A review of the components of brown seaweeds as potential candidates in cancer therapy

Ejaz Hussain abc, Li-Jun Wangab, Bo Jiangab, Saba Riazd, Ghazala Yasmeen Butt*d and Da-Yong Shi *ab
aKey Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences (CAS), Qingdao 266071, China. E-mail: shidayong@qdio.ac.cn; Fax: +86 532 8289 8741; Tel: +86 532 8289 8719
bFunction Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology (QNLM), Qingdao 266235, China
cUniversity of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
dPhycology Lab, Department of Botany, Government College University, Lahore, Pakistan. E-mail: dr.ghazalayasmeen@gcu.edu.pk; Tel: +92 42 111 000 010 ext. 257

Received 13th November 2015 , Accepted 4th January 2016

First published on 13th January 2016


Abstract

Finding novel anticancer agents is very important for the treatment of cancer, and marine organisms are a valuable source for developing novel agents for clinical application. Therefore, the isolation and identification of novel anticancer components from brown seaweeds and a study of their mode of action is very attractive in current scenarios to assess their potential as an unexplored source for pharmacological applications. This review will reveal the active components of brown algae, together with their antitumor potential toward cancer treatment according to their structure. This should provide useful information for medicinal chemists in their attempts to develop potent anticancer agents.


image file: c5ra23995h-p1.tif

Ejaz Hussain

Ejaz Hussain received his B.S 4 year degree in Botany from Bahauddin Zakariya University Multan, Pakistan, and his M.Phil. degree in Botany from the Government College University, Lahore, Pakistan. In September 2015, he joined Prof. Da-yong Shi's group for his doctoral degree (PhD) at the Institute of Oceanology, the Chinese Academy of Sciences, CAS. His current research interests are in the isolation, purification and biological potential of natural products with anti-diabetic or antitumor activities.

image file: c5ra23995h-p2.tif

Da-Yong Shi

Prof. Da-yong Shi received his B.Sc. degree in Chemistry from the Fermentation Engineering, Shandong Institute of Light Industry, his M.Sc. degree in Biochemistry from Qingdao University of Science and Technology, and his PhD degree in Medicinal Chemistry from the Institute of Oceanology, the Chinese Academy of Sciences (CAS). From 2005 to 2006, he was a postdoctoral fellow at the Natural Products Research & Infection Biology-Hans Knöll Institute (Jena, Germany). From 2006, he worked at the Institute of Oceanology, CAS. His research interests are in the isolation and synthesis of natural products with anti-diabetes mellitus or antitumor activities.


1 Introduction

Cancer is known as a malignant tumor and is part of a group of diseases involving abnormal cell growth with the potential to kill normal cells and to spread rapidly to all parts of the body. Cancers can be classified on the basis of the type of cell such as carcinoma (cancers derived from the epithelial cells, which are the most common in the aged group of people), sarcoma (cancers originating from connective tissues), lymphoma and leukemia (both classes of cancer derived from hematopoietic cells such as blood cancer), germ cell tumor (cancers arising from pluripotent cells such as dysgerminoma and seminoma), and blastoma (cancer originating from immature embryos, which is the most common type in children).1 There are about 100 known kinds of cancer so far that have been found to affect humans. There are many causes of cancer; for instance, tobacco contributes 25–30%, obesity, poor diet and excessive, and the use of alcohol 30–35%, genetic defects 15–20%, and 10% is following radiation.2 Chemotherapy or CTX is considered a novel strategy to treat cancer cells today, and is defined as the use of chemical substances, namely, anticancer drugs, especially one or more chemotherapeutic agents (alone or combined), to stop the growth of cancer cells. There has also been a tremendous increase in the use of herbal drugs by cancer patients all around the world, which are chosen due to their potential effects against cancer diagnosis and easy access3–6 and which people mostly take as part of a regime comprising multiple complementary and alternative medicine modalities.7,8

Natural products from marine sources that have a strong medicinal potential have attracted much interest in the field of cancer research and in the development of novel anticancer drugs.9 So far, 15[thin space (1/6-em)]000 novel compounds have been discovered from seaweed, and several antitumor compounds are currently being investigated through clinical trials.10,11 Seaweed consumption and its health benefits are correlated and seaweed is considered a potential source for the development of anticancer drugs, functional foods, and pharmacological products.12–17 In the current scenario, pharmaceutical companies are gaining interest in those compounds (sulfated polysaccharides, halogenated furanones, kahalalide F, lectins, kainoidsfucoidans, and aplysiatoxins) and are utilizing them in drug development after isolation from marine algae, especially phaeophyta.12 The phyto-chemical constituents of brown seaweeds, such as carbohydrates, flavonoid, phenols, alkaloids, and proteins, play a key role against pathogens. These compounds also have significant potential as antitumor, antioxidants, and anticoagulants, and due to their immunomodulating activities.18–22 In this review, we discuss the detailed structural composition of different components of brown algae, together with their antitumor potential toward cancer treatment.

