Next Article in Journal
Pyranoxanthones from Mesua ferrea
Previous Article in Journal
Synthesis and Antifungal Activities of Some Novel Pyrimidine Derivatives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Survey of Marine Natural Compounds and Their Derivatives with Anti-Cancer Activity Reported in 2010

Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Fondation de Recherche Cancer et Sang, Hôpital Kirchberg, 9, rue Steichen L-2540, Luxembourg
*
Author to whom correspondence should be addressed.
Molecules 2011, 16(7), 5629-5646; https://doi.org/10.3390/molecules16075629
Submission received: 11 May 2011 / Revised: 31 May 2011 / Accepted: 31 May 2011 / Published: 30 June 2011

Abstract

:
Although considerable progress in oncology therapeutics has been achieved in the last century, cancer remains one of major death causes in the World and for this reason, the development of novel cancer drugs remains a pressing need. Natural marine compounds represent an interesting source of novel leads with potent chemotherapeutic or chemo-preventive activities. In the last decades, structure-activity-relationship studies have led to the development of naturally-derived or semi-synthetic analogues with improved bioactivity, a simplified synthetic target or less toxicity. We aim here to review a selection of natural compounds with reported anticancer activity isolated of marine sources and their associated analogues published in 2010.

1. Introduction

Despite the continuous and important advances in biomedical research, the World Health Organization predicts that there will be more then 11 million cancer-related deaths per annum by 2030 [1]. Recent research highlights the isolation of promising compounds with effective anticancer activities from natural sources. An example of these compounds is trabectedin (PharmaMar’s Yondelis®) [2], which represents the first anticancer drug isolated from a marine source. Almost 50 percent of the antitumor agents approved over the last 50 years have consisted of compounds either derived from natural sources or (hemi-) synthetic analogues of these products [3]. Natural compounds remain a rich source of promising chemotherapeutic or chemo-preventive agents [4,5,6,7,8].
The sea covers over seventy percent of the Earth’s surface, and ecosystems such as coral reefs contain high levels of biodiversity compared to rainforests. The sea contains many untapped sources of drugs with promising activities due to the extensive variety of marine habitats (influenced by factors such as UV-exposure, the presence of sunlight, and salinity levels) [9]. Over 2700 scientists from over 80 nations, who assessed the diversity, distribution and abundance of marine life, conducted a marine census. The census resulted in the discovery of over 6000 potentially new species [10,11,12,13]. As a consequence of this research effort, it is clear that the marine environment represents a largely unexploited reservoir of unknown natural compounds, which need to be evaluated for potential medicinal applications.
Natural derivatives of potent bioactive compounds from marine organisms can be bio-synthesized. It is well known that in some cases, like that of the plant-derived polyphenol curcumin, the synthetic analog exerts a higher activity compared to the parent compound [14,15]. In addition to natural analogs, chemical modification is an extensive and exceptionally powerful tool for the development of novel drug candidates [16]. Structure-activity-relationship studies of marine compounds can lead to the design of analogs that have greater activity together with a simplified synthetic approach, as reported with bryostatin 1, a compound produced by the marine bryozoan Bugula neritina, which has been studied for several years by the Wender research group [17,18].
Many reviews on natural compounds from marine environments have been published [6,19,20,21,22,23,24,25]. Here, however, we will focus only on selected marine anti-cancer agents and analogues either discovered or synthesized in 2010 or whose biological activity was discussed in that year.

2. Marine Natural Compounds and Their Derivatives Published in 2010

2.1. Cryptosphaerolide (1)

Fenical et al. isolated cryptosphaerolide (1, Figure 1), an ester-substituted sesquiterpenoid, in 2010 from the ascomycete strain CNL-523 (Cryptosphaeria sp.) [26]. This marine product exerts cytotoxicity (IC50 of 4.5 μM) on the HCT-116 colon carcinoma cell line. A biochemical study revealed that this compound inhibited myeloid leukemia cell differentiation protein Mcl-1, a critical player involved in life/death decisions of individual cells [27], with an IC50 of 11.4 μM. Studies on a hydrolyzed analog of this compound demonstrated that the presence of a hydroxylated ester side chain, linked to the core sesquiterpenoid group, is responsible for the observed anti-cancer activity [26] (Figure 2).

2.2. Manzamine A (2)

The alkaloid manzamine A (2, Figure 1), which has been isolated from various marine sponges, exerted a cytotoxic effect against AsPC-1 pancreatic cancer cells, with an IC50 in a range of 4.2 μM, after 3 days of treatment [28]. Additionally, it was reported that manzamine A inhibited AsPC-1pancreatic cancer cell migration in vitro, and it decreased their overall metastatic potential. Fluorescent microscopy after staining with annexin V indicated an onset of apoptosis.
Figure 1. Chemical structures of marine compounds 1–31.
Figure 1. Chemical structures of marine compounds 1–31.
Molecules 16 05629 g001
Figure 2. Summary of structure-activity relationship (SAR) results of the marine compound cyptosphaerolide (1) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Figure 2. Summary of structure-activity relationship (SAR) results of the marine compound cyptosphaerolide (1) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Molecules 16 05629 g002
Wright’s group reported the antimetastatic and proapoptotic effects of this alkaloid. Consequently, manzamine A can be used in combination of therapies, as it sensitizes cancer cells to TRAIL-induced apoptosis, an effect resulting from the inhibition of glycogen synthase kinase GSK3β [28]. Structure-activity-relationship studies revealed that neither the additional hydroxy group at position 8, nor a small variation in the amine group excessively altered the activity of the compound (Figure 3). Instead, the double bond between positions 15 and 16 was found to be crucial for the observed activity, as described by Hamann et al., who indicated that the phenyl group of the heterocycle carboline group fits well into a pocket of this enzyme. Additionally, manzamine A inhibited cyclin-dependent kinase (CDK) 5, which decreased tau hyperphosphorylation in human neuroblastoma cells [29].
Figure 3. Summary of structure-activity relationship (SAR) results of the marine compound manzamine A (2) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Figure 3. Summary of structure-activity relationship (SAR) results of the marine compound manzamine A (2) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Molecules 16 05629 g003

2.3. Cholestanes (3)

Polyoxygenated cholestanes 3 (Figure 1) were isolated from the sea whip Leptogorgia sarmentosa. Four of these steroids exhibited cytotoxic activity against mouse lymphoid neoplasma (P-388), human lung carcinoma (A 549), human colon carcinoma (HT-29) and human melanoma (MEL 28) cells, with an ED50 in the range of 1 μg/mL [30].
Because this potent bioactive product can only be obtained in small quantities from marine sources, Kongkathip’s research group developed a method to produce these cholestanes synthetically. These steroids have an α,β-unsaturated ketone that exhibited significant cytotoxicity activity against human lung cancer cell lines (NCI) (IC50 of 6.2–10.5 μM), moderate activity against MCF7 breast cancer cell lines (IC50 of 30.7–31.4 μM) and human oral cancer KB (IC50 of 41.7–42.2 μM) cell lines [31]. Updated structure-activity-relationship studies have been recently published (Figure 4) and Kongkathip et al. reported that the hydroxyl groups at the C-3 and C-16 positions, as well as the cholesterol-like side chain, are crucial for the cytotoxic activity observed with these four steroids. Interestingly, a steroid with an aromatic A-ring exhibited the most potent cytotoxic effects, greater than that of the natural derivatives, against MCF7 and KB cell lines [32]. The biological pathway affected by this group of steroids remains unidentified.
Figure 4. Summary of structure-activity relationship (SAR) results of the marine compounds cholestanes (3) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Figure 4. Summary of structure-activity relationship (SAR) results of the marine compounds cholestanes (3) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Molecules 16 05629 g004

