Applications of Curcumin

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Trends in Food Science & Technology 88 (2019) 445–458

Contents lists available at ScienceDirect

Trends in Food Science & Technology


journal homepage: www.elsevier.com/locate/tifs

Review

Application of curcumin-loaded nanocarriers for food, drug and cosmetic T


purposes
Zahra Rafieea, Mohammad Nejatianb, Marjan Daeihamedc, Seid Mahdi Jafaria,∗
a
Department of Food Materials and Process Design Engineering, Gorgan University of Agricultural Science and Natural Resources, Gorgan, Iran
b
Department of Food Science and Technology, Tarbiat Modares University, Tehran, Iran
c
Department of Pharmaceutics, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran

A R T I C LE I N FO A B S T R A C T

Keywords: Background: Curcumin, the major bioactive material of turmeric, has wide functional features such as anti-
Curcumin diabetic, anti-cancer, anti-inflammatory, and antioxidant activities. However, curcumin exhibits some major
Nanocarriers limitations including low water solubility, degradation during processing or within the gastrointestinal tract and
Bioavailability alkaline conditions, leading to decrease of its bioavailability. For these reasons, many studies have tried to
Foods
improve functionality of curcumin and overcome its limitations through delivery systems, especially na-
Pharmaceuticals
noencapsulation.
Cosmetics
Scope and approach: In this review, practical applications of the nanoencapsulated curcumin, prepared by var-
ious techniques in different food, pharmaceutical and cosmetic products have been considered and explained.
The importance of nanoencapsulation in improving functional effects of curcumin in processed foods, such as
coloring, antioxidant and antimicrobial activity has been discussed thoroughly and its potential applications for
medicinal proposes including cancer, cardiovascular and neurodegenerative diseases, viral and bacterial infec-
tions have been reviewed.
Key findings and conclusions: Nano-delivery systems, because of exclusive characteristics of nanoparticles as well
as conventional properties of an encapsulation medium, can provide chemical stability, hydrophilicity, sustained
release and adequate dispersibility of curcumin in final products (food, cosmetics and drug). Consequently, the
pharmacokinetic properties of curcumin including biological half-life and bioavailability (or bioaccessibility)
can be improved resulting in better clinical and functional efficacy in vivo. Although the potentials of curcumin
nanoformulations have been extensively studied in the literature, future studies can better help to find optimum
formulations for clinical and industrial applications of nanoencapsulated curcumin.

1. Introduction curcumin. It has been approved by the Scientific Committee for Food of
the European Community and presents the number of E100 in the list of
Curcumin is a polyphenol of low molecular weight (368.37 g mol−1) additives. Curcumin can be used as an antioxidant, flavoring agent
existing in the Curcuma longa (turmeric) rhizomes (ARAIZA-Calahorra, (warm, bitter taste) and natural colorant (a bright yellow color) in
Akhtar, & Sarkar, 2018). This perennial herb contains 3–5% of the four various foods or even culinary applications (Aguilar et al., 2010).
types of curcuminoid derivatives including curcumin, demethox- Turmeric and curcumin have been known and widely applied in tra-
ycurcumin, bisdemethoxycurcumin, and cyclocurcumin, so that cur- ditional medicine for treating various diseases like acne, injuries, am-
cumin is the main (77%) bioactive ingredient (Tayyem, Heath, AL- bustions, eye infections, skin diseases, stress, and depression (Hatcher,
Delaimy, & Rock, 2006; Heger, Van Golen, Broekgaarden, & Michel, Planalp, Cho, Torti, & Torti, 2008). Researches in the recent three
2014). Chemically, curcumin belongs to diarylheptanoids and consists decades have shown the remedial effects of this molecule on different
of two aromatic rings bearing two hydroxyl and two methoxyl groups. kinds of diseases including cancer, pulmonary and chronic kidney dis-
The phenolic rings are joined by the aliphatic unsaturated carbon chain orders, diseases of the nervous system, metabolic disease, cardiovas-
with two carbonyl groups placed at C-3 and C-5 as shown in Fig. 1 cular disease and other inflammatory diseases (Aggarwal & Sung, 2009;
(Nelson et al., 2017; Rafiee, Nejatian, Daeihamed, & Jafari, 2018). Kannappan, Gupta, Kim, Reuter, & Aggarwal, 2011). According to
Many functional and biological features have been reported for current scientific evidences, curcumin is severely an anti-inflammation


Corresponding author.
E-mail address: smjafari@gau.ac.ir (S.M. Jafari).

https://doi.org/10.1016/j.tifs.2019.04.017
Received 6 December 2018; Received in revised form 19 April 2019; Accepted 19 April 2019
Available online 24 April 2019
0924-2244/ © 2019 Elsevier Ltd. All rights reserved.
Z. Rafiee, et al. Trends in Food Science & Technology 88 (2019) 445–458

Fig. 1. Chemical structure of curcumin, its loading within nanocarriers (liposome) and application of nanoencapsulated curcumin in different industries.

and wound healing agent and shows antimicrobial effects against yeasts 2. Nanoencapsulated curcumin for the food industry
and molds, as well as various species of Gram-negative and Gram-po-
sitive bacteria (Silva et al., 2018). However, poor water solubility is the Fortification of food products with bioactive compounds such as
main challenge in industrial applications of curcumin (11 ng/mL curcumin has attracted growing consideration due to increment of
to < 10 μg/mL) (Tomren, Masson, Loftsson, & Tønnesen, 2007; Wang awareness and tendency of consumers towards healthy diets
et al., 2008). Additionally, it may be degraded under processing and (Assadpour, Maghsoudlou, Jafari, Ghorbani, & Aalami, 2016; Faridi
within the gastrointestinal tract (GIT) conditions which consequently Esfanjani & Jafari, 2016). Curcumin is not only commonly utilized as a
decreases its bioavailability. Curcumin is stable in the systems with food colorant, but also it can be utilized within food products as a safe
physiological pH values as well as acidic conditions, but under alkaline and natural antimicrobial and antioxidant agent instead of synthetic
conditions, it is easily decomposed (ARAIZA-Calahorra et al., 2018). ones (Assadpour & Jafari, 2018; Gómez-Estaca, Gavara, & Hernández-
Incorporation of curcumin into nanocarriers by different techniques is a Muñoz, 2015). Previous research works have confirmed notable anti-
suitable and efficient option for improving its solubility, stability and oxidant and antimicrobial efficiency of curcumin toward different
functionality (Assadpour, Jafari, & Esfanjani, 2017; Faridi Esfanjani, strains of bacteria plus some fungal species (Bhawana et al., 2011;
Assadpour, & Jafari, 2018). Moreover, the nanocarriers are capable of Dovigo et al., 2013; Dubey, Sharma, Narain, Misra, & Pati, 2008; Hu,
releasing curcumin in target sites and therefore have being exploited in Huang, & Chen, 2013; Mun et al., 2013; Sathishkumar et al., 2015).
pharmaceutical and food supplements industries. However, food applications of curcumin have been limited by some
In a recent article, we reviewed the existing literature on different obstacles. Curcumin displays a low water solubility and inadequate
techniques of curcumin nanoencapsulation for the food and nu- dispersibility in food systems owing to its hydrophobic nature (Huang
traceutical practices (Rafiee et al., 2018). Here, feasible applications of et al., 2016; Nguyen et al., 2017). It is also decomposed in contact with
such curcumin-loaded nanoformulations in food, pharmaceutical and adverse circumstances during storage and food processing, reducing its
cosmetic sectors were comprehensively underlined. This would be efficiency and bioactivity. Moreover, direct usage of curcumin in food
useful for readers and researchers to expand their perspective in man- products may cause negative effects on sensorial attributes along with
ufacturing of functional products. unpleasant interactions with food components (Fang & Bhandari, 2010;
Mehanny, Hathout, Geneidi, & Mansour, 2016; Paramera, Konteles, &

446
Z. Rafiee, et al. Trends in Food Science & Technology 88 (2019) 445–458

Karathanos, 2011). Nanoencapsulation techniques can improve anti- containing nanocurcumin received a similar sensory score. Moreover,
microbial and antioxidant capacity of phenolic compounds such as nanocurcumin remained stable against the processing conditions of ice
curcumin, because they provide sustained release, good stability, en- cream and its encapsulation efficiency was slightly decreased. Hence,
hanced solubility in aqueous mediums for bioactives, therefore leading ice cream could be suggested as a suitable delivery system for nano-
to promote their functionality and bioactivity without any considerable curcumin.
change in quality and sensory properties of fortified food (Abdou, In a recent work by Almeida et al. (2018), curcumin as a natural
Galhoum, & Mohamed, 2018; Faridi Esfanjani & Jafari, 2016; Hussain colorant agent was applied in yogurt samples in diverse forms of water-
et al., 2017; Jafari, 2017b). dispersible, powder and nanoparticles prepared via solid dispersion.
Several researches have been carried out on nanoencapsulation of Curcumin powder displayed the maximum antioxidant, antimicrobial,
curcumin by various carriers to solve its limitations when directly cytotoxic and anti-inflammatory effects as compared to other forms of
added into food systems. As an example, Sadeghi et al. (2014) for- curcumin which is explainable by considering protective effect of en-
mulated curcumin-loaded nanoparticles via desolvation method with capsulation on curcumin and decease of its availability for a short
the aid of bovine serum albumin (BSA) and different solvents. Based on period of time. Yogurt samples containing curcumin had acceptable
their results, acetone gave the highest nanoparticulation efficiency sensory properties and curcumin made no remarkable effects on their
(99.2%) and nanoparticles with rod and spherical morphology. More- fatty acids profiles as well as nutritional quality over storage time.
over, all samples had a PDI lower than 0.2 and encapsulation efficiency However, yogurt samples treated with modified curcumin were pre-
was dependent on concentrations of curcumin and BSA. Glutaraldehyde ferred to those containing free (unencapsulated) curcumin regarding
as crosslinking agent led to enhancement of storage stability in nano- sensory attributes especially color and dispersibility which are sa-
particles. Evaluation of release behavior of loaded nanoparticles by tisfactory for food applications (Almeida et al., 2018).
dialysis tubing method revealed high ability of BSA nanoparticles for
the gradual release of curcumin (Sadeghi et al., 2014). another study, 2.2. Antioxidant activity of nanoencapsulated curcumin
starch nanoparticles were synthetized through nanoprecipitation tech-
nique using different corn starches for encapsulation of curcumin Curcumin as a phenolic compound presents an outstanding anti-
(Sadeghi, Daniella, Uzun, & Kokini, 2017). Nanoparticles produced by oxidant activity resulted from hydroxyl group of phenol rings and
ethanol and normal corn starch had a particle size below 100 nm and protects foods from oxidation. Besides, it plays a protective role in
the highest uniformity. Moreover, curcumin-loaded starch nano- human body against free radicals (Abdel Fattah, 2016). Curcumin exert
particles were more stable compared to free one when stored for 10 antioxidant activity through multiple ways. It is able to trap various
days at room temperature in PBS (pH = 7). A mixture of BSA and poly- kinds of free radicals mainly reactive nitrogen and oxygen species.
D-lysine (PDL) with low and high molecular weights were also used to Curcumin not only improves performance of enzymes with free radicals
design soluble coacervate nanoparticles for the loading of curcumin. scavenging activity like glutathione peroxidase, superoxide dismutase,
According to the results, molecular weight, pH, concentration of salt and catalase, but also shows inhibitory effects on enzymes which gen-
and PDL/BSA ratio significantly affected characteristics of coacervate erate free radicals like xanthine oxidase/hydrogenase and cycloox-
nanoparticles. Glutaraldehyde could enhance stability of nanoparticles ygenase/lipoxygenase. Also, lipophilic nature of curcumin allows it to
during three weeks of storage at refrigerated and room temperatures. effectively scavenge peroxyl radicals. Thus, this compound can act as a
BSA in combination with low molecular weight PDL fabricated loaded chain-breaking antioxidant and interrupt propagation chain of oxida-
nanoparticles with a high stability when stored for three weeks tion by eliminating peroxyl radicals from system (Hewlings & Kalman,
(Maldonado, Sadeghi, & Kokini, 2017). In a recent work, Sadati 2017; Marchiani, Rozzo, Fadda, Delogu, & Ruzza, 2014; Menon &
Behbahani, Ghaedi, Abbaspour, Rostamizadeh, and Dashtian (2019) Sudheer, 2007). Nonetheless, antioxidant efficiency of curcumin ex-
made nanostructured lipid carriers (NLCs) containing curcumin via periences a considerable loss in food matrices due to its instability,
microemulsion–ultrasonication technique by Tween 80 and Pluronic reaction with food components, poor solubility, and high sensitivity to
F167 as surfactant with the aid of stearic acid and caprylic/capric tri- alkaline conditions and elevated temperatures. Hence, researchers have
glycerides as lipid phase. Based on results, all samples had a nanometric recently made efforts for nanoencapsulation of curcumin to overcome
size and were semi-spherical in shape. In addition, NLCs showed out- these drawbacks (Aadinath, Bhushani, & Anandharamakrishnan, 2016;
standing capability to boost stability of curcumin within simulated Chen et al., 2015). Some effects of nanoencapsulation on antioxidant
gastric medium up to 2 h (Sadati Behbahani et al., 2019). In the next activity of curcumin have been presented in Table 1.
section, effects of nanoencapsulation on coloring, antioxidant and an- As an example, ctrospinning method was employed to enclose cur-
timicrobial activities of curcumin have been underlined. cumin with the aid of pullulan and amaranth protein isolate and phy-
siochemical characteristics, release behavior and antioxidant activity of
2.1. Coloring activity of nanoencapsulated curcumin curcumin-loaded fibers were investigated. Curcumin concentration and
pullulan/amaranth protein isolate ratio showed an obvious impact on
Many studies have investigated nanoencapsulation of curcumin, but particle size, encapsulation efficiency and morphology of nanofibers.
there were only a few ones regarding its usage as a natural colorant, Curcumin was liberated sustainably under in vitro digestion conditions
preservative, and antioxidant within food matrices. For instance, as well as in a buffer solution (pH = 7.4). Furthermore, loaded cur-
Gomez-Estaca, Balaguer, Gavara, and Hernandez-Munoz (2012) en- cumin retained its antioxidant potential after digestion and exhibited a
capsulated curcumin by electrospraying technique and added it into the better free radical scavenging property in ABST assay than its free form.
semi-skimmed milk. They reported that nanoencapsulation promoted Encapsulation also protected curcumin from degradation when exposed
storage stability and facilitated dispersion of curcumin. Consequently, to high temperatures (up to 600 °C). Thus, usage of food grade biopo-
nanocurcumin exhibited a higher coloring capacity than commercial lymers for incorporation allows the food industry to take advantages of
curcumin in semi-skimmed milk (Gomez-Estaca et al., 2012). In another bioactive compounds more efficiently (Blanco-Padilla, López-Rubio,
study, curcumin nanoemulsions were developed by milk proteins (so- Loarca-PIña, Gómez-Mascaraque, & Mendoza, 2015).
dium caseinate) as emulsifiers and then were used in ice cream for- Protein-polysaccharide complex is a suitable choice for enhancing
mulations (Kumar et al., 2016). Produced curcumin-loaded nanoemul- stability of bioactive compounds (Ghasemi et al., 2017, 2018). In the
sions had good physiochemical characteristics regarding particle size study of Yi et al. (2016), α-lactalbumin-dextran conjugates were formed
(333.8 nm), surface charge (−44.1 mV) and encapsulation efficiency and confirmed their protective effect on curcumin against harsh en-
(96.9%). Moreover, nanoencapsulated curcumin exhibited a consider- vironmental conditions and oxidation. Moreover, water dispersibility
able stability under simulated GIT conditions. Both control and samples and antioxidant activity of curcumin promoted upon

