TSBAS2 Bahan Ujian

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

Seminars in Cancer Biology


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

Review

Dietary flavonoids: Nano delivery and nanoparticles for cancer therapy


Paola Aielloa,b,c, Sara Consalvid, Giovanna Poced, Anna Raguzzinia, Elisabetta Totia,
Maura Palmeryb, Mariangela Biavad, Marco Bernardib, Mohammad A. Kamale,f,g,
George Perryh, , Ilaria Pelusoa,
⁎⁎ ⁎

a
Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), Rome, Italy
b
Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Italy
c
Universidad Católica San Antonio de Murcia (UCAM), Murcia, Spain
d
Department of Chemistry and Drug Technologies, University “La Sapienza”, Rome, Italy
e
King Fahd Medical Research Center, King Abdulaziz University, Saudi Arabia
f
Enzymoics, 7 Peterlee Place, Hebersham, NSW, 2770, Australia
g
Novel Global Community Educational Foundation, Australia
h
Department of Biology, University of Texas at San Antonio, TX, USA

ARTICLE INFO ABSTRACT

Keywords: Application of nanotechnologies to cancer therapy might increase solubility and/or bioavailability of bioactive
Epigallocatechin gallate compounds of natural or synthetic origin and offers other potential benefits in cancer therapy, including se-
Quercetin lective targeting. In the present review we aim to evaluate in vivo studies on the anticancer activity of nano-
Flavonoids particles (NPs) obtained from food-derived flavonoids. From a systematic search a total of 60 studies were
Cancer
identified. Most of the studies involved the flavanol epigallocatechin-3-O-gallate and the flavonol quercetin, in
Nanoparticle
both delivery and co-delivery (with anti-cancer drugs) systems. Moreover, some studies investigated the effects
of other flavonoids, such as anthocyanins aglycones anthocyanidins, flavanones, flavones and isoflavonoids. NPs
inhibited tumor growth in both xenograft and chemical-induced animal models of cancerogenesis.
Encapsulation improved bioavailability and/or reduced toxicity of both flavonoids and/or co-delivered drugs,
such as doxorubicin, docetaxel, paclitaxel, honokiol and vincristine. Moreover, flavonoids have been successfully
applied in molecular targeted nanosystems. Selectivity for cancer cells involves pH- and/or reactive oxygen

Abbreviations: 2-AAF, 2-acetylaminofluorene; 67LR, laminin receptor 67 kDa; ALP, alkaline aminotransferate; ALT, alanine aminotransferate; API, apigenin; AST,
aspartate aminotransferate; ATC, anthocyanin; AUC, area under the curve; BaP, Benzo[a]pyrene; BCL, baicalein; BR, biotin receptor; BSA, bovine serum albumin;
BW, body weight; CAT, catalase; CD44R, CD44 receptor; CH, chitin; CHOL, cholesterol; CIS, cisplatin; Cmax, maximum plasma concentration; CMS, colloidal
mesoporous silica; COX-2, cyclooxygenase-2; CS, chitosan; CSU, chondroitin sulfate; CTX, cyclophosphamide; Cy, cyanidin; DEN, diethyl nitrosamine; DMBA, 12-
dimethyl(a)benz anthracene; DOX, doxorubicin; Dp, delphinidin; DSPE-MPEG, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-methoxy(polyethylene glycol);
EAC, Ehrlich ascites carcinoma; EGCG, epigallocatechin-3-O-gallate; EGFR, epidermal growth factor receptor; EIF5A2, oncogene eukaryotic translation initiation
factor 5A2; ER, estrogen receptor; ERK, extracellular signal-regulated kinase; Eudragit, aminoalkyl methacrylate copolymer E; FA, folic acid; FITC, fluorescein
isothiocyanate; FR, folate receptors; FU, fucose; GEN, genistein; GPNLC, GeluPearl stabilized by Gelucire; GPSLN, GeluPearl with Precirol ATO 5 stabilized by
Gelucire; GSH, glutatione; GST, glutatione-S-transferase; HA, hyaluronic acid; Hb, haemoglobin; HES, hesperetin; IC50, half inibitory concentration; IFN, interferon;
IL, interleukin; IR, integrin receptor; KpK, kaempferol; MDA, malondialdeide; MDR, multidrug resistance; MNU, N-methyl-Nitrosurea; MPEG–PCL, monomethoxy
poly (ethylene glycol)–poly(ε-caprolactone); MPEG-PLA, methoxy poly (ethylene glycol)-poly(lactide); M-PLGA–TPGS, copolymer mannitol-functionalized Poly
(lactic-co-glycolic acid); MPO, myeloperoxidase; MPP, matrix metalloproteinases; MSN, mesoporous silica nanoparticles; Mv, malvidin; NF-κB, nuclear factor-κB;
NPs, nanoparticles; Nrf2, nuclear transcription factor-erythroid 2-related factor 2; PAA, poly (acrylic acid); PBS, phosphate buffered saline; PC, phosphatidylcholine;
PCL, poly(ε-caprolactone); PCNA, proliferating cell nuclear antigen; Pe, peonidin; PE, phosphatidylethanolamine; PEG, polyethylene glycol; PEG-DSPE, poly(ethy-
lene glycol)–distearoylphosphatidylethanolamine; PEG-PCL, poly(ethylene glycol)-poly(-caprolactone); Pg, pelargonidin; PLA, polylactic acid; PLGA, poly(lactic co-
glycolic acid); PMSN, arginine-glycine-aspartic acid peptide-MNS; pPhe-pHis-PEG, poly(phenylalanine)-b-poly(L-histidine) -b- poly(ethylene glycol); PS, particle
size; PSA, prostate specific antigen; PSMA, prostate-specific membrane antigen; Pt, petunidin; PTX, paclitaxel; PVA, poly-vinyl alcohol; QCT, quercetin; ROS, reactive
oxygen species; SA, stearic acid; SCID, severe combined immunodeficient; SNEDDS, self-nano-emulsifying drug delivery system; SOD, superoxide dismutase; SPC,
soybean phosphatidylcholine; Tf, transferrin; TfR, transferrin receptor; Tmax, time to reach peak concentration; TMX, tamoxifen; TNF, tumor necrosis factor; TPGS,
d-α-tocopheryl polyethylene glycol 1000 succinate; TPP, triphenylphosphine; TPs, tea polyphenols; TPT, topotecan; UVB, ultraviolet-B; VEGF, vascular endothelial
growth factor

Corresponding author at: Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), via Ardeatina 546, Rome, Italy.
⁎⁎
Corresponding author.
E-mail addresses: george.perry@utsa.edu (G. Perry), ilaria.peluso@crea.gov.it (I. Peluso).

https://doi.org/10.1016/j.semcancer.2019.08.029
Received 9 July 2019; Received in revised form 8 August 2019; Accepted 22 August 2019
1044-579X/ © 2019 Elsevier Ltd. All rights reserved.

Please cite this article as: Paola Aiello, et al., Seminars in Cancer Biology, https://doi.org/10.1016/j.semcancer.2019.08.029
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

species-mediated mechanisms. Furthermore, flavonoids are good candidates as drug delivery for anticancer
drugs in green synthesis systems. In conclusion, although human studies are needed, NPs obtained from food-
derived flavonoids have promising anticancer effects in vivo.

1. Introduction polyphenols (i.e.curcumin, resveratrol) were excluded. Studies that met


the following criteria were included: studies appeared in an edited
The main dietary sources of flavonoids, bioactive polyphenols of journal (peer-review criterion), written in English (language criterion),
natural origin, are cocoa products (1589–3410 mg/100 g) and green tea focused on the anticancer effect of flavonoid nanoparticles (topic cri-
(7.93–27.16 mg/100 ml) for flavanols, blackberries (595–1316 mg/ terion) and measured tumor growth in vivo (method criterion). Studies
100 g) for anthocyanins, soy products (148–466 mg/100 g) for iso- were included when they met the above criteria regardless of dose and
flavonoids, spinach and onions (119–158 mg/100 g) for flavonols, black treatment duration and/or type of animal model of cancerogenesis
olives and artichokes (27–58 mg/100 g) for flavones, orange and (xenograft or chemical-induced). Through these criteria, a total of 60
grapefruit juices (38–51 mg/100 ml) for flavanone [1,2]. However, studies were identified (Tables 1–5). Most of the studies involved EGCG
several studies reported differences in foods consumed in different (Table 1) [40–46] and QCT (Table 2) [47–66], but some surveys in-
countries. In general, while tea [3–9] and fruits [4,8–11] are the most vestigated the effects of other flavonoids, such as anthocyanins, agly-
important food sources of flavonoids, beer [8,9], cocoa products [5], cones, anthocyanidins, flavanones, flavones and isoflavonoids (Table 3)
and beans [12] are within the major food sources in the US, Poland and [67–76]. Furthermore, some studies investigated the effects of flavo-
Brazil, respectively. Moreover, onions and apples are the main dietary noids as drug delivery and in co-delivery formulation (Table 4)
sources of flavonoids in Finland [7] and in The Netherlands [3], [77–83], as well as in molecular targeted nanosystems (Table 5)
whereas in the Mediterranean countries the most important source of [84–99]. Studies on bioavailability and in vitro evidence were main-
polyphenols are olives and olive oil [11]. In Italy, within the main tained for discussion.
dietary sources of flavonoids there are various vegetables, such as ar-
tichokes and olives (flavones), spinach and beans (flavonols), and 3. Flavanols
cherries (anthocyanins), as well as citrus fruits (flavanones) and red
wine (flavanols) [13]. Encapsulation improved both stability and cellular uptake and/or
Preclinical studies reported that flavonoids, such as the flavanol cytotoxic effect of EGCG [42,46,100–102] and cocoa procyanidins [103].
epigallocatechin-3-gallate (EGCG), the flavonol quercetin (QCT), the The effect of EGCG on tumor growth was investigated in vivo in studies
flavone apigenin (API), the flavanone hesperetin (HES) and the iso- that used chitosan (CS) [40,41], fucose (FU)-CS-polyethylene glycol
flavone genistein (GEN) exerted anticancer activity by inhibiting cell (PEG)-CS [42], gold [43–45] and polylactic acid (PLA)-PEG [46] NPs
cycle and/or by inducing apoptosis [14–21]. Moreover QCT and GEN (Table 1). EGCG NPs showed lower half maximal inhibitory concentra-
interact with androgen receptors and estrogen receptor β, respectively tion (IC50) on cell viability/proliferation in vitro [43,46,102,104–106]
[14,20], and others suggested mechanisms for flavonoid anticancer and induced high tumor growth inhibition in xenograft cancer models
activities involving epidermal growth factor receptor (EGFR), vascular compared to pure EGCG (Table 1). There was a dose-dependent inhibi-
endothelial growth factor (VEGF), nuclear factor-κB (NF-κB) and matrix tion of tumor growth by EGCG NPs [46] and the growth-suppressive
metalloproteinases (MPP) [14–20,22,23]. Although an inverse asso- effect of EGCG NPs was found to be more effective in the group viain-
ciation between dietary flavonoid intake and certain cancers (upper tratumor (i.t.) or intraperitoneally (i.p.) than that orally (p.o.) [44]
aero-digestive tract, colorectal, breast, and lung cancers) has been re- (Table 1). It has been reported, by using excised mouse jejunum in
ported from case-control studies, further prospective cohorts assessing chambers, that encapsulation significantly enhanced intestinal absorp-
dietary polyphenol metabolism are needed to confirm health benefits tion [107]. Mass balance studies conducted during the experiment,
[24–26]. Despite the potential activity, pharmacokinetic studies re- measuring the disappearance of catechins in the donor chamber, in-
vealed low bioavailability and consequent controversial efficacy, re- dicated that enhancement of intestinal absorption most likely occurs as a
sulting from these agents being phytochemicals substrates of both phase result of the stabilization of catechins in the donor chamber, which
I and phase II detoxification enzymes and phase III transporters, as well subsequently drives their flux across the tissue [107]. Administration of
as of the microbial flora in the colon [22,23,27–29]. Estimated max- the CS EGCG NPs increased the concentrations of EGCG in the jejunum
imum plasma concentration (Cmax) and estimated time to reach this (2.3-fold increase) of mice and enhanced the plasma exposure of total
peak concentration (Tmax) for flavonoids and their metabolites are EGCG by a factor of 1.5 relative to an EGCG solution (plasma AUC 179.3
very variable within different flavonoids classes. Cmax is about 10−3M and 116.4 nM/h, respectively) [108]. Despite the reported hepatotoxicity
for flavanones (Tmax range: 4.0–5.0 h) and anthocyanins (Tmax of EGCG in humans [109–113], no toxicity was observed in vivo in terms
range: < 1–1.5 h), between 10-5–10−3M for flavanols (Tmax range: of differences in body weight (BW) gain, liver, kidney, spleen, and
1.6–2.6 h) and isoflavones (Tmax range: 1.6–17.1 h) and between 10-5- uterine weight between control animals and treated groups [41,44], as
10-4M for flavonols (Tmax range: < 1-5.4 h) [29]. In order to improve well as in terms of renal and hepatic toxicities (serum levels of creatinine,
bioavailability and reduce toxicity and multidrug resistance (MDR), blood urea nitrogen, bilirubin, alkaline phosphatase: ALP, alanine ami-
research is focusing, as previously reviewed, on nanodelivery systems notransferase: ALT, and aspartate aminotransferase: AST) [45]. In Ehr-
[14,18,19,22,23,27,30–39]. In the present review we aim to evaluate in lich ascites carcinoma (EAC), biochemical parameters in blood, such as
vivo studies on the anticancer activity of nanoparticles (NPs) obtained level of haemoglobin (Hb), glucose, cholesterol, triglycerides, urea, ALP,
from food-derived flavonoids. ALT, AST, bilirubin and total protein improved, compared to controls, in
mice treated with and without tea polyphenols (TPs) NPs (10 mg/kg).
2. Study selection These mice were injected before tumor induction and after 2 days re-
ceived intraperitoneally EAC cells [114]. Lipid peroxidation was reduced
We performed a systematic search on the Medline database for lit- in EAC bearing mice administered (pre- and post-treated) with TPs NPs,
erature examining the effects of flavonoid nanoparticles on cancer in whereas it was high in EAC bearing control mice and EAC bearing mice
vivo until May 2019. The search terms were: flavonoid AND nano- treated with unloaded NPs. The activities of the antioxidant enzymes
particle AND cancer. The aim of the present review focused on the food- catalase (CAT) and superoxide dismutase (SOD) in EAC bearing mice
derived flavonoids, studies on herbal extracts, silymarin or other were significantly reduced and ameliorated by the oral administration of