2 Categorization of anticancer compounds isolated from brown seaweeds

2.1 Polysaccharides

Polysaccharides are the main components of brown algae rather than green and red algae, and are composed of alginate 1 (Fig. 1), laminarin 2 (Fig. 1), fucoidan 3 (Fig. 1), and their derivatives. Some constituents of porphyran 4 (Fig. 1) and alginic acid 5 (Fig. 1), have also been reported in some species of brown algae.23 The polysaccharides regulate the primary functions of brown algae, like strength and flexibility of the cell wall, prevent desiccation, and keep the ionic equilibrium. The polysaccharide content in brown algae, their structural composition, and functional and anticancer properties have been reported.24–33 Polysaccharides are extensively studied as novel antitumor agents, especially those found in brown seaweeds, such as 8 to 12% of compound 3 (Fig. 1) present in Sargassum sp. and Fucus sp. on a dry weight basis.34 Compound 3 (Fig. 1) comprise complex sulfated polysaccharides and account for 10–20% Dw in brown algae, and are considered as a unique polymer with a heterogeneous composition and structure, mainly composed of sulfated L-fucose and small fractions of mannose, rhamnose, xylose, and glucose.35,36 The polysaccharides isolated from brown seaweeds contain similar repeating sugars but these are different in terms of sulphation and molecular weights, probably due to the different isolation techniques and geographical locations.37–40 Compound 3 (Fig. 1) isolated from five different brown seaweeds consists of 13 to 36% fucose and an 8 to 25% variation in the degree of sulfation.41 Several studies have been carried out about how the sulfation variation in the polysaccharides of brown seaweeds affects their antitumor activity.42,43 Marine algae phaeophyta possess sulfated polysaccharides as the major components of their cell wall and are considered as a valuable bioactive compound with several beneficial biological activities, such as antitumor,44 anticoagulant,45 antiviral,46,47 and antiinflammatory,48,49 as well as immunomodulating activities.50 Marine macro algae, especially phaeophyceae – a class of brown seaweeds – contain fucoidan complex polysaccharides (FCSPs), and their specific biological activities depend on the source of the seaweed, the method of extraction, and their compositional and structural traits.34 Ye et al. isolated highly sulfated polysaccharide fractions, SP-1, SP-2, and SP-3, from Sargassum pallidum and the in vitro results showed significant cytotoxicity against A549 cells, HepG2 cells, and MGC-803 cells.51 Compound 3 (Fig. 1) fractions from Dictyopteris delicatula and Dictyopteris polypodioides induced 60–90% and 28% tumor growth inhibition for Hela and RPMI-7951 cancer cells, respectively.52,53 The polysaccharides extracted from Quebec's Ascophyllum nodosum, Fucus vesiculosus, and Saccharina longicruris contain significant constituents of sulfates, total sugars and uronic acids that have diverse industrial and pharmacological applications.54 Compound 3 (Fig. 1) increased IFN gama T cells production and showed a significant increase in NK cells activity when applied to mice inoculated with P-388 tumor cells.55 Compound 3 (Fig. 1) fractions isolated from Sargassum hornery, Eclonia cava, and Costaria costata induced the inhibition of colony formation in human melanoma and colon cancer cells, and might potentially be effective antitumor agents.56 Compound 3 (Fig. 1) and fucose isolated from brown seaweeds are rich in sulfated polysaccharides and have potential biomedical properties in immunostimulatory,57 immunomodulation, anti-inflammatory, anticoagulant,58 antithrombotic, anticancer, and antiproliferative activities.59 Compound 3 (Fig. 1) isolated from Undaria pinnatifida has the potential to repress the differentiation of adipose cells by inhabiting inflammatory cytokines and is considered as a potent therapeutic agent against obesity and diabetes.60–63 There are authentic reports that low molecular weight compound 3 (Fig. 1) has better antitumor activity than high molecular weight compound 1 (Fig. 1).64 O-Acetylated sulfated galactofucan polysaccharide isolated from the brown seaweed Undaria pinnatifida suppresses the proliferation of PC-3 (prostate cancer), A549 (alveolar carcinoma), and Hela (cervical cancer) and HepG2 (heptacellular carcinoma) cells in a similar way to that of commercial fucoidan.65 Ascophyllan (6, Fig. 1), extracted from the brown alga Ascophyllum nodosum reduced the growth of U937 cells and also induced apoptosis and DNA fragmentation.66,67 Compound 3 (Fig. 1) isolated from the brown seaweed Undaria pinnatifida was tested against AGS stomach cancer cells and it was found that the low molecular fraction was more effective i.e. <30 kDa compared to the >30 kDa fraction.68 Compound 3 (Fig. 1) from Laminaria brasiliensis was found to be cytotoxic to Hela cells at doses of 2.5–40 μg mL−1.69 Polysaccharide fractions extracted from the brown alga Coccophora langsdorfii, with a similar linear backbone to compound 3 (Fig. 1), exhibited a significant colony formation of SK-ML-5 and SK-ML-28 melanoma cells (the percentage of inhibition was 28 and 76%, respectively).70 In vitro and in vivo studies of polysaccharides and sulfated polysaccharides proved their significance as a novel source of anticancer agents and are hence the most studied group of macromolecules, but still none of the agents have entered into clinical trials, which may be due to issues around purifying and identifying their specific structure. Polysaccharides are a diverse group of molecules, where even pure fractions contain a diversity of sugar units, and it is difficult to find a specific administrative route. Polysaccharides from brown seaweeds have provided promising results but still identifying and formulating their molecular structures and determining the mechanism of their administration route is required and is an essential step in the development of anticancer drugs for human deadly diseases (Table 1).
image file: c5ra23995h-f1.tif
Fig. 1 Structures of compounds 1–10.
Table 1 Anticancer components and their efficacy against cancer cells isolated from brown algae
Seaweed Active agent(s) Activity Test type(s) References
Esenia bicycles Laminarin, EBL 1[thin space (1/6-em)]:[thin space (1/6-em)]3[thin space (1/6-em)]:[thin space (1/6-em)]1[thin space (1/6-em)]:[thin space (1/6-em)]6 B. D-Glucan (2, Fig. 1) Anticancer activity at 200 μg mL−1, human melanoma SK-MEL-28 cells and colon cancer DLD1-cells In vitro 58
Coccophoa langsdorfii α-L-Fucoidan (3, Fig. 1) 28% and 78% anticancer activity at 100 μg mL−1 on SK-ML-5 and SK-ML-28 melanoma cells In vitro 70
Ishigeo kamurae Diphlorethohydroxycarmalol (DPHC) (50, Fig. 5) 75% anticancer activity at 100 μg mL−1 on HL60 cancer cells In vitro 143
Saccharina gurjanovae Sulfated galactofucan SgF (MW 123 kDa) (3, Fig. 1) 21% anticancer activity at 800 μg mL−1 on colon cancer DLD-1 cells In vitro 144
Turbinaria conoides Fucoidan (3, Fig. 1) 73.5% anticancer activity at 500 μg mL−1 on A549 cell line In vitro 145
Fucus evanescens Fucoidan (3, Fig. 1) 70%, 63% anticancer activity at 400 μg mL−1 on SK-MEL-28 and SK-MEL-5 cells In vitro 146
Alaria angusta Fucoidan AaF3 (3, Fig. 1) laminaran AaL (2, Fig. 1) 29% and 22% anticancer activity at 400 μg mL−1 on HT29 cells, respectively In vitro 58
Ecklonia cava 8,8-Bieckol (11, Fig. 2) (phlorotannins) Anti-inflammatory In vivo 147
Ecklonia cava Phloroglucinol (PG) (14, Fig. 2) Decreased CD44+ cancer cell population and expression of CSC regulators, such as Sox2, CD44, Oct4, Notch2 and β-catenin In vitro and in vivo 90
Stoechospermum marginatum Spatane derivatives compounds 4 (51, Fig. 5), 1b (52, Fig. 5) 2a (53, Fig. 5), and 4a (54, Fig. 5) Anticancer activity on B16F10 cancer cell line with IC50 values of 3.28, 3.45, 3.62, and 4.11 μg mL−1, respectively, as compared to the standard drug etoposide IC50 = 4.12 μg mL−1 In vitro 148
Sargassum cichorioides Fucoidan ScF2, (55, Fig. 5) 26% anti-proliferation activity at 200 μg mL−1 on DLD-1 cells In vitro 59
F. evanescens Fucoidan FeF2 (56, Fig. 6), 46% anti-proliferation activity at 200 μg mL−1 on RPMI-7951 cells
U. pinnatifida Galactofucan UpF2 (57, Fig. 6) 60% inhibition activity at 200 μg mL−1 on T-47D cells
Fucus vesiculosus Fucoidan (3, Fig. 1) Inhibit growth and apoptosis of HT-29 and HCT116 cells In vitro 149
Ecklonia cava Dieckol (9, Fig. 1) 50% anticancer activity at 84.3 μg mL−1 and 99.6 μg mL−1 on A2780 and SKOV3 cells In vitro 91
Laminaria japonica Phlorotannins (7, Fig. 1) 30% and 43% anti-proliferation activity at 100 μg mL−1 on BEL-7402 and P388 cells In vitro 92
Lobophora variegata Fraction rich in fucans (FRF) (3, Fig. 1) 54% anticancer activity at 25 μg mL−1 on HepG2 cells In vitro 150
Sargassum heterophyllum Sargaquinoic acid (SQA) (28, Fig. 3) SQA displayed an IC50 of 67.4 ± 5.9 μM against MDA-MB-231cells via caspase-3 activity and the down-regulation of the Bcl-2, cell cycle arrest in the G1 phase In vitro 151
Sargassum wightii Methanolic extract 29% and 41% anticancer activity at 200 μg mL−1 on HeLa and MDA-MB 231 cell lines In vitro 152
Pylaiella littoralis PLE extract 67.9%, 37%, 21.9%, and 20.2% antiproliferative activity at 100 μg mL−1 on HT-29, AGS, SK-HEP, NCI-H1299 cell lines In vitro 153
Laminaria japonica Fucoidan (3, Fig. 1) 2% osteoblast differentiation at 10 μg mL−1 in hABM-MSCs In vitro 154
Sargassum macrocarpum Sargafuran (58, Fig. 6) At 15 μg mL−1 kills completely P. acnes In vitro 155
D. polypodioides and Sargassum sp. Fucoidan (3, Fig. 1) 44% and 28% anticancer activity at 200 μg mL−1 on RPMI-7951 cells In vitro 53
Fucus serratus, Laminaria digitata, Ascophyllmnodosum, Pelvetia canaliculata Astaxanthin (59, Fig. 6), β-carotene (25, Fig. 3), zeaxanthin (22, Fig. 3) Improves immune system, protects against eye diseases and anticancer effects In vivo 156 and 157
Leathesia difformes Methanolic extract IC50 of 12.6 μg mL−1 and 40.6 μg mL−1 against KB and HT-29 cells In vitro 158
Turbinaria ornata Sulfated fucan-like polysaccharide (3, Fig. 1) 50% antiproliferative effect at 6.7 μg mL−1 on NSCLC-N6 cell line In vitro 61
Leathesia nana Six bromophenol derivatives 6-(2,3-dibromo-4,5-dihydroxybenzyl)-2,3-dibromo-4,5-dihydroxybenzyl methyl ether (16, Fig. 2), (+)-3-(2,3-dibromo-4,5-dihydroxyphenyl)-4-bromo-5,6-dihydroxy-1,3-dihydroisobenzofuran (17, Fig. 2), 3-bromo-4-(2,3-dibromo-4,5-dihydroxybenzyl)-5-methoxymethyl-pyrocatechol (18, Fig. 2), 2,3,3-tetrabromo-4,4,5,5-tetrahydroxy-diphenylmethane (19, Fig. 2), bis(2,3-dibromo-4,5-dihydroxybenzyl)ether (20, Fig. 3), 2,2,3-tribromo-3,4,4,5-tetrahydroxy-6-ethyloxymethyldiphenylmethane (21, Fig. 3) 17, 19, 20 (Fig. 2 and 3) compounds ethanolic extract of Leathesia nana (EELN) All six exhibited 50% anticancer activity at 10 μg mL−1 on A549, BGC-823, MCF-7, B16-BL6, HT-1080, A2780, Bel7402 and HCT-8 cell lines 77.5%, 80.1%, and 71.4% protein tyrosine kinase (PTK) inhibition activity. Inhibit the growth of sarcoma 180 tumor cells and increase the indices of thymus and spleen to improve the immune system In vitro, in vivo, in vivo 98
Sargassum vulgare C. Agardh, Laminaria digitata Alginates (SVHV and SVLV) (1, Fig. 1) 51.8%, 74.8%, 66.2%, and 88.8% inhibition of sarcoma 180 cells in mice at the doses of 50 and 100 mg per m2 per day for SVLV and SVHV, respectively In vivo 30 and 31
Sargassum fulvellum Pheophytin a (26, Fig. 3) Enhances the neuro differentiation of PC12 cells at 3.9 μg mL−1 concentration In vivo 159
Undaria pinnatifida and Hijikia fusiformis Fucoxanthin, 5,6-epoxy-3′-ethanoyloxy-3,5′-dihydroxy-6′,7′-didehydro-5,6,7,8,5′,6′-hexahydro-β,β-caroten-8-one (23, Fig. 3) Regulates the white adipose tissue (WAT) weight gain and hyperglycemia in diabetic/obese KK-Ay mice In vivo 62
Eisenia bicyclis Pyropheophytin a (60, Fig. 6) Antioxidant activity In vitro 111
Undaria pinnatifida Fucoxanthin and its metabolite, fucoxanthinol (23, Fig. 3) Inhibits adipocyte differentiation in 3T3-L1 cells In vitro 160
Undaria pinnatifida Fucoidan (3, Fig. 1) 15.2%, 29.8%, 39.3%; 45.1% inhibited growth of PC-3 cells and induced apoptosis at 10 μg mL−1 50 μg mL−1, 100 μg mL−1, and 200 μg mL−1, respectively In vivo 161
Fucus vesiculosus Fucoidan-sulfated polysaccharides (3, Fig. 1) 80% anticancer activity at 100 μg mL−1 on DC cells and it has dose-dependent cytoprotective activity In vitro 63
Sargassum mcclurei Fucoidan polysaccharides SmF1, SmF2, SmF3, and SmF3-DS (3, Fig. 1) 17%, 48%, 20%, and 18% inhibited the colony formation of DLD-1 colon cancer cells at 100 μg mL−1 respectively In vitro 162
Sargassum sp., Fucus vesiculosus Crude fucoidan MTA and SIG (3, Fig. 1) 40% and 36% reduction in viability of LLC and B16 cells at 1 μg mL−1 in a dose-dependent manner, respectively In vitro 34
Sargassum filipendula SF-0.7v fucoidan (3, Fig. 1) 38.1% and 31.0%, growth inhibition at 2.0 mg mL−1 on HepG2 and PC3, respectively In vitro 163
Dictyopterispolypodioides Fucoidan (3, Fig. 1) 44% and 28% growth inhibition at 200 μg mL−1 on RPMI-7951 cells, respectively In vitro 53
Ecklonia cava, Sargassum horneri, Costaria costata Fucoidan (3, Fig. 1) 8–55% anticancer activity at 1–200 μg mL−1 on SK-ML-28, DLD-1 cells In vitro 56
Undaria pinnatifida Fucoidan of sporophyll (3, Fig. 1) 10–20% antitumor activity at 0–0.8 mg mL−1 on PC-3, Hela, A549, HepG2 cancer cells In vitro 65
Dictyopterisdelicatula Fucoidan (3, Fig. 1) 60–90% inhibition in tumor growth at 2 mg mL−1 on Hela cancer cells In vitro 52
Sargassum stenophyllum Sarg A fucoidan polysaccharides (3, Fig. 1) 40% and 80% decrease in B16F10 melanoma cell tumors with the dose of 1.5 or 150 μg per animal per day for 3 days In vivo 164
Laminaria digitata Fucoidan (3, Fig. 1) Inhibited inflammation and heterotypic tumor cell adhesions on MDA-MB-231 tumor cells at a significant level In vitro 165
Sargassum thunbergii Fucoidan fractions (3, Fig. 1) Injection of 20 mg kg per day for 10 days increases the survival of Ehrlich carcinoma implanted IP in ICR/Slc mice as compared to control In vivo 166
Alaria esculenta Crude extract Crude extract reduced viability of Caco-2 cancer cells In vivo 167
Laminaria digitata Laminarin (2, Fig. 1) Induced tumor growth HT-29 Bcl-2 cells by the decrease in cytochrome c expression and the increase in Bad and Bax, restricted phosphorylation of ErbB2 and accumulation of cells in sub-G1 and G2-M phase In vivo 93
Ascophyllumnodosum Ascophyllan (6, Fig. 1) Reduced the growth of U937 cells and also induced apoptosis and DNA fragmentation In vivo 66
Ascophyllumnodosum Ascophyllan (6, Fig. 1) Inhibited tumor growth of Vero and XC cells and increased the growth of MDCK cells at concentrations 0–1000 μg mL−1 In vivo 67
Leathesia nana Bis(2,3-dibromo-4,5-dihydroxybenzyl) ether (20, Fig. 3) Induced apoptosis in mouse breast cancer by a mitochondrial-mediated pathway and ROS generation. Inhibited topoisomerase I and cell cycle activity in the S phase In vivo 168
Undaria pinnatifida Fucoidan extract (3, Fig. 1) Inhibited the angiogenesis by human umbilical vein endothelial cells In vivo 116
Fucus vesiculosus Fucoidan (3, Fig. 1) Induced apoptosis of human lymphoma HS-sultan cancer cells by the down-regulation of ERK and activation of caspase-3 pathways In vivo 169
Cladosiphon okamuranus Fucoidan (3, Fig. 1) Inhibited cell growth in MKN-45 cancer cells at 1 mg mL−1 In vitro 170
Sargassum hemiphyllum Hedaol A, B, and C (61, 62, 63, Fig. 6) 50% anticancer activity at 50 μg mL−1 to P-388 cells for Heladaol A, B and C, respectively In vitro 171
Hizikia fusiforme Ethanolic extract 50–60% inhibition in tumor growth with doses of 30–50 μg mL−1. Increased caspase-3, 8,9 PARP and decreased IAP-2, Bcl-2, IAP-1 and XIAP In vivo 172
Ecklonia cava Dieckol (9, Fig. 1) Induced SK-Hep1 human hepatoma cell motility through the suppression of matrix metalloproteinase-9 pathways In vivo 85
Sargassum fulvellum Sodium alginate (8, Fig. 1) Inhibited the tumor growth of S-180 in mice In vivo 83
Stypopodim flabelliforme Stypodiol (38, Fig. 4) Induced anti-proliferation activity to SH-SY5Y cells with an IC value of ≤50 μM and was nontoxic to V79 normal cells In vitro 123
Stypopodim flabelliforme Two mero-diterpenoids derivatives 2β,3α-epitaondiol (41, Fig. 4), and flabellinol (42, Fig. 4) All three showed cytotoxic to neuro-2a cells at 2–11 μM and 9–24 μM to NCI-H460 cells, respectively In vivo 122
Bifurcaria bifurcata Elaganolone (64, Fig. 6) Strong antiprotozoal activity against T. bruceirhodesiense and a selective SI of 12.4 in LC6 cells In vitro 173