447
Z. Rafiee, et al. Trends in Food Science & Technology 88 (2019) 445–458

Table 1
Some selected studies on antioxidant activity of nanoencapsulated curcumin.
Nanoencapsulation method/Formulation Assay Results Reference

Cellulose acetate nanofibers DPPH Antioxidant effect of curcumin maintained after the Suwantong, Opanasopit,
electrospinning process Ruktanonchai, and Supaphol
(2007)
Nanoprecipitation method DPPH Significant increment of antioxidant potency of Yen, Wu, Tzeng, Lin, and Lin
curcumin (2010)
Chitosan/poly (lactic acid) nanofibers DPPH Antioxidant activity retained after high electric field Dhurai et al. (2013)
Spray-drying warm aqueous ethanol solution ABTS Nanoencapsulation enhanced antioxidant activity of Pan, Zhong, and Baek (2013)
with co-dissolved sodium caseinate curcumin due to the improved dispersibility
Lipid-biopolymer nanoparticles TBARS and radical scavenging Improved antioxidant activity of nanocurcumin Pathak, Kanwal, and Agrawal
(DPPH, NO, H2O2, reducing power) compared to crystalline curcumin in aqueous media (2015)
activity assays
Amaranth protein isolate/pullulan electrospun ABTS - Maintenance of antioxidant potency of Blanco-Padilla et al. (2015)
nanofibers nanocurcumin after an in vitro digestion process
- Higher antioxidant activity of incorporated
curcumin than free counterpart
Starch nanoparticles DPPH and ABTS Stability of curcumin against UV irradiation Li, Shin, Lee, Chen, and Park
improved and its antioxidant ability retained (2016)
Nanocomplexes prepared with both α- DPPH Higher free radical scavenging activity of loaded Yi et al. (2016)
lactalbumin (ALA) and ALA-dextran curcumin compared to free one
conjugates
Nanoemulsion DPPH, ABTS and TABRS Lower Lipid oxidation in milk samples fortified with Joung et al. (2016)
nanocurcumin compared to samples containing free
one
Nanoemulsion DPPH Better performance of incorporated curcumin than Shah et al. (2016)
its free counterpart
Nano spray drying Redispersibility and antioxidant activity of curcumin Chang, Wang, Hu, and Luo (2017)
significantly improved after drying
SLNs ABTS assay and Reducing power Remarkable increase in antioxidant activity of Xue et al. (2018)
curcumin after encapsulation
NLCs ABTS Better maintenance of antioxidant activity of loaded Sadati et al. (2019)
curcumin during storage than free one

DPPH: 2,2-diphenyl-1-picrylhydrazy; ABTS: 2,2 -azino-bis(3- ethylbenzothiazoline-6- sulfonic acid), SLNs: Solid lipid nanoparticles, NLCs: Nanostructured lipid
carriers, TBARS: Thiobarbituric acid reactive substances.

nanoencapsualtion. Dextran in combination with α-lactalbumin re- corn oil for trapping large molecules such as curcumin by formation of
sulted in a higher free radical trapping capacity and stability for cur- several hydrophobic cores than emulsions with MCT. Regarding anti-
cumin than α-lactalbumin alone (Yi et al., 2016). oxidant activity, encapsulated curcumin showed a better performance
In terms of lipid-based nanocarriers, Shah, Zhang, Li, and Li (2016) than its free counterpart, proving the positive impact of emulsion for-
fabricated two various types of lipid carriers including oil-in-water (O/ mulations on antioxidant efficiency of curcumin. Oil type and en-
W) Pickering emulsions and nanoemulsions for encapsulation of cur- capsulation method influenced antioxidant capacity of all samples.
cumin using corn oil along with medium chain triglycerides (MCT) as Radical scavenging activity of nanoemulsions and Pickering emulsions
oil phase. Chitosan-tripolyphosphate nanoparticles were used as aqu- with corn oil increased from 4.06% in control sample to 37.03% and
eous phase in formulation of Pickering emulsions in order to increase 49.58%, respectively. Also, MTC as oil phase improved radical
their stability. A mixture of Span 80 and Tween 80 were also applied as scavenging activity of nanoemulsions and Pickering emulsions from
surfactant for preparation of nanoemulsions. A simulated GIT model 12.21% to 29.80% and 35.18%, respectively (Shah et al., 2016).
composed of different phases (i.e. stomach, intestinal and mouth) was In another work, Joung et al. (2016) prepared curcumin-loaded
applied for determination of digestion pattern of samples. Then, impact nanoemulsions via high pressure homogenization and they were added
of carrier type and their characteristics were assessed on bioaccessi- into milk for evaluation of the lipid oxidation during storage. Anti-
bility by measuring release amount of curcumin after digestion into oxidant activity was determined by various assays including DPPH,
mixed micelles plus radical trapping activity of curcumin. According to ABTS and TABRS (thiobarbituric acid reactive substances). Nanoe-
their results, droplet size and surface charge of all samples evidently mulsions containing curcumin showed a good physical stability during
influenced by digestion. For instance, droplet size of Pickering emul- storage in ambient temperatures. Lipid oxidation was also significantly
sions and nanoemulsions formed by corn oil was reached to 86.4 from lower in milk samples fortified with nanocurcumin compared to sam-
18.8 μm and to 1221 from 109 nm after subjecting to the simulated ples containing free one (Joung et al., 2016).
small intestinal fluids, respectively. Also, ζ-potential of Pickering Xue, Wang, Hu, Zhou, and Luo (2018) successfully formulated solid
emulsions with corn oil changed from 13.17 mV to −2.46, 13.77, and lipid nanoparticles (SLNs) for loading of curcumin by hot emulsification
−27.12 mV in the mouth, stomach and intestine, respectively. On the approach using stearic acid, pectin and sodium caseinate as solid lipid,
other hand, a decline in droplet size caused a significant increase in stabilizing additive and emulsifying agent, respectively. Neither toxic
bioaccessibility as well as lipid digestion. Bioaccessibility of nanoe- solvents nor synthetic surfactants were used in production of SLNs and
mulsions formed by MCT and corn oil were 32% and 65%, respectively antioxidant potential of loaded curcumin was determined by reducing
while 21% and 53% of bioaccessibility were achieved by Pickering power and DPPH assays. Nanoencapsulation boosted antioxidant ac-
emulsions containing MCT and corn oil, respectively. Superiority of tivity of curcumin. On the other hand, curcumin content had a sig-
nanoemulsions to Pickering emulsions concerning bioaccessibility can nificant effect on antioxidant activity and designed food grade SLNs
be attributed to their rapid digestion. Moreover, higher bioaccessibility provided more oxidative stability in higher concentrations of curcumin
of curcumin emulsions obtained from corn oil in comparison with MCT- (Xue et al., 2018).
derived samples was related to higher ability of emulsions containing Recently, Chang et al. (2019) used egg white protein (EWP) to