2.4. epi-Malyngamide C (4)

Malyngamide C, a chlorinated amide derivative of lyngbic acid, was first isolated by Moor’s research team in 1985, from the cyanobacterium Lynbya majuscula [33]. In 2010, an epi-isomer of malyngamide C (4) was isolated and identified. Kwan et al. evaluated the bioactivity of this new stereoisomer and compared it to the activity of the native compound. The epi-isomer was less cytotoxic to HT-29 colon cancer cells [IC50 values of 5.2 and 15.4 (epi-isomer), respectively] [34]. The R-configuration of the alcohol group on the six-membered cyclic ketone ring is crucial for cytotoxic activity (Figure 5). To date, no studies on the signaling pathway used by malyngamides have been published.
Figure 5. Summary of structure-activity relationship (SAR) results of the marine compound 8-epi-malyngamide C (4) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Figure 5. Summary of structure-activity relationship (SAR) results of the marine compound 8-epi-malyngamide C (4) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Molecules 16 05629 g005

2.5. HESA-A, a Drug from Herbal-Marine Origin

HESA-A (patented by researchers in Iran) is composed of both plant and marine materials, including material from Penaeus latisculatus (king prawn), Carum carvi (Persian cumin) and Apiumgraveolens (celery). HESA-A consists of both mineral and organic constituents and a small amount of water (45%, 50% and 5%, respectively) [35]. The exact biological targets of HESA-A have not been determined to date, but it is thought that this multi-component drug acts using a variety of pharmacological mechanisms [36]. Its efficiency as a non-toxic, chemotherapeutic agent has been confirmed recently in various in vivo and pre-clinical studies [35,37,38,39,40].

2.6. Spongistatin 1 (5)

The Pettit group isolated the macrocyclic lactone spongistatin 1 (5) from a marine sponge of the genus Spongia in 1993; this marine lactone exerted strong cytotoxicity on a panel of 60 types of human cancer cells [41]. Spongistatin 1 was reported to inhibit glutamate-induced tubulin polymerization (IC50 of 3.6 μM in PtK1 kangaroo rat kidney cells) through its interaction with the Vinca alkaloid domain of tubulin, which leads to the inhibition of mitosis [42].
After treatment of A549 lung cancer cells with spongistatin 1 (1 nM), cell cycle arrest at the G2-M phase, the simultaneous up-regulation of GADD45α-γ and down-regulation of c-Myc were observed [43]. Various studies have reported that this marine product triggers caspase-dependent apoptosis in leukemia cells, even in primary leukemia cell lines, at low concentrations (1 nM) [43,44,45].
This compound did not induce significant apoptosis in healthy peripheral blood cells, highlighting its potential use as a therapeutic drug [44]. Several structure-activity-relationship studies have been conducted to date (Figure 6); Kishi et al. noted that a C-23 epimer and spongistatin 1 had similar cytotoxic effects [46]. Paterson et al. reported that dehydration of the E-ring (C35-C36) led to an increase in cytotoxic potency, but that altering the side chain resulted in an important loss of activity [47]. A hydrogen-chlorine substitution in spongistatin 1 resulted in a 10-fold reduction in cytotoxicity [41]. In 2008, Heathcock et al. evaluated the toxicity of acyclic spongistatin 2 analogs, which contained only the E- and F-rings, as well as cyclic EF, ABEF and ABCD ring derivatives. In all derivatives tested, cytotoxicity was lost [48]. More recently, Smith et al. noted that an ABEF analog had cytotoxic effects when used in the nanomolar range, although its potency was 1,000 times weaker than that of spongistatin 1 [49]. In summary, these results demonstrate that the ABEF ring system, as well as the triene side chain, are crucial for spongistatin 1 cytotoxicity.

2.7. Bromopyrrole Akaloids

Marine sponges from the genera Agelas, Axinella and Hymeniacidon are known to synthesize bromopyrrole alkaloids [50,51]. In 1990, various compounds from a similar chemical class, namely hymenialdisine (6), debromo-hymeniaidisine (7) and agelasine G (8) were shown to exert significant cytotoxic activity against murine lymphoma cells (ED50 of 2.0–3.1 μg/mL). In contrast to these compounds, axinohydantoin (9) was significantly less active (IC50 of 18 μg/mL) [51,52]. Additionally, dibromophakellstatin (10) exhibited cytotoxicity against various human cancer cells at sub-micromolar concentrations, but replacement of the urea group with a guanidine resulted in a decrease in activity [53]. In 2010, Xu et al. demonstrated that bromopyrrole alkaloids have anti-cancer activity in vivo. This was observed with a novel bromopyrrole, N-(4,5-dibromopyrrole-2-carbonyl)-L-aminoisovaleric acid methyl ester (11). This compound inhibited the proliferation of human cancer cells in vitro (IC50 of 3.8–17.2 μg/mL) and in xenografted mice (MIC at 40 mg/kg). It has also been reported that bromopyrrole triggered cell cycle arrest in the G1 phase, and that it induced caspase-dependent apoptosis [54]. Another compound, sceptrin (12), composed of two bromopyrroles bound to a cyclobutane unit, halted cell motility in a variety of cancer cells (at 40 μM) [55] but had no effect on cell proliferation or survival [55]. It has been reported to be noncytotoxic to monkey kidney cells (at 200 μg/disk) [56]. The three sceptrin derivatives, namely nakamuric acid (13), its methyl ester derivative and debromosceptrin, exhibited lower anti-motility effects compared to sceptrin [55]. However, detailed mechanistic studies on the pathways affected by sceptrin still need to be conducted.
Figure 6. Summary of structure-activity relationship (SAR) results of the marine compound spongistatin 1 (5) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Figure 6. Summary of structure-activity relationship (SAR) results of the marine compound spongistatin 1 (5) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Molecules 16 05629 g006

2.8. β-Carbolines

Crews et al. isolated a novel β-carboline, named hyrtiocarboline (14), from the marine sponge Hyrtios reticulates. This β-carboline exhibited antiproliferative activity when applied to a panel of 13 cancer cell types, with an IC50 as low as 1.2 μg/mL for non-small cell lung cancer (H522-T1) cells [57]. The core structure of β-carboline may represent a promising chemical class, as reported in several publications [58,59]. To date, a few β-carboline alkaloids have been isolated from marine sources, namely 5-bromo-8-methoxy-1-methyl-β-carboline (15) [60], norharman (16) [61], 2-methyl-eudistomins-J-D (1718), and 14-methyleudistomin C (19) [62]. Only the latter had cytotoxic effects in sub-micromolar range. Further investigations on the affected pathways need to be conducted.