2
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Table 1
Anticancer effect of EGCG nanoparticles in animal models.
Formulation Animal model Treatment and Tumor growth Ref.
(Mean PS) administration route

CS Xenograft: Control (Chit-nano) 25-60 days [40]


(EGCG NPs) Athymic nude mice, human prostate carcinoma 22Rv1 EGCG (40 mg/) EGCG ↓
(200-207 nm) cells EGCG NPs (3 mg/kg), EGCG NPs (3) ↓↓
EGCG NPs (6 mg/kg) EGCG NPs (6) ↓↓↓
Orally (treatment started one day post inoculation, 5/ week)
CS Xenograft: Control 27-32 days [41]
(EGCG NPs) Athymic nude mice, human melanoma Mel 98 cells EGCG (1 mg/mice), EGCG 1 mg ↓↓
(PS n.r.) EGCG (100 μg/mice) EGCG 100 μg ↓
EGCG NPs (100 μg/mice) EGCG NPs ↓↓↓
Orally (treatment started one day post inoculation, 5/ week)
FU-CS-PEG-CS Xenograft: Control (blank NPs) 16 days [42]
(EGCG NPs) Athymic nude mice, gastric adenocarcinoma Luc MKN45 EGCG (40 mg/kg) EGCG ↓
(250-540 nm) cells EGCG NPs (40 mg/kg) EGCG NPs ↓↓
Orally (treatment started 14 days post inoculation, once daily).
Gold Xenograft: Control (PBS), 28 days [43]
(EGCG NPs) C57/BL6 mice, murine melanoma B16F10 cells EGCG (2 mg /mouse) EGCG ↓
(64.7 nm) EGCG NPs (2 mg/mouse) EGCG NPs ↓
Intratumorally (treatment started on day 14 post inoculation, 2/
week)
Gold Xenograft: Control (water) 29 days [44]
(EGCG NPs) C3H/HeN mice, murine bladder tumor MBT-2 cells EGCG (2 mg /mouse) EGCG (TR p.o.) ↔
(20-1200 nm) EGCG NPs (2 mg /mouse) EGCG (PR p.o.) ↓
Prevention (PR): orally EGCG NPs (PR p.o.) ↓↓
Treatment (TR): orally (p.o.), ↓
intraperitoneally (i.p.) EGCG NPs (TR p.o.) ↓
or intratumor (i.t.). EGCG NPs (TR i.p.) ↓↓
PR: once daily every other day for 7 days before inoculation. EGCG NPs (TR i.t.) ↓↓
TR: every other day on day 14 post inoculation.
Gold Xenograft: Control 29 days [45]
(EGCG NPs) C3H/He mice, MBT-2 murine bladder tumor cells EGCG (2 mg /mouse) EGCG (p.o.) ↓
(50 nm) EGCG NPs (2 mg /mouse) EGCG NPs (p.o.) ↓↓
Orally (o.p.) or intraperitoneally (i.p) EGCG NPs (p.o.) ↓↓↓
(treatment started 14 days post inoculation).
LA-PEG Xenograft: Control (no treatment) Weeks 5-7 [46]
(EGCG NPs) Athymic nude mice, 22Rv1 cells (human prostate EGCG (1 mg/mice) EGCG ↓
(285 nm) carcinoma) EGCG NPs (100 mg/mice) EGCG NPs ↓
(treatment started after inoculation, 3/week)

CS: Chitosan; EGCG: Epigallocatechin-3-O-gallate; FU: Fucose; NPs: nanoparticles; PEG: Polyethylene glycol: PLA: Polylactic acid; PS: particle size (mean).

TPs NPs [114]. In this context, oral administration of TPs NPs for 3 days treated with EGCG NPs compared with free EGCG [41,45,46,102].
prior to irradiation exposure has been shown to protect mice from he- Furthermore, EGCG NPs showed to inhibit angiogenesis [45,46].
matological injuries that result in the reduction of radiation-induced Khan et al. [40] observed a 5.7- to 8.2-fold expression of CD31 and 4.1-
lethality and of radiation-induced oxidative damage and apoptosis, by to 5.9-fold VEGF-positive cells in control group than in EGCG NPS
restoring the redox status through the nuclear transcription factor-ery- treated groups. Similar decreased VEGF levels after treatment with
throid 2-related factor 2 (Nrf2)-extracellular signal-regulated kinase EGCG NPs were found by Hsieh et al. [44,45] and inhibition of neo-
(ERK) pathway and reducing Bax expression, respectively [115]. angiogenesis have been observed also in zebrafish xenotransplants for
On the other hand, there was a significant inhibition in serum catechin NPs (50 μg/ml) [101].
prostate-specific antigen (PSA, marker of disease progression) in EGCG Gold EGCG NPs also showed immunomodulating activity in tumor-
treated mice. The EGCG-NPs treatment led to an additional decrease in bearing mice, decreasing the expression levels of interleukin-4 (IL-4)
the levels of serum PSA [40,46]. High efficacy and lower toxicity in and IL-6 significantly and increasing the expression levels of IL-2 and
cancerogenesis in vivo models can be due to NPs biodistribution com- interferon-γ (IFN-γ) [44]. These results suggested that the anti-tumor
pared to non-conjugates. effect of EGCG plus pNG viaoral administration could be correlated to
In vivo tumor accumulation and clearance of green tea [116] and the cell-mediated immunity. To verify this hypothesis, authors ex-
EGCG [117] NPs have been reported. Studies presented in Table 1 re- amined the cytotoxic activity of natural killer (NK)-cells in tumor-
port inhibition of proliferation in tumor tissues, by measuring Ki-67, bearing mice and found an increased NK cell-activity in the group
proliferating cell nuclear antigen (PCNA) and CDK 4 and 6 [40–42]. subjected to early intervention of EGCG plus pNG [44]. Moreover, a
Fucose-carboxymethyl chitosan graft EGCG with gold NPs possible involvement of Th1 (by virtue of IFN-γ production) and Th17
(30–70 nm) displayed cancer weakening as suggested by the lowly cells (viaproduction of IL-17A) in cancer immunosurveillance has been
nuclear thickness of cancers in mice undergoing pre-cancerous vacci- suggested [44].
nation with gastric tumor cells (MKN45) [117].
Moreover, in tumor tissues isolated from EGCG NPs treated mice a 4. Flavonols
shift in the Bax/Bcl-2 ratio in favor of apoptosis [41,45] and an increase
in the activation of caspases-3, -8 and -9 were observed [40,45]. Ac- Although studies reported both the anti-cancer effect in vitro of
cordingly, enhanced apoptosis and/or inhibition of proliferation, by morin NPs [118] and kaempferol (KPF) NPs [119,120] and the brain
modulations of Bcl-2/Bax, cyclin dependent kinase inhibitors WAF1/ delivery of intranasal administration of KPF NPs [119], among flavo-
p21 and CIP1/p27, cyclins and caspases, were observed in cancer cells nols only QCT NPs have been assessed for tumor growth inhibition

3
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Table 2
Anticancer effect of quercetin (QCT) nanoparticles in animal models.
Formulation Animal model Treatment and Tumor growth Ref.
administration route

Gold DMBA-induced mammary carcinoma in Sprague-Dawley Control (NPs) 8 days [47]