2.2 Phenolic compounds

Polyphenols act as antioxidants and have an ability to scavenge free radicals and up-regulate certain metal chelation reactions to improve the body's own antioxidant system.71 Phenolics are secondary metabolites of seaweeds and are composed of aromatic rings bearing one or more hydroxyl groups and include flavonoids, lignans, tannins, and phlorotannins and may have an in vivo affect in receptor sensitivity,28,72 cell signaling pathways, and in gene regulation or inflammatory enzyme activity.73 Phlorotannins (7, Fig. 1), are produced in abundance through secondary metabolism in phaeophyceae.74–76 Compound 7 (Fig. 1) consists of different molecular sizes (400–400[thin space (1/6-em)]000 Da) and accounts for 0.5–20% Dw in brown algae. The phenolic content quantitative analysis and comparative studies of Fucaceae, Sargassaceae, Cystoseiraceae, and Laminariaceae have been reported.77–81 A flavonoid content of 88 μg mL−1 have been isolated from a methanol extract of the brown alga Turbinaria ornate and exhibited significant anti-proliferation activity on A549, PC-3, HCT-15, and MG-63 tumor cells in vivo.82 Sodium alginate (8, Fig. 1), isolated from Sargassum fulvellum, inhibited the tumor growth of S-180 mice.83,84 Dieckol (9, Fig. 1), from brown alga when applied to SK Hep-1 cells decreased TPA cell motility and MMP-9 activity associated with AP-1 in MAPK signaling pathways.85 Methanol extracts of Sargassum fulvellum and Sargassum thunbergii inhibited by 79.1% and 72.1% an inflammatory symptom of mouse ear edema without any toxicity, respectively.74 A polyphenolic crude ethanolic extract from Ecklonia cava induced the inhibition of MMP-2 and MMP-9 activity, and the link was thought to be associated with polyphenols and anticancer activity.86

Sargassum muticum, Fucus vesiculosus, Gelidium sesquipedale, and Cystoseira compressa extracts were evaluated to find out the total flavonoid and phenolic contents in order to investigate the cytotoxic and mutagenic potential. Hexane extracts of these isolates had no significant cytotoxic and mutagenic activity against the human hepatocellular carcinoma Hep 3B cell line when applied as 5–50 μg mL−1. The finding suggests that the phytochemical constituent of brown seaweeds might be a suitable agent for the control of human deadly diseases.87–89 Phlorotannin compounds, such as eckol (10, Fig. 1), 8,8′-bieckol (11, Fig. 2), 6,6′-bieckol (12, Fig. 2), and 6,8′-bieckol (13, Fig. 2), have been isolated from several brown algae, namely Sargassum fulvellum, Sargassum thunbergii, Ecklonia cava, Hizikia fusiformis, Ishige okamurae, Ecklonia cava, Eisenia arborea, and Eisenia arborea.74 Phloroglucinol (14, Fig. 2), isolated from Ecklonia cava, decreases the CD44+ cancer cell population and the expression of CSC regulators such as Sox2, CD44, Oct4, Notch2, and β-catenin.90 Compound 9 (Fig. 1) from Ecklonia cava exhibited 50% anticancer activity at 84.3 μg mL−1 and 99.6 μg mL−1 on A2780 and SKOV3 cells, respectively.91 The growth of two tumor cell lines BEL-7402 and P388 cells was inhibited to 30 to 43% at 100 μg mL−1 of compound 7 (Fig. 1)-rich extracts from Laminaria japonica and apoptosis was also observed.92 Compound 2 (Fig. 1), isolated from the brown alga Laminaria digitata, induced apoptosis in HT-29 Bcl-2 cells in a dose-dependent manner, via increasing the percentage of cells in the sub-G1, G2-M phase and inhibited the heregulin-stimulated phosphorylation of ErbB2.93 Similarly, compound 1 (Fig. 1) was isolated from the brown alga Sargassum vulgare and studied in vivo and exhibited 27 to 88% inhibition of tumor growth in Swiss mice with S-180 implanted SC, supplemented at doses of 50 and 100 mg m2 per day for 10 days.94 Kimiya et al. studied various extracts of brown algae in vivo and in vitro, including Ecklonia cava, Codium fragile, Ulva japonica, Undarina pinnatifida, and P. binghamiae, against RBL-2H3 cells at 100 to 200 μg mL−1; among these, P. binghamia exhibited the highest degranulation of both RBL-2H3 cells as well mouse esinophils.95 A phlorotannin compound, dioxinodehydroeckol (15, Fig. 2), induced apoptosis in MCF-7, MDA-MB-231 cells and increased the activities of caspase-3 and -9, Bax, p53, and PARP pathways via the down-regulation of NF-kB and Bcl2.20,28 Bromophenol compounds from marine organisms, especially from brown algae, have proven to be valuable characteristic natural products with a range of potential biological activities, including antioxidant, antidiabetic, anticancer, and as an α-glucosidase inhibitor. Leathesia nana, a marine brown seaweed, possess six unique bromophenols compounds (16–21, Fig. 2 and 3) and all six exhibited 50% anticancer activity at 10 μg mL−1 on A549, BGC-823, MCF-7, B16-BL6, HT-1080, A2780, Bel7402, and HCT-8 cell lines, respectively.96–99


image file: c5ra23995h-f2.tif
Fig. 2 Structures of compounds 11–19.s

image file: c5ra23995h-f3.tif
Fig. 3 Structures of compounds 20–28.

2.3 Carotenoids

Carotenoids are natural pigments and dietary fibers and contain functional compounds primarily found in both plants and animals. Brown seaweed's cell wall contains catatonic compounds, including lutein, zeaxanthin (22, Fig. 3), and fucoxanthin (23, Fig. 3).100,101 Brown algae is considered to be a rich marine reservoir of secondary metabolites, especially carotenoids, and they possess a range of bioactivities, including, antioxidant, anticancer, anti-inflammatory, and antiviral.102 Epidemiological investigations have evidenced that there is a clear link between seaweed carotenoids diet and cancer risk, and considered that these have potential as important pharmaceutical compounds, which might be a promising anticancer marine drug.103 Carotenoids are colored terpenes produced by a secondary metabolism in brown algae, and have an abundant structural variety, such as a-carotene (24, Fig. 3), and b-carotene (25, Fig. 3), chlorophyll a, phaeophytin a, (26, Fig. 3), lutein, zeaxanthin (22, Fig. 3), and fucoxanthin (23, Fig. 3). They play a key role in human nutrition, providing provitamin A, as well as acting in hormone synthesis, photomorphogenesis, photoprotection, and against cerebrovascular diseases.104–107 Compound 23 (Fig. 3) content values in the range 1–6 mg g−1,15,18 structural variability,108,109 and the bioactivity of some brown seaweeds have been reported up to 16 mg g−1 for Turbinaria sp.,106 and even higher for Laminaria sp. and Undaria pinnatifida.105 Compound 23 (Fig. 3) and fucoxanthinol both have a reduced proliferation of HUVECs without affecting their chemotaxis and inhibit the growth in ex vivo rat aortic rings through the suppression of micro vessel (CD31+ve) formation.110 A compound 23 (Fig. 3) derivative from two brown algae Undaria pinnatifida and Hijikia fusiformis regulates the white adipose tissue (WAT) weight gain and hyperglycemia in diabetic/obese KK-Ay mice in vivo.62 Similarly, compound 23 (Fig. 3) and its metabolite, fucoxanthinol were isolated from the brown alga Undaria pinnatifida and exhibited significant inhibition to adipocyte differentiation in 3T3-L1cells.111 Compound 23 (Fig. 3) induced apoptosis via the activation of caspase-3 and -9, and reduced the expression of Bax and Bcl-2 proteins, but not Bcl-X(L).112 However, apoptosis, DNA fragmentation, a reduction in Bcl-2, Bax, and caspase-3 activation has been observed in DU145, PC-3 and LNCaP prostate cancer cell lines.113,114 Compound 23 (Fig. 3) reduced cell viability and induced apoptosis via a decrease in Bcl-2 expression on HT-29 and DLD-1 cells.115 Compound 23 (Fig. 3) increased the NFκB-regulated Bax/Bcl-2 mRNA ratio, via the inhibition of ERCC1 and NF-kB expression through blocking the P13K/AKT pathways. It also induced cell inhibition of human hepatoma HepG2 cells and improved cisplatin treatment.116 Compound 23 (Fig. 3) induced the apoptosis G2/M1 cell cycle arrest via down-regulating the expression of CyclinB1, which was linked with the JAK/STAT pathway.117 Compound 23 (Fig. 3) induced apoptosis via the reduction of cell viability in EJ-1 cancer cells and increased hypodiploid cells, the DNA ladder, and caspase-3 activities.118

