You are on page 1of 14

Trends in Food Science & Technology 21 (2010) 510e523

Review

Encapsulation of
polyphenols e phase or payload. On the other hand, the packaging mate-
rials are called coating material, wall material, capsule,
membrane, carrier or shell, which can be made of sugars,
a review gums, proteins, natural and modified polysaccharides,
lipids and synthetic polymers (Gibbs, Kermasha, Alli, &
Mulligan, 1999; Mozafari, 2006)
Zhongxiang Fanga,b,* and Microcapsules are small vesicles or particulates that may
Bhesh Bhandaria range from sub-micron to several millimeters in size
(Dziezak, 1998). Many morphologies can be produced for
a
School of Land, Crop and Food Sciences, encapsulation, but two major morphologies are more com-
The University of Queensland, Brisbane, monly seen (Fig. 1): one is mononuclear capsules, which
Qld 4072, Australia have a single core enveloped by a shell, while the other
b is aggregates, which have many cores embedded in a matrix
School of Biosystems Engineering and Food Science,
(Schrooyen, van der Meer, & De Kruif, 2001). Their spe-
Zhejiang University, Hangzhou 310029, China
cific shapes in different systems are influenced by the pro-
(School of Land, Crop and Food Sciences, The
cess technologies, and by the core and wall materials from
University of Queensland, Brisbane, Qld 4072,
which the capsules are made.
Australia. Tel.: D61 7 33469187;
Various techniques are used for encapsulation. In gen-
e-mail: z.fang2@uq.edu.au)
eral, three steps are involved in the encapsulation of bioac-
tive agents: (i) the formation of the wall around the material
Research on and the application of polyphenols, have recently to be encapsulated; (ii) ensuring that undesired leakage
attracted great interest in the functional foods, nutraceutical and does not occur; (iii) ensuring that undesired materials are
pharmaceutical industries, due to their potential health benefits kept out (Gibbs et al., 1999; Mozafari et al., 2008). The
to humans. However, the effectiveness of polyphenols depends current encapsulation techniques include spray drying,
on preserving the stability, bioactivity and bioavailability of the spray cooling/chilling, extrusion, fluidized bed coating, co-
active ingredients. The unpleasant taste of most phenolic com- acervation, liposome entrapment, inclusion complexation,
pounds also limits their application. The utilization of encapsu- centrifugal suspension separation, lyophilization, cocrystal-
lated polyphenols, instead of free compounds, can effectively lization and emulsion, etc. (Augustin & Hemar, 2009;
alleviate these deficiencies. The technologies of encapsulation Desai & Park, 2005; Gibbs et al., 1999).
of polyphenols, including spray drying, coacervation, liposome The main objective of encapsulation is to protect the
entrapment, inclusion complexation, cocrystallization, nanoen- core material from adverse environmental conditions,
capsulation, freeze drying, yeast encapsulation and emulsion, such as undesirable effects of light, moisture, and oxygen,
are discussed in this review. Current research, developments thereby contributing to an increase in the shelf life of the
and trends are also discussed. product, and promoting a controlled liberation of the encap-
sulate (Shahidi & Han, 1993). In the food industry, the
microencapsulation process can be applied for a variety
Introduction
of reasons, which have been summarized by Desai and
Microencapsulation, developed approximately 60 years
Park (2005) as follows: (i) protection of the core material
ago, is defined as a technology of packaging solids, liquids,
from degradation by reducing its reactivity to its outside en-
or gaseous materials in miniature, sealed capsules that can
vironment; (ii) reduction of the evaporation or transfer rate
release their contents at controlled rates under specific con-
of the core material to the outside environment; (iii) mod-
ditions (Desai & Park, 2005; Vilstrup, 2001). The packaged
ification of the physical characteristics of the original mate-
materials can be pure materials or a mixture, which are also
rial to allow easier handling; (iv) tailoring the release of the
called coated material, core material, actives, fill, internal
core material slowly over time, or at a particular time; (v) to
mask an unwanted flavor or taste of the core material; (vi)
* Corresponding author. dilution of the core material when only small amounts are
0924-2244/$ - see front matter Ó 2010 Elsevier Ltd. All rights reserved.
doi:10.1016/j.tifs.2010.08.003
Z. Fang, B. Bhandari / Trends in Food Science & Technology 21 (2010) 510e523 511

major polyphenols from different plant sources are present


Wall material Wall material in Table 1, which shows their limited stability and condi-
tioned solubility. Another unfortunate trait of polypheonls
Core material Core material
is their potential unpleasant taste, such as astringency
(Table 1), which needs to be masked before incorporation
Fig. 1. Two major forms of encapsulation: mononuclear capsule (left) into food products (Haslam & Lilley, 1988).
and aggregate (right). The utilization of encapsulated polyphenols instead of
free compounds can overcome the drawbacks of their insta-
bility, alleviate unpleasant tastes or flavors, as well as im-
required, while achieving uniform dispersion in the host prove the bioavailability and half-life of the compound in
material; (vii) to help separate the components of the mix- vivo and in vitro. There have been a number of recent re-
ture that would otherwise react with one another. Food in- views or mini-reviews on the encapsulation of foods or
gredients of acidulants, flavoring agents, sweeteners, food ingredients (Augustin & Hemar, 2009; Desai &
colorants, lipids, vitamins and minerals, enzymes and Park, 2005; de Vos, Faas, Spasojevic, & Sikkema, 2010;
microorganisms, are encapsulated using different technolo- Flanagan & Singh, 2006; Gouin, 2004; Jafari, Assadpoor,
gies (Desai & Park, 2005). He, & Bhandari, 2008; Khaled & Jagdish, 2007;
Recently, research and application of polyphenols have McClements, Decker, Park, & Weiss, 2009; Mozafari,
been areas of great interest in the functional foods, nutra- 2005; Mozafari, 2006; Mozafari et al., 2008; Peter &
ceutical and pharmaceutical industries (Manach, Scalbert, Given, 2009). This review focuses on the encapsulation
Morand, Rémésy, & Jiménez, 2004; Scalbert, Manach, of the more widely used polyphenols, discussing their
Morand, Rémésy, & Jiménez, 2005). Polyphenols consti- effectiveness, variations, developments and trends.
tute one of the most numerous and ubiquitous groups of
plant metabolites, and are an integral part of both human Spray drying
and animal diets which possess a high spectrum of biolog- Spray drying encapsulation has been used in the food in-
ical activities, including antioxidant, anti-inflammatory, an- dustry since the late 1950s. Because spray drying is an eco-
tibacterial, and antiviral functions (Bennick, 2002; Haslam, nomical, flexible, continuous operation, and produces
1996; Quideau & Feldman, 1996). A large body of preclin- particles of good quality, it is the most widely used micro-
ical research and epidemiological data suggests that plant encapsulation technique in the food industry and is typi-
polyphenols can slow the progression of certain cancers, re- cally used for the preparation of dry, stable food additives
duce the risks of cardiovascular disease, neurodegenerative and flavors (Desai & Park, 2005). For encapsulation pur-
diseases, diabetes, or osteoporosis, suggesting that plant poses, modified starch, maltodextrin, gum or other sub-
polyphenols might act as potential chemopreventive and stances are hydrated to be used as the wall materials. The
anti-cancer agents in humans (Arts & Hollman, 2005; core material for encapsulation is homogenized with the
Scalbert, Johnson, & Saltmarsh, 2005; Scalbert, Manach wall materials. The mixture is then fed into a spray dryer
et al., 2005; Surh, 2003). and atomized with a nozzle or spinning wheel. Water is
Unfortunately, the concentrations of polyphenols that evaporated by the hot air contacting the atomized material.
appear effective in vitro are often of an order of magnitude The capsules are then collected after they fall to the bottom
higher than the levels measured in vivo. The effectiveness of the drier (Gibbs et al., 1999). The typical shape of spray
of nutraceutical products in preventing diseases depends dried particles is spherical, with a mean size range of
on preserving the bioavailability of the active ingredients 10e100 mm (Fig. 2).
(Bell, 2001). This is a big challenge, as only a small propor- One limitation of the spray-drying technology is the lim-
tion of the molecules remain available following oral ad- ited number of shell materials available, since the shell ma-
ministration, due to insufficient gastric residence time, terial must be soluble in water at an acceptable level (Desai
low permeability and/or solubility within the gut, as well & Park, 2005). Maltodextrins are widely used for encapsula-
as their instability under conditions encountered in food tion of flavours (Bhandari, 2007), which are also used for
processing and storage (temperature, oxygen, light), or in polyphenol encapsulation. The ethanol extracts of black car-
the gastrointestinal tract (pH, enzymes, presence of other rots, which contain a high level of anthocyanins
nutrients), all of which limit the activity and potential (125  17.22 mg/100 g), have been spray dried using malto-
health benefits of the nutraceutical components, including dextrins as a carrier and coating agents (Ersus & Yurdagel,
polyphenols (Bell, 2001). The delivery of these compounds 2007). High air inlet temperatures (>160e180  C) caused
therefore requires product formulators and manufacturers greater anthocyanin losses, while the maltodextrin of
to provide protective mechanisms that can maintain the ac- 20e21 DE gave the highest anthocyanin content powder at
tive molecular form until the time of consumption, and de- the end of drying process (Ersus & Yurdagel, 2007). The
liver this form to the physiological target within the maltodextrin can also be mixed with gum arabic as wall ma-
organism (Chen, Remondetto, & Subirade, 2006). Some terial. A mixture of maltodextrin (60%) and gum arabic
physicochemical characteristics and food properties of the (40%) has been used for encapsulation of procyanidins
512 Z. Fang, B. Bhandari / Trends in Food Science & Technology 21 (2010) 510e523

Table 1. Major polyphenols, sources and their properties.