(QCT NPs) rats DMBA QCT ↓
(5.2 nm) QCT (25 mg/kg); QCT NPs ↓↓
QCT NPs (25 mg/kg)
Intratumoural (treatment started after 60 days, 8
days)
n.r. DEN-induced hepatocarcinogenesis in rats Control (medium/NPs) 18 weeks [48]
(QCT NPs) QCT Preneoplastic lesions
(40-100 nm) QCT NPs (8.98 μmol/kg) QCT ↔
Intravenously (once a week for 16 weeks) QCT NPs ↓
PLGA- rutin (RT quercetin-3- DEN-induced hepatocarcinogenesis in rats Control (olive oil) 22 weeks [49]
O-rutinoside) RT (5 mg/kg) RT (5) ↓
(RT NPs) RT (100 mg/kg) RT (100) ↓↓
(211 nm) RT NPs (5 mg/kg) RT NPs ↓↓↓
Orally (daily for 22 weeks)
PLGA DEN-induced hepatocarcinoma in rats Control (olive oil/NPs) 18 weeks [50]
(QCT NPs) DEN (200 mg/kg) QCT ↔
(270 nm) QCT (8.98 mmol/l) QCT NPs ↓
QCT NPs (8.98 mmol/l)
Orally (weekly for 16 weeks)
PLGA DEN-induced hepatocarcinoma in rats Control (untreated/NPs) 18 weeks [51]
(QCT NPs) DEN Preneoplastic
(270 nm) QCT (8.98 mmol/l) lesions:
QCT NPs (8.98 mmol/l) QCT ↔
Orally (weekly for 16 weeks) QCT NPs ↓
DSPE-MPEG Xenograft: Control (saline) 40 days [52]
(QCT NPs) Balb/c nude mice, human prostate cancer cells PC-3. QCT (30 mg/kg) QCT ↓
(13.21 nm) QCT NPs (30 mg/kg). QCT NPs ↓↓
Intravenously (treatment started when the tumor
volume reached 150 mm3, at days 23, 26, 29, 32, 35
and 38)
CS Xenograft: Control (PBS) Weeks 4 [53]
(QCT NPs) Immunocompromised C57BL6 mice, A549 and MDA MB QCT (25 mg/kg) QCT ↓
(100-200 nm) 468 cells QCT NPs (25 mg/kg) QCT NPs ↓↓
Intravenously (treatment started when the tumor
volume reached 80-100mm3, twice in a week)
Freeze-dried Xenograft: Control (saline) 28 days [54]
(FD QCT NPs) Mice, C6 glioma cells QCT (25 mg/kg) QCT ↓
(56.5 nm, QCT NPs QCT NPs (25 mg/kg) QCT NPs ↓↓
104.6 nm) FD QCT NPs (25 mg/kg) FD QCT NPs ↓↓
Orally (treatment started when tumors reached
40–100 mm3, every 7 days)
QCT GPSLN Xenograft: Control (No treatment) 14-22 days [55]
QCTGPNLC C57BL/6 mice, 105 B16F10 melanoma cells QCT (50 mg/Kg) QCT ↓
(400 nm) QCT GPSLN (50 mg/Kg) QCT GPSLN ↓↓↓
QCT GPNLC (50 mg/Kg) QCT GPNLC ↓↓
Orally (treatment started after tumor induction, days
1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21)
Gold –PLA Xenograft: Inoculated cells treated with/without 5 weeks [56]
(QCT NPs) BALB/c nu/nu nude mice, Caski cells QCT NPs Low (10 mg/ml) QCT NPs Low ↓
(PS n.r.) QCT NPs High (20 mg/ml) QCT NPs High ↓↓
Gold –PLA Xenograft: Control (PBS) 5 weeks [57]
(QCT NPs) BALB/c nu/nu nude mice, MHCC97H cells QCT NPs (30 mg/kg) QCT NPs (30) ↓
(106.7 nm) QCT NPs (40 mg/kg) QCT NPs (40) ↓↓
QCT NPs (50 mg/kg) QCT NPs (50) ↓↓↓
Intra-peritoneally (treatment started when the tumor
reached 3 mm, every day)
Gold –PLA Xenograft: Control (PBS) 5 weeks [58]
(QCT NPs) BALB/c-nude mice, U87 cells QCT NPs Low (40 mg/kg) QCT NPs Low ↓
(106.7 nm) QCT NPs High (80 mg/kg) QCT NPs High ↓↓
Intraperitoneally (treatment started after two weeks,
when the tumor reached to 3 mm, every day)
MPEG-PLA Xenograft: Control (saline/NPs) 22 days [59]
(QCT NPs) BALB/c mice, T1 cancer cells QCT (4 mg/kg) QCT ↓
(155.3 nm) QCT NPs (0.5 mg/kg) QCT NPs (0.5) ↓
QCT NPs (4 mg/kg) QCT NPs (4) ↓↓
Peritumoral (treatment started on day 10 when
tumors reached 100–200 mm3, every 3rd day till day
19)
MPEG–PCL Xenograft: Control (saline/NPs) 22 days [60]
(QCT NPs) BALB/c mice, CT26 colon carcinoma cells. QCT (50 mg/kg) QCT ↓
(34.8 nm) QCT NPs (50 mg/kg) QCT NPs ↓↓
(continued on next page)

4
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Table 2 (continued)

Formulation Animal model Treatment and Tumor growth Ref.


administration route

Intravenously (treatment started when the tumor


reached 6 mm, once daily for nine consecutive days).
PLGA and Xenograft: Control (medium/NPs) 10 days [61]
TPGS Kunming mice, HCa-F cells QCT (10 mg/kg) QCT ↔
(QCT NPs: QNPs (10 mg/kg) QPNs ↓
QPNs and QPTNs) QPTNs (10 mg/kg) Intravenously (treatment for 10 QPTNs ↓↓
(100-200 nm) consecutive days, once a day)
PEG-TPP Xenograft: Control 14 days [62]
(QCT NPs) Mice, H22 cells QCT QCT ↔
(80-100 nm) QCT NPs QCT NPs ↓
SPC-CHOL Xenograft: Control (PBS/NPs) 16 days [63]
(QCT NPs) Mice, U14 cervical cancer ascites QCT (2 mg/kg) QCT ↓
(143.1 nm) QCT NPs (2 mg/kg) QCT NPs ↓↓
Intravenously (treatment started 24 h after
inoculation, 15 days)
PEG Xenograft: Control (PBS/NPs) 17 days [64]
(QCT NPs) C57BL/6 mice, B16-F10 melanoma cells QCT (5 mg/ kg) QCT ↓
(20 nm) QCT NPs (5 mg/ kg) QCT NPs ↓↓
Orally (treatment started 7 days after tumor
induction, twice/week)
PEG-PE Xenograft: Control (untreated) 10 weeks [65]
(QCT NPs) Rag-2 M mice, human lung A549 cells. QCT (30 mg/kg) QCT ↔
(15.4-18.5 nm) QCT NPs QCT NPs↓
Orally (treatment started when tumor volume
reached 80 mm3, 3 times/week, 3 weeks)
PEG-liposomal Xenograft: Control (saline/NPs) 31 days [66]
(QCT NPs) BALB/c athymic nude mice, cisplatin-sensitive (A2780s) or QCT (50 mg/kg) Both tumor models
(163 nm) cisplatin-resistant (A2780cp) human ovarian cancer cells QCT NPs (50 mg/kg) QCT ↓
Intravenously (treatment started when tumor volume QCT NPs ↓↓
reached 100 mm3, every three-day for 27 days)

CHOL cholesterol; CS: Chitosan; DEN: diethyl nitrosamine; DMBA: 12-dimethyl(a)benz anthracene; DSPE-MPEG: 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-
N-methoxy (polyethylene glycol); GPNLC: GeluPearl stabilized by Gelucire; GPSLN: GeluPearl with Precirol ATO 5 stabilized by Gelucire; MPEG–PCL Monomethoxy
poly (ethylene glycol)–poly(ε-caprolactone); MPEG-PLA: Methoxy poly (ethylene glycol)-poly(lactide); NPs: nanoparticles; PE: phosphatidylethanolamine; PEG:
Polyethylene glycol; PLA Polylactic acid; PLGA: Poly(lactic co-glycolic acid); PS: particle size (mean); SPC: soybean phosphatidylcholine; TPGS: d-α-tocopheryl
polyethylene glycol 1000 succinate; TPP: triphenylphosphine.

(Table 2). The in vivo anticancer effect of QCT NPs has been in- pathways and QUE NPs, at high concentrations, induced necrotic cell
vestigated in 7,12-dimethyl(a)benz anthracene (DMBA)-induced death [122,123].
(n = 1) and diethyl nitrosamine (DEN)-induced (n = 4), of which one Among molecular mechanisms, reductions in the protein expression
with the glycosylated form of QCT: rutin (RT) quercetin-3-O-rutinoside) of MMPs, p-EGFR and VEGFR-2 have been reported in tumor bearing
carcinogenesis, as well as in xenograft models (Table 2). animals [47,48]. Moreover, QCT NPs inhibited angiogenesis in vivo
RT NPs, through oral route, reduced tumor growth and exhibited [60,66] and new blood vessel formation in chick embryos [47], more
significant improvement in BW, hematologic (white blood cells (WBC); efficiently compared with free QCT.
red blood cells (RBC) and Hb) and hepatic/non-hepatic (AST, ALP, ALT, Regardless the formulation and the animal model, NPs improved
total protein, albumin and bilirubin) parameters. Moreover, RT NPS in- QCT efficacy in reducing tumor growth (Table 2). In agreement, in vitro
hibited the DEN-induced increases in serum α-fetoprotein and alterations results showed significantly higher cytotoxicity and/or cellular uptake
of Phase I enzymes, such as NADPH-cytochrome P450 reductase, NADH- of QCT NPs compared to free QCT [52,53,61,63,121,124–128]. Com-
cytochrome-b5-reductase, cytochrome P450 and cytochrome-b5 in rats pared to the control group the tumor growth was significantly inhibited
[49]. Furthermore, higher activity was observed in regulating oxidative in vivo, after treatment, by around 15% with free QCT, by 66% with
stress (malondialdehyde (MDA); glutathione (GSH); GSH-S-transferase QCT NPs and by 59% with the positive control drug cisplatin (CIS) at a
(GST), CAT, SOD, myeloperoxidase (MPO) and protein carbonyls) and dose of 1 mg/kg [64]. Similarly, by using cyclophosphamide (CTX,
inflammatory markers (IL-1β, tumor necrosis factor (TNF)-α, IL-6 and 10 mg/kg) as the positive control, CTX, free QCT and QCT NPs sig-
NF-κB) [49]. Similarly, QCT NPs improved inflammatory (TNF-α, IL-6, nificantly inhibited tumor growth and the inhibition rates were
cyclooxygenase-2 (COX-2) and NF-κB) [48,57] and oxidative stress 65.99%, 23.03% and 54.06%, respectively [63].QCT NPs showed the
markers (CAT, SOD, GST, GSH, protein carbonyls, conjugated diene, li- highest tumor suppression of about 65.7% compared with control (0%),
pohydroperoxides and Reactive Oxygen Species (ROS) production) free QCT (11.7%) and positive control doxorubicin (DOX) (54.2%)
[48,50,53] in vivo; and significantly inhibited hydrogen peroxide-in- [62]. The BW increase rate of the QCT NPs group was similar to the
duced DNA damage in HepG2 cells [121]. Immunochemistry in tumor control group, while a slower rate was visualized in the DOX group; and
bearing animals revealed decreases in proliferation (assessed by PCNA, blood biochemistry assessment confirmed the safety of QCT NPs [62].
KI-67 and Cyclin-D1) [52,56,58,60,66] and increases in apoptosis Other studies reported that QCT NPs were well tolerated by the
(evaluated by apoptotic index, Bcl-2, Bax, Bad, and caspases) animals, as reflected by changes in the BW of animals, altered behavior,
[52,54,56–58,66]. The suppression of cancer-induced JAK2 activation histological pathologic changes in the heart, liver, spleen, lung, kidney,
has been suggested for QCT NPs, having the JAK2 inhibitor similar anti- and/or intestine, as well as by biochemical markers of liver (AST, ALT)
cancer effects in vitro and in vivo [56]. However, in vitro evidence sug- and/or renal (urea and creatinine) impairment [52,53,57,60,63–66].
gested that there are several components such as ROS, mitochondrial, Pharmacokinetics profile of QCT NPs revealed higher AUC in
Bcl-2 family and JAK2/STAT3 shared by the necrotic and apoptotic plasma and/or tumor tissue, compared to free QCT [59–61]. Besides, in

5
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Table 3
Anticancer effect of Anthocyanidins, Flavanones, Flavones and Isoflavonoids in animal models.
Flavonoid Formulation Animal model Treatment and administration route Tumor growth Ref.
(mean PS)

