monteiro N, Serquiz R, Santos P, et al. A lactose-binding lectin from the marine sponge Cinachyrella apion (Cal) induces cell death in human cervical adenocarcinoma cells. Marine Drugs. 2012;10:727-743
  30. 30. Rothmeier E, Pfaffinger G, Hoffmann C, Harrison CF, et al. Activation of Ran GTPase by a Legionella effector promotes microtubule polymerization, pathogen vacuole motility and infection. PLoS Pathogens. 2013;9:e1003598. DOI: 10.1371/journal.ppat.1003598
  31. 31. Inman WD, Bray WM, Gassner NC, Lokey RS, et al. β-Carboline alkaloid from the Papau New Guinea marine sponge Hyrtios reticulates. Journal of Natural Products. 2010;73:255-257
  32. 32. Lyakhova EG, Kolesnikova SA, Kalinovsky AI, Afiyatullov SS, et al. Bromine-containing alkaloids from the marine sponge Penares sp. Tetrahedron Letters. 2012;53:6119-6122
  33. 33. Pham CD, Hartmann R, Muller WE, de Voogd N, Lai D, Proksch P. Aaptamine derivatives from the Indonesian sponge Aaptos suberitoides. Journal of Natural Products. 2013;76:103-106
  34. 34. Laville R, Thomas OP, Berrué F, Marquez D, Vacelet J, Amade P. Bioactive guanidine alkaloids from two Caribbean marine sponges. Journal of Natural Products. 2009;72:1589-1594
  35. 35. Daikuhara N, Tada Y, Yamaki S, Charupant K, et al. Renieramycins T and U, novel renieramycin–ecteinascidin hybrid marine natural products from Thai sponge Xestospongia sp. Tetrahedron Letters. 2009;50:4276
  36. 36. Prawat H, Mahidol C, Kaweetripob W, Wittayalai S, Ruchirawat S. Iodo–sesquiterpene hydroquinone and brominated indole alkaloids from the Thai sponge Smenospongia sp. Tetrahedron. 2012;68:6881-6886
  37. 37. Momose R, Tanaka N, Fromont J, Kobayashi J. Hyrtimomines A-C, new heteroaromatic alkaloids from a sponge Hyrtios sp. Organic Letters. 2013;15:2010-2013
  38. 38. Tran TD, Pham NB, Fechner G, Hooper JN, Quinn RJ. Bromotyrosine alkaloids from the Australian marine sponge Pseudoceratina verrucosa. Journal of Natural Products. 2013;76:516-523
  39. 39. Kubota T, Kura KI, Fromont J, Kobayashi JI. Tetrahedron. 2013;69:96
  40. 40. Ibrahim SRM, Mohamed GA, Elkhayat ES, Fouad MA, Proksch P. Marine pyridoacridine alkaloids: Biosynthesis and biological activities. Bulletin of Faculty of Pharmacy, Cairo University. 2013;51:229
  41. 41. Makarieva TN, Tabakmaher KM, Guzii AG, Denisenko VA, et al. Monanchocidins B-E: Polycyclic guanidine alkaloids with potent antileukemic activities from the sponge Monanchora pulchra. Journal of Natural Products. 2011;74:1952
  42. 42. Sun YT, Lin B, Li SG, Liu M, et al. New bromopyrrole alkaloids from the marine sponge Agelas sp. Tetrahedron. 2017;73:2786-2792
  43. 43. Nguyen HT, Chau VM, Tran TH, Phan VK, et al. C29 sterols with a cyclopropane ring at C-25 and 26 from the Vietnamese marine sponge Ianthella sp. and their anticancer properties. Bioorganic & Medicinal Chemistry Letters. 2009;19:4584-4585
  44. 44. Ushiyama S, Umaoka H, Kato H, Suwa Y, et al. Manadosterols A and B, sulfonated sterol dimers inhibiting the Ubc13-Uev1A interaction, isolated from the marine sponge Lissodendryx fibrosa. Journal of Natural Products. 2012;75:1495
  45. 45. Harinantenaina L, Brodie PJ, Maharavo J, Bakary G, et al. Antiproliferative homoscalarane sesterterpenes from two Madagascan sponges. Bioorganic & Medicinal Chemistry. 2013;21:2912-2917
  46. 46. Rateb ME, Houssen WE, Schumacher M, Harrison WTA, et al. Bioactive diterpene derivatives from the marine sponge Spongionella sp. Journal of Natural Products. 2009;72:1471-1476
  47. 47. Wang CY, Wang BG, Wiryowidagdo S, Wray V, et al. Melophlins C–O, thirteen novel tetramic acids from the marine sponge Melophlus sarassinorum. Journal of Natural Products. 2003;66(1):51-56
  48. 48. Hahn D, Won DH, Mun B, Kim H, et al. Cytotoxic scalarane sesterterpenes from a Korean marine sponge Psammocinia sp. Bioorganic & Medicinal Chemistry Letters. 2013;23(8):2336-2339
  49. 49. Lamoral-Theys D, Fattorusso E, Mangoni A, Perinu C, et al. Evaluation of the antiproliferative activity of diterpene Isonitriles from the Sponge Pseudoaxinella flava in apoptosis-sensitive and apoptosis-resistant cancer cell lines. Journal of Natural Products. 2011;74(10):2299-2303
  50. 50. Pettit GR, Tang Y, Zhang Q, Bourne GT, et al. Isolation and structures of axistatins 1-3 from the Republic of Palau marine sponge Agelas axifera Hentschel(1). Journal of Natural Products. 2013;76(3):420-422
  51. 51. Ovenden SPB, Nielson JL, Liptrot CH, Willis RH, et al. Metachromins U–W: Cytotoxic merosesquiterpenoids from an Australian specimen of the sponge Thorecta reticulata. Journal of Natural Products. 2011;73(3):467-471