2.4 Terpenoids

Terpenes and polyketides, for example, account for most of the secondary metabolites and can be recognized as oligomers of the primary metabolites isoprene and acetate, respectively. Terpenes and polyketides in brown algae frequently occur as secondary metabolites and their structure ranges from acyclic entities with a linear chain to complex polycyclic molecules, which are considered to be the main components for cancer treatment.119 The antitumor activity of meroterpenoid metabolites isolated from Sargassum fallax against p388 human cancer cells were found to have IC50 values of 17, 14, 32, and >27–29 μM when treated with 1 mg mL−1 for sargaquinone (27, Fig. 3), sargaquinoic acid (28, Fig. 3), sargahydroquinoic acid (29, Fig. 4), and fallachromonoic acid (30, Fig. 4), fallahydroquinone (31, Fig. 4), fallaquinone (32, Fig. 4), and sargachromenol (33, Fig. 4), respectively.120 Atomarianone A (34, Fig. 4), and B (35, Fig. 4), compounds were isolated from Taonia atomaria and both were found to be cytotoxic to NSCLC-N6, A549 cell lines with IC50 values of <7.35 μM.121 Diterpenoid metabolites isolated from the brown alga Cystoseira mediterranea were tested in vivo against in mouse P388 leukemia cells and found to have significant cell inhibition activity.122 The isolated terpenoid derivatives sargaquinone (27, Fig. 4), taondiol (36, Fig. 4), isoepitaondiol (37, Fig. 4), stypodiol (38, Fig. 4), stypoldione (39, Fig. 4), and sargaol (40, Fig. 4) were found to have strong antioxidant potential, with specific biological activities, such as, compound 39 (Fig. 4) which inhibits the microtubule polymerization, compound 36 (Fig. 4) which exhibits anticancer activity, compound 37 (Fig. 4) related to insecticidal activity, and cytotoxicity against P-388 lymphocytic cells related to metabolites of compounds 27 and 40 (Fig. 4). Two mero-diterpenoids, including, 2β,3α-epitaondiol (41, Fig. 4) and flabellinol (42, Fig. 4), were isolated from Stypopodium flabelliforme, and in vivo studies showed that all three were cytotoxic to neuro-2a cells at 2–11 μM and 9–24 μM to NCI–H460 cells, respectively.123 Compound 38 (Fig. 4) induced anti-proliferation activity to SH-SY5Y cells with an IC value of ≤50 μM and was also found to be nontoxic to V79 normal cells.124
image file: c5ra23995h-f4.tif
Fig. 4 Structures of compounds 29–42.

2.5 Proteins, lipids, sterols and quinones, vitamins, fatty acids, and amino acids

The protein structure of brown seaweeds and their biological potential are still poorly studied so far, but their amino acid composition has been well documented by several studies.125 The protein contents in the brown seaweed are usually considered small and different from species to species, such as, Undaria sp., which has the highest ratio of 24% dry weight, followed by 17–21% for Fucus, Sargassum and Laminaria, while the lowest content of 10% is for Ascophyllum sp. Brown seaweed proteins generally contain the highest content of threonine, alanine, valine, glycine, leucine, and lysine, together with several amino acids, such as histidine, tryptophan, cysteine, methionine, and tyrosine with lower levels.126,127 Combined glutamic acid and aspartic acid levels of 22–44%, 39–41%, and 18% wet weight of the total amino acid fraction are reported for Fucus sp., Sargassum sp. and Laminaria digitata, respectively.128–130 Amino acids isolated from the brown seaweeds Sargassum vulgare (C. agardh) and Sargassum thunbergii extracts have been considered as potential sources of new treatments for parasitic diseases such as anti-helmintics.131 Ishihara et al. isolated two polyunsaturated fatty acids 18[thin space (1/6-em)]:[thin space (1/6-em)]4n − 3 and 16[thin space (1/6-em)]:[thin space (1/6-em)]4n − 3 from two brown marine algae Ulva pertusa and Ulva pinnatifida and extensively studied them in vivo, and they exhibited a strong inhibition on Leukotriene B4, 5-hydroxyeicosatetraenoic acid and leukotriene C4 in MC/9 mice mast cells.132 Deoxylapachol a 1,4-naphthoquinone (43, Fig. 5), derivative isolated from Landsburgia quercifolia induced apoptosis to p-388 human cancer cells (IC > 0.6 pg mL−1).133 Sargachromanol E (44, Fig. 5), from Sargassum siliquastrum, induced caspase-3-mediated apoptosis in HL-60 cancer cells.134 Two steroidal compounds, named 3-keto-22-epi-28-nor-cathasterone and cholest-4-ene-3,6-dion, were isolated from Cystoseira myrica and induced cytotoxicity to HEPG-2 and HCT116 cells in the range of 12.38–1.16 μM in selective patterns.135 Ergosterols (45, Fig. 5), isolated from the brown alga Lyengaria stellata, exhibited a noticeable hematopoietic effect when it was applied orally at the doses of 10 mg/200 g body weight to rabbits for 30 days.136,137 Another compound, fucosterol (46, Fig. 5), isolated from the brown seaweeds Pelvetia siliquosa, Cystoseira foeniculacea, and Sargassum angustifolium, exhibited a significant cytotoxic effect to HT-29, Caco-2 and T47D cells.138,139 Laminaria japonica glycoprotein (LJGP) induced apoptosis and cell cycle arrest in AGS, HepG2, and HT-29 cancer cells in a dose-dependent manner via mediation by a Fas signaling pathway, caspas-3 activation, and a mitochondrial pathway.140 The PGE2 production and histamine release were lowered in the canine mastocytoma cell line C2 and RBL-2H3 cells treated with alpha-linolenic acid (47, Fig. 5), γ-linolenic acid (48, Fig. 5), and docosahexaenoic acid (49, Fig. 5), respectively.141,142
image file: c5ra23995h-f5.tif
Fig. 5 Structures of compounds 43–55.

3 Mechanisms of action and comparison of the toxicity of the compounds with other chemotherapeutic drugs

In recent decades, scientists have devoted much attention to investigating the nature of the initialization and progression of malignant tumors through the advancement of genetics and molecular biology. In some research papers, there are some mechanistic studies that prove the role of these compounds in regulating the biological and physiological processes of the cell, and this has research field has occupied many researchers around the chemotherapeutic world.20,65,66,138 In some reports, researchers have identified specific inhibitory activities of natural compounds from brown seaweeds, in a number of key cellular processes, including, antimetastatic, antiangiogenic, telomerase, proapoptotic, tumor angiogenesis, and apoptosis pathways.174,175 Compound 6 (Fig. 1) has been reported to induce cytokine release TNF and granulocyte colony-stimulating factor (GCSF) from macrophage-derived RAW264.7 cells though apoptosis and DNA fragmentation,66 while a sulfated polysaccharide compound 3 (Fig. 1) extracted to Hydroclathrus clathratus was found to increase the tumor necrosis factor (TNF-α) in mouse serum.64 Compound 14 (Fig. 2) has been evidenced to inhibit the epithelial-mesenchymal cell transition (EMT) process and to suppress the metastatic ability of breast cancer cells through a decrease in expression of SNAIL-related zinc-finger transcription factors and by the inhibition of PI3K/AKT and Ras/Raf-1/ERK signaling pathways.176 Compound 22 (Fig. 3) proved its anti-carcinogenic activity was interrelated with mutagens such as 1-nitropyrene,177 and aflatoxin B1 (AFB1),178 by regulating specific genes involved in T-cell transformations.179 Compound 23 (Fig. 3) has been evidenced to induce apoptosis through caspase-3, -7, -9 activation and to suppress Bax and Bcl-2 proteins expression through inhibition of the NF-kB pathway in several cancer cell lines, including the HL-60 cell line,114 MDA-MB-231,180 MCF-7 breast cancer cells,181 Caco-2 colon cancer cells,182 Caco-2, HT-29, DLD-1 cells,115 prostate cancer cell line,112 and the urinary bladder cancer EJ-1 cell line.118 Compound 25 (Fig. 3) prevents the carcinogenesis by preventing DNA damage,183 the onset of cancers, especially lung cancer,184 and regulates several biological functions, including the hormones, and tissue growth and differentiation, and is a mediator of cell signaling and a regulator of cells.104 A compound 59 (Fig. 6)-rich algal extract, has been evidenced to provide potent protection against UVA-induced DNA damage to melanocytes, intestinal CaCo-2 cells,185 and to inhibit the androgen-induced proliferation of human prostate cancer cells.113,186 In the current scenario of chemotherapy, there are several studies that have proven the synergistic effects of natural bioactive compounds when in combination with complementary or conventional anticancer drugs.187–189 These chemotherapeutics have been received promising results for the application of natural compounds when in combination with commonly used anticancer drugs, such as in the activation of different molecular mechanistic pathways, improved drug absorption, enhanced anticancer drug efficiency, and increased clinical responses.27,175,190 A compound 7 (Fig. 1)-rich extract exhibited significant antiproliferative effect over that of 5-fluorouracil (a commercial chemotherapy drug), against P388 and BEL-7402 cancer cells at doses of 120 μg mL−1 and >200 μg mL−1, respectively.92 A recent finding for compound 22 (Fig. 3) has evidenced a significant antiproliferative effect against MCF-7 and MDA-MB-231 cells, through apoptosis and cell cycle arrest when co-treated with cisplatin, tamoxifen, and paclitaxel. The suppression of Bcl-2 proteins expression, ERK and AKT signaling pathway, the regulation of estrogen receptors, and the production of oxidative stress are possible mechanisms.27 Compound 23 (Fig. 3) has been found to have promising results when combined in treatment with oxaliplatin/5-fluorouracil/leucovorin or irinotecan/fluorouracil/leucovorin, such as a decrease in fatigue and an increased survival rate of patients receiving the co-treatment.191 Compound 50 (Fig. 5) has been found to be more potent than acarbose, a commercial carbohydrate digestive enzyme inhibitor, against alpha-glucosidase and alpha-amylase with IC50 values of 0.16 mM and 0.53 mM, respectively, and can be considered a potent chemotherapeutic drug for treating diabetes.143 However, despite the strong potential of these studies, these natural compounds need more comprehensive studies carried out before they can be translated into useful modern chemotherapeutic drugs (Fig. 7 and 8).
image file: c5ra23995h-f6.tif
Fig. 6 Structures of compounds 56–64.