Anthocyanidins (Antho, Exosomal: Xenograft: Control (50 mg Exo protein/kg) 5 weeks [67]
extract of (ExoAnthos) Female athymic nude mice, Anthos (10 mg/kg) Anthos ↓
bilberry): (83 nm) human lung A549 cells. ExoAnthos (5 mg Anthos) ExoAnthos ↓
Dp, Cy, Mv, Pe and Pt ratio: Orally (treatment started when tumor volume Weeks 6-7
33:28:16:16:7 reached 80 mm3, 3 times/ week) ExoAnthos ↓
Flavanone Soluthin- Xenograft: Control (saline solution/ Physical mixture: PM) 30 days [68]
Naringenin maltodextrin BALB/c mice, colon-26 cells. NAR (40 mg/kg) NAR ↓
(NAR) (NAR NPs) NAR NPs (40 mg/kg) PM ↓↓
(< 250 nm) Orally (treatment started when the tumor was˜8 NAR NPs ↓↓↓
mm3, every day)
Flavanone Eudragit E100 Xenograft: Control (saline solution) 30 days [69]
Naringenin (NAR NPs) BALB/c mice, colon-26 cells. NAR (40 mg/kg) NAR ↓
(NAR) (430 nm) physical mixture (PM) PM ↓↓
NAR NPs (40 mg/kg) NAR NPs ↓↓↓
Orally (treatment started when the tumor was˜8
mm3, every day)
Flavanone PVA- DMBA-induced oral Control (liquid paraffin) 14 weeks [70]
Naringenin Eudragit carcinogenesis in hamsters NAR (50 mg/kg) NAR ↓
(NAR) (NAR NPs) NAR NPs NAR NPs ↓↓↓
(90 nm) Orally (treatment started 1 week before exposure
to the carcinogen, days alternate to DMBA)
Flavanone Eudragit DMBA-induced oral Control (liquid paraffin) 14 weeks [71]
Naringenin (NAR NPs) carcinogenesis in hamsters NAR (50 mg/kg) NAR ↓↓
(NAR) (90 nm) NAR NPs NAR NPs ↓↓↓↓
Orally (treatment started 1 week before exposure
to the carcinogen, days alternate to DMBA)
Flavone PLA-PEG Xenograft: Control (PBS/PEG) 19 days [72]
Luteolin (LUT NPs) Athymic nude mice, Tu212 cells LUT (3.3 mg/kg): LUT↔
(LUT) (115 nm) LUT NPs LUT NPs ↓
Intraperitoneally (treatment started 7 days before
cells injection and alternate day for 30 days)
Flavone FITC labelled DEN and 2-AAF-induced hepato- API (20 mg/kg) API ↔ [73]
Apigenin (API NPs) carcinogenesis in rats API NPs (20 mg/kg) API NPs ↓
(API) (270 nm) Intravenously (single dose/week for 12 weeks)
Flavone PLGA BaP and UVB induced skin cancer Control (untreated) 90 days [74]
Apigenin (API NPs) in mice topical API NPs (5 lg/mouse), Topical ↓
(API) (101.3 nm) oral API NPs (6 mg/kg) Oral ↓
API NPs topical and oral topical + oral ↓↓
topical (2 lg/mouse) + oral (3 mg/kg). Dose response
topical (5 lg/mouse) +oral (6 mg/kg)
topical (8 lg/mouse) + oral (9 mg/kg)
Isoflavonoid GEN PCL Xenograft: Control (saline) 16 days [75]
Genistein (225.8 nm) BALB/c nude mice, HeLa cells pristine-GEN (50 mg/kg) pristine-GEN ↓
(GEN) GEN TPGS-b-PCL GEN PCL NPs (50 mg/kg) GEN PCL ↓↓
NPs GEN TPGS-b-PCL NPs (50 mg/kg) GEN TPGS-bPCL NPs ↓
(181.8 nm) Intraperitoneally (treatment started when tumor ↓↓
was touchable, every 2 days)
Isoflavonoid M-PLGA–TPGS Xenograft: Control (saline) 12 days [76]
Genistein (GEN NPs) Female SCID mice, liver pristine-GEN (50 mg/kg) pristine-GEN ↓
(GEN) (225.7 nm) carcinoma HepG2 cells GEN NPs (50 mg/kg) GEN NPs ↓
Intratumoral (treatment started when tumor
volume reached 50 mm3, 0, 4 and 8 days)

2-Acetylaminofluorene (2-AAF); BaP: Benzo[a]pyrene; Cy: cyanidin; Dp: delphinidin; Pt: petunidin; Pe: peonidin; Pg: pelargonidin; Mv: malvidin; DEN: diethyl
nitrosamine; DMBA: 12-dimethyl(a)benz anthracene; Eudragit: Aminoalkyl Methacrylate Copolymer E; Exosomal: isolated from bovine milk; FITC: Fluorescein
isothiocyanate; MPEG-PCL: monomethoxy poly(ethylene glycol)-poly(-caprolactone); M-PLGA–TPGS: copolymer mannitol-functionalized Poly (lactic-co-glycolic
acid) - d-α-tocopheryl polyethylene glycol 1000 succinate; PC: phosphatidylcholine; PCL: poly(ε-caprolactone); PVA: poly-vinyl alcohol; SCID: severe combined
immunodeficient; UVB: ultraviolet-B.

vivo biodistribution experiments revealed that NPs prolonged the re- 5. Other flavonoids
tention of drug in the whole body, reduced the accumulation in liver
and spleen, and promoted the accumulation in tumor as compared to Table 3 summarizes the in vivo studies on anticancer effect of NPs
vital organs [52,53]. These results can account for the high anti-cancer containing anthocyanidins (anthocyanin aglycones) from bilberry
efficacy in the absence of toxicity. Moreover, QCT NPs reduced the (Antho), flavanone naringenin (NAR), flavones API and luteolin (LUT)
toxicity of DEN [48,50], DMBA [47], evaluated by measuring changes and isoflavone GEN. While both Antho and ExoAnthos (by oral gavage)
in BW [47], serum ALP, AST, ALT, urea, and/or creatinine [48,50]. QCT reduced tumor growth [67], NPs of NAR [68,69], LUT [72] and GEN
in soluble form reduced the oxidative stress mediated (evaluated by [75,76] decreased the tumor growth more than flavonoid solutions.
measuring GSH and MDA) in kidney tissues) nephrotoxicity (increases Higher inhibition of tumor growth were found for flavonoids NPs
in serum urea, uric acid and creatinine) caused by gold NPs [129] and compared to free compounds were also observed on DMBA-induced
QCT NPs reduced Ultraviolet B (UVB)-induced skin damage and in- oral carcinogenesis in hamsters (NAR [70,71]), on Benzo[a]pyrene
flammation, inhibiting NF-κB/COX-2 signaling pathway [130,131]. (BaP) and ultraviolet-B (UVB)-induced skin cancer of mice (API [74])

6
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Table 4
Flavonoids in delivery and co-delivery systems of anticancer drugs.
Drug Flavonoid - Animal model Treatment and Tumor growth Ref.
Formulation Administration route
(mean PS)

Doxorubicin ATC Xenograft: Control (no treatment) 12 days: [77]


(DOX) DOX-delivery Balb/c nude mice, HCT-116 cells. DOX NPs (1 mg) ATC-DOX NPs↓↓
CSU ATC (1.5 mg)–DOX NPs DOX NPs↓
(ATC–DOX NPs) Intravenously (tumor volumes reached 100 mm3
(174.8 nm) before treatment, once)
Doxorubicin QCT Xenograft: DOX 21 days [78]
(DOX) Co-delivery Mice, SCC-7 cells. QCT DOX ↓↓
pPhe-pHis-PEG DOX/QCT QCT ↓
(DOX/QCT NPs) DOX-NPs DOX/QCT ↓↓↓
(82 nm) DOX/QCT NPs (5 mg/kg) DOX-NPs ↓↓↓
Intravenously (treatment started when tumor DOX/QCT NPs ↓↓↓↓
volumes reached 50 mm3, days 1, 4, and 7)
Docetaxel EGCG Xenograft: Control (untreated) 14 days [79]
(DTX) DTX Delivery Male hairless mice (HRS/J), A- DTX (30 mg/kg, 0.3%) DTX ↓
EGCG-SPC 375 human melanoma cells EGCG (0.2%) + DTX (0.3%) EGCG + DTX ↓↓
(DTX/EGCG NPs) NPs NPs ↔
(72.4 nm) DTX NPs (0.3%) DTX NPs ↓↓
EGCG NPs (0.2%) EGCG NPs ↔
DTX/EGCG NPs DTX/EGCG NPs ↓↓↓
Topically (treatment started at 14 days when tumor
volume reached 203 mm3, every 3 d)
Paclitaxel BCL Xenograft: 5 groups: day 30: [80]
(PTX) Co-delivery: Female Balb/C nude mice, MCF-7/ Control (saline) PTX ↔
PTX-CHOL and BCL-phospholipid Tax PTX (10 mg/kg), PTX/BCL ↔
(PTX/BCL NPs) PTX/BCL PTX NPs ↓
(170 nm) PTX NPs, PTX/BCL NPs ↓↓
PTX/BCL NPs
Intravenously (treatment started when tumor volume
reached 80–100 mm3, 0, 4, 8 and 12)
Paclitaxel GEN Xenograft: Control (nanoparticles) 8 days [81]
(PTX) Co-delivery Ehrlich Ascites Tumor (EAT) in mice G12P0: GEN (12 mg/kg) G0P0.2 ↓
PLGA NPs coated with a lipid G0P10: PTX (10 mg/kg) G12P0.2 ↓↓
bilayer containing GEN G0P2: PTX (2 mg/kg) G12P2 and G0P2 ↓↓↓
(150 nm) G0P0.2: PTX (0.2 mg/kg)
G12P10: GEN + PTX 12 + 10
G12P2: GEN + PTX 12 + 2
G12P0.2: GEN + PTX 12 + 0.2
Intraperitoneal (treatment started at day 1, 7/every
day)
Honokiol EGCG Xenograft: Control (2% glucose solution) 21 days [82]
(HK) HK delivery Male BALB/c Nude mice, Human liver free HK (40 mg/kg) HK ↓
EGCG functionalized CH derivative cancer cell line (HepG2) HK NPs (20 mg/kg) HK NPs 20 ↓↓
(HK NPs) HK NPs (40 mg/kg) HK NPs 40 ↓↓↓
(80 nm) Intertumoral (treatment started when tumor volume
reached 110mm^3, 3/ week)
Vincristine QCT Xenograft: Control (saline/ blank NPs) 18 days [83]
(VCR) Co-delivery BALB/c mice, Raji/VCR cells VCR VCR ↓
PLGA-CHOL-SA-PEG-DSPE (lymphoma) QCT QCT ↓
(VCR/QCT PNs) VCR/QQCT, VCR/QCT ↓↓
(115.7 nm) VCR PNs VCR NPs ↓↓
QCT PNs QCT NPs ↓↓
VCR/QCTPNs VCR/QCT NPs ↓↓↓
20 mg drug/kg
Intravenously (treatment started one week after
cancer induction)

ATC: Anthocyanin (extracted from black soy bean); CH: chitin; CHOL cholesterol; CSU: Chondroitin sulfate; PEG-DSPE: Poly(ethylene glycol)–distearoylpho-
sphatidylethanolamine; PLGA: Poly(lactic co-glycolic acid); pPhe-pHis-PEG: poly(phenylalanine)-b-poly(L-histidine) -b- poly(ethylene glycol); PS: particle size; SA:
stearic acid; SPC: soybean phosphatidylcholine.

