Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Journal of Controlled Release 296 (2019) 190–201

Contents lists available at ScienceDirect

Journal of Controlled Release


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

Review article

Recent advances in topical delivery of flavonoids: A review T



Ruchika L. Nagula, Sarika Wairkar
Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra 400056, India

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

Keywords: Flavonoids are one of the vital classes of bioactive chemicals, abundantly found in plants. These are natural
Drug delivery systems polyphenolic compounds derived from plant metabolites. Their lipophilic nature and poor solubility lead to
Flavonoids variable and limited oral bioavailability. The substantial pharmacological properties of flavonoids include an-
Novel formulations tioxidant, anti-inflammatory, antiproliferative, photoprotective, depigmentation, anti-aging which are very
Topical route
promising in the treatment of several skin disorders. Thus, various topical delivery systems of flavonoids have
been extensively studied. Mostly, colloidal carriers of flavonoids were reported which are very efficient for
topical route with good encapsulation potential, reduced toxicity, and overcome the limitations of conventional
dosage forms. This review focuses on various formulations aspects, in vitro characterization and in vivo studies of
different classes of flavonoids administered by topical route. Although flavonoids offer tremendous potential in
healing the skin conditions categorically, its clinical translation needs in depth safety and efficacy data, meeting
established regulatory standards.

1. Introduction several flavonoids [7]. However, stratum corneum acts as the major
barrier during skin penetration which can be addressed by preparing
Since ancient times several plants and its products are being used for various delivery systems of flavonoids such as nanoparticles, lipid na-
treating diseases and most of these plants are rich in bioflavonoids. nocapsules, microparticle, microsponges, etc. These novel delivery
Bioflavonoids are the naturally occurring compounds which are sec- systems help in formulating both hydrophilic and lipophilic compounds
ondary metabolites of plants having numerous therapeutic activities thus increasing the concentration of the drug at target site. Other ad-
[1]. They are low molecular weight polyphenolic compounds and stated vantages of topical route are controlled drug release at predetermined
as non‑nitrogenous plant pigments. It is said that a diet rich in fruits and rate and no interaction with the gastric and intestinal fluids, preventing
vegetables is a healthy diet, flavonoids may be one of the reasons [2,3]. degradation of product. The topical route is also very useful in delivery
Flavonoids consist of C6-C3-C6 ring as basic structure, with varying of drugs with shorter half-life and narrow therapeutic index [8]. While
substitutes to create different subclasses such as flavones, flavanol, designing topical formulation, various points need to be considered like
flavonol, isoflavones, anthocyanins, as shown in Fig. 1 [3]. partition coefficient, pH, polymorphism, particle size, molecular
Bioflavonoids are valuable in both, for health promoting activities weight, vehicles, release characteristics, etc.
and therapeutic benefits [4]. Health promoting effect improves well- Several topical preparations of flavonoids have been formulated and
being and reduces the chances of developing risk that include beneficial patented, showcasing wide range of activities as described in Table 1.
cardiovascular effect, increased eyesight, improved blood circulation, Patent overview of flavonoids would help to understand the relevant
immune system, etc. [5]. Therapeutic applications of bioflavonoids flavonoids, their molecular targets, delivery systems and commercial
have extensively been reported in various diseases such as inflamma- significance and thus, important to capture the gaps in flavonoids
tion, cardiovascular, respiratory, diabetes, etc. Categorically, these spectrum and to decide future research strategies.
biologically active compounds possess powerful antioxidant property With due respect to tremendous therapeutic potential of flavonoids
and act by several pathways as seen in Fig. 2 [6]. combined with certain inherent limitations leads to development of
Flavonoids are highly lipophilic compounds and therefore difficult new formulations for alternative routes of administration. In this re-
to absorb orally. They undergo excessive first pass metabolism on oral view, we have attempted to take comprehensive review of topical de-
administration that hampers their oral bioavailability. Thus, an alter- livery systems of flavonoids with recent advances and lingering chal-
native topical route of administration has been explored for delivery of lenges for their successful delivery.


Corresponding author.
E-mail address: sarikawairkar@gmail.com (S. Wairkar).

https://doi.org/10.1016/j.jconrel.2019.01.029
Received 6 December 2018; Received in revised form 18 January 2019; Accepted 18 January 2019
Available online 22 January 2019
0168-3659/ © 2019 Elsevier B.V. All rights reserved.
R.L. Nagula, S. Wairkar Journal of Controlled Release 296 (2019) 190–201

Fig. 1. Classification of flavonoids based on chemical structure.

Fig. 2. Antioxidant effect of flavonoids by (a) ROS inhibition (b) leukocyte immobilization (c) nitric oxide inhibition (d) inhibition of xanthine oxidase.

2. Structure of skin and barriers in topical delivery keratinocytes and acts as the barrier to fluids such as water. Stratum
corneum is the last layer which acts as semipermeable membrane and
The largest organ of body, skin, helps in body protection, tem- inhibits entry of microbes and chemicals. It is the hardest layer and
perature regulation and acts as an organ of sensation. The skin is di- keratinocytes in this layer gets keratinized and the nucleus is lost.
vided into three layers namely epidermis, dermis and subcutaneous Dermis is rich in blood capillaries which provide support and nu-
layer as depicted in Fig. 3. trition to the cells in epidermis that is divided into two layers, papillary
Epidermis is the outermost layer of the skin which is divided into dermis and reticulate dermis. Papillary dermis is thin layer comprises of
four layers and acts as major barrier. Stratum basalis is present next to loosely arranged collagen and reticulate dermis is thick portion con-
dermis layer and is the bottommost layer of epidermis. Second layer taining subcutaneous fat. The major cells in dermis layer are fibroblasts,
above stratum basalis is stratum spinosum in which keratinocytes are mast cells and macrophages. Subcutaneous layer consists of adipocytes
bound to each other by desmosomes which maintains the structural which provide thermoregulation and insulation to body and it also acts
integrity of skin. The next layer stratum granulosm is made up of flatter as energy storage system. Adipocytes are widely distributed in

191
R.L. Nagula, S. Wairkar Journal of Controlled Release 296 (2019) 190–201

Table 1
List of patents on topical delivery of flavonoids.
Patent no. Patent title Original assignee/applicant Grant date Reference

US6471973B2 Flavonide esters and their use notably in cosmetics Coletica 29-10-2002 [9]
WO2001049285A1 Flavonoid drug and dosage form, its production and use Slk Foundation 12-7-2001 [10]
US8613910B2 Flavonoids as synergists for enhancing the action of self-tanning substances Merck patent GmbH 24-12-2013 [11]
US6409996B1 Composition comprising one or more flavonoids, method of obtaining such composition Flavone Sunproducts AS 25-6-2002 [12]
and use thereof as UV-absorbing agent
EP0774249A2 Topical composition containing specific flavanones Unilever PLC, Unilever NV 21-5-1997 [13]
US8440704B2 Quercetin containing compositions Quercegen Pharmaceuticals LLC 14-5-2013 [14]
US20170020798A1 Hesperidin containing compositions and methods for treatment of skin disorders University of California 26-1-2017 [15]
WO2014090586A1 Delphinidin for combating melanoma cells Sapiotech Gmbh 19-6-2014 [16]
US20040191327A1 Methods of treating and/or preventing asthma using natural compound luteolin Council of scientific and industrial 30-09-2004 [17]
research
US20150342843A1 Topical liposome composition containing phenolic anti-inflammatory agents and their D&J Rosee Inc 3-12-2015 [18]
method of preparation
US20100069476A1 Composition and methods for reduction of cutaneous photoaging Mitsui Norin Co Ltd. 18-3-2010 [19]
US20120213842A1 Methods of making and using compositions comprising flavonoids API Genesis LLC 23-08-2012 [20]

subcutaneous layer; two types are white and brown adipocytes. between the corneocyte and need to cover the gap of 400 μm which
[21–23]. decreases the rate of permeability. Additionally, the shunt pathway is
The major barrier for topical delivery is the complicated physiology also reported for skin transport which consists of lipophilic follicular
of the skin. Among all these layers stratum corneum acts as the major ducts or hydrophilic sweat glands and also called as appendageal route
barrier as it consists of densely packed lipid mortar and protein bricks [24].
which limits the drug permeability. The stratum granulosm contains
desmosomes which forms the interlock with neighboring cells and acts 3. Advances in topical drug delivery
as barrier. It also includes keratinocytes that produce keratin and the
glycolipid and acts as water repellant. Along with this, the other in- Topical delivery is one of the most preferable routes to overcome
terruptions are the characteristics of the drug such as molecular weight, the drawbacks of other routes such as parenteral, oral etc. [25]. Oral
degree of ionization, partition coefficient, diffusion coefficient, and delivery of phytoceuticals is undesirable due to its characteristic taste
physicochemical nature of the drug. Drugs with only molecular weight and odor, also there are chances of gastrointestinal (GIT) degradation of
less than 500 Da show good permeability. The diffusion of the drug the drug till absorption [26] whereas patient compliance is compro-
from the vehicle is also important and so the characteristics of the ve- mised in parenteral route. Nevertheless, the most difficult challenge in
hicle should also be considered. topical delivery is the impermeable nature of the skin. The therapeutic
There are different skin penetration pathways (Fig. 3) and thereby effect produced by the conventional topical dosage form is not very
targeting the optimum one is equally important for effective topical effective, so the research has been shifted to development of novel drug
delivery. In transcellular pathway, the drug passes through stratum delivery systems [23,24]. Conventionally, gels are the most commonly
corneum which comprises of keratinocytes and lipid membrane thus, it used dosage form in topical delivery, mainly olegels and hydrogels but
is difficult for the drug to pass through both the lipophilic and hydro- the recent studies have reported wide range of gels such as niosomal,
philic layers. The alternative and the most commonly used pathway is proniosomal gels, emulgels, bigels, aerogels, xerogels which assists in
intercellular pathway, in this the drug travels through the space producing the desirable therapeutic effect [25,26,27].

Fig. 3. Structure of skin with penetration pathways.