448
Z. Rafiee, et al. Trends in Food Science & Technology 88 (2019) 445–458

accommodate curcumin. The results confirmed effect of pH and adding et al., 2017).
ethanol on structure of protein and characteristics of nanoparticles. In a recent study, Abdou et al. (2018) prepared nanoemulsions
EWP in both forms of solution and nanoparticles showed a higher containing curcumin through the emulsion inversion point technique
ABTS+ scavenging activity as compared to pure curcumin dissolved in with the aid of different oils (cinnamon essential oil, sunflower oil or
water or ethanol. Free radical scavenging activity diminished during garlic essential oil) and Tween 80 as surfactant. In the next step, high
storage for three days at ambient temperature except for curcumin ester pectin solution was mixed with nanoemulsions to prepare dis-
dissolved in ethanol. However, encapsulation lowered antioxidant ac- persions for covering chicken fillets before packaging and subsequently
tivity loss of curcumin over time. Moreover, antioxidant activity of coated samples were analyzed in terms of microbial, chemical, physical
curcumin was preserved at pH = 3.0 better than pH = 3.8 (Chang and sensorial characteristics during storage for 12 days at 4 °C. Ac-
et al., 2019). cording to their results, size, polydispersity index (PDI) and mor-
phology of nanoemulsion droplets were dependent on oil type. Na-
2.3. Antimicrobial activity of nanoencapsulated curcumin noemulsions could inhibit growth of bacteria and fungi, extending shelf
life of product. Nanoemulsions obtained from cinnamon essential oil
Curcumin shows its antimicrobial activity by interaction with pro- showed the highest capability to control microbial population
tein FtsZ which has a vital role in cell division of microorganisms. The throughout storage period as compared to other oils. At zero time, the
oxygen molecules of phenol, two carbonyl groups and methoxyl func- highest rate of reduction in total bacterial count was found in samples
tional groups linked to phenolic rings of curcumin form hydrogen and treated with curcumin-cinnamon essential oil nanoemulsion/pectin
hydrophobic bonds with the catalytic site of this protein which disturbs coating (CCNC) (80.3%) followed by those with curcumin-garlic oil
GTPase activity of FtsZ, leading to cell death (Kaur, Modi, Panda, & nanoemulsion/pectin coating (CGNC) and curcumin-sunflower oil na-
Roy, 2010; Rai, Singh, Roy, & Panda, 2008). Nevertheless, different noemulsion/pectin coating (CSNC) which showed 47.4% and 24.7%
factors such as food components and processing and storage conditions reduction, respectively in comparison with control. The same trend was
could negatively influence the antimicrobial potency of curcumin in observed after 12 days of storage and the decline was reached to 97.8%,
food systems. Furthermore, direct usage of curcumin in food systems 93.6% and 40.8% in samples with CCNC, CGNC and CSNC, respec-
may decrease consumer satisfaction. For instance, application of cur- tively. Moreover, CCNC, CGNC and CSNC caused 95.1%, 72.9% and
cumin as a natural preservative in minced meat showed that curcumin 42.6% decrease in psychrophilic bacterial count of fillets compared to
has an strong antimicrobial efficacy against some food pathogens (Es- control sample, respectively. Regarding yeast and mold counts, the
cherichia coli O157:H7, Listeria monocytogenes, Salmonella typhimurium, lowest growth rates were related to samples treated with CCNC
and staphylococcus aureus) but samples treated with higher concentra- (0.56 d−1) and CGNC (0.49 d−1), which was attributed to inhibitory
tions of curcumin were not acceptable in terms of sensory attributes effect of cinnamaldehyde and allicin as main bioactive compounds of
especially color (Altunatmaz et al., 2016). Nanoencapsulation of phe- cinnamon essential oil and garlic essential oil against different micro-
nolic compounds like curcumin in suitable carriers presents great po- organisms. Furthermore, fillets treated with nanoemulsions bearing
tential to promote antimicrobial properties of phenolic compounds cinnamon essential oil or garlic essential oil exhibited a higher water
through overcoming drawbacks associated with their direct applica- holding capacity, oxidative stability and texture score as well as less
tions such as poor water solubility, low stability and unfavorable in- total volatile nitrogen content than control sample and those coated
teraction with food ingredients (Akhavan, Assadpour, Katouzian, & with sunflower oil-derived nanoemulsion. Control samples also re-
Jafari, 2018; Rezaei, Fathi, & Jafari, 2019). Moreover, encapsulated ceived the least degree of acceptability in sensory features by panelists
form of curcumin can effectively interact with microbial cells and fa- (Abdou et al., 2018).
cilitate cell perturbation, increasing inhibitory effect against micro- Some researchers have encapsulated turmeric extract in different
organisms (Pagnussatt et al., 2016, Da Silva et al., 2018). nanocarriers and evaluated their antimicrobial activities. For instance,
In this regard, Wang et al. (2017a) employed electrospinning antioxidant and antimicrobial activities of free and loaded turmeric
method to enclose curcumin in zein fibers and then evaluated their methanolic extracts were determined by Abdel Fattah and Tawfik
antioxidant and antimicrobial capabilities against E.coli and S.aureus. (2016) and Alnashiand and Abdel Fattah (2016), respectively. They
Encapsulated curcumin had a nanometric size and a high entrapment found that nanoencapsulation improved antioxidant and antimicrobial
efficiency. Concentration of curcumin influenced the particle size, activity of turmeric extract against tested microorganisms (L.mono-
shape and transition temperature of fibers. Antioxidant capacity of cytogenes, E.coli O157:H7, S.aureus, S.typhimurium, B.cereus, Candida
curcumin was preserved upon encapsulation. Besides, fibers bearing albicans, Geotricum candidum, Fusarium moniliform, Aspergillus flavus,
curcumin showed inhibitory action against bacterial strains tested, but and Aspergillus. niger) (Abdel Fattah, 2016; Alnashi, 2016). The effect of
they were more effective on S.aureus. These authors suggested cur- nanocurcumin on pathogenic microorganisms has been reviewed in
cumin-loaded zein fibers as an encouraging alternative to retard mi- section 3.4.
crobial spoilage and prolong shelf life of food products (Wang et al.,
2017a). 3. Nanoencapsulated curcumin for medicinal purposes
Deng, Kang, Liu, Feng, and Zhang (2017) encapsulated curcumin via
electrospinning technique by means of three various surfactants (i.e. Curcumin is historically valuable in medical applications and has
cetyltrimethyl ammonium bromide (CTAB), anionic sodium dodecyl been used as a traditional therapy in various Asian medicines (Sharma,
sulfonate (SDS) and Tween 80) and gelatin as polymer. They revealed Gescher, & Steward, 2005). Due to its polyphenolic structure and highly
correlation between surfactant type and bioactivity, release pattern and safe nature, it has been known as a potential therapeutic molecule with
morphology of nanocurcumin formulations. SDS caused an evident in- versatile pharmacological activities (Noorafshan & Ashkani-Esfahani,
crease in diameter of nanofibers and also had a strong interaction with 2013; Yallapu, Nagesh, Jaggi, & Chauhan, 2015). Its therapeutic ap-
gelatin by formation of hydrophobic and ionic bonds which decreased plications are highly variant including anti-oxidant, anti-neoplasm,
release rate of curcumin in contrast to other surfactants. CTAB made anti-bacterial, anti-diabetic and anti-inflammatory properties; while it
nanofibers with a higher antioxidant activity (determined by DPPH and is proven to have positive effects in the treatment of cardiovascular and
reducing power assays) than SDS and Tween 80. Moreover, regardless Alzheimer's disease, liver cirrhosis, GIT problems and wound healing
of surfactant type, nanofibers bearing curcumin were effective on (Gera et al., 2017; Gupta, Patchva, & Aggarwal, 2013; Noorafshan &
S.aureus, but only those with CTAB could control growth of E.coli. Ashkani-Esfahani, 2013; Yallapu et al., 2015).
Therefore, entrapping bioactive materials in nanofibers by food grade However, curcumin poses some critical drawbacks which impede its
surfactants can be helpful for developing novel functional foods (Deng efficacy as a therapeutic drug molecule. Its highly lipophilic structure

449
Z. Rafiee, et al. Trends in Food Science & Technology 88 (2019) 445–458

results in very low aqueous solubility and intrinsic dissolution rate. bioavailability compared to free curcumin suspensions and suggested
Additionally, it shows instability at alkaline pH values and undergoes this nanoformulations as effective delivery systems for oral applications
rapid metabolism and elimination in vivo. Hence, it has very poor of curcumin (Baek & Cho, 2017).
pharmacokinetic properties including poor absorption and low oral Some other studies have proven the anti-cancer efficacy of curcumin
bioavailability along with low penetration and targeting efficacy (Gera nanoformulations in animal models. For example, Gong et al. (2013a)
et al., 2017; Naksuriya, Okonogi, Schiffelers, & Hennink, 2014; Yallapu evaluated curcumin-loaded polymeric micelles for pulmonary carci-
et al., 2015). Encapsulation of curcumin in different nanocarriers has noma. The results showed a sustained in vitro release profile and in-
been proposed as an emerging solution for these limitations (Jafari & creased in vitro anti-angiogenesis, cellular uptake and cytotoxicity
Mcclements, 2017). As mentioned in previous sections, nanocarriers compared to free curcumin. The micelles also showed a superior anti-
can help to increase the aqueous solubility of curcumin while protecting tumor efficacy in subcutaneous and pulmonary metastatic LL/2 tumor
it from degrading environment. These formulations can modify the models compared to free form (Gong et al., 2013a). Lin et al. (2012)
pharmacokinetic properties of the loaded drug and may increase ther- used 270 nm-sized cationic liposome-PEG-PEI complexes for en-
apeutic efficiency at the site of action with passive and active targeting capsulation of curcumin. This nanoformulation caused a 5-fold and 20-
mechanisms, and also higher cellular uptake rates (Gera et al., 2017; fold increase in cytotoxicity against curcumin-sensitive and curcumin-
Yallapu et al., 2012, 2013). Different curcumin-loaded nanocarriers resistant cells, respectively. The cytotoxicity was attributed to induced
have been designed and studied for various disease conditions and some apoptosis by cell cycle arrest at G2/M phase and rapid accumulation in
formulations have reached the clinical trials (Naksuriya et al., 2014). the cell. These nanocarriers also inhibited the tumor growth up to
Some important and current pharmaceutical applications of these na- 60–90% in mice bearing CT-26 or B16F10 cells (Lin et al., 2012).
noformulations are summarized in the following sections. Yallapu et al. (2014) evaluated the therapeutic potential of curcumin-
loaded PLGA nanoparticles in prostate cancer. They showed that these
3.1. Nanoencapsulated curcumin against cancer nanoparticles can enter the prostate cancer cells and curcumin is re-
leased in cytosolic compartment. PLGA nanoparticles showed superior
Curcumin exhibits anti-cancer effects against numerous kinds of in vivo anti-tumor effects compared to free curcumin in xenograft mice
malignancies including head and neck squamous cell carcinoma, mul- with additional antineoplastic effects in prostate cancer (Yallapu et al.,
tiple myeloma, and many other malignancies like colorectal, pan- 2014).
creatic, breast, prostate, lung, and oral cancer. It can suppress different In a comprehensive study conducted by Zaman et al. (2016), the
cellular processes including cell transformation or proliferation, inva- efficacy of PLGA-based curcumin nanoparticles in cervical cancer was
sion and metastasis with a combination of complicated mechanisms. assessed in vitro and in vivo. The nanoformulations improved accumu-
The main anti-cancer mechanisms involve modulation of pro-in- lation of curcumin in cervical cancer cells and caused cell arrest in G1-S
flammatory cytokines (like tumor necrosis factor [TNF]-α and inter- transition phase, inducing apoptosis. The designed nanoparticles sig-
leukin IL-6 and [IL]-1β), modulation of numerous apoptotic proteins nificantly reduced tumor growth in cervical cancer orthotopic xenograft
including NF–κB, cyclooxygenase (COX)-2, IKKβ, STAT3, and en- mouse model while the level of oncogenic HPV E6/E7 and Ki67 pro-
dothelin-1 (Vallianou, Evangelopoulos, Schizas, & Kazazis, 2015; teins were also decreased. Additionally, the formulation decreased the
Yallapu et al., 2012). levels of miRNA-21, in vitro and in vivo, in comparison to free curcumin
Although curcumin is potentially effective against different cancers, while decreasing the levels of IL-6 (Zaman et al., 2016).
but poor pharmacokinetic properties limit its clinical efficacy. The anti-cancer efficacy of milk-derived exosomes containing cur-
Utilization of different nanoformulations is a promising approaches in cumin was investigated by Aqil, Munagala, Jeyabalan, Agrawal, and
this regard. A wide range of in vitro and in vivo studies obviously reveal Gupta (2017). The exosomes were taken up by cancer cells via ca-
the therapeutic efficacy of curcumin-loaded nanocarriers including veolae/clathrin-mediated endocytosis and showed 3–5 times higher
polymeric nanoparticles, liposomes, SLNs, micelles, etc. in diverse tissue accumulations compared to free curcumin. Also, the curcumin-
cancer types (Naksuriya et al., 2014; Yallapu et al., 2012, 2013). The loaded exosomes showed a higher anti-proliferative activity against
results of these studies show that nanocarriers can enhance the anti- multiple cancer cell lines including cervical, breast, and lung cancer cell
cancer efficacy of curcumin by modulation of pharmacokinetic prop- lines. This formulation showed a significant inhibition of the cervical
erties (higher bioavailability and longer biological half-life) and/or CaSki tumor xenograft in nude mice while no systemic toxicity was
increasing site-specific drug delivery. This can be achieved by pro- observed in the rats (Aqil et al., 2017).
tecting curcumin from degradation, increasing its solubility, controlling Novel formulations of curcumin-loaded nanoparticles (like
the drug release, promoting membrane transport and/or absorption, Theracurcumin®, a brand name for curcumin-loaded nanoparticles)
enhancing cellular uptake, and bypassing of cellular efflux pumps have shown improved pharmacokinetic properties in recent clinical
(Naksuriya et al., 2014; Yallapu et al., 2012, 2013). trials in human volunteers, but still a high-throughput research activity
A vast majority of studies on the literature have been focused on to develop more efficient curcumin-loaded nanocarriers for cancer
preparation, characterization and cellular uptake and cytotoxicity of therapy is needed (Naksuriya et al., 2014). Some studies on application
nanoencapsulated curcumin in different types of cancer cell lines which of curcumin nanoformulations in cancer therapy are illustrated in
have been thoroughly reviewed in previously published review articles Table 2.
(Gera et al., 2017; Naksuriya et al., 2014; Yallapu et al., 2012, 2013,
2015). In some studies, the pharmacokinetics of formulations have also 3.2. Nanoencapsulated curcumin against cardiovascular diseases
been assessed and compared to solutions (Gera et al., 2017; Mohanty,
Das, & Sahoo, 2012; Naksuriya et al., 2014; Yallapu et al., 2012, 2013). Curcumin is also effective in mitigation and treatment of cardio-
For instance, Khalil et al. (2013) suggested PLGA and PLGA-PEG na- vascular diseases and vascular dysfunctions. The cardioprotective me-
noparticles as potential nanocarriers for oral delivery of curcumin. chanisms of curcumin are various and mostly related to its anti-in-
These nanoparticles provided a sustained drug delivery and increased flammatory effects. For instance, curcumin can reduce NO oxidation
the bioavailability of curcumin up to 15.6- and 55.4-fold compared to and shows membrane stabilizing effects in MI, prevents doxorubicin
its suspensions, respectively (Khalil et al., 2013). induced cardiomyopathy, and diabetic complications. Curcumin can
Baek and Cho (2017) in a recent study prepared stable N-carbox- also inhibit p300 and NF-κB which are associated with prevention of
ymethyl chitosan-coated SLNs loaded with curcumin which showed a cardiac hypertrophy and cardiac fibrosos, respectively (Wongcharoen &
higher cellular uptake and cytotoxicity on MCF-7 cells. They also re- Phrommintikul, 2009).
ported a 6.3-fold higher lymphatic uptake and 9.5-fold higher oral Sunagawa et al. (2012) prepared a new curcumin delivery system

450
Z. Rafiee, et al. Trends in Food Science & Technology 88 (2019) 445–458

consisting colloidal nanoparticles dissolved in gum ghatti solution. This

Aqil et al. (2017)


Lin et al. (2012)
formulation showed a 27-fold increase in oral bioavailability of cur-

Yallapu et al.