  52. 52. Bokesch HR, Pannell LK, McKee TC, Boyd MR. Coscinamides A, B and C, three new bis indole alkaloids from the marine sponge Coscinoderma sp. Tetrahedron Letters. 2000;41:6305-6308
  53. 53. Kim CK, Song IH, Park HY, Lee YJ, et al. Suvanine sesterterpenes and deacyl irciniasulfonic acids from a tropical Coscinoderma sp. sponge. Journal of Natural Products. 2014;77(6):1396-1403
  54. 54. Oku N, Takada K, Fuller RW, Wilson JA, et al. Isolation, structural elucidation, and absolute stereochemistry of enigmazole A, a cytotoxic phosphomacrolide from the Papua New Guinea marine sponge Cinachyrella enigmatica. Journal of the American Chemical Society. 2010;132(30):10278-10285
  55. 55. Gala F, D’Auria MV, De Marino S, Sepe V, et al. Jaspamides M–P: New tryptophan modified jaspamide derivatives from the sponge Jaspis splendens. Tetrahedron. 2009;65:51-56
  56. 56. Singh AJ, Razzak M, Teesdale-Spittle P, Gaitanos TN, et al. Structure-activity studies of the pelorusides: New congeners and semi-synthetic analogues. Organic & Biomolecular Chemistry. 2011;9(12):4456-4466
  57. 57. Sorres J, Martin MT, Petek S, Levaique H, et al. Pipestelides A-C: Cyclodepsipeptides from the Pacific marine sponge Pipestela candelabra. Journal of Natural Products. 2012;75(4):759-763
  58. 58. Zhang J, Tang X, Li J, Li P, et al. Cytotoxic polyketide derivatives from the South China sea sponge Plakortis simplex. Journal of Natural Products. 2013;76(4):600-606
  59. 59. Jiménez-Romero C, Ortiz I, Vicente J, Vera B, Rodríguez AD, et al. Bioactive cycloperoxides isolated from the Puerto Rican sponge Plakortis halichondrioides. Journal of Natural Products. 2010;73(10):1694-1700
  60. 60. Pelay-Gimeno M, García-Ramos Y, Martin MJ, Jan S, et al. The first total synthesis of the cyclodepsipeptide pipecolidepsin A. Nature Communications. 2013;4:2352
  61. 61. Zampella A, Sepe V, Luciano P, Bellotta F, et al. Homophymine A, an anti-HIV cyclodepsipeptide from the sponge Homophymia sp. The Journal of Organic Chemistry. 2008;73(14):5319-5327
  62. 62. Tran TD, Pham NB, Fechner G, Zencak D, et al. Cytotoxic cyclic depsipeptides from the Australian marine sponge Neamphius huxleyi. Journal of Natural Products. 2012;75(12):2200-2208
  63. 63. Williams DE, Yu K, Behrisch HW, Van Soest R, Andersen RJ. Rolloamides A and B, cytotoxic cyclic heptapeptides isolated from the Caribbean marine sponge Eurypon laughlini. Journal of Natural Products. 2009;72(7):1253-1257
  64. 64. Wang X, Morinaka BI, Molinski TF. Structures and solution conformational dynamics of stylissamides G and H from the Bahamian sponge Stylissa caribica. Journal of Natural Products. 2014;77(3):625-630
  65. 65. Coello L, Reyes F, Martín MJ, Cuevas C, Fernández R. Isolation and structures of pipecolidepsins A and B, cytotoxic cyclic depsipeptides from the Madagascan sponge Homophymia lamellosa. Journal of Natural Products. 2014;77(2):298-303
  66. 66. Regalado EL, Jimenez-Romero C, Genta-Jouve G, Tasdemir D, et al. Acanthifoliosides, minor steroidal saponins from the Caribbean sponge Pandaros acanthifolium. Tetrahedron. 2011;67:1011-1018
  67. 67. Ye J, Zhou F, Al-Kareef AMQ, Wang H. Anticancer agents from marine sponges. Journal of Asian Natural Products Research. 2015;17:64-88
  68. 68. Jiao W-H, Huang X-J, Yang J-S, Yang F, et al. Dysidavarones A–D, new sesquiterpene quinones from the marine sponge Dysidea avara. Organic Letters. 2012;14(1):202-205
  69. 69. Daletos G, de Voogd NJ, Müller WE, Wray V, et al. Cytotoxic and protein kinase inhibiting nakijiquinones and nakijiquinols from the sponge Dactylospongia metachromia. Journal of Natural Products. 2014;77(2):218-226
  70. 70. Jiao WH, Xu TT, Yu HB, Chen GD, et al. Dysideanones A-C, unusual sesquiterpene quinones from the South China Sea sponge Dysidea avara. Journal of Natural Products. 2014;77(2):346-350
  71. 71. Shaala LA, Bamane FH, Badr JM, Youssef DT. Brominated arginine-derived alkaloids from the red sea sponge Suberea mollis. Journal of Natural Products. 2011;74(6):1517-1520
  72. 72. Williams A, Bax NJ, Kloser RJ, Althaus F, et al. Australia’s deep-water reserve network: implications of false homogeneity for classifying abiotic surrogates of biodiversity. ICES Journal of Marine Science. 2009;66:214-224
  73. 73. Hwang IH, Oh J, Kochanowska-Karamyan A, et al. A novel natural phenyl alkene with cytotoxic activity. Tetrahedron Letters. 2013;54(29):3872-3876
  74. 74. Farokhi F, Wielgosz-Collin G, Robic A, Debitus C, et al. Antiproliferative activity against human non-small cell lung cancer of two O-alkyl-diglycosylglycerols from the marine sponges Myrmekioderma dendyi and Trikentrion laeve. European Journal of Medicinal Chemistry. 2012;49:406-410