192
R.L. Nagula, S. Wairkar Journal of Controlled Release 296 (2019) 190–201

Novel drug delivery of phytochemicals is the most preferred one due showed less than 15% release after 24 h, signifying sustained behavior
to minimum side effects, less toxicity, increased solubility and bioa- in the skin. Anti-inflammatory activity of quercetin LNC (5 μg/ml) was
vailability. It also helps in providing site specific delivery and with evaluated on THP-1 cells of human leukemic monocytes and it was
better pharmacokinetic parameters [30]. Phytochemicals possess wide found that THP-1 cells were unharmed, as quercetin prevents
range of pharmacological properties such as anti-inflammatory, anti- inflammation by scavenging free radical, protecting keratinocytes on
oxidant, apoptosis, photoprotective, antiproliferative and opportunely, skin. It was observed that quercetin LNCs decreased ROS in cell, thus
all these activities can be directly linked to the skin and thus, topical reducing inflammation. LNCs also showed sustained release with no
delivery [31]. Extensive research has evolved in the investigation of toxic effect [42]. Thus, LNCs act as a good carrier for topical delivery of
new carrier systems for phytochemicals such as phospholipid carriers, quercetin and provide triple protection by preventing its degradation,
nanoparticles, microparticles, liposomes, etc. [28–31]. These carriers increasing the occlusive effect on skin and improving water solubility.
can later be formulated into into gels, creams and other dosage forms
which help in increasing the therapeutic line of phytochemicals [32- 4.1.1.2. Quercetin nanoparticles. Nanoparticles have a greater efficiency
36]. to permeate and accumulate within the skin to produce therapeutic
activity. Quercetin nanoparticles (QN) prepared from lecithin, chitosan,
4. Topical delivery systems of flavonoids tween 80, and TPGS had perfect round shape due to interaction
between the polycationic chitosan and negative lecithin. Lecithin
Various novel formulations of flavonoids for topical route are clas- formed core and chitosan shaped the outer protective layer of
sified as per their chemical class as mentioned below. nanoparticles. The particle size of QN was 95.3 nm and zeta potential
was 10.85 ± 0.05 mV. In vitro evaluation of QN on dorsal skin of mice
4.1. Flavonols showed significant amount of drug in the dermis and epidermis layer of
skin. In vivo study was performed in mice using 0.5% w/w quercetin
Flavonols are O-glycosidic, ketonic compounds with sugar moiety at loaded nanoparticle suspension, the concentration of drug in dermis
the 3-position. Flavonols act as antioxidants and protects reactive and epidermis was found to be 8.4 ± 1.53 μg and 2.21 ± 0.01 μg
oxygen species (ROS) formation. Skin is the most common target for respectively [43]. It was seen that nanoparticles altered the skin surface
oxidative stress due to UV, ozone radiations, and other harmful che- and stratum corneum and disrupted the conjugation between
micals. The antioxidant property of flavonols is due to the combination corneocyte layers that resulted in increased permeation of quercetin
of conjugated double bond present in the C-ring along with neighboring and also increased retention time in the skin.
hydroxyl group in the B-ring [37]. Compounds belonging to this class
are quercetin, kaempferol, myricetin, etc. 4.1.1.3. Quercetin microemulsion. Roberta Censi et al. studied quercetin
microemulsions to determine its penetrability through the skin and to
4.1.1. Quercetin evaluate drug concentration in systemic circulation. Ternary phase
Quercetin is a flavonol abundantly found in leafy vegetables, citrus diagram was plotted with Labrafil®M1944CS, Labrasol® and
fruits, berries etc. Quercetin reduces inflammation by decreasing the Capryol™90 and Transcutol® P. The physicochemical parameters of
edema, leukocyte formation and irritation [38]. It triggers tissue re- microemulsion such as pH, particle size distribution, % drug content
generation by promoting the growth of new collagen fibers and pro- was found to be 5.3, 10-50 nm, 98.00 ± 1.70% respectively. In vitro
ducing ground substance to restore the skin structure [39]. Also, study through pig skin revealed the good diffusion rate of quercetin in
quercetin inhibits various inflammatory mediators such as interleukins microemulsion formulation [44]. But the disadvantage of
(IL), prostaglandins (PGs), produced by COX, LOX, LPS. Nitric oxide microemulsions is that, their retention time in skin is very less due to
(NO) is one of the inflammatory mediators synthesized from nitric rapid transdermal absorption.
oxide synthase which generates reactive nitrogen species such as per-
oxynitrite. An anti-inflammatory activity of quercetin and various other 4.1.1.4. Quercetin nanostructured lipid carrier and solid lipid
flavonoids is depicted in Fig. 4. Quercetin helps in inhibiting all the nanoparticles. Lipid based nanosystems such as quercetin
mediators which causes oxidative stress, thus producing the antioxidant nanostructured lipid carrier (NLC) and solid lipid nanoparticles (SLN)
effect [40]. It prevents cell death by inhibiting caspase-3 pathway and have been a promising approach for the various hydrophobic drugs.
decreases the level of histidine decarboxylase, IL-6, monocyte che- Quercetin NLCs prepared by probe ultrasonication method made up of
moattractant protein (MCP-1) by inhibiting the mast cell secretion that Compritol® 888. SLN was prepared in the same way except Compritol
leads to anti-allergic effect [41]. 888 was substituted with oleic acid.
The poor water solubility of quercetin results in its limited bioa- Zeta potential of spherical shaped quercetin loaded NLC and SLN
vailability. In spite of having poor bioavailability, it produces several was found to be −35.64 ± 1.13 mV and − 35.83 ± 2.11 mV, in-
therapeutic benefits and it is also an important intermediary of its bio- dicating good stability. In vitro drug release of NLC was found to be
actives. Quercetin is unstable compound with less skin permeability. So, biphasic with initial burst release followed by controlled release. The
it is formulated into various forms such as nanoemulsion, nanocapsules, release of quercetin from NLC was approximately 55% and that of SLN
solid lipid nanoparticles, and microemulsion etc. which will not only was 45% in 2 h [45]. The addition of the liquid lipid decreased the
increase its solubility but also improve the skin permeability. viscosity of the NLC, resulting in the rapid release of the drug, thus
increasing the permeability. It also exhibited anti-inflammatory activity
4.1.1.1. Quercetin lipid nanocapsules. Among the various by suppressing edema and scavenging the superoxide radicals. Hence
nanoformulations, lipid nanocapsules (LNCs) act as a promising quercetin NLCs and SLN have proven to be good option to target topical
approach to encapsulate quercetin, due to the presence of lipid delivery.
structure. Hatahet et al. reported spherical LNCs prepared with
Cremophor EL having particle size ranged from 26 ± 3 nm - 4.1.1.5. Quercetin penetration enhancer containing vesicle. Penetration
54 ± 3 nm. Addition of Cremophor EL showed higher encapsulating enhancer containing vesicles (PEVs) act as excellent carrier due to the
efficiency of quercetin due to its OH groups which interact with the presence of both, phospholipid and penetration enhancers which
polar groups of quercetin. LNCs formed were perfectly spherical and the provide synergistic effect on permeation. Quercetin loaded PEV's
particle size ranged from 26 ± 3 nm to 54 ± 3 nm. Quercetin loaded were formulated using 4 penetration enhancers namely Transcutol® P
LNCs exhibited 5000-fold increase in water solubility and revealed (Trc), Labrasol® (lab), PEG 400, PG. The size of the liposomal vesicles
good stability at 4 °C and 25 °C. In vitro release study of quercetin LNC containing PEG and Trc which was around 200 nm, greater than that of

193
R.L. Nagula, S. Wairkar Journal of Controlled Release 296 (2019) 190–201

Fig. 4. Anti-inflammatory effect of flavonoids [COX: Cyclooxygenase; LOX: Lipoxygenase; PGs: Prostaglandins; LTs: Leukotrienes; I kB: Inhibitory factor Kappa B].

Lab and PG i.e. approximately 80 nm. Zeta potential value of PEG and efficient for the quercetin delivery.
Trc PEV's was higher (around -50 mV), indicating high stability without Quercetin being lipophilic in nature, various nanoformulations have
any aggregation of the vesicles but the Lab-PEVs showed lower zeta been developed to increase its bioavailability. There is a need of pe-
potential (-30 mV). The outcome of rheology studies confirmed that the netration enhancers for quercetin topical delivery, however, the extent
shear rate increased with the increase in the shear stress and the shear of skin penetration was found to be dependent on lipid content and its
viscosity was independent of shear stress. The viscosity of the four type. Also, application of quercetin in other skin disorders such as
formulations with Trc, PG, PEG 400 and Lab-PEVs were 5, 6, 9 and 40 psoriasis and atopic dermatitis can be explored in future.
mPas respectively. The higher value of viscosity was attributed to the
high hydrodynamic volume occupied by the lamellar structure of the
vesicles. It was also seen that the amount of quercetin that can be 4.1.2. Kaempferol
delivered decreases with the increase in the viscosity. Ex vivo study was Kaempferol is mostly found in berries and plants belonging to spe-
performed by Franz diffusion cell on skin of new born pig. Amount of cies of allium and brassica. Kaempferol possesses anticancer, anti-
drug in various layers of the skin was evaluated i.e. stratum corneum, oxidant, anti-inflammatory, and anti-allergic activity [48–51]. Kaemp-
epidermis, dermis and receptor fluid, the highest concentration of drug ferol inhibits nitric oxide synthase which forms nitric oxide, a pro
was found in the epidermis and lowest in the stratum corneum [46]. inflammatory mediator to act as anti-inflammatory agent. It also in-
hibits the nuclear factor kappa B (NF-kappa B), with help of nuclear
4.1.1.6. Quercetin liposomes and PEVs. PEV's help in increasing the factor inducing kinase (NIK) and mitogen activated protein kinase
permeation of the drug though the skin and promotes deposition in all (MAPKs) [52]. In addition, kaempferol obstructs COX-2 by inhibiting
three layers of skin. In this study, 10% of quercetin was loaded in in all nitric oxide synthase and TNF-α to produce anti-inflammatory effect.
three formulations such as liposomes and PEVs (5% PEG-PEVs, 10% But, kaempferol undergoes excessive first pass metabolism and there-
PEG-PEVs), prepared using Lipoid (S75) and soya lecithin. The size of fore, bioavailability is only 2% and hence topical route is preferred for
the liposomes was 100-150 nm, with zeta potential of -10 mV, its delivery.
indicating poor stability. Whereas the size of 5%PEG-PEVs and 10% Kaempferol acts as novel agent in treating the UVB induced tu-
PEG-PEVs was approximately 190 nm with -50 mV zeta potential morigenesis and photo-inflammation. Kaempferol was studied in skin
representing good stability. In in vivo anti-inflammatory study, 5% cancer where level of the COX 2 enzymes is found to be elevated. It
PEG-PEV showed 4.7-fold decrease in myeloperoxidase (MPO) activity inhibits the AP-1 (Activator protein) activity by suppressing COX-2 in
while quercetin loaded liposome and 10%PEG-PEV showed 2.5-fold JB6 P+ mouse epidermal cells. Thus, to check the transactivation of
decrease in the MPO activity. The MPO activity of the quercetin 5%v/v AP-1, JB6 P+ mouse epidermal cells was transfected by the luciferase
PEG/ PEV was more effective than diclofenac in same PEG/PEV system reporter plasmid bearing AP-1 and it was found that kaempferol suc-
[47]. Thus, quercetin has superior anti-inflammatory activity, ROS cessfully inhibits the COX and AP-1 activities in dose dependent manner
scavenging and increased fibroblast proliferation which helps in the assisting in antitumor promoting activity. Proto-oncogene tyrosine
rapid wound healing. PEVs helps in localization of quercetin into the protein-kinase Src (Src) is the protooncogene molecule which has main
cells at the site of inflammation which causes the degradation of the function in the cell growth, differentiation and survival. Kaempferol
cells by the lysosomal enzymes. Confocal images of the treated mice competes with ATP for the Src binding site and blocks the Src activity.
skin showed large number of collagen fibers and ground substance Topical application of kaempferol on the dorsal skin of mouse inhibited
demonstrating healing has occurred. Hence the vesicular system is COX-2 expression thus, producing anti-inflammatory effect [53,54].