Polyphenol groups Examples Sources Properties


Anthocyanidins Cyanidin, delphinidin, malvidin, Fruit, flowers Natural pigments; Highly sensitive to temperature,
pelargonidin, peonidin, petunidin and oxidation, pH, and lights; water soluble
their glycosides.
Catechins Catechin, epicatechin, gallocatechin, Tea Sensitive to oxidation, lights and pH; astringent
epigallocatechin and epigallocatechin and bitter; slightly soluble in water
gallate
Flavanones Hesperetin, hesperidin, homoeriodictyol, Citrus Sensitive to oxidation, lights and pH; aglycones
naringenin, naringin insoluble but glycosides soluble in water
Flavones Apigenin, luteolin, tangeritin Fruit/vegetables Natural pigments; sensitive to oxidation and pH;
aglycones slightly soluble but glycosides soluble in water
Flavonols Kaempferol, myricetin, quercetin and Fruit/vegetables Sensitive to oxidation, lights and pH; aglycones slightly
their glycosides soluble but glycosides soluble in water
Isoflavones Daidzein, genistein, glycitein Soybeans, Sensitive to alkaline pH; astringent and bitter; soy smell;
peanuts water soluble
Hydroxybenzoic Gallic acid, p-hydroxybenzoic, Berries, tea, Sensitive to temperature, oxidation, pH, and lights; most
acids vanillic acid wheat soluble in water
Hydroxycinnamic Caffeic acid, ferulic acid, p-coumaric Fruit, oats, rice Sensitive to oxidation and pH; Most slightly soluble
acids acid, sinapic acid in water
Lignans Pinoresinol, podophyllotoxin, Flax, sesame, Relatively stable under normal conditions; unpleasant
steganacin. vegetables flavour; water soluble.
Tannins Castalin, pentagalloyl glucose, Tea, berries, Sensitive to high temperature and oxidation; astringent
(proanthocyanidines) procyanidins wines, and bitter; water soluble
chocolate

from grape seeds (Zhang, Mou, & Du, 2007). The ratio of 333, showed a lower degradation of its polyphenol content
core substance to wall material was 30:70 w/w, while the and lower reduction of its antioxidant activity, suggesting
concentration of the slurry was 20% w/v. The encapsulation that the correct selection of the drying excipients is an im-
efficiency was up to 88.84%, and the procyanidin was not portant step in guaranteeing the stability and the quality of
changed during drying. The stability of the products was ob- the finished product. The results also indicated that the inlet
viously improved by spray drying. gas temperature had a significant effect on the total poly-
Chitosan has also been used as a wall material in spray phenol, protein and genistein contents of the dried extracts
drying of olive leaf extract (OLE) (Kosaraju, D’ath, & (Georgetti et al., 2008). Another wall material successfully
Lawrence, 2006). The loading percent of polyphenolic used for encapsulation of polyphenol was protein-lipid (so-
compounds was 27%, and the OLE-loaded microspheres dium caseinate-soy lecithin) emulsion, which has been used
normally had a smooth surface morphology. The FTIR in spray drying of grape seed extract, apple polyphenol ex-
spectroscopy results indicated that the majority of the tract and olive leaf extract (Kosaraju, Labbett, Emin,
OLE in the chitosan microsphere were physically encapsu- Konczak, & Lundin, 2008). Optical microscopy and parti-
lated in the chitosan matrix. Chiou and Langrish (2007) in- cle size distribution analysis indicated that the encapsulated
troduced citrus fruit fiber as an encapsulating agent for particles all had spherical morphology and uniform size
spray drying of bioactives extracted from Hibiscus sabdar- distribution. Radical scavenging activity studies demon-
iffa L. The main bioactive compounds in H. sabdariffa L. strated a significant retention of antioxidant activity after
extract are polyphenols, or more specifically, the anthocya- encapsulation by the spray-drying process (Kosaraju
nin complexes. The presence of the bioactive material in et al., 2008).
the fibers did not appear to significantly affect the product
size or shape. The results demonstrated that natural fruit Coacervation
fibers might be a potential replacement carrier for spray The concept behind coacervation microencapsulation is
drying sticky materials. This encapsulation process com- the phase separation of one or many hydrocolloids from
bined two products (fruit fiber and polyphenols) into one the initial solution and the subsequent deposition of the
multipurpose functional food, creating a novel nutraceutical newly formed coacervate phase around the active ingredi-
product suitable for a variety of applications in functional ent suspended or emulsified in the same reaction media
food manufacturing (Chiou & Langrish, 2007). (Gouin, 2004). Coacervation encapsulation can be achieved
More recently, the effects of drying aids comprising col- simply with only one colloidal solute such as gelatin, or
loidal silicon dioxide (tixosil 333), maltodextrin and starch through a more complex process, for example, with gelatin
on spray drying of soybean extract have been studied and gum acacia. Complex coacervation is usually associ-
(Georgetti, Casagrande, Souza, Oliveira, & Fonseca, ated with no definite forms (Fig. 2), and is considered an
2008). The resulting product, to which was added tixosil expensive method for encapsulating food ingredients
Z. Fang, B. Bhandari / Trends in Food Science & Technology 21 (2010) 510e523 513

Fig. 2. Illustration of the characteristics of encapsulated polyphenolic capsules produced by various encapsulation processes.

(Gouin, 2004); however, this process should be related to chitosan. The polyphenols can be retained in a chitosan-al-
the potential benefits it might offer, especially to high- ginate membrane, but maximum release in water was
value, labile functional ingredients, such as the encapsula- achieved in a shorter time for chitosan coated beads than
tion of polyphenols. with the alginate beads. These results implied that the
Yerba mate (Ilex paraguariensis) extract (containing wall materials can affect the release of the natural antioxi-
62.11  1.16 mg of gallic acid/g yerba mate) has been en- dants of yerba mate.
capsulated with two different systems: calcium alginate and Gelatin is a protein containing many glycine, proline and
calcium alginate-chitosan (Deladino, Anbinder, Navarro,& 4-hydroxyproline residues. A new type of protein/polyphe-
Martino, 2008). A high load of active compound (>85%) nol microcapsule based on ()-epigallocatechin gallate
was obtained in the alginate beads but in chitosan coated (EGCG) and gelatin (type A), has been produced using
beads the entrapment was lower (around 50%), on account the layer-by-layer (LbL) assembly method (Shutava,
of the active compound being lost during immersion in Balkundi, & Lvov, 2009). The first layer was a gelatin layer
514 Z. Fang, B. Bhandari / Trends in Food Science & Technology 21 (2010) 510e523

over the MnCO3 microcores, over which an EGCG layer Mozafari & Mortazavi, 2005; Mozafari et al., 2008;
was formed by adding EGCG solution to the gelatin-coated Watwe & Bellare, 1995). A variety of liposome techniques
microparticles. The MnCO3 microcores were dissolved in have been employed for the encapsulation of polyphenols.
EDTA solution to form the stable (Gel A/EGCG)4 capsules. Fan, Xu, Xia, and Zhang (2007) compared the effects of
The EGCG content of the protein/polyphenol film material five different liposome methods on the encapsulation of sal-
was as high as 30% w/w, while the EGCG in the LbL as- idroside e thin film evaporation, sonication, reverse phase
semblies retained its antioxidant activity (Shutava, evaporation, melting, and freezing-thawing. Multilamellar
Balkundi & Lvov, 2009). vesicles can be obtained by thin film evaporation, larger
Glucan is a polysaccharide, and also a thermoreversible unilamellar vesicles by reverse phase evaporation, and
gelling agent, whose gelling behavior depends on its molec- small unilamellar vesicles by sonication or extrusion tech-
ular weight and concentration (Vaikousi, Biliaderis, & nique (Bangham et al., 1965; Cevce, 1993.). The freez-
Izydorczyk, 2004). During cooling of the glucan water so- ing-thawing treatment leads to the production of special
lution from 80  C to room temperature, a network structure freezing-thawing multilamellar vesicles (Maestrelli,
can be formed through an interaction of the chain segment Gonzalez-Rodriguez, Rabasco, & Mura, 2006). The encap-
association and aggregated junction zones (Morgan & sulating efficiency of liposomes is highest when they are
Ofman, 1998). Black currant extract has been encapsulated prepared by freezing-thawing, followed by thin film evapo-
in glucan by simply mixing with hot dispersed glucan gel, ration, then reverse phase evaporation, while melting and
followed by cooling and cutting into cubes, or being drop- sonication has the lowest efficiency. Loading capacity of
ped into oil to produce a bead morphology (Xiong, Melton, salidroside can have significant effects on encapsulating
Easteal, & Siew, 2006). Recovery of 73e79% of encapsu- efficiency, average diameter, and z potential of liposomes.
lated anthocyanins was achieved using normal oven drying Liposomal systems prepared by sonication, melting, and re-
to dehydrate the gel matrix. Larger amounts of anthocya- verse phase evaporation, displayed better dispersivity. Sali-
nins were released from cubes than from beads using the droside liposomes show a slower increase in particle size
same drying process. The encapsulated anthocyanins ex- than liposomes without salidroside, suggesting salidroside
hibited little difference as free radical scavengers, with an plays an important role in preventing the aggregation and
increase in their reducing ability with time. fusion of liposomes. Fan et al. (2007) illustrated that these
Other coacervation coating systems such as gliadin, hep- differences might come from the different morphologies of
arin/gelatin, carrageenan, soy protein, polyvinyl alcohol, liposomes prepared by different methods.
gelatin/carboxymethylcellulose, b-lactoglobulin/gum aca- The nature of the core materials is another factor that af-
cia, and guar gum/dextran have also been studied (Gouin, fects the efficiency of liposome encapsulation. The isomers
2004). However, most of the core materials in these studies of (þ)-catechin and ()-epicatechin entrapped in lipo-
were essential oils rather than polyphenols. somes show similar encapsulation levels and release rates
(Fang, Hwang, Huang, & Fang, 2006). However, another
Liposomes type of catechin, ()-epigallocatechin-3-gallate (EGCG),
Liposomes were first described by Bangham and co- has been observed to have a much higher level of encapsu-
workers in 1965 at Cambridge University (Bangham, lation for the same liposome system. EGCG contains a gal-
Standish, & Watkins, 1965). They are colloidal particles con- loyl group, indicating a greater lipophilicity. Hence it is
sisting of a membranous system formed by lipid bilayers en- possible that EGCG was stronger when to locate within
capsulating aqueous space(s) (Fig. 2). Owing to the the liposome bilayers, thereby increasing the entrapment.
possession of both lipid and aqueous phases, liposomes Liposome encapsulation efficiency can be increased by
can be utilized in the entrapment, delivery, and release of wa- the addition of ethanol (15%) to the preparation hydration
ter soluble, lipid-soluble, and amphiphilic materials. The un- solution (Fang, Lee, Shen, & Huang, 2006). In response,
derlying mechanism for the formation of liposomes and the core material of EGCG in the liposomes showed
nanoliposomes is basically a hydrophilicehydrophobic in- a high rate of encapsulation of nearly 100%, compared to
teraction between phospholipids and water molecules. A ma- 84.6% encapsulation for conventional liposome. It was
jor advantage of their use is the ability to control the release also shown that ethanolic solutions of phospholipids exhibit
rate of the incorporated materials and deliver them to the high encapsulation efficiency for both hydrophilic and lipo-
right place at the right time (Schäfer et al., 1992). Bioactive philic actives (Dayan & Touitou, 2000). The liposomes
agents encapsulated into liposomes can be protected from di- made in the presence of ethanol had a relatively small
gestion in the stomach, and show significant levels of absorp- size of 133.1 nm. The further addition of deoxycholic
tion in the gastrointestinal tract, leading to the enhancement acid (DA) significantly increased the size of the vesicles
of bioactivity and bioavailability (Takahashi et al., 2007). to 378.2 nm. EGCG encapsulated in liposomes with ethanol
There are several methods for producing liposomes, and and DA gave a 20 fold increase in active deposition in basal
there are a number of excellent books and published re- cell carcinomas relative to the free form (Fang et al., 2006).
views that provide details of the most common production The larger vesicle size of this formulation was suggested to
techniques (Betageri & Kulkarni, 1999; Frezard, 1999; be the predominant factor governing this enhancement.
Z. Fang, B. Bhandari / Trends in Food Science & Technology 21 (2010) 510e523 515