  75. 75. Lee Y-J, Han S, Lee H-S, Kang JS, et al. Cytotoxic psammaplysin analogues from a Suberea sp. marine sponge and the role of the spirooxepinisoxazoline in their activity. Journal of Natural Products. 2013;76(9):1731-1736
  76. 76. Kobayashi M. In search for biologically active substances from marine sponges. In: Fusetani N, editor. Drugs from the Sea. Basel, Switzerland: Karger; 2000. pp. 46-58
  77. 77. Amade PG, Chariou G, Baby C, Vacelet J. Antimicrobial activity of marine sponges of Mediterranean. Sea. Marine Biology. 1987;94:271-275
  78. 78. McCaffrey EJ, Endeau R. Antimicrobial activity of tropical and subtropical sponges. Marine Biology. 1985;89:1-8
  79. 79. Mayer AM, Hamann MT. Marine pharmacology in 2000: Marine compounds with antibacterial, anticoagulant, antifungal, anti-inflammatory, antimalarial, antiplatelet, antituberculosis, and antiviral activities; affecting the cardiovascular, immune, and nervous systems and other miscellaneous mechanisms of action. Marine Biotechnology New York. 2004;6(1):37-52
  80. 80. Sipkema D, Franssen MC, Osinga R, Tramper J, Wijffels RH. Marine sponges as pharmacy. Marine Biotechnology New York. 2005;7(3):142-162
  81. 81. Torres YR, Berlink RGS, Nascimento GGF, Fortier SC, et al. Antibacterial activity against resistant bacteria and cytotoxicity of four alkaloid toxins isolated from the marine sponge. Arenosclera brasiliensis. Toxicon. 2002;40(7):885-891
  82. 82. De Silva ED, Scheuer PJ. Manoalide, an antibiotic sesterpenoid from the marine sponge Luffariella variablis. Tetrahedron Letters. 1980;21:1611-1614
  83. 83. Urban S, Leone Pde A, Carroll AR, Fechner GA, et al. Axinellamines A−D, novel imidazo−azolo−imidazole alkaloids from the Australian marine sponge Axinella sp. The Journal of Organic Chemistry. 1999;64(3):731-735
  84. 84. Rao KV, Kasanah N, Wahyuono S, Tekwani BL, et al. Three new manzamine alkaloids from a common Indonesian sponge and their activity against infectious and tropical parasitic diseases. Journal of Natural Products. 2004;67(8):1314-1318
  85. 85. Torres YR, Berlinck RG, Magalhães A, Schefer AB, Ferreira AG, et al. Arenosclerins A-C and haliclonacyclamine E, new tetracyclic alkaloids from a Brazilian endemic Haplosclerid sponge Arenosclera brasiliensis. Journal of Natural Products. 2000;63(8):1098-1105
  86. 86. Oh KB, Mar W, Kim S, Kim JY, et al. Antimicrobial activity and cytotoxicity of bis(indole) alkaloids from the sponge Spongosorites sp. Biological & Pharmaceutical Bulletin. 2006;29(3):570-573
  87. 87. Pettit GR, Knight JC, Collins JC, Herald DL, et al. Antineoplastic agents 430. Isolation and structure of cribrostatins 3, 4, and 5 from the Republic of Maldives Cribrochalina species. The Journal of Natural Products. 2000;63:793-798
  88. 88. Pettit GR, Collins JC, Knight JC, Herald DL, et al. Antineoplastic agents 485. Isolation and structure of cribrostatin 6, a dark blue cancer cell growth inhibitor from the marine sponge Cribrochalina sp. Journal of Natural Products. 2003;66:544-547
  89. 89. Carroll AR, Ngo A, Quinn RJ, Redburn J, Hooper JN. Petrosamine B, an inhibitor of the Helicobacter pylori enzyme aspartyl semialdehyde dehydrogenase from the Australian sponge Oceanapia sp. Journal of Natural Products. 2005;68(5):804-806
  90. 90. Ford J, Capon RJ. Discorhabdin R: A new antibacterial pyrroloiminoquinone from two latrunculiid marine sponges, Latrunculia sp. and Negombata sp. Journal of Natural Products. 2000;63(11):1527-1528
  91. 91. Gupta L, Talwar A, Chauhan PMS. Bis and tris indole alkaloids from marine organisms: New leads for drug discovery. Current Medicinal Chemistry. 2007;14:1789-1803
  92. 92. Oliveira JHHL, Grube A, Köck M, Berlinck RGS, et al. Ingenamine G and cyclostelletamines G–K from the new Brazilian species of marine sponge Pachychalina sp. Journal of Natural Products. 2004;67:1685-1689
  93. 93. Oliveira JHHL, Seleghim MHR, Timm C, Grube A, et al. Antimicrobial and antimycobacterial activity of cyclostellettamine alkaloids from sponge Pachychalina sp. Marine Drugs. 2006;4:1-8
  94. 94. Vik A, Hedner E, Charnock C, Samuelsen O, et al. (+)-Agelasine D: Improved synthesis and evaluation of antibacterial and cytotoxic activities. Journal of Natural Products. 2006;69(3):381-386
  95. 95. Rubio BK, Soest RW, Crews P. Extending the record of meroditerpenes from Cacospongia marine sponges. Journal of Natural Products. 2007;70(4):628-631
  96. 96. Gul FA, Jaggi BL, Krishnan GV. Auditor independence: Evidence on the joint effects of auditor tenure and nonaudit fees. Auditing: A Journal of Practice & Theory. 2007;26(2):117-142