2.9. Makaluvamine Analogs

Makaluvamines (20) (a type of pyrroloiminoquinone) were first isolated from marine sponges belonging to the genera Zyzzya and Histodermella [63,64]. These products exert cytotoxic activity against various cell lines by inhibiting DNA topoisomerase II [65]. Anti-cancer activity was observed with bioactive synthetic analogs of these compounds. Imidazoquinoxalinone derivatives 21 have been reported to be less active due to the presence of an electro-deficient benzimidazole ring; the naturally occurring compound contains an indole ring. Under physiological conditions, the cationic makaluvamines were reported to be active, whereas imidazoquinoxalinones analogs are not charged, which explains the decreased activity of the latter [66,67]. Velu et al. evaluated the biological activity of makaluvamines containing various substitutions at the 7-position of the pyrroloiminoquinone ring. Observations from many natural derivatives allowed them to conclude that the presence of functional groups at this position greatly increased cytotoxic potential (Figure 7). In vitro testing with a NCI panel of 60 human cancer cell types indicated that the 7-benzyl- and 7-(4-fluorobenzyl) analogs (BA-TPQ and FBA-TPQ, respectively) exhibited the greatest cytotoxic effects [68,69]. These promising results lead various groups to perform pre-clinical studies. In breast cancer cell lines, both products significantly decreased cancer cell growth, induced apoptosis and caused cell cycle arrest at submicromolar concentrations (0.5 μM). Additionally, it has been reported that the anticancer activity was independent from the activity of p53 in cancer cells [70,71]. FBA-TPQ also strongly inhibited cancer cell proliferation, activated apoptosis and caused cell cycle arrest in prostate cancer cells, in the low micromolar range (2 μM). Furthermore, androgen receptor (AR) and prostate-specific antigen (PSA) levels, as well as the expression of apoptosis-related proteins were reduced [72]. A pharmacological study conducted in mice revealed that intravenously-injected BA-TPQ accumulated in the lungs, kidneys and spleen; it even reached low concentrations in the brain. However, this product was systemically toxic (indicated by animal weight loss) when administered at a concentration of 10 mg/kg [73]. The data presented here clearly indicated that makaluvamine analogs represent a promising choice for future clinical trails, and they may promote the development of novel anticancer drugs.
Figure 7. Summary of structure-activity relationship (SAR) results of the marine compounds makaluvamines (20) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Figure 7. Summary of structure-activity relationship (SAR) results of the marine compounds makaluvamines (20) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Molecules 16 05629 g007

2.10. Cyclopentenones

In 1977, cyclopentenones were shown to possess antitumor properties and to down-regulate cellular metabolism [74,75]. It is has been noted that prostaglandins A1, A2 and J2 (compounds 2224), which are known cancer-proliferation inhibitors, carry an a,b-unsaturated cyclopentenone ring linked to alkyl chains [76]. It is not surprising that the bioactivity of prostaglandins is mainly due to the presence of the cyclopentenone ring. An α,β-unsaturated ketone group is thought to act as an important alkylating center through a Michael-type reaction with cysteine [74,75,76]. Detailed mechanistic studies indicated that cyclopentenone (25) caused cell cycle arrest by the repression of cyclin D1, inhibited constitutive NF-kB activity and lead to the induction of apoptosis [77,78]. Novel cytotoxic cyclopentenones, namely didemnenone (26) and trichoderone (27), have been isolated from the didemnid ascidian Lissoclinum sp., and from the marine-derived fungus Trichoderma sp. [79,80]. In both cases, the IC50 was in the micromolar range. These findings highlight the anticancer potential of cyclopentenone groups, making them interesting compounds on which to focus future studies.

2.11. Heteronemin and Semi-Synthetic Derivatives

The pentacyclic scalarane heteronemin (28) was first isolated in large quantities from the sponges Heteronema erecta and Hytios sp. in 1976 [81]. Crews et al. reported heteronemin had cytotoxic effects when applied to brine shrimp and giant kelp (Macrocystis pyrifera) gametes [82]. In cytotoxicity assays, this sesterterpene induced cell death in human thyroid carcinoma cells and an analog, 12-deacetoxy-21-hydroxyheteronemin, exhibited significant cytotoxicity against K562 cells [83,84]. The biological pathways affected by this marine product have since been identified. It has also been shown that heteronemin exerts antitubercular activity by inhibiting farnesyl transferase [85,86]. Furthermore, results from our laboratory have clearly shown that heteronemin attenuates NF-κB pathway activation through the down-regulation of proteasome activity [87]. Heteronemin triggered caspase-dependent apoptosis in K562 cells, and it sensitized K562 cells to TNFα-induced apoptosis [87].
Two structure-activity-relationship studies were published in 2009, with focus on the anti-carcinogenic effects induced by this compound [88,89] (Figure 8). Despite the fact that half of the tested analogs showed little or no cytotoxicity towards normal human oral fibroblasts or monkey kidney epithelial cells, these studies demonstrated that the oxygen atoms at positions C-25 and C-16 were crucial for the cytotoxic activity of heteronemin, whereas the double bond at position C-17-C-24 was of marginal importance [88,89]. To summarize, these promising results indicate that heteronemin and some of its derivatives represent interesting candidates for future chemotherapeutic drug research.

2.12. Latrunculin A and B

Latrunculins A and B (compounds 29,30) were first isolated from the Red Sea sponge Negombata magnifica [90,91]. The core structure of both compounds consists of a macrolide fused to a tetrahydropyran moiety, where the latter is linked to a 2-thiazolidinone side chain. These were the first marine natural products reported to bind reversibly to actin, leading to its disorganization [91]. These compounds also exerted potent activity on the angiogenesis, migration and proliferation of cells [92,93]. Latrunculin A has a therapeutic index (T/C) of 146% in mice [94], a remarkable result, considering the actin-active agents jaspamide and cucurbitacin did not have comparable therapeutic indices in case study conducted by scientists in the Developmental Therapeutics Program (DTP) at the NCI [95,96]. X-ray crystallography demonstrated that 2-thiazolidinone fits perfectly into actin pockets, and each polar oxygen, except for the O2-ester, forms a hydrogen bond with actin [97].
Figure 8. Summary of structure-activity relationship (SAR) results of the marine compound heteronemin (28) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Figure 8. Summary of structure-activity relationship (SAR) results of the marine compound heteronemin (28) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Molecules 16 05629 g008
Several structure-activity-relationship studies have been completed [92,98,99,100,101]. The deletion of methyl groups from the macrocycle group of latrunculin B resulted in an increased activity and a simplified synthetic target [99]. The composition of latrunculin A’s 16-membered macrolide, as well as the conformation of its thiazolidinone ring play critical roles in its anticancer activities, as observed with human solid cancer cell lines HCT-116 and MDA-MB-435 [98]. Carbamate derivatives exerted 2.5- to 5-fold greater anti-invasive activity against the extremely metastatic human prostate PC-3M cancer cells, with lower actin binding properties [101] (Figure 9). El Sayed et al. demonstrated that both 17-O-phenylethyl- and N-hydroxymethyl-analogs of latrunculin A had higher activity than the parent product [100].