image file: c5ra23995h-f7.tif
Fig. 7 Molecular mechanisms and targets of phloroglucinol, fucoxanthin, and fucoidan mediating anticancer activity in breast cancer.192

image file: c5ra23995h-f8.tif
Fig. 8 Anticancer effects of fucoxanthin and fucoxanthinol on various types of cancer cells and their mechanism of action.193

4 Conclusions and prospective

Cancer is a multi-faceted molecular disease that cannot be cured by drugs alone to date. Many academic and research institutes, even after their intensive efforts, are still unable to find potential antitumor compounds rather than just a few useful products.194 Therefore, there is a growing trend for scientists to seek novel compounds to treat cancer. Although a few marine antitumor compounds are being tested and utilized to treat human deadly diseases such as cancer, they are known to have some side effects such as sleepiness, nervousness, tiredness, and drowsiness. To eliminate these side effects, researchers have paid great attention to finding potential drugs from marine sources with potent efficacy and specificity for the treatment of cancer. Brown seaweeds have a diversity of compounds and novel entities such as polysaccharides, polyphenolic contents, carotenoids, terpenoids, bromophenols, proteins, lipids, amino acids, vitamins, sterols, and quinines. Therefore, in vitro and in vivo studies of these compounds have proven their strong potential against cancer cells without toxicity problems. Considerably, there are still many issues that persist to develop a marine drug such as potential toxic side effects and large-scale production issues. However, biochemical combinatorial genetic and metabolic engineering can be helpful for the development of natural drugs by modification or by eliminating the toxic groups from these natural compounds to obtain a pure compound that is more specific and less cytotoxic. In addition, the anticancer activity and specificity of active compounds can be increased to determine the exact mechanism of action, structural activity relationship, synthetic methods, and drug metabolism. There is a need for extensive study to overcome the issues related to finding these desired compounds such as large-scale production issues that may be improved through new aquaculture and fermentation processes. The brown seaweeds studied to date have exhibited strong potential against various cancer cells without producing toxicity and therefore, there is a need to explore the marine brown algae for the development of new pharmaceutical products. Thus, this review might be useful for developing potential anticancer drugs from brown seaweeds.

Abbreviations

IFNInterferon factor
NKNatural killer
FCSPsFucoidan complex sulfated polysaccharides
AGSHuman stomach cancer cell line
RPMI-7951Human malignant melanoma obtained
P-388Murine leukemic cells
Pc-3Prostate cancer
A549Alveolar carcinoma
HelaCervical cancer
HepG2Heptacellular carcinoma
U937Human leukemic monocyte lymphoma
kDaKilo dalton
SK-ML-5Human malignant melanoma
SK-ML-28Human malignant melanoma
HCT-15Human colon cancer cell line
MG-63Human osteosarcoma
MCF-7Breast cancer
Hep-2Liver cancer
S-180Sarcoma 180
DwDry weight
MAPKMitogen-activated protein kinase
TPATetradecanoylphorbol acetate
MMP-9Matrix metalloproteinase-9
AP-1Activator protein-1
Hep 3BHuman hepatoma cell line
BGC-823Human gastric cancer cell line
B16-BL6Murine melanoma
HT-1080Human fibrosarcoma cells
A2780Human ovarian cancer cell line
Bel7402Human hepatocellular carcinoma
HCT-8Human colon cancer cells
CSCsCancer stem-like cells
SKOV3Human ovarian carcinoma cell line
P388Human leukemia cells
HT-29Human colon adenocarcinoma cells
RBL-2H3Basophilic leukemia cell line
MDA-MB-231Human mammary adenocarcinoma
WATWhite adipose tissues
HUVECsHuman umbilical vein endothelial cells
3T3-L1Mouse adipose tissue cell line
DU145Human prostate cancer cell line
LNCaPHuman prostate adenocarcinoma cell line
DLD-1Human colorectal adenocarcinoma
ERCC1Expression of excision repair cross complementation 1
PI3K/AKTPhosphatidylinositol 3-kinase
NFκBNuclear transcription factor kappa B
EJ-1Human bladder cancer cells
MGC-803Human gastric adenocarcinoma cancer cells
JAK/STATJanus kinase/signal transducer and activator of transcription
Neuro-2aMouse neuroblastoma cell line
V79Chinese hamster lung fibroblast cell line
MC/9Mice mast cells
HCT116Human colon cancer cells
Caco-2Human epithelial colorectal
T47DBreast cancer cell line
LJGPLaminaria japonica glycoprotein
EBLEisenia bicyclis laminaran
SgFSulfated glactofucan
AaFAlaria agusta fucoidan
AaLAlaria agusta laminaran
ScFSargassum cichorioides fucoidan
FeFFicus evanescens fucoidan
UpFUndaria pinnatifida galactofucan
FRFFraction rich in fucans
SQASargaquinoic acid
PLEPylaiella littoralis extract
NCI-H1299Human lung cancer cell line
hABM-MSCsHuman alveolar bone marrow-derived mesenchymal stem cells
ERKExtracellular signal-related kinase
JNKc-Jun N-terminal kinase
KBHuman leukemia-lymphoma cell line
NSCLC-N6Human non-small cell bronchopulmonary carcinoma line
PTKProtein tyrosine kinase
SVLVSargassum vulgare low viscosity
SVHVSargassum vulgare high viscosity
PC12Clonal rat pheochromocytoma cell line
DCDendritic cells
SmFSargassum mcclurei fucoidan
LCCLewis lung carcinoma cells
MCB16Melanoma cells B16
MDCKMadine–Darby canine kidney
ROSReactive oxygen species
MKN-45Human gastric adenocarcinoma
SK-Hep1Human hepatoma cell line
LC6Large cell lung cancer cell line
SH-SY5YHuman neuroblastoma cell line
NCI-H460Lung cancer cell line
EELNEthanolic extract of Leathesia nana
GCSFGranulocyte colony-stimulating factor
EMTEpithelial-mesenchymal cell transition

Acknowledgements

The study was supported by the National High-level personnel of special support program (“people plan”), Program for Talent of Aoshan, National Natural Science Foundation of China (No. 41276167, 41206066, 41506169 and 41306157), China Postdoctoral Science Foundation (No. 2015T80755 and 2014M551972), the National science and technology support project (2013BAB01B02-2), Focus on research and development plan in Shandong province (2015GGF01039), Science and Technology Development Program of Qingdao Shinan District (No. 2013-12-008-SW), and Basic Research Projects of Qingdao Science and Technology Plan (No. 14-2-4-35-jch).