  97. 97. Matsunaga S, Okada Y, Fusetani N, van Soest RWM. An antimicrobial C14 acetylenic acid from a marine sponge Oceanapia species. Journal of Natural Products. 2000;63(5):690-691
  98. 98. Linington RG, Robertson M, Gauthier A, Finlay B, et al. Caminosides B−D, antimicrobial glycolipids isolated from the marine sponge Caminus sphaeroconia. Journal of Natural Products. 2006;69(2):173-177
  99. 99. Grube A, Assmann M, Lichte E, Sasse F, et al. Bioactive metabolites from the Caribbean Sponge Aka coralliphagum. Journal of Natural Products. 2007;70(4):504-509
  100. 100. Moura RM, Queiroz AF, Fook JM, Dias AS, et al. CvL, a lectin from the marine sponge Cliona varians: Isolation, characterization and its effects on pathogenic bacteria and Leishmania promastigotes. Comparative Biochemistry and Physiology. Part A, Molecular & Integrative Physiology. 2006;145(4):517-523
  101. 101. El Sayed KA, Youssef DT, Marchetti D. Bioactive natural and semisynthetic latrunculins. Journal of Natural Products. 2006;69(2):219-223
  102. 102. Nicolaou KC, Hughes R, Pfefferkorn JA, Barluenga S, Roecker AJ. Combinatorial synthesis through disulfide exchange: Discovery of potent Psammaplin A type antibacterial agents active against methicillin-resistant Staphylococcus aureus (MRSA). Chemistry—A European Journal. 2001;7(19):4280-4295
  103. 103. Donia M, Hamann M. Marine natural products and their potential applications as anti-infective agents. The Lancet Infectious Diseases. 2003;3:338-348
  104. 104. Blunt JW, Copp BR, Munro MH, Northcote PT, Prinsep MR. Marine natural products. Natural Product Reports. 2004;21:1-49
  105. 105. Bergmann W, Feeney RJ. Contribution to the study of marine products of sponges. The Journal of Organic Chemistry. 1951;16:981-987
  106. 106. De Clerq E. New anti-HIV agents and targets. Medicinal Research Reviews. 2002;22:531-565
  107. 107. Collum LM, O’Connor M, Logan P. Comparison of the efficacy and toxicity of acyclovir and of adenine arabinoside when combined with dilute betamethasone in herpetic disciform keratitis: Preliminary results of a double-blind trial. Transactions of the Ophthalmological Societies of the United Kingdom. 1983;103:597-599
  108. 108. Whitley RJ, Gnann JW Jr, Hinthorn D, Liu C, et al. The NIAID Collaborative Antiviral Study Group. Disseminated herpes zoster in the immunocompromised host: A comparative trial of acyclovir and vidarabine. The Journal of Infectious Diseases. 1992;165:450-455
  109. 109. Kamiyama T, Kurokawa M, Shiraki K. Characterization of the DNA polymerase gene of varicellazoster viruses resistant to acyclovir. The Journal of General Virology. 2001;82:2761-2765
  110. 110. Souza TM, Abrantes JL, Epifanio R, Leite-Fontes CF, Frugulhetti IC. The alkaloid 4-methylaaptamine isolated from the sponge Aaptos aaptos impairs Herpes simplex virus type 1 penetration and immediate-early protein synthesis. Planta Medica. 2007;73(3):200-205
  111. 111. Cutignano A, Bifulco G, Bruno I, Casapullo A, et al. Dragmacidin F: A new antiviral bromoindole alkaloid from the Mediterranean sponge Halicortex spp. Tetrahedron. 2000;56:3743-3748
  112. 112. Yousaf M, Hammond NL, Peng J, Wahyuono S, et al. New manzamine alkaloids from an Indo-Pacific sponge. Pharmacokinetics, oral availability, and the significant activity of several manzamines against HIV-I, AIDS opportunistic infections, and inflammatory diseases. Journal of Medicinal Chemistry. 2004;47(14):3512-3517
  113. 113. Perry WL, Hustad CM, Swing DA, O'Sullivan TN, et al. The itchy locus encodes a novel ubiquitin protein ligase that is disrupted in a18H mice. Nature Genetics. 1998;18(2):143-146
  114. 114. Ford PW, Gustafson KR, McKee TC, Shigematsu N, et al. Papuamides A−D, HIV-inhibitory and cytotoxic depsipeptides from the Sponges Theonella mirabilis and Theonella swinhoei collected in Papua New Guinea. Journal of the American Chemical Society. 1999;121(25):5899-5909
  115. 115. Rashid MA, Gustafson KR, Cartner LK, Shigematsu N, et al. Microspinosamide, a new HIV-inhibitory cyclic depsipeptide from the marine sponge Sidonops microspinosa. Journal of Natural Products. 2001;64(1):117-121
  116. 116. Qureshi A, Faulkner DJ. Haplosamates A and B: New steroidal sulfamate esters from two haplosclerid sponges. Tetrahedron. 1999;55(28):8323-8330
  117. 117. Muller WEG, Sobel C, Diehl-Seifert B, Maidhof A, Schroder HC. Influence of the antileukemic and anti-human immunodeficiency virus agent avarol on selected immune responses in vitro and in vivo. Biochemical Pharmacology. 1987;36:1489-1494
  118. 118. Perry NB, Blunt JW, Munro MHG, Thompson AM. Antiviral and antitumor agents from a New Zealand sponge, Mycale sp. The Journal of Organic Chemistry. 1990;55:223-227