and on DEN-induced and 2-acetylaminofluorene (2-AAF)-induced he- increased solubilization and dissolution improved the absorption from
patocarcinogenesis in rats (API [73]). However, in chemically induced enterocytes [68,69], whereas enhanced endocytic activity in human
cancers, treatment typically started before the exposure to the carci- colon cancer cell line colon-26 has been suggested within the me-
nogens (Table 3), whereas in the xenografts’ models, treatment started chanisms involved in the higher cytotoxic effect of physical mixture
when tumor volume reached a defined size in most of the studies (PM) and optimized NAR NPs were found to be approximately 6/7-fold
(Table 3). The administration route in vivo was variable (Table 3) and in and 16/21-fold higher as compared with pure NAR [68,69].
vivo pharmacokinetic studies reported that nano-incapsulation im- Self-emulsifying phospholipid pre-concentrates for oral delivery of
proved oral bioavailability of NAR [68,69] and API [74]. It has been GEN have been tested in ex-vivo permeation study using non-everted
suggested that the presence of the bioactive molecule in the core of NPs gut sac technique [132]. Das et al. [74] reported high efficacy for
prevented its degradation in the gastrointestinal tract [68] and that the oral + topical API NPs versusoral or topical administrations. Tumor

7
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Table 5
Molecular targeted nanoparticles containing flavonoids.
Target Flavonoid - Animal model Treatment and Tumor growth Ref.
Formulation Administration route
(mean PS)

ER QCT DMBA-induced breast Control (saline) 30 Days [84]


(antagonist TMX) Co-delivery carcinogenesis in rat TMX (3 mg/kg) TMX ↓
PLGA TMX/QCT (3 mg/6 mg/Kg) TMX/QCT↓
(TMX-QCT-NPs) TMX-QCT-NPs TMX-QCT-NPs ↓↓
(185.3 nm) Orally (treatment started on tumor bearing
animals, once in 3 days)
ER NAR DMBA-induced breast Control 30 days [85]
(antagonist TMX) Co-delivery carcinogenesis in rat TMX (10 mg) TMX↓
SNEDDS TMX (10 mg) /NAR (20 mg) TMX/NAR↓↓
(52-73 nm) TMX NPs TMX NPs↓↓↓
TMX/NAR NPs TMX/NAR NPs↓↓↓↓
Orally, (treatment started after 10 weeks,
repeated 1 in 3 days)
Breast tumor-homing EGCG delivery Xenograft: Control (Saline and blank nanoparticles) 18 days [86]
cell- CMS@peptide@EGCG Female Balb/c nude mice, MCF-7 100 mg/kg EGCG: EGCG↓
penetrating (150 -180 nm) breast cancer cells EGCG CMS@EGCG ↓↓
peptide CMS@EGCG CMS@peptide@
CMS@peptide@EGCG. EGCG ↓↓↓
Intravenously (treatment started when tumor
volume reached 50 mm3, 5 times at 3-day
intervals).
PSMA EGCG delivery Xenograft Control (PBS/NPs) 7-21 days [87]
small organic molecules targeting Athymic nude mice, 22Rv1 cells EGCG (1 mg/mice/day) EGCG ↓
PSMA (human prostate carcinoma). EGCG NPs (100 mg/mice/day) EGCG NPs ↓↓
(EGCG-NPs) targeted EGCG NPs (100 mg/mice/day) targeted EGCG NPs
(130-250 nm) (treatment started 24 h post cell inoculation, 5 ↓↓↓
times/week)
67LR EGCG Xenograft: Control (Saline) 14-42 Days [88]
198
Delivery Gold (Au) radioactive SCID mice, human prostate cancer EGCG AuNP-EGCG ↓
(198AuNP-EGCG) PC-3 cells. 198
AuNP-EGCG
(535 nm) Intratumoral (Single dose 1 week after cancer
induction, day 6)
67LR EGCG Xenograft: Control (PBS) 2 weeks [89]
Delivery Ruthenium SCID mice, RuBB-Loaded EGCG-RuNPs (2.5 mg/kg) RuBB-Loaded
(EGCG-RuNPs) SMMC-7721 hepatocellular Intratumoral (treatment started after the EGCG-RuNPs ↓
(73.59 nm) carcinoma (HCC) cells tumors become ˜50 mm3, every day)
EIF5A2 CAT Xenograft: Control (PBS) 24-48 days [90]
Delivery siRNA targeting EIF5A2 Athymic mice, T24 cells Mg(II)-CAT Xenograft
(Mg(II)-CAT/siEIF5A2) MNU-induced bladder cancer in rats Mg(II)-CAT/siEIF5A2 (20 mg of siEIF5A2) Mg(II)-CAT ↓
(10-20 nm) Intravenously Mg(II)-CAT/
(treatment started 12 days after T24 siEIF5A2 ↓↓
inoculation, 1/ week for 5 times) MNU-induced:
(treatment started after successful MNU tumor Mg(II)-CAT ↓
induction 16th week, 1/ week for 5 times) Mg(II)-CAT/
siEIF5A2 ↓↓
EGFR EGCG-PEG Xenograft Control (PBS/BSA-NPs) 35 days [91]
(HER)-2 Delivery anticancer protein Athymic nude mice, human breast Herceptin (0.5 mg/ ml) Herceptin-
herceptin (monoclonal antibody cancer BT-474 cells. Herceptin-NPs (0.5- EGCG 0.26 mg/ml) NPs > BSA-NPs
Trastuzumab), Intravenously (treatment started when tumours Herceptin-NPs >
(Herceptin-NPs) reached 270 mm3, twice weekly) Herceptin
(100-500 nm)
FR FA- MSN Xenograft Control (untreated) 15 Days [92]
(QCT NPs) Chicken embryos, MDAMB-231cells QCT NPs (5 μg/mL) QCT NPs ↓
(< 200 nm) Intravenously (treatment started after tumor
induction, on days 12 and 14)
FR-CD44R BCL-PTX Xenograft Control (saline) 24 days [93]
Co-delivery Kunming mice, A549/PTX human PTX (10 mg/kg) PTX ↔
FA-HA lung cancer cells BCL (50 mg/ kg) BCL ↓
(PTX-BCL NPs) PTX-BCL (10 mg/kg + 50 mg/kg) PTX-BCL ↓↓
(90 nm) PTX NPs PTX NPs ↓↓↓
BCL NPs BCL NPs ↓↓↓
PTX-BCL NPs PTX-BCL NPs ↓↓↓↓
Intravenous (treatment started when the tumor
volume reached 90–100 mm3, every 4 days)
CD44R HA-EGCG-cisplatin NPs Xenograft: Control xenograft 38 days [94]
(57 nm) athymic NCR nude mice, ovarian Cisplatin (1 mg) Cisplatin ↓
cancer cell line (SKOV-3) HA-EGCG HA-EGCG ↔
Peritoneal metastatic model: HA-EGCG-cisplatin NPs HA-EGCG-cisplatin
SCID mice, SKOV-3 cells. Intravenously (weekly/3 weeks) NPs ↓↓
metastatic model:
cisplatin ↔
(continued on next page)

8
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Table 5 (continued)

Target Flavonoid - Animal model Treatment and Tumor growth Ref.


Formulation Administration route
(mean PS)

HA-EGCG-cisplatin
NPs ↓
CD44R BCL-DOX Xenograft BCL/DOX 21 days [95]
Co-delivery BALB/c nude mice, MCF-7/ADR DOX NPs, BCL/DOX ↓
HA decorated cells. BCL NPs, DOX NPs ↓↓
(HA-BCL/DOX NPs) HA-BCL/DOX NPs BCL NPs ↓↓↓
(103.5 nm) BCL/DOX-NPs HA-BCL/DOX NPs ↓
(treatment started when tumor volume reached ↓↓↓↓
100 mm3, weekly) BCL/DOX-NPs ↓↓↓↓
CD44R QCT-DOX Xenograft: Control (saline) 14 days [96]
Co-delivery BALB/c nude mice, SGC7901/ADR QCT + DOX (5 mg/kg +5 mg/kg) QCT + DOX ↓
HA - silica cells HA-SiLN/D (DOX 5 mg/kg) HA-SiLN/D ↓↓
(HA-SiLN/QD) HA-SiLN/Q (QCT 5 mg/kg) HA-SiLN/Q ↓↓
(100 nm) HA-SiLN/QD HA-SiLN/QD ↓↓↓
Intravenous (treatment started when the tumor
volume reached 100 mm3, every 2 days for 7
times)
TfR BCL-DOX Xenograft Control (saline/blank NPs) 21 days [97]
Co-delivery BALB/c nude mice, MCF-7/ADR DOX (20 mg) DOX ↓
Tf decorated cells. BCL (20 mg) BCL ↓↓
(Tf-D/B NPs) DOX/BCL DOX/BCL ↓↓↓
(91.8 nm) Tf-D-SLNs (DTX 10 mg) Tf-D-SLNs ↓↓↓↓
Tf-B-SLNs (BCL 10 mg) Tf-B-SLNs ↓↓↓↓↓
Tf-D/B-SLNs Tf-D/B-SLNs ↓↓↓↓↓↓
D/ B-SLNs ↓
Intravenously, (treatment started when tumor D/ B-SLNs ↓↓↓↓↓↓
volume reached 100–200 mm3, weekly)
BR QCT-DOX Xenograft: Control (saline) 21 day [98]
Co-delivery BALB/c nude mice, DOX (5 mg/kg) DOX ↓
MPEG-PCL injecting MCF-7/ADR cells. DOX + QCT, DOX + QCT ↓
(MNDQ) MN DOX MN DOX ↓↓
100.2 MN QCT, MN QCT ↔
Biotin-decorated PEG-PLC MNDQ MNDQ ↓↓↓
(BNDQ) BNDQ BNDQ ↓↓↓↓
105.8 Intravenously (treatment started when tumor
volumes reached 50 mm3, once every three
days for a total of seven doses)
IR QCT-TPT Xenograft Control (saline) 24 days [99]
Co-delivery Athymic nude mice, breast TPT (5 mg/kg) TPT ↓
(MSN-PAA-CS) carcinoma MDA-MB-231 cells QCT (5 mg/kg) QCT ↓
45–55 MSN-PAA-CS (2.5 TPT /2.5 QCT mg/kg) MSN-PAA-CS ↓↓
Arginine-glycine-aspartic acid PMSNs (2.5 TPT /2.5 QCT mg/kg) PMSNs ↓↓↓
peptide Intravenous (Treatment started after 8 days,
(PMSN) weekly)
65–75

67LR: Laminin receptor 67 kDa; BCL: Baicalein; Bovine serum albumin (BSA); BR: biotin receptor; Catechin (CAT); CD44R: CD44 receptor; CMS: colloidal meso-
porous silica; CS: Chitosan; DMBA: 7,12-dimethyl(a)benz anthracene; DOX: Doxorubicin; EGFR: Epidermal growth factor receptor; EIF5A2: Oncogene eukaryotic
translation initiation factor 5A2; ER: Estrogen receptor; FA: folic acid; FR: folate receptors; HA: hyaluronic acid; IR: integrin receptor; MNU: N-methyl-Nitrosurea;
MSN: mesoporous silica nanoparticles; MPEG-PCL: monomethoxy poly(ethylene glycol)-poly(-caprolactone); PAA: poly (acrylic acid); PEG: Polyethylene glycol; PEG-
PCL: poly(ethylene glycol)-poly(-caprolactone)PBS: Phosphate buffered saline; PLGA: Poly (lactic-co-glycolic acid); PSMA: Prostate-specific membrane antigen
PMSN: Arginine-glycine-aspartic acid peptide-MNS; PTX: Paclitaxel; QCT Quercetin; SCID: severe combined immune deficiency; SNEDDS: Self-nano-emulsifying drug
delivery system; Tf: Transferrin; TfR: Transferrin receptor; TPT: Topotecan. TMX: tamoxifen.