Zaman et al.
Gong et al. cumin and significantly improved perivascular fibrosis in post-MI rats
Reference

(2013a) compared to free curcumin (Sunagawa et al., 2012). Carlson, Cote,

(2014)

(2016)
Alani, and Rao (2014) designed resveratrol and curcumin-loaded
Pluronic-F127 micelles for prevention of doxorubicin-induced cardio-
angiogenesis; anti-tumor efficacy in HUVECs, transgenic zebrafish models, and

proteins in cervical cancer xenograft mouse model; Decreased levels of miRNA-


cancer cells; Superior tumor regression compared to free curcumin in xenograft
against curcumin-resistant cells compared to curcumin; Inhibition of 60–90% of

Improved accumulation in cervical cancer cells and induction of apoptosis via


5-fold higher cytotoxicity on curcumin-sensitive cells and 20-fold more active

free curcumin; Significant inhibition of the cervical CaSki tumor xenograft in


toxicity. The results revealed that this formulation could enhance so-

proliferative activity and 3–5 times higher tissue accumulations compared to


Effective inhibition of proliferation and colony formation ability of prostate

cell arrest in G1-S transition phase; Reduced tumor growth and oncogenic
lubility of these compounds and can mitigate cardiomyopathy by re-

Cellular uptake via caveolae/clathrin-mediated endocytosis; Higher anti


duction of apoptosis and ROS level, while improving doxorubicin
Increased cellular uptake, and cytotoxicity; in vitro and in vivo anti-

efficacy in ovarian cancer cells (Carlson et al., 2014). Soltani,


Bodaghabadi, Mahpour, Ghaemi, and Sadeghizadeh (2016) in-
subcutaneous and pulmonary metastatic LL/2 tumor models

21 and IL-6 in vitro and in vivo, compared to free curcumin.


vestigated the potential of a dendrosomal formulation of curcumin in
tumor growth in mice bearing CT-26 or B16F10 tumors.

prevention of radiation induced atherosclerosis. The formulation sig-


nificantly inhibited the Co60 gamma rays-induced changes in human
umbilical vein endothelial cells by activation of the Nrf-2 pathway

nude mice with no systemic toxicity in the rats.


(Soltani et al., 2016).

3.3. Nanoencapsulated curcumin against neurodegenerative diseases

Curcumin has shown therapeutic potentials in improvement of


neurodegenerative diseases. These effects are related to its anti-oxidant,
anti-inflammatory and anti-protein aggregation activities. The accu-
mulation of toxic β-amyloid peptide (Aβ) in senile plaques is typically
Study outcome

seen in Alzheimer's disease (AD). Curcumin can target the amyloid


pathway and has received great attention in the treatment of AD
mice.

(Darvesh et al., 2012).


Doggui, Sahni, Arseneault, Dao, and Ramassamy (2012) studied
- Cytotoxicity, apoptosis induction, and cellular uptake on LL/

- In vivo studies including systemic toxicity, tumor uptake and

cell cycle analysis in Caski and SiHa cervical cancer cell lines.
- Cellular uptake, cell proliferation, colony forming assay and

effects of curcumin-loaded PLGA nanoparticles on human neuro-


- In vivo suppression of tumor growth in mice bearing tumors

- Cell proliferation assay in lung (H1299 and A549), cervical


- In vitro antiangiogenic activity in primary human umbilical

- Cell proliferation, clonogenic, and Western blot analyses in


- In vivo anti-tumor efficacy in subcutaneous and pulmonary

xenograft mouse model, In vivo assessment of miRNA-21in


- Cytotoxic activity on 10 different curcumin-sensitive and

human prostate cancer cell lines (DU-145 and PC-3 cells)

(HeLa) and breast cancer (MDA-MB-231 and T47D) cells

blastoma SK-N-SH cells. The nanoparticles were able to protect cells


- Tumor growth in NOD SCID gamma (NSG) orthotopic

against H2O2 and prevented H2O2-induced ROS formation and glu-


tumor targeting capability in C4-2 xenograft mice.

tathione consumption (Doggui et al., 2012). Cheng et al. (2013) de-


- In vivo drug extravasation and anti-angiogenesis

- Tissue distribution in the lung, liver, and brain

signed polyethyleneglycol-polylactide (PEG-PLA) di-block polymeric


micelles to increase bioavailability of curcumin and enhance its pene-
tration into the Blood Brain Barrier (BBB). After oral administration in
- Cellular uptake studies in H1299 cells

Tg2576 AD model mice, a significant improvement was seen over


- In vivo Tumor Xenograft Studies
vein endothelial cells (HUVECs)

metastatic LL/2 tumor models.

curcumin resistant cancer cells

control in working and cue memory. The nanoparticles significantly


increased curcumin plasma concentrations and bioavailability with a 6-
Examples of comprehensive studies on curcumin nanoformulations for treatment of cancer.

fold higher AUC in brain (Cheng et al., 2013). Mourtas et al. (2011)
proposed a new formulation of curcumin-loaded nanoliposomes which
had the proper planar structure required for interaction with Aβ and
they reported a high affinity of these nanoparticles for Aβ1-42 fibrils
mice tissues.

(Mourtas et al., 2011). In another study conducted by this group in


Model used

2 cells

2014, anti-Transferrin-conjugated PEGylated nanoliposomes were de-


signed and characterized. They reported that both ligand-targeted li-
posomes and simple liposomes showed a high affinity for the Aβ de-
posits on samples obtained from patients with AD and could retard Aβ1-
Monomethyl poly (ethylene glycol)-poly

42 peptide aggregation in Thioflavin assay. However, active-targeting


(ε-caprolactone) (MPEG-PCL) micelles

Cationic liposome-PEG-PEI complexes

with anti-Transferrin significantly improved the uptake in BBB cellular


model (Mourtas, Lazar, Markoutsa, Duyckaerts, & Antimisiaris, 2014).
Another attempt for active targeting of curcumin-loaded nanolipo-
somes to BBB was conducted by Mathew et al. (2012). They suggested
Milk-derived exosomes

PLGA nanoparticles to increase the solubility of curcumin, and deco-


PLGA nanoparticles

PLGA nanoparticles

rated the surface of nanoparticles with Tet-1 peptide to enhance BBB


Characteristics

uptake. The designed nanoparticles showed a high in vitro cellular up-


take into GI-1 glioma cells and were able to destroy amyloid aggregates,
and exhibited anti-oxidant properties showing no toxicity (Mathew
et al., 2012). Sandhir et al. (2014) proposed curcumin-loaded SLNs for
Huntington's disease. The cell culture and in vivo studies showed that
Polymeric nanoparticles

Polymeric nanoparticles

SLNs can cause a significant improvement in neuromotor coordination


Type of nanocarrier

Polymeric micelles

of Huntington's disease-induced rats. Hence, they suggested these cur-


cumin-loaded nanoparticles as a promising formulation to ameliorate
mitochondrial dysfunctions in Huntington's disease (Sandhir et al.,
Exosomes
Liposome

2014).
Table 2

451
Z. Rafiee, et al.

Table 3
Studies considering the antimicrobial properties of curcumin-loaded nanocarriers.
Type of nanocarrier Characteristics Study design Study outcome Reference

Curcumin nanoparticles Prepared by a wet-milling technique MIC was examined against Staphylococcus aureus, Bacillus subtilis, The activity of nanoformulations was more pronounced against Bhawana et al.
E.coli, Pseudomonas aeruginosa, Penicillium notatum and Aspergillus Gram-positive than Gram-negative bacteria; Better anti-bacterial (2011)
niger activity than anti-fungal activity.
Curcumin nanoparticles Prepared by supercritical CO2 The anti-bacterial activity (MIC) was examined against gram The MIC of nanoparticles was %50 lower than free curcumin Xie et al. (2015)
positive (S. aureus) and gram negative (E. coli) bacteria solution.
Curcumin nanoparticles Prepared by precipitation of curcumin from Anti-bacterial activity against E. coli, S. aureus, and Pseudomonas The nanoparticles exhibited in vitro anti-bacterial activity against Pandit et al.
organic solution in boiling water aeruginosa using Kierby–Bauer disc diffusion method all tested bacteria. (2015)
Curcumin-loaded chitosan Prepared by polymerization of chitosan in the Colony forming unit (CFU) quantification was performed on CFU quantification showed that curc-np exerted a 97.0% reduction Krausz et al.
nanoparticles presence of PEG 400 and hydrolyzed MRSA and P. aeruginosa isolates to determine antimicrobial of MRSA growth (Fig. 2C) and 59.2% reduction of P. aeruginosa (2015)
Tetramethyl orthosilicate (TMOS- HCl) activity/Antimicrobial activity was also evaluated in vivo by growth/Curc-np reduce bacterial burden in MRSA-infected burn
homogenizing infected burn wounds and quantifying CFUs wounds.
present in tissue on day 3 and 7
Curcumin-loaded polymeric Prepared from chitosan or PLGA Rats were inoculated with Giardia lamblia cysts then treated with The parasite was successfully eradicated from stool and intestine. Said et al. (2012)

452
nanoparticles curcumin formulations and the number of Giardia cysts in stools
and trophozoites in intestinal sections were detected.
Nanostructured lipid carriers Prepared by nanoemulsion technique employing In vivo antimalarial activity was assessed in malaria induced 2-fold increase in anti-malaria activity of nanoparticles was seen Nayak et al.
high-speed homogenizer and ultrasonic probe Albino mice after injection of formulation for 3 consecutive days. compared to curcumin in in vivo model. (2010)
Liposome PEGylated liposomes containing Artemisinin, Artemisinin-based liposomal formulations was tested in Artemisinin and curcumin-loaded PEGylated liposomes showed an Isacchi et al.
alone or in combination with curcumin Plasmodium berghei NK-65 infected mice immediate antimalarial effect. (2012)
Chitosan nanoparticles Curcumin was bound to the surface of chitosan In vitro and in vivo stability was assessed in mouse plasma, oral Reduced curcumin degradation in mouse plasma in vitro, increased Akhtar et al.
nanoparticles bioavailability and pharmacokinetic properties were evaluated in oral bioavailability and survival of mice infected with a lethal (2012)
mice and in vivo antimalarial activity was tested on Plasmodium strain of Plasmodium yoelii (N-67).
yoelii infected mice.
Curcumin nanoparticles Produced by high-frequency ultrasonication Antimicrobial activity of nanocurcumin, microcurcumin and Fivefold increase in antimicrobial activity by nanoencapsultion of Gopal, Muthu, and
macro-turmeric were evaluated aginst E.coli, Salmonella curcumin; Higher solubility, penetration and antimicrobial activity Chun (2016)
enteritidis, Streptococcus mutans and S. aures of nanocurcumin compared to macro-turmeric and
microcurcumin; The most susceptible bacteria: E.coli and S.
enteritids
Curcumin nanoparticles Prepared by a solvent and antisolvent Using nanocurumin along with silver nanoparticles as an anti- Inhibition of biofilm formation with the combination of silver and Loo et al. (2015)
precipitation technique biofilm agent against Pseudomonas aeruginosa and S. aureus curcumin nanoparticles
Significant effect of nanocurcumin content on anti-biofilm activity
Trends in Food Science & Technology 88 (2019) 445–458
Z. Rafiee, et al. Trends in Food Science & Technology 88 (2019) 445–458