194
R.L. Nagula, S. Wairkar Journal of Controlled Release 296 (2019) 190–201

4.1.2.1. Kaempferol submicron emulsion. Yun chao et al. reported


submicron emulsion of 0.1% of kaempferol, prepared by pseudo
ternary phase diagram in which ethanol or PEG 400 were used as co-
surfactant. The area of PEG 400 was very less compared to ethanol.
Hence, ethanol was preferred co-surfactant and it might help to
penetrate more easily due to its smaller chains. The size of
microemulsions was found between 75.3 and 85.4 nm and in vitro
permeation study on rat skin followed zero order kinetics [55]. Increase
in the drug deposition with the reduction in lag time was attained,
which proves that submicron emulsion act as excellent carriers along
with the increased retention time.
Research papers reported that various pharmacological activities of
kaempferol and its presence in several herbal sources Thus, a wide
range of nanoformulations like nanoemulsion, SLN, liposomes etc. can
also be evaluated further. A comprehensive clinical data is required to
decide the effective dose of kaempferol.

4.2. Flavanones

Flavanones are aromatic ketones derived from flavones, abundantly Fig. 5. Depigmentation effect of flavonoids [α- MSH: Melanocyte stimulating
hormone, MC1R: Melanocortin 1 receptor, MiTF: Microphthalmia associated
found in many citrus fruits like oranges, lemons. Byproduct of the citrus
transcription factor, Tyrp-1: Tyrosinase related protein 1, Tyrp-2: Tyrosinase
cultivation also yields huge amount of the hesperidin. They have cy-
related protein 2, ASIP: Agouti signaling protein].
totoxic activity and inhibit tumor growth and can act as anticancer
agent. Flavanones also inhibit protein tyrosine kinase which decreases
cell growth, differentiation, mitosis, apoptosis and produce anti-in- contact with physiological fluid it converts to gel. Hence high amount
flammatory activity [56]. of drug can be loaded without using the organic. In vitro studies,
95.3 ± 1.3% hesperetin was released within 120 min. Ex vivo study
4.2.1. Hesperetin and hesperidin was performed on the whole eye globes acquired from pel-Freez and
Hesperidin has various activities like anti-inflammatory, anti- were placed in tissue culture plates containing 12-wells, with cornea on
oxidant, Antidiabetic, neuroprotective, etc. [57–60]. Hesperetin is the upper side. The 10%w/w hesperetin film was applied at corneo-scleral-
aglycone moiety of hesperidin that shows antioxidant, anti-in- limbus for 3 h and the concentration of the drug in the aqueous humor,
flammatory activity by interfering with arachidonic acid and also in- vitreous humor (VH), retina choroid (RC) was found to be
hibits COX and LOX enzymes and prevents formation of inflammatory 5.8 ± 0.2 μg/g, 5.6 ± 1.4 μg/g, 14.2 ± 3.8 μg/g of tissue. In vivo
mediators [60]. Hesperetin has the ability to reduce hyperpigmentation study was performed on albino rabbits where 10%w/w and 20% w/w
caused by UV rays, producing whitening effect. Hesperetin and he- HT film were kept in the conjunctival sacs. The hesperetin level for 10%
speridin inhibits the release of histamine from mast cells to produce was observed in VH (0.05 ± 0.03 μg/g of tissue) RC (7.8 ± 0.2 μg/g
anti-allergic activity. They also have the ability to reduce HMG CoA of tissue) and for 20% the concentration in VH (1.4 ± 0.5 μg/g of
reductase and acyl CoA, thus producing the hypolipidemic action. He- tissue) RC (23.5 ± 6.9 μg/g of tissue) respectively which showed the
speretin has log P value 1.7 to 2.20, which makes it lipophilic in nature linear rise in drug concentration with the dose [62]. The film on ap-
and would be difficult to absorb orally. Considering its pharmacological plication transformed into gel and did not show any allergic condition
actions and physicochemical properties, topical route is more suitable such as redness, selling, irritation, excess tear, etc.
for the hesperetin delivery.
4.2.1.3. Hesperidin microemulsions. Hesperidin has the capacity to
4.2.1.1. Hesperetin microemulsion. Microemulsions of hesperetin were decrease the capillary permeability and the fragility, therefore it can
prepared having particle size ranged from 101.5-233 μm. be used in treatment of various blood vessel related diseases. Yet, it is
Microemulsion formulations exhibits lower lag time and therefore, very slightly soluble in water and unstable at the gastric pH and
increased the permeation rate and localization of hesperetin. In vitro undergoes hydrolysis to produce its aglycone hesperetin. But hesperidin
permeation study on rat skin exhibited distinct increase in the shows high permeability through skin which can be utilized to
permeation rate with decrease in the surfactant concentration. formulate its topical formulations. Microemulsions based hesperidin
Hesperetin was evaluated in vivo for the whitening effect on guinea ointment (MBO) was prepared with eucalyptus oil acts as penetration
pigs' dorsal skin after the UV irradiation exposure [61]. UV rays cause enhancer. The permeability and the bioavailability of MBO was more as
hyperpigmentation which leads to dark skin and application of compared to the plain microemulsions. In vivo studies for MBO and
hesperetin microemulsion decreases hyperpigmentation. The microemulsion on the dorsal skin surface of wistar rats demonstrated
mechanism of action of depigmentation of hesperetin and other Cmax of 30.3 and 11.3 μg/ml, the AUC of 66.7 ± 0.2 and 43.26 ± 0.1
flavonoids is demonstrated in Fig. 5. Additionally, hesperetin for MBO and microemulsion correspondingly. But there was no
microemulsion decreased the skin irritation and revealed anti- difference observed in Tmax of both formulations [63].
inflammatory action. Hesperetin microemulsion has proved to be more efficient in pro-
ducing whitening effect by decreasing hyperpigmentation, therefore
4.2.1.2. Hesperetin topical film for eye delivery. Hesperetin is strong various topical nano-formulations can be studied for skin. Hesperidin
antioxidant activity than hesperidin in scavenging ROS. It has the shows improved bioavailability to treat venous diseases by decreasing
property to increase the ocular blood flow and restore the retina. It is permeability of capillaries. Thus, vasoprotective activity of hesperetin
used as dietary supplement to provide vasoprotective effect. Posterior can be utilized by preparing ocular formulations.
of the eye has unique structure which acts as physiological barrier,
therefore the treatment of back of the eye is a big challenge. 4.2.2. Naringenin
Poly ethylene oxide N10 film with 10–20% of hesperetin was pre- Naringenin possess various activities like antioxidant, anti-in-
pared the by melt cast method by Adellia et al. When the drug comes in flammatory, antidiabetic, anticancer, etc. [64–67]. It shows antioxidant

