You are on page 1of 21

Food &

Function
View Article Online
REVIEW View Journal

Fucoxanthin activities motivate its nano/micro-


Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

Cite this: DOI: 10.1039/d0fo02176h


encapsulation for food or nutraceutical
application: a review
Chunyan Wang,a Xing Chen, *b Yoshimasa Nakamura, c
Chenxu Yu a,d
and
Hang Qi *a

Fucoxanthin is a xanthophyll carotenoid abundant in marine brown algae. The potential therapeutic
effects of fucoxanthin on tumor intervention have been well documented, which have aroused great
interests in utilizing fucoxanthin in functional foods and nutraceuticals. However, the utilization of fucox-
anthin as a nutraceutical in food and nutrient supplements is currently limited due to its low water solubi-
lity, poor stability, and limited bioaccessibility. Nano/micro-encapsulation is a technology that can over-
come these challenges. A systematic review on the recent progresses in nano/micro-delivery systems to
encapsulate fucoxanthin in foods or nutraceuticals is warranted. This article starts with a brief introduction
of fucoxanthin and the challenges of oral delivery of fucoxanthin. Nano/micro-encapsulation technology
is then covered, including materials and strategies for constructing the delivery system. Finally, future pro-
spective has been discussed on properly designed oral delivery systems of fucoxanthin for managing
cancer. Natural edible materials such as whey protein, casein, zein, gelatin, and starch have been success-
fully utilized to fabricate lipid-based, gel-based, or emulsion-based delivery systems, molecular nano-
complexes, and biopolymer nanoparticles with the aid of advanced processing techniques, such as
freeze-drying, high pressure homogenization, sonication, anti-solvent precipitation, coacervation, ion
crosslinking, ionic gelation, emulsification, and enzymatic conjugation. These formulated nano/micro-
Received 18th August 2020, capsules have proven to be effective in stabilizing and enhancing the bioaccessibility of fucoxanthin. This
Accepted 21st October 2020
review will inspire a surge of multidisciplinary research in a broader community of foods and motivate
DOI: 10.1039/d0fo02176h material scientists and researchers to focus on nano/micro-encapsulated fucoxanthin in order to facilitate
rsc.li/food-function the commercialization of orally-deliverable tumor intervention products.

1. Introduction including ocular-protective, anti-obesity, and tumor intervention


effects.2–4 However, due to the difficulty in achieving cost-effective-
Fucoxanthin is a natural pigment carotenoid, which is unique in ness in the production and extraction of fucoxanthin biomass, the
marine brown algae.1 Its structural formula is shown in Fig. 1, production of fucoxanthin from macroalgae or microalgae is
including an unusual allenic bond, a 5,6-monoepoxide, and seven limited despite its high proportion in natural carotenoids.
conjugated double bonds. Similar to lutein, another popular caro- Tumor is a major public health problem, with an estimated
tenoid from microalgae, fucoxanthin can give food an orange 9.6 million deaths in 2018 worldwide, in which, China
color. In recent years, it has been shown that consuming fucox- accounts for almost a quarter of the total.7 It has been esti-
anthin-rich food is associated with multiple health benefits, mated that the number of worldwide tumor-related deaths will
increase to 11.4 million by 2030.8,9 Lung tumor and breast
tumor are the most common cancers, followed by prostate
a
School of Food Science and Technology, Dalian Polytechnic University, National
tumor and colorectal tumor.7 Moreover, the most common
Engineering Research Center of Seafood, Dalian 116034, P. R. China.
E-mail: xingchen@jiangnan.edu.cn; Tel: +86 510 85197367
causes of death related to tumor are also lung tumor in men
b
State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, and breast tumor in women. Tumor is a huge burden on
Jiangsu 214122, P.R. China. E-mail: qihang@dlpu.edu.cn; Fax: +86411 86323262; public health and economy around the world. One obvious
Tel: +86411 86318785 phenomenon is that the incidence rate and mortality rate of
c
Graduate School of Environmental and Life Science, Okayama University, Okayama,
tumor are relatively low in countries with advanced environ-
Japan
d
Department of Agricultural and Biosystems Engineering, Iowa State University,
ment, economy, and healthcare.9 It was also confirmed by the
Ames, IA, USA report of World Health Organization that Asia has the most

This journal is © The Royal Society of Chemistry 2020 Food Funct.


View Article Online

Review Food & Function


Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

Fig. 1 Fucoxanthin’s origin and its representative molecular structure and metabolism.

cancer patients and the highest case fatality rate, followed by there.10 Prevention and early diagnosis and intervention has
Europe and America. A notable trend in the past few decades been shown to be the most effective way of reducing tumor-
has seen a decline in tumors attributable to environmental related deaths as most tumors have become treatable due to
pollution in developed countries, while the opposite is true for the advancement of medical sciences.11
developing countries as industrialization and population Diet plays an important role in tumor intervention and
growth have brought more pollution exposure to people treatment. The oral administration of nutraceuticals or func-

Food Funct. This journal is © The Royal Society of Chemistry 2020


View Article Online

Food & Function Review

tional foods containing bioactive ingredients could be instru- tenoids, accounting for more than 10% of the estimated total
mental in improving human physical or mental health, which production of carotenoids on earth.16 This yellowish-brown
would in turn help with tumor intervention and treatment. For pigment exhibits a broad range of protective functionalities,
this reason, there have been considerable efforts in the mining including anti-oxidant, anti-obesity, anti-diabetic, anti-cancer,
of bioactive agents including carotenoids, polyphenols, flavo- anti-inflammatory, hepato-protective, skin-protective, anti-
noids, essential fatty acids, peptides, and glucosinolates from angiogenic, cerebrovascular, bone-protective, ocular-protective,
natural edible materials and incorporating them into foods, cardiovascular-protective, and anti-malarial activities.3 As a
nutrient supplements, and pharmaceuticals in the last xanthophyll carotenoid extracted from marine algae (Table 1),
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

decade.12–15 Fucoxanthin is the most abundant of natural caro- fucoxanthin is safe for animals and humans.17 Although fucox-

Table 1 Selected examples of fucoxanthin extraction and purification process from marine algae

Macroalgae/microalgae Extraction process Ref.

Laminaria japonica, Undaria ● The algae were first extracted using microwave-assisted extraction, then the Xiao et al., (2012)1
pinnatifida, Sargassum fusiforme dried extract was dissolved and directly introduced into the high-speed
countercurrent chromatography system with a two-phase solvent system
consisting of hexane–ethyl acetate–ethanol–water (5 : 5 : 6 : 4, v/v/v/v).
Phaeodactylum tricornutum ● Fucoxanthin was isolated and structurally identified through Kim et al., (2012)17
chromatographic and spectroscopic methods, including 100% ethanol extracts,
LC-MS and HPLC analysis, isolation by silica gel chromatography, purification
by NMR, and quantification by reverse-phase high-performance liquid
chromatography.
Isochrysis zhangjiangensis ● Lyophilized biomass was extracted with pure methanol. The methanol layer Li et al., (2019)113
was collected by centrifugation at 5000 rpm for 10 min. The extracts were
centrifuged for 15 min at 18 000 rpm before subjecting to high performance
liquid chromatography (HPLC) analysis.
Sargassum thunbergii ● The powder was extracted three times with 90% methanol at 40 °C for 3 h, Wang et al., (2017)25
then purified by column chromatography with silica gel and further purified by
HPLC.
Nitzschia laevis ● The lyophilized powder was ground with liquid nitrogen. Ethanol was added Guo et al., (2019)24
to the ground cells for extraction, followed by shaking for 1 h. Supernatants
were collected by centrifugation. Extracts was obtained by evaporating with
nitrogen at 22 °C.
Phaeodactylum tricornutum ● Pigments were extracted from harvested cells by several methanol extractions Staleva-Musto et al.,
with brief (1.5 min) sonication. Crude pigment extracts were pooled, dried (2018)23
under vacuum, and dissolved in methanol for HPLC purification.
Fucus vesiculosus ● Fucoxanthin extraction using enzymes (viscozyme), water, low-temperature Shannon & Abu-
(40 °C for 12 h) dehydration, and mechanical blending (optimum extraction Ghannam, (2018)114
parameters: enzyme-to-water ratio 0.52%, seaweed-to-water ratio 5.37%, and
enzyme incubation time 3.05 h).
Phaeodactylum tricornutum ● Fucoxanthin was extracted from the disrupted biomass by pressurized liquid Neumann et al.,
extraction for a static extraction time of 20 min at 100 °C and 100 bar using (2019)22
adequate subcritical organic solvents. Fucoxanthin was purified by multiple
separation steps using filters (0.25 µm) consisting of polytetrafluoroethylene to
a final purity of 99.2% (w/w) (HPLC).
Sargassum wightii Greville ● The dried powder was sequentially extracted with hexane and ethyl acetate Raji et al., (2020)29
for 6 h. The ethyl acetate extract was fractionated by silica column
chromatography to obtain fucoxanthin-rich fraction. The fraction was dissolved
in ethyl acetate, centrifuged to remove silica, and concentrated by the vacuum
rotary evaporator under reduced pressure to obtain fucoxanthin.
Undaria pinnatifida ● Dried samples were ground into a fine powder and mixed with 75% (v/v) Liu et al., (2019)27
ethanol. After three extractions, the crude extracts were purified using
macroporous resins. Fucoxanthin was finally collected, concentrated in vacuo,
and then stored at −20 °C.
Phaeodactylum tricornutum ● Wet biomass was loaded into the mixing extraction vessel of the subcritical Aslanbay Guler et al.,
fluid extraction system and operated in the constant pressure mode to supply (2019)16
methanol with heating until the pressure was increased up to 20 MPa. The
extracted samples were collected and centrifuged at 6000 rpm for 10 min.
Fucoxanthin was finally collected after rotary evaporation.
Chaetoceros gracilis ● Freeze-dried algal samples was suspended in methanol and shaken overnight Peraman &
at about 100 rpm and filtered. The residues were re-extracted under the same Nachimuthu, (2019)28
conditions. The contents were stored for a period of 5 days at room temperature
(25 ± 2 °C) and then filtered. The solvent was removed under vacuum to
produce the dry crude extract.
Nitzschia laevis ● Fucoxanthin was determined by ultra-performance liquid chromatography Sun et al., (2018)20
coupled to photodiode array detector-quadrupole/travelling-wave ion mobility
mass spectrometry/time-of-flight mass spectrometry. Furthermore, solid-phase
extraction and thin-layer chromatography techniques were used to isolate and
purify fucoxanthinol and fucoxanthin from the extracts of N. laevis.

This journal is © The Royal Society of Chemistry 2020 Food Funct.


View Article Online

Review Food & Function

anthin can be produced by both macroalgae and microalgae, released from the food matrix to be incorporated into mixed
including Laminaria japonica, Undaria pinnatifida, and lipid micelles.37,38 Meanwhile, fucoxanthin can be hydrolyzed
Sargassam fusiforme, as well as Phaeodactylum tricornutum, into fucoxanthinol (Fig. 1) in the gastrointestinal tract and is
Odontella aurita, Isochrysis galbana, and Nitzschia laevis,18–21 further converted into amarouciaxanthin A (Fig. 1) in the
which is much richer in some microalgae, such as liver.39,40 Similar to the other carotenoids, the absorption of
Phaeodactylum tricornutum.22 In the same line, the production fucoxanthin may also occur in the intestinal mucosal epithelial
of fucoxanthin from microalgae is economically feasible due cells from the proximal half of the small intestine through the
to the advantages of high fucoxanthin content and extraction facilitated diffusion of scavenger receptor B1 (SR-B1).41,42
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