3.4. Anti-pathogenic effects of nanoencapsulated curcumin 3.5. Anti-viral effects of nanoencapsulated curcumin

Curcumin has been known as a traditional anti-microbial agent and It has been thoroughly reported that curcumin poses inhibitory ef-
a variety of studies have proved its efficacy against numerous Gram- fects against viruses interfering some critical steps of virus replication
negative and Gram-positive bacteria. However, efforts have been made cycle. Curcumin has been studied as an anti-viral agent against nu-
to increase its anti-microbial efficacy by incorporating into different merous viruses like Herpes simplex virus (HSV), Human
nanoparticles. A general review in this regard has been published by Immunodeficiency Virus (HIV), Hepatitis viruses, influenza type A
(Silva et al., 2018). Table 3 summarized some of the recent studies on virus, and Ebola virus. This natural compound can directly interfere
anti-microbial effects of nanoencapsulated curcumin. with the viral replication machinery or suppress the essential cellular
Bhawana et al. (2011) prepared curcumin nanoparticles with a wet- signaling pathways in viral replication cycle such as inflammation, and
milling technique. The solubility of curcumin was increased in this apoptosis and including PI3K/Akt, NF-κB s (Mathew & Hsu, 2018).
nanoformulation. The minimum inhibitory concentration (MIC) of In a study conducted by Gandapu, Chaitanya, Kishore, Reddy, and
curcumin was examined against Aspergillus niger, Bacillus subtilis, E.coli, Kondapi (2011), the potentials of curcumin-loaded apotransferrin na-
S.aureus, Pseudomonas aeruginosa, and Penicillium notatum. The results noparticles for inhibition of HIV-1 replication was studied in vitro.
demonstrated that this nanoformulation of curcumin was more active Nanoparticles showed a sustained delivery of curcumin and decreased
against Gram-positive than Gram-negative bacteria. Furthermore, it curcumin cytotoxicity up to 50% compared with free curcumin. The
exhibited better anti-bacterial activity than anti-fungal activity. The nanoformulation of curcumin showed about 3-fold higher anti-HIV
transmission electron microscopy (TEM) analysis revealed that these activity compared to its free form and inhibited the HIV-1 induced
particles can cause cell death by entering the bacterial cell through expression of IL-1β,Topo II α, and COX-2 and completely blocked the
complete breaking of the cell wall (Bhawana et al., 2011). Xie et al. synthesis of viral cDNA (Gandapu et al., 2011). Yang, Li, and Huang
(2015) prepared curcumin nanoparticles via supercritical CO2. They (2016) introduced curcumin modified silver nanoparticles for sy-
reported a higher solubility and increased anti-bacterial, antioxidant nergistic anti-viral effects of curcumin and AgNPs in respiratory syn-
and anti-cancer efficacy for this curcumin-loaded nanoparticles. The cytial virus (RSV) infection. These nanoparticles showed a high in-
results also revealed a better anti-microbial efficacy of nanoparticles hibitory effect against RSV infection in tissue culture infectious dose
and the MIC of nanoparticles was 50% lower than free curcumin so- assay and 2-fold lower viral titers were resulted. The nanoformulations
lution (Xie et al., 2015). In another study conducted by Pandit, could inactivate the RSV and prevent the host cells from infecting (Yang
Gaikwad, Agarkar, Gade, and Rai (2015), curcumin nanoparticles were et al., 2016).
prepared by precipitation of the bioactive from organic solution in
boiling water and their anti-bacterial activity against some pathogenic 3.6. Wound healing activities of nanoencapsulated curcumin
bacteria was evaluated. They reported that curcumin nanoformulations
exhibit in vitro anti-bacterial activity against E.coli, S.aureus, and Curcumin has shown notable wound healing properties. It can
P.aeruginosa (Pandit et al., 2015). Krausz et al. (2015) synthesized promote the wound healing process through different mechanisms in-
curcumin-loaded chitosan nanoparticles and reported in vitro growth cluding involvement in the tissue remodeling process, granulation or
inhibition of methicillin-resistant S.aureus (MRSA) and P.aeruginosa in a tissue formation, and collagen deposition. It can also interfere with
dose-dependent manner. The synthesized nanoparticles could inhibit epithelial regeneration and promote fibroblast proliferation and vas-
planktonic growth of both microorganisms and induced cellular da- cular density (Akbik, Ghadiri, Chrzanowski, & Rohanizadeh, 2014).
mage of MRSA. The nanoparticles could also decrease bacterial burden A nanoformulation of curcumin was developed by Li et al. (2012)
of full-thickness burns in vivo (Krausz et al., 2015). with methoxy poly (ethylene glycol)-b-poly(ɛ-caprolactone) copolymer
Some studies have shown anti-parasite activity of curcumin nano- (MPEG-PCL) and then it was incorporated into the N,O-carboxymethyl
formulations. Said, Elsamad, and Gohar (2012) reported the positive chitosan/oxidized alginate hydrogel. This nanoformulation showed an
role of curcumin-loaded nanoparticles (including chitosan and PLGA improved stability and slow release. In vivo wound healing experiments
polymeric nanoparticles) in treatment of the intestinal giardiasis, and revealed improved re-epithelialization of epidermis and collagen de-
regarded it as a curable and safe treatment (Said et al., 2012). Nayak, position in the injured tissue (Li et al., 2012). Gong et al. (2013b)
Tiyaboonchai, Patankar, Madhusudhan, and Souto (2010) showed a 2- prepared curcumin-loaded polymeric micelles in a thermosensitive
fold increase in anti-malaria activity of lipid-based curcuminoid nano- hydrogel formulation for wound healing. This formulation exhibited
particles over free curcumin after parenteral administration in an in vivo proper tissue-adhesion and provided extended release of curcumin. The
model (Nayak et al., 2010). Furthermore, Isacchi et al. (2012) designed in vivo wound healing effects of formulation was examined on linear
PEGylated liposomes containing Artemisinin and curcumin, which incision and full-thickness excision wound models. The results in-
provided a highly significant therapeutic benefit in vivo in mice infected dicated that curcumin-loaded polymeric micelles could enhance cuta-
with Plasmodium berghei NK-65 (Isacchi et al., 2012). Chitosan-bound neous wound repair and caused higher collagen content, better gran-
curcumin nanoformulations were also evaluated for their anti-malaria ulation and wound maturity, a highly significant decrease in superoxide
effects by Akhtar, Rizvi, and Kar (2012). The designed nanoparticles dismutase, and slight increase in catalase (Gong et al., 2013b).
showed an increased stability in mouse plasma in vitro and better up- Krausz et al. (2015) prepared curcumin polymeric nanoparticles
take by mouse RBC compared to free curcumin. Curcumin-loaded from chitosan and PEG 400 as an anti-microbial and wound healing
chitosan nanoparticles improved the oral bioavailability of curcumin agent. This formulation accelerated the wound healing process in a
and increased the survival of mice infected with a lethal strain of murine burn model and could enhance collagen deposition, formation
Plasmodium yoelii (N-67) (Akhtar et al., 2012). of granulation tissue, and neoangiogenesis (Krausz et al., 2015).
In a recent work, Karimi, Ghanbarzadeh, Hamishehkar, Mehramuz, As mentioned above, curcumin nanoformulations are extensively
and Kafil (2018) incorporated turmeric extract into NLCs by high shear under study and some formulations have reached the clinical trial.
homogenization. Nanoparticles (112.4 nm) presented a sustained re- Table 4 summarizes some clinical trials conducted on curcumin nano-
lease pattern as well as a high physical stability. A significant increase formulations.
was observed in antioxidant and antimicrobial activity of curcumin
against Gram-negative bacteria (E.coli, Acinetobacter juni and P.aerugi- 4. Application of nanoencapsulated curcumin in the cosmetic
nosa) after nanoencapsulation (Karimi et al., 2018). industry

Curcumin has been considered in cosmetic applications due to its

453
Table 4
Clinical trials conducted on curcumin nanoformulations.
Formulation type Study title Disease/condition Aims ClinicalTrials.gov
Z. Rafiee, et al.

Identifier

Surface-controlled water soluble Phase I study of surface-controlled water soluble curcumin Advanced cancer Evaluation of the highest tolerable dose and safety of formulation NCT01201694
curcumin (THERACURMIN CR-011L)
Liposomal curcumin Safety, tolerability and pharmacokinetics of liposomal curcumin Drug safety Evaluation of the safety, tolerability and pharmacokinetics of NCT01403545
in healthy volunteers - a phase I dose escalation study increasing doses of intravenous liposomal curcumin in healthy
subjects
A phase IB dose escalation study on the safety, tolerability and Patients with advanced cancer Determination of the safety profile and maximum tolerated dose NCT02138955
activity of liposomal curcumin in patients with locally advanced who have failed standard of (MTD) of increasing doses of intravenous liposomal curcumin in
or metastatic cancer care therapy cancer patients
Nanocurcumin capsules (containing Effects of oral nanocurcumin on expression levels of micrornas Ankylosing spondylitis Number of subjects with a reduction in signs and symptoms of NCT03140657
curcumin nanoparticles, and treg cells and th17 cells development factors in ankylosing Ankylosing spondylitis 4 month after treatment
Exirnanosina) spondylitis patients
Nanomicelle curcumin The effects of nanomicelles curcumin on glycemic control, serum Metabolic syndrome Evaluation of blood pressure and some serum biomarkers related NCT03534024
lipid profile, blood pressure and anthropometric measurements to metabolic syndrome including blood glucose level and serum
in patients with metabolic syndrome lipid profile (HDL, LDL, Cholesterol and TG)
Effects of nanocurcumin on serum oxidative stress, Metabolic syndrome Evaluation of some serum biomarkers like total antioxidant NCT03514667
inflammation, adiponectin and NF-kB in blood mononuclear capacity (TAC), serum malondialdehyde (MDA), and serum C-
cells in metabolic syndrome patients (nuclear factor-κB) reactive protein (CRP)
Curcumin conjugated with plant Phase I clinical trial investigating the ability of plant exosomes Colon cancer Comparison of the effect and the concentration of curcumin in NCT01294072
exosomes to deliver curcumin to normal and malignant colon tissue normal and cancerous tissue after administration of the curcumin
formulations; Safety, immune responses and histochemical
staining are also evaluated as secondary outcomes

454
Table 5
Studies considering cosmetic applications of curcumin nanoformulations.
Type of nanocarrier Characteristics Study design Study outcome Reference

Lipid-core nanocapsules Curcumin and resveratrol encapsulating LNCs were In vitro antioxidant activity and photostability study was In vitro antioxidant activity of polyphenols against OH Coradini et al.
(LNC) prepared by precipitation of preformed polymer method conducted. radicals was enhanced by nanoencapsulation, and a better (2014)
using PCL, grape seed oil, sorbitan monoestearate effect was observed after their co-nanoencapsulation.
Lipid vehicles Curcumin was incorporated into lipid vehicle consisting Propionibacterium acnes growth inhibition of the Curcumin-loaded lipid vesicles could efficiently accumulate Liu and Huang
of lauric acid, isopropanol, and surfactants (Tween 80 and formulation was evaluated in vitro. The mechanism of in the neonate pig skin and inhibit propionibacterium acnes (2013)
F127) in order to inhibit the growth of propionibacterium curcumin accumulation in porcine skin was evaluated
acnes as an anti-bacterial and anti-infammatory agent. using neonate pig skin.
Vesicular carriers Curcumin-loaded liposomes, ethosomes, and Effects of vesicular carriers on ultraviolet radiation- Curcumin-loaded vesicles showed good skin penetration and Kaur and Saraf
transfersomes were prepared as cream. damaged skin were assessed through examination of the photoprotective effects with the following order: curcumin- (2011)
skin hydration level and sebum content. loaded transfersomal creams > ethosomal
creams > liposomal creams > curcumin creams > empty
transfersome cream > empty ethosome cream > empty
liposome cream > base cream
Cyclodextrin-based Cyclodextrin/curcumin complex Cyclodextrin/curcumin complex or curcumin solution Solubilized curcumin in an aqueous cyclodextrin vehicle Konradsdottir et al.
nanoformulations were placed on top of excised porcine skin and left on for (HPγCD) was delivered deep into the skin hair follicles. The (2009)
5 min then the samples were frozen in liquid nitrogen and HPγCD complex appears to selectively target the hair
10-μm-thick slices were prepared and viewed with follicles.
fluorescence microscopy.
Liposomes Anionic liposomes of curcumin/anionic, cationic and Liposomes containing carboxyfluoresceine or curcumin as Liposomal formulations penetrated considerably deeper into Jung et al. (2006)
amphoteric liposomes of carboxyfluoresceine hydrophilic and lipophilic model drugs were applied on the hair follicles and showed approximately 48% of relative
pig ear skin and the depth of penetration was measured by penetration depth at day 3 after application.
laser scanning microscopy in histological sections.
Trends in Food Science & Technology 88 (2019) 445–458
Z. Rafiee, et al. Trends in Food Science & Technology 88 (2019) 445–458