  119. 119. Mehta A, Zitzmann N, Rudd PM, et al. Alpha-glucosidase inhibitors as potential broad based anti-viral agents. FEBS Letters. 1998;430:17-22
  120. 120. Kelve M, Kuusksalu A, Lopp A, Reintamm T. Sponge (2′,5′)oligoadenylate synthetase activity in the whole sponge organism and in a primary cell culture. Journal of Biotechnology. 2003;100:177-180
  121. 121. Wellington KD, Cambie RC, Rutledge PS, Bergquist PR. Chemistry of sponges. 19. Novel bioactive metabolites from Hamigera tarangaensis. Journal of Natural Products. 2000;63:79-85
  122. 122. Sun HH, Cross SS, Gunasekera M, Koehn FE. Weinbersterol disulfates A and B, antiviral steroid sulfates from the sponge Petrosia weinbergi. Tetrahedron. 1991;47:1185-1190
  123. 123. García-Ruiz JC, Amutio E, Pontón J. Invasive fungal infection in immunocompromised patients. Revista Iberoamericana de Micología. 2004;21:55-62
  124. 124. Walsh TJ, Groll A, Hiemenz J, Fleming R, et al. Infections due to emerging and uncommon medically important fungal pathogens. Clinical Microbiology and Infection. 2004;10(S1):48-66
  125. 125. Giusiano G, Mangiaterra M, Rojas F, Gámez V. Yeasts species distribution in Neonatal Intensive Care Units in northeast Argentina. Mycoses. 2004;47:300-303
  126. 126. Giusiano G, Mangiaterra M, Rojas F, Gámez V. Azole resistance in neonatal intensive care units in Argentina. Journal of Chemotherapy. 2005;17:347-350
  127. 127. Rashid MA, Gustafson KR, Boswell JL, Boyd MR. Haligramides A and B, two new cytotoxic hexapeptides from the marine sponge Haliclona nigra. Journal of Natural Products. 2000;63:956-959
  128. 128. Dunbar DC, Rimoldi JM, Clark AM, Kelly M, Hamann MT. Anti-cryptococcal and nitric oxide synthase inhibitory imidazole alkaloids from the Calcareous sponge Leucettacf chagosensin. Tetrahedron. 2000;56:8795-8798
  129. 129. Kon Y, Kubota T, Shibazaki A, Gonoi T, Kobayashi J. Ceratinadins A–C, new bromotyrosine alkaloids from an Okinawan marine sponge Pseudoceratina sp. Bioorganic & Medicinal Chemistry Letters. 2010;20:4569-4572
  130. 130. Stout EP, Yu LC, Molinski TF. Antifungal diterpene alkaloids from the Caribbean sponge Agelas citrina: Unified configurational assignments of agelasidines and agelasines. European Journal of Organic Chemistry. 2012;2012:5131-5135
  131. 131. Arevabini C, Crivelenti YD, de Abreu MH, Bitencourt TA, Santos MF, et al. Antifungal activity of metabolites from the marine sponges Amphimedon sp. and Monanchora arbuscula against Aspergillus flavus strains isolated from peanuts (Arachis hypogaea). Natural Product Communications. 2014;9:33-36
  132. 132. Ettinger-Epstein P, Tapiolas DM, Motti CA, Wright AD, Battershill CN, de Nys R. Production of manoalide and its analogues by the sponge Luffariella variabilis is hardwired. Marine Biotechnology. 2008;10(1):64-74
  133. 133. Zhang X, Jacob MR, Rao RR, Wang YH, et al. Antifungal cyclic peptides from the marine sponge. Research and Reports in Medicinal Chemistry. 2012;2:7-14
  134. 134. Xu WH, Ding Y, Jacob MR, Agarwal AK, Clark AM, Ferreira D, et al. Puupehanol, a sesquiterpene-dihydroquinone derivative from the marine sponge Hyrtios sp. Bioorganic & Medicinal Chemistry Letters. 2009;19:6140-6143
  135. 135. Boonlarppradab C, Faulkner DJ. Eurysterols A and B, cytotoxic and antifungal steroidal sulfates from a marine sponge of the Genus Euryspongia. Journal of Natural Products. 2007;77(4):818-823
  136. 136. Digirolamo JA, Li XC, Jacob MR, Clark AM, Ferreira D. Reversal of fluconazole resistance by sulfated sterols from the marine sponge Topsentia sp. Journal of Natural Products. 2009;72:1524-1528
  137. 137. Gunasekera SP, Pomponi SA, McCarthy PJ. Discobahamins A and B, new peptides from the Bahamian deep water marine sponge Discodermia sp. Journal of Natural Products. 1994;57(1):79-83
  138. 138. Scott VR, Boehme R, Matthews TR. New class of antifungal agents: Jasplakinolide, a cyclodepsipeptide from the marine sponge, Jaspis species. Antimicrobial Agents and Chemotherapy. 1988;32(8):1154-1157
  139. 139. Sepe V, D’Osri R, Borbone N, D'Auria MV, et al. Towards new ligands of nuclear receptors. Discovery of malaitasterol A, and unique bis-secosterol from marine sponge Theonella swinhoei. Tetrahedron. 2006;62:833-840
  140. 140. Youssef DT, Shaala LA, Mohamed GA, Badr JM, et al. Theonellamide G, a potent antifungal and cytotoxic bicyclic glycopeptide from the Red Sea marine sponge Theonella swinhoei. Marine Drugs. 2014;12:1911-1923
  141. 141. Matsunaga S, Fusetani N, Hashimoto K, Walchli M. Theonellamide F. A novel antifungal bicyclic peptide from a marine sponge Theonella sp. Journal of the American Chemical Society. 1989;111:2582-2588