efficiency with a short growth cycle.23,24 Furthermore, micro- However, pharmacokinetic studies have showed that after
algae could be cultivated in bioreactors with a high propa- fucoxanthin administration (a single oral dose of 31 mg or
gation rate and good control.17 Table 1 shows several kinds of 6.1 mg day−1 for a week), only fucoxanthinol was found in the
extraction methods for fucoxanthin from different marine circulation of human bodies, while both fucoxanthinol and
algae. The unique allenic bond structure of fucoxanthin is amarouciaxanthin A were found in the plasma and other
unstable and can be easily damaged by heating, aerial tissues of mice.43,44 It demonstrated that the metabolic rate of
exposure, or illumination.25 However, it is the structure of fucoxanthin in mice was faster than that in humans, which
allenic bond in fucoxanthin that gives it anti-oxidative, anti- should be considered when conducting studies for the biologi-
inflammatory, anti-obesity, selective bacteriostasis, and anti- cal activities of fucoxanthin in mice. It was recently found that
cancer activities.22,26–29 degradation components (apo-13-fucoxanthinone and apo-9′-
Despite the potential health benefits of fucoxanthin, its fucoxanthinone) of fucoxanthin obtained by ozonolysis had
utilization as a nutraceutical ingredient by the food and nutra- the same anti-proliferative effect against Caco-2 cells as fucox-
ceutical industry is currently limited due to its poor stability anthin itself as long as the allene structure remained intact.45
and low bioaccessibility caused by aqueous insolubility. All of Moreover, fucoxanthin seemed to exert its biological effect in
these characteristics are interrelated and associated with different tissues as different forms of metabolites. For
fucoxanthin’s molecular hydrophobicity, unstable chemical instance, after the administration of dietary fucoxanthin sup-
structure, and its low release from food substrates or their plementation (including: 160 nmol as a single oral dose,
complexes.22,30 160 nmol day−1 for a week, and 128 nmol day−1 for 14 days) in
When it is difficult to directly introduce functional ingredi- mice, fucoxanthinol and amarouciaxanthin A but not fucox-
ents into food, nano/micro-encapsulation-based approaches, anthin were found in the plasma and different tissues.40,43,44
in which nanocarriers are utilized as delivery systems, could be It is clear that oral fucoxanthin is easy to degrade in the
sought as alternatives. In recent years, several kinds of nano- stomach and thus lose its biological activity, or be hydrolyzed
carriers have been developed to encapsulate, protect, trans- to produce key functional derivatives that can be absorbed by
port, and eventually release fucoxanthin, and to overcome the small intestine, thus entering the liver metabolism and
some of the limitations currently compromising fucoxanthin blood circulation system to play an anti-cancer role. But such a
utilization.5,31–34 To date, relevant discussions on fucoxanthin direct oral administration of pure free fucoxanthin may lead to
mostly center on its bioactivity related to human health or the a reduced absorption rate due to its degradation in the diges-
benefits of its direct oral administration (including combined tive tract; and eating kelp may also have a low absorption rate
use with other drugs) to human health.35,36 There is no com- due to its low fucoxanthin content (compared with pure fucox-
prehensive review on how the bioaccessibility issue can be anthin), which seriously restricts the tumor intervention
addressed to improve the functional bioactivity of fucoxanthin efficacy of diet fucoxanthin.
with nano/micro-encapsulation approaches. This review is
aimed at filling this void. The challenges of fucoxanthin deliv- 2.2. Tumor intervention efficacy
ery are discussed as well as the current knowledge on utilizing The tumor intervention efficacy of fucoxanthin has been
nano/micro-encapsulation in order to enhance its bioaccessi- reported in various tumor models.35 Fig. 2 shows several
bility. Furthermore, future trends regarding the nano/micro- reported mechanistic pathways for the tumor intervention
encapsulation systems of fucaxanthin and their potential use efficacy of fucoxanthin. Fucoxanthin can exert effects on cell
as a tumor intervention ingredient in functional foods, nutra- cycle arrest and apoptosis, which are related to tumor interven-
ceuticals, or nutrient supplements are prospected. tion and treatment.46 Moreover, fucoxanthin is an anti-angio-
genic agent, which could suppress the expression of pro-angio-
genic factor (FGF-2), trans-activation factor, and their receptor,
2. Tumor intervention efficacy of hence contributing to the inhibition of tumor metastasis.47,48
fucoxanthin Hereby, different regulatory and protective mechanisms or
related signal transduction pathways of fucoxanthin and its
2.1. Metabolism of fucoxanthin metabolites are discussed in detail.
The digestion, absorption, and transportation of fucoxanthin Fucoxanthin and, to a greater extent, its metabolite fucox-
is a systemic and complicated course. As a hydrophobic and anthinol, have been shown to reduce the cell viability or
lipid-soluble carotenoid, ingested fucoxanthin should be inhibit cell proliferation by inducing cell cycle arrest in human

Food Funct. This journal is © The Royal Society of Chemistry 2020


View Article Online

Food & Function Review


Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

Fig. 2 Several reported mechanistic pathways of the tumor intervention efficacy of fucoxanthin.

skin tumor cells (B16-F10),49 human osteosarcoma cells (Saos- bladder tumor cell line cells (T24),56 and human cervical
2, LM8, and 143B),50 human primary effusion lymphoma tumor cells (SiHa).59 Fucoxanthin and fucoxanthinol can also
cells,51 human gastric adenocarcinoma cells (SGC-7901 and suppress angiogenesis in embryo blood vessels. Fucoxanthin
BGC-823),52 human liver tumor cells (HepG2),46 human non- at concentrations over 10 µM was shown to inhibit the prolifer-
small cell lung tumor cells (A549 and H1299),53 human colon ation and tube formation of human umbilical vein endothelial
adenocarcinoma cells (DLD-1, Caco-2 and HT-29),54 human cells, and fucoxanthin in the concentration range of 10–20 µM
prostate cancer cells (LNCaP, PC-3 and DU 145),55 and human could also inhibit the differentiation of endothelial progenitor
bladder cancer cells (T24).56 The inductive effect of fucox- cells into endothelial cells, which was associated with new
anthin on cell cycle arrest was mediated by increased blood vessel formation in the tumor.60 Furthermore, studies
expressions of cyclin-dependent kinase-inhibitory protein have shown that fucoxanthin could inhibit the proliferation of
( p21), and decreased expressions of cyclin-dependent kinase-4 murine astroglioma cells by inhibiting the expression of angio-
and cyclin Ein human osteosarcoma and bladder tumor cells. genic indicators such as vascular endothelial growth factor
It could also increase gap junction intercellular communi- and epidermal growth factor receptor.47,48
cation between the tumor cells, which could then result in In addition, the inhibitory effect of fucoxanthin on the
increased intracellular signaling, leading to cell cycle arrest migration or invasion of tumor cells might be related to its
and apoptosis.46,50 anti-angiogenic activity. A previous study has shown that fucox-
In fact, fucoxanthin and its metabolites could induce the anthin could down-regulate the phosphorylation of FGF-2
apoptosis of cells by reducing the expression of anti-apoptotic caused by extracellular signal-regulated kinase and protein
protein (Bcl-2, Bcl-xL, XIAP, and survivin), anti-apoptotic tran- kinase B in endothelial cells.48 Moreover, fucoxanthin could
scription factor (signal transducer and activator of transcrip- suppress the migration of human glioma tumor cells by redu-
tion 3, STAT3), and their phosphorylated active forms, while cing the expression of matrix metalloproteinases 2 and −9,
increasing the expression of pro-apoptotic protein (Bax) and which play vital roles in tumor invasion and metastasis.57
activating the caspases family (caspases-3, -8, and -9) in Consistently, fucoxanthin has been found to reduce the
human cancer cells, which has been discovered so far in migration and invasion of melanoma cells (B16-F10) in mice
human glioma tumor cells (U87 and U251),57 human gastric by reducing the expression of matrix metalloproteinases 9.61
adenocarcinoma cells (SGC-7901 and BGC-823),52 human liver Therefore, fucoxanthin and fucoxanthinol could be effective in
tumor cells (HepG2),46 human non-small cell lung tumor cells tumor intervention and management. However, there are still
(A549 and H1299),53 human colon cells (Caco-2),58 human some serious challenges such as how to stably and efficiently

This journal is © The Royal Society of Chemistry 2020 Food Funct.


View Article Online

Review Food & Function

deliver fucoxanthin to the tumor site and then exert its anti- based on its digestion, absorption, metabolism, distribution,
cancer activity. and bioactivity.5 The evaluation of bioaccessibility is usually
done in a simulated gastrointestinal digestion model in vitro
that is relatively simple, inexpensive, and repeatable.68–71 The
3. Challenges in the oral delivery of evaluation of bioaccessibility and bioavailability of fucoxanthin
fucoxanthin is helpful to understand the interaction between fucoxanthin
and food components, and the effects of pH, enzymes, and
The annual production of fucoxanthin from brown algae and food processing on its absorption rate or absorption poten-
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

microalgae is on the rise, and its potential utilization as a tial.62 Fucoxanthin, as a non-polar and hydrophobic marine
tumor nutraceutical and/or preventive agent is getting more phytochemical, is not water soluble, which leads to its low
and more attention from the food and pharmaceutical indus- bioaccessibility. In fact, fucoxanthin can form aggregates by
tries.17 Oral administration of free fucoxanthin is not effective intermolecular bonding due to the thermodynamic inaccessi-
due to its poor solubility and gastrointestinal stability, leading bility between its non-polar groups and water.72 The bioacces-
to its reduced bioaccessibility.43,45 Moreover, the processing sibility of fucoxanthin may be influenced by the food matrix
and storage instability of fucoxanthin tends to influence its and the gut microbiota composition based on various diets.26
utilization in functional foods and nutraceuticals as well.62 For instance, fucoxanthin may bind to proteins or dietary
fibers in the food matrix and be decomposed and utilized by
3.1. Instability some intestinal microorganisms, which may be excreted via
The issues of instability of fucoxanthin can be analyzed from faeces, thus reducing the bioaccessibility of fucoxanthin in
two aspects—processing and storage instability, and gastroin- gastrointestinal fluids.26,73 Moreover, fucoxanthin can promote
testinal digestion instability. As previously mentioned, the the growth of Escherichia coli and Lactobacilli, the main micro-
unique allenic bond structure of the fucoxanthin molecule is organisms in the gut, and can be deacetylated by these two
unstable and could be easily damaged by heating as well as microorganisms in turn to produce fucoxanthinol, which may
oxygen, acid, and light exposure,17,25 which makes fucoxanthin increase its bioaccessibility.27 For tumor patients, their diges-
susceptible to unfavorable environmental exposures. Fung, tive tract could be affected by the tumor so that the digestion,
Hamid, and Lu (2013)63 found that the fucoxanthin content metabolism, absorption, and biodistribution of fucoxanthin
and anti-oxidant activity of processed (rinsing, cutting, blanch- could be changed, and the intestinal flora related to fucox-
ing, salting, and oven drying) Undaria pinnatifida were lower anthin metabolism could be disordered as well, thus affecting
than that of the freeze-dried unprocessed ones. It was reported its bioaccessibility.5,27 In addition, the transportation and
that with the increase in temperature (2 °C, 10 °C, 26 °C), the absorption of the ingested fucoxanthin may be mediated
stability of fucoxanthin in water decreased significantly, i.e., through the facilitated diffusion of SR-B1 in the intestinal
after 4 weeks of storage at 26 °C, the content of fucoxanthin in tract.41 Hence, the absorption rate of fucoxanthin in the intes-
water degraded to about 30% of the initial amount, indicating tinal epithelium cells could also influence the bioaccessibility
the instability of fucoxanthin in water.5 Moreover, fucoxanthin of fucoxanthin, which may in turn cause a difference of its
may also interact with other components of the food matrix, effective concentration of fucoxanthin in the circulatory
which could also alter its stability. For example, fucoxanthin is system, thus affecting its ultimate protective effects. Although
prone to degradation in the food matrix containing transition not proven, some possible hydrophilic metabolites of fucox-
metals, while it is more stable in a food matrix containing anthin produced during biotransformation may be excreted
milk protein.64–66 As we already discussed, the metabolism with urine, reducing the effective concentration of fucoxanthin
and biotransformation of fucoxanthin released from the food in the target tissues.42 Such types of biotransformation are
matrix mainly occurs in the gastrointestinal tract and the liver more likely to occur during the oral administration of fucox-
after hydrolysis, dehydrogenation, and isomerization.37,38,67 In anthin and further lower its bioaccessibility.
the gastrointestinal environment, fucoxanthin is extremely sus-
ceptible to degradation or transformation. The acidic environ-
ment in the stomach and the cholesterol esterase and lipase 4. Nano/micro-encapsulation
located in the gut play key roles in the degradation and bio- technology for fucoxanthin (Table 2)
transformation of fucoxanthin. Since the bioaccessibility and
potential protective effects of fucoxanthin are dependent on its In recent decades, the application of encapsulation technology
physical and chemical structure, they could change notably to enhance the stability, solubility, and bioaccessibility of bio-
after metabolic transformation.41,42 active compounds has seen an exponential growth, fueled by
the rapid progress of key enabling technologies such as micro-
3.2. Insolubility and low bioaccessibility/bioavailability nano technology, industrial biotechnology, advanced
The bioaccessibility of the ingested bioactive compound is materials, and advanced manufacturing technologies. Silva
evaluated based on the ease of its release from the food matrix et al. (2017)74 found that the water solubility of lutein and its
and subsequent absorption through the digestive tract. The efficacy in improving murine declarative memory were
bioavailability of the ingested bioactive compound is evaluated increased by PVP nanoparticle delivery systems, which were

Food Funct. This journal is © The Royal Society of Chemistry 2020


View Article Online

Food & Function Review

Table 2 Encapsulation systems of fucoxanthin

Application Encapsulation Encapsulation


fields Encapsulation materials system techniques Results Ref.