195
R.L. Nagula, S. Wairkar Journal of Controlled Release 296 (2019) 190–201

effect by chelating metal ions and by inhibiting the xanthine oxidase scavenging free radicals at the site of wound.
thus preventing the formation of oxygen radicals and lipid peroxida-
tion. It also has the ability to scavenge ROS by –OH substitution. Nar- 4.3.1.1. Nano-transfersomes epigallocatechin-3-gallate and hyaluronic
ingenin being hydrophobic has low solubility and poor bioavailability, acid. Epigallocatechin-3-gallate (EGCG) is abundantly found in green
so the topical route can be explored for effective formulations [68]. tea, possessing a wide range of above said therapeutic effects [74].
Since poor absorption and low bioavailability are of major concerns of
4.2.2.1. Naringenin submicron emulsion. Preparation of the submicron oral catechins, the topical formulation was attempted by Avadhani
emulsion is easy, economical and high amount of drug can be loaded et al. for its antioxidant and anti-aging effects. Therefore, formulating
with a good stability. Naringenin submicron emulsions showed 4.5–9.4 EGCG into transferosomes may increase its permeability and
times increased drug deposition as compared to saturated aqueous bioavailability [80]. Incorporation of EGCG along with the hyaluronic
solution of naringenin as control group. Skin irritation test did not show acid (HA) was studied to check synergistic effects of anti-aging and
any erythema and edema compared to the control and was found to be antioxidant. In vitro cytotoxicity study in the human keratinocytes cells
biocompatible with skin. [69]. was evaluated and the cell viability of the transferosomes containing
HA along with EGCG was greater compared to the EGCG
4.2.2.2. Naringenin gel. Naringenin based Carbopol gel (0.5%) was transferosomes. In vitro permeation study was performed on male
evaluated topically for antioxidant and anti-inflammatory activity. In Wistar rat skin with plain EGCG, EGCG transferosomes and EGCG
vitro antioxidant study by the FRAP assay showed that the reducing with HA transferosomes. The Q12 (cumulative amount of EGCG
effect of 0.5% naringenin was 0.157 μg/ml. The IC50 value for the ABTS permeated at the end of 12 h) values were found to be
[2,20-azinobis (3ethylbenzothiazoline- 6-sulfonicacid], OH radical, 56.08 ± 4.42 μg/cm2, 159.90 ± 6.90 μg/cm2 and 199.1 ± 6.30 μg/
iron independent LPO assay was 0.71 μg/ml, 183 μg/ml and 159 μg/ cm2 correspondingly. It was concluded that that HA possesses
ml respectively. In vivo evaluation of naringenin in the inflammation moisturizing effect which increase deposition, retention time of
induced HRS/J mice inhibited the skin edema, production of the EGCG. Also, permeability of EGCG with HA transferosomes was 2.8
cytokines, catalase activity and reduced glutathione levels [70]. times greater compared to the plain EGCG [81]. In addition, topical
application of EGCG.
4.2.2.3. Naringenin elastic liposome. Ming-JunTsai et al. have prepared decreases the erythema, COX activity and decreases the level of PGs,
the naringenin loaded liposomes (5 mg/ml) and evaluated for in vitro NO and H2O2 production.
study permeation rate. The permeation rate of the saturated aqueous
solution (0.25 ± 0.1 μg/h/cm3) was less than that of naringenin
loaded liposomes (0.37 ± 0.15 μg/h/cm3) at 24 h. There was no skin 4.3.1.2. Grape seed extract cream. Grape seed extract (GSE) is rich in
irritation observed after the topical application and stability study polyphenols containing proanthocyanidins [77–79] acts as a
performed at the 4 °C showed neither aggregation nor creaming, therapeutic aid the wound healing [82-84]. The experiment was
indicating the stable formulation for three months [71]. conducted by using hydroalcoholic GSE in Eucerin base for the
wound healing and was compared against phenytoin cream for
4.2.2.4. Naringenin nanoparticles-based sunscreen cream. Naringenin 13 days [85]. Complete wound healing was found as compared to the
nanoparticles were prepared to explore its photo-protective and control group. Wound healing was due to the presence of
antioxidant effect in the sunscreen cream with polyvinyl alcohol and proanthocyanidins (condensed tannins) which increase the tissue
poly (D, L-lactide-co-glycolide). In vitro permeation study of naringenin oxidation at the site of wound and promotes the wound healing by
and naringenin nanoparticles at 12 h was found to be inducing apoptosis. The pathway of apoptosis for catechins and
124.88 ± 2.24 μg/cm2 and 184.03 ± 3.37 μg/cm2 respectively. It apigenin is described in Fig. 6 [86]. Therefore, it could be used
was observed that the retention of the nanoparticles in epidermis was topically for the wound contraction and closure.
considerably higher than plain naringenin. In vivo study was performed Although catechins have been extensively studied for anticancer
on Wistar rats that displayed significant skin retention (100 ng/cm2) property, the group also possesses the properties helpful in treating skin
without any toxicity [72]. disorders. Stability is the major disadvantage of catechins with respect
The wide spectrum of naringenin therapeutic activities and reported to pH, temperature and light which leads to rapid epimerization.
scientific evidences makes it an interesting flavonoid to be worked on.
However, it lacks the sufficient clinical data for commercial utilization.
Although above delivery systems have been reported for naringenin,
topical formulations like microsponges, nanosponges, microneedles can
be attempted for skin disorders [73].

4.3. Flavanols

They are different from the flavonol and not to be confused, they
lack ketone group. The class includes epigallocatechin-3-gallate,
proanthocyanidins, etc. They are naturally found in tea, cocoa, and
various vegetables and fruits [74]. FDA has approved the use of ca-
techins and its derivatives in various pharmaceutical formulations.

4.3.1. Catechins
The various activities of catechins include antioxidants, photo-
protective, antiaging, anti-inflammatory, anticancer, neuroprotective,
cardioprotective, antiviral and antibacterial [75–77]. Grape seed ex-
tract and tea polyphenols are rich in epigallocatechin, epicatechin. It
shows antioxidant effect by scavenging free radicals [78]. Inhibition of
the COX enzyme, PGs, NO and H2O2 production leads to the anti-in-
flammatory activity. Catechins promote the wound healing by Fig. 6. Apoptosis by flavonoids [FADD: Fas associated death domain].

196
R.L. Nagula, S. Wairkar Journal of Controlled Release 296 (2019) 190–201

Therefore research should be focused on improving its stability in sui- 4.5. Flavones
table formulation [80].
Flavones provide additional nutritional value to our diet. Flavones
are having anti-microbial anti-oxidant, anti-inflammatory, and anti-
4.4. Isoflavones carcinogenic activity promotes apoptosis [93]. Apigenin and luteolin
have been studied in different formulations.
Isoflavones are the naturally occurring isoflavonoids and most
commonly found in soyabeans, soy foods and legumes. Isoflavones are 4.5.1. Apigenin
non-steroidal compounds having estrogen like activity as they mimic The most important property of apigenin is inhibition of COX and
17-eatradiol structure and called as phytoestrogens [87]. Most of the inactivation of nuclear transcription factor (NF-kB), induces apoptosis,
compounds belonging to Fabaceae family contain isoflavones and the activating transcription factor 3 to assist in the reduction of in-
common isoflavones are genistein, daidzein, glycitein, formononetin. flammation. It also acts as chemo-preventive by inhibiting enzyme
They show antioxidant property by scavenging free radical, protection CYP2C9 and preventing the metabolism of many drugs and xenobiotics
against lipid peroxidation and also decreases oxidative stress caused by [94].
UV damage. Genistein has been reported to show anti-osteoporotic
activity [88]. Isoflavones being phytoestrogen and their ability for in- 4.5.1.1. Apigenin-phospholipid phytosome. Phytosomes are the
creased production of hyaluronic acid which provides moisturizing ef- phospholipid carriers which help in the delivery of the poorly soluble
fect, can be used in topical delivery. drugs. Oral administration leads to rapid metabolism of apigenin.
Apigenin-phospholipid phytosomes (APLC) were prepared and
4.4.1. Genistein and daidzein evaluated in albino rats (25 mg/kg) to estimate antioxidant enzymes.
Genistein and daidzein inhibit tyrosine kinase of the growth re- Also, hepatoprotective activity and CCl4 induced hepatotoxin
ceptor factor which reduced cell growth and proliferation [89]. They demonstrated increase in the liver marker enzymes, showing excellent
also have the ability to bind all the three-peroxisome proliferator-acti- hepatoprotection. The level of several antioxidant enzymes was
vated receptors isoforms. Isoflavones were evaluated as topical photo- elevated proving its antioxidant potential. The Cmax, Tmax, and AUC(0-
protective agent in the pig skin by Jing-Yi Lin et al. The erythema re- ∞) of the APLC were greater than that of the aqueous solution of

sponse and the sunburn cells were studied for photoprotective effect of apigenin, indicates enhanced bioavailability [95].
isoflavones. Solutions of Isoflavones (0.5%) i.e. genistein, daidzein,
equol, biochanin A, formononetin were compared to various standard 4.5.1.2. Apigenin ethosomes. Apigenin has the ability to inhibit COX
antioxidants such as α-tocopherol, ascorbic acid, ferulic acid. Out of enzyme which helps in prevention of inflammation caused by UV, X-
which, genistein, daidzein and biochanin A showed significant photo- rays. Apigenin loaded ethosomes, liposomes and deformable liposomes
protection. Genistein inhibits tyrosine receptor kinase activity and de- formulations were prepared and studied in vivo in Sprague-dawley rats
creases the UV damage. Formononetin has no effect on sunburn cells and konmin mice. In vivo skin deposition study performed on shaved
but effective for the treatment of erythema. Isoflavones act as potent abdominal skin of rats showed that there was highest deposition at 2 h
photoprotective and also the good candidate ingredient for UV damage found with ethosomes and liposome i.e. 0.176 ± 0.010 μg/cm2 and
[90]. 0.167 ± 0.047 μg/cm2 whereas deposition of deformable liposomes
was lower i.e. 0.123 ± 0.011 μg/cm2. The effect of UVB induced skin
inflammation of all formulations and plain apigenin was studied on
4.4.1.1. Effect of penetration enhancers on topical delivery of genistein and mice which showed inhibitory effect on COX-2 level and prevented
daidzein. It is known that genistein is potent inhibitor of the protein inflammation but the inhibitory effect of apigenin ethosomes was more
tyrosine kinase and possesses good topical permeability. Genistein as compared to others [96].
decreases the H2O2 level in the skin, showing radical scavenging
activity. As it belongs to polyphenolic class, they donate H-atom to 4.5.1.3. Chamomile flower extract liposomes. Chamomile flower extract
the oxygen radical and form the phenoxyl radicals which are less (CFE) rich in apigenin was extracted by the percolation. Liposomes
reactive. Daidzein contains 1 less OH group than genistein, so it was were prepared using nattermann phospholipid and apigenin mixture
assumed to have less antioxidant effect which was not true [91]. Pka of (1:2) and then incorporated into oil in water cream containing various
daidzein was 7.2, shows higher water solubility and decreased topical excipients. In vitro dissolution study showed increased release
permeability through skin. The retention time of genistein was more compared to the non-liposomal cream containing plain CFE apigenin.
than that of daidzein due to its lipophilicity. Log K′ of genistein and Clinical study on the 19 human volunteers between age 24–65 years
daidzein was 0.23 and − 0.05 respectively indicating the greater was performed and the in vivo study showed significant anti-
hydrophilicity of daidzein. inflammatory activity with apigenin cream [97]. Apigenin liposomal
Subcutaneous layer acts as the major barrier for genistein and cream was effective, tolerated well and showed much less adverse
daidzein and thus, α-terpineol and oleic acid was evaluated as pene- effects than corticosteroids.
tration enhancers by Zih-Rou Huanga. Melting point of the compounds Majority of the apigenin research include in vitro studies which
genistein, daidzein, aglycones were 305°, 338°, 238 °C but when mix- creates difficulty in drawing concrete conclusions. Therefore, more in
ture was formed the melting point reduced, showing eutectic effect. vivo studies of formulations can be performed to prove its safety and
This helped to increase skin permeability as compounds in the eutectic efficacy in various chronic and acute skin disorders.
form have high solubility subcutaneously. In vivo study in the mouse
showed the increase in the uptake of genistein with no toxic effects and 4.5.2. Luteolin
decrease in the erythema. Daidzein also showed similar effect but in- Luteolin is found in various plants such as celery, green pepper,
creased the pH of skin [92]. chamomile flower, etc. Although it has wide range of therapeutic ac-
Genistein and daidzein were not explored earlier due to their es- tivities as other flavonoids, it shows antioxidant effect by scavenging
trogenic properties but later its anti-inflammatory, anti-oxidant prop- free radicals and nitrogen species. But the most important use of lu-
erties were discovered. The advantage could be taken as these com- teolin is in treatment of arthritis due to its ability to inhibit IL, TNFα,
pounds are non-steroidal but possess estrogenic property. Currently, and necrosis factor [98,99]. Luteolin was evaluated on the psoriatic
isoflavones are being used for cancer treatment but novel formulations skin for keratinocyte activation and the level of NF-kB factor. The effect
of isoflavones are very few to be used for therapeutic benefits. of luteolin (1–100 μM) on HaCaT keratinocytes showed concentration