antioxidant and anti-inflammatory effects on the skin. It poses anti- to assess the efficacy and/or safety of formulations inside the living
aging, anti-wrinkle, sunscreen and skin regenerative activities and can cells in in vitro conditions. Furthermore, cell culture studies can be
lead to a better skin appearance. Curcumin can be used as a coloring utilized to understand the related mechanisms of action and also to
agent in nail-care and lip-care products. Nanoencapsulation of cur- estimate cell permeability and absorption of formulations as well. As
cumin can enhance its efficacy, providing better skin penetration and curcumin has significant anti-tumor efficacy, cell culture experiments
retention (Ganesan & Choi, 2016). There are few studies considering are extensively employed to evaluate the anti-tumor activity and its
the cosmetic applications of curcumin-loaded nanocarriers which are related mechanism of action via cellular uptake, cell proliferation,
discussed in the following section and have been summarized in colony forming assay or cell cycle analysis in different cell lines
Table 5. (Naksuriya et al., 2014). Yallupa et al. (2010) evaluated the transport
Coencapsulation of resveratrol and curcuminin in nanocapsules and retention efficiency of curcumin-loaded PLGA nanoparticles in
with lipid core have showed enhanced delivery of these agents and A2780CP and MDA-MB-231 cancer cell lines. They also used cell cy-
increased antioxidant activities; it is suggested that this formulation can totoxicity studies to evaluate the anti-tumor activity and assessed the
delay aging effects on the skin (Coradini et al., 2014; Friedrich et al., expression of apoptosis associated proteins in both cell lines using im-
2015). Liu and Huang (2013) suggested curcumin-loaded lipid vehicles munoblot assay. The results showed 2 and 6-fold increase in curcumin
for the treatment of acne vulgaris as an anti-bacterial and anti-in- uptake into the metastatic cell lines, compared to free curcumin. De-
flammatory agent. They examined skin accumulation and propioni- tailed experiments showed that the nanoparticles can inhibit the pro-
bacterium acnes growth inhibition of the formulation in vitro. The results lifereation and colony formation of cancer cells by enhanced apoptosis
revealed that curcumin-loaded lipid vesicles could considerably accu- (Yallapu, Gupta, Jaggi, & Chauhan, 2010). However, during in vitro
mulate in the neonate pig skin and inhibit propionibacterium acnes (Liu studies cells are exposed to high static concentrations of curcumin for a
& Huang, 2013). Kaur and Saraf (2011) examined the photoprotective long time and interpretation of the results to the dynamic in vivo con-
effects of vesicular systems of curcumin including liposomes, etho- centrations may be difficult.
somes, and transfersomes on ultraviolet radiation-damaged skin. The On the other hand, in vivo pharmacokinetic profile of curcumin-
skin hydration level and sebum content was assessed and the following encapsulated nanoformulations can significantly influence the efficacy
order was seen for all cream formulations: of formulations. Tumor Xenograft animal models are generally used to
curcumin-loaded transfersomal > curcumin-loaded ethosomal > examine the anti-tumor activity of nanoformulations. Zaman et al.
curcumin-loaded liposomal > curcumin-loaded > empty transfer- (2016) studied cellular uptake, cell proliferation, colony forming assay
some > empty ethosome > empty liposome > base cream and cell cycle inhibition of PLGA nanoparticles in cervical cancer cell
They concluded that curcumin-loaded vesicles could have a better lines. They reported up to about 2-fold more accumulation of curcumin
skin penetration and show good photoprotective effects (Kaur & Saraf, in cervical cancer cells compared to the free form. Curcumin-loaded
2011). nanoparticles could also reduce cell viability and clonogenicity and
Some studies have shown that curcumin-loaded nanocarriers can induce apoptosis in vitro with superior effects to free curcumin. The
effectively penetrate into the hair follicles and can be used for delivery nanoparticles reduced the tumor burden in vivo better than free cur-
of curcumin deep into and through the hair follicles. For example, cumin and could suppress the expression of HPV oncogenic proteins
Konradsdottir, Ogmundsdottir, Sigurdsson, and Loftsson (2009) re- and cell proliferation marker in mice. Curcumin-loaded nanoparticles
ported deep penetration of cyclodextrin-based curcumin nanoformula- showed a superior in vitro and in vivo anti-tumor efficacy compared to
tions in porcine hair follicle (Konradsdottir et al., 2009). Jung et al. free drug (Zaman et al., 2016).
(2006) used curcumin-loaded liposomes as a follicular delivery system In vitro antibacterial and antifungal activity tests are also used to
in pig ear skin. The liposomal formulations penetrated deeply into the evaluate the efficacy of curcumin nanoformulations against proposed
hair follicles and reached a penetration depth of approximately 48% of microorganisms including G+ or G-bacteria. Different in vitro methods
the hair follicle length (Jung et al., 2006). are available to assess susceptibility of bacteria to formulations.
Quantitative dilution methods have been widely used to evaluate the
5. In vitro versus in vivo tests for curcumin-loaded nanocarriers antimicrobial efficacy of curcumin-loaded nanoformulations compared
to the free form (Silva et al., 2018). Krausz et al. (2015) evaluated the
A wide variety of in vitro and in vivo studies can be conducted to growth inhibitory effects of curcumin nanoparticles on methicillin-re-
evaluate the safety and efficacy of curcumin nanoformulations ac- sistant S. aureus (MRSA) and P. aeruginosa in vitro. Both free and nano-
cording to their potential application. In vivo studies can better predict encapsulated curcumin showed in vitro antibacterial effects but the re-
the efficacy and/or safety of nanoformulations inside the body, while, sults did not show a significant difference between curcumin in MRSA
in vitro studies are conducted in a highly controlled condition which isolates, while a significant difference was seen for P. aeruginosa iso-
allows better understanding of details and mechanisms. Release profiles lates. The in vivo efficacy of formulation was also tested in MRSA-in-
are generally defined in a simulated medium that the formulation is oculated murine wound model. Curcumin nanoformulations sig-
intended to be used. The medium is usually a buffered solution with nificantly reduced bacterial counts on days 3 and 7 compared to control
pH = 7.4, simulating release in body fluids, or it can be selected as a and also free curcumin. The same results were seen considering wound
specific medium according to the rout of administration. Lin et al. healing efficacy of formulations (Krausz et al., 2015). Comparison of
(2012) proposed cationic liposome-PEG-PEI complex for encapsulation the in vitro and in vivo results shows that the results may be correlated or
of curcumin as an anti-tumor agent. The release profile was conducted different depending on study conditions; both type of studies are ne-
in phosphate buffered saline at 37 °C and showed a sustained release of cessary and will help us to make better conclusions.
90% of curcumin in about 120 h which can result in controlled release
of drug in tumor area (Lin et al., 2012). Liu, Jing, Han, Zhang, and Tian 6. Conclusion
(2019) examined curcumin-loaded zein nanoparticles for oral delivery
of curcumin. Curcumin release in simulated gastric and intestinal fluids Curcumin is a natural compound with polyphenolic nature. This
(SGF and SIF) was evaluated and compared to its free form in ethanol. bioactive ingredient has been proved to have a wide range of functional
They reported controlled release of curcumin from nanoparticles sug- and biological activities. In addition to uses as food additives such as
gesting the formulation as a proper oral delivery carrier for curcumin colorant and antioxidant, it is used in remedies purposes.
(Liu et al., 2019). Nanoencapsulation techniques have been commonly applied to en-
Cell culture studies are another category of in vitro studies used for hance the functional properties of curcumin. In according to mentioned
evaluation of curcumin nanoformulations. These studies are conducted content in different sections of this review, nanoformulations of