Food Casein/chitosan Biopolymer Freeze drying and Improved bioavailability of Koo et al.,
nanoparticles ionic gelation fucoxanthin (2016)5
Food Zein/caseinate Biopolymer Anti-solvent Improved stability, bioaccessibility, Li et al.,
nanoparticles precipitation anti-oxidant activity, and anti- (2018)30
proliferative activity of fucoxanthin
Food Gum arabic/gelatin/ Gel-based delivery Freeze drying Improved gastrointestinal stability Li et al.,
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

alginate hydrogel system and oral efficacy of fucoxanthin (2019)21


Food Medium-chain o/w nanoemulsion- High pressure Improved stability and Ma et al.,
triacylglycerol/whey based delivery homogenization bioaccessibility of fucoxanthin (2019)33
protein isolate system
Food Medium-chain o/w Microchannel Improved chemical stability and Ma et al.,
triacylglycerol/whey microemulsion- emulsification sustained release of fucoxanthin (2020)93
protein isolate/modified based delivery
lecithin/Tw20 system
Food Seaweed oil/sunflower o/w nanoemulsion- Ultrasound-assisted Improved encapsulation efficiency Saravana et al.
oil/κ-carrageenan based delivery emulsification (98.99%), stability, and (2019)108
system bioaccessibility of fucoxanthin, and
good cell inhibition against a few
cancer cell lines
Food Porous starch/ Molecular Solvent exchange/ Improved the storage stability of Oliyaei et al.,
maltodextrin/gum arabic nanocomplexes freeze drying/vacuum fucoxanthin (2020)86
drying
Food Whey protein isolate/ Molecular Complex coacervation Improved the storage and simulated Zhu et al.,
lysozyme nanocomplexes gastrointestinal stability of (2019)78
fucoxanthin
Food Whey proteins Molecular Molecular self- Improved The stability and water Zhu et al.,
nanocomplexes assembly solubility of fucoxanthin (2017)32
Food Cetylpalmitate-based Lipid-based Ultrasonic and freeze Improved stability and sustained Quan et al.,
solid lipid core and fish delivery system drying release of fucoxanthin (2013)85
gelatin–gum arabic
coacervate shell
Food Chitosan/glycolipid Gel-based delivery Ionic gelation and Improved stability, bioaccessibility, Ravi &
system freeze drying and anti-tumor activity of Baskaran,
fucoxanthin (2015);31 Ravi
et al., (2018)58
Cosmetics Self-emulsifying wax/ Lipid-based High pressure Improved stability and Tavares et al.,
liquid polymer/UV filters delivery system homogenization and photoprotective potential of (2020)82
emulsification sunscreens
Food and Palm stearin/fish gelatin/ Lipid-based Ultrasonic, Improved the storage stability and Wang et al.,
drugs gum arabic delivery system emulsification and sustained release of fucoxanthin (2017)25
freeze drying

prepared using solvent evaporation. Chen et al. (2019)71 con- ture of the entrapped compound; (2) to facilitate the incorpor-
structed zein/carrageenan core–shell nanoparticles based on ation of multiple mutually incompatible compounds; (3) to
an anti-solvent precipitation method, which enhanced the protect the bioactive compound from being degraded by
bioaccessibility and provided excellent protection through cur- environmental factors such as sunlight, oxygen, and heat; (4)
cumin against simulated gastrointestinal environments. Liu to increase the bioaccessibility of the bioactive compound; (5)
et al. (2018)75 successfully prepared β-lactoglobulin/chitosan to control the rate or location of the release of the bioactive
oligosaccharides nanocomplexes by spontaneous co-assembly compound.76,77 Apart from functional food applications,
and freeze-drying to improve the stability and anti-oxidant nanoencapsulated products are also widely used in pharma-
activity of astaxanthin with high effectiveness. All these reports ceuticals, cosmetics, healthcare, and other related industries.
suggested that suitable encapsulation systems could be essen-
tial to improve the bioaccessibility and thereby the in vivo 4.1. Natural materials for fucoxanthin nano/micro-
efficacy of fucoxanthin. With a reliable encapsulation system, encapsulation
fucoxanthin can be delivered to the target without losing its The encapsulating material plays an important role in the
active structure in the process of product manufacturing, efficiency of entrapping fucoxanthin. Various materials, such
transport and storage, as well as metabolism and as semiconductor, inorganic salts, as well as artificial and
biotransformation.5 natural polymers, have been explored as nanoencapsulating
In general, the nano/micro-encapsulation of a hydrophobic carriers in a series of ordered and delicate bottom-up nano/
bioactive compound such as fucoxanthin is usually carried out micro-encapsulation methods.4,34,78 Among them, edible col-
due to the following reasons: (1) to preserve the physical struc- loids and polymers from natural materials have been gaining

This journal is © The Royal Society of Chemistry 2020 Food Funct.


View Article Online

Review Food & Function

popularity, especially in the food sector, due to strict regulat- by Trp213, Leu237, Val240, Leu259, Ala260, and Ala290 of BSA
ory requirements. Proteins and polysaccharides, including (Fig. 3A and D). Fucoxanthin could also bind to the top of the
whey protein, casein, zein, alginate, chitosan, gum arabic, and calyx of β-Lg through van der Waals force, hydrogen bond
gelatin, have been confirmed as safe nanoencapsulating car- (Asp28), and hydrophobic force (Leu31, Pro38, Ile71, Ile72,
riers for fucoxanthin for improving its bioaccessibility.79 The Ile84) (Fig. 3B and E). In the case of α-La, residues of Phe31,
best performing ones are discussed in detail below. Tyr36, Ile41, Val42, and Trp118 were shown to be involved in
Whey protein. Nowadays, there is a considerable interest in the formation of α-La-fucoxanthin nanocomplex mainly via
the application of whey protein for the encapsulation and con- hydrophobic interaction (Fig. 3C and F). The binding affinities
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

trolled release of fucoxanthin. Whey protein ( particularly, the of various WPI components to fucoxanthin showed a decreas-
whey protein isolate, WPI) extracted from milk is well known ing order as BSA > β-Lg > α-La. In another report, nanocom-
for its high nutritional value and ease of digestion and absorp- plexes composed of fucoxanthin, oleic acid, and BSA were
tion. It is one of the most widely commercialized high-quality created and characterized by Liu et al. (2019).4 According to
protein supplements for humans, containing bovine serum their results, fucoxanthin molecules were bound at the sites of
albumin (BSA), β-lactoglobulin (β-Lg), α-lactoalbumin (α-La), the hydrophobic cavity of BSA with amino acid residues
lactoferrin, immunoglobulin, and many other active ingredi- 183–291 through intermolecular π–π interactions, van der
ents. WPI is also widely used in food, pharmaceutical, and Waals forces, and hydrogen bonds, while oleic acid was bound
other industries due to its various physiological functions.4,80 at the region of amino acid residues 115–185 through van der
In addition to its nutritional value, whey proteins can also Waals forces. Fucoxanthin and oleic acid were bound to BSA at
serve as important transporters in the body, which are involved completely different sites, while the presence of oleic acid
in the absorption and transport of a variety of small molecule could strengthen the bonding of fucoxanthin to the hydro-
nutrients and amino acids in cells.81 The existence of hydro- phobic cavities of the BSA. As a conclusion, WPI could be
phobic cavities in whey protein molecules makes WPI a good effectively utilized to encapsulate fucoxanthin in food systems.
nano-carrier for hydrophobic nutrients.79 Nanocomplexes of Casein. As a major protein family in milk, caseins also
fucoxanthin binding to BSA, β-Lg, and α-La have been success- demonstrated the same ligand binding properties as well as
fully constructed and characterized (diameter size < 300 nm).32 the self-assembly ability as that of WPI. The potential of casein
Molecular docking results suggested that fucoxanthin mole- micelles as a delivery system for bioactive compounds and
cules can bind tightly to the Sudlow sites I of BSA where they nutraceuticals has been showcased in either self-assembled
form hydrogen bonds with the Tyr149 and Arg256 residues of micelles from casein fractions (mostly β-casein) or reas-
BSA, and interacted with the hydrophobic pocket constituted sembled micelles from commercial caseins (caseinates), which

Fig. 3 TEM photographs and molecular docking of fucoxanthin into BSA (A), β-Lg (B), and a-La (C). [BSA] = [β-Lg] = [α-La] = 5 mM, [fucoxanthin] =
15 mM. The magnification of TEM photographs is 300 000 in BSA, 400 000 in β-Lg, and 200 000 in α-La. For molecular docking, whey proteins are
showed as cartoon colored in cyan (helix), magenta (sheet), and salmon (loop). The spherical model displays fucoxanthin colored as orange. Details
of protein residues interacting with fucoxanthin in BSA (D), β-Lg (E), and a-La (F) are also shown. The stick models represent fucoxanthin and key
amino acid residues, colored as orange and green, respectively. Dashed line colored as yellow shows the hydrogen bond (reproduced with per-
mission from Zhu et al., 2017;32 refer to this text for more details).

Food Funct. This journal is © The Royal Society of Chemistry 2020


View Article Online

Food & Function Review

can protect the entrapped substances against environmentally cells is reduced.85 These shortcomings severely limit the tra-
induced degradation and oxidation.82,83 The unique properties ditional application of gelatin/gum arabic microcapsule-based
of caseins and the casein micelles, and how these properties carriers for the oral delivery of various functional active ingre-
can enhance the successful delivery of sensitive ingredients dients. To solve such problems, a novel delivery system fabri-
and bioactives, have been well covered in a previous review.83 A cated by the complex coacervation of gelatin/gum arabic micro-
recent study has showed that casein could be combined with capsules and alginate hydrogel beads has been developed as
chitosan to generate fucoxanthin-loaded nanoparticles.5 The specialized oral vehicles of fucoxanthin. The results of simu-
casein-based nanoparticles were mixed micelles and vesicles lated gastrointestinal digestion in vitro indicated that the
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

containing fucoxanthin in the micelle phases, and fucoxanthin obtained beads could protect fucoxanthin while being incu-
could be released more easily from the matrices, resulting in bated in simulated gastric fluid and the amount of fucox-
better fucoxanthin bioaccessibility compared to the free ones. anthin released in the simulated intestinal fluid was more
In addition, due to the increase in the adsorption capacity of than 90%. In vivo experiments on mice have shown that the
chitosan to mucin, the chitosan coated casein-based nano- encapsulated fucoxanthin could more effectively lower the
particles had a higher accumulation of fucoxanthinol in the blood lipid and oxidative stress level compared to the free
blood circulation of C57BL/6 mice. As a conclusion, caseins ones.21
could also be effectively facilitated to serve as fucoxanthin car- Porous starch. Porous starch is a new type of modified
riers in order to enhance its water solubility and starch with a honeycomb structure. After natural raw starch is
bioaccessibility. hydrolyzed, small pores (about 1 µm in diameter) are formed
Zein. Zein, a protein derived from maize seeds (Zea mays L.), on the surfaces of the starch granules. These small pores
has been approved for oral use by the US Food and Drug extend to the inside of the granules, which can then entrap
Administration (FDA). Due to its inherent hydrophobicity, zein various substances in them. In porous starch, the porosity
can self-assemble into a variety of structures through a simple accounts for about 50% of the particle volume. Thus, the
process commonly known as anti-solvent precipitation. porous starch particles have a good carrying capacity and can
Besides, zein is resistant to digestive enzymes in the gastroin- be used as a new type of organic embedding material for the
testinal tract and zein colloidal particles can deliver a con- encapsulation of various functional active ingredients. For
trolled release of functional components loaded in them. fucoxanthin, porous starch carriers are on the rise because of
These characteristics have made zein a great candidate to their large pore size, high specific surface area, and ease of
generate encapsulating carriers for a variety of bioactive com- synthesis.86 Oliyaei et al. (2020)86 produced a fucoxanthin-
pounds.84 Fucoxanthin-loaded zein/caseinate nanoparticles loaded delivery system based on porous starch, maltodextrin,
have been successfully fabricated where zein interacted with and gum arabic by using methods of solvent exchange and
fucoxanthin through hydrophobic interactions.30 The obtained freeze drying. According to the results, the storage stability
nanoparticles were stabilized by caseinate; they greatly and the anti-oxidant/anti-proliferative activities of fucoxanthin
improved the anti-cancer activity and bioaccessibility of the were greatly improved by this porous starch-based delivery
encapsulated fucoxanthin. system. Oliyaei et al. (2020)34 developed a new and original
Gelatin and gum arabic. Gelatin and gum arabic, two oppo- encapsulation system for fucoxanthin (encapsulation efficiency
sitely charged biopolymers, are safe, biocompatible, and abun- up to 94.05%). Porous starch and halloysite nanotubes were
dant. Both of them could be co-assembled by complex coacer- combined via solvent exchange and vacuum hybrid to improve
vation to encapsulate hydrophobic bioactive compounds such the stability and sustained release of fucoxanthin, and a
as fucoxanthin.85 It has been reported that gelatin–arabic gum release rate of 27.09 ± 0.17% of fucoxanthin in 6 h was
microcapsules, as a protective shell, could reduce the contact achieved by the simulated digestive fluids in vitro. Nowadays,
of fucoxanthin with light, oxygen, heat, and other degrading with the rapid development of colloidal/polymer science and
factors, and enhance the dispersibility of this insoluble ingre- nanotechnology, more nano-encapsulation materials are con-
dient in water so as to improve its stability in food systems and stantly being developed to fully take advantage of the potential
promote its effective oral delivery to the target tissue.85 health benefit of fucoxanthin so as to produce functional
However, this microcapsule shell composed of gelatin and foods and pharmaceuticals, especially for the prevention and
gum arabic is unstable in the stomach. In the presence of treatment of chronic diseases such as tumor.
pepsin and gastric acid, the shell protein is prone to hydrolysis
and protonation, resulting in the breaking of the hydrogen 4.2. Types of delivery systems for nanoencapsulated
bond and the weakening of electrostatic adsorption between fucoxanthin
the protein and the polysaccharide.21 Therefore, when the Various delivery systems for nanoencapsulated fucoxanthin
microcapsules are used for the oral delivery of fucoxanthin, have been developed to cope with different encapsulation
some of these encapsulated active ingredients could be materials and to meet specific needs. The interactions
released directly into the stomach due to the swelling and dis- between fucoxanthin and the encapsulated material play sig-
integration of the capsules in the gastric environment. nificant roles in determining the performance of the delivery
Consequently, the target release amount of fucoxanthin in the system applied in functional foods and nutraceuticals.
small intestine and its absorption and utilization in the target Fucoxanthin can interact with encapsulation materials both