197
R.L. Nagula, S. Wairkar Journal of Controlled Release 296 (2019) 190–201

dependent inhibition of various mediators such as IL-6, IL-8, and VEGF showed down regulation of caspase 14 and infiltrating macrophages. It
whereas its 100 μM concentration showed complete inhibition of all also decreased the keratin-14 which causes hyperproliferation. Thus,
three mediators. Due to the lipid solubility of luteolin, it can be used for delphinidin acts and antiproliferative by inhibiting keratin-14. Appli-
topical formulation for treating psoriasis [100].Luteolin has the ability cation of delphinidin to flaky mice skin decreases the level of patho-
to inhibit the release of histamine from the mast cells which helps in logical markers of psoriasis lesions.
inhibiting inflammation and allergy. The cutaneous reaction was in-
duced in ICR mice by injecting histamine or serotonin intradermally. 4.6.1.1. Anthocyanin niosome gel. Anthocyanin niosomes were prepared
Also, the contact dermatitis was induced by applying 2,4-Dinitrohlor- and in vivo anti-inflammatory activity was evaluated on croton oil
obenzene (DNCB). The concentration of luteolin 2,20 and 100 μg/site induced inflamed ears of rats with niosomes (0.5 and 5 mg/cm2). It was
dissolved in olive oil was applied at the site of inflammation that sig- found that niosomes improved permeation rate and prolonged anti-
nificantly reduced the ear swelling and thickness by inhibiting vascular inflammatory effect. Percent ear edema inhibition of niosome
permeability, showed antagonist effect on inflammatory mediators and containing 5 mg/cm2 of anthocyanin complex was greater compared
also decreased the scratching behaviour [101]. to 0.5 mg/cm2 [108]. Thus, topical niosomes prove to be the
appropriate and potential candidate for treating inflammatory
4.5.2.1. Luteolin niosomes. Niosomes are a non-ionic surfactant-based condition.
colloidal system and have ability to encapsulate hydrophobic as well as
hydrophilic drugs. Luteolin niosomes were prepared by Lubna Abidin 4.6.1.2. Anthocyanin microemulsion. Purple sweet potato (Ipomoea
et al. and were formulated into the gel (niotransgel). The ex vivo study batatas L.) is rich in anthocyanins and possesses the ability to
of luteolin niosomes performed on the rat skin showed excellent scavenge free radicals. As skin is easily susceptible to free radicals,
permeability with transdermal flux value 93.21 ± 13.14 mg/cm2/h. topical treatment would be more preferable. Anthocyanins
In vitro antiarthritic study of niotransgel compared to diclofenac gel microemulsion was prepared consisted to evaluate antioxidant
(omnigel) showed 81.82% inhibition and the diclofenac with 85.21% property using DPHH method for plain ethanolic extract of purple
inhibition. In vivo analgesic study in rats by tail flick hot water sweet potato and microemulsion. It was found that the ethanolic extract
immersion method showed 32.6 s much greater than that of showed 50% inhibition whereas microemulsion containing extract
diclofenac gel [102]. Hence, luteolin loaded niosomes can be used as showed 80% inhibition of ROS [109].
an alternative treatment of arthritis. Delphinidin is extensively used in food industry as colorants. It has
wide range of pharmacological activities and yet, more can be learnt on
4.5.2.2. Luteolin nanoemulsion. Apart from aforementioned the therapeutic line of anthocyanins. Development of novel formula-
pharmacological actions, luteolin, baicalin and alpinetin have tions of anthocyanin is the basic need at present as it is not been much
property to induce alkaline phosphatase which plays an important explored.
role in hair growth. As flavonoids are lipophilic in nature, formulating A brief summary of pharmacological properties of flavonoids and in
them into nano-formulation will help in increasing bioavailability. vivo/cell line study is described in Table 2.
Luteolin nanoemulsion prepared and radical scavenging activity was
evaluated using DPHH method and was compared with ascorbic acid. It 5. Future perspective and conclusion
was found that ascorbic acid showed 10% inhibition at dose 250 μM
whereas luteolin nanoemulsion exhibited strong inhibitory activity in Current generation has become more vigilant and interested to-
concentration dependent manner till dose 50 μM. In vivo study for hair wards the use of natural products. Flavonoid is an important class of
growth was performed on dorsal skin of C57BL/6 mice using minoxidil phytoproducts for its therapeutic and preventive benefits. They possess
as positive control [103]. The results showed that the skin was fully an ability to decrease the biotic stress in the human body by acting as
covered with soft silky hairs, showing excellent growth promoting phytoalexins. Their combination therapy aids in providing the sy-
effect whereas minoxidil showed uneven hair growth with stiff hairs nergistic effect as well as it helps in digging the ground to discover new
due to epidermal keratinization. This shows that luteolin nanoemulsion therapeutic activities. However, oral delivery of flavonoids is the big-
proves to be more efficient for follicular delivery. gest challenge and thus the focus of formulation development has been
In depth study of structural activity relationship of luteolin is re- shifted towards alternative route of delivery like topical route. Various
quired to develop a luteolin derivative with more specific activity. novel formulations for topical delivery have been attempted to increase
Additional in vivo studies and their models should be conducted to the solubility and permeability of flavonoids to cross the skin barrier,
evaluate the inflammatory activity and use in autoimmune disease. yet there is need to overcome other limitations of topical delivery such
as inadequate residence time and sustained release profile, etc. Various
4.6. Anthocyanins methods such as iontophoresis, electroporation, magnetophoresis, ul-
trasound, radiofrequency, microneedles can also be explored for ef-
Anthocyanin are colored glycosylated, water-soluble pigments and fective topical delivery of flavonoids. Although many novel formula-
are accountable for imparting blue, red, purple colors to fruits and tions have been evaluated at laboratory, its scale up at commercial level
vegetables. Various scientific studies have proved that anthocyanins is major challenge and therefore, there is need to look at scalability of
possess antioxidant, anti-inflammatory, depigmentation properties formulation and manufacturing process.
[104,105]. A continuous growth in the research of flavonoids is envisioned,
however a quality research providing concrete safety, toxicity and ef-
4.6.1. Delphinidin ficacy data of flavonoids is essential. Studies on controlled, long dura-
Delphinidin is known to inhibit osteoclastogenesis in osteoporosis, tion clinical trials indicating important endpoints should be conducted
and has antioxidant, anti-inflammatory, antitumorigenic and anti- for determining their dosage regimen and evaluating adverse effects.
angiogenic properties. Delphinidin reduces the levels of inflammatory The limitations from various in vitro, animal and human studies should
mediators including iNOS, NO, IL-6, MCP-1, and TNF-α induced by LPS, be considered while making conclusions which assists in future re-
downregulates NF-κB pathway and MEK1/2-ERK1/2 signaling [106]. search. This can be later extrapolated to clinical conditions to reach the
Topical application of delphinidin diminishes psoriasiform lesions in actual ripening phase.
the flaky skin mouse model by inducing epidermal differentiation and Even for oral formulations of flavonoids, the other strategies can be
inhibiting inflammation [107]. Delphinidin was evaluated for psoriasis adopted to increase solubility of flavonoids including co-crystallization,
on the flaky skin mice decreased the thickness of the epidermis. Also modification with esters or formation of glycosides etc. At the same

198
R.L. Nagula, S. Wairkar Journal of Controlled Release 296 (2019) 190–201

Table 2
Pharmacological properties of flavonoids in-vivo/ cell line study.
Sr.no Formulation of flavonoid Therapeutic activity Animal/cell line Dose Ref.

Quercetin
1. Lipid nanocapsules Antioxidant, anti-inflammatory Acute monocytic leukemia cell line (THP1–1 5 μg/ml [42]
cell)
2. Nanoparticle suspension Antioxidant Mice 0.5% w/w [43]
3. Liposomes with penetration enhancing vesicles Anti-inflammatory Female CD-1 mice 5% PEG-PEV [47]
(PEV) 10%PEG-PEV

Hesperetin, hesperidin
4. Microemulsion Whitening effect Guinea pigs 2 mg/cm2 [61]
5. Topical matrix film Release of hesperetin in posterior of eye Albino rabbits 10% w/w and 20% [62]
w/w
6. Microemulsion based ointment Skin irritation Wistar rats 1 ml/g [63]

Naringenin
7. Gel Antioxidant, Anti-inflammatory HRS/J mice 0.5% [70]
8. Nanoparticles Photoprotective, antioxidant Wistar rats 2 mg/cm2 [72]

Catechins
9. Cream Wound healing Iranian rabbits 2% Grape seed extract [85]
10. Nano-transfersomes Antioxidant, antiaging effect HaCaT (human keratinocytes) cells 10 mg [81]

Apigenin
11. Apigenin phospholipid phytosome Antioxidant Albino rats 25 mg/kg [95]
12. Apigenin ethosomes Anti-inflammatory Konmin mice 0.02% w/v [96]

Luteolin
13. Luteolin in olive oil Anti-inflammatory ICR mice 2,20 and 100 μg [101]
14. Luteolin nanoemulsion Growth promoting effect C57BL/6 mice 50 μM [103]