Notes and references

  1. R. Weinberg, The biology of cancer, Garland Science, New York, 2007, vol. 255 Search PubMed.
  2. R. J. B. King and M. W. Robins, Cancer biology, Pearson Education, 2006 Search PubMed.
  3. B. Poonthananiwatkul, R. H. Lim, R. L. Howard, P. Pibanpaknitee and E. M. Williamson, J. Ethnopharmacol., 2015, 168, 100 CrossRef PubMed.
  4. A. Goey, J. Beijnen and J. Schellens, Clin. Pharmacol. Ther., 2014, 95, 354 CrossRef CAS PubMed.
  5. T. Zeller, K. Muenstedt, C. Stoll, J. Schweder, B. Senf, E. Ruckhaeberle, S. Becker, H. Serve and J. Huebner, J. Cancer Res. Clin. Oncol., 2013, 139, 357 CrossRef CAS PubMed.
  6. G. Cragg and D. Newman, Phytochem. Rev., 2009, 8, 313 CrossRef CAS.
  7. S. M. Alsanad, E. M. Williamson and R. L. Howard, Phytother. Res., 2014, 28, 1749 CrossRef PubMed.
  8. J. McLay, D. Stewart, J. George, C. Rore and S. Heys, Eur. J.Clin. Pharmacol., 2012, 68, 811 CrossRef CAS PubMed.
  9. A. M. Ibrahim, M. H. Mostafa, M. H. El masry and M. A. El-Naggar, Egypt. J. Aquat. Res., 2005, 31, 146 CAS.
  10. N. A. Shoeib, M. C. Bibby, G. Blunden, P. A. Linley, D. J. Swaine, R. T. Wheelhouse and C. W. Wright, J. Nat. Prod., 2004, 67, 1445 CrossRef CAS PubMed.
  11. J. W. Blunt, B. R. Copp, R. A. Keyzers, M. H. Munro and M. R. Prinsep, Nat. Prod. Rep., 2012, 29, 144 RSC.
  12. A. J. Smit, J. Appl. Phycol., 2004, 16, 245 CrossRef CAS.
  13. K. H. Cardozo, T. Guaratini, M. P. Barros, V. R. Falcão, A. P. Tonon, N. P. Lopes, S. Campos, M. A. Torres, A. O. Souza and P. Colepicolo, Comp. Biochem. Physiol., Part C: Toxicol. Pharmacol., 2007, 146, 60 CrossRef PubMed.
  14. W.-L. Chu, Curr. Top. Nutraceutical Res., 2011, 9, 83 CAS.
  15. S. Lordan, R. P. Ross and C. Stanton, Mar. Drugs, 2011, 9, 1056 CrossRef CAS PubMed.
  16. N. V. Thomas and S.-K. Kim, Environ. Toxicol. Pharmacol., 2011, 32, 325 CrossRef CAS PubMed.
  17. R. Pangestuti and S.-K. Kim, in Handbook of Anticancer Drugs from Marine Origin, Springer, 2015, p. 165 Search PubMed.
  18. S. L. Holdt and S. Kraan, J. Appl. Phycol., 2011, 23, 543 CrossRef CAS.
  19. S. Mohamed, S. N. Hashim and H. A. Rahman, Trends Food Sci. Technol., 2012, 23, 83 CrossRef CAS.
  20. Y.-X. Li, I. Wijesekara, Y. Li and S.-K. Kim, Process Biochem., 2011, 46, 2219 CrossRef CAS.
  21. K. Queiroz, V. Medeiros, L. Queiroz, L. Abreu, H. Rocha, C. Ferreira, M. Juca, H. Aoyama and E. Leite, Biomed. Pharmacother., 2008, 62, 303 CrossRef CAS PubMed.
  22. K. Rupapara, N. Joshi and K. Vyas, Int. J. Curr. Microbiol. Appl. Sci., 2015, 4, 300 Search PubMed.
  23. T. N. Zvyagintseva, N. M. Shevchenko, A. O. Chizhov, T. N. Krupnova, E. V. Sundukova and V. V. Isakov, J. Exp. Mar. Biol. Ecol., 2003, 294, 1 CrossRef CAS.
  24. L. O'Sullivan, B. Murphy, P. McLoughlin, P. Duggan, P. G. Lawlor, H. Hughes and G. E. Gardiner, Mar. Drugs, 2010, 8, 2038 CrossRef PubMed.
  25. S. Cofrades, I. López-López, L. B. Bravo, C. Ruiz-Capillas, S. Bastida, M. T. Larrea and F. Jiménez-Colmenero, Food Sci. Technol. Int., 2010, 16(5), 361–370 CrossRef CAS PubMed.
  26. M. Elleuch, D. Bedigian, O. Roiseux, S. Besbes, C. Blecker and H. Attia, Food Chem., 2011, 124, 411 CrossRef CAS.
  27. T. Alekseyenko, S. Y. Zhanayeva, A. Venediktova, T. Zvyagintseva, T. Kuznetsova, N. Besednova and T. Korolenko, Bull. Exp. Biol. Med., 2007, 143, 730 CrossRef CAS PubMed.
  28. C.-S. Kong, J.-A. Kim, N.-Y. Yoon and S.-K. Kim, Food Chem. Toxicol., 2009, 47, 1653 CrossRef CAS PubMed.
  29. M. C. R. de Souza, C. T. Marques, C. M. G. Dore, F. R. F. da Silva, H. A. O. Rocha and E. L. Leite, J. Appl. Phycol., 2007, 19, 153 CrossRef PubMed.
  30. A. P. A. de Sousa, M. R. Torres, C. Pessoa, M. O. de Moraes, F. D. Rocha Filho, A. P. N. N. Alves and L. V. Costa-Lotufo, Carbohydr. Polym., 2007, 69, 7 CrossRef.
  31. J. Jongaramruong and N. Kongkam, J. Asian Nat. Prod. Res., 2007, 9, 743 CrossRef CAS PubMed.
  32. J. L. Abrantes, J. Barbosa, D. Cavalcanti, R. C. Pereira, C. F. L. Fontes, V. L. Teixeira, T. M. L. Souza and I. C. Paixão, Planta Med., 2010, 76, 339 CrossRef CAS PubMed.
  33. P. Ruperez and F. Saura-Calixto, Eur. Food Res. Technol., 2001, 212, 349 CrossRef CAS.
  34. M. T. Ale, J. D. Mikkelsen and A. S. Meyer, Mar. Drugs, 2011, 9, 2106 CrossRef CAS PubMed.
  35. V. H. Pomin and P. A. Mourão, Glycobiology, 2008, 18, 1016 CrossRef CAS PubMed.
  36. A. I. Usov and M. I. Bilan, Russ. Chem. Rev., 2009, 78, 785 CrossRef CAS.
  37. M. Funaki, M. Nishizawa, T. Sawaya, S. Inoue and T. Yamagishi, Fish. Sci., 2001, 67, 295 CrossRef CAS.
  38. P. Ghosh, U. Adhikari, P. K. Ghosal, C. A. Pujol, M. a. J. Carlucci, E. B. Damonte and B. Ray, Phytochemistry, 2004, 65, 3151 CrossRef CAS PubMed.
  39. S. M. Colegate and R. J. Molyneux, Bioactive natural products: detection, isolation, and structural determination, CRC press, 2007 Search PubMed.
  40. Y. Hou, J. Wang, W. Jin, H. Zhang and Q. Zhang, Carbohydr. Polym., 2012, 87, 153 CrossRef CAS.
  41. K. Senthilkumar, P. Manivasagan, J. Venkatesan and S.-K. Kim, Int. J. Biol. Macromol., 2013, 60, 366 CrossRef CAS PubMed.
  42. T. Teruya, T. Konishi, S. Uechi, H. Tamaki and M. Tako, Int. J. Biol. Macromol., 2007, 41, 221 CrossRef CAS PubMed.
  43. D. O. Croci, A. Cumashi, N. A. Ushakova, M. E. Preobrazhenskaya, A. Piccoli, L. Totani, N. E. Ustyuzhanina, M. I. Bilan, A. I. Usov and A. A. Grachev, PLoS One, 2011, 6, e17283 CAS.
  44. A. M. Gamal-Eldeen, E. F. Ahmed and M. A. Abo-Zeid, Food Chem. Toxicol., 2009, 47, 1378 CrossRef CAS PubMed.
  45. O. Berteau and B. Mulloy, Glycobiology, 2003, 13, 29R CrossRef CAS PubMed.
  46. K. Hayashi, T. Nakano, M. Hashimoto, K. Kanekiyo and T. Hayashi, Int. Immunopharmacol., 2008, 8, 109 CrossRef CAS PubMed.
  47. J.-H. Chen, J.-D. Lim, E.-H. Sohn, Y.-S. Choi and E.-T. Han, Parasitol. Res., 2009, 104, 245 CrossRef PubMed.
  48. M. J. Abad, L. M. Bedoya and P. Bermejo, Mini-Rev. Med. Chem., 2008, 8, 740 CrossRef CAS PubMed.
  49. S. Ananthi, H. R. B. Raghavendran, A. G. Sunil, V. Gayathri, G. Ramakrishnan and H. R. Vasanthi, Food Chem. Toxicol., 2010, 48, 187 CrossRef CAS PubMed.
  50. I. Wijesekara, R. Pangestuti and S.-K. Kim, Carbohydr. Polym., 2011, 84, 14 CrossRef CAS.
  51. H. Ye, K. Wang, C. Zhou, J. Liu and X. Zeng, Food Chem., 2008, 111, 428 CrossRef CAS PubMed.
  52. K. D. Magalhaes, L. S. Costa, G. P. Fidelis, R. M. Oliveira, L. T. D. B. Nobre, N. Dantas-Santos, R. B. G. Camara, I. R. L. Albuquerque, S. L. Cordeiro and D. A. Sabry, Int. J. Mol. Sci., 2011, 12, 3352 CrossRef CAS PubMed.
  53. R. Sokolova, S. Ermakova, S. Awada, T. Zvyagintseva and H. Kanaan, Chem. Nat. Compd., 2011, 47, 329 CrossRef CAS.
  54. L.-E. Rioux, S. L. Turgeon and M. Beaulieu, Carbohydr. Polym., 2007, 69, 530 CrossRef CAS.
  55. H. Maruyama, H. Tamauchi, M. Hashimoto and T. Nakano, In Vivo, 2002, 17, 245 Search PubMed.
  56. S. Ermakova, R. Sokolova, S.-M. Kim, B.-H. Um, V. Isakov and T. Zvyagintseva, Appl. Biochem. Biotechnol., 2011, 164, 841 CrossRef CAS PubMed.
  57. W. Wijesinghe and Y.-J. Jeon, Carbohydr. Polym., 2012, 88, 13 CrossRef CAS.
  58. R. V. Menshova, S. D. Anastyuk, S. P. Ermakova, N. M. Shevchenko, V. I. Isakov and T. N. Zvyagintseva, Carbohydr. Polym., 2015, 132, 118 CrossRef CAS PubMed.
  59. O. S. Vishchuk, S. P. Ermakova and T. N. Zvyagintseva, Food Chem., 2013, 141, 1211 CrossRef CAS PubMed.
  60. K.-J. Kim and B.-Y. Lee, Nutr. Res., 2012, 32, 439 CrossRef CAS PubMed.
  61. E. Deslandes, P. Pondaven, T. Auperin, J. Guézennec, V. Stiger and C. Payri, J. Appl. Phycol., 2000, 12, 257 CrossRef.
  62. M. Hosokawa, T. Miyashita, S. Nishikawa, S. Emi, T. Tsukui, F. Beppu, T. Okada and K. Miyashita, Arch. Biochem. Biophys., 2010, 504, 17 CrossRef CAS PubMed.
  63. B.-E. Jeong, E.-J. Ko and H.-G. Joo, Food Chem. Toxicol., 2012, 50, 1480 CrossRef CAS PubMed.
  64. H. Wang, L. Chiu, V. E. Ooi and P. O. Ang, Bot. Mar., 2010, 53, 265 CAS.