This journal is © The Royal Society of Chemistry 2020 Food Funct.


View Article Online

Review Food & Function

covalently to form conjugation bonds and non-covalently via Gel-based delivery systems. The nanogel delivery system for
hydrogen bond, hydrophobic interaction, electrostatic inter- the encapsulation of fucoxanthin could be constructed by
action, and van der Waals forces.32,72,78 Based on the encapsu- using ionic gelation method through ionic interaction between
lation materials and mechanisms, various delivery systems, as two kinds of oppositely charged embedded materials in the
summarized in Fig. 4, including lipid-based delivery systems, presence of ions.21,71 Spherically cross-linked hydrophilic
gel-based delivery systems, emulsion-based delivery systems, polymer-based nanogel beads could be made with this
molecular nanocomplexes, and biopolymer nanoparticles, method, in which the stable embedding of active substances
have been studied and developed in order to increase the solu- could be achieved.21 Ravi & Baskaran (2015)31 encapsulated
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

bility of fucoxanthin in water, and thus, to improve its fucoxanthin using chitosan and glycolipid by the ionic gela-
bioaccessibility.5,31,33,58 tion of sodium tripolyphosphate. The smooth spherical gel
Lipid-based delivery systems. As the commonly used delivery beads (200–550 nm) were characterized by scanning electron
system for lipophilic active substances, lipid-based delivery microscopy and dynamic light scattering analyses. The yield,
systems include liposomes and solid lipid cores. Liposomes encapsulation efficiency, and loading capacity of the nanogel
have been widely used in the pharmaceutical and cosmetic beads were 70%, 90%, and 47%, respectively. Based on the
industries to improve chemical solubility and to facilitate con- zeta potential analysis, fucoxanthin-loaded nanogel beads with
tinuous targeted material delivery with improved protective glycolipid (+30 to +50 mV) were more stable than that of the
effects and reduced chemical resistance.87 Liposomes, as the ones without glycolipid (+15 mV). The release of fucoxanthin
name suggests, are mainly composed of lipids, including phos- from these beads was controlled by the swelling and relaxation
pholipids and cholesterol.88 The size distribution of liposomes of the beads in the simulated digestive juice, while the stabi-
is wide, from tens of nanometers to several microns. The lity, bioaccessibility, and anti-tumor activity of fucoxanthin
surface of liposomes is usually coated with a layer of 4–5 nm were all improved by the nano/micro-encapsulation.58 Li et al.
hydrophilic membrane. Hydrogen bonding and van der Waals (2019)21 synthesized complex nanocarriers containing gum
interactions are the main driving forces in the formation of arabic–gelatin microcapsules and calcium-crosslinked alginate
liposomes. The formation and function of liposome as a deliv- hydrogel beads to improve the gastrointestinal stability and
ery system for lipophilic and hydrophilic functional com- oral efficacy of fucoxanthin. The obtained composite nanogel
ponents have been fully discussed in previous reviews.89,90 It beads exhibited a uniform spherical shape and a brownish-
was suggested that soybean lecithin can be used to prepare yellow color arose from the loaded fucoxanthin, which was dis-
fucoxanthin-loaded soybean phosphatidylcholine-liposomes, tributed uniformly in the microcapsule core instead of being
which could effectively improve the anti-oxidant and tumor cross-linked to the gel shell. The gel-based approach could be
intervention efficacy of the delivered fucoxanthin compared to applied to produce various delivery vectors from different
that of the free one when administered orally.91,92 However, encapsulation materials by selecting suitable conditions.58
due to the low loading capacity, the stimulation of the human However, the gelation process usually takes a long time, which
digestive tract caused by a large amount of surfactants, and may result in a large number of pores in the gel beads due to
the possible leakage of active ingredients during storage, the the escape of gas and organic compounds, which may lead to
application of liposomes in foods is quite limited.92 To over- gel shrinkage in the subsequent drying step.21
come these shortcomings, several technologies have been Emulsion-based delivery systems. Previous studies have
studied. Quan et al. (2013)85 fabricated fucoxanthin-loaded demonstrated that emulsion-based delivery systems can
microspheres using cetylpalmitate solid lipid core coated with improve the bioaccessibility of fucoxanthin compared to direct
fish gelatin–gum arabic coacervate shell, which exhibited a administration.33 Table 2 shows in detail the nano/micro-
greatly enhanced stability and sustained release of fucoxanthin emulsion of fucoxanthin encapsulation that was fabricated.
under simulated gastrointestinal conditions. Wang et al. They are used in the pharmaceutical and cosmetic industries,
(2017)25 constructed fucoxanthin-loaded microcapsules with which refer to emulsion systems with nanoscale dimensions,
palm stearin as the solid lipid core and fish gelatin–gum typically smaller than 1 μm in size. The nanoemulsion could
arabic complex coacervates as the protective coverings, which be more easily digested and absorbed by human body, which
also exhibited improved stability against light, humidity, and could lead to higher bioaccessibility.77,79 In addition, the
temperature, and a sustained release of fucoxanthin under nanoemulsion also has much better stability against floccula-
simulated gastrointestinal condition. The encapsulation tion and coalescence compared to conventional emulsion.76
efficiency and loading capacity reported for fucoxanthin were The oil phase core may be comprised of oil and one or more
98.3% and 0.04%, respectively, in this system. The images of non-polar lipophilic active ingredients, including phytosterols,
optical microscopy and scanning electron microscopy reveal curcumin, vitamins, carotenoids, and fucoxanthin. The water
that the obtained microcapsules have smooth surfaces with a phase shell surrounding the core may also be comprised of
mean diameter of 19.19 µm. Although the biopolymer-coated one or more polar or amphiphilic materials, including pro-
solid lipid cores in these systems could better protect fucox- teins and polysaccharides.33 When the nanoemulsion droplets
anthin against light, heat, and simulated gastrointestinal reach their target locations, the lipophilic nutrients could only
environment compared to the liposomes, the low loading be released from the droplets after the oil phase has been
capacity still limits its widespread use in the industry. digested and they could then be incorporated into mixed

Food Funct. This journal is © The Royal Society of Chemistry 2020


View Article Online

Food & Function Review


Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

Fig. 4 Types of nanostructured delivery systems applied in fucoxanthin encapsulation. The green triangle represents fucoxanthin, which is actually
yellow-brown (adapt with permission from Wei et al., 201979 and Katouzian89).

micelles to exert their effects.77 The release rate of the encapsu- shown to decrease in the order of whey protein isolate > modi-
lated components in these systems depends on the rate of fied lecithin > gum arabic, with bioaccessibility indexes of
enzyme digestibility of the nanoemulsion droplets. 92.5%, 44.6%, and 36.8%, respectively. In order to improve the
Nanoemulsion droplets have a much higher oil–water inter- chemical stability of fucoxanthin during storage, modified
facial surface area than that of conventional emulsions, and lecithin, whey protein isolate, and Tween 20 were used as
therefore they could be digested more rapidly.76 emulsifiers for the preparation of fucoxanthin-loaded O/W
Ma et al. (2019)33 formulated fucoxanthin-loaded oil-in- emulsion by using the microchannel emulsification (MCE)
water nanoemulsion by using medium-chain triacylglycerol oil method.93 The sketch diagram in Fig. 5A showed the gene-
as the surfactant, sodium azide as the anti-microbial agent, ration of droplets through MCE. According to the results,
and dissolving emulsifiers (whey protein isolate, modified monosized droplets (Fig. 5B) were successfully generated and
lecithin, and gum arabic) in water as the continuous phases. evenly distributed on the microchannel array plates (MCs),
The oil-in-water (O/W) nanoemulsion was produced via high and closely-packed droplets (Fig. 5C) were observed under a
pressure homogenization technique at ambient temperature, microscope. This emulsion-based delivery system could signifi-
in which the ratio of the continuous phase to the dispersed cantly improve the chemical stability (Fig. 5D and E) of fucox-
phase is 9 : 1. The obtained emulsion droplets ranged from anthin, and performed well for the slow release of fucoxanthin
136 nm (whey protein isolate), 140 nm (modified lecithin), to and free fatty acid (Fig. 5F). In vitro digestion showed that the
897 nm (gum arabic), and had improved stability and bioacces- bioaccessibility of fucoxanthin could be enhanced by these
sibility for the loaded fucoxanthin. It was discovered that the emulsion-based delivery systems.93 Both the liquid state and
type and concentration of the emulsifier used significantly powder form of fucoxanthin-loaded nanoemulsion could be
influenced the stability and bioaccessibility of the entrapped used to improve the shelf life and nutritional value of func-
fucoxanthin. The release of fucoxanthin from these systems tional foods with their increased bioaccessibility for
was studied in an in vitro digestion model; the release rate was fucoxanthin.33

This journal is © The Royal Society of Chemistry 2020 Food Funct.


View Article Online

Review Food & Function


Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

Fig. 5 The formation, structure, and morphology of the o/w emulsions and the performance of the encapsulated fucoxanthin (FX): sketch diagram
used for the generation of droplets from microchannel array plates (A). Photographs of droplet generation (B) and the closely-packed droplets (C)
under the microscope. Storage stability photos (D), chemical stability (E), and the FFAs released (F) of monodisperse O/W emulsions encapsulating
FX under different temperatures and times. WPI: Whey protein isolates, ML: modified lecithin HPH: high pressure homogenization, FFAs: free fatty
acids (reproduced with permission from Ma et al., 2020;93 refer to this text for more details).