Evidence of liposomes enhancing the bioactivity and Inclusion encapsulation


bioavailability of polyphenols has been reported by a num- Molecular inclusion is generally achieved by using cy-
ber of researchers. Curcumin [1,7-bis (4-hydroxy-3-me- clodextrins (CDs) as the encapsulating materials. CDs are
thoxyphenyl)-1,6-hepadiene-3,5-dion] is the principle a group of naturally occurring cyclic oligosaccharides de-
curcuminoid of the popular India spice turmeric, which ex- rived from starch, with six, seven or eight glucose residues
hibits anti-HIV, antitumor, antioxidant, and anti-inflamma- linked by a (1-4) glycosidic bonds in a cylinder-shaped
tory activities (Maheshwari, Singh, Gaddipati, & Srimal, structure, and denominated as a-, b- and g-cyclodextrins,
2006). However, curcumin is poorly absorbed from the gas- in which b- cyclodextrin is commonly applied
trointestinal (GI) tract after oral administration, due to its (Pagington, 1986.) The external part of the cyclodextrin
low water solubility and low stability against GI fluids molecules is hydrophilic, whereas the internal part is hy-
and/or alkaline/higher pH conditions. To enhance the bio- drophobic (Fig. 2). This structure characteristic makes
availability and food functionality of curcumin, liposome- CDs a satisfactory medium for encapsulation of less polar
encapsulated curcumin (LEC) can be prepared from com- molecules (such as essential oils) into the apolar internal
mercially available lecithins (SLP-PC70) and curcumin, cavity through a hydrophobic interaction (Bhandari,
by using a microfluidizer (Takahashi, Uechi, Takara, D’Arcy, & Padukka, 1999; Dziezak, 1998).
Asikin, & Wada, 2009). The resulting LEC is composed One outstanding advantage of the inclusion of polyphe-
of small unilamellar vesicles with a diameter of approxi- nols in CDs is the effect in improving their water solubility,
mately 263 nm, with encapsulation efficiency for curcumin especially for the less water soluble phytochemicals. The
of 68.0%. A faster rate and better absorption were observed inclusion of hesperetin and hesperidin in (2-hydroxy-
for LEC relative to other forms. The results indicated that propyl)-b-cyclodextrin (HP-b-CD) (Tommasini et al.,
curcumin enhanced the gastrointestinal absorption by lipo- 2005), resveratrol in b-CD and maltosyl-b-CDs (Lucas-
some encapsulation, while the plasma antioxidant activity Abellán, Fortea, López-Nicolás, & Núñez-Delicado, 2007),
following oral LEC was significantly higher than that of olive leaf extract (rich in oleuropein) in b-CD (Mourtzinos,
other treatments. Another example reported was quercetin Salta, Yannakopoulou, Chiou, & Karathanos, 2007), querce-
liposomes prepared from egg phosphatidylcholine/choles- tin and myricetin in HP-b-CD, maltosyl-b-CDs and b-CDs,
terol (2:1) (Priprem, Watanatorn, Sutthiparinyanont, (Lucas-Abellán, Fortea, Gabaldón, & Núñez eDelicado,
Phachonpai, & Muchimapura, 2008). The resulting lipo- 2008), kaempferol, quercetin and myricetin in HP-b-CD
somes were approximately 200 nm in mean particle diam- (Mercader-Ros, Lucas-Abellán, Fortea, Gabaldón, &
eter with a negative surface charge and a range of Núñez-Delicado, 2010), 3-hydroxyflavone (3-OHeF),
encapsulation efficiency between 60% and 80%. Both con- morin and quercetin in a- and b-CDs (Calabrò et al.,
ventional and quercetin liposomes have shown anxiolytic 2004), rutin in b-CD (Ding, Chao, Zhang, Shuang, & Pan,
and cognitive-enhancing effects. A lower dose (20 mg/kg 2003) have been studied, and their water solubilities im-
body weight day) and a faster rate of absorption were ob- proved by inclusion encapsulation. In addition, their antiox-
served with intranasal quercetin liposomes when compared idant activities all increased in these CDs encapsulated
with oral quercetin (300 mg/kg body weight/day). The re- systems. The improved antioxidant efficacy of the inclusion
sults suggested that intranasal delivery of quercetin in the complex may come from the protection of the polyphenols
form of liposomes to the brain could allow a reduction in against rapid oxidation by free radicals (Mercader-Ros
the dose and thereby reduce the potential of toxicity of et al., 2010), which may in part be explained by an increase
the quercetin (Priprem et al., 2008). in their solubility in the biological moiety (Ding et al., 2003).
A modified liposome system encapsulating of resveratrol The encapsulation efficacy of CDs inclusion is affected
has been developed, with the encapsulated particles being by the core materials. Generally, the higher the hydropho-
called “acoustically active lipospheres” (AALs) or “microbub- bicity and smaller the molecule is, the greater the affinity
bles” (Fang et al., 2007). The liposome is prepared using dis- for the CDs. For example, based on their relative CDs affin-
solved soybean phosphatidylcholine, cholesterol, co- ity, hesperetin was more effective than hesperidin
emulsifier, and resveratrol in chloroformemethanol. After (Tommasini et al., 2005), and 3-OHeF was more effective
evaporation of the organic solvent and rehydration, AALs than morin or quercetin (Calabrò et al., 2004). On the other
are formed by stabilization using coconut oil and perfluorocar- hand, different wall materials affect the encapsulation ca-
bons. The benefits of AALs are high core material loading ca- pacity for the same core material. For example, a number
pacity (>90%) with a small droplet size (mean diameter of of studies have been reported on the encapsulation of cur-
w300 nm), together an acceptable level of safety, sustained cumin in different CD variants (Tang, Ma, Wang, &
core material release, and high sensitivity to ultrasound treat- Zhang, 2002; Tomren, Masson, Loftsson, & Tonnesen,
ment. The ultrasound sensitivity of AALs is very useful, as 2007; Tonnesen, Masson, & Loftsson, 2002). It has been
they possess the potential to be “magic bullet” agents for shown that HP-b-CD has the highest encapsulation capacity
the delivery of core materials to precise locations in the for curcumin (Tomren et al., 2007). For the core materials
body, with the locations being determined by focusing the ul- of quercetin and myricetin, the affinity to CDs was HP-
trasound energy. b-CD > maltosyl-b-CDs > b-CDs, reflecting the greater
516 Z. Fang, B. Bhandari / Trends in Food Science & Technology 21 (2010) 510e523