2.13. Dermacozines

Dermacozines (31), phenazine-type pigments, have been isolated from marine actinomycetes isolated from Mariana Trench sediment from a depth of 10.898 meters by Jaspar’s research group [102]. This novel class of phenazines has been characterized and confirmed through in-depth analysis of 1D-,2D-NMR data combined to high-resolution MS, UV-data and CD spectroscopy. Hence these marine products exerted a cytotoxicity activity versus leukemic K562 cancer cells with an IC50-range from 7 to 220 mM. A structure-activity-relationship study showed that a carboxamide moiety nor a lactone ring or a benzyl function majorly affected the observed cytotoxicity of the products (Figure 10). However, an additional carboxylic anhydride linked to the phenazine core structure led to a 20-fold increase in observed cytotoxicity in contrast to an imide ring. The latter did not alter the observed activity [102]. Further mechanistic studies on the pathways affected by dermacozines need to be conducted.
Figure 9. Summary of structure-activity relationship (SAR) results of the marine compound latrunculin A (29) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Figure 9. Summary of structure-activity relationship (SAR) results of the marine compound latrunculin A (29) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Molecules 16 05629 g009
Figure 10. Summary of structure-activity relationship (SAR) results of the marine compounds dermacozines (31) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Figure 10. Summary of structure-activity relationship (SAR) results of the marine compounds dermacozines (31) concerning the observed anti-cancer cytotoxicity. The pharmacophores identified during the studies are highlighted in color (X: inactive; Molecules 16 05629 i001, =, Molecules 16 05629 i002: higher, similar, lower cytotoxicity).
Molecules 16 05629 g010

4. Conclusions

This review provides insight into the current literature regarding marine natural products and their derivatives which was published in 2010. The data presented here indicate the great value of natural marine products, as well as their synthetic analogs. The data suggest that these synthetic analogs, in particular, could be important candidates for further studies involving structural modifications to improve the pharmacological profile of native marine metabolites. Furthermore, a simplified analog with equipotent activity can lead to the development of a simplistic synthesis process, which would guarantee a sufficient supply if bioactive products for further investigation. In conclusion, the isolation or modification of novel marine products, as well as their analogs, and the subsequent evaluation of their bioactivity will push the discovery of novel promising chemotherapeutic drugs forward.

Acknowledgments

Research in MD’s lab is supported by the “Recherche Cancer et Sang”, the “Recherches Scientifiques Luxembourg” association, the “Een Häerz fir kriibskrank Kanner” association, the Action Lions “Vaincre le Cancer” association and by Télévie Luxembourg. MK is supported by an AFR postdoctoral fellowship from the Fonds National de la Recherche Luxembourg. Editing and publication costs are covered by the Fonds National de la Recherche Luxembourg.

Conflict of Interest

The authors declare no conflict of interest.