Anthocyanin
15. Niosome gel Anti-inflammatory Male wistar rats 0.5 and 5 mg/cm2 [108]

time, suitable formulation strategies and other administration routes (2018) 1–25, https://doi.org/10.1016/j.pharmthera.2018.09.009.
can be explored as per pharmacological activity of flavonoids. [6] F. Perez-Vizcaino, C.G. Fraga, Research trends in flavonoids and health, Arch.
Biochem. Biophys. 646 (2018) 107–112, https://doi.org/10.1016/j.abb.2018.03.
In summary, flavonoids have been established as the excellent 022.
natural compounds, especially for topical delivery that aids in treating [7] S.J. Shelke, D.M. Shinkar, R.B. Saudagar, Topical gel: a novel approach for de-
various skin ailments with minimum side effects. There has been an velopment of topical drug delivery system, Int. J. Pharm. Technol. 5 (2013)
2739–2763, https://doi.org/10.1016/j.mpsur.2011.06.010.
exponential growth in research of flavonoids for several other disorders [8] F. Menaa, A. Menaa, B. Menaa, Polyphenols Nano-formulations for Topical,
such as cardiovascular, neurological, diabetes and cancer which de- Elsevier Inc, 2014, https://doi.org/10.1016/B978-0-12-398456-2.00065-7.
monstrates deviance from synthetic drugs to healthier regimen of [9] Eric Perrier, et al., Flavonoide Esters and their Use Notably in Cosmetics,
US6471973B2 (2002).
dietary supplements and nutraceuticals. Topical route is the best tar-
[10] L. Kurppa, Flavonoid Drug and Dosage Form, its Production and Use
geted route for flavonoids due to its lipophilic nature. At present a wide (WO2001049285A1), (2001).
range of novel drug delivery are available which show promising results [11] H. buchholz Thomas rudolph, Flavonoids as Synergists for Enhancing the Action of
Self-tanning Substances, US8613910B2 (2013).
for topical delivery. Though preclinical results are promising, sub-
[12] K. Plaschke, Composition Comprising One or More Flavonoids, Method of
stantial clinical trials need to be conducted for translation of research to Obtaining Such Composition and Use Thereof as UV-absorbing Agent,
commercial products. US6409996B1 (2002).
[13] S.H. Gottlieb, Topical Composition Containing Specific Flavanones, EP 0 774 249
A2, (1996).
Funding [14] Thomas Christian Lines, Quercetin-containing Compositions, US 8,440,704 B2
(2013).
This research did not receive any specific grant from funding [15] M.-Q.M. Peter, M. Elias, Hesperidin-containing Compositions and Methods for
Treatment of Skin Disorders, US 2017/0020798 A1, (2017).
agencies in the public, commercial, or not-for-profit sectors. [16] N.R. Broscheit, Delphinidin for Combating Melanoma Cells, WO 2014/090586 AI,
(2014).
Declaration of interest [17] M. das Balaram gosh, Method of Treating and/or Preventing Asthama Using
Natural Compound Luteolin, US20040191327A1, (2004).
[18] D.L. Simmons, Topical Liposome Compositions Containing Phenolic Anti-in-
None. flammatory Agents and Their Methods of Preparation, US 2015/0342843 A1,
(2015).
[19] Y. Santosh k katiyar Hara, Compositions and Methods for Reduction of Cutaneous
References Photoageing, US 2010/0069476A1, (2010).
[20] Philip J. Birbara, Methods of Making and Using Compositions Comprising
[1] D. Procházková, I. Boušová, N. Wilhelmová, Antioxidant and prooxidant proper- Flavonids, US 2012/0213842 A1, (2013).
ties of flavonoids, Fitoterapia 82 (2011) 513–523, https://doi.org/10.1016/j. [21] J.S. Barbieri, K. Wanat, J. Seykora, Skin: Basic Structure and Function, Elsevier
fitote.2011.01.018. Inc., 2014, https://doi.org/10.1016/B978-0-12-386456-7.03501-2.
[2] A.D.D. and S.R.C. A. N. Panche, Flavonoids: an overview, J. Nutr. Sci. 5 (2016) [22] B.M. Carlson, Skin, Hum. Body, (2019), pp. 65–85, https://doi.org/10.1016/
1–15, https://doi.org/10.1017/jns.2016.41. B978-0-12-804254-0.00003-X.
[3] B. Yang, H. Liu, J. Yang, V.K. Gupta, Y. Jiang, New insights on bioactivities and [23] J. Marks, G. J, J. Miller, 2 – Structure and Function of the Skin, Lookingbill Marks.
biosynthesis of flavonoid glycosides, Trends Food Sci. Technol. 79 (2018) Princ. Dermatology, (2019), pp. 2–10, https://doi.org/10.1016/B978-0-323-
116–124, https://doi.org/10.1016/j.tifs.2018.07.006. 43040-1.00002-6.
[4] T. yang Wang, Q. Li, K. shun Bi, Bioactive flavonoids in medicinal plants: struc- [24] K.W. Ng, W.M. Lau, Skin deep: the basics of human skin structure and drug pe-
ture, activity and biological fate, Asian J. Pharm. Sci. 13 (2017) 12–23, https:// netration, Percut. Penet. Enhanc. Chem. Methods Penetration Enhanc. Drug
doi.org/10.1016/j.ajps.2017.08.004. Manip. Strateg. Veh. Eff. (2015) 3–11, https://doi.org/10.1007/978-3-662-45013-
[5] K.R.R. Rengasamy, H. Khan, S. Gowrishankar, R.J.L. Lagoa, F.M. Mahomoodally, 0_1.
Z. Khan, S. Suroowan, D. Tewari, G. Zengin, S.T.S. Hassan, S.K. Pandian, The role [25] S.A. Souyoul, K.P. Saussy, M.P. Lupo, Nutraceuticals: a review, Dermatol. Ther.
of flavonoids in autoimmune diseases: therapeutic updates, Pharmacol. Ther. (Heidelb) 8 (2018) 5–16, https://doi.org/10.1007/s13555-018-0221-x.