affinity of modified cyclodextrins (Lucas-Abellán, Fortea, extract containing caffeoyl derivatives and flavonoids, by
& Gabaldón, 2008) cocrystallization in a supersaturated sucrose solution. The
To illustrate the structures of polyphenol-CD inclusion co-crystallized product had a typically cluster-like agglom-
complexes, some advanced analytical instruments were ap- erate structure with void spaces and a sucrose crystal size
plied. The spatial configuration of the complex of rutin with varying between 2 and 30 mm. An extra layer of a network
b-CD has been proposed, based on NMR and molecular mod- with neat edges covered the crystals. The microstructure
eling (Ding et al., 2003), which revealed that the binding site was further confirmed by differential scanning calorimetry,
for rutin is a single ring of rutin molecule penetrating into the X-ray diffraction and scanning electron microscopy
b-CD cavity in the shallow position, forming a 1:1 inclusion (Deladino, Navarro, & Martino, 2010). The cocrystallization
complex. This structure can be confirmed by 1H NMR and cir- of yerba mate extract changed it from a cohesive material to
cular dichroism spectroscopy (Calabrò et al., 2005). The struc- be a non-cohesive product, and notably reduced its hygro-
ture of the inclusion complex of ferulic acid (FA) with a-CD scopic characteristics without affecting its high solubility;
has been analyzed by rotating frame nuclear overhouser effect this demonstrated that cocrystallization is a good alternative
spectroscopy (ROESY) (Anselmi et al., 2008). Based on this for the preservation and handling of yerba mate extract for
technology and modeling simulation, the insertion of the FA further application in food products. There have been very
into the lipophilic interior of a-CD involves the -COOH and few reports of the application of the cocrystallization
a, b-unsaturated groups and part of its aromatic moiety. The process.
phenol and methoxyl groups of FA lie on the plane of the wider
rim. This encapsulation increases the photo-stability of FA, Nanoencapsulation
slows FA release, and would provide safer and longer-lasting Nanoencapsulation involves the formation of active-
protection of the skin against solar radiation, if applied in cos- loaded particles with diameters ranging from 1 to
metic formulations (Anselmi et al., 2008) 1000 nm (Reis, Neufeld, Ribeiro, & Veiga, 2006). The
With the exception of CDs, other types of biopolymers term nanoparticle is a collective name for both nano-
have been employed in the molecular inclusion of polyphe- spheres and nanocapsules. Nanospheres have a matrix
nols, such as curcumin being encapsulated in hydrophobi- type of structure. Actives may be absorbed at the sphere
cally modified starch (HMS) (Yu & Huang, 2010). The surface or encapsulated within the particle. Nanocapsules
complexed curcumin showed a 1670 fold increase in solu- are vesicular systems in which the active is confined to
bility, possibly reflecting the hydrophobic interaction and a cavity consisting of an inner liquid core surrounded by
hydrogen bonding between curcumin and HMS. The encap- a polymeric membrane (Fig. 2) (Couvreur, Dubernet, &
sulated curcumin revealed enhanced in vitro anti-cancer ac- Puisieux, 1995). The active substances are usually dis-
tivity compared to the free form. solved in the inner core but may also be adsorbed to the
capsule surface (Allémann, Gurny, & Doekler, 1993). It
Cocrystallization is proposed that any target actives, while incorporated
Co-crystallization is an encapsulation process in which the into a complex of polymers, which result in nanoscale-
crystalline structure of sucrose is modified from a perfect to an sized particles, might be called ‘encapsulated nanopar-
irregular agglomerated crystal, to provide a porous matrix in ticles’. Compared to micron-sized particles, nanoparticles
which a second active ingredient can be incorporated (Chen, provide a greater surface area and have the potential to in-
Veiga, & Rizzuto, 1988). Spontaneous crystallization of super- crease solubility due to a combination of large interfacial
saturated sucrose syrup is achieved at high temperature (above adsorption of the core compound, enhanced bioavailabil-
120  C) and low moisture (95e97  Brix). If a second ingredi- ity, improved controlled release, which enable better pre-
ent is added at the same time, the spontaneous crystallization cision targeting of the encapsulated materials (Mozafari
results in the incorporation of the second ingredient into the et al., 2008). A variety of techniques have been employed
void spaces inside the agglomerates of the microsized crystals to develop polyphenol nanoparticles.
(Fig. 2), with a size less than 30 mm (Bhandari, Datta, D’Arcy, Barras et al. (2009) describe the loading of quercetin and
& Rintoul, 1998). The main advantages of cocrystallization EGCG by lipid nanocapsules (LNC) through the applica-
are improved solubility, wettability, homogeneity, dispersibil- tion of the phase inversion process. Briefly, the actives
ity, hydration, anticaking, stability and flowability of the en- were mixed in the oil phase prior to preparation. Soybean
capsulated materials (Beristain, Vázquez, Garcı́a, & Vernon- lecithin, surfactant, NaCl and distilled water were then
Carter, 1996). Other advantages are that the core materials mixed and heated to form a W/O emulsion. The mixture
in a liquid form can be converted to a dry powdered form was cooled, and distilled cold water (0  C) added, with stir-
without additional drying, and the products offer direct tablet- ring to form O/W nanocapsules. The benefits of this method
ing characteristics because of their agglomerated structure, are that the average volume sizes of particles are a function
and thus offer significant advantages to the candy and pharma- of the formulation composition, which means the LNC size
ceutical industries (Desai & Park, 2005). can be tailored by formulation design. The higher encapsu-
Deladino, Anbinder, Navarro, and Martino (2007) re- lated quercetin LNC increased its apparent aqueous solubil-
ported on the encapsulation of yerba mate (I. paraguariensis) ity by a factor of 100. The encapsulated quercetin and
Z. Fang, B. Bhandari / Trends in Food Science & Technology 21 (2010) 510e523 517

()-EGCG have proved to be more stable compared to the (Shao et al., 2009). The mPEGePCL based nanoparticles
free ones. are composed of a hydrophilic segment and a hydrophobic
The nanoprecipitation technique has been used for cur- segment, which are capable of loading the target active by
cumin entrapment, based on poly (lactide-co-glycolide) self assembling into nanoscale spherical structures with
(PLGA) and a stabilizer polyethylene glycol (PEG)-5000 a hydrophilic outer shell and a hydrophobic inner core
(Anand et al., 2010). The nanoprecipitation technique in- (Liu et al., 2008). In this way, lipophilic actives can be en-
volves three steps: First, the target actives and a polymer trapped into the hydrophobic core of the nanosphere, while
are mixed in an organic solution; second, the mixture is its hydrophilic outer shell is maintained as a stabilizer for
added, drop wise, to an aqueous solution, normally contain- the system. Other actives with lipophilicity can also be in-
ing a surfactant; third, the resulting dispersion of nanopar- corporated into this nanoparticle system to enhance their
ticles is vacuum evaporated to eliminate the organic bioavailability.
solvent, and then centrifuged or filtered to obtain the parti- Tea catechins have been successfully encapsulated in chi-
cles. In the case of curcumin-loaded nanoparticles, the en- tosan- tripolyphosphate (CS-TPP) nanoparticles using a sim-
capsulation efficiency was reported to have reached 97.5%, ple ionotropic gelation method (Hu et al., 2008). By
with the particle diameter being about 80.9 nm, which en- controlling the critical fabricating parameters of the CS mo-
hanced its cellular uptake, and increased in vitro bioactivity, lecular mass, CS concentration, and CS-TPP mass ratio, de-
resulting in superior in vivo bioavailability over free curcu- sirable CS-TPP nanoparticles can be spontaneously formed
min (Anand et al., 2010). Other quercetin loaded nanopar- when the freshly prepared CS solution containing tea cate-
ticles were developed by using a similar technique, with chins is added with TPP solution, while stirring at room tem-
a particle size of <85 nm, and encapsulation efficiency of perature (Hu et al., 2008). In comparison with this simple
over 99% (Wu et al., 2008). The encapsulated active of method, a relatively complicated method has been developed
quercetin might have an amorphous state, which formed in- for the encapsulation of polyphenols of EGCG, tannic acid,
termolecular hydrogen bonding with carriers. The release curcumin, and theaflavin (Shutava, Balkundi, Vangala et al.,
of the nanoparticals was 74-fold times higher when com- 2009). First, gelatin nanoparticles are prepared using a two-
pared with the pure active, and possessed more effective an- step desolvation method. These particles are then further
tioxidant activities. encapsulated in polyelectrolytes using a layer-by-layer shell
A method based on the concept of emul- assembly method. Finally, the polyphenols are loaded into
sionediffusioneevaporation, using polyethylene glycol the prepared nanoparticles by adsorption under certain pH
(PEG) 400 as a co-solvent, has been applied on ellagic values. The adsorption of polyphenols to the nanoparticles
acid (EA) loaded PLGA nanoparticles (Bala, Bhardwaj, depends on the chemical nature of the molecules. Adsorption
Hariharan, Kharade, Roy, & Ravi Kumar, 2006). Didode- of polyphenols with higher molecular weights and a larger
cyldimethylammomium bromide (DMAB) and polyvinyl number of phenolic -OH groups was found to be higher.
alcohol (PVA), alone and in combination with chitosan The amount of theaflavin, the polyphenol with the highest
(CS), were used as the stabilizer. The basis of this technique molecular weight among those investigated, was as high as
is as follows: the stirring of the EA-PLGA-PEG 400 mix- 70% of the mass of nanoparticle solid material. Loading of
ture causes the dispersion of the solvent in the form of ir- tannic acid and EGCG is lower, while it is almost negligible
regularly sized droplets in equilibrium with the for curcumin (Shutava, Balkundi, Vangala et al., 2009).
continuous phase, while the stabilizer is adsorbed on to
the larger interface, thereby creating the first emulsion Freeze drying
stage; then, the homogenization results in smaller droplets Freeze drying, also known as lyophilization or cryode-
with more homogenous size distribution; the addition of siccation, is a process used for the dehydration of almost
water and subsequent heating destabilizes the equilibrium all heat-sensitive materials and aromas. Freeze-drying
and causes the organic solvent to diffuse into the aqueous works by freezing the material and then reducing the sur-
phase and then out of the system, leading to precipitation rounding pressure and adding enough heat, to allow the fro-
of the polymer along with the active as very small particles zen water in the material to sublimate directly from the
(Kumar, Bakowsky, & Lehr, 2004). The initial release of solid phase to the gas phase (Oetjen & Haseley, 2004). En-
EA from nanoparticles in pH 7.4 phosphate buffer is rapid, capsulation by freeze drying is achieved as the core
followed by a slower sustained release. An in situ intestinal materials homogenize in matrix solutions and then co-ly-
permeability study in rats showed a higher uptake of active ophilize, usually resulting in uncertain forms (Fig. 2).
encapsulated in nanoparticles prepared using PVA, Except for the long dehydration period required (generally
PVAeCS blend and DMAB as stabilizers, than pure active 20 h), freeze-drying is a simple technique for encapsulating
(Bala et al., 2006). water-soluble essences and natural aromas, as well as drugs
Resveratrol is incorporated into amphiphilic copolymers (Desai & Park, 2005). Freeze dried samples of pomace con-
of mPEGePCL (methoxy poly(ethylene glycol)-poly(cap- taining anthocyanin and maltodextrin DE20 have shown
rolactone)), with an active loading content of good shelf life stability during storage at 50  C/0.5 water
19.4  2.4% and an encapsulation efficiency of >90% activity for up to two months (Delgado-Vargas, Jimenez,
518 Z. Fang, B. Bhandari / Trends in Food Science & Technology 21 (2010) 510e523