References and Notes

  1. WHO. Fact Sheet N°297. Available online: http://www.who.int/mediacentre/factsheets/fs297/en/ (accessed on 20 March 2011).
  2. D'Incalci, M.; Galmarini, C.M. A review of trabectedin (ET-743): A unique mechanism of action. Mol. Cancer Ther. 2010, 9, 2157–2163. [Google Scholar] [CrossRef]
  3. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the last 25 years. J. Nat. Prod. 2007, 70, 461–477. [Google Scholar] [CrossRef]
  4. Dumontet, C.; Jordan, M.A. Microtubule-binding agents: A dynamic field of cancer therapeutics. Nat. Rev. Drug Discov. 2010, 9, 790–803. [Google Scholar] [CrossRef]
  5. Gullett, N.P.; Ruhul Amin, A.R.; Bayraktar, S.; Pezzuto, J.M.; Shin, D.M.; Khuri, F.R.; Aggarwal, B.B.; Surh, Y.J.; Kucuk, O. Cancer prevention with natural compounds. Semin. Oncol. 2010, 37, 258–281. [Google Scholar] [CrossRef]
  6. Villa, F.A.; Gerwick, L. Marine natural product drug discovery: Leads for treatment of inflammation, cancer, infections, and neurological disorders. Immunopharmacol. Immunotoxicol. 2010, 32, 228–237. [Google Scholar] [CrossRef]
  7. Juncker, T.; Cerella, C.; Teiten, M.H.; Morceau, F.; Schumacher, M.; Ghelfi, J.; Gaascht, F.; Schnekenburger, M.; Henry, E.; Dicato, M.; et al. NBS1450, a steroid cardiac glycoside inducing apoptotic cell death in human leukemia cells. Biochem. Pharmacol. 2011, 81, 13–23. [Google Scholar]
  8. Folmer, F.; Jaspars, M.; Dicato, M.; Diederich, M. Photosynthetic marine organisms as a source of anticancer compounds. Phytochem. Rev. 2010, 9, 557–479. [Google Scholar] [CrossRef]
  9. Scheuer, P.J. Some marine ecological phenomena: Chemical basis and biomedical potential. Science 1990, 248, 173–177. [Google Scholar]
  10. Miloslavich, P.; Diaz, J.M.; Klein, E.; Alvarado, J.J.; Diaz, C.; Gobin, J.; Escobar-Briones, E.; Cruz-Motta, J.J.; Weil, E.; Cortes, J.; et al. Marine biodiversity in the Caribbean: Regional estimates and distribution patterns. PLoS One 2010, 5, e11916. [Google Scholar]
  11. Fautin, D.; Dalton, P.; Incze, L.S.; Leong, J.A.; Pautzke, C.; Rosenberg, A.; Sandifer, P.; Sedberry, G.; Tunnell, J.W., Jr.; Abbott, I.; et al. An overview of marine biodiversity in United States waters. PLoS One 2010, 5, e11914. [Google Scholar]
  12. Butler, A.J.; Rees, T.; Beesley, P.; Bax, N.J. Marine biodiversity in the Australian region. PLoS One 2010, 5, e11831. [Google Scholar]
  13. Census of marine life. Available online: http://www.coml.org/about-census/ (accessed on 20 March 2011).
  14. Anand, P.; Thomas, S.G.; Kunnumakkara, A.B.; Sundaram, C.; Harikumar, K.B.; Sung, B.; Tharakan, S.T.; Misra, K.; Priyadarsini, I.K.; Rajasekharan, K.N.; et al. Biological activities of curcumin and its analogues (Congeners) made by man and Mother Nature. Biochem. Pharmacol. 2008, 76, 1590–1611. [Google Scholar] [CrossRef]
  15. Yadav, B.; Taurin, S.; Rosengren, R.J.; Schumacher, M.; Diederich, M.; Somers-Edgar, T.J.; Larsen, L. Synthesis and cytotoxic potential of heterocyclic cyclohexanone analogues of curcumin. Bioorg. Med. Chem. 2010, 18, 6701–6707. [Google Scholar]
  16. Floss, H.G. Combinatorial biosynthesis—Potential and problems. J. Biotechnol. 2006, 124, 242–257. [Google Scholar] [CrossRef]
  17. Wender, P.A.; Baryza, J.L.; Brenner, S.E.; Dechristopher, B.A.; Loy, B.A.; Schrier, A.J.; Verma, V.A. Organic synthesis toward small-molecule probes and drugs special feature: Design, synthesis, and evaluation of potent bryostatin analogs that modulate PKC translocation selectivity. Proc. Natl. Acad. Sci. USA 2011, 108, 6721–6726. [Google Scholar]
  18. Wender, P.A.; Miller, B.L. Synthesis at the molecular frontier. Nature 2009, 460, 197–201. [Google Scholar]
  19. Penesyan, A.; Kjelleberg, S.; Egan, S. Development of novel drugs from marine surface associated microorganisms. Mar. Drugs 2010, 8, 438–459. [Google Scholar] [CrossRef]
  20. Ebada, S.S.; Lin, W.; Proksch, P. Bioactive sesterterpenes and triterpenes from marine sponges: Occurrence and pharmacological significance. Mar. Drugs 2010, 8, 313–346. [Google Scholar] [CrossRef]
  21. Lin, X.; Liu, M.; Hu, C.; Liao, D.J. Targeting cellular proapoptotic molecules for developing anticancer agents from marine sources. Curr. Drug Targets 2010, 11, 708–715. [Google Scholar] [CrossRef]
  22. Blunt, J.W.; Copp, B.R.; Munro, M.H.; Northcote, P.T.; Prinsep, M.R. Marine natural products. Nat. Prod. Rep. 2010, 27, 165–237. [Google Scholar] [CrossRef]
  23. Schumacher, M.; Kelkel, M.; Dicato, M.; Diederich, M. Gold from the sea: Marine compounds as inhibitors of the hallmarks of cancer. Biotechnol. Adv. 2011. [Google Scholar] [CrossRef]
  24. Folmer, F.; Jaspars, M.; Schumacher, M.; Dicato, M.; Diederich, M. Marine natural products targeting phospholipases A2. Biochem. Pharmacol. 2010, 80, 1793–1800. [Google Scholar] [CrossRef]
  25. Folmer, F.; Jaspars, M.; Dicato, M.; Diederich, M. Marine natural products as targeted modulators of the transcription factor NF-kappaB. Biochem. Pharmacol. 2008, 75, 603–617. [Google Scholar] [CrossRef]
  26. Oh, H.; Jensen, P.R.; Murphy, B.T.; Fiorilla, C.; Sullivan, J.F.; Ramsey, T.; Fenical, W. Cryptosphaerolide, a cytotoxic Mcl-1 inhibitor from a marine-derived ascomycete related to the genus Cryptosphaeria. J. Nat. Prod. 2010, 73, 998–1001. [Google Scholar] [CrossRef]
  27. Michels, J.; Johnson, P.W.; Packham, G. Mcl-1. Int. J. Biochem. Cell Biol. 2005, 37, 267–271. [Google Scholar] [CrossRef]
  28. Guzman, E.A.; Johnson, J.D.; Linley, P.A.; Gunasekera, S.E.; Wright, A.E. A novel activity from an old compound: Manzamine A reduces the metastatic potential of AsPC-1 pancreatic cancer cells and sensitizes them to TRAIL-induced apoptosis. Invest. New Drugs 2010. Epub ahead of print. [Google Scholar]
  29. Hamann, M.; Alonso, D.; Martin-Aparicio, E.; Fuertes, A.; Perez-Puerto, M.J.; Castro, A.; Morales, S.; Navarro, M.L.; Del Monte-Millan, M.; Medina, M.; et al. Glycogen synthase kinase-3 (GSK-3) inhibitory activity and structure-activity relationship (SAR) studies of the manzamine alkaloids. Potential for Alzheimer's disease. J. Nat. Prod. 2007, 70, 1397–1405. [Google Scholar] [CrossRef]