Molecular nanocomplexes. Molecular nanocomplexes here formed. Zhu et al. (2019)78 also constructed a different type of
refer to the autonomous assembly of host molecules (biopoly- nanocomplexes using whey protein isolate through its ligand-
mers such as proteins and polysaccharides) and ligand mole- binding and complex coacervation property with fucoxanthin
cules (functional active ingredients) bound together by non- and lysozyme, respectively. FT-IR spectroscopy and molecular
covalent interactions between them.4,32 Zhu et al. (2017)32 docking analyses revealed that fucoxanthin was successfully
assembled nanocomplexes consisting of fucoxanthin and whey encapsulated into the heteroprotein complex coacervates, and
proteins. Spectroscopic and molecular docking analyses the heteroprotein complexes exhibited a higher loading
showed that the van der Waals force, hydrogen bonding, and efficiency compared to the single-protein complexes (82.36%
hydrophobic interaction were the major driving forces for the of whey protein isolate-fucoxanthin-lysozyme, while 55.60% of
formation of whey protein–fucoxanthin nanocomplexes. In the whey protein isolate-fucoxanthin) and better protection for
presence of fucoxanthin, the skeleton structures of whey pro- fucoxanthin against heating, long-term storage, and simulated
teins (including BSA, β-Lg, and α-La) were opened, and then gastrointestinal environments. Liu et al. (2019)4 produced
nanoscale protein aggregates with fucoxanthin molecules were fucoxanthin-loaded nanocomplexes composed by oleic acid

Food Funct. This journal is © The Royal Society of Chemistry 2020


View Article Online

Food & Function Review

and bovine serum albumin (BSA) that were stabilized via the 4.3. Processing techniques for nano/micro-encapsulation
ligand-binding property of BSA with fucoxanthin and oleic
acid. When the molar ratio of fucoxanthin, oleic acid, and BSA Processing techniques to prepare nano/micro-encapsulation
was 5 : 4 : 1, the obtained complexes (size ∼265 nm) with good systems developed nutraceuticals have been gradually applied
transparency (transmittance nearly 90%) were acquired. for the preparation of functional nanoencapsulated
Besides, the nanocomplexes with oleic acid as the protein co- fucoxanthin.31–33,78,95 Several common nano/micro-encapsula-
ligand exhibited 5 times higher loading efficiency than that of tion methods for fucoxanthin are summarized in Table 3.
the ones with no oleic acid, and the anti-oxidative capability of Generally speaking, nano/micro-encapsulation techniques
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

fucoxanthin in mice eyes was improved by 71.02%. could be categorized into several classes: physical mixing,
Biopolymer nanoparticles. Biopolymer nanoparticles used chemical cross-linking, enzymatic conjugation, and physico-
for the delivery of hydrophobic bioactive ingredients in food, chemical gelation. Physical mixing methods for nano/micro-
pharmaceutical and cosmetic industries are typically smaller encapsulation include freeze drying, high pressure homogeniz-
than 1 μm in size.71,76 Typically, fucoxanthin-loaded bio- ation, microchannel emulsification, and sonication.75,93
polymer nanoparticles consist of hydrophobic cores and Chemical cross-linking techniques include anti-solvent pre-
hydrophilic shells.5,30 Hydrophobic proteins (whey protein, cipitation, solvent evaporation, ion crosslinking, and
casein, zein, etc.) firstly assemble to form the spherical inner polymerization.5,69,71,96 Enzymatic conjugation technique
core and hydrophilic polysaccharides ( pectin, carrageenan, involves the generation of nano/micro-encapsulation via
chitosan, etc.) can then be coated on the core surface to form enzyme-aided mechanisms, which is different from Maillard
the outer shell.5,70,71 Fucoxanthin can be embedded in the reaction that is another reaction that can form protein-polysac-
protein core primarily via hydrophobic interactions and charide conjugates.97–99 In physicochemical methods, nano/
soluble polysaccharides were adsorbed on the surface of micro-encapsulation is generated via complex coacervation
protein core mainly through electrostatic interactions in order and ionic gelation through synergistic physical and chemical
to improve the stability, water dispersibility, and bioaccessibil- procedures.75,80 Hereafter, advances in some specific nano/
ity of the entire nanoparticle system.6,30 The design of an micro-encapsulation techniques are discussed in more detail.
appropriate shell structure in biopolymer nanoparticles is very Physical methods. Among all the physical methods, freeze-
conducive for the stability and efficient delivery of bioactive drying is the one most widely used in pharmaceutical and
components. For example, the use of alginate or chitosan functional food industries, especially for the preparation of
instead of pectin to form the shell layer of the nanoparticles thermally sensitive loading liposomes with small particle size
can significantly improve the stability of zein or casein-based (within 200 nm) and good stability, which could be con-
core–shell nanoparticles, which could be advantageous to veniently sterilized and stored.85 With the freeze-drying
improve the water dispersion and slow-release of bioactive method, hydrophobic fucoxanthin can be easily entrapped in
components.5,94 Natural biopolymers are preferred as raw liposomes and its bioactivity could also be fully utilized. Koo
materials to prepare core–shell nanoparticles for the nano- et al. (2016)5 developed procedures to prepare fucoxanthin-
encapsulation of hydrophobic bioactive components includ- loaded casein–chitosan composite nanoparticles by the freeze-
ing fucoxanthin to improve their water solubility and drying method. In their approach, casein (CN) was completely
bioaccessibility.69–71 Koo et al. (2016)5 used casein/chitosan hydrated and mixed with fucoxanthin-enriched fraction (FX-fr)
core–shell nanoparticles for encapsulating fucoxanthin and of ethanol solution. Crosslinking between FX and CN was
their morphology, structure, encapsulation efficiency, water induced by calcium ions, and the FX–CN complexes were pre-
dispersibility, and bioavailability were investigated. pared by electrospray method. Chitosan was coated onto the
According to their results, the fucoxanthin-loaded core–shell surface of the complexes and the fucoxanthin-loaded casein–
nanoparticles with a smooth spherical surface were chitosan composite nanoparticles (FX-CS–CN) were obtained
∼277 nm in size with an encapsulation efficiency over 71% after freeze-drying at −120 °C for 48 h. Fig. 6 shows the freeze-
and a polydispersity index less than 0.40 in water. The drying process and the resultant nanoencapsulated fucox-
bioaccessibility (simulated digestion in vitro) of fucoxanthin anthin. After storage at 26 °C for 4 weeks, the free FX in water
from the core–shell nanoparticles (36–61%) was higher than was degraded to about 30% of the initial amount, while more
that of free fucoxanthin (26–48%). The pharmacokinetic than 80% of FX in the two nanoparticles were retained under
study indicated that the bioavailability (280 nmol h L−1 to the same conditions. Koo et al. (2016)5 suggested that FX-CS–
531 nmol h L−1) and mucin adsorption level (34.7% to CN nanoparticles obtained by freeze-drying showed enhanced
61.2%) of fucoxanthin from the core–shell nanoparticles bioaccessibility (26%–61%), bioavailability (280 nmol h/L–
were greatly enhanced. In addition, the existence of calcium 531 nmol h L−1) and water dispersibility compared to that of
ions in the core–shell nanoparticles helps to create a more free fucoxanthin and binary FX–CN particles, which could be
compact protective structure, thus reducing the degradation more suitable for food industry. As an alternative physical
and destruction of fucoxanthin by light, oxygen, heat, and mixing method, Ma et al. (2019)33 constructed a novel fucox-
gastric acid. The obtained fucoxanthin-embedded core–shell anthin-loaded oil-in-water emulsion by using high-pressure
nanoparticles can be used in functional beverages including homogenization at 100 MPa with 4 passes. In brief, due to the
milk.65,66 special purpose of encapsulation, a variety of physical techno-

This journal is © The Royal Society of Chemistry 2020 Food Funct.


View Article Online

Review Food & Function

Table 3 Common nano/micro-encapsulation techniques for fucoxanthin

Classes Techniques Advantages Disadvantages

Physical mixing Freeze drying - Maintains the natural properties of the - Limitation in types of coating material
method material to the greatest extent and minimize its
loss; especially suitable for handling heat
sensitive materials.
- Suitable for encapsulating many types of - Loss of compound bioactivity during freezing
compounds process
- The operation is simple. - High operational cost
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

- The rehydration of the material is easy.


- Easily adaptable at larger scales
High pressure - The pressure applied to the material is - Energy consumption and the equipment is
homogenization uniform. vulnerable to damage
- The particle size is small and uniform. - Limitation in types of coating material and
the viscosity cannot be too high
- Less heat, thus basically keep the material
properties unchanged.
- Is able to ration the material
Sonication - Saves time - The temperature rises sharply during the
treatment.
- Minimizes sample loss - Materials are susceptible to inactivation.
- Is able to process lots of samples
- Simple technique
Emulsification - High reproducibility - Not suitable for volatile compounds and high
affinity molecules for continuous phase
- High loading efficiency - The active compound might be lost during
- Easier to industrialize due to simple and solvent evaporation.
flexible steps
Chemical Anti-solvent - Simple preparation process The organic solvents involved may make the
crosslinking method precipitation - The preparation conditions are easy to control. whole encapsulation system contaminated with
- Short synthesis period odor and its removal is more troublesome.
- Has a low cost
Ion crosslinking - Easy approach - Only certain compounds can be
encapsulated.
- Controlled release of encapsulated compounds - Positively charged coating material might
- Longer period of encapsulation cause a change in the electron donating affinity
of the encapsulated bioactive compound.
Enzymatic Enzymatic - Strong combination, good stability - Limitation in the types of coating material
conjugation method conjugation and usuallys occur between proteins and
reducing sugars
- Safe, efficient and pollution-free - Only certain enzymes work
- Simple operation and low cost
Physicochemical Coacervation - High value active substances can be - Depends on the electrostatic interaction of
method encapsulated, especially unstable compounds. the compounds with the coating material
- Low production cost for simple coacervation - Only few selected compounds can be
encapsulated
- High loading efficiency - High production cost for complex
coacervation
Ionic gelation - It facilitates easy reaction and the temperature - Raw materials are expensive and may involve
is low. organic solvents.
- The mixture is uniform at the molecular level. - The gelation process takes a long time, days
or weeks.
- Various new materials can be prepared by - There are a large number of pores in the gel
selecting suitable conditions. due to gas and organic material escaping.
Shrinkage may occur during drying.

logies and methods with different characteristics can be relatively expensive for the food industry. Li et al. (2018)30 suc-
selected to precisely process the delivery system of the bio- cessfully fabricated fucoxanthin-loaded zein/caseinate compo-
active compound. site nanoparticles by using such a facile anti-solvent precipi-
Chemical methods. At present, several research studies are tation method at neutral pH. These conditions are easy to be
focused on the anti-solvent precipitation method to construct controlled, the synthesis cycle is short, and the material cost is
natural biopolymer-based delivery systems for the application low. However, the stability, bioaccessibility, anti-oxidative
of bioactive compounds in functional foods.30,69,94 Since the activity, and anti-proliferative activity of encapsulated fucox-
high-energy emulsifying devices, such as high-pressure hom- anthin were appreciably higher compared to the free ones.
ogenizer, were widely used to manufacture the stable and However, such a simple preparation process may involve the
uniform emulsions in pharmaceuticals and cosmetics, were use of organic solvents, which are troublesome to remove and

Food Funct. This journal is © The Royal Society of Chemistry 2020


View Article Online

Food & Function Review


Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

Fig. 6 Study of fucoxanthin-loaded casein–chitosan nanoparticles using the freeze-drying process. FX: Fucoxanthin, CN: casein, CS: chitosan,
FXOH: fucoxanthinol (reproduced with permission from Koo et al., 2016;5 refer to this text for more details).

easily pollute the environment. Another chemical method that cancer.101,102 In addition, protein–polysaccharide conjugated
also involves the use of organic solvents is the solvent particles for encapsulation could also be obtained by another
exchange method. Oliyaei et al. (2020)34 developed a nano- covalent crosslinking reaction, namely, the Maillard reaction,
carrier based on porous starch and halloysite nanotube by which does not require enzymes. Wang et al. (2016)98 produced
using the methods of solvent exchange and vacuum hybrid for special O/W emulsions with the cross-linked interfacial film of
improving the stability and sustained release of fucoxanthin. BSA–dextran conjugated particles for encapsulation. Also, such
The outcome of this nanocarrier suggested that the modified a BSA–dextran conjugated particle, as the emulsifier and the
starch prepared by the solvent exchange method has a stabilizer of the O/W emulsions, was formed by the Maillard
uniform and dense porous structure. This structure can sig- reaction during the heat treatment. The improvement of oral
nificantly increase the encapsulation efficiency, loading bioavailability in mice suggested that this kind of emulsion
capacity, stability and slow-release of fucoxanthin after it is system produced by Maillard reaction is a good oral delivery
mixed with halloysite nanotube in vacuum. system for hydrophobic compounds (fucoxanthin may also be
Enzymatic conjugation and maillard reactions. Enzymatic included).
conjugation is noted for its strong specificity and mild reaction Physicochemical methods. Complex coacervation is one of
conditions. Also, it is a kind of covalently cross-linking tech- the most convenient techniques in physicochemical methods
nique to create protein–polysaccharide conjugated particles for to encapsulate high value active substances to prevent their
encapsulation, especially in the presence of oxidases or molecular degradation and oxidation, especially for unstable
TGase.100,101 The working mechanism of the two types of bioactive compounds, such as essential oils, polyphenols, and
enzymes is that TGase catalyzes simple acyl transfer reactions carotenoids.103,104 The formation of complex coacervates
between glutamine residues and free amino groups, while oxi- usually occurs between two biomacromolecules with opposite
dases (including laccase and tyrosinase) catalyze oxidation charges, such as proteins or polysaccharides, which can be
reactions between highly reactive quinone residues of the oxi- used for the stable nano/micro-encapsulation of hydrophobic
dized proteins and nucleophilic substituents of polysacchar- bioactive compounds.103,105 By combining all the advantages
ides.97 The green fluorescent protein–chitosan conjugated par- of two or more different wall materials, the obtained coacer-
ticles for encapsulation, produced by such a enzymatic conju- vates may have new functional properties. The nano/micro-
gation method, were observed to have the smart attribute of encapsulation system constructed by complex coacervation
pH-responsiveness, which will provide the possibility for the technology has a high loading capacity but its stability
stable delivery and even the targeted controlled release of the depends on the electrostatic interaction of the embedded com-
encapsulated fucoxanthin in the treatment of hypoxic pound with the coating material.103 Also, only few selected

This journal is © The Royal Society of Chemistry 2020 Food Funct.