& Pardes-Lopez, 2000). Recently, Laine, Kylli, Heinonen, Emulsions


and Jouppila (2008) encapsulated phenolic-rich cloudberry Emulsion technology is generally applied for the encapsu-
extract by freeze drying, using maltodextrins DE5-8 and lation of bioactives in aqueous solutions, which can either be
DE18.5 as wall materials. The microencapsulated cloud- used directly in the liquid state or can be dried to form powders
berry extract offered better protection for phenolics during (e.g., by spray, roller, or freeze drying) after emulsification.
storage, while the antioxidant activity remained the same or Therefore it is actually a part of encapsulation process. Basi-
even improved slightly. cally, an emulsion consists of at least two immiscible liquids,
However, there is also some evidence of freeze drying usually as oil and water, with one of the liquids being dis-
induced encapsulation being unable to improve stability persed as small spherical droplets in the other (Friberg,
or bioactivity. When Hibisus anthocyanin extract was en- Larsson, & Sjoblom, 2004; McClements, 2005). Typically,
capsulated in pullulan by freeze drying, it was only when the diameters of the droplets in food systems range from 0.1
samples stored at higher relative humidity levels to100 mm (McClements et al., 2009). Emulsions can be clas-
(aw>0.75) that the free anthocyanins showed w1.5e1.8 sified according to the spatial organization of the oil and water
times faster degradation than the pullulan-anthocyanin co- phases. A system that consists of oil droplets dispersed in an
lyophilized materials (Gradinaru, Biliaderis, Kallithraka, aqueous phase is called an oil-in-water (O/W) emulsion,
Kefalas, & Garcia-Viguera, 2003). Obviously, this was whereas a system that consists of water droplets dispersed
not a big difference. Furthermore, both free and co-lyophi- in an oil phase is called a water-in-oil (W/O) emulsion
lized with pullulan, Hibiscus anthocyanins exhibited good (Fig. 2). With the exception of the simple O/W or W/O sys-
antiradical activity throughout storage, and no significant tems, various types of multiple emulsions can be developed,
differences were observed between them, suggesting that such as oil-in-water-in-oil (O/W/O) or water-in-oil-in-water
the encapsulation might not be necessary if the Hibisus an- (W/O/W) emulsions (Benichou, Aserin, & Garti, 2004; van
thocyanin extract is to be freeze dried. der Graaf, Schroen, & Boom, 2005). To obtain a kinetically
stable solution, stabilizers such as emulsifiers or texture mod-
ifiers, are commonly added in the emulsion systems. The use
of this technology for delivering food components and nutri-
Yeast encapsulation ceuticals has been comprehensively reviewed by Augustin
Encapsulation of essential oils and flavours by using yeast and Hemar (2009), Flanagan and Singh (2006) and
cells (Saccharomyces cerevisiae) as wall material have McClements et al. (2009).
proven to to be a low cost, high volume process (Bishop, A US patent named “functional emulsions”, relates to
Nelson, & Lamb, 1998). Yeast encapsulation depends on dissolved polyphenols in ethanol (polyglycerol oleic acid
the yeast cells, which allow the actives to pass freely through ester added), which are then stirred with vegetable
the cell wall and membrane, while remaining passively oil in a homogenizer, or emulsified, to obtain E/O type or
within the cells (Fig. 2). Encapsulation by yeast cells can E/O/W type emulsions (Nakajima, Nabetani, Ichikawa, &
control the diffusion of actives through the cell wall and Xu, 2003). These emulsions can be used in pharmaceutical,
membrane, using a defined temperature and time, in nutriceutical or food industries as polyphenol delivery sys-
a pre-determined solution mix, with the wall of the yeast cells tems. Naturally these polyphenols are insoluble or have low
providing protection of the liquid active ingredients against solubility in water and oil, so the obvious advantage of
evaporation, extrusion, oxidation and light (MICAP PLC, these emulsions is that they contain a high concentration
2004).This technology has been typically used for encapsu- of polyphenols. Most recent researches in relation to poly-
lation of small lipophilic molecules such as essential oils. phenol emulsions have been used for the reduction of lipid
The yeast cells have proved to be able to absorb and re- oxidation or increase lipid stability. In one study, after dis-
tain water-soluble flavor compounds when pre-treated with solving Tween 20 in water containing lyophilized tea infu-
a plasmolyser (Serozym Laboratories, 1973). This tech- sion and bovine serum albumin (BSA), sunflower oil (from
nique has been adopted in water soluble polyphenol encap- which tocopherols has been removed) was added dropwise
sulation. After treatment with 5% sodium chloride at 54  C to the aqueous sample in an ice bath and sonicated for
for 24 h for autolysis, the yeast cells can be used to encap- 5 min (Almajano, Carbó, Jiménez, & Gordon, 2008). The
sulate water soluble polyphenol of chlorogenic acid, with W/O emulsions containing tea extracts have shown strong
an encapsulation efficiency of 12.6% (Shi et al., 2007). antioxidant activity against oil oxidation. Another W/O
The yeast encapsulated chlorogenic acid was found to be emulsion prepared by the same research group using a sim-
highly stable under wet and thermal stresses, with the re- ilar method but containing caffeic acid as an antioxidant
lease profiles suggesting that the yeast cells could prevent and Fe (III) as a pro-oxidant ion, also noted the antioxidant
chlorogenic acid from change, without significantly slow- activity of caffeic acid (Almajano, Carbó, Delgado, &
ing down the release. Another obvious benefit of this tech- Gordon, 2007). However, when different polyphenols
nique is that no additives apart from water, yeast and core were used in the W/O emulsions, their antioxidant activities
materials are used during processing, thereby ensuring its showed different characteristics. In the Tween 20-phos-
safety in the food industries (Blanquet et al., 2005). phate buffer-olive oil emulsion system, gallic acid can
Z. Fang, B. Bhandari / Trends in Food Science & Technology 21 (2010) 510e523 519

Table 2. Technologies for encapsulation of polyphenols.