  142. 142. Vik A, Hedner E, Charnock C, Tangen LW, et al. Antimicrobial and cytotoxic activity of agelasine and agelasimine analogs. Bioorganic & Medicinal Chemistry. 2007;15(12):4016-4037
  143. 143. Jamison MT, Molinski TF. Antipodal crambescin A2 homologues from the marine sponge Pseudaxinella reticulata antifungal structure–activity relationships. Journal of Natural Products. 2015;78:557-561
  144. 144. Nishimura S, Arita Y, Honda M, Iwamoto K, et al. Marine antifungal theonellamides target 3beta-hydroxysterol to activate Rho1 signaling. Nature Chemical Biology. 2010;6:519-526
  145. 145. Kumar R, Subramani R, Feussner KD, Aalbersberg W. Aurantoside K, a new antifungal tetramic acid glycoside from a Fijian marine sponge of the genus Melophlus. Marine Drugs. 2012;10:200-208
  146. 146. Rudi A, Kashman Y. Three new cytotoxic metabolites from the marine sponge Plakortis halichondrioides. Journal of Natural Products. 1993;56:1827-1830
  147. 147. El-Amraoui B, Biard JF, Fassouane A. Haliscosamine: A new antifungal sphingosine derivative from the Moroccan marine sponge Haliclona viscosa. Springer Plus. 2013;2:252
  148. 148. Sionov E, Roth D, Sandovsky-Losica H, Kashman Y, et al. Antifungal effect and possible mode of activity of a compound from the marine sponge Dysidea herbacea. The Journal of Infection. 2005;50:453-460
  149. 149. Chen Y, McCarthy PJ, Harmody DK, Schimoler-O'Rourke R, et al. New bioactive peroxides from marine sponges of the family plakiniidae. Journal of Natural Products. 2002;65:1509-1512
  150. 150. Vuong D, Capon RJ, Lacey E, Gill JH, et al. Onnamide F: A new nematocide from a southern Australian marine sponge, Trachycladus laevispirulifer. Journal of Natural Products. 2001;64:640-642
  151. 151. Edrada RA, Ebel R, Supriyono A, Wray V, et al. Swinhoeiamide A, a new highly active calyculin derivative from the marine sponge, Theonella swinhoei. Journal of Natural Products. 2002;65:1168-1172
  152. 152. Wright AD, McCluskey A, Robertson MJ, MacGregor KA, et al. Anti-malarial, anti-algal, anti-tubercular, anti-bacterial, anti-photosynthetic, and anti-fouling activity of diterpene and diterpene isonitriles from the tropical marine sponge Cymbastela Hooperi. Organic & Biomolecular Chemistry. 2011;9:400-407
  153. 153. Chen Y, Killday KB, McCarthy PJ, Schimoler R, et al. Three new peroxides from the sponge Plakinastrella species. Journal of Natural Products. 2001;64:262-264
  154. 154. Rifai S, Fassouane A, Kijjoa A, VanSoest R. Antimicrobial activity of Untenospongin B, a metabolite from the marine sponge Hippospongia communis collected from the Atlantic Coast of Morocco. Marine Drugs. 2004;2:147-153
  155. 155. Piao SJ, Song YL, Jiao WH, Yang F, et al. Hippolachnin A, a new antifungal polyketide from the South China sea sponge Hippospongia lachne. Organic Letters. 2013;15:3526-3529
  156. 156. Herecia F, Ubeda A, Ferrandiz ML, Terencio MC, et al. Anti-inflammatory activity in mice of extracts from Mediterranean marine invertebrates. Life Sciences. 1998;62:PL115
  157. 157. Sturm C, Paper DH, Franz G. Screening for immune response modifiers from marine origin. Pharmaceutical and Pharmacological Letters. 1999;9:76
  158. 158. Jiang YH, Ryu S-H, Ahn E-Y, You S, et al. Antioxidant activity of (8E,13Z,20Z)-strobilinin/(7E,13Z,20Z)-felixinin from a marine sponge Psammocinia sp. Natural Product Sciences. 2004;10(6):272-276
  159. 159. Keyzers RA, Northcote PT, Zubkov OA. Novel anti-inflammatory spongian diterpenes from the New Zealand marine sponge Chelonaplysilla violacea. European Journal of Organic Chemistry. 2004;(2):419-425
  160. 160. Posadas I, Terencio MC, De Rosa S, Paya M. Cavernolide: A new inhibitor of human sPLA2 sharing unusual chemical features. Life Sciences. 2000;67:3007
  161. 161. Lucas R, Casapullo A, Ciasullo L, Gomez-Paloma L, Payá M. Cycloamphilectenes, a new type of potent marine diterpenes: Inhibition of nitric oxide production in murine macrophages. Life Sciences. 2003;72(22):2543-2552
  162. 162. Cichewicz RH, Kenyon VA, Whitman S, Morales NM, et al. Redox inactivation of human 15-lipoxygenase by marine-derived meroditerpenes and synthetic chromanes: Archetypes for a unique class of selective and recyclable inhibitors. Journal of the American Chemical Society. 2004;126:14910-14920
  163. 163. Keysers C, Wicker B, Gazzola V, Anton JL, et al. A touching sight: SII/PV activation during the observation and experience of touch. Neuron. 2004;42(2):335-346
  164. 164. Ferrandiz ML, Sanz MJ, Bustos G, Paya M, et al. Avarol and avarone, two new anti-inflammatory agents of marine origin. European Journal of Pharmacology. 1994;253:75-82