View Article Online

Review Food & Function

compounds can be encapsulated by such a technology with a helpful to improve the cardiovascular and cerebrovascular dis-
high production cost. As mentioned above, a fucoxanthin eases, and it has the effects of lowering the blood pressure as
vector based on the complex coacervation of whey protein iso- well as reducing blood lipid, anti-oxidation, and ocular
lates and lysozyme was successfully constructed.78 The process protection.106,107 Similarly, Saravana et al. (2019)108 success-
for preparing molecular nanocomplexes through heteroprotein fully developed a κ-carrageenan-based nano-emulsion, which
complex coacervation was easy to carry out but the obtained can be used to encapsulate fucoxanthin-rich seaweed oil
encapsulation system exhibited excellent loading efficiency through ultrasound-assisted emulsification in functional
and provided remarkable protection by fucoxanthin against foods, beverage systems, and pharmaceuticals.
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

heat, light, and simulated gastrointestinal environments. The research on fucoxanthin is at the stage of stable embed-
Another physicochemical method frequently used is ionic gela- ding and delivery. Owing to the lack of rigorous in vivo metab-
tion technology, and various delivery systems can be prepared olism research and clinical trials, the practical application of
by selecting suitable coating materials and conditions. In this fucoxanthin in food and nutraceutical is still scarce (Table 4),
respect, the chitosan-sodium tripolyphosphate-glycolipid which leads to few fucoxanthin embedded foods and nutraceu-
nanogel was successfully prepared by ionic interactions ticals in the market. However, with the development of science
between two oppositely charged materials in order to improve and technology and the deepening of related research, there
the stability and bioavailability of fucoxanthin with highly will be a boom in the development of fucoxanthin-fortified
efficacy.31 In addition, Zhu et al. (2019)72 reported that the foods and nutraceuticals in the next few years.
polymerization between whey protein and fucoxanthin endow
the nanocomplexes system with good delivery ability through
simple self-assembly, which is demonstrated by its physical
stability, encapsulation efficiency, and bioaccessibility.
6. Challenges and future trends
It is still a challenge to incorporate fucoxanthin into food or a
nutrient supplement due to its poor stability, low solubility,
5. Food or nutraceutical applications and weak bioaccessibility. Although nano/micro-encapsulation
of fucoxanthin could be utilized to effectively improve its stabi-
Adding the encapsulated fucoxanthin into foods and nutraceu- lity, as shown by in vitro investigations,5,31,33,58 major draw-
ticals as a functional component can offer special nutritional backs still exist: the nanoemulsion requires more complex pro-
value to the product, cover, or preserve the flavor as well as cesses and surfactants than conventional emulsions, which
reduce the degradation or loss of fucoxanthin, which provides may increase the costs and potential toxicity; there may be
many advantages for food processing. The effective encapsula- more regulatory concerns for nanoemulsions than convention-
tion of fucoxanthin based on casein and whey protein in milk al emulsions. Furthermore, the low loading capacity of the
has been reported. Mok et al. (2016, 2018)65,66 incorporated current approaches could limit the amount of fucoxanthin
fucoxanthin extracted from brown algae into skimmed and full reaching the targeted sites efficiently in vivo. Reducing the use
fat milk by using spray drying methods, which not only of surfactants or exploring some natural surfactants should be
improved the flavor, nutritional, and functional value of ordin- considered when developing nano/micro-encapsulation
ary milk but also enhanced its stability and bioavailability due systems. The improvement and optimization of coating
to the ligand binding characteristics between milk protein and materials, structural modification, and/or the introduction of
fucoxanthin during the encapsulation. Koduvayur new carrier materials are still needed for nano/micro-encapsu-
Habeebullah, Surendraraj, & Jacobsen (2018)106 encapsulated lation delivery systems of fucoxanthin to increase the targeting
fucoxanthin in fish oil, which can be either formulated into and loading performance. Research in the relevant fields need
fucoxanthin-loaded oil in the water emulsion for functional to move beyond the stages of construction, characterization,
foods or directly processed into fucoxanthin-loaded fish oil and in vitro simulated digestion to focus more on the related
soft capsules. Encapsulated fucoxanthin in fish oil is also pharmacokinetics, metabolomics, and even clinical studies to

Table 4 Fucoxanthin products in food-nutraceutical applications

Products Encapsulation system Patent number Technical fields

Concentrated extracts of fucoxanthin Unencapsulated CN201110155266. Natural products’ separation and


X extraction
Microencapsulated fucoxanthin powder β-Cyclodextrin CN201410490630.1 Natural products’ application
microcapsule
A water soluble fucoxanthin dry powder O/W emulsion system CN201510407570.7 Natural products’ application
Fucoxanthin fish oil soft capsule O/W emulsion system CN201610698526.0 Health products and
nutraceutical
applications
Full fat and skimmed milk powder containing Molecular nanocomplexes Not found Food processing
fucoxanthin

Food Funct. This journal is © The Royal Society of Chemistry 2020


View Article Online

Food & Function Review

examine their practical tumor intervention performance tional foods, nutraceuticals, or nutrient supplement products
in vivo. At the same time, the work on safety and reliability is for the prevention and management of tumor. This article pro-
also quite lacking. vides a comprehensive review on the latest progress of nano/
In the future, the pharmaceutical industry will benefit the micro-encapsulation methods in enhancing the bioaccessibil-
most from the nano/micro-encapsulation technology of fucox- ity of fucoxanthin especially with the aim of fully realizing its
anthin, which makes it possible for the full therapeutic poten- tumor intervention efficacy. Natural edible materials such as
tial of fucoxanthin to be realized against chronic diseases such whey protein, casein, zein, gelatin, and starch could be used to
as cancer.36 Tumor patients often suffer from poor appetite fabricate lipid-based, gel-based, or emulsion-based nano/
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

and reduced digestive effectiveness due to the side effects pro- micro-delivery systems for fucoxanthin. Based on the pub-
duced by chemotherapy. They are more likely to suffer from lished literature, it seems that the nano/micro-delivery system
malnutrition than normal healthy people. Therefore, a variety with whey protein as the encapsulation material seems to be
of natural functional ingredients including fucoxanthin more conducive for the stable encapsulation and safe delivery
should be considered as nutritional supplements for tumor of fucoxanthin. To make nano/micro-capsules, various
patients. Functional foods that can satisfy both the gastro- advanced processing techniques can be utilized, each with their
nomic and dietary protective needs of tumor patients could be own advantages and disadvantages, e.g., freeze-drying, high
developed for the benefit of tumor patients. Certainly, for such pressure homogenization, sonication, anti-solvent precipitation,
foods, sensory characteristics, such as flavor, texture, and coacervation, ion crosslinking, ionic gelation, emulsification,
taste, along with the nutritional/dietary needs, should all be and enzymatic conjugation. Past researches have indicated that
considered for their proper design and production.109 In nano/micro-capsules formulated using either coacervation (best
addition, nano/micro-carriers with unique functionality, performance in terms of fucoxanthin loading) or freeze-drying
including magnetic responsiveness and pH responsiveness, (best performance in terms of retaining fucoxanthin activity)
can be used for the targeted delivery of fucoxanthin to maxi- were among the most effective in stabilizing and enhancing the
mize its tumor intervention efficacy. Magnetic nanoparticles bioaccessibility of fucoxanthin. It is conceivable that the explora-
have been explored as tumor drug carriers due to their high tion and utilization of encapsulated fucoxanthin will continue
specific surface area, favorable biocompatibility, low toxicity, in the coming years to bring more health benefits to functional
and strong magnetic responsiveness.110 Also, they are usually foods, nutraceuticals, and nutrient supplements. For the poten-
composed of magnetic cores with polymer coating over the tial of fucoxanthin to be fully realized, further studies on the
surfaces so as to reduce the agglomeration of nanoparticles metabolomics of fucoxanthin and its derivatives in vivo, the con-
and to improve the affinity of the delivery system towards tar- tinued improvement of performance of nano/micro-encapsula-
geted organs.111 Furthermore, such magnetic nanoparticles tion techniques, and the investigation into new synergistic
can generate heat in an external alternating magnetic field and nanocarriers are all needed. The information presented in this
by moving the magnetic field position, they can be artificially paper could be utilized by a broad community of multidisciplin-
controlled to produce hyperthermia at specific focal sites and ary researchers from foods, clinical, materials, and colloid
kill the tumors.112 With the rapid growth of solid tumor, the sciences working at the interface of chemistry and biology to
hypoxia caused by the imbalance of oxygen supply and con- keep the march towards taking full advantage of fucoxanthin to
sumption tends to cause systemic or local acidulated environ- improve human health.
ment.102 In this respect, a delivery system with pH responsive-
ness, which may be produced by green fluorescent protein–
chitosan conjugate, is critically important for the stable deliv- Author contributions
ery and targeted controlled release of fucoxanthin in the treat-
ment of hypoxic tumor.101 In the future, the nano/micro- Hang Qi and Xing Chen: Conceptualization, methodology,
encapsulation technology of fucoxanthin will inevitably focus software. Chunyan Wang: Resources, investigation, writing,
on its application in targeted controlled release, which is of original draft preparation. Yoshimasa Nakamura:
great significance for the development of fucoxanthin-based Visualization, validation, reviewing. Chenxu Yu: Reviewing and
tumor intervention products. editing. All the authors read, edited, and approved the final
manuscript.

7. Conclusion Conflicts of interest


Fucoxanthin is a natural carotenoid abundant in marine There are no conflicts to declare.
brown algae with a huge potential of tumor intervention
efficacy. The direct addition of fucoxanthin and its derivatives
into foods and nutraceuticals is neither easy nor effective due Acknowledgements
to their low solubility, stability, and bioaccessibility. Nano/
micro-encapsulation offers a possible solution to this problem. This work was financially supported by The National Key
By this technique, fucoxanthin could be introduced into func- Research and Development Program of China

This journal is © The Royal Society of Chemistry 2020 Food Funct.


View Article Online

Review Food & Function

(2019YFD0902001), Young Top Talents Project of Liaoning gallate delivery, Crit. Rev. Food Sci. Nutr., 2020, 60, 1243–
Revitalization Talents Program (XLYC1807166), and Dalian 1264.
Science and Technology Foundation for Distinguished Young 15 L. Sun and M. Miao, Dietary polyphenols modulate starch
Scholars (2018RL07). digestion and glycaemic level: A review, Crit. Rev. Food Sci.
Nutr., 2020, 60, 541–555.
16 B. A. Guler, I. Deniz, Z. Demirel, O. Yesil-Celiktas and
E. Imamoglu, A novel subcritical fucoxanthin extraction
References with a biorefinery approach, Biochem. Eng. J., 2020, 153,
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