Encapsulation Technologies Polyphenols References


Spray Drying
Wall materials:
Maltodextrins black carrot extracts (anthocyanins) Ersus & Yurdagel, 2007
Maltodextrin and gum arabic procyanidins Zhang et al., 2007
Chitosan olive leaf extract Kosaraju et al., 2006
Citris fruit fiber Hibiscus sabdariffa L. extract Chiou & Langrish, 2007
(anthocyanins)
Colloidal silicon dioxide, maltodextrin and starch soybean extract Georgetti et al., 2008
Sodium caseinate-soy lecithin grape seed extract, apple polyphenol Kosaraju et al., 2008
extract and olive leaf extract
Coacervation
Wall materials:
Calcium alginate and calcium alginateechitosan yerba mate extract Deladino, Anbinder, Navarro, &
Martino, 2008
Gelatin (type A) EGCG Shutava et al., 2009a
Glucan black currant extract Xiong et al., 2006
Liposome
Specific methods:
Thin film evaporation, sonication, reverse phase salidroside Fan et al., 2007
evaporation, melting, and freezing-thawing
Thin film evaporation (þ)-catechin, ()-epicatechin, EGCG Fang, Hwang, Huang, & Fang C.-C, 2006a
Using microfluidizer curcumin Takahashi et al., 2009
Lipid thin film formation and extrusion quercetin Priprem et al., 2008
Thin film evaporation and sonication resveratrol Fan et al., 2007
Inclusion encapsulation
Wall materials:
HP-b-CD hesperetin and hesperidin Tommasini et al., 2005
b-CD and maltosyl-b-CDs resveratrol Lucas-Abellán et al., 2007
b-CD olive leaf extract (rich in oleuropein) Mourtzinos et al., 2007
HP-b-CD, maltosyl-b-CDs and b-CDs quercetin and myricetin Lucas-Abellán et al., 2008
HP-b-CD kaempferol, quercetin and myricetin Mercader-Ros et al., 2010
a- and b-CDs 3-hydroxyflavone, morin and quercetin Calabrò et al., 2004
b-CD rutin Ding et al., 2003
HP- b-CD curcumin Tomren et al., 2007
HP- b-CD, maltosyl-b-CDs, b-CDs, quercetin and myricetin Lucas-Abellán et al., 2008
b-CD rutin Ding et al., 2003
a-CD ferulic acid Anselmi et al., 2008
hydrophobically modified starch curcumin Yu & Huang, 2010
Cocrystallization
Yerba mate extract Deladino et al., 2007
Freeze drying
Wall materials:
Maltodextrin DE20 anthocyanin Delgado-Vargas et al., 2000
Maltodextrins DE 5-8 and DE18.5 cloudberry extract Laine et al., 2008
Pullulan Hibisus anthocyanin Gradinaru et al., 2003
Nanoencapsulation
Specific methods:
Phase inversion quercetin and EGCG Barras et al., 2009
Nanoprecipitation curcumin Anand et al., 2010
Nanoprecipitation quercetin Wu et al., 2008
Emulsionediffusioneevaporation ellagic acid Bala et al., 2006
Amphiphilic copolymers resveratrol Shao et al., 2009
Ionotropic gelation tea catechins Hu et al., 2008
Adsorption to prepared nanoparticles EGCG, tannic acid, curcumin, Shutava et al., 2009b
(layer-by-layer assembly) and theaflavin
Yeast cells
chlorogenic acid Shi et al., 2007
Emulsions
Systems:
Tween 20-BSA- Fe (III)- sunflower oil O/W emulsion caffeic acid Almajano et al., 2007
Tween 20-BSA- sunflower oil O/W emulsion Tea extract Almajano et al., 2008
Tween 20-phosphate buffer-olive oil O/W emulsion Gallic acid, catechin, quercetin Di Mattia et al., 2009
520 Z. Fang, B. Bhandari / Trends in Food Science & Technology 21 (2010) 510e523

stabilise the colloidal properties towards physical instabil- Acknowledgement


ity, while showing low activity towards secondary oxida- This work is supported by the Postdoctoral Research
tion. Catechin showed an interfacial localisation which Fellowship of The University of Queensland. The authors
was reflected in the enhancement of primary oxidation thank Dr John Schiller for his professional proof reading.
and in the inhibition of secondary oxidation. Quercetin
was poorly partitioned in the aqueous phase and had no ef- References
fect on slowing down the bimolecular phase of auto-oxida-
tion (Di Mattia, Sacchetti, Mastrocola, & Pittia, 2009). Allémann, E., Gurny, R., & Doekler, E. (1993). Drug-loaded nanopar-
ticles preparation methods and drug targeting issues. European
These results suggested that not only polarity but also anti- Journal of Pharmaceutics and Biopharmaceutics, 39, 173e191.
oxidant activity, can affect the polyphenol protective role Almajano, M. P., Carbó, R., Delgado, M. E., & Gordon, M. H. (2007).
towards lipids auto-oxidation in emulsions. Effect of pH on the antimicrobial activity and oxidative stability of
oil-in-water emulsions containing caffeic acid. Journal of Food
Science, 72, C258eC263.
Almajano, M. P., Carbó, R., Jiménez, J. A., & Gordon, M. H. (2008).
Antioxidant and antimicrobial activities of tea infusions. Food
Chemistry, 108, 55e63.
Summary and trends Anand, P., Nair, H. B., Sung, B., Kunnumakkara, A. B., Yadav, V. R.,
The abundant work on encapsulation of polyphenols is Tekmal, R. R., et al. (2010). Design of curcumin-loaded PLGA
nanoparticles formulation with enhanced cellular uptake, and in-
summarized in this paper. The characteristics of capsules
creased bioactivity in vitro and superior bioavailability in vivo.
produced by the various encapsulation processes are illus- Biochemical Pharmacology, 79, 330e338.
trated in Fig. 2, which also shows that the different morphol- Anselmi, C., Centini, M., Maggiore, M., Gaggelli, N., Andreassi, M.,
ogies can be achieved by these techniques. All of the work Buonocore, A., et al. (2008). Non-covalent inclusion of ferulic acid
reported and summarized in this paper, has been undertaken with a-cyclodextrin improves photo-stability and delivery: NMR
and modeling studies. Journal of Pharmaceutical and Biomedical
since the year 2000 (Table 2), with the research and related
Analysis, 46, 645e652.
reporting indicating the current worldwide interest in the Arts, I. C. W., & Hollman, P. C. H. (2005). Polyphenols and disease risk
subject. From the literature, it is clear that the utilization in epidemiologic studies. American Journal of Clinical Nutrition,
of encapsulated polyphenols instead of free compounds, 81, 317se325s.
can lead to improvements in both the stability and bioavail- Augustin, M. A., & Hemar, Y. (2009). Nano- and micro-structured as-
semblies for encapsulation of food ingredients. Chemical Society
ability of the compounds in vivo and in vitro, and optimize
Reviews, 38, 902e912.
routes for their administration. Although most of the encap- Bala, I., Bhardwaj, V., Hariharan, S., Kharade, S. V., Roy, N., & Ravi
sulation technologies employed for other chemicals have Kumar, M. N. (2006). Sustained release nanoparticulate formula-
been adopted in polyphenol encapsulation, there are still tion containing antioxidant-ellagic acid as potential prophylaxis
some technologies not being applied for these special phyto- system for oral administration as potential prophylaxis system for
oral administration. Journal of Drug Targeting, 14, 27e34.
chemicals, including spray cooling/chilling, spinning disk
Bangham, A. D., Standish, M. M., & Watkins, J. C. (1965). Diffusion of
and centrifugal coextrusion, extrusion and fluidized bed. univalent ions across the lamellae of swollen phospholipids. Jour-
However, this does not necessary mean that these technolo- nal of Molecular Biology, 13, 238e252.
gies are not suitable for polyphenol encapsulation. Barras, A., Mezzetti, A., Richard, A., Lazzaroni, S., Roux, S.,
Because there is still a lack of direct evidence for the use Melnyk, P., et al. (2009). Formulation and characterization of
polyphenol-loaded lipid nanocapsules. International Journal of
of polyphenols in preventing and treating of human dis-
Pharmaceutics, 379, 270e277.
eases (Scalbert, Manach et al., 2005), most of the polyphe- Bell, L. N. (2001). Stability testing of nutraceuticals and functional
nol encapsulated particles are classified as ‘functional foods. In R. E. C. Wildman (Ed.), Handbook of nutraceuticals and
foods’ or ‘nutriceuticals’, which limits their potential mar- functional foods (pp. 501e516). New York: CRC Press.
kets. In food grade products, cost is an important factor for Benichou, A., Aserin, A., & Garti, N. (2004). Double emulsions sta-
bilized with hybrids of natural polymers for entrapment and slow
their industrialization. Yeast encapsulation of chlorogenic
release of active matters. Advances in Colloid and Interface Sci-
acid is an example of a successful low cost but high volume ence, 108e109, 29e41.
processing (Shi et al., 2007). Future research of polyphenol Bennick, A. (2002). Interaction of plant polyphenols with salivary
encapsulation is likely to focus on aspects of delivery and proteins. Critical Reviews in Oral Biology & Medicine, 13,
the potential use of co-encapsulation methodologies, where 184e196.
Beristain, C. I., Vázquez, A., Garcı́a, H. S., & Vernon-Carter, E. J.
two or more bioactive ingredients can be combined to have
(1996). Encapsulation of orange peel oil by co-crystallization.
a synergistic effect. It can be foreseen that, with a deep un- Lebensmittel-Weissenchaft Und-Technologie, 29, 645e647.
derstanding of the health benefits of polyphenols, improve- Betageri, G. V., & Kulkarni, S. B. (1999). Preparation of liposomes. In
ments in manufacturing technologies, new strategies for R. Arshady (Ed.), Microspheres, microcapsules and liposomes.
stabilization of fragile nutraceuticals, and the development Preparation and chemical applications, Vol. 1 (pp. 489e521).
London: Citus Books.
of novel approaches to site-specific carrier targeting, encap-
Bhandari, B. R. (2007). Spray drying and powder properties. In
sulated polyphenols will play an important role in increas- Y. H. Hui, C. Clary, M. M. Farid, O. O. Fasina, A. Noomhorm, &
ing the efficacy of functional foods or even J. Welti-Chanes (Eds.), Food drying science and technology
pharmaceuticals, over the next decade. (pp. 215e248). USA: DEStech Publications, Inc.
Z. Fang, B. Bhandari / Trends in Food Science & Technology 21 (2010) 510e523 521