455
Z. Rafiee, et al. Trends in Food Science & Technology 88 (2019) 445–458

curcumin are potentially useful in the near future for the different bio- stabilized curcumin nanoparticles tested in an in vitro blood-brain barrier model and
products but there is a need for further studies, especially on cosme- in Alzheimer's disease Tg2576 mice. The AAPS Journal, 15, 324–336.
Chen, X., Zou, L.-Q., Niu, J., Liu, W., Peng, S.-F., & Liu, C.-M. (2015). The stability,
ceutical and food fields, to give the researchers and industries a broad sustained release and cellular antioxidant activity of curcumin nanoliposomes.
range of deeper information. Moreover, creating more economical Molecules, 20, 14293–14311.
scalable methods to nanoencapsulate curcumin is an industrial ne- Coradini, K., Lima, F. O., Oliveira, C. M., Chaves, P. S., Athayde, M. L., Carvalho, L. M.,
et al. (2014). Co-encapsulation of resveratrol and curcumin in lipid-core nano-
cessity for reducing production costs providing better competition with capsules improves their in vitro antioxidant effects. European Journal of Pharmaceutics
synthetic additives and drugs. Additionally, there is still a pressing need and Biopharmaceutics, 88, 178–185.
to investigate toxicological safety of the curcumin application as a na- Da Silva, A. C., De Freitas Santos, P. D., Do Prado Silva, J. T., Leimann, F. V., Bracht, L., &
Goncalves, O. H. (2018). Impact of curcumin nanoformulation on its antimicrobial
nomedicine for the prevention and treatment of various diseases or/and activity. Trends in Food Science & Technology, 72, 74–82.
as a nanoadditive in the food products. Clinical case-by-case trials can Darvesh, A. S., Carroll, R. T., Bishayee, A., Novotny, N. A., Geldenhuys, W. J., & Van der
be suitable to assure general consumption of these novel bio-sub- Schyf, C. J. (2012). Curcumin and neurodegenerative diseases: A perspective. Expert
Opinion on Investigational Drugs, 21, 1123–1140.
stances.
Deng, L., Kang, X., Liu, Y., Feng, F., & Zhang, H. (2017). Effects of surfactants on the
formation of gelatin nanofibres for controlled release of curcumin. Food Chemistry,
References 231, 70–77.
Dhurai, B., Saraswathy, N., Maheswaran, R., Sethupathi, P., Vanitha, P., Vigneshwaran,
S., et al. (2013). Electrospinning of curcumin loaded chitosan/poly (lactic acid) na-
Aadinath, W., Bhushani, A., & Anandharamakrishnan, C. (2016). Synergistic radical nofilm and evaluation of its medicinal characteristics. Frontiers of Materials Science, 7,
scavenging potency of curcumin-in-β-cyclodextrin-in-nanomagnetoliposomes. 350–361.
Materials Science and Engineering: C, 64, 293–302. Doggui, S., Sahni, J. K., Arseneault, M., Dao, L., & Ramassamy, C. (2012). Neuronal up-
Abdel Fattah, A. A. A., & T.M, F. (2016). Antioxidant activities of raw and nano turmeric take and neuroprotective effect of curcumin-loaded PLGA nanoparticles on the
(Curcuma longa L.) powders. J. Food and Dairy Sci, 7, 443–450. human SK-N-SH cell line. Journal of Alzheimer's Disease, 30, 377–392.
Abdou, E. S., Galhoum, G. F., & Mohamed, E. N. (2018). Curcumin loaded nanoemul- Dovigo, L., Carmello, J., De Souza Costa, C., Vergani, C., Brunetti, I., Bagnato, V., et al.
sions/pectin coatings for refrigerated chicken fillets. Food Hydrocolloids, 83, 445–453. (2013). Chemistry of curcumin: from extraction to therapeutic agent.,“Curcumin-
Aggarwal, B. B., & Sung, B. (2009). Pharmacological basis for the role of curcumin in mediated photodynamic inactivation of Candida albicans in a murine model of oral
chronic diseases: An age-old spice with modern targets. Trends in Pharmacological candidiasis. Medical Mycology, 51, 243–251.
Sciences, 30, 85–94. Dubey, S. K., Sharma, A. K., Narain, U., Misra, K., & Pati, U. (2008). Design, synthesis and
Aguilar, F., Dusemund, B., Galtier, P., Gilbert, J., Gott, D., Grilli, S., et al. (2010). characterization of some bioactive conjugates of curcumin with glycine, glutamic
Scientific opinion on the re-evaluation of curcumin (E 100) as a food additive. EFSA J, acid, valine and demethylenated piperic acid and study of their antimicrobial and
8, 46. antiproliferative properties. European Journal of Medicinal Chemistry, 43, 1837–1846.
Akbik, D., Ghadiri, M., Chrzanowski, W., & Rohanizadeh, R. (2014). Curcumin as a Fang, Z., & Bhandari, B. (2010). Encapsulation of polyphenols–a review. Trends in Food
wound healing agent. Life Sciences, 116, 1–7. Science & Technology, 21, 510–523.
Akhavan, S., Assadpour, E., Katouzian, I., & Jafari, S. M. (2018). Lipid nano scale cargos Faridi Esfanjani, A., Assadpour, E., & Jafari, S. M. (2018). Improving the bioavailability of
for the protection and delivery of food bioactive ingredients and nutraceuticals. phenolic compounds by loading them within lipid-based nanocarriers. Trends in Food
Trends in Food Science & Technology, 74, 132–146. Science & Technology, 76, 56–66.
Akhtar, F., Rizvi, M. M., & Kar, S. K. (2012). Oral delivery of curcumin bound to chitosan Faridi Esfanjani, A., & Jafari, S. M. (2016). Biopolymer nano-particles and natural nano-
nanoparticles cured Plasmodium yoelii infected mice. Biotechnology Advances, 30, carriers for nano-encapsulation of phenolic compounds. Colloids and Surfaces B:
310–320. Biointerfaces, 146, 532–543.
Almeida, H. H., Barros, L., Barreira, J. C., Calhelha, R. C., Heleno, S. A., Sayer, C., et al. Friedrich, R. B., Kann, B., Coradini, K., Offerhaus, H. L., Beck, R. C., & Windbergs, M.
(2018). Bioactive evaluation and application of different formulations of the natural (2015). Skin penetration behavior of lipid-core nanocapsules for simultaneous de-
colorant curcumin (E100) in a hydrophilic matrix (yogurt). Food Chemistry, 261, livery of resveratrol and curcumin. European Journal of Pharmaceutical Sciences, 78,
224–232. 204–213.
Alnashi, B. A. A. A. F., & A, A. (2016). Antimicrobial activity of raw and nano turmeric Gandapu, U., Chaitanya, R. K., Kishore, G., Reddy, R. C., & Kondapi, A. K. (2011).
powder extracts. Middle East Journal of Applied Sciences, 6, 787–796. Curcumin-loaded apotransferrin nanoparticles provide efficient cellular uptake and
Altunatmaz, S. S., Aksu, F. Y., Issa, G., Kahraman, B. B., Altiner, D. D., & Buyukunal, S. effectively inhibit HIV-1 replication in vitro. PLoS One, 6, e23388.
(2016). Antimicrobial effects of curcumin against L. Monocytogenes, S. aureus, S. Ganesan, P., & Choi, D. K. (2016). Current application of phytocompound-based nano-
Typhimurium and E. coli O157: H7 pathogens in minced meat. Veterinarni cosmeceuticals for beauty and skin therapy. International Journal of Nanomedicine, 11,
Medicina, 61. 1987–2007.
Aqil, F., Munagala, R., Jeyabalan, J., Agrawal, A. K., & Gupta, R. (2017). Exosomes for the Gera, M., Sharma, N., Ghosh, M., Huynh, D. L., Lee, S. J., Min, T., et al. (2017).
enhanced tissue bioavailability and efficacy of curcumin. The AAPS Journal, 19, Nanoformulations of curcumin: An emerging paradigm for improved remedial ap-
1691–1702. plication. Oncotarget, 8, 66680–66698.
ARAIZA-Calahorra, A., Akhtar, M., & Sarkar, A. (2018). Recent advances in emulsion- Ghasemi, S., Jafari, S. M., Assadpour, E., & Khomeiri, M. (2017). Production of pectin-
based delivery approaches for curcumin: From encapsulation to bioaccessibility. whey protein nano-complexes as carriers of orange peel oil. Carbohydrate Polymers,
Trends in Food Science & Technology, 71, 155–169. 177, 369–377.
Assadpour, E., & Jafari, S. M. (2018). A systematic review on nanoencapsulation of food Ghasemi, S., Jafari, S. M., Assadpour, E., & Khomeiri, M. (2018). Nanoencapsulation of d-
bioactive ingredients and nutraceuticals by various nanocarriers. Critical Reviews in limonene within nanocarriers produced by pectin-whey protein complexes. Food
Food Science and Nutrition, 1–47. Hydrocolloids, 77, 152–162.
Assadpour, E., Jafari, S. M., & Esfanjani, A. F. (2017). Protection of phenolic compounds Gomez-Estaca, J., Balaguer, M., Gavara, R., & Hernandez-Munoz, P. (2012). Formation of
within nanocarriers. CAB Reviews, 12, 1–8. zein nanoparticles by electrohydrodynamic atomization: Effect of the main proces-
Assadpour, E., Maghsoudlou, Y., Jafari, S.-M., Ghorbani, M., & Aalami, M. (2016). sing variables and suitability for encapsulating the food coloring and active in-
Evaluation of folic acid nano-encapsulation by double emulsions. Food and Bioprocess gredient curcumin. Food Hydrocolloids, 28, 82–91.
Technology, 9, 2024–2032. Gómez-Estaca, J., Gavara, R., & Hernández-Muñoz, P. (2015). Encapsulation of curcumin
Baek, J. S., & Cho, C. W. (2017). Surface modification of solid lipid nanoparticles for oral in electrosprayed gelatin microspheres enhances its bioaccessibility and widens its
delivery of curcumin: Improvement of bioavailability through enhanced cellular uses in food applications. Innovative Food Science & Emerging Technologies, 29,
uptake, and lymphatic uptake. European Journal of Pharmaceutics and 302–307.
Biopharmaceutics, 117, 132–140. Gong, C., Deng, S., Wu, Q., Xiang, M., Wei, X., Li, L., et al. (2013a). Improving anti-
Bhawana, R. K. B., Singh Buttar, H., Jain, V. K., & Jain, N. I. (2011). Curcumin nano- angiogenesis and anti-tumor activity of curcumin by biodegradable polymeric mi-
particles: Preparation, characterization, and antimicrobial study. Journal of celles. Biomaterials, 34, 1413–1432.
Agricultural and Food Chemistry, 59, 2056–2061. Gong, C., Wu, Q., Wang, Y., Zhang, D., Luo, F., Zhao, X., et al. (2013b). A biodegradable
Blanco-Padilla, A., López-Rubio, A., Loarca-PIña, G., Gómez-Mascaraque, L. G., & hydrogel system containing curcumin encapsulated in micelles for cutaneous wound
Mendoza, S. (2015). Characterization, release and antioxidant activity of curcumin- healing. Biomaterials, 34, 6377–6387.
loaded amaranth-pullulan electrospun fibers. LWT-Food Science and Technology, 63, Gopal, J., Muthu, M., & Chun, S. (2016). Bactericidal property of macro-, micro-and
1137–1144. nanocurcumin: An assessment. Arabian Journal for Science and Engineering, 41,
Carlson, L. J., Cote, B., Alani, A. W., & Rao, D. A. (2014). Polymeric micellar co-delivery 2087–2093.
of resveratrol and curcumin to mitigate in vitro doxorubicin-induced cardiotoxicity. Gupta, S. C., Patchva, S., & Aggarwal, B. B. (2013). Therapeutic roles of curcumin:
Journal of Pharmaceutical Sciences, 103, 2315–2322. Lessons learned from clinical trials. The AAPS Journal, 15, 195–218.
Chang, C., Meikle, T. G., Su, Y., Wang, X., Dekiwadia, C., Drummond, C. J., et al. (2019). Hatcher, H., Planalp, R., Cho, J., Torti, F., & Torti, S. (2008). Curcumin: From ancient
Encapsulation in egg white protein nanoparticles protects anti-oxidant activity of medicine to current clinical trials. Cellular and Molecular Life Sciences, 65, 1631–1652.
curcumin. Food Chemistry, 280, 65–72. Heger, M., Van Golen, R. F., Broekgaarden, M., & Michel, M. C. (2014). The molecular
Chang, C., Wang, T., Hu, Q., & Luo, Y. (2017). Caseinate-zein-polysaccharide complex basis for the pharmacokinetics and pharmacodynamics of curcumin and its meta-
nanoparticles as potential oral delivery vehicles for curcumin: Effect of poly- bolites in relation to cancer. Pharmacological Reviews, 66, 222–307.
saccharide type and chemical cross-linking. Food Hydrocolloids, 72, 254–262. Hewlings, S., & Kalman, D. (2017). Curcumin: A review of its' effects on human health.
Cheng, K. K., Yeung, C. F., Ho, S. W., Chow, S. F., Chow, A. H., & Baum, L. (2013). Highly Foods, 6, 92.