107403.
1 X. Xiao, X. Si, Z. Yuan, X. Xu and G. Li, Isolation of fucox- 17 S. M. Kim, et al., A potential commercial source of fucox-
anthin from edible brown algae by microwave-assisted anthin extracted from the microalga Phaeodactylum tri-
extraction coupled with high-speed countercurrent chrom- cornutum, Appl. Biochem. Biotechnol., 2012, 166, 1843–
atography, J. Sep. Sci., 2012, 35, 2313–2317. 1855.
2 K. N. Kim, et al., Inhibition of tumor growth in vitro and 18 S. Xia, et al., Production, characterization, and antioxidant
in vivo by fucoxanthin against melanoma B16F10 cells, activity of fucoxanthin from the marine diatom Odontella
Environ. Toxicol. Pharmacol., 2013, 35, 39–46. aurita, Mar. Drugs, 2013, 11, 2667–2681.
3 A. Zarekarizi, L. Hoffmann and D. Burritt, Approaches for 19 S. Xia, B. Gao, J. Fu, J. Xiong and C. Zhang, Production of
the sustainable production of fucoxanthin, a xanthophyll fucoxanthin, chrysolaminarin, and eicosapentaenoic acid
with potential health benefits, J. Appl. Phycol., 2018, 31, by Odontella aurita under different nitrogen supply
281–299. regimes, J. Biosci. Bioeng., 2018, 126, 723–729.
4 Y. Liu, et al., Oleic acid as a protein ligand improving 20 P. Sun, et al., A novel strategy for isolation and purifi-
intestinal absorption and ocular benefit of fucoxanthin in cation of fucoxanthinol and fucoxanthin from the diatom
water through protein-based encapsulation, Food Funct., Nitzschia laevis, Food Chem., 2019, 277, 566–572.
2019, 10, 4381–4395. 21 Y. Li, et al., Improvement of fucoxanthin oral efficacy via
5 S. Y. Koo, I. K. Mok, C. H. Pan and S. M. Kim, Preparation vehicles based on gum Arabic, gelatin and alginate hydro-
of fucoxanthin-loaded nanoparticles composed of casein gel, J. Funct. Foods, 2019, 63, 103573.
and chitosan with improved fucoxanthin bioavailability, 22 U. Neumann, et al., A carotenoid derived from
J. Agric. Food Chem., 2016, 64, 9428–9435. Phaeodactylum tricornutum exerts antiproliferative and
6 H. Li, Y. Yuan, J. Zhu, T. Wang and Y. Xu, Zein/soluble antioxidant activities in vitro, Antioxidants, 2019, 8, 183–
soybean polysaccharide composite nanoparticles for 193.
encapsulation and oral delivery of lutein, Food 23 H. Staleva-Musto, et al., Nonconjugated acyloxy group de-
Hydrocolloids, 2020, 103, 105715. activates the intramolecular charge-transfer state in the
7 F. Bray, et al., Global cancer statistics 2018: globocan esti- carotenoid fucoxanthin, J. Phys. Chem. B, 2018, 122, 2922–
mates of incidence and mortality worldwide for 36 2930.
cancers in 185 countries, CA Cancer J. Clin., 2018, 68, 394– 24 B. Guo, et al., Gastrointestinal bioaccessibility and colonic
424. fermentation of fucoxanthin from the extract of the micro-
8 A. Jemal, R. Siegel, J. Xu and E. Ward, Cancer statistics, alga Nitzschia laevis, J. Agric. Food Chem., 2019, 68, 1844–
2010, CA Cancer J. Clin., 2010, 60, 277–300. 1850.
9 R. L. Siegel, K. D. Miller and A. Jemal, Cancer statistics, 25 X. Wang, et al., Isolation of fucoxanthin from Sargassum
2018, CA Cancer J. Clin., 2018, 68, 7–30. thunbergii and preparation of microcapsules based on
10 D. Hashim and P. Boffetta, Occupational and environ- palm stearin solid lipid core, Front. Mater. Sci., 2017, 11,
mental exposures and cancers in developing countries, 66–74.
Ann. Glob. Health, 2014, 80, 393–411. 26 B. Guo, B. Yang, X. Pang, T. P. Chen and K. W. Cheng,
11 U. T. Sankpal, et al., Environmental factors in causing Fucoxanthin modulates cecal and fecal microbiota differ-
human cancers: emphasis on tumorigenesis, Tumour ently based on diet, Food Funct., 2019, 10, 5644–5655.
Biol., 2012, 33, 1265–1274. 27 Z. Liu, X. Sun, X. Sun, S. Wang and Y. Xu, Fucoxanthin
12 S. Cheng, M. Tu, H. Liu, G. Zhao and M. Du, Food-derived isolated from Undaria pinnatifida can interact with
antithrombotic peptides: Preparation, identification, and Escherichia coli and Lactobacilli in the intestine and
interactions with thrombin, Crit. Rev. Food Sci. Nutr., inhibit the growth of pathogenic bacteria, J. Ocean Univ.
2019, 59, S81–S95. China, 2019, 18, 926–932.
13 D. Julian and D. J. McClements, Advances in nanoparticle 28 M. Peraman and S. Nachimuthu, Identification and quanti-
and microparticle delivery systems for increasing the dis- fication of fucoxanthin in selected carotenoid-producing
persibility, stability, and bioactivity of phytochemicals, marine microalgae and evaluation for their chemothera-
Biotechnol. Adv., 2018, 38, 107287. peutic potential, Phcog. Mag., 2019, 15, S243–S249.
14 Q. Q. Yang, et al., Nanochemoprevention with therapeutic 29 C. L. V. Raji, G. Sadhasivam, S. Kandasamy, K. Poomani
benefits: An updated review focused on epigallocatechin and P. Thayumanavan, Purification of fucoxanthin from

Food Funct. This journal is © The Royal Society of Chemistry 2020


View Article Online

Food & Function Review

Sargassum wightii Greville and understanding the inhi- 44 T. Hashimoto, et al., Pharmacokinetics of fucoxanthinol
bition of angiotensin 1-converting enzyme: An in vitro and in human plasma after the oral administration of kombu
in silico studies, Int. J. Biol. Macromol., 2020, 148, 696– extract, Br. J. Nutr., 2012, 107, 1566–1569.
703. 45 S. Komba, E. Kotake-Nara and W. Tsuzuki, Degradation of
30 H. Li, et al., Stability, bioactivity, and bioaccessibility of fucoxanthin to elucidate the relationship between the
fucoxanthin in zein-caseinate composite nanoparticles fucoxanthin molecular structure and its antiproliferative
fabricated at neutral pH by antisolvent precipitation, Food effect on Caco-2 cells, Mar. Drugs, 2018, 16, 275–283.
Hydrocolloids, 2018, 84, 379–388. 46 C. L. Liu, Y. S. Huang, M. Hosokawa, K. Miyashita and
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

31 H. Ravi and V. Baskaran, Biodegradable chitosan-glyco- M. L. Hu, Inhibition of proliferation of a hepatoma cell
lipid hybrid nanogels: A novel approach to encapsulate line by fucoxanthin in relation to cell cycle arrest and
fucoxanthin for improved stability and bioavailability, enhanced gap junctional intercellular communication,
Food Hydrocolloids, 2015, 43, 717–725. Chem.–Biol. Interact., 2009, 182, 165–172.
32 J. Zhu, X. Sun, S. Wang, Y. Xu and D. Wang, Formation of 47 S. Y. Shin, et al., Egr-1 is necessary for fibroblast growth
nanocomplexes comprising whey proteins and fucox- factor-2-induced transcriptional activation of the glial cell
anthin: Characterization, spectroscopic analysis, and line-derived neurotrophic factor in murine astrocytes,
molecular docking, Food Hydrocolloids, 2017, 63, 391–403. J. Biol. Chem., 2009, 284, 30583–30593.
33 Z. Ma, et al., Fucoxanthin-loaded oil-in-water emulsion- 48 P. Ganesan, K. Matsubara, T. Sugawara and T. Hirata,
based delivery systems: Effects of natural emulsifiers on Marine algal carotenoids inhibit angiogenesis by down-
the formulation, stability, and bioaccessibility, ACS regulating FGF-2-mediated intracellular signals in vascu-
Omega, 2019, 4, 10502–10509. lar endothelial cells, Mol. Cell. Biochem., 2013, 380, 1–9.
34 N. Oliyaei, M. Moosavi-Nasab, A. M. Tamaddon and 49 K. N. Kim, et al., Inhibition of tumor growth in vitro and
M. Fazaeli, Encapsulation of fucoxanthin in binary in vivo by fucoxanthin against melanoma B16F10 cells,
matrices of porous starch and halloysite, Food Environ. Toxicol. Pharmacol., 2013, 35, 39–46.
Hydrocolloids, 2020, 100, 105458. 50 T. Rokkaku, et al., Anticancer effects of marine caroten-
35 L. J. Martin, Fucoxanthin and its metabolite fucoxanthi- oids, fucoxanthin and its deacetylated product, fucox-
nol in cancer prevention and treatment, Mar. Drugs, 2015, anthinol, on osteosarcoma, Int. J. Oncol., 2013, 43, 1176–
13, 4784–4798. 1186.
36 M. Bae, M. B. Kim, Y. K. Park and J. Y. Lee, Health 51 K. Yamamoto, C. Ishikawa, H. Katano, T. Yasumoto and
benefits of fucoxanthin in the prevention of chronic dis- N. Mori, Fucoxanthin and its deacetylated product, fucox-
eases, Biochim. Biophys. Acta, Mol. Cell Biol. Lipids, 2020, anthinol, induce apoptosis of primary effusion lympho-
1865, 158618. mas, Cancer Lett., 2011, 300, 225–234.
37 E. H. Harrison, Mechanisms involved in the intestinal 52 R. X. Yu, R. T. Yu and Z. Liu, Inhibition of two gastric
absorption of dietary vitamin A and provitamin A caroten- cancer cell lines induced by fucoxanthin involves downre-
oids, BBA-Mol. Cell Biol. L., 2012, 1821, 70–77. gulation of Mcl-1 and STAT3, Hum. Cell, 2018, 31, 50–63.
38 E. Reboul, Absorption of vitamin A and carotenoids by the 53 C. Mei, et al., Antitumor effects of laminaria extract fucox-
enterocyte: Focus on transport proteins, Nutrients, 2013, 5, anthin on lung cancer, Mar. Drugs, 2017, 15, 39–50.
3563–3581. 54 S. K. Das, et al., Fucoxanthin induces cell cycle arrest at
39 T. Sugawara, V. Baskaran, W. Tsuzuki and A. Nagao, g0/g1 phase in human colon carcinoma cells through up-
Brown algae fucoxanthin is hydrolyzed to fucoxanthinol regulation of p21waf1/cip1, Biochim. Biophys. Acta, 2005,
during absorption by Caco-2 human intestinal cells and 1726, 328–335.
mice, J. Nutr., 2002, 132, 946–951. 55 E. Kotake-Nara, et al., Carotenoids affect proliferation of
40 A. Asai, T. Sugawara, H. Ono and A. Nagao, human prostate cancer cells, J. Nutr., 2001, 131, 3303–
Biotransformation of fucoxanthinol into amarouciax- 3306.
anthin A in mice and HepG2 cells: Formation and cyto- 56 L. Wang, et al., Fucoxanthin induces growth arrest and
toxicity of fucoxanthin metabolites, Drug Metab. Dispos., apoptosis in human bladder cancer T24 cells by up-regu-
2004, 32, 205–211. lation of p21 and down-regulation of mortalin, Acta
41 C. Kiefer, E. Sumser and M. F. Wernet, A class B scavenger Biochim. Biophys. Sin., 2014, 46, 877–884.
receptor mediates the cellular uptake of carotenoids in 57 Y. Liu, et al., Fucoxanthin activates apoptosis via inhibition
drosophila, Proc. Natl. Acad. Sci. U. S. A., 2002, 99, 10581– of PI3 K/Akt/mTOR pathway and suppresses invasion and
10586. migration by restriction of p38-MMP-2/9 pathway in human
42 V. Shete and L. Quadro, Mammalian metabolism of glioblastoma cells, Neurochem. Res., 2016, 41, 1–24.
β-Carotene: Gaps in knowledge, Nutrients, 2013, 5, 4849–4868. 58 H. Ravi, N. Kurrey, Y. Manabe, T. Sugawara and
43 T. Hashimoto, Y. Ozaki, M. Taminato, S. K. Das and V. Baskaran, Polymeric chitosan-glycolipid nanocarriers
K. Kanazawa, The distribution and accumulation of fucox- for an effective delivery of marine carotenoid fucoxanthin
anthin and its metabolites after oral administration in for induction of apoptosis in human colon cancer cells
mice, Br. J. Nutr., 2009, 102, 242–248. (Caco-2 cells), Mater. Sci. Eng., C, 2018, 91, 785–795.

This journal is © The Royal Society of Chemistry 2020 Food Funct.