  65. A. Synytsya, W.-J. Kim, S.-M. Kim, R. Pohl, A. Synytsya, F. Kvasnička, J. Čopíková and Y. I. Park, Carbohydr. Polym., 2010, 81, 41 CrossRef CAS.
  66. S. Nakayasu, R. Soegima, K. Yamaguchi and T. Oda, Biosci., Biotechnol., Biochem., 2009, 73, 961 CrossRef CAS PubMed.
  67. Z. Jiang, T. Okimura, T. Yokose, Y. Yamasaki, K. Yamaguchi and T. Oda, J. Biosci. Bioeng., 2010, 110, 113–117 CrossRef CAS PubMed.
  68. M. L. Cho, B.-Y. Lee and S. G. You, Molecules, 2010, 16, 291 CrossRef PubMed.
  69. F. Stevan, M. Oliveira, D. Bucchi, M. Noseda, M. Iacomini and M. Duarte, J. Submicrosc. Cytol. Pathol., 2001, 33, 477 CAS.
  70. T. I. Imbs, S. P. Ermakova, O. S. Malyarenko, V. V. Isakov and T. N. Zvyagintseva, Carbohydr. Polym., 2016, 135, 162 CrossRef CAS PubMed.
  71. S. Cox, N. Abu-Ghannam and S. Gupta, Int. Food Res. J., 2010, 17, 205 CAS.
  72. C. D. Amsler and V. A. Fairhead, Adv. Bot. Res., 2005, 43, 1 Search PubMed.
  73. A. A. Alfadda and R. M. Sallam, J. Biomed. Biotechnol., 2012, 2012, 936486 Search PubMed.
  74. J. Kang, M. Khan, N. Park, J. Cho, M. Lee, H. Fujii and Y. Hong, J. Ethnopharmacol., 2008, 116, 187 CrossRef CAS PubMed.
  75. R. Koivikko, J. Loponen, K. Pihlaja and V. Jormalainen, Phytochem. Anal., 2007, 18, 326 CrossRef CAS PubMed.
  76. J. Serrano, R. Puupponen-Pimiä, A. Dauer, A. M. Aura and F. Saura-Calixto, Mol. Nutr. Food Res., 2009, 53, S310 Search PubMed.
  77. T. Shibata, S. Kawaguchi, Y. Hama, M. Inagaki, K. Yamaguchi and T. Nakamura, J. Appl. Phycol., 2004, 16, 291 CrossRef CAS.
  78. K. P. Devi, N. Suganthy, P. Kesika and S. K. Pandian, BMC Complementary Altern. Med., 2008, 8, 38 CrossRef PubMed.
  79. A. Dellai, S. Laajili, V. Le Morvan, J. Robert and A. Bouraoui, Ind. Crops Prod., 2013, 47, 252 CrossRef CAS.
  80. S. Connan, F. Delisle, E. Deslandes and E. Ar Gall, Bot. Mar., 2006, 49, 39 CAS.
  81. M. Nakai, N. Kageyama, K. Nakahara and W. Miki, Mar. Biotechnol., 2006, 8, 409 CrossRef CAS PubMed.
  82. K. Murugan and V. V. Iyer, J. Food Biochem., 2014, 38, 92 CrossRef CAS.
  83. M. Fujihara, N. Iizima, I. Yamamoto and T. Nagumo, Carbohyd. Res., 1984, 125, 97 CrossRef CAS.
  84. J.-S. Yang, Y.-J. Xie and W. He, Carbohydr. Polym., 2011, 84, 33 CrossRef CAS.
  85. S.-M. Oh, C. G. Park, J. H. Kang, E.-J. Kim, H. Y. Chee, B. H. Lee and K. B. Lee, J. Korean Soc. Appl. Biol. Chem., 2011, 54, 376 CrossRef CAS.
  86. M.-M. Kim, Q. Van Ta, E. Mendis, N. Rajapakse, W.-K. Jung, H.-G. Byun, Y.-J. Jeon and S.-K. Kim, Life Sci., 2006, 79, 1436 CrossRef CAS PubMed.
  87. P. V. Moorthi and C. Balasubramanian, J. Coastal Life Med., 2015, 3, 122 Search PubMed.
  88. A. Güner, Ç. Köksal, Ş. B. Erel, H. Kayalar, A. Nalbantsoy, A. Sukatar and N. Ü. K. Yavaşoğlu, Cytotechnology, 2015, 67, 135 CrossRef PubMed.
  89. M. El Wahidi, B. El Amraoui, M. El Amraoui and T. Bamhaoud, Ann. Pharm. Fr., 2015, 73, 190 CrossRef CAS PubMed.
  90. R.-K. Kim, N. Uddin, J.-W. Hyun, C. Kim, Y. Suh and S.-J. Lee, Toxicol. Appl. Pharmacol., 2015, 143–150 CrossRef CAS PubMed.
  91. J.-H. Ahn, Y.-I. Yang, K.-T. Lee and J.-H. Choi, J. Cancer Res. Clin. Oncol., 2015, 141, 255 CrossRef CAS PubMed.
  92. H. Yang, M. Zeng, S. Dong, Z. Liu and R. Li, Chin. J. Oceanol. Limnol., 2010, 28, 122 CrossRef CAS.
  93. H.-K. Park, I.-H. Kim, J. Kim and T.-J. Nam, Int. J. Mol. Med., 2013, 32, 291 CAS.
  94. M. Fertah, A. Belfkira, M. Taourirte and F. Brouillette, Arabian J. Chem., 2014 DOI:10.1016/j.arabjc.2014.05.003.
  95. T. Kimiya, K. Ohtani, S. Satoh, Y. Abe, Y. Ogita, H. Kawakita, H. Hamada, Y. Konishi, S. Kubota and A. Tominaga, Fish. Sci., 2008, 74, 1157 CrossRef CAS.
  96. X. L. Xu, X. Fan, F. H. Song, J. L. Zhao, L. J. Han and J. G. Shi, J. Nat. Prod., 2004, 63, 1661 CrossRef PubMed.
  97. D. Shi, F. Xu, J. Li, S. Guo, H. Su and L. Han, Zhongguo Zhongyao Zazhi, 2008, 33, 2238 CAS.
  98. D. Shi, J. Li, S. Guo, H. Su and X. Fan, Chin. J. Oceanol. Limnol., 2009, 27, 2772 Search PubMed.
  99. D. Shi, X. Li, J. Li, S. Guo, H. Su and X. Fan, Chin. J. Oceanol. Limnol., 2010, 28, 96 CrossRef CAS.
  100. G. Britton, FASEB J., 1995, 9, 1551 CAS.
  101. H. Nishino, M. Murakoshi, H. Tokuda and Y. Satomi, Arch. Biochem. Biophys., 2009, 483, 165 CrossRef CAS PubMed.
  102. Y. Nakazawa, T. Sashima, M. Hosokawa and K. Miyashita, J. Funct. Foods, 2009, 1, 88 CrossRef CAS.
  103. M. Boominathan and A. Mahesh, in Handbook of Anticancer Drugs from Marine Origin, Springer, 2015, vol. 185 Search PubMed.
  104. X. Yan, Y. Chuda, M. Suzuki and T. Nagata, Biosci., Biotechnol., Biochem., 1999, 63, 605 CrossRef CAS PubMed.
  105. A. R.-B. de Quirós, S. Frecha-Ferreiro, A. Vidal-Pérez and J. López-Hernández, Eur. Food Res. Technol., 2010, 231, 495 CrossRef.
  106. K. L. Lann, C. Ferret, E. VanMee, C. Spagnol, M. Lhuillery, C. Payri and V. Stiger-Pouvreau, Phycol. Res., 2012, 60, 37 CrossRef.
  107. M. Terasaki, A. Hirose, B. Narayan, Y. Baba, C. Kawagoe, H. Yasui, N. Saga, M. Hosokawa and K. Miyashita, J. Phycol., 2009, 45, 974 CrossRef CAS.
  108. S.-J. Heo, W.-J. Yoon, K.-N. Kim, G.-N. Ahn, S.-M. Kang, D.-H. Kang, C. Oh, W.-K. Jung and Y.-J. Jeon, Food Chem. Toxicol., 2010, 48, 2045 CrossRef CAS PubMed.
  109. J. Peng, J.-P. Yuan, C.-F. Wu and J.-H. Wang, Mar. Drugs, 2011, 9, 1806 CrossRef CAS PubMed.
  110. T. Sugawara, K. Matsubara, R. Akagi, M. Mori and T. Hirata, J. Agric. Food Chem., 2006, 54, 9805 CrossRef CAS PubMed.
  111. A. Herry Cahyana, Y. Shuto and Y. Kinoshita, Biosci., Biotechnol., Biochem., 1992, 56, 1533 CrossRef.
  112. E. Kotake-Nara, A. Asai and A. Nagao, Cancer Lett., 2005, 220, 75 CrossRef CAS PubMed.
  113. E. Kotake-Nara, M. Kushiro, H. Zhang, T. Sugawara, K. Miyashita and A. Nagao, J. Nutr., 2001, 131, 3303 CAS.
  114. E. Kotake-Nara, M. Terasaki and A. Nagao, Biosci., Biotechnol., Biochem., 2005, 69, 224 CrossRef CAS PubMed.
  115. M. Hosokawa, M. Kudo, H. Maeda, H. Kohno, T. Tanaka and K. Miyashita, BBA, Biochim. Biophys. Acta, Gen. Subj., 2004, 1675, 113 CrossRef CAS PubMed.
  116. F. Liu, J. Wang, A. K. Chang, B. Liu, L. Yang, Q. Li, P. Wang and X. Zou, Phytomedicine, 2012, 19, 797 CrossRef CAS PubMed.
  117. R.-x. Yu, X.-m. Hu, S.-q. Xu, Z.-j. Jiang and W. Yang, Eur. J. Pharmacol., 2011, 657, 10 CrossRef CAS PubMed.
  118. Z. Zhang, P. Zhang, M. Hamada, S. Takahashi, G. Xing, J. Liu and N. Sugiura, Oncol. Rep., 2008, 20, 1099 CAS.
  119. J. A. Maschek and B. J. Baker, in Algal chemical ecology, Springer, 2008, p. 1 Search PubMed.
  120. P. Reddy and S. Urban, Phytochemistry, 2009, 70, 250 CrossRef CAS PubMed.
  121. D. Abatis, C. Vagias, D. Galanakis, J. N. Norris, D. Moreau, C. Roussakis and V. Roussis, Tetrahedron Lett., 2005, 46, 8525 CrossRef CAS.
  122. C. Francisco, B. Banaigs, J. Teste and A. Cave, J. Org. Chem., 1986, 51, 1115 CrossRef CAS.
  123. O. M. Sabry, S. Andrews, K. L. McPhail, D. E. Goeger, A. Yokochi, K. T. LePage, T. F. Murray and W. H. Gerwick, J. Nat. Prod., 2005, 68, 1022 CrossRef CAS PubMed.
  124. D. M. Pereira, J. Cheel, C. Areche, A. San-Martin, J. Rovirosa, L. R. Silva, P. Valentao and P. B. Andrade, Mar. Drugs, 2011, 9, 852 CrossRef CAS PubMed.
  125. M. Murata and J.-i. Nakazoe, JARQ, 2001, 35, 281 Search PubMed.
  126. C. Dawczynski, R. Schubert and G. Jahreis, Food Chem., 2007, 103, 891 CrossRef CAS.
  127. T. Fujiwara-Arasaki, N. Mino and M. Kuroda, Hydrobiologia, 1984, 116, 513 CrossRef.
  128. H. Augier and M. Santimone, B. Soc. Phycol. FR., 1978, 23, 19 Search PubMed.
  129. J. Fleurence and R. Yada, Proteins in food processing, 2004, p. 197 Search PubMed.
  130. I.-M. Munda, Aquat. Bot., 1977, 3, 273 CrossRef CAS.
  131. R. Moo-Puc, D. Robledo and Y. Freile-Pelegrin, J. Ethnopharmacol., 2008, 120, 92 CrossRef CAS PubMed.
  132. K. Ishihara, M. Murata, M. Kaneniwa, H. Saito, K. Shinohara and M. Maeda-Yamamoto, Biosci., Biotechnol., Biochem., 1998, 62, 1412 CrossRef CAS PubMed.
  133. N. B. Perry, J. W. Blunt and M. H. Munro, J. Nat. Prod., 1991, 54, 9785 CrossRef.