Bhandari, B. R., D’Arcy, B. D., & Padukka, I. (1999). Encapsulation of with rutin. Spectrochimica Acta Part A: Molecular and Biomolec-
lemon oil by paste method using b-cyclodextrin: encapsulation ular Spectroscopy, 59, 3421e3429.
efficiency and profile of oil volatiles. Journal of Agricultural and Dziezak, J. D. (1998). Microencapsulation and encapsulated food in-
Food Chemistry, 47, 5194e5197. gredients. Food Technology, 42, 136e151.
Bhandari, B. R., Datta, N., D’Arcy, B. R., & Rintoul, G. B. (1998). Co- Ersus, S., & Yurdagel, U. (2007). Microencapsulation of anthocyanin
crystallization of honey with sucrose. Lebensmittel-Wissenschaft pigments of black carrot (Daucus carota L.) by spray drier. Journal
und-Technologie, 31, 138e142. of Food Engineering, 80, 805e812.
Bishop, J. R. P., Nelson, G., & Lamb, J. (1998). Microencapsulation in Fan, M. H., Xu, S. Y., Xia, S. Q., & Zhang, X. M. (2007). Effect of
yeast cells. Journal of Microencapsulation, 15, 761e773. different preparation methods on physicochemical properties of
Blanquet, S., Garrait, G., Beyssac, E., Perrier, C., Denis, S., salidroside liposomes. Journal of Agricultural and Food Chemistry,
Hébrard, G., et al. (2005). Effects of cryoprotectants on the viability 55, 3089e3095.
and activity of freeze dried recombinant yeasts as novel oral drug Fang, J.-Y., Hwang, T.-L., Huang, Y.-L., & Fang, C.-L. (2006).
delivery systems assessed by an artificial digestive system. Euro- Enhancement of the transdermal delivery of catechins by liposomes
pean Journal of Pharmaceutics and Biopharmaceutics, 61, 32e39. incorporating anionic surfactants and ethanol. International Journal
Calabrò, M. L., Tommasini, S., Donato, P., Stancanelli, R., Raneri, D., of Pharmaceutics, 310, 131e138.
Catania, S., et al. (2005). The rutin/ b-cyclodextrin interactions in fully Fang, J.-Y., Lee, W.-R., Shen, S.-C., & Huang, Y.-L. (2006). Effect of
aqueous solution: spectroscopic studies and biological assays. Jour- liposome encapsulation of tea catechins on their accumulation in
nal of Pharmaceutical and Biomedical Analysis, 36, 1019e1027. basal cell carcinomas. Journal of Dermatological Science, 42,
Calabrò, M. L., Tommasini, S., Donato, P., Raneri, D., Stancanelli, R., 101e109.
Ficarra, P., et al. (2004). Effects of a- and b-cyclodextrin com- Fang, J.-Y., Hung, C.-F., Liao, M.-H., & Chien, C.-C. (2007). A study of
plexation on the physico-chemical properties and antioxidant ac- the formulation design of acoustically active lipospheres as carriers
tivity of some 3-hydroxyflavones. Journal of Pharmaceutical and for drug delivery. European Journal of Pharmaceutics and Bio-
Biomedical Analysis, 35, 365e377. pharmaceutics, 67, 67e75.
Cevce, G. (1993). Lipid properties as a basis for membrane modeling and Flanagan, J., & Singh, H. (2006). Microemulsions: a potential delivery
rational liposome design. In G. Gregoriadis (Ed.) (2nd ed.).Liposome system for bioactives in food. Critical Reviews in Food Science and
technology, Vol. 1 (pp. 1e36) Boca Raton: CRC Press. Nutrition, 46, 221e237.
Chen, A. C., Veiga, M. F., & Rizzuto, A. B. (1988). Cocrystallization: an Frezard, F. (1999). Liposomes: from biophysics to the design of peptide
encapsulation process. Food Technology, 11, 87e90. vaccines. Brazilian Journal of Medical and Biological Research, 32,
Chiou, D., & Langrish, T. A. G. (2007). Development and character- 181e189.
isation of novel nutraceuticals with spray drying technology. Jour- Friberg, S., Larsson, K., & Sjoblom, J. (2004). Food emulsions, (4th ed.).
nal of Food Engineering, 82, 84e91. New York: Marcel Dekker.
Chen, L., Remondetto, G. E., & Subirade, M. (2006). Food protein- Georgetti, S. R., Casagrande, R., Souza, C. R. F., Oliveira, W. P., &
based materials as nutraceutical delivery systems. Trends in Food Fonseca, M. J. V. (2008). Spray drying of the soybean extract: effects
Science & Technology, 17, 272e283. on chemical properties and antioxidant activity. LWT -Food
Couvreur, P., Dubernet, C., & Puisieux, F. (1995). Controlled drug Science and Technology, 41, 1521e1527.
delivery with nanoparticles: current possibilities and future trends. Gibbs, B. F., Kermasha, S., Alli, I., & Mulligan, C. N. (1999). Encap-
European Journal of Pharmaceutics and Biopharmaceutics, 41, sulation in the food industry: a review. International Journal of
2e13. Food Sciences and Nutrition, 50, 213e224.
Dayan, N., & Touitou, E. (2000). Carriers for skin delivery of trihexy- Gouin, S. (2004). Microencapsulation: industrial appraisal of existing
phenidyl HCl: ethosomes vs. liposomes. Biomaterials, 21, technologies and trends. Trends in Food Science and Technology,
1879e1885. 15, 330e347.
de Vos, P., Faas, M. M., Spasojevic, M., & Sikkema, J. (2010). Encapsu- Gradinaru, G., Biliaderis, C. G., Kallithraka, S., Kefalas, P., &
lation for preservation of functionality and targeted delivery of bio- Garcia-Viguera, C. (2003). Thermal stability of Hibiscus sabdariffa
active food components. International Dairy Journal, 20, 292e302. L. anthocyanins in solution and in solid state: effects of copig-
Deladino, L., Anbinder, P. S., Navarro, A. S., & Martino, M. N. (2007). mentation and glass transition. Food Chemistry, 83, 423e436.
Co-crystallization of yerba mate extract (Ilex paraguariensis) and Haslam, E. (1996). Natural polyphenols (vegetable tannins) as drugs:
mineral salts within a sucrose matrix. Journal of Food Engineering, possible modes of action. Journal of Natural Product, 59,
80, 573e580. 205e215.
Deladino, L., Anbinder, P. S., Navarro, A. S., & Martino, M. N. (2008). Haslam, E., & Lilley, T. H. (1988). Natural astringency in foodstuffsda
Encapsulation of natural antioxidants extracted from Ilex para- molecular interpretation. Critical Reviews of Food Science and
guariensis. Carbohydrate Polymers, 71, 126e134. Nutrition, 27, 1e40.
Deladino, L., Navarro, A. S., & Martino, M. N. (2010). Microstructure Hu, B., Pan, C., Sun, Y., Hou, Z., Ye, Y., Hu, B., et al. (2008). Opti-
of minerals and yerba mate extract co-crystallized with sucrose. mization of fabrication parameters to produce chitosan-tripoly-
Journal of Food Engineering, 96, 410e415. phosphate nanoparticles for delivery of tea catechins. Journal of
Delgado-Vargas, F., Jimenez, A. R., & Pardes-Lopez, O. (2000). Natural Agricultural and Food Chemistry, 56, 7451e7458.
pigments: carotenoids, anthocyanins and betalainsecharacteris- Jafari, S. M., Assadpoor, E., He, Y., & Bhandari, B. (2008). Encapsula-
tics, biosynthesis, processing and stability. Critical Reviews in Food tion efficiency of food flavours and oils during spray drying. Drying
Science and Nutrition, 40, 173e289. Technology, 26, 816e835.
Desai, K. G. H., & Park, H. J. (2005). Recent developments in micro- Khaled, Al-T., & Jagdish, S. (2007). Smart polymer based delivery
encapsulation of food ingredients. Drying Technology, 23, systems for peptides and proteins. Recent Patents on Drug Delivery
1361e1394. & Formulation, 1, 65e71.
Di Mattia, C. D., Sacchetti, G., Mastrocola, D., & Pittia, P. (2009). Kosaraju, S. L., D’ath, L., & Lawrence, A. (2006). Preparation and
Effect of phenolic antioxidants on the dispersion state and chemical characterisation of chitosan microspheres for antioxidant delivery.
stability of olive oil O/W emulsions. Food Research International, Carbohydrate Polymers, 64, 163e167.
42, 1163e1170. Kosaraju, S. L., Labbett, D., Emin, M., Konczak, I., & Lundin, L. (2008).
Ding, H., Chao, J., Zhang, G., Shuang, S., & Pan, J. (2003). Preparation Delivering polyphenols for healthy ageing. Nutrition & Dietetics,
and spectral investigation on inclusion complex of b-cyclodextrin 65, S48eS52.
522 Z. Fang, B. Bhandari / Trends in Food Science & Technology 21 (2010) 510e523