View Article Online

Review Food & Function

59 Y. Jin, S. Qiu, N. Shao and J. Zheng, Fucoxanthin and fermentation of fucoxanthin from the extract of the micro-
tumor necrosis factor-related apoptosis-inducing ligand alga Nitzschia laevis, J. Agric. Food Chem., 2020, 68, 1844–
(TRAIL) synergistically promotes apoptosis of human cer- 1850.
vical cancer cells by targeting PI3 K/Akt/NF-kappaB signal- 74 J. T. D. P. Silva, J. M. T. Geiss, S. M. Oliveira,
ing pathway, Med. Sci. Monit., 2018, 24, 11–18. E. D. S. Brum, S. C. Sagae, D. Becker, F. V. Leimann,
60 T. Sugawara, K. Matsubara, R. Akagi, M. Mori and R. P. Ineu, G. P. Guerra and O. H. Goncalves, Nano/micro-
T. Hirata, Antiangiogenic activity of brown algae fucox- encapsulation of lutein and its effect on mice’s declarative
anthin and its deacetylated product, fucoxanthinol, memory, Mater. Sci. Eng., C, 2017, 76, 1005–1011.
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

J. Agric. Food Chem., 2007, 54, 9805–9810. 75 C. Liu, et al., Fabrication and characterization of
61 T. W. Chung, et al., Marine algal fucoxanthin inhibits the β-Lactoglobulin-based nanocomplexes composed of chito-
metastatic potential of cancer cells, Biochem. Biophys. Res. san oligosaccharides as vehicles for delivery of astax-
Commun., 2013, 439, 580–585. anthin, J. Agric. Food Chem., 2018, 66, 6717–6726.
62 N. Thakur, P. Raigond, Y. Singh, T. Mishra and S. Dutt, 76 D. J. McClements, Edible nanoemulsions: fabrication,
Recent updates on bioaccessibility of phytonutrients, properties, and functional performance, Soft Matter, 2011,
Trends Food Sci. Technol., 2020, 97, 366–380. 7, 2297–2316.
63 A. Fung, N. Hamid and J. Lu, Fucoxanthin content and 77 L. Chen, C. Gnanaraj, P. Arulselvan, H. El-Seedi and
antioxidant properties of Undaria pinnatifida, Food Chem., H. Teng, A review on advanced microencapsulation
2013, 136, 1055–1062. technology to enhance bioavailability of phenolic com-
64 C. S. Boon, D. J. Mcclements, J. Weiss and E. A. Decker, pounds: Based on its activity in the treatment of Type 2
Factors influencing the chemical stability of carotenoids Diabetes, Trends Food Sci. Technol., 2019, 85, 149–162.
in foods, Crit. Rev. Food Sci. Nutr., 2010, 50, 515–532. 78 J. Zhu, H. Li, Y. Xu and D. Wang, Construction of fucox-
65 I. K. Mok, J. R. Yoon, C. H. Pan and S. M. Kim, anthin vector based on binding of whey protein isolate
Development, quantification, method validation, and and its subsequent complex coacervation with lysozyme,
stability study of a novel fucoxanthin-fortified milk, J. Agric. Food Chem., 2019, 67, 2980–2990.
J. Agric. Food Chem., 2016, 64, 6196–6202. 79 Z. Wei and Q. Huang, Assembly of protein−polysaccharide
66 I. K. Mok, J. K. Lee, J. H. Kim, C. H. Pan and S. M. Kim, complexes for delivery of bioactive ingredients: A perspec-
Fucoxanthin bioavailability from fucoxanthin-fortified milk: tive paper, J. Agric. Food Chem., 2019, 67, 1344–1352.
In vivo and in vitro study, Food Chem., 2018, 258, 79–86. 80 A. L. Chapeau, G. M. Tavares, P. Hamon, T. Croguennec,
67 I. Hossen, et al., Phytochemicals and inflammatory bowel D. Poncelet and S. Bouhallab, Spontaneous co-assembly
disease: A review, Crit. Rev. Food Sci. Nutr., 2019, 60, 1321– of lactoferrin and β-lactoglobulin as a promising biocar-
1345. rier for vitamin B9, Food Hydrocolloids, 2016, 57, 280–290.
68 Z. Liu, et al., Nano/micro-encapsulation of cyanidin-3-o- 81 F. Kimpel and J. J. Schmitt, Review: Milk proteins as nano-
glucoside enhances protection against UVB-induced epi- carrier systems for hydrophobic nutraceuticals, J. Food
dermal damage through regulation of p53-mediated apop- Sci., 2015, 80, R2361–R2366.
tosis in mice, J. Agric. Food Chem., 2018, 66, 5359–5367. 82 G. M. Tavares, T. Croguennec, A. F. Carvalho and
69 X. Huang, Y. Liu, Y. Zou, X. Liang, Y. Peng, S. Bouhallab, Milk proteins as encapsulation devices and
D. J. Mcclements and K. Hu, Encapsulation of resveratrol delivery vehicles: Applications and trends, Trends Food Sci.
in zein/pectin core-shell nanoparticles: Stability, bioacces- Technol., 2014, 37, 5–20.
sibility, and antioxidant capacity after simulated gastroin- 83 C. S. Ranadheera, W. S. Liyanaarachchi, J. Chandrapala,
testinal digestion, Food Hydrocolloids, 2019, 93, 261–269. M. Dissanayake and T. Vasiljevic, Utilizing unique pro-
70 X. Yan, X. Zhang, D. J. Mcclements, L. Zou, X. Liu and perties of caseins and the casein micelle for delivery of
F. Liu, Co-encapsulation of epigallocatechin gallate sensitive food ingredients and bioactives, Trends Food Sci.
(EGCG) and curcumin by two proteins-based nano- Technol., 2016, 57, 178–187.
particles: Role of EGCG, J. Agric. Food Chem., 2019, 67, 84 A. R. Patel and K. P. Velikov, Zein as a source of functional
13228–13236. colloidal nano- and microstructures, Curr. Opin. Colloid
71 S. Chen, Q. Li, D. J. Mcclements, Y. Han and Y. Gao, Co- Interface Sci., 2014, 19, 450–458.
delivery of curcumin and piperine in zein-carrageenan 85 J. Quan, S. M. Kim, C. H. Pan and D. Chung,
core-shell nanoparticles: Formation, structure, stability Characterization of fucoxanthin-loaded microspheres
and in vitro gastrointestinal digestion, Food Hydrocolloids, composed of cetyl palmitate-based solid lipid core and
2019, 99, 105334. fish gelatin-gum arabic coacervate shell, Food Res. Int.,
72 J. Zhu, C. Wang, J. Gao, H. Wu and Q. Sun, Aggregation of 2013, 50, 31–37.
fucoxanthin and its effects on binding and delivery pro- 86 N. Oliyaei, M. Moosavi-Nasab, A. M. Tamaddon and
perties of whey proteins, J. Agric. Food Chem., 2019, 67, M. Fazaeli, Double encapsulation of fucoxanthin using
10412–10422. porous starch through sequential coating modification
73 B. Guo, T. Oliviero, V. Fogliano, Y. Ma, F. Chen and with maltodextrin and gum Arabic, Food Sci. Nutr., 2020,
E. Capuano, Gastrointestinal bioaccessibility and colonic 8, 1226–1236.

Food Funct. This journal is © The Royal Society of Chemistry 2020


View Article Online

Food & Function Review

87 O. Temidayo, H. A. Rita, I. Daniel, S. James and E. Amal, 102 W. Zhang, et al., Oxygen-generating MnO2 nanodots-
Liposomal drug delivery systems and anticancer drugs, anchored versatile nanoplatform for combined chemo-
Molecules, 2018, 23, 907–923. photodynamic therapy in hypoxic cancer, Adv. Funct.
88 H. Rostamabadi, S. R. Falsafi and S. M. Jafari, Nano/ Mater., 2018, 28, 1706375.
micro-encapsulation of carotenoids within lipid-based 103 R. Shaddel, et al., Use of gelatin and gum Arabic for
nanocarriers, J. Controlled Release, 2019, 298, 38–67. encapsulation of black raspberry anthocyanins by
89 I. Katouzian and S. M. Jafari, Nano-encapsulation as a complex coacervation, Int. J. Biol. Macromol., 2018, 107,
promising approach for targeted delivery and controlled 1800–1810.
Published on 22 October 2020. Downloaded by University of New England on 11/5/2020 4:18:34 PM.

release of vitamins, Trends Food Sci. Technol., 2016, 53, 104 M. Zare, Z. N. Roshan, E. Assadpour and S. M. Jafari,
34–48. Improving the cancer prevention/treatment role of caro-
90 I. Shishir, M. Rezaul, N. Karim, V. Gowd, X. Zheng and tenoids through various nano-delivery systems, Crit. Rev.
W. Chen, Liposomal delivery of natural product: A promis- Food Sci. Nutr., 2020, DOI: 10.1080/
ing approach in health research, Trends Food Sci. Technol., 10408398.2020.1738999.
2019, 85, 177–200. 105 S. Tie, X. Zhang, H. Wang, Y. Song and M. Tan,
91 M. K. Airanthi, M. Hosokawa and K. Miyashita, Procyanidins-loaded complex coacervates for improved
Comparative antioxidant activity of edible Japanese brown stability by self-crosslinking and calcium ions chelation,
seaweeds, J. Food Sci., 2011, 76, C104–C111. J. Agric. Food Chem., 2020, 68, 3163–3170.
92 W. Tokarek, et al., The meaning of lipid carriers for fucox- 106 S. F. K. Habeebullah, A. Surendraraj and C. Jacobsen,
anthin anti-cancer properties, New Biotechnol., 2016, 33, Isolation of fucoxanthin from brown algae and its anti-
S75–S75. oxidant activity: In vitro and 5% fish oil-in-water emulsion,
93 Z. Ma, Y. Zhao, N. Khalid, G. Shu and M. Nakajima, J. Am. Oil Chem. Soc., 2018, 95, 835–843.
Comparative study of oil-in-water emulsions encapsulat- 107 M. Daniela, et al., Maternal supplementation of inositols,
ing fucoxanthin formulated by microchannel emulsifica- fucoxanthin, and hydroxytyrosol in pregnant murine
tion and high-pressure homogenization, Food models of hypertension, Am. J. Hypertens, 2020, 33, 652–
Hydrocolloids, 2020, 105977. 659.
94 X. Huang, et al., Enhancement of curcumin water disper- 108 P. S. Saravana, et al., Ultrasound-mediated fucoxanthin
sibility and antioxidant activity using core-shell protein- rich oil nanoemulsions stabilized by κ-carrageenan:
polysaccharide nanoparticles, Food Res. Int., 2016, 87, 1–9. process optimization, bio-accessibility and cytotoxicity,
95 M. Liu, W. Li, Y. Chen, X. Wan and J. Wang, Fucoxanthin: Ultrason. Sonochem., 2019, 55, 105–116.
A promising compound for human inflammation-related 109 L. T. Galaniha, D. J. McClements and A. Nolden,
diseases, Life Sci., 2020, 255, 117850. Opportunities to improve oral nutritional supplements for
96 X. Sun, et al., Construction and multifunctionalization of managing malnutrition in cancer patients: A food design
chitosan-based three-phase nano-delivery system, Food approach, Trends Food Sci. Technol., 2020, 102, 254–260.
Hydrocolloids, 2019, 96, 402–411. 110 J. Shi, H. Fu, X. Sun, J. Shen and H. Zhang, Magnetic,
97 Z. Wei and Q. Huang, Assembly of protein-polysaccharide long persistent luminescent and mesoporous nano-
complexes for delivery of bioactive ingredients: A perspec- particles as trackable transport drug carriers, J. Mater.
tive paper, J. Agric. Food Chem., 2019, 67, 1344–1352. Chem. B, 2014, 3, 635–641.
98 C. Wang, Z. Liu, G. Xu, B. Yin and P. Yao, BSA-dextran 111 K. J. Lee, J. H. An, J. S. Shin and D. H. Kim, Synthesis and
emulsion for protection and oral delivery of curcumin, characterization of bicalutamide-loaded magnetic nano-
Food Hydrocolloids, 2016, 61, 11–19. particles as anti-tumor drug carriers, J. Nanosci.
99 J. J. Fu, C. Sun, X. B. Xu, D. Y. Zhou, L. Song and Nanotechnol., 2012, 12, 1611–1615.
B. W. Zhu, Improving the functional properties of bovine 112 H. Peng, C. Hu, J. Hu, X. Tian and T. Wu, Fe3O4@mZnO
serum albumin-glucose conjugates in natural deep eutec- nanoparticles as magnetic and microwave responsive drug
tic solvents, Food Chem., 2020, 328, 127122. carriers, Microporous Mesoporous Mater., 2016, 226, 140–
100 P. D. Pierro, B. Chico, R. Villalonga, L. Mariniello, P. Masi 145.
and R. Porta, Transglutaminase-catalyzed preparation of 113 Y. Li, et al., Storage carbon metabolism of Isochrysis zhang-
chitosan-ovalbumin films, Enzyme Microb. Technol., 2007, jiangensis under different light intensities and its appli-
40, 437–441. cation for co-production of fucoxanthin and stearidonic
101 T. Chen, D. A. Small, L. Q. Wu, G. W. Rubloff and acid, Bioresour. Technol., 2019, 282, 94–102.
G. F. Payne, Nature-inspired creation of protein-polysac- 114 E. Shannon and N. Abu-Ghannam, Enzymatic extraction
charide conjugate and its subsequent assembly onto a of fucoxanthin from brown seaweeds, Int. J. Food Sci.
patterned surface, Langmuir, 2003, 19, 9382–9386. Technol., 2018, 53, 2195–2204.

This journal is © The Royal Society of Chemistry 2020 Food Funct.

You might also like