  165. 165. Muller WEG, Bohm M, Batel R, De Rosa S, et al. Application of cell culture for the production of bioactive compounds from sponges: Synthesis of avarol by primmorphs from Dysidea avara. Journal of Natural Products. 2000;63:1077-1081
  166. 166. Posadas I, Terencio MC, Giannini C, D’Auria MV, Paya M. Dysidotronic acid, a new sesquiterpenoid, inhibits cytokine production and the expression of nitric oxide synthase. European Journal of Pharmacology. 2001;415:285-292
  167. 167. Gunasekera SP, Isbrucker RA, Longley RF, Wright AE, et al. Plakolide A, a new gamma-lactone from the marine sponge Plakortis sp. Journal of Natural Products. 2004;67:110
  168. 168. Lu PH, Chueh SC, Kung FL, Pan SL, et al. Ilimaquinone, a marine sponge metabolite, displays anticancer activity via GADD153-mediated pathway. European Journal of Pharmacology. 2007;556:45-54
  169. 169. Glase KB, Lock YW. Regulation of prostaglandin H synthase 2 expression in human monocytes by the marine natural products manoalide and scalaradial. Novel effects independent of inhibition of lipid mediator production. Biochemical Pharmacology. 1995;50:913-922
  170. 170. Pastor PG, De Rosa S, De Giulio A, Paya M, Alcaraz MJ. Modulation of acute and chronic inflammatory processes by cacospongionolide B, a novel inhibitor of human synovial phospholipase A2. British Journal of Pharmacology. 1999;126:301-311
  171. 171. Giannini C, Debitus C, Lucas R, Ubeda A, et al. New sesquiterpene derivatives from the sponge Dysidea species with a selective inhibitor profile against human phospholipase A2 and other leukocyte functions. Journal of Natural Products. 2001;64:612-615
  172. 172. Miyaoka H, Yamanishi M, Mitome H. PLA2 inhibitory activity of marine sesterterpenoids cladocorans, their diastereomers and analogues. Chemical & Pharmaceutical Bulletin. 2006;54:268-270
  173. 173. Dal-Piaz F, Casapullo A, Randazzo A, Riccio R, Pucci P, et al. Molecular basis of phospholipase A2 inhibition by petrosaspongiolide M. Chembiochem. 2002;3:664-671
  174. 174. Garcia P, Randazzo A, Gomez-Paloma L, Alcaraz MJ, Paya M. Effects of petrosaspongiolide M, a novel phospholipase A2 inhibitor, on acute and chronic inflammation. The Journal of Pharmacology and Experimental Therapeutics. 1999;289:166-172
  175. 175. Barnette MS, Rush J, Marshall LA, Foley JJ, et al. Effects of scalaradial, a novel inhibitor of 14 kDa phospholipase A2, on human neutrophil function. Biochemical Pharmacology. 1994;47:1661-1667
  176. 176. Fontana A, Mollo E, Ortea J, Cavagnin M, Cimino G. Scalarane and homoscalarane compounds from the nudibranchs Glossodoris sedna and Glossodoris dalli: Chemical and biological properties. Journal of Natural Products. 2000;63:527-530
  177. 177. Amagata T, Whitman S, Jonson TA, Stessman CC, et al. Exploring sponge-derived terpenoids for their potency and selectivity against 12-human, 15-human, and 15-soybean lipoxygenases. Journal of Natural Products. 2003;66:230-235
  178. 178. D'acquisto F, Lanzotti V, Carnuccio R. Cyclolinteinone, a sesterterpene from sponge Cacospongia linteiformis, prevents inducible nitric oxide synthase and inducible cyclo-oxygenase protein expression by blocking nuclear factor-kappa B activation in J774 macrophages. The Biochemical Journal. 2000;346(3):793-798
  179. 179. Fukami A, Ikeda Y, Kondo S, Naganawa H, et al. Akaterpin, a novel bioactive triterpene from the marine sponge Callyspongia sp. Tetrahedron Letters. 1997;38:1201-1202
  180. 180. Keyzers RA, Norticote PT, Webb V. Clathriol, a novel polyoxygenated 14β steroid isolated from the New Zealand marine sponge Clathria lissosclera. Journal of Natural Products. 2002;65:598-600
  181. 181. Mandeaua A, Debitus C, Ariès M-F, David B. Isolation and absolute configuration of new bioactive marine steroids from Euryspongia n. sp. Steroids. 2005;70:873-878
  182. 182. Sharma V, Lansdell TA, Jin G, Tepe JJ. Inhibition of cytokine production by hymenialdisine derivatives. Journal of Medicinal Chemistry. 2004;47(14):3700-3703
  183. 183. Tasdemir D, Mallon R, Greenstein M, Feldberg LR, et al. Aldisine alkaloids from the Philippine sponge Stylissa massa are potent inhibitors of mitogen-activated protein kinase kinase-1 (MEK-1). Journal of Medicinal Chemistry. 2002;45:529-532
  184. 184. Buchanan MS, Carroll AR, Addepalli R, Avery VM, Hooper JN, Quinn RJ. Natural products, stylissadines A and B, specific antagonists of the P2X7 receptor, an important inflammatory target. The Journal of Organic Chemistry. 2007;72:2309-2317
  185. 185. Sakamoto H, Furukawa K-I, Matsunaga K, Nakamura H, Ohizumi Y. Xestoquinone activates skeletal muscle actomyosin ATPase by modification of the specific sulfhydryl group in the myosin head probably distinct from sulfhydryl groups SH1 and SH2. Biochemistry. 1995;34:12570-12575