onset, induced by UVB–BaP radiation was significantly faster in control urinary bladder as well as in the intestinal region were noticed in mice
mice compared to mice receiving oral + topical API NPs, lowering and rats, as well as modulation of cytochrome P-450 (cyt p-450) con-
chromosomal aberrations and ROS, and inducing apoptosis (caspases tent, glutathione-S-transferase (GST) activity in the cytosolic fraction
and bax/bcl-2 modulation) [74]. On the contrary, in a xenograft mouse and, UDP-glucuronyl transferase (UDPGT) activity in the microsomal
model of head and neck cancer developed by subcutaneous injection of hepatic fraction in rats [73]. Moreover, Das et al. [134] demonstrated
Tu212 cells in athymic nude mice, the efficacy of LUT NPs (in- the ability of API NPs, injected intraperitoneally in mice, to cross the
traperitoneally administered) was not related to apoptosis, whereas blood-brain barrier.
inhibition of proliferation was observed in tumor tissue [72]. The higher efficacy of nanoparticles was confirmed by in vitro ex-
Although in 6 out of 10 studies, that investigated the anticancer periments, where reduced IC50 for proliferation, viability and/or in-
effect, NPs were orally administered (Table 3), also intraperitoneal duction of apoptosis were found for encapsulated/coniugated compared
(LUT [72], GEN [75]), intratumoral (GEN [76]) and intravenous (API to free HES [135], API [72,73], GEN [136–138] and LUT [139]. LUT
[73]) treatments were applied. monomethoxy poly(ethylene glycol)-poly(ε-caprolactone) (MPEG-PCL)
In vivo pharmacokinetics studies in rats reported that encapsulation micelles maintained the cytotoxicity of LUT on 4T1 breast cancer cells
increased AUC and Cmax of LUT intravenously administered [133]. (IC50 6.4 μg/mL) and C-26 colon carcinoma cells (IC50 12.62 μg/mL) in
Radiolabeled API NPs (20 mg/kg single i.v.) accumulations in liver, vitro [133]. ExoAnthos (Table 3) had higher anti-proliferative effects on

9
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

various cancer cell lines (breast MCF7 and MDA-MB-231; lung H1299 DOX in tumor tissue was higher in ATC–DOX NPs treated mice than
and A549; colon HCT116, pancreatic Panc1 and Mia PaCA2; prostate those treated with DOX NPs [77]. In tumor sections, percentage of
DU145 and PC3; ovarian Ova432), compared to Anthos [67]. IC50 cancer cells expressing CD31 (marker for angiogenesis and vascular
values are different depending on cell type, ranging from 2μM (H1299) permeability) and Ki-67 (tumor proliferation marker) were significantly
to 68μM (MDA-MB-231) for ExoAnthos and from 59μM (H1299) to lower in the DOX/QCT NPs treated group than in other groups [77],
960μM (Mia PaCa2) for Anthos, but irrespective of the cancer cell type accounting for the synergistic effect on cancer growth (Table 4). DOX/
ExoAnthos had significantly higher anti-proliferative effects compared QCT NPs did not result in BW loss in mice, whereas DOX and drug
to Anthos, within the same cell type [67]. Furthermore, the anti-in- cocktail (DOX/QCT)-treated groups experienced nearly 11% and 5%
flammatory activity in vitro, measured by the TNF-α-induced NF-kB loss of BW, respectively, indicating severe drug-related toxicity [78].
activity was higher for ExoAnthos compared to Anthos in H1299 [67]. ROS are involved in DOX-mediated hepatotoxicity and cardiotoxicity
Although no in vivo studies on tumor growth inhibition are available, in and QCT, reducing the GSH/GSSG ratio, potentiated the cytotoxic effect
vitro studies investigated the cytotoxic effect of anthocyanin nano- of DOX on the cancer cells and in vitro data, on HCT-116 human colon
particles (Tetraethyl orthosilicate Si(OC2H5)4 (TEOS), hydrolyzed si- cancer cells, suggested a H2O2 dependent drug delivery and cell death
lica, PAMAM (Polyamidoamine) from black carrots [140] and cyanidin- [77], whereas CSU/ATC had free radical scavenging activity in LPS
3-O-Glucoside nanoliposomes [141]. In particular, Yesil-Celiktas et al. treated Hela cells [146]. The histopathologic analysis of hearts in vivo
[140] reported differential effects on Neuro-2A brain neuroblastoma and cellular uptake in MCF-7/Adr cells suggested that combination
from mouse and Vero (African green monkey kidney) normal cell lines therapy with EGCG and DOX in the NPs core could protect the cardi-
and reported that neither empty NPs nor anthocyanin loaded NPs were omyocytes from DOX-mediated cardiotoxicity and overcome MDR, re-
toxic for Vero normal cells, whereas anthocyanin NPs reduced Neuro spectively [147]. EGCG-based NPs were used for the delivery of hon-
2A cells proliferation. Similar results of selective cytotoxic effects on okiol (HK) [82] and intertumoral injection of HK NPs (40 mg/kg)
cancer cells have been reported for CS encapsulated NAR towards inhibited tumor growth by 83.55% (p < 0.05), which was far higher
normal fibroblast 3T3 cells and A549 lung cancer cells and authors also than the 30.15% inhibition of free HK (40 mg/kg). Improved efficacy
observed a free radical scavenging activity in NAR NPs [142]. (Table 4) and delivery of docetaxel (DTX) were reported for topic
Among the other potential mechanisms, there is the modulation of EGCG-based NPs and pharmacokinetics after both intravenous and to-
the early alteration in metabolic activity in cancer cells and tissue. In pical dermal administrations revealed that DTX tumor exposure, as
the DMBA-induced oral carcinogenesis in hamsters, by monitoring the measured by AUC of the concentration-time curve, was 2.48-fold higher
NADH and FAD fluorescence, Gurushankar et al. [143] reported a in mice receiving 30 mg/kg by topically administered DTX/EGCG NPs
greater inhibition of the increased metabolic activity of tumor tissue than those treated with 10 mg/kg DTX by tail vein injection [79].
with HES NPs oral treatment (dose equivalent to 40 mg/kg BW/day of Moreover, the dermal administration decreased toxicity of DTX to the
HES), starting 1 week before exposure to the carcinogen and continuing liver, lung and spleen because the maximum concentrations of DTX by
on days alternate to DMBA for 14 weeks, than HES (40 mg/kg BW/day). dermal administration were lower than those by intravenous adminis-
By using the same DMBA-induced oral carcinogenesis in hamsters, tration [79]. On the other hand, in vivo tissue distribution results found
Krishnakumar et al. [144] reported that oral administration of free NAR that NPs pulmonary administration enhanced the lung PTX and QCT
(50 mg/kg) and NAR NPs significantly increased lipids and the gly- concentrations and decreased their concentration in other tissues, re-
cogen contents and decreased the levels of proteins and nucleic acid ducing the clearance rate of drugs [148]. Moreover, pharmacokinetic
contents in tumor. and intestinal permeation studies reported high etoposide AUC after
When the safety profile was assessed by measuring the changes in treatment with a QCT-based NPs and the involvement of P-glycoprotein
BW during the cancerogenesis protocol, NAR [68,69], LUT [72], API (Pgp) [149].
[74] and GEN [75,76] NPs inhibited the cancer-induced BW reduction. Flavone baicalein (BCL) was used as MDR reversal agent in co-en-
On the contrary, pristine-GEN solution induced BW loss in mice [76]. capsulated paclitaxel (PTX) NPs and were reported synergistic effects
On the other hand, Antho was nontoxic in wild-type mice at a dose and improved cellular uptake in vitro [80] and inhibition of tumor
(40 mg/kg weekly dose) higher than those used for anti-tumor study growth in vivo (Table 4). Also GEN improved PTX efficacy in vivo
(weekly dose of 30 mg/kg as free Anthos or 15 mg/kg in bovine milk (Table 4) [81].
exosomal formulation, ExoAnthos) [67]. No effects on blood counts, In a co-delivery system tested for tumor growth inhibition in lym-
liver and kidney function enzymes were observed in the toxicity study phoma-bearing BALB/c nude mice model (Table 4), both vincristine
[67]. (VCR) and QCT distribution of VCR/QCT PNs were higher in the tumor
tissue than VCR/QCT solution, mainly distributed in heart and kidney
6. Flavonoids in delivery and co-delivery systems of anticancer [83].
drugs
7. Molecular targeted nanoparticles containing flavonoids
Chemotherapy for cancer treatment causes side effects on healthy
tissues and MDR of the tumor cells, which limits therapy. To address Molecular targeted cancer therapy mediated by NPs is a promising
these limitations, combination therapies based on NPs and flavonoids strategy to overcome the lack of specificity of conventional che-
administration have been suggested to both improve efficacy (through motherapeutic agents and to improve efficacy and reduce toxicity. The
delivering different agents simultaneously with synergistic effect) and estrogen receptor antagonist tamoxifen (TMX) can be considered a
minimize toxicity of anticancer drugs (by targeting towards cancer cells “pioneer" of this approach. Kumar et al. [150] studied NPs for the de-
and by counteracting ROS-mediated damage). Combination therapies livery of TMX with physically adsorbed QCT (TMX-QCT-NPs), the latter
included co-administration and co-delivery. Co-administration of QCT being a mild P-gp efflux inhibitor, in order to treat MDR-cancer cells
NPs [64] or theaflavin-EGCG NPs [145] with cisplatin improved its MDA-MB-231. Authors reported reduced IC50 values for TMX-QCT-NPs
efficacy in mice. compared to TMX and TMX-NPs. Moreover, the pharmacokinetic in rats
Studies that evaluated tumor growth after treatment with flavonoids (intravenous injection) of TMX (2 mg/Kg), TMX-NPs or TMX-QCT-NPs
in delivery and co-delivery systems of anticancer drugs are summarized (equivalent to 2 mg/Kg of TMX and 10 mg/Kg of QCT) turned out high
in Table 4. plasma drug concentrations from TMX-QCT-NPs (AUC ng·mL−1·h
Anthocyanins from black soy bean (ATC) [77] loaded NPs, for DOX 9138.36) and TMX-NPs (AUC ng·mL−1·h 7770.58) by comparison with
delivery, increased DOX NPs efficacy in vivo (Table 4). Histological pure TMX (AUC ng·mL−1·h 3435.62) [150]. Jain et al. [84] in-
analysis revealed apoptosis after treatment with ATC–DOX NPs and that vestigated TMX association with quercetin in NPs. The plasma