Kumar, M. N. V. R., Bakowsky, U., & Lehr, C. M. (2004). Preparation Priprem, A., Watanatorn, J., Sutthiparinyanont, S., Phachonpai, W., &
and characterization of cationic PLGA nanospheres as DNA car- Muchimapura, S. (2008). Anxiety and cognitive effects of quercetin
riers. Biomaterials, 25, 1771e1777. liposomes in rats. Nanomedicine: Nanotechnology, Biology, and
Laine, P., Kylli, P., Heinonen, M., & Jouppila, K. (2008). Storage stability Medicine, 4, 70e78.
of microencapsulated cloudberry (Rubus chamaemorus) phenolics. Quideau, S., & Feldman, K. S. (1996). Ellagitannin chemistry. Chemi-
Journal of Agricultural and Food Chemistry, 56, 11251e11261. cal Reviews, 96, 475e503.
Liu, B., Yang, M., Li, X., Qian, X., Shen, Z., Ding, Y., et al. (2008). Reis, C. P., Neufeld, R. J., Ribeiro, A. J., & Veiga, F. (2006). Nanoencapsula-
Enhanced efficiency of thermally targeted taxanes delivery in tion I. Methods for preparation of drug-loaded polymeric nanoparticles.
a human xenograft model of gastric cancer. Journal of Pharma- Nanomedicine: Nanotechnology, Biology, and Medicine, 2, 8e21.
ceutical Sciences, 97, 3170e3181. Scalbert, A., Johnson, I. T., & Saltmarsh, M. (2005). Polyphenols: an-
Lucas-Abellán, C., Fortea, M. I., Gabaldón, J. A., & Núñez tioxidants and beyond. American Journal of Clinical Nutrition, 81,
-Delicado, E. (2008). Encapsulation of quercetin and myricetin in 215Se217S.
cyclodextrins at acidic pH. Journal of Agricultural and Food Scalbert, A., Manach, C., Morand, C., Rémésy, C., & Jiménez, L.
Chemistry, 56, 255e259. (2005). Dietary polyphenols and the prevention of diseases. Criti-
Lucas-Abellán, C., Fortea, M. I., López-Nicolás, J. M., & Núñez- cal Reviews in Food Science and Nutrition, 45(20), 287e306.
Delicado, E. (2007). Cyclodextrins as resveratrol carrier system. Schäfer, V., von Briesen, H., Andreesen, R., Steffan, A. M., Royer, C.,
Food Chemistry, 104, 39e44. Tröster, S., et al. (1992). Phagocytosis of nanoparticles by human
Maestrelli, F., Gonzalez-Rodriguez, M. L., Rabasco, A. M., & Mura, P. immunodeficiency virus (HIV)-infected macrophages: a possibility
(2006). Effect of preparation technique on the properties of lipo- for antiviral drug targeting. Pharmaceutical Research, 9, 541e546.
somes encapsulating ketoprofen-cyclodextrin complexes aimed for Schrooyen, P. M. M., van der Meer, R., & De Kruif, C. G. (2001). Mi-
transdermal delivery. International Journal of Pharmaceutics, 298, croencapsulation: its application in nutrition. Proceedings of the
53e60. Nutrition Society, 60, 475e479.
Maheshwari, R. K., Singh, A. K., Gaddipati, J., & Srimal, R. C. (2006). Serozym Laboratories. (1973). French patent specification no. 2179528.
Multiple biological activities of curcumin: a short review. Life Shahidi, F., & Han, X. Q. (1993). Encapsulation of food ingredients.
Sciences, 78, 2081e2087. Critical Reviews in Food Science and Nutrition, 33, 501e547.
Manach, C., Scalbert, A., Morand, C., Rémésy, C., & Jiménez, L. Shao, J., Li, X., Lu, X., Jiang, C., Hu, Y., Li, Q., et al. (2009). Enhanced
(2004). Polyphenols: food sources and bioavailability. American growth inhibition effect of resveratrol incorporated into biode-
Journal of Clinical Nutrition, 79, 727e747. gradable nanoparticles against glioma cells is mediated by the in-
McClements, D. J. (2005). Food emulsions: Principles, practice, and duction of intracellular reactive oxygen species levels. Colloids
techniques, (2nd ed.). Boca Raton: CRC Press. and Surfaces B: Biointerfaces, 72, 40e47.
McClements, D. J., Decker, E. A., Park, Y., & Weiss, J. (2009). Structural Shi, G., Rao, L., Yu, H., Xiang, H., Pen, G., Long, S., et al. (2007). Yeast-
design principles for delivery of bioactive components in nutra- cell-based microencapsulation of chlorogenic acid as a water-sol-
ceuticals and functional foods. Critical Reviews in Food Science uble antioxidant. Journal of Food Engineering, 80, 1060e1067.
and Nutrition, 49, 577e606. Shutava, T. G., Balkundi, S. S., & Lvov, Y. M. (2009). ()-Epigalloca-
Micap Plc. (2004). Micro-organism microcapsules, UK patent appli- techin gallate/gelatin layer-by-layer assembled films and micro-
cation, GB2396107A. capsules. Journal of Colloid and Interface Science, 330, 276e283.
Mercader-Ros, M. T., Lucas-Abellán, C., Fortea, M. I., Gabaldón, J. A., Shutava, T. G., Balkundi, S. S., Vangala, P., Steffan, J. J., Bigelow, R. L.,
& Núñez-Delicado, E. (2010). Effect of HP-b-cyclodextrins com- Cardelli, J. A., et al. (2009). Layer-by-layer-coated gelatin nano-
plexation on the antioxidant activity of flavonols. Food Chemistry, particles as a vehicle for delivery of natural polyphenols. ACS
118, 769e773. Nano, 3, 1877e1885.
Morgan, K., & Ofman, J. (1998). Glucagel, a gelling b-glucan from Surh, Y. J. (2003). Cancer chemoprevention with dietary phytochemi-
barley. Cereal Chemistry, 75, 879e881. cals. Nature Reviews Cancer, 3, 768e780.
Mozafari, M. R. (2005). Liposomes: an overview of manufacturing Tang, B., Ma, L., Wang, H. Y., & Zhang, G. Y. (2002). Study on the
techniques. Cellular & Molecular Biology Letters, 10, 711e719. supramolecular interaction of curcumin and beta-cyclodextrin by
Mozafari, M. R. (2006). Bioactive entrapment and targeting using spectrophotometry and its analytical application. Journal of Agri-
nanocarrier technologies: an introduction. In M. R. Mozafari (Ed.), cultural and Food Chemistry, 50, 1355e1361.
Nanocarrier technologies: Frontiers of nanotherapy (pp. 1e16). Takahashi, M., Inafuku, K., Miyagi, T., Oku, H., Wada, K., Imura, T.,
The Netherlands: Springer. et al. (2007). Efficient preparation of liposomes encapsulating
Mozafari, M. R., Khosravi-Darani, K., Borazan, G. G., Cui, J., food materials using lecithins by a mechanochemical method.
Pardakhty, A., & Yurdugul, S. (2008). Encapsulation of food ingre- Journal of Oleo Science, 56, 35e42.
dients using nanoliposome technology. International Journal of Takahashi, M., Uechi, S., Takara, K., Asikin, Y., & Wada, K. (2009).
Food Properties, 11, 833e844. Evaluation of an oral carrier system in rats: bioavailability and
Mozafari, M. R., & Mortazavi, S. M. (2005). Nanoliposomes: From antioxidant properties of liposome-capsulated curcumin. Journal of
fundamentals to recent developments. Oxford: Trafford Pub. Ltd. Agricultural and Food Chemistry, 57, 9141e9146.
Mourtzinos, I., Salta, F., Yannakopoulou, K., Chiou, A., & Karathanos, V. T. Tommasini, S., Calabrò, M. L., Stancanelli, R., Donato, P., Costa, C.,
(2007). Encapsulation of olive leaf extract in becyclodextrin. Journal Catania, S., et al. (2005). The inclusion complexes of hesperetin
of Agricultural and Food Chemistry, 55, 8088e8094. and its 7-rhamnoglucoside with (2-hydroxypropyl)-b-cyclodextrin.
Nakajima, M., Nabetani, H., Ichikawa, S., & Xu, Q.Y. (2003). Func- Journal of Pharmaceutical and Biomedical Analysis, 39, 572e580.
tional emulsions. US Patent, US6538019B1. Tomren, M. A., Masson, M., Loftsson, T., & Tonnesen, H. H. (2007).
Oetjen, G.-W., & Haseley, P. (2004). Freeze-drying. Weinheim: Wiley- Studies on curcumin and curcuminoids XXXI. Symmetric and
VCH Verlag Gmbh & Co. KGaA. asymmetric curcuminoids: stability, activity and complexation
Pagington, J. S. (1986). a-Cyclodextrin and its uses in the flavour in- with cyclodextrin. International Journal of Pharmaceutics, 338,
dustry. In G. G. Birch, & M. G. Lindley (Eds.), Developments in 27e34.
food flavours. London: Elsevier Applied Science. Tonnesen, H. H., Masson, M., & Loftsson, T. (2002). Studies of cur-
Peter, S., & Given Jr., (2009). Encapsulation of flavors in emulsions for cumin and curcuminoids. XXVII. Cyclodextrin complexation: sol-
beverages. Current Opinion in Colloid & Interface Science, 14, ubility, chemical and photochemical stability. International Journal
43e47. of Pharmaceutics, 244, 127e135.
Z. Fang, B. Bhandari / Trends in Food Science & Technology 21 (2010) 510e523 523

Vaikousi, H., Biliaderis, C. G., & Izydorczyk, M. S. (2004). Solution flow antioxidant effects of quercetin nanoparticles. International Journal
behavior and gelling properties of water-soluble barley (1/3, 1/4)- of Pharmaceutics, 346, 160e168.
b-glucans varying in molecular size. Cereal Science, 39, 119e137. Xiong, S., Melton, L. D., Easteal, A., & Siew, D. (2006). Stability and
van der Graaf, S., Schroen, C. G. P. H., & Boom, R. M. (2005). Prep- antioxidant activity of black currant anthocyanins in solution and
aration of double emulsions by membrane emulsificationea encapsulated in glucan gel. Journal of Agricultural and Food
review. Journal of Membrane Science, 251, 7e15. Chemistry, 54, 6201e6208.
Vilstrup, P. (2001). Microencapsulation of food ingredients. Surrey: Yu, H., & Huang, Q. (2010). Enhanced in vitro anti-cancer activity of
Leatherhead Pub. curcumin encapsulated in hydrophobically modified starch. Food
Watwe, R., & Bellare, J. (1995). Manufacture of liposomes: a review. Chemistry, 119, 669e674.
Current Science, 68, 715e724. Zhang, L., Mou, D., & Du, Y. (2007). Procyanidins: extraction and
Wu, T.-H., Yen, F.-L., Lin, L.-T., Tsai, T.-R., Lin, C.-C., & Cham, T.-M. micro-encapsulation. Journal of Agricultural and Food Chemistry,
(2008). Preparation, physicochemical characterization, and 87, 2192e2197.

You might also like