  186. 186. Nakamura M, Kakuda T, Oba Y, Ojika M, Nakamura H. Synthesis of biotinylated xestoquinone that retains inhibitory activity against Ca2+ ATPase of skeletal muscle myosin. Bioorganic & Medicinal Chemistry. 2003;11:3077-3082
  187. 187. Alvi KA, Rodriguez J, Diaz MC, Moretti R, et al. Protein tyrosine kinase inhibitory properties of planar polycyclics obtained from the marine sponge Xestospongia cf. carbonaria and from total synthesis. The Journal of Organic Chemistry. 1993;58:4871-4880
  188. 188. Laurent D, Jullian V, Parenty A, Knibiehler M, et al. Antiplasmodial marine natural products in the perspective of current chemotherapy and prevention of malaria. A review. Marine Biotechnology. 2006;14:433-477
  189. 189. Oku N, Nagai K, Shindoh N, Terada Y, et al. Three new cyclostellettamines, which inhibit histone deacetylase, from a marine sponge of the genus Xestospongia. Bioorganic & Medicinal Chemistry Letters. 2004;14:2617-2620
  190. 190. Bourguet-Kondracki ML, Rakotoarisoa MT, Martin MT, Guyot M. Bioactive bromoacetylenes from marine sponge Xestospongia testudinaria. Tetrahedron Letters. 1992;33:225-226
  191. 191. Roll DM, Scheuer PJ, Matsumoto GK, Clardy J. Halenaquinone, a pentacyclic polyketide from a marine sponge. Journal of the American Chemical Society. 1983;105:6177-6178
  192. 192. Nakamura H, Kobayashi J, Kobayashi M, et al. Xestoquinone, a novel cardiotonic marine natural product isolated from the Okinawan sea sponge Xestospongia sapra. Chemistry Letters. 1985;(6):713-716
  193. 193. Kobayashi M, Shimizu N, Kitagawa I, Kyogoku Y, et al. Absolute stereostructures of halenaquinol and halenaquinol sulfate, pentacyclic hydroquinones from the okinawan marine sponge xestospongia sapra, as determined by theoretical calculation of CD spectra. Tetrahedron Letters. 1985;26:3833-3836
  194. 194. Concepcion GP, Foderaro TA, Eldredge GS, Lobkovsky E, et al. Topoisomerase II-mediated DNA cleavage by adocia- and xestoquinones from the Philippine sponge Xestospongia sp. Journal of Medicinal Chemistry. 1995;38:4503-4507
  195. 195. Cao S, Foster C, Brisson M, Lazo JS, KDG. Halenaquinone and xestoquinone derivatives, inhibitors of Cdc25B phosphatase from a Xestospongia sp. Bioorganic & Medicinal Chemistry. 2005;13:999-1003
  196. 196. Gunasekera SP, Cranick S, Longley RE. Immunosuppressive compounds from a deep water marine sponge, Agelas flabelli. Journal of Natural Products. 1989;52:757-761
  197. 197. Chairman K, Jeyamala M, Sankar S, Murugan A, Singh R. Immunomodulating properties of bioactive compounds present in Aurora globostellata. International Journal of Marine Science. 2013;3:151-157
  198. 198. Schumacher M, Cerella C, Eifes S, Chateauvieux S, et al. Heteronemin, a spongean sesterterpene, inhibits TNF alpha-induced NF-kappa B activation through proteasome inhibition and induces apoptotic cell death. Biochemical Pharmacology. 2010;79:610-622
  199. 199. Shin BA, Kim YR, Lee IS, Sung CK, et al. Lyso-PAF analogues. and lysophosphatidylcholines from the marine sponge Spirastrella abata as inhibitors of cholesterol biosynthesis. Journal of Natural Products. 1999;62:1554-1557
  200. 200. Zhao Q, Mansoor TA, Hong J, Lee CO, et al. New lysophosphatidylcholines and monoglycerides from the marine sponge Stelletta sp. Journal of Natural Products. 2003;66:725-728
  201. 201. Shen S, Liu D, Wei C, Proksch P, Lin W. Purpuroines A–J, halogenated alkaloids from the sponge Iotrochota purpurea with antibiotic activity and regulation of tyrosine kinases. Bioorganic & Medicinal Chemistry. 2012;20:6924-6928
  202. 202. Lee Y, Jang KH, Jeon JE, Yang WY, et al. Cyclic bis-1,3-dialkylpyridiniums from the Sponge Haliclona sp. Marine Drugs. 2012;10:2126-2137
  203. 203. Yang F, Hamann MT, Zou Y, Zhang MY, et al. Antimicrobial metabolites from the Paracel Islands sponge Agelas mauritiana. Journal of Natural Products. 2012;75:774-778
  204. 204. Laport MS, Marinho PR, Santos OC, de Almeida P, et al. Antimicrobial activity of marine sponges against coagulase-negative staphylococci isolated from bovine mastitis. Veterinary Microbiology. 2012;155:362-368
  205. 205. Fusetani N. Biofouling and antifouling. Natural Product Reports. 2004;21:94-104
  206. 206. Blihoghe D, Manzo E, Villela A, Cutignano A, et al. Evaluation of the antifouling properties of 3-alyklpyridine compounds. Biofouling: Journal of Bioadhesion and Biofilm Research. 2010;27:90-109

Written By

Musarat Amina and Nawal M. Al Musayeib

Submitted: 04 June 2017 Reviewed: 05 January 2018 Published: 25 April 2018