10
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

concentration time profiles of TMX and QCT after single oral adminis- anticancer protein followed by complexation of PEG-EGCG [91], which
tration of free TMX (10 mg/kg), free QCT (20 mg/kg), a combination of have greater anticancer effects in vitro and in vivo (Table 5) than the free
TMX and QCT (TMX/QCT) or TMX-QCT-NPs (equivalent to 10 mg/kg protein. Herceptin NPs showed the improved tumor selectivity to sur-
TMX) revealed that TMX-QCT-NPs increased the bioavailability of TMX rounding normal organs/tissues, exhibited 2.3-fold higher accumula-
by 5.31- and 2.69-fold as compared to free TMX and TMX/QCT. Co- tion in tumor and 0.3-, 0.3- and 0.6-fold lower accumulation in liver,
formulated NPs increased the oral bioavailability of QCT by 2.9-fold in kidney and lung, respectively, as compared to free Herceptin at 24 h
comparison with free QCT and its combination with free TMX. More- post-injection and were observed throughout the extravascular space,
over, TMX-QCT-NPs had higher in vivo antitumor efficacy (Table 5), as which was consistent with the localization of overexpressed HER2/neu
well as anti-angiogenic potential measured by plasma MMP-2 and receptors on the tumor [91].
MMP-3 after 30 days of treatment. Interestingly, TMX-QCT-NPs, but not On the other hand, folic acid (FA), having high selectivity for folate
TMX/QCT, reduced TMX hepatotoxicity, measured by liver markers in receptors (FR), which are overexpressed on the surface of cancer cells,
plasma (AST and ALT), oxidative stress markers in liver tissue (thio- has been employed in the development of EGCG NPs and tested on
barbituric acid reactive substances (TBARS) and GSH) and liver histo- human breast cancer MCF-7 cells [102]. The in vivo tumor accumulation
pathology (parenchymal degeneration, lymphocyte infiltration and [153] and the in vitro targeting of FA armed QCT NPs to folate-ex-
apoptosis of cells) [84]. Similar results were found with TMX/NAR NPs, pressing cells [153], were confirmed on cancer growth in Chicken
with improved pharmacokinetic, reduced hepatotoxicity [85] and in- embryos [92] (Table 5). In biodistribution experiments, QCT was co-
creased efficacy (Table 5) compared to TMX in solution. delivered with DTX by using hyaluronic acid (HA)-modified NPs as
In a xenograft model of breast cancerogenesis, the anticancer effect tumor targeting system [154]. Dual-targeted ligands of FA and HA have
of EGCG was increased by loading of EGCG into colloidal mesoporous been successful employed for BCL and PTX co-delivery in a xenograft
silica (CMS) containing a breast tumor-homing cell-penetrating peptide mice model with injection of A549/PTX resistant human lung cancer
NPs (CMS@peptide) [86] (Table 5). On tissue sections, at 18 days after cells [93]. HA on the surface of the complex can bind to CD44 receptor,
treatment with EGCG, CMS@EGCG, and CMS@peptide@EGCG, ne- which is overexpressed in some cancer cells [93,95,96]. HA-EGCG NPs
crosis was observed in histological sections from the different treatment improved cisplatin anticancer effect in both xenograft and peritoneal
groups, whereas no sign of organ damage was found [86]. The prostate- metastatic in vivo models [94], whereas orally EGCG (20 mg/kg) and
specific membrane antigen (PSMA) targeting has been also used to se- HA-based NPs (intravenously) improved DOX efficacy [155]. HA de-
lectively deliver EGCG to cancer cells in vitro [151] and in vivo corated BCL/DOX NPs reduced tumor growth induced by injection of
(Table 5). In a tumor xenograft model EGCG NPs showed better efficacy MCF-7/ADR resistant cells better than BCL/DOX-NPs, single drug NPs
than native EGCG and decreased serum PSA [87]. and free BCL/DOX (Table 5) [95]. Similar results were obtained with
On the other hand, in vivo studies investigated the use of the high QCT and DOX in HA-modified silica NPs [96] and the biodistribution
binding affinity of EGCG to the 67-kDa laminin receptor (67LR) study revealed that a high dose of HA intravenously injected prior to
(Table 5), overexpressed in cancer cells, in order to increase the de- administration of NPs to saturate CD44 receptors reduced tumor tar-
livery of NPs. EGCG (used as a capping agent) improved the efficacy of geting [96]. Tumor targeting has been reported also for HA-EGCG-cis-
both radioactive gold NPs derived from the Au-198 isotope [88] and platin NPs and HA-EGCG formulation inhibited cisplatin-induced liver
ruthenium NPs [89] in xenografts models with 67LR overexpression in toxicity (ALT and AST) [94].
human prostate cancer cells (PC-3) [88] and hepatocellular carcinoma It has been suggested that QCT decreased the expression of Pgp,
(HCC) [89] cells (Table 5). In vitro experiments showed that RuBB- thus reversing MDR and facilitate DOX activity against SGC7901/ADR
loaded EGCG-RuNPs accumulated in the cytoplasm of SMMC-7721 resistant cells [96]. On the other hand, transferrin receptor(TfR)-
cells, where direct ROS-dependent cytotoxic and pro-apoptotic effects mediated, biotin receptor (BR)-mediated and integrin receptor (IR)-
were observed, with no toxic effect in normal L-02 cells [89]. Phar- mediated (arginine-glycine-aspartic acid peptide) endocytosis improved
macokinetic and therapeutic studies in PC-3 xenograft SCID mice in vivo efficacy of BCL/DOX [97], DOX/QCT [98] and topotecan(TPT)/
showed 72% retention of NPs in tumors 24 h after intratumoral ad- QCT [99] co-delivery NPs, respectively (Table 5). Tumor targeting
ministration [89] and efficacy (Table 5). Shukla et al. [88] did observe viareceptor-mediated endocytosis has been also suggested for DOX/
neither EGCG therapeutic effects nor blood parameters alterations EGCG NPs linked with an aptamer (DOX/EGCG NP-Apt) [156]. In a
(RBC, WBC, Hb and lymphocytes) within the tumor-bearing mice. xenograft mice model, it has been reported that DOX accumulated
Analysis of radioactivity revealed that 72% NPs were retained in preferentially in liver and kidney after free DOX intravenous injection,
prostate tumors at 24 h. Intratumoral injected NPs exhibited slow whereas DOX/EGCG NPs and to a great extent DOX/EGCG NP-Apt ac-
leakage into the blood (0.06%), and variable uptakes were observed: cumulated in the tumor.
lungs (0.33% at 24 h), pancreas (0.22% at 24 h), kidneys (0.12% at
24 h), spleen (1.56% at 24 h), stomach (5% at 2 h, decreasing to 0.03% 8. Discussion and conclusion
at 24 h), small intestines (0.91% at 2 h, decreasing to 0.01% at 24 h)
and liver (0.51% after 30 min, increasing to 6.13% after 24 h). Application of nanotechnologies to cancer therapy might increase
Catechin [90] and EGCG containing NPs have been also used to solubility and/or bioavailability of bioactive compounds of natural or
deliver small interfering RNA (siRNA), targeting the oncogene eu- synthetic origin and offers other potential benefits in cancer therapy,
karyotic translation initiation factor 5A2 (EIF5A2) and herceptin including selective targeting [157,158]. In both xenograft and che-
(Trastuzumab), the approved monoclonal antibody (mAb) for EGFR mical-induced animal models of cancerogenesis, NPs (formulated with
(HER)-2 positive breast cancer and gastric or gastroesophageal junction CS, PEG, PLGA (Fig. 1) and other chemicals (Tables 1–3) of food-de-
adenocarcinoma [152], into cells in vitro [90,91] and into tumor site in rived flavonoids resulted in better efficacy for tumor growth than free
vivo (Table 5). compounds (Tables 1–3), and synergistic effects in co-delivery systems
Chen et al. [90] reported that Mg(II)-CAT/siEIF5A2 inhibited on- (both drug and flavonoid in the core, Fig. 1) have been reported
cogenic PI3K/Akt signal pathway and demonstrated, in mice, effective (Table 4). Besides, when EGCG [147] and BCL [80] were used as MDR
delivery in heart, lung, liver, kidneys, spleen, and most importantly reversal agent in co-encapsulated (DOX and PTX) NPs increased bioa-
tumor. Accordingly, Mg(II)-CAT/siEIF5A2 had high efficacy on tumor vailability and efficacy of drugs. QCT has been used as MDR reversal
growth in both xenograft and N-methyl-N-nitrosourea (MNU)– induced agent as well [96,150]. Moreover, flavonoids’ loaded NPs for delivery
cancer models (Table 5). of anticancer drugs (Table 4), as well as decorated NPs targeting cancer
Self-assembly of the EGCG derivative with anticancer herceptin cells (Table 5), increased the efficacy and/or reduced the systemic
formed NPs, obtained by complexation of oligomerized EGCG with the toxicity of drugs.

11
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

References

[1] J. Perez-Jimenez, V. Neveu, F. Vos, A. Scalbert, Systematic analysis of the content