  134. S.-J. Heo, K.-N. Kim, W.-J. Yoon, C. Oh, Y.-U. Choi, A. Affan, Y.-J. Lee, H.-S. Lee and D.-H. Kang, Food Chem. Toxicol., 2011, 49, 1998 CrossRef CAS PubMed.
  135. A.-H. A. Hamdy, E. A. Aboutabl, S. Sameer, A. A. Hussein, A. R. Díaz-Marrero, J. Darias and M. Cueto, Steroids, 2009, 74, 927 CrossRef CAS PubMed.
  136. K. Usmanghani, M. Shameel, S. Siddiqui and M. Alam, Pak. J. Bot., 1987, 19, 249 CAS.
  137. B. Riaz, R. Najam, I. Azhar and S. S. Khan, J. Pharma Res., 2013, 7, 215 CrossRef.
  138. M. Khanavi, R. Gheidarloo, N. Sadati, M. R. S. Ardekani, S. M. B. Nabavi, S. Tavajohi and S. N. Ostad, Pharmacogn. Mag., 2012, 8, 60 CrossRef CAS PubMed.
  139. N. Bouzidi, Y. Viano, A. Ortalo-Magné, H. Seridi, Z. Alliche, Y. Daghbouche, G. Culioli and M. El Hattab, Arabian J. Chem., 2014 DOI:10.1016/j.arabjc.2014.11.004.
  140. H. Go, H.-J. Hwang and T.-J. Nam, Toxicol. In Vitro, 2010, 24, 1546 CrossRef CAS PubMed.
  141. M. Kawasaki, M. Toyoda, R. Teshima, J. Sawada and Y. Saito, Biol. Pharm. Bull., 1994, 17, 1321 CAS.
  142. T. Gueck, A. Seidel, D. Baumann, A. Meister and H. Furhamann, Vet. Dermatol., 2004, 15, 309 CrossRef PubMed.
  143. S.-M. Kang, A.-D. Kim, S.-J. Heo, K.-N. Kim, S.-H. Lee, S.-C. Ko and Y.-J. Jeon, J. Funct. Foods, 2012, 4, 433 CrossRef CAS.
  144. N. M. Shevchenko, S. D. Anastyuk, R. V. Menshova, O. S. Vishchuk, V. I. Isakov, P. A. Zadorozhny, T. V. Sikorskaya and T. N. Zvyagintseva, Carbohydr. Polym., 2015, 121, 207 CrossRef CAS PubMed.
  145. T. Marudhupandi, T. T. A. Kumar, S. Lakshmanasenthil, G. Suja and T. Vinothkumar, Int. J. Biol. Macromol., 2015, 72, 919 CrossRef CAS PubMed.
  146. S. D. Anastyuk, N. M. Shevchenko, S. P. Ermakova, O. S. Vishchuk, E. L. Nazarenko, P. S. Dmitrenok and T. N. Zvyagintseva, Carbohydr. Polym., 2012, 87, 186 CrossRef CAS.
  147. Y.-I. Yang, S.-H. Jung, K.-T. Lee and J.-H. Choi, Int. Immunopharmacol., 2014, 23, 460 CrossRef CAS PubMed.
  148. B. Chinnababu, S. P. Reddy, P. S. Rao, V. L. Reddy, B. S. Kumar, J. V. Rao, R. Prakasham and K. S. Babu, Bioorg. Med. Chem. Lett., 2015, 25, 2479 CrossRef CAS PubMed.
  149. E. J. Kim, S. Y. Park, J.-Y. Lee and J. H. Park, BMC Gastroenterol., 2010, 10, 96 CrossRef PubMed.
  150. L. S. E. P. W. Castro, T. de Sousa Pinheiro, A. J. G. Castro, M. d. S. N. Santos, E. M. Soriano and E. L. Leite, J. Appl. Phycol., 2014, 27, 1315 CrossRef.
  151. J.-A. de la Mare, J. C. Lawson, M. T. Chiwakata, D. R. Beukes, A. L. Edkins and G. L. Blatch, Invest. New Drugs, 2012, 30, 2187 CrossRef PubMed.
  152. I. Shaik, A. Shameem and P. S. B. Rao, in Biotechnology biofortification, Springer, 2015, vol. 3 Search PubMed.
  153. B.-R. Ye, J. Kim, M.-S. Kim, J. Jang, C. Oh, D.-H. Kang, Z.-J. Qian, W.-K. Jung, I.-W. Choi and S.-J. Heo, OSJ, 2013, 48, 339 Search PubMed.
  154. B. S. Kim, H.-J. Kang, J.-Y. Park and J. Lee, Exp. Mol. Med., 2015, 47, e128 CrossRef CAS PubMed.
  155. Y. Kamei, M. Sueyoshi, K.-i. Hayashi, R. Terada and H. Nozaki, J. Antibiot., 2009, 62, 259 CrossRef CAS PubMed.
  156. J. A. Haugan, Biochem. Syst. Ecol., 1994, 22, 31 CrossRef CAS.
  157. H. M. Jamieson, FASEB J., 2013, 27, 638 Search PubMed.
  158. N. Xu, X. Fan, X. Yan and C. Tseng, J. Appl. Phycol., 2004, 16, 451 CrossRef CAS.
  159. A. Ina, K.-I. Hayashi, H. Nozaki and Y. Kamei, Int. J. Dev. Neurosci., 2007, 25, 63 CrossRef CAS PubMed.
  160. H. Maeda, M. Hosokawa, T. Sashima, N. Takahashi, T. Kawada and K. Miyashita, Int. J. Mol. Med., 2006, 18, 147 CAS.
  161. H.-J. Boo, J.-Y. Hong, S.-C. Kim, J.-I. Kang, M.-K. Kim, E.-J. Kim, J.-W. Hyun, Y.-S. Koh, E.-S. Yoo and J.-M. Kwon, Mar. Drugs, 2013, 11, 2982 CrossRef PubMed.
  162. P. D. Thinh, R. V. Menshova, S. P. Ermakova, S. D. Anastyuk, B. M. Ly and T. N. Zvyagintseva, Mar. Drugs, 2013, 11, 1456 CrossRef CAS PubMed.
  163. L. S. Costa, G. P. Fidelis, C. B. S. Telles, N. Dantas-Santos, R. B. G. Camara, S. L. Cordeiro, M. S. S. P. Costa, J. Almeida-Lima, R. F. Melo-Silveira and R. M. Oliveira, Mar. Drugs, 2011, 9, 952 CrossRef CAS PubMed.
  164. P. F. Dias, J. M. Siqueira Jr, L. F. Vendruscolo, T. de Jesus Neiva, A. R. Gagliardi, M. Maraschin and R. M. Ribeiro-do-Valle, Cancer Chemother. Pharmacol., 2005, 56, 436 CrossRef CAS PubMed.
  165. A. Cumashi, N. A. Ushakova, M. E. Preobrazhenskaya, A. D'Incecco, A. Piccoli, L. Totani, N. Tinari, G. E. Morozevich, A. E. Berman and M. I. Bilan, Glycobiology, 2007, 17, 541 CrossRef CAS PubMed.
  166. C. Zhuang, H. Itoh, T. Mizuno and H. Ito, Biosci., Biotechnol., Biochem., 1995, 59, 563 CrossRef CAS PubMed.
  167. F. Nwosu, J. Morris, V. A. Lund, D. Stewart, H. A. Ross and G. J. McDougall, Food Chem., 2011, 126, 1006 CrossRef CAS.
  168. M. Xue, Y. Ge, J. Zhang, Q. Wang, L. Hou, Y. Liu, L. Sun and Q. Li, PLoS One, 2012, 7, e43483 CAS.
  169. Y. Aisa, Y. Miyakawa, T. Nakazato, H. Shibata, K. Saito, Y. Ikeda and M. Kizaki, Am. J. Hematol., 2005, 78, 7 CrossRef CAS PubMed.
  170. H. Kawamoto, Y. Miki, T. Kimura, K. Tanaka, T. Nakagawa, M. Kawamukai and H. Matsuda, Food Sci. Technol. Res., 2006, 12, 218 CrossRef CAS.
  171. N. Takada, R. Watanabe, K. Suenaga, K. Yamada and D. Uemura, J. Nat. Prod., 2001, 64, 653 CrossRef CAS.
  172. C.-H. Kang, S.-H. Kang, S.-H. Boo, S.-Y. Park, D.-O. Moon and G.-Y. Kim, Trop. J. Pharm. Res., 2011, 10, 739 Search PubMed.
  173. J.-B. Gallé, B. Attioua, M. Kaiser, A.-M. Rusig, A. Lobstein and C. Vonthron-Sénécheau, Mar. Drugs, 2013, 11, 599 CrossRef PubMed.
  174. K. Miyashita, S. Nishikawa, F. Beppu, T. Tsukui, M. Abe and M. Hosokawa, J. Sci. Food Agric., 2011, 91, 1166 CrossRef CAS PubMed.
  175. S. R. Kumar, M. Hosokawa and K. Miyashita, Mar. Drugs, 2013, 11, 5130 CrossRef CAS PubMed.
  176. R. K. Kim, Y. Suh, K. C. Yoo, Y. H. Cui, E. Hwang, H. J. Kim, J. S. Kang, M. J. Kim, Y. Y. Lee and S. J. Lee, Cancer Sci., 2015, 106, 94 CrossRef CAS PubMed.
  177. E. G. de Mejía, G. Ramos and P. Loarca, Environ. Mol. Mutagen., 1997, 30, 346 CrossRef.
  178. E. G. de Mejía, G. Loarca-Piña and M. Ramos-Gómez, Mutat. Res., 1997, 389, 219 Search PubMed.
  179. J. S. Park, B. P. Chew, T. S. Wong, J.-X. Zhang and N. S. Magnuson, Nutr. Cancer, 1999, 33, 206 CrossRef CAS PubMed.
  180. A. Rwigemera, J. Mamelona and L. J. Martin, Cell Biol. Toxicol., 2014, 30, 157 CrossRef CAS PubMed.
  181. A. Rwigemera, J. Mamelona and L. J. Martin, Anticancer Res., 2015, 35, 207 CAS.
  182. I. Konishi, M. Hosokawa, T. Sashima, H. Kobayashi and K. Miyashita, Comp. Biochem. Physiol., Part C: Toxicol. Pharmacol., 2006, 142, 53 CrossRef PubMed.
  183. I.-M. Lee, N. R. Cook, J. E. Manson, J. E. Buring and C. H. Hennekens, J. Natl. Cancer Inst., 1999, 91, 2102 CrossRef CAS PubMed.
  184. P. Astorg, S. Gradelet, R. Bergès and M. Suschetet, Nutr. Cancer, 1997, 29, 60 CrossRef CAS PubMed.
  185. N. M. Lyons and N. M. O'Brien, J. Dermatol. Sci., 2002, 30, 73 CrossRef CAS PubMed.
  186. J. Levy, Presented at 13th Int. In Carotenoid Symp., Honolulu, 2002, vol. 135, p. 6 Search PubMed.
  187. H. Fujiki and M. Suganuma, Cancer Lett., 2012, 324, 119 CrossRef CAS PubMed.
  188. G. J. Kapadia, G. S. Rao, C. Ramachandran, A. Iida, N. Suzuki and H. Tokuda, J. Complementary Integr. Med., 2013, 10, 113 CAS.
  189. G.-S. Wu, J.-J. Lu, J.-J. Guo, M.-Q. Huang, L. Gan, X.-P. Chen and Y.-T. Wang, Pharmacol. Rep., 2013, 65, 453 CrossRef CAS PubMed.
  190. S. Y. Eid, M. Z. El-Readi and M. Wink, Phytomedicine, 2012, 19, 977 CrossRef CAS PubMed.
  191. M. Ikeguchi, M. Yamamoto, Y. Arai, Y. Maeta, K. Ashida, K. Katano, Y. Miki and T. Kimura, Oncol. Lett., 2011, 2, 319 CAS.
  192. D. Pádua, E. Rocha, D. Gargiulo and A. Ramos, Phytochem. Lett., 2015, 14, 91 CrossRef.
  193. S.-K. Kim, Handbook of Anticancer Drugs from Marine Origin, Springer, 2015, p. 185 Search PubMed.
  194. C. Murphy, S. Hotchkiss, J. Worthington and S. R. McKeown, J. Appl. Phycol., 2014, 26, 2211 CrossRef CAS.

This journal is © The Royal Society of Chemistry 2016
Click here to see how this site uses Cookies. View our privacy policy here.