199
R.L. Nagula, S. Wairkar Journal of Controlled Release 296 (2019) 190–201

[26] N.M. Zaki, Progress and problems in nutraceuticals delivery, J. Bioequiv. [51] S.H. Kim, J.G. Park, J. Lee, W.S. Yang, G.W. Park, H.G. Kim, Y.S. Yi, K.S. Baek,
Bioavailab. 6 (2014) 075–077, https://doi.org/10.4172/jbb.1000183. N.Y. Sung, M.J. Hossen, M.N. Lee, J.H. Kim, J.Y. Cho, The dietary flavonoid
[27] S. Saraf Ajazuddin, Applications of novel drug delivery system for herbal for- kaempferol mediates anti-inflammatory responses via the src, syk, IRAK1, and
mulations, Fitoterapia 81 (2010) 680–689, https://doi.org/10.1016/j.fitote.2010. IRAK4 molecular targets, Mediat. Inflamm. 2015 (2015), https://doi.org/10.
05.001. 1155/2015/904142.
[28] K. Rehman, M.H. Zulfakar, Recent advances in gel technologies for topical and [52] A.Y. Chen, Y.C. Chen, A Review of the Dietary Flavonoid, Kaempferol on Human
transdermal drug delivery, Drug Dev. Ind. Pharm. 40 (2014) 433–440, https://doi. Health and Cancer Chemoprevention, Vol. 138 (2014), pp. 2099–2107, https://
org/10.3109/03639045.2013.828219. doi.org/10.1016/j.foodchem.2012.11.139.A.
[29] B. Ghanbarzadeh, A. Babazadeh, H. Hamishehkar, Nano-phytosome as a potential [53] K.M. Lee, K.W. Lee, S.K. Jung, E.J. Lee, Y. Heo, A.M. Bode, R.A. Lubet, H.J. Lee,
food-grade delivery system, Food Biosci. 15 (2016) 126–135, https://doi.org/10. Z. Dong, Suppressing Src Kinase Activity, Vol. 80 (2011), pp. 2042–2049, https://
1016/j.fbio.2016.07.006. doi.org/10.1016/j.bcp.2010.06.042.
[30] A. Semalty, M. Semalty, M.S.M. Rawat, F. Franceschi, Supramolecular phospho- [54] X. Yi, J. Zuo, C. Tan, S. Xian, C. Luo, S. Chen, L. Yu, Y. Luo, Kaempferol, a fla-
lipids-polyphenolics interactions: the PHYTOSOME®strategy to improve the vonoid compound from gynura medica induced apoptosis and growth inhibition in
bioavailability of phytochemicals, Fitoterapia 81 (2010) 306–314, https://doi. MCF-7 breast cancer cell, African J. Tradit. Complement. Altern. Med. 13 (2016)
org/10.1016/j.fitote.2009.11.001. 210–215, https://doi.org/10.21010/ajtcam.v13i4.27.
[31] S. Saraf, J. Khan, A. Alexander, S. Saraf Ajazuddin, Recent advances and future [55] Y. Chao, C. Huang, L. Fu, Y. Huang, Y. Tsai, P. Wu, The Effect of Submicron
prospects of phyto-phospholipid complexation technique for improving pharma- Emulsion Systems on Transdermal Delivery of Kaempferol, Vol. 60 (2012), pp.
cokinetic profile of plant actives, J. Control. Release 168 (2013) 50–60, https:// 1171–1175.
doi.org/10.1016/j.jconrel.2013.02.025. [56] L. Shi, X.E. Feng, J.R. Cui, L.H. Fang, G.H. Du, Q.S. Li, Synthesis and biological
[32] N. Karimi, B. Ghanbarzadeh, H. Hamishehkar, A. Pezeshki, Phytosome as Novel activity of flavanone derivatives, Bioorganic Med. Chem. Lett. 20 (2010)
Delivery System for Nutraceutical Materials, Vol. 4 (2015), pp. 152–159. 5466–5468, https://doi.org/10.1016/j.bmcl.2010.07.090.
[33] A. Alexander, R.J. Ajazuddin, S. Patel, S. Saraf, Recent expansion of pharmaceu- [57] W.Y. Liu, S.S. Liou, T.Y. Hong, I.M. Liu, Protective effects of hesperidin (Citrus
tical nanotechnologies and targeting strategies in the field of phytopharmaceu- flavonone) on high glucose induced oxidative stress and apoptosis in a cellular
ticals for the delivery of herbal extracts and bioactives, J. Control. Release 241 model for diabetic retinopathy, Nutrients 9 (2017) 1–15, https://doi.org/10.
(2016) 110–124, https://doi.org/10.1016/j.jconrel.2016.09.017. 3390/nu9121312.
[34] K. Zhang, M. Zhang, Z. Liu, Y. Zhang, L. Gu, G. Hu, X. Chen, J. Jia, Development of [58] B. Hemanth Kumar, B. Dinesh Kumar, P.V. Diwan, Hesperidin, a citrus flavonoid,
quercetin-phospholipid complex to improve the bioavailability and protection protects against l-methionine-induced hyperhomocysteinemia by abrogation of
effects against carbon tetrachloride-induced hepatotoxicity in SD rats, Fitoterapia oxidative stress, endothelial dysfunction and neurotoxicity in wistar rats, Pharm.
113 (2016) 102–109, https://doi.org/10.1016/j.fitote.2016.07.008. Biol. 55 (2017) 146–155, https://doi.org/10.1080/13880209.2016.1231695.
[35] A.A. Date, N. Desai, R. Dixit, M. Nagarsenker, Self-nanoemulsifying drug delivery [59] D. Stanisic, A.F. Costa, G. Cruz, N. Durán, L. Tasic, Applications of Flavonoids,
systems: formulation insights, applications and advances, Nanomedicine 5 (2010) With an Emphasis on Hesperidin, as Anticancer Prodrugs: Phytotherapy as an
1595–1616, https://doi.org/10.2217/nnm.10.126. Alternative to Chemotherapy, (2018), https://doi.org/10.1016/B978-0-444-
[36] P.F. Kador, Topical applied nutraceutical antioxidant formulation reduces ocular 64056-7.00006-4.
oxidative stress, Funct. Foods Heal. Dis. 7 (2017) 17–35. [60] J. Cho, Antioxidant and neuroprotective effects of hesperidin and its aglycone
[37] H. Wang, K. Helliwell, Determination of Favonols in Green and Black Tea Leaves hesperetin, Arch. Pharm. Res. 29 (2006) 699–706, https://doi.org/10.1007/
and Green Tea Infusions by High-performance Liquid Chromatography, 34 (2001), BF02968255.
pp. 223–227, https://doi.org/10.1016/S0963-9969(00)00156-3. [61] Y. Tsai, K. Lee, Y. Huang, C. Huang, In Vitro Permeation and In Vivo Whitening
[38] A. Gupta, K. Birhman, I. Raheja, S.K. Sharma, H.K. Kar, Quercetin: a wonder Effect of Topical Hesperetin Microemulsion Delivery System, Vol. 388 (2010), pp.
bioflavonoid with therapeutic potential in disease management, Asian Pacific J. 257–262, https://doi.org/10.1016/j.ijpharm.2009.12.051.
Trop. Dis. 6 (2016) 248–252, https://doi.org/10.1016/S2222-1808(15)61024-6. [62] G.R. Adelli, T. Hingorani, N. Punyamurthula, S. Prachetan, S. Majumdar,
[39] G.D. Andrea, Fitoterapia quercetin : a flavonol with multifaceted therapeutic ap- European Journal of Pharmaceutics and Biopharmaceutics Evaluation of topical
plications? Fitoterapia 106 (2015) 256–271, https://doi.org/10.1016/j.fitote. hesperetin matrix film for back-of-the-eye delivery, Eur. J. Pharm. Biopharm. 92
2015.09.018. (2015) 74–82, https://doi.org/10.1016/j.ejpb.2015.02.006.
[40] M. Lesjak, I. Beara, N. Simin, D. Pintać, T. Majkić, K. Bekvalac, D. Orčić, [63] V. Kilor, N. Sapkal, G. Vaidya, Design and development of novel microemulsion
N. Mimica-Dukić, Antioxidant and anti-inflammatory activities of quercetin and its based topical formulation of hesperidin, Int J Pharm Pharm Sci 7 (2015), https://
derivatives, J. Funct. Foods 40 (2018) 68–75, https://doi.org/10.1016/j.jff.2017. doi.org/10.1208/s12249-009-9242-1.
10.047. [64] W. Sangpheak, J. Kicuntod, R. Schuster, T. Rungrotmongkol, P. Wolschann,
[41] E.J. Park, J.Y. Kim, M.S. Jeong, K.Y. Park, K.H. Park, M.W. Lee, S.S. Joo, S.J. Seo, N. Kungwan, H. Viernstein, M. Mueller, P. Pongsawasdi, Physical properties and
Effect of topical application of quercetin-3-O-(2″-gallate)-α-l-rhamnopyranoside biological activities of hesperetin and naringenin in complex with methylated p-
on atopic dermatitis in NC/Nga mice, J. Dermatol. Sci. 77 (2015) 166–172, cyclodextrin, Beilstein J. Org. Chem. 11 (2015) 2763–2773, https://doi.org/10.
https://doi.org/10.1016/j.jdermsci.2014.12.005. 3762/bjoc.11.297.
[42] T. Hatahet, M. Morille, A. Shamseddin, A. Aubert-pouëssel, J.M. Devoisselle, À. C, [65] Z. Xu, B. Huang, J. Liu, X. Wu, N. Luo, X. Wang, X. Zheng, X. Pan, Combinatorial
Dermal quercetin lipid nanocapsules : in fl uence of the formulation on antioxidant anti-proliferative effects of tamoxifen and naringenin: the role of four estrogen
activity and cellular protection against hydrogen peroxide, Int. J. Pharm. 518 receptor subtypes, Toxicology (2018), https://doi.org/10.1016/j.tox.2018.08.
(2017) 167–176, https://doi.org/10.1016/j.ijpharm.2016.12.043. 013.
[43] Q. Tan, W. Liu, Preparation and evaluation of quercetin-loaded lecithin-chitosan [66] M.F. Manchope, R. Casagrande, W.A. Verri Jr., M.F. Manchope, R. Casagrande,
nanoparticles for topical delivery, Int. J. Nanomedicine (2015) 1621–1630, J.W.A. Verri, Naringenin: an analgesic and anti-inflammatory citrus flavanone,
https://doi.org/10.2147/IJN.S22411. Oncotarget 8 (2017) 3766–3767, https://doi.org/10.18632/oncotarget.14084.
[44] R. Censi, V. Martena, E. Hoti, L. Malaj, P. Di Martino, Permeation and Skin [67] M.A. Alam, N. Subhan, M.M. Rahman, S.J. Uddin, H.M. Reza, S.D. Sarker, Effect of
Retention of Quercetin from Microemulsions Containing Transcutol ® P, Vol. 38 citrus flavonoids, naringin and naringenin, on metabolic syndrome and their
(2012), pp. 1128–1133, https://doi.org/10.3109/03639045.2011.641564. mechanisms of action, Adv. Nutr. An Int. Rev. J. 5 (2014) 404–417, https://doi.
[45] S. Bose, B. Michniak-kohn, European Journal of Pharmaceutical Sciences org/10.3945/an.113.005603.
Preparation and characterization of lipid based nanosystems for topical delivery of [68] V.R. P, S. Kiran, P. Rohini, P. Bhagyasree, Flavonoid : a review on Naringenin, J.
quercetin, Eur. J. Pharm. Sci. 48 (2013) 442–452, https://doi.org/10.1016/j.ejps. Pharmacogn. Phytochem. 6 (2017) 2778–2783.
2012.12.005. [69] M. Tsai, Y. Huang, J. Fang, Y. Fu, P. Wu, Preparation and evaluation of submicron-
[46] M. Chessa, C. Caddeo, D. Valenti, M. Manconi, C. Sinico, Effect of Penetration carriers for naringenin topical application, Int. J. Pharm. 481 (2015) 84–90,
Enhancer Containing Vesicles on the Percutaneous Delivery of Quercetin through https://doi.org/10.1016/j.ijpharm.2015.01.034.
New Born Pig Skin, (2011), pp. 497–509, https://doi.org/10.3390/ [70] R.M. Martinez, F.A. Pinho-Ribeiro, V.S. Steffen, T.C.C. Silva, C.V. Caviglione,
pharmaceutics3030497. C. Bottura, M.J.V. Fonseca, F.T.M.C. Vicentini, J.A. Vignoli, M.M. Baracat,
[47] M.M. Carla Caddeoa, C. Octavio Díez-Salesb, Ramon Ponsd, F.G. Xavier S.R. Georgetti, W.A. Verri, R. Casagrande, Topical formulation containing nar-
Fernàndez-Busquetse, A. Anna Maria Faddaa, Topical anti-inflammatory potential ingenin: efficacy against ultraviolet B irradiation-induced skin inflammation and
of quercetin in lipid-based nanosystems: in vivo and in vitro evaluation Carla oxidative stress in mice, PLoS One 11 (2016) 1–21, https://doi.org/10.1371/
Caddeo, Pharm. Res. 31 (2014) 958–968, https://doi.org/10.1007/s11095-013- journal.pone.0146296.
1215-0. [71] M. Tsai, Y. Huang, J. Fang, Y. Fu, P. Wu, Preparation and Characterization of
[48] S.H. Kim, K.C. Choi, Anti-cancer effect and underlying mechanism(s) of Naringenin-loaded Elastic Liposomes for Topical Application, (2015), pp. 1–12,
Kaempferol, a phytoestrogen, on the regulation of apoptosis in diverse cancer cell https://doi.org/10.1371/journal.pone.0131026.
models, Toxicol. Res. 29 (2013) 229–234, https://doi.org/10.5487/TR.2013.29.4. [72] Haritima Joshi, Papr reduction using scs-slm technique in stfbc mimo-ofdm, ARPN
229. J. Eng. Appl. Sci. 12 (2017) 3218–3221, https://doi.org/10.1111/ijlh.12426.
[49] H. Li, L. Yang, Y. Zhang, Z. Gao, Kaempferol inhibits fibroblast collagen synthesis, [73] R. Joshi, Y.A. Kulkarni, S. Wairkar, Pharmacokinetic, pharmacodynamic and
proliferation and activation in hypertrophic scar via targeting TGF-β receptor type formulations aspects of Naringenin: an update, Life Sci. 215 (2018) 43–56,
I, Biomed. Pharmacother. 83 (2016) 967–974, https://doi.org/10.1016/j.biopha. https://doi.org/10.1016/j.lfs.2018.10.066.
2016.08.011. [74] M. Grzesik, G. Bartosz, A. Dziedzic, D. Narog, J. Namiesnik, I. Sadowska-Bartosz,
[50] J. Wang, X. Fang, L. Ge, F. Cao, L. Zhao, Z. Wang, W. Xiao, Antitumor, antioxidant Antioxidant properties of ferrous flavanol mixtures, Food Chem. 268 (2018)
and anti-inflammatory activities of kaempferol and its corresponding glycosides 567–576, https://doi.org/10.1016/j.foodchem.2018.06.076.
and the enzymatic preparation of kaempferol, PLoS One 13 (2018) 1–12, https:// [75] D.G. Nagle, D. Ferreira, Y.D. Zhou, Epigallocatechin-3-gallate (EGCG): chemical
doi.org/10.1371/journal.pone.0197563. and biomedical perspectives, Phytochem. 67 (2006) 1849–1855 https://doi.org/