  30. Garrido, L.; Zubia, E.; Ortega, M.J.; Salva, J. Isolation and structure elucidation of new cytotoxic steroids from the gorgonian Leptogorgia sarmentosa. Steroids 2000, 65, 85–88. [Google Scholar] [CrossRef]
  31. Boonananwong, S.; Kongkathip, B.; Kongkathip, N. First synthesis of 3,16,20-polyoxygenated cholestanes, new cytotoxic steroids from the gorgonian Leptogorgia sarmentosa. Steroids 2008, 73, 1123–1127. [Google Scholar] [CrossRef]
  32. Bunyathaworn, P.; Boonananwong, S.; Kongkathip, B.; Kongkathip, N. Further study on synthesis and evaluation of 3,16,20-polyoxygenated steroids of marine origin and their analogs as potent cytotoxic agents. Steroids 2010, 75, 432–444. [Google Scholar] [CrossRef]
  33. Ainnslie, R.D.; Barchi, J.J.; Kuniyoshi, M.; Moore, R.E.; Myndersee, J.S. Structure of Malyngamide C. J. Org. Chem. 1985, 50, 2859–2862. [Google Scholar] [CrossRef]
  34. Kwan, J.C.; Teplitski, M.; Gunasekera, S.P.; Paul, V.J.; Luesch, H. Isolation and biological evaluation of 8-epi-malyngamide C from the Floridian marine cyanobacterium Lyngbya majuscula. J. Nat. Prod. 2010, 73, 463–466. [Google Scholar] [CrossRef]
  35. Ahmadi, A.; Mohagheghi, M.; Karimi, M.; Golestanha, S.A.; Naseri, M.; Faghihzadeh, S.; Habibi, G. Therapeutic effects of HESA-A in patients with end-stage metastatic cancers. Integr. Cancer Ther. 2010, 9, 32–35. [Google Scholar] [CrossRef]
  36. Ahmadi, A.; Naderi, G.; Asgary, S. Evaluation of hepatoprotective potential of HESA-A (a marine compound) pretreatment against thioacetamide-induced hepatic damage in rabbits. Drugs Exp. Clin. Res. 2005, 31, 1–6. [Google Scholar]
  37. Amrollah, A.; Gholamreza, H.; Mehdi, F. Anticancer effects of HESA-A: An herbal marine compound. Chin. J. Integr. Med. 2010, 16, 366–367. [Google Scholar] [CrossRef]
  38. Moallem, S.; Ahmadi, A.; Moshafi, M.; Taghavi, M. Teratogenic effects of HESA-A, a natural anticancer product from Iran, in mice. Hum. Exp. Toxicol. 2010. [Google Scholar] [CrossRef]
  39. Ahmadi, A.; Mohagheghi, M.; Karimi, M.; Seyed Ali, G.; Naseri, M. Anticancer effects of HESA-A in patients with metastatic colon cancer. Integr. Cancer Ther. 2009, 8, 71–74. [Google Scholar] [CrossRef]
  40. Ahmadi, A.; Ghanbari, H.; Soheilian, M.; Naseri, M. The EFFEct of HESA-A (natural drug) on visual acuity in age related macular degeneration: A randomized double blind controlled clinical trial. Afr. J. Tradit. Complement. Altern. Med. 2009, 6, 549–553. [Google Scholar]
  41. Pettit, G.R.; Cichacz, Z.A.; Gao, F.; Herald, C.L.; Boyd, M.R.; Schmidt, J.M.; Hooper, J.N. Isolation and structure of spongistatin 1. J. Org. Chem. 1993, 58, 1302–1304. [Google Scholar] [CrossRef]
  42. Bai, R.; Cichacz, Z.A.; Herald, C.L.; Pettit, G.R.; Hamel, E. Spongistatin 1, a highly cytotoxic, sponge-derived, marine natural product that inhibits mitosis, microtubule assembly, and the binding of vinblastine to tubulin. Mol. Pharmacol. 1993, 44, 757–766. [Google Scholar]
  43. Catassi, A.; Cesario, A.; Arzani, D.; Menichini, P.; Alama, A.; Bruzzo, C.; Imperatori, A.; Rotolo, N.; Granone, P.; Russo, P. Characterization of apoptosis induced by marine natural products in non small cell lung cancer A549 cells. Cell. Mol. Life Sci. 2006, 63, 2377–2386. [Google Scholar] [CrossRef]
  44. Schyschka, L.; Rudy, A.; Jeremias, I.; Barth, N.; Pettit, G.R.; Vollmar, A.M. Spongistatin 1: A new chemosensitizing marine compound that degrades XIAP. Leukemia 2008, 22, 1737–1745. [Google Scholar] [CrossRef]
  45. Schneiders, U.M.; Schyschka, L.; Rudy, A.; Vollmar, A.M. BH3-only proteins Mcl-1 and Bim as well as endonuclease G are targeted in spongistatin 1-induced apoptosis in breast cancer cells. Mol. Cancer Ther. 2009, 8, 2914–2925. [Google Scholar] [CrossRef]
  46. Hayward, M.M.; Roth, R.M.; Duffy, K.J.; Dalko, P.I.; Stevens, K.L.; Guo, J.; Kishi, Y. Total synthesis of altohyrtin A (Spongistatin 1): Part 2. Angew. Chem. Int. Ed. 1998, 37, 192–196. [Google Scholar]
  47. Paterson, I.; Chen, D.Y.K.; Coster, M.J.; Aceña, J.L.; Bach, J.; Wallace, D.J. The stereocontrolled total synthesis of altohyrtin A/spongistatin 1: Fragment couplings, completion of the synthesis, analogue generation and biological evaluation. Org. Biomol. Chem. 2005, 3, 2431–2440. [Google Scholar] [CrossRef]
  48. Wagner, C.E.; Wang, Q.; Melamed, A.; Fairchild, C.R.; Wild, R.; Heathcock, C.H. Synthesis and biological evaluation of analogs of altohyrtin C (spongistatin 2). Tetrahedron 2008, 64, 124–136. [Google Scholar]
  49. Smith, A.B. Design, synthesis, and biological evaluation of EF- and ABEF- analogues of (+)-spongistatin 1. Org. Lett. 2010, 12, 1792–1795. [Google Scholar] [CrossRef]
  50. Kobayashi, J.; Tsuda, M.; Murayama, T.; Nakamura, H.; Ohizumi, Y.; Ishibashi, M.; Iwamura, M.; Ohta, T.; Nozoe, S. Ageliferins, potent actomyosin Atpase activators from the Okinawan marine sponge Agelas sp. Tetrahedron 1990, 46, 5579–5586. [Google Scholar] [CrossRef]
  51. Pettit, G.R.; Herald, C.L.; Leet, J.E.; Gupta, R.; Schaufelberger, D.E.; Bates, R.B.; Clewlow, P.J.; Doubek, D.L.; Manfredi, K.P.; Rützler, K.; et al. ntineoplastic agents. Can. J. Chem. 1990, 68, 1621–1624. [Google Scholar] [CrossRef]
  52. Ishida, K.; Ishibashi, M.; Shigemori, H.; Sasaki, T.; Kobayashi, J. Agelasine G, a new antileukemic alkaloid from the Okinawan marine sponge Agelas sp. Chem. Pharm. Bull. (Tokyo) 1992, 40, 766–767. [Google Scholar] [CrossRef]
  53. Pettit, G.R.; McNulty, J.; Herald, D.L.; Doubek, D.L.; Chapuis, J.C.; Schmidt, J.M.; Tackett, L.P.; Boyd, M.R. Antineoplastic agents. 362. Isolation and X-ray crystal structure of dibromophakellstatin from the Indian ocean sponge Phakellia mauritiana. J. Nat. Prod. 1997, 60, 180–183. [Google Scholar] [CrossRef]
  54. Xiong, S.; Pang, H.D.; Fan, J.; Ge, F.; Yang, X.X.; Liu, Q.Y.; Liao, X.J.; Xu, S.H. In vitro and in vivo antineoplastic activity of a novel bromopyrrole and its potential mechanism of action. Br. J. Pharmacol. 2010, 159, 909–918. [Google Scholar] [CrossRef]
  55. Cipres, A.; O'Malley, D.P.; Li, K.; Finlay, D.; Baran, P.S.; Vuori, K. Sceptrin, a marine natural compound, inhibits cell motility in a variety of cancer cell lines. ACS Chem. Biol. 2010, 5, 195–202. [Google Scholar] [CrossRef]