of 502 polyphenols in 452 foods and beverages: an application of the phenol-
explorer database, J. Agric. Food Chem. 58 (8) (2010) 4959–4969, https://doi.
org/10.1021/jf100128b.
[2] I. Peluso, M. Serafini, Antioxidants from black and green tea: from dietary mod-
ulation of oxidative stress to pharmacological mechanisms, Br. J. Pharmacol. 174
(11) (2017) 1195–1208, https://doi.org/10.1111/bph.13649.
[3] M.G. Hertog, P.C. Hollman, M.B. Katan, D. Kromhout, Intake of potentially an-
ticarcinogenic flavonoids and their determinants in adults in The Netherlands,
Nutr. Cancer 20 (1) (1993) 21–29, https://doi.org/10.1080/
01635589309514267.
[4] R. Zamora-Ros, V. Knaze, J.A. Rothwell, B. Hemon, A. Moskal, K. Overvad,
A. Tjonneland, C. Kyro, G. Fagherazzi, M.C. Boutron-Ruault, M. Touillaud,
V. Katzke, T. Kuhn, H. Boeing, J. Forster, A. Trichopoulou, E. Valanou, E. Peppa,
D. Palli, C. Agnoli, F. Ricceri, R. Tumino, M.S. De Magistris, P.H. Peeters,
H.B. Bueno-De-Mesquita, D. Engeset, G. Skeie, A. Hjartaker, V. Menendez,
A. Agudo, E. Molina-Montes, J.M. Huerta, A. Barricarte, P. Amiano, E. Sonestedt,
L.M. Nilsson, R. Landberg, T.J. Key, K.T. Khaw, N.J. Wareham, Y. Lu, N. Slimani,
I. Romieu, E. Riboli, A. Scalbert, Dietary polyphenol intake in Europe: the
European Prospective Investigation into Cancer and Nutrition (EPIC) study, Eur. J.
Nutr. 55 (4) (2016) 1359–1375, https://doi.org/10.1007/s00394-015-0950-x.
[5] G. Grosso, U. Stepaniak, R. Topor-Madry, K. Szafraniec, A. Pajak, Estimated
Fig. 1. Nanodelivery and nanoparticles of dietary flavonoids. dietary intake and major food sources of polyphenols in the Polish arm of the
CS: chitosan; NPs: nanoparticles; PEG: Polyethylene glycol; PLGA: Poly(lactic HAPIEE study, Nutrition 30 (11–12) (2014) 1398–1403, https://doi.org/10.1016/
co-glycolic acid); ROS: reactive oxygen species. j.nut.2014.04.012.
[6] C. Taguchi, Y. Fukushima, Y. Kishimoto, N. Suzuki-Sugihara, E. Saita,
Y. Takahashi, K. Kondo, Estimated dietary polyphenol intake and major food and
beverage sources among elderly Japanese, Nutrients 7 (12) (2015) 10269–10281,
https://doi.org/10.3390/nu7125530.
Furthermore, reductions in the MMPs have been reported among [7] P. Knekt, R. Jarvinen, A. Reunanen, J. Maatela, Flavonoid intake and coronary
molecular mechanisms [47,48,84], suggesting inhibition of invasion, mortality in Finland: a cohort study, Bmj 312 (7029) (1996) 478–481, https://doi.
org/10.1136/bmj.312.7029.478.
migration and angiogenesis [159]. [8] O.K. Chun, S.J. Chung, W.O. Song, Estimated dietary flavonoid intake and major
Mechanisms of cellular uptake include diffusion, endocytosis and food sources of U.S. adults, J. Nutr. 137 (5) (2007) 1244–1252, https://doi.org/
specific molecular target NPs (Fig. 1). In addition to receptor-mediated 10.1093/jn/137.5.1244.
[9] W. Bai, C. Wang, C. Ren, Intakes of total and individual flavonoids by US adults,
targeting, it has been reported that EGCG, due to its high binding af-
Int. J. Food Sci. Nutr. 65 (1) (2014) 9–20, https://doi.org/10.3109/09637486.
finity to the 67LR (Table 5), overexpressed in cancer cells, can be used 2013.832170.
as capping agent. Cancer cells have lower pH and higher ROS content [10] J. Perez-Jimenez, L. Fezeu, M. Touvier, N. Arnault, C. Manach, S. Hercberg,
and within mechanisms, ROS-dependent [77,89] and pH-dependent P. Galan, A. Scalbert, Dietary intake of 337 polyphenols in French adults, Am. J.
Clin. Nutr. 93 (6) (2011) 1220–1228, https://doi.org/10.3945/ajcn.110.007096.
[62], drug delivery and/or cell death have been suggested. pH-re- [11] A. Tresserra-Rimbau, A. Medina-Remon, J. Perez-Jimenez, M.A. Martinez-
sponsive drug carriers loaded with QCT were synthesized with CS Gonzalez, M.I. Covas, D. Corella, J. Salas-Salvado, E. Gomez-Gracia, J. Lapetra,
chemically modified with 2-chloro-N,N-diethylethylamine hydro- F. Aros, M. Fiol, E. Ros, L. Serra-Majem, X. Pinto, M.A. Munoz, G.T. Saez, V. Ruiz-
Gutierrez, J. Warnberg, R. Estruch, R.M. Lamuela-Raventos, Dietary intake and
chloride and dodecyl aldehyde [160] and PEG modified viaa pH-re- major food sources of polyphenols in a Spanish population at high cardiovascular
sponsive coordination bond to form triphenylphosphine-QCT-PEG NPs risk: the PREDIMED study, Nutr. Metab. Cardiovasc. Dis. 23 (10) (2013) 953–959,
[62]. https://doi.org/10.1016/j.numecd.2012.10.008.
[12] M.A. Nascimento-Souza, P.G. De Paiva, J. Perez-Jimenez, S. Do Carmo Castro
On the other hand, results with ROS-sensitive drug carriers, in- Franceschini, A.Q. Ribeiro, Estimated dietary intake and major food sources of
cluding ATC in NPs for DOX delivery, demonstrated that higher DOX polyphenols in elderly of Vicosa, Brazil: a population-based study, Eur. J. Nutr. 57
release occurred compared to ATC-free NPs [77]. Authors suggested (2) (2018) 617–627, https://doi.org/10.1007/s00394-016-1348-0.
[13] J. Godos, S. Marventano, A. Mistretta, F. Galvano, G. Grosso, Dietary sources of
that DOX was almost always released before reaching at the tumor site
polyphenols in the Mediterranean healthy Eating, Aging and Lifestyle (MEAL)
in the ATC-free DOX NPs, whereas DOX was released from ATC-con- study cohort, Int. J. Food Sci. Nutr. 68 (6) (2017) 750–756, https://doi.org/10.
taining NPs at the tumor site, by ROS mediated ATC degradation and 1080/09637486.2017.1285870.
[14] K. Bhise, S.K. Kashaw, S. Sau, A.K. Iyer, Nanostructured lipid carriers employing
NPs destruction [77]. Similarly, it has been reported that NPs can ef-
polyphenols as promising anticancer agents: quality by design (QbD) approach,
fectively protect the carotenoid lycopene against degradation due to Int. J. Pharm. 526 (1–2) (2017) 506–515, https://doi.org/10.1016/j.ijpharm.
EGCG's anti-oxidant property [161]. Authors applied self-assembly of 2017.04.078.
oligomerized (-)-epigallocatechin-3-O-gallate (OEGCG) for delivery of [15] I.S. Santos, B.M. Ponte, P. Boonme, A.M. Silva, E.B. Souto, Nanoencapsulation of
polyphenols for protective effect against colon-rectal cancer, Biotechnol. Adv. 31
lycopene [161]. (5) (2013) 514–523, https://doi.org/10.1016/j.biotechadv.2012.08.005.
In this context, polyphenols act as both reducing and capping agents [16] M.M. Mocanu, P. Nagy, J. Szollosi, Chemoprevention of breast Cancer by dietary
of green chemistry process [19,161,162] and tea [161] and Mangosteen polyphenols, Molecules 20 (12) (2015) 22578–22620, https://doi.org/10.3390/
molecules201219864.
pericarp waste [163] extracts have been employed to produce NPs by [17] R.Y. Gan, H.B. Li, Z.Q. Sui, H. Corke, Absorption, metabolism, anti-cancer effect
eco-friendly synthesis. Other suggested natural excipients include α- and molecular targets of epigallocatechin gallate (EGCG): an updated review, Crit.
tocopherol [164] and squalene [165]. On the other hand, non-flavonoid Rev. Food Sci. Nutr. 58 (6) (2018) 924–941, https://doi.org/10.1080/10408398.
2016.1231168.
bioactive phytochemicals, including curcumin, has been included in [18] B. Peter, S. Bosze, R. Horvath, Biophysical characteristics of proteins and living
nanoformulations [166] or used as a model for the synthesis of analogs cells exposed to the green tea polyphenol epigallocatechin-3-gallate (EGCg): re-
with better bioavailability and higher anti-cancer activity [167]. view of recent advances from molecular mechanisms to nanomedicine and clinical
trials, Eur. Biophys. J. 46 (1) (2017) 1–24, https://doi.org/10.1007/s00249-016-
In conclusion, although human studies are needed, NPs obtained
1141-2.
from food-derived flavonoids show promising anticancer effects in vivo, [19] A. Granja, M. Pinheiro, S. Reis, Epigallocatechin gallate nanodelivery systems for
improve cancer drug efficacy and minimize their toxicity. Cancer therapy, Nutrients 8 (5) (2016), https://doi.org/10.3390/nu8050307.
[20] E. Shankar, A. Goel, K. Gupta, S. Gupta, Plant flavone apigenin: an emerging
anticancer agent, Curr. Pharmacol. Rep. 3 (6) (2017) 423–446, https://doi.org/10.
1007/s40495-017-0113-2.
Declaration of Competing Interest [21] G.P. Nagaraju, S.F. Zafar, B.F. El-Rayes, Pleiotropic effects of genistein in meta-
bolic, inflammatory, and malignant diseases, Nutr. Rev. 71 (8) (2013) 562–572,
https://doi.org/10.1111/nure.12044.
The authors declare that there are no conflicts of interest.

12
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

[22] M. Iriti, E.M. Varoni, Chemopreventive potential of flavonoids in oral squamous a novel approach for cancer control: proof of principle with green tea polyphenol
cell carcinoma in human studies, Nutrients 5 (7) (2013) 2564–2576, https://doi. epigallocatechin-3-gallate, Cancer Res. 69 (5) (2009) 1712–1716, https://doi.org/
org/10.3390/nu5072564. 10.1158/0008-5472.CAN-08-3978.
[23] T. Khushnud, S.A. Mousa, Potential role of naturally derived polyphenols and their [47] S. Balakrishnan, F.A. Bhat, P. Raja Singh, S. Mukherjee, P. Elumalai, S. Das,
nanotechnology delivery in cancer, Mol. Biotechnol. 55 (1) (2013) 78–86, https:// C.R. Patra, J. Arunakaran, Gold nanoparticle-conjugated quercetin inhibits epi-
doi.org/10.1007/s12033-012-9623-7. thelial-mesenchymal transition, angiogenesis and invasiveness via EGFR/VEGFR-
[24] P. Knekt, J. Kumpulainen, R. Jarvinen, H. Rissanen, M. Heliovaara, A. Reunanen, 2-mediated pathway in breast cancer, Cell Prolif. 49 (6) (2016) 678–697, https://
T. Hakulinen, A. Aromaa, Flavonoid intake and risk of chronic diseases, Am. J. doi.org/10.1111/cpr.12296.
Clin. Nutr. 76 (3) (2002) 560–568, https://doi.org/10.1093/ajcn/76.3.560. [48] A.K. Mandal, D. Ghosh, S. Sarkar, A. Ghosh, S. Swarnakar, N. Das, Nanocapsulated
[25] G. Grosso, J. Godos, R. Lamuela-Raventos, S. Ray, A. Micek, A. Pajak, S. Sciacca, quercetin downregulates rat hepatic MMP-13 and controls diethylnitrosamine-
N. D’orazio, D. Del Rio, F. Galvano, A comprehensive meta-analysis on dietary induced carcinoma, Nanomedicine 9 (15) (2014) 2323–2337, https://doi.org/10.
flavonoid and lignan intake and cancer risk: level of evidence and limitations, Mol. 2217/nnm.14.11.
Nutr. Food Res. 61 (4) (2017), https://doi.org/10.1002/mnfr.201600930. [49] P. Pandey, M. Rahman, P.C. Bhatt, S. Beg, B. Paul, A. Hafeez, F.A. Al-Abbasi,
[26] L.O. Dragsted, M. Strube, T. Leth, Dietary levels of plant phenols and other non- M.S. Nadeem, O. Baothman, F. Anwar, V. Kumar, Implication of nano-antioxidant
nutritive components: could they prevent cancer? Eur. J. Cancer Prev. 6 (6) (1997) therapy for treatment of hepatocellular carcinoma using PLGA nanoparticles of
522–528. rutin, Nanomedicine 13 (8) (2018) 849–870, https://doi.org/10.2217/nnm-2017-
[27] I.A. Siddiqui, V. Sanna, Impact of nanotechnology on the delivery of natural 0306.
products for cancer prevention and therapy, Mol. Nutr. Food Res. 60 (6) (2016) [50] A. Ghosh, D. Ghosh, S. Sarkar, A.K. Mandal, S. Thakur Choudhury, N. Das,
1330–1341, https://doi.org/10.1002/mnfr.201600035. Anticarcinogenic activity of nanoencapsulated quercetin in combating diethylni-
[28] I. Peluso, M. Palmery, Flavonoids at the pharma-nutrition interface: is a ther- trosamine-induced hepatocarcinoma in rats, Eur. J. Cancer Prev. 21 (1) (2012)
apeutic index in demand? Biomed. Pharmacother. 71 (2015) 102–107, https:// 32–41, https://doi.org/10.1097/CEJ.0b013e32834a7e2b.
doi.org/10.1016/j.biopha.2015.02.028. [51] X. Wei, X. Gao, B. Wang, K. Men, F. Zheng, Y. Zhou, Y. Zheng, M. Gou, M. Huang,
[29] D. Del Rio, A. Rodriguez-Mateos, J.P. Spencer, M. Tognolini, G. Borges, A. Crozier, G. Guo, N. Huang, Z. Qian, Y. Wei, Anticancer effect and mechanism of polymer
Dietary (poly)phenolics in human health: structures, bioavailability, and evidence micelle-encapsulated quercetin on ovarian cancer, Nanoscale 4 (22) (2012)
of protective effects against chronic diseases, Antioxid. Redox Signal. 18 (14) 7021–7030, https://doi.org/10.1039/c2nr32181e.
(2013) 1818–1892, https://doi.org/10.1089/ars.2012.4581. [52] J. Liu, J. Zhao, T. Wei, X. Ma, Q. Cheng, S. Huo, C. Zhang, Y. Zhang, X. Duan,
[30] J.S. Nam, A.R. Sharma, L.T. Nguyen, C. Chakraborty, G. Sharma, S.S. Lee, X.J. Liang, Quercetin-loaded nanomicelles to circumvent human castration-re-
Application of bioactive quercetin in oncotherapy: from nutrition to nanomedi- sistant prostate cancer in vitro and in vivo, Nanoscale 8 