200
R.L. Nagula, S. Wairkar Journal of Controlled Release 296 (2019) 190–201

10.1016/j.phytochem.2006.06.020. Delivery and Potential Dermal Use of Soy Isoflavones Genistein and Daidzein, Vol.
[76] Y.H. Lin, M.J. Tsai, Y.P. Fang, Y.S. Fu, Y. Bin Huang, P.C. Wu, Microemulsion 364 (2008), pp. 36–44, https://doi.org/10.1016/j.ijpharm.2008.08.002.
formulation design and evaluation for hydrophobic compound: catechin topical [93] N. Jiang, A. Doseff, E. Grotewold, Flavones: from biosynthesis to health benefits,
application, Colloids Surf. B Biointerf. 161 (2018) 121–128, https://doi.org/10. Plan. Theory 5 (2016) 27, https://doi.org/10.3390/plants5020027.
1016/j.colsurfb.2017.10.015. [94] X. Zhou, F. Wang, R. Zhou, X. Song, M. Xie, Apigenin: a current review on its
[77] D. Chen, S.B. Wan, H. Yang, J. Yuan, T.H. Chan, Q.P. Dou, EGCG, green tea beneficial biological activities, J. Food Biochem. 41 (2017), https://doi.org/10.
polyphenols and their synthetic analogs and prodrugs for human cancer preven- 1111/jfbc.12376.
tion and treatment, Adv. Clin. Chem. 53 (2011) 155–177, https://doi.org/10. [95] D.R. Telange, A.T. Patin, Formulation and Characterization of an Apigenin-
1016/B978-0-12-385855-9.00007-2. phospholipid Phytosome (APLC) for Improved Solubility, In Vivo Bioavailability,
[78] M. Grzesik, K. Naparło, G. Bartosz, I. Sadowska-Bartosz, Antioxidant properties of and Antioxidant Potential, (2016), pp. 36–49, https://doi.org/10.1016/j.ejps.
catechins: comparison with other antioxidants, Food Chem. 241 (2018) 480–492, 2016.12.009.
https://doi.org/10.1016/j.foodchem.2017.08.117. [96] L.N. Shen, Y.T. Zhang, Q. Wang, L. Xu, N.P. Feng, Enhanced in vitro and in vivo
[79] R.M. Brand, J.L. Jendrzejewski, Topical treatment with (−)-epigallocatechin-3- skin deposition of apigenin delivered using ethosomes, Int. J. Pharm. 460 (2014)
gallate and genistein after a single UV exposure can reduce skin damage, J. 280–288, https://doi.org/10.1016/j.ijpharm.2013.11.017.
Dermatol. Sci. 50 (2008) 69–72, https://doi.org/10.1016/j.jdermsci.2007.11.008. [97] I. Arsić, V. Tadić, D. Vlaović, I. Homåek, S. Vesić, G. Isailović, G. Vuleta,
[80] P.V. Gadkari, M. Balaraman, Catechins: Sources, Extraction and Encapsulation: A Preparation of novel apigenin-enriched, liposomal and non-liposomal, antiin-
Review, Institution of Chemical Engineers, 2015, https://doi.org/10.1016/j.fbp. flammatory topical formulations as substitutes for corticosteroid therapy, Phyther.
2013.12.004. Res. 25 (2011) 228–233, https://doi.org/10.1002/ptr.3245.
[81] K.S. Avadhani, J. Manikkath, M. Tiwari, A. Godavarthi, S.M. Vidya, C. Raghu, [98] N. Aziz, M.Y. Kim, J.Y. Cho, Anti-inflammatory effects of luteolin: a review of in
G. Kalthur, N. Udupa, K.S. Avadhani, J. Manikkath, M. Tiwari, M. Chandrasekhar, vitro, in vivo, and in silico studies, J. Ethnopharmacol. 225 (2018) 342–358,
A. Godavarthi, Skin Delivery of Epigallocatechin-3-gallate (EGCG) and Hyaluronic https://doi.org/10.1016/j.jep.2018.05.019.
Acid Loaded Nano-transfersomes for Antioxidant and Anti-aging Effects in UV [99] G. Odontuya, J.R.S. Hoult, P.J. Houghton, Structure-activity relationship for an-
Radiation Induced Skin Damage Loaded Nano-transfersomes for Antioxidant and tiinflammatory effect of luteolin and its derived glycosides, Phyther. Res. 19
Anti-aging Effects, 7544, (2017), https://doi.org/10.1080/10717544.2016. (2005) 782–786, https://doi.org/10.1002/ptr.1723.
1228718. [100] Z. Weng, A.B. Patel, M. Vasiadi, A. Therianou, T.C. Theoharides, Luteolin Inhibits
[82] H. Zhang, S. Liu, L. Li, S. Liu, S. Liu, J. Mi, G. Tian, The impact of grape seed Human Keratinocyte Activation and Decreases NF- k B Induction that Is Increased
extract treatment on blood pressure changes: a meta-analysis of 16 randomized in Psoriatic Skin, Vol. 9 (2014), pp. 1–8, https://doi.org/10.1371/journal.pone.
controlled trials, Medicine (Baltimore) 95 (2016) e4247, , https://doi.org/10. 0090739.
1097/MD.0000000000004247. [101] L. Baolin, W. Weiwei, T. Ning, Topical application of luteolin inhibits scratching
[83] B.N. Singh, S. Shankar, R.K. Srivastava, Green tea catechin, epigallocatechin-3- behavior associated with allergic cutaneous reaction in mice, Planta Med. 71
gallate (EGCG): mechanisms, perspectives and clinical applications, Biochem. (2005) 424–428, https://doi.org/10.1055/s-2005-864137.
Pharmacol. 82 (2011) 1807–1821, https://doi.org/10.1016/j.bcp.2011.07.093. [102] L. Abidin, M. Mujeeb, S.S. Imam, M. Aqil, Enhanced Transdermal Delivery of
[84] P.D. Taylor, P.W. Hamilton-Miller, J.M.T. Stapleton, Antimicrobial properties of Luteolin Via Non-ionic Surfactant-based Vesicle : Quality Evaluation and Anti-ar-
green tea catechins, Food Sci. Technol. Bull. 2 (1985) 332, https://doi.org/10. thritic Assessment Surfactant-based Vesicle : Quality Evaluation and Anti-arthritic,
1616/1476-2137.14184. vol. 7544, (2016), https://doi.org/10.3109/10717544.2014.945130.
[85] A.A. Hemmati, N. Aghel, A. Gholampur-aghdami, Topical Grape (Vitis vinifera) [103] K. Shin, H. Choi, S.K. Song, J.W. Yu, J.Y. Lee, E.J. Choi, D.H. Lee, S.H. Do,
Seed Extract Promotes Repair of Full Thickness Wound in Rabbit, 8 (2011), pp. J.W. Kim, Nanoemulsion vehicles as carriers for follicular delivery of luteolin, ACS
514–520, https://doi.org/10.1111/j.1742-481X.2011.00833.x. Biomater. Sci. Eng. 4 (2018) 1723–1729, https://doi.org/10.1021/
[86] C. Agarwal, R.P. Singh, R. Agarwal, Grape seed extract induces apoptotic death of acsbiomaterials.8b00220.
human prostate carcinoma DU145 cells via caspases activation accompanied by [104] H.E. Khoo, Anthocyanidins and anthocyanins : colored pigments as food, phar-
dissipation of mitochondrial membrane potential and cytochrome c release, maceutical ingredients, and the potential health benefits, Food Nutr. Res. 61
Carcinogenesis 23 (2002) 1869–1876. (2017), https://doi.org/10.1080/16546628.2017.1361779.
[87] J. Yu, X. Bi, B. Yu, D. Chen, Isoflavones: anti-inflammatory benefit and possible [105] C.S. Bowen-Forbes, Y. Zhang, M.G. Nair, Anthocyanin content, antioxidant, anti-
caveats, Nutrients 8 (2016) 1–16, https://doi.org/10.3390/nu8060361. inflammatory and anticancer properties of blackberry and raspberry fruits, J. Food
[88] A. Accorsi-Neto, M. Haidar, R. Simões, M. Simões, J. Soares-Jr, E. Baracat, Effects Compos. Anal. 23 (2010) 554–560, https://doi.org/10.1016/j.jfca.2009.08.012.
of isoflavones on the skin of postmenopausal women: a pilot study, Clinics 64 [106] A. Sobiepanek, M. Milner-Krawczyk, K. Bobecka-Wesołowska, T. Kobiela, The
(2009) 505–510, https://doi.org/10.1590/S1807-59322009000600004. effect of delphinidin on the mechanical properties of keratinocytes exposed to UVB
[89] N. Irrera, G. Pizzino, R. D'Anna, M. Vaccaro, V. Arcoraci, F. Squadrito, D. Altavilla, radiation, J. Photochem. Photobiol. B Biol. 164 (2016) 264–270, https://doi.org/
A. Bitto, Dietary management of skin health: the role of genistein, Nutrients 9 10.1016/j.jphotobiol.2016.09.038.
(2017) 1–10, https://doi.org/10.3390/nu9060622. [107] H.C. Pal, J.C. Chamcheu, V.M. Adhami, G.S. Wood, C.A. Elmets, H. Mukhtar,
[90] J. Lin, J.A. Tournas, J.A. Burch, N.A. Monteiro-riviere, J. Zielinski, Topical F. Afaq, Topical application of delphinidin reduces psoriasiform lesions in the
Isoflavones Provide Effective Photoprotection to Skin, (2008), pp. 61–66, https:// flaky skin mouse model by inducing epidermal differentiation and inhibiting in-
doi.org/10.1111/j.1600-0781.2008.00329.x. flammation, Br J Dermatol 172 (2015) 354–364 https://doi.org/10.1111/bjd.
[91] Y. Zhang, S. Hendrick, Bioavailability and Biological Effects of the Isoflavone 13513.
Glycitein and Isoflavone Glucuronides: Role of Glucuronide in Human Natural [108] A. Priprem, S. Limsitthichaikoon, S. Thappasarapong, A.A. Complex,
Killer Cell Modulation In Vitro, 9990506 (2000), pp. 17–18 http://search. A.C. Preparation, Anti-inflammatory Activity of Topical Anthocyanins by
proquest.com/docview/304604582?accountid=13042%5Cnhttp://oxfordsfx. Complexation and Niosomal Encapsulation, 9 (2015), pp. 133–137.
hosted.exlibrisgroup.com/oxford?url_ver=Z39.88-2004&rft_val_fmt=info:ofi/ [109] R. Desnita, S. Wahdaningsih, S. Hervianti, Topical microemulsion's formulation of
fmt:kev:mtx:dissertation&genre=dissertations+&+theses&sid=ProQ:ProQuest purple sweet potato (Ipomoea batatas L.) ethanol extract as antioxidant by using
+Dissertations+&+Theses+Global. various concentration of span 80, Int. J. PharmTech Res. 9 (2016) 198–203.
[92] Z. Huang, C. Hung, Y. Lin, J. Fang, In Vitro and In Vivo Evaluation of Topical

201

You might also like