  56. Keifer, P.A.; Schwartz, R.E.; Koker, M.E.S.; Hughes, R.G.J.; Rittschof, D.; Rinehart, K.L. Bioactive bromopyrrole metabolites from the Caribbean sponge Agelas conifera. J. Org. Chem. 1991, 56, 2965–2975. [Google Scholar]
  57. Inman, W.D.; Bray, W.M.; Gassner, N.C.; Lokey, R.S.; Tenney, K.; Shen, Y.Y.; Tendyke, K.; Suh, T.; Crews, P. A beta-carboline alkaloid from the Papua New Guinea marine sponge hyrtios reticulatus. J. Nat. Prod. 2010, 73, 255–257. [Google Scholar] [CrossRef]
  58. Daugan, A.; Grondin, P.; Ruault, C.; Le Monnier de Gouville, A.C.; Coste, H.; Linget, J.M.; Kirilovsky, J.; Hyafil, F.; Labaudiniere, R. The discovery of tadalafil: A novel and highly selective PDE5 inhibitor. 2: 2,3,6,7,12,12a-hexahydropyrazino[1',2':1,6]pyrido[3,4-b]indole-1,4-dione analogues. J. Med. Chem. 2003, 46, 4533–4542. [Google Scholar] [CrossRef]
  59. Trujillo, J.I.; Meyers, M.J.; Anderson, D.R.; Hegde, S.; Mahoney, M.W.; Vernier, W.F.; Buchler, I.P.; Wu, K.K.; Yang, S.; Hartmann, S.J.; et al. Novel tetrahydro-beta-carboline-1-carboxylic acids as inhibitors of mitogen activated protein kinase-activated protein kinase 2 (MK-2). Bioorg. Med. Chem. Lett. 2007, 17, 4657–4663. [Google Scholar]
  60. Till, M.; Prinsep, M.R. 5-Bromo-8-methoxy-1-methyl-beta-carboline, an alkaloid from the New Zealand marine bryozoan Pterocella vesiculosa. J. Nat. Prod. 2009, 72, 796–798. [Google Scholar] [CrossRef]
  61. Zheng, L.; Yan, X.; Han, X.; Chen, H.; Lin, W.; Lee, F.S.; Wang, X. Identification of norharman as the cytotoxic compound produced by the sponge (Hymeniacidon perleve)-associated marine bacterium Pseudoalteromonas piscicida and its apoptotic effect on cancer cells. Biotechnol. Appl. Biochem. 2006, 44, 135–142. [Google Scholar] [CrossRef]
  62. Rashid, M.A.; Gustafson, K.R.; Boyd, M.R. New cytotoxic N-methylated beta-carboline alkaloids from the marine ascidian Eudistoma gilboverde. J. Nat. Prod. 2001, 64, 1454–1456. [Google Scholar] [CrossRef]
  63. Carney, J.R.; Scheuer, P.J.; Kelly-Borges, M. Makaluvamine G, a cytotoxic pigment from an an Indonesian Sponge Histodermella sp. Tetrahedron 1993, 49, 8483–8486. [Google Scholar] [CrossRef]
  64. Radisky, D.C.; Radisky, E.S.; Barrows, L.R.; Copp, B.R.; Kramer, R.A.; Ireland, C.M. Novel cytotoxic topoisomerase II inhibiting pyrroloiminoquinones from Fijian sponges of the genus Zyzzya. J. Am. Chem. Soc. 1993, 115, 1632–1638. [Google Scholar] [CrossRef]
  65. Matsumoto, S.S.; Haughey, H.M.; Schmehl, D.M.; Venables, D.A.; Ireland, C.M.; Holden, J.A.; Barrows, L.R. Makaluvamines vary in ability to induce dose-dependent DNA cleavage via topoisomerase II interaction. Anticancer Drugs 1999, 10, 39–45. [Google Scholar]
  66. Hoang, H.; LaBarbera, D.V.; Mohammed, K.A.; Ireland, C.M.; Skibo, E.B. Synthesis and biological evaluation of imidazoquinoxalinones, imidazole analogues of pyrroloiminoquinone marine natural products. J. Med. Chem. 2007, 50, 4561–4571. [Google Scholar] [CrossRef]
  67. Labarbera, D.V.; Skibo, E.B. Synthesis of imidazo[1,5,4-de]quinoxalin-9-ones, benzimidazole analogues of pyrroloiminoquinone marine natural products. Bioorg. Med. Chem. 2005, 13, 387–395. [Google Scholar]
  68. Shinkre, B.A.; Raisch, K.P.; Fan, L.; Velu, S.E. Analogs of the marine alkaloid makaluvamines: Synthesis, topoisomerase II inhibition, and anticancer activity. Bioorg. Med. Chem. Lett. 2007, 17, 2890–2893. [Google Scholar] [CrossRef]
  69. Shinkre, B.A.; Raisch, K.P.; Fan, L.; Velu, S.E. Synthesis and antiproliferative activity of benzyl and phenethyl analogs of makaluvamines. Bioorg. Med. Chem. 2008, 16, 2541–2549. [Google Scholar] [CrossRef]
  70. Wang, W.; Rayburn, E.R.; Velu, S.E.; Chen, D.; Nadkarni, D.H.; Murugesan, S.; Zhang, R. A novel synthetic iminoquinone, BA-TPQ, as an anti-breast cancer agent: In vitro and in vivo activity and mechanisms of action. Breast Cancer Res. Treat. 2010, 123, 321–331. [Google Scholar] [CrossRef]
  71. Wang, W.; Rayburn, E.R.; Velu, S.E.; Nadkarni, D.H.; Murugesan, S.; Zhang, R. In vitro and in vivo anticancer activity of novel synthetic makaluvamine analogues. Clin. Cancer Res. 2009, 15, 3511–3518. [Google Scholar] [CrossRef]
  72. Wang, F.; Ezell, S.J.; Zhang, Y.; Wang, W.; Rayburn, E.R.; Nadkarni, D.H.; Murugesan, S.; Velu, S.E.; Zhang, R. FBA-TPQ, a novel marine-derived compound as experimental therapy for prostate cancer. Invest. New Drugs 2010, 28, 234–241. [Google Scholar] [CrossRef]
  73. Ezell, S.J.; Li, H.; Xu, H.; Zhang, X.; Gurpinar, E.; Rayburn, E.R.; Sommers, C.I.; Yang, X.; Velu, S.E.; Wang, W.; et al. Preclinical pharmacology of BA-TPQ, a novel synthetic iminoquinone anticancer agent. Mar. Drugs 2010, 8, 2129–2141. [Google Scholar] [CrossRef]
  74. Lee, K.H.; Hall, I.H.; Mar, E.C.; Starnes, C.O.; ElGebaly, S.A.; Waddell, T.G.; Hadgraft, R.I.; Ruffner, C.G.; Weidner, I. Sesquiterpene antitumor agents: Inhibitors of cellular metabolism. Science 1977, 196, 533–536. [Google Scholar]
  75. Haven, G.T.; Krzemien, J.R.; Nguyen, T.T. Study of a renal microsomal inhibitor of hepatic cholesterol synthesis in intact cell systems. Res. Commun. Chem. Pathol. Pharmacol. 1973, 6, 253–262. [Google Scholar]
  76. Santoro, M.G.; Garaci, E.; Amici, C. Prostaglandins with antiproliferative activity induce the synthesis of a heat shock protein in human cells. Proc. Natl. Acad. Sci. USA 1989, 86, 8407–8411. [Google Scholar] [CrossRef]
  77. Ciucci, A.; Gianferretti, P.; Piva, R.; Guyot, T.; Snape, T.J.; Roberts, S.M.; Santoro, M.G. Induction of apoptosis in estrogen receptor-negative breast cancer cells by natural and synthetic cyclopentenones: Role of the IkappaB kinase/nuclear factor-kappaB pathway. Mol. Pharmacol. 2006, 70, 1812–1821. [Google Scholar] [CrossRef]
  78. Hsiang, C.H.; Straus, D.S. Cyclopentenone causes cell cycle arrest and represses cyclin D1 promoter activity in MCF-7 breast cancer cells. Oncogene 2002, 21, 2212–2226. [Google Scholar] [CrossRef]
  79. Ogi, T.; Margiastuti, P.; Teruya, T.; Taira, J.; Suenaga, K.; Ueda, K. Isolation of C11 cyclopentenones from two didemnid species, Lissoclinum sp. and Diplosoma sp. Mar. Drugs 2009, 7, 816–832. [Google Scholar] [CrossRef]