456
Z. Rafiee, et al. Trends in Food Science & Technology 88 (2019) 445–458

Huang, X., Huang, X., Gong, Y., Xiao, H., Mcclements, D. J., & Hu, K. (2016). Mohanty, C., Das, M., & Sahoo, S. K. (2012). Emerging role of nanocarriers to increase the
Enhancement of curcumin water dispersibility and antioxidant activity using core–- solubility and bioavailability of curcumin. Expert Opinion on Drug Delivery, 9,
shell protein–polysaccharide nanoparticles. Food Research International, 87, 1–9. 1347–1364.
Hu, P., Huang, P., & Chen, M. W. (2013). Curcumin reduces Streptococcus mutans biofilm Mourtas, S., Canovi, M., Zona, C., Aurilia, D., Niarakis, A., LA Ferla, B., et al. (2011).
formation by inhibiting sortase A activity. Archives of Oral Biology, 58, 1343–1348. Curcumin-decorated nanoliposomes with very high affinity for amyloid-beta1-42
Hussain, Z., Thu, H. E., Amjad, M. W., Hussain, F., Ahmed, T. A., & Khan, S. (2017). peptide. Biomaterials, 32, 1635–1645.
Exploring recent developments to improve antioxidant, anti-inflammatory and anti- Mourtas, S., Lazar, A. N., Markoutsa, E., Duyckaerts, C., & Antimisiaris, S. G. (2014).
microbial efficacy of curcumin: A review of new trends and future perspectives. Multifunctional nanoliposomes with curcumin-lipid derivative and brain targeting
Materials Science and Engineering: C, 77, 1316–1326. functionality with potential applications for Alzheimer disease. European Journal of
Isacchi, B., Bergonzi, M. C., Grazioso, M., Righeschi, C., Pietretti, A., Severini, C., et al. Medicinal Chemistry, 80, 175–183.
(2012). Artemisinin and artemisinin plus curcumin liposomal formulations: Mun, S.-H., Joung, D.-K., Kim, Y.-S., Kang, O.-H., Kim, S.-B., Seo, Y.-S., et al. (2013).
Enhanced antimalarial efficacy against Plasmodium berghei-infected mice. European Synergistic antibacterial effect of curcumin against methicillin-resistant
Journal of Pharmaceutics and Biopharmaceutics, 80, 528–534. Staphylococcus aureus. Phytomedicine, 20, 714–718.
Jafari, S. M. (2017b). Nanoencapsulation technologies for the food and nutraceutical in- Naksuriya, O., Okonogi, S., Schiffelers, R. M., & Hennink, W. E. (2014). Curcumin na-
dustries. Academic Press. noformulations: A review of pharmaceutical properties and preclinical studies and
Jafari, S. M., & Mcclements, D. J. (2017). Nanotechnology approaches for increasing nutrient clinical data related to cancer treatment. Biomaterials, 35, 3365–3383.
bioavailability. Advances in food and nutrition research. Academic Press. Nayak, A. P., Tiyaboonchai, W., Patankar, S., Madhusudhan, B., & Souto, E. B. (2010).
Joung, H. J., Choi, M. J., Kim, J. T., Park, S. H., Park, H. J., & Shin, G. H. (2016). Curcuminoids-loaded lipid nanoparticles: Novel approach towards malaria treat-
Development of food‐grade curcumin nanoemulsion and its potential application to ment. Colloids and Surfaces B: Biointerfaces, 81, 263–273.
food beverage system: Antioxidant property and in vitro digestion. Journal of Food Nelson, K. M., Dahlin, J. L., Bisson, J., Graham, J., Pauli, G. F., & Walters, M. A. (2017).
Science, 81. The essential medicinal chemistry of curcumin. Journal of Medicinal Chemistry, 60,
Jung, S., Otberg, N., Thiede, G., Richter, H., Sterry, W., Panzner, S., et al. (2006). 1620–1637.
Innovative liposomes as a transfollicular drug delivery system: Penetration into Nguyen, T. T. H., Si, J., Kang, C., Chung, B., Chung, D., & Kim, D. (2017). Facile pre-
porcine hair follicles. Journal of Investigative Dermatology, 126, 1728–1732. paration of water soluble curcuminoids extracted from turmeric (Curcuma longa L.)
Kannappan, R., Gupta, S. C., Kim, J. H., Reuter, S., & Aggarwal, B. B. (2011). powder by using steviol glucosides. Food Chemistry, 214, 366–373.
Neuroprotection by spice-derived nutraceuticals: You are what you eat!. Molecular Noorafshan, A., & Ashkani-Esfahani, S. (2013). A review of therapeutic effects of cur-
Neurobiology, 44, 142–159. cumin. Current Pharmaceutical Design, 19, 2032–2046.
Karimi, N., Ghanbarzadeh, B., Hamishehkar, H., Mehramuz, B., & Kafil, H. S. (2018). Pagnussatt, F. A., DE Lima, V. R., Dora, C. L., Costa, J. A. V., Putaux, J.-L., & Badiale-
Antioxidant, antimicrobial and physicochemical properties of turmeric extract- Furlong, E. (2016). Assessment of the encapsulation effect of phenolic compounds
loaded nanostructured lipid carrier (NLC). Colloid and Interface Science from Spirulina sp. LEB-18 on their antifusarium activities. Food Chemistry, 211,
Communications, 22, 18–24. 616–623.
Kaur, S., Modi, N. H., Panda, D., & Roy, N. (2010). Probing the binding site of curcumin in Pandit, R. S., Gaikwad, S. C., Agarkar, G. A., Gade, A. K., & Rai, M. (2015). Curcumin
Escherichia coli and Bacillus subtilis FtsZ–a structural insight to unveil antibacterial nanoparticles: Physico-chemical fabrication and its in vitro efficacy against human
activity of curcumin. European Journal of Medicinal Chemistry, 45, 4209–4214. pathogens. 3 Biotech, 5, 991–997.
Kaur, C. D., & Saraf, S. (2011). Topical vesicular formulations of Curcuma longa extract Pan, K., Zhong, Q., & Baek, S. J. (2013). Enhanced dispersibility and bioactivity of cur-
on recuperating the ultraviolet radiation-damaged skin. Journal of Cosmetic cumin by encapsulation in casein nanocapsules. Journal of Agricultural and Food
Dermatology, 10, 260–265. Chemistry, 61, 6036–6043.
Khalil, N. M., Do Nascimento, T. C., Casa, D. M., Dalmolin, L. F., DE Mattos, A. C., Hoss, I., Paramera, E. I., Konteles, S. J., & Karathanos, V. T. (2011). Microencapsulation of cur-
et al. (2013). Pharmacokinetics of curcumin-loaded PLGA and PLGA-PEG blend na- cumin in cells of Saccharomyces cerevisiae. Food Chemistry, 125, 892–902.
noparticles after oral administration in rats. Colloids and Surfaces B: Biointerfaces, 101, Pathak, L., Kanwal, A., & Agrawal, Y. (2015). Curcumin loaded self assembled lipid-
353–360. biopolymer nanoparticles for functional food applications. Journal of Food Science and
Konradsdottir, F., Ogmundsdottir, H., Sigurdsson, V., & Loftsson, T. (2009). Drug tar- Technology, 52, 6143–6156.
geting to the hair follicles: A cyclodextrin-based drug delivery. AAPS PharmSciTech, Rafiee, Z., Nejatian, M., Daeihamed, M., & Jafari, S. M. (2018). Application of different
10, 266–269. nanocarriers for encapsulation of curcumin. Critical Reviews in Food Science and
Krausz, A. E., Adler, B. L., Cabral, V., Navati, M., Doerner, J., Charafeddine, R. A., et al. Nutrition, 1–77.
(2015). Curcumin-encapsulated nanoparticles as innovative antimicrobial and wound Rai, D., Singh, J. K., Roy, N., & Panda, D. (2008). Curcumin inhibits FtsZ assembly: An
healing agent. Nanomedicine, 11, 195–206. attractive mechanism for its antibacterial activity. Biochemical Journal, 410, 147–155.
Kumar, D. D., Mann, B., Pothuraju, R., Sharma, R., & Bajaj, R. MINAXI. (2016). Rezaei, A., Fathi, M., & Jafari, S. M. (2019). Nanoencapsulation of hydrophobic and low-
Formulation and characterization of nanoencapsulated curcumin using sodium soluble food bioactive compounds within different nanocarriers. Food Hydrocolloids,
caseinate and its incorporation in ice cream. Food & Function, 7, 417–424. 88, 146–162.
Li, X., Chen, S., Zhang, B., Li, M., Diao, K., Zhang, Z., et al. (2012). In situ injectable nano- Sadati Behbahani, E. S., Ghaedi, M., Abbaspour, M., Rostamizadeh, K., & Dashtian, K.
composite hydrogel composed of curcumin, N,O-carboxymethyl chitosan and oxi- (2019). Curcumin loaded nanostructured lipid carriers: In vitro digestion and release
dized alginate for wound healing application. International Journal of Pharmacy, 437, studies. Polyhedron.
110–119. Sadeghi, R., Daniella, Z., Uzun, S., & Kokini, J. (2017). Effects of starch composition and
Lin, Y.-L., Liu, Y.-K., Tsai, N.-M., Hsieh, J.-H., Chen, C.-H., Lin, C.-M., et al. (2012). A type of non-solvent on the formation of starch nanoparticles and improvement of
Lipo-PEG-PEI complex for encapsulating curcumin that enhances its antitumor effects curcumin stability in aqueous media. Journal of Cereal Science, 76, 122–130.
on curcumin-sensitive and curcumin-resistance cells. Nanomedicine: Nanotechnology, Sadeghi, R., Moosavi-Movahedi, A., Emam-Jomeh, Z., Kalbasi, A., Razavi, S., Karimi, M.,
Biology and Medicine, 8, 318–327. et al. (2014). The effect of different desolvating agents on BSA nanoparticle prop-
Li, J., Shin, G. H., Lee, I. W., Chen, X., & Park, H. J. (2016). Soluble starch formulated erties and encapsulation of curcumin. Journal of Nanoparticle Research, 16, 2565.
nanocomposite increases water solubility and stability of curcumin. Food Said, D. E., Elsamad, L. M., & Gohar, Y. M. (2012). Validity of silver, chitosan, and cur-
Hydrocolloids, 56, 41–49. cumin nanoparticles as anti-Giardia agents. Parasitology Research, 111, 545–554.
Liu, C. H., & Huang, H. Y. (2013). In vitro anti-propionibacterium activity by curcumin Sandhir, R., Yadav, A., Mehrotra, A., Sunkaria, A., Singh, A., & Sharma, S. (2014).
containing vesicle system. Chemical & Pharmaceutical Bulletin, 61, 419–425. Curcumin nanoparticles attenuate neurochemical and neurobehavioral deficits in
Liu, Q., Jing, Y., Han, C., Zhang, H., & Tian, Y. (2019). Encapsulation of curcumin in zein/ experimental model of Huntington's disease. NeuroMolecular Medicine, 16, 106–118.
caseinate/sodium alginate nanoparticles with improved physicochemical and con- Sathishkumar, P., Hemalatha, S., Arulkumar, M., Ravikumar, R., Yusoff, A. R. M.,
trolled release properties. Food Hydrocolloids, 93, 432–442. Hadibarata, T., et al. (2015). Curcuminoid extraction from turmeric (Curcuma longa
Loo, C.-Y., Rohanizadeh, R., Young, P. M., Traini, D., Cavaliere, R., Whitchurch, C. B., L.): Efficacy of bromine‐modified curcuminoids against food spoilage flora. Journal of
et al. (2015). Combination of silver nanoparticles and curcumin nanoparticles for Food Biochemistry, 39, 325–333.
enhanced anti-biofilm activities. Journal of Agricultural and Food Chemistry, 64, Shah, B. R., Zhang, C., Li, Y., & Li, B. (2016). Bioaccessibility and antioxidant activity of
2513–2522. curcumin after encapsulated by nano and Pickering emulsion based on chitosan-tri-
Maldonado, L., Sadeghi, R., & Kokini, J. (2017). Nanoparticulation of bovine serum al- polyphosphate nanoparticles. Food Research International, 89, 399–407.
bumin and poly-D-lysine through complex coacervation and encapsulation of cur- Sharma, R. A., Gescher, A. J., & Steward, W. P. (2005). Curcumin: The story so far.
cumin. Colloids and Surfaces B: Biointerfaces, 159, 759–769. European Journal of Cancer, 41, 1955–1968.
Marchiani, A., Rozzo, C., Fadda, A., Delogu, G., & Ruzza, P. (2014). Curcumin and cur- Silva, A. C. D., Santos, P. D. D. F., Silva, J. T. D. P., Leimann, F. V., Bracht, L., & Goncalves,
cumin-like molecules: From spice to drugs. Current Medicinal Chemistry, 21, 204–222. O. H. (2018). Impact of curcumin nanoformulation on its antimicrobial activity.
Mathew, A., Fukuda, T., Nagaoka, Y., Hasumura, T., Morimoto, H., Yoshida, Y., et al. Trends in Food Science & Technology, 72, 74–82.
(2012). Curcumin loaded-PLGA nanoparticles conjugated with Tet-1 peptide for po- Soltani, B., Bodaghabadi, N., Mahpour, G., Ghaemi, N., & Sadeghizadeh, M. (2016).
tential use in Alzheimer's disease. PLoS One, 7, e32616. Nanoformulation of curcumin protects HUVEC endothelial cells against ionizing ra-
Mathew, D., & Hsu, W.-L. (2018). Antiviral potential of curcumin. Journal of Functional diation and suppresses their adhesion to monocytes: Potential in prevention of ra-
Foods, 40, 692–699. diation-induced atherosclerosis. Biotechnology Letters, 38, 2081–2088.
Mehanny, M., Hathout, R. M., Geneidi, A. S., & Mansour, S. (2016). Exploring the use of Sunagawa, Y., Wada, H., Suzuki, H., Sasaki, H., Imaizumi, A., Fukuda, H., et al. (2012). A
nanocarrier systems to deliver the magical molecule; curcumin and its derivatives. novel drug delivery system of oral curcumin markedly improves efficacy of treatment
Journal of Controlled Release, 225, 1–30. for heart failure after myocardial infarction in rats. Biological and Pharmaceutical
Menon, V. P., & Sudheer, A. R. (2007). Antioxidant and anti-inflammatory properties of Bulletin, 35, 139–144.
curcumin. The molecular targets and therapeutic uses of curcumin in health and disease. Suwantong, O., Opanasopit, P., Ruktanonchai, U., & Supaphol, P. (2007). Electrospun
Springer. cellulose acetate fiber mats containing curcumin and release characteristic of the

457
Z. Rafiee, et al. Trends in Food Science & Technology 88 (2019) 445–458

herbal substance. Polymer, 48, 7546–7557. cancer cells. Journal of Colloid and Interface Science, 351, 19–29.
Tayyem, R. F., Heath, D. D., AL-Delaimy, W. K., & Rock, C. L. (2006). Curcumin content of Yallapu, M. M., Jaggi, M., & Chauhan, S. C. (2012). Curcumin nanoformulations: A future
turmeric and curry powders. Nutrition and Cancer, 55, 126–131. nanomedicine for cancer. Drug Discovery Today, 17, 71–80.
Tomren, M., Masson, M., Loftsson, T., & Tønnesen, H. H. (2007). Studies on curcumin and Yallapu, M. M., Jaggi, M., & Chauhan, S. C. (2013). Curcumin nanomedicine: A road to
curcuminoids: XXXI. Symmetric and asymmetric curcuminoids: Stability, activity and cancer therapeutics. Current Pharmaceutical Design, 19, 1994–2010.
complexation with cyclodextrin. International Journal of Pharmaceutics, 338, 27–34. Yallapu, M. M., Khan, S., Maher, D. M., Ebeling, M. C., Sundram, V., Chauhan, N., et al.
Vallianou, N. G., Evangelopoulos, A., Schizas, N., & Kazazis, C. (2015). Potential antic- (2014). Anti-cancer activity of curcumin loaded nanoparticles in prostate cancer.
ancer properties and mechanisms of action of curcumin. Anticancer Research, 35, Biomaterials, 35, 8635–8648.
645–651. Yallapu, M. M., Nagesh, P. K., Jaggi, M., & Chauhan, S. C. (2015). Therapeutic applica-
Wang, H., Hao, L., Wang, P., Chen, M., Jiang, S., & Jiang, S. (2017a). Release kinetics and tions of curcumin nanoformulations. The AAPS Journal, 17, 1341–1356.
antibacterial activity of curcumin loaded zein fibers. Food Hydrocolloids, 63, 437–446. Yang, X. X., Li, C. M., & Huang, C. Z. (2016). Curcumin modified silver nanoparticles for
Wang, X., Jiang, Y., Wang, Y.-W., Huang, M.-T., Ho, C.-T., & Huang, Q. (2008). Enhancing highly efficient inhibition of respiratory syncytial virus infection. Nanoscale, 8,
anti-inflammation activity of curcumin through O/W nanoemulsions. Food Chemistry, 3040–3048.
108, 419–424. Yen, F.-L., Wu, T.-H., Tzeng, C.-W., Lin, L.-T., & Lin, C.-C. (2010). Curcumin nanoparticles
Wongcharoen, W., & Phrommintikul, A. (2009). The protective role of curcumin in car- improve the physicochemical properties of curcumin and effectively enhance its
diovascular diseases. International Journal of Cardiology, 133, 145–151. antioxidant and antihepatoma activities. Journal of Agricultural and Food Chemistry,
Xie, M., Fan, D., Zhao, Z., Li, Z., Li, G., Chen, Y., et al. (2015). Nano-curcumin prepared 58, 7376–7382.
via supercritical: Improved anti-bacterial, anti-oxidant and anti-cancer efficacy. Yi, J., Fan, Y., Zhang, Y., Wen, Z., Zhao, L., & Lu, Y. (2016). Glycosylated α-lactalbumin-
International Journal of Pharmacy, 496, 732–740. based nanocomplex for curcumin: Physicochemical stability and DPPH-scavenging
Xue, J., Wang, T., Hu, Q., Zhou, M., & Luo, Y. (2018). Insight into natural biopolymer- activity. Food Hydrocolloids, 61, 369–377.
emulsified solid lipid nanoparticles for encapsulation of curcumin: Effect of loading Zaman, M. S., Chauhan, N., Yallapu, M. M., Gara, R. K., Maher, D. M., Kumari, S., et al.
methods. Food Hydrocolloids, 79, 110–116. (2016). Curcumin nanoformulation for cervical cancer treatment. Scientific Reports, 6,
Yallapu, M. M., Gupta, B. K., Jaggi, M., & Chauhan, S. C. (2010). Fabrication of curcumin 20051.
encapsulated PLGA nanoparticles for improved therapeutic effects in metastatic

458

You might also like