  80. You, J.; Dai, H.; Chen, Z.; Liu, G.; He, Z.; Song, F.; Yang, X.; Fu, H.; Zhang, L.; Chen, X. Trichoderone, a novel cytotoxic cyclopentenone and cholesta-7, 22-diene-3 beta, 5 alpha, 6 beta-triol, with new activities from the marine-derived fungus Trichoderm sp. J. Ind. Microbiol. Biotechnol. 2010, 37, 245–252. [Google Scholar] [CrossRef]
  81. Kazlauskas, R.; Murphy, P.T.; Quinn, R.J.; Wells, R.J. Heteronemin, a new scalarin type sesterterpene from the sponge Heteronema erecta. Tetrahedron Lett. 1976, 17, 2631–2634. [Google Scholar] [CrossRef]
  82. Walker, R.P.; Thompson, J.E.; Faulkner, D.J. Sesterpenes from Spongia idia. J. Org. Chem. 1980, 45, 4976–4979. [Google Scholar] [CrossRef]
  83. Fontana, A.; Cavaliere, P.; Ungur, N.; D'Souza, L.; Parameswaram, P.S.; Cimino, G. New scalaranes from the nudibranch Glossodoris atromarginata and its sponge prey. J. Nat. Prod. 1999, 62, 1367–1370. [Google Scholar] [CrossRef]
  84. Song, J.; Jeong, W.; Wang, N.; Lee, H.S.; Sim, C.J.; Oh, K.B.; Shin, J. Scalarane Sesterterpenes from the Sponge Smenospongia sp. J. Nat. Prod. 2008, 71, 1866–1871. [Google Scholar] [CrossRef]
  85. Wonganuchitmeta, S.N.; Yuenyongsawad, S.; Keawpradub, N.; Plubrukarn, A. Antitubercular sesterterpenes from the Thai sponge Brachiaster sp. J. Nat. Prod. 2004, 67, 1767–1770. [Google Scholar] [CrossRef]
  86. Ledroit, V.; Debitus, C.; Ausseil, F.; Raux, R.; Menou, J.L.; Hill, B.T. Heteronemin as a protein farnesyl transferase inhibitor. Pharm. Biol. 2004, 42, 454–456. [Google Scholar] [CrossRef]
  87. Schumacher, M.; Cerella, C.; Eifes, S.; Chateauvieux, S.; Morceau, F.; Jaspars, M.; Dicato, M.; Diederich, M. Heteronemin, a spongean sesterterpene, inhibits TNF alpha-induced NF-kappa B activation through proteasome inhibition and induces apoptotic cell death. Biochem. Pharmacol. 2010, 79, 610–622. [Google Scholar] [CrossRef]
  88. Jaisamut, S.; Thengyai, S.; Yuenyongsawad, S.; Karalai, C.; Plubrukarn, A.; Suwanborirux, K. Structure-activity relationships of antitubercular scalaranes: Heteronemin revisited. Pure Appl. Chem. 2009, 81, 1019–1026. [Google Scholar] [CrossRef]
  89. Kamel, H.N.; Kim, Y.B.; Rimoldi, J.M.; Fronczek, F.R.; Ferreira, D.; Slattery, M. Scalarane sesterterpenoids: Semisynthesis and biological activity. J. Nat. Prod. 2009, 72, 1492–1496. [Google Scholar] [CrossRef]
  90. Kashman, Y.; Groweiss, A.; Shmueli, U. Latrunculin, a new 2-thiazolidinone macrolide from the marine sponge latrunculia magnifica. Tetrahedron Lett. 1980, 21, 3629–3632. [Google Scholar] [CrossRef]
  91. Spector, I.; Shochet, N.R.; Kashman, Y.; Groweiss, A. Latrunculins: Novel marine toxins that disrupt microfilament organization in cultured cells. Science 1983, 219, 493–495. [Google Scholar]
  92. El Sayed, K.A.; Youssef, D.T.; Marchetti, D. Bioactive natural and semisynthetic latrunculins. J. Nat. Prod. 2006, 69, 219–223. [Google Scholar] [CrossRef]
  93. Hayot, C.; Debeir, O.; Van Ham, P.; Van Damme, M.; Kiss, R.; Decaestecker, C. Characterization of the activities of actin-affecting drugs on tumor cell migration. Toxicol. Appl. Pharmacol. 2006, 211, 30–40. [Google Scholar] [CrossRef]
  94. Longley, R.E.; McConnell, O.J.; Essich, E.; Harmody, D. Evaluation of marine sponge metabolites for cytotoxicity and signal transduction activity. J. Nat. Prod. 1993, 56, 915–920. [Google Scholar] [CrossRef]
  95. Newman, D.J.; Cragg, G.M. Marine natural products and related compounds in clinical and advanced preclinical trials. J. Nat. Prod. 2004, 67, 1216–1238. [Google Scholar] [CrossRef]
  96. Newman, D.J.; Cragg, G.M.; Holbeck, S.; Sausville, E.A. Natural products and derivatives as leads to cell cycle pathway targets in cancer chemotherapy. Curr. Cancer Drug Targets 2002, 2, 279–308. [Google Scholar] [CrossRef]
  97. Morton, W.M.; Ayscough, K.R.; McLaughlin, P.J. Latrunculin alters the actin-monomer subunit interface to prevent polymerization. Nat. Cell Biol. 2000, 2, 376–378. [Google Scholar] [CrossRef]
  98. Amagata, T.; Johnson, T.A.; Cichewicz, R.H.; Tenney, K.; Mooberry, S.L.; Media, J.; Edelstein, M.; Valeriote, F.A.; Crews, P. Interrogating the bioactive pharmacophore of the latrunculin chemotype by investigating the metabolites of two taxonomically unrelated sponges. J. Med. Chem. 2008, 51, 7234–7242. [Google Scholar]
  99. Furstner, A.; Kirk, D.; Fenster, M.D.; Aissa, C.; De Souza, D.; Nevado, C.; Tuttle, T.; Thiel, W.; Muller, O. Latrunculin analogues with improved biological profiles by "diverted total synthesis": Preparation, evaluation, and computational analysis. Chemistry 2007, 13, 135–149. [Google Scholar] [CrossRef]
  100. Khanfar, M.A.; Youssef, D.T.; El Sayed, K.A. Semisynthetic latrunculin derivatives as inhibitors of metastatic breast cancer: Biological evaluations, preliminary structure-activity relationship and molecular modeling studies. ChemMedChem 2010, 5, 274–285. [Google Scholar] [CrossRef]
  101. El Sayed, K.A.; Khanfar, M.A.; Shallal, H.M.; Muralidharan, A.; Awate, B.; Youssef, D.T.; Liu, Y.; Zhou, Y.D.; Nagle, D.G.; Shah, G. Latrunculin A and its C-17-O-carbamates inhibit prostate tumor cell invasion and HIF-1 activation in breast tumor cells. J. Nat. Prod. 2008, 71, 396–402. [Google Scholar] [CrossRef]
  102. Abdel-Mageed, W.M.; Milne, B.F.; Wagner, M.; Schumacher, M.; Sandor, P.; Pathom-aree, W.; Goodfellow, M.; Bull, A.T.; Horikoshi, K.; Ebel, R.; et al. Dermacozines, a new phenazine family from deep-sea dermacocci isolated from a Mariana Trench sediment. Org. Biomol. Chem. 2010, 8, 2352–2362. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Schumacher, M.; Kelkel, M.; Dicato, M.; Diederich, M. A Survey of Marine Natural Compounds and Their Derivatives with Anti-Cancer Activity Reported in 2010. Molecules 2011, 16, 5629-5646. https://doi.org/10.3390/molecules16075629

AMA Style

Schumacher M, Kelkel M, Dicato M, Diederich M. A Survey of Marine Natural Compounds and Their Derivatives with Anti-Cancer Activity Reported in 2010. Molecules. 2011; 16(7):5629-5646. https://doi.org/10.3390/molecules16075629

Chicago/Turabian Style

Schumacher, Marc, Mareike Kelkel, Mario Dicato, and Marc Diederich. 2011. "A Survey of Marine Natural Compounds and Their Derivatives with Anti-Cancer Activity Reported in 2010" Molecules 16, no. 7: 5629-5646. https://doi.org/10.3390/molecules16075629

Article Metrics

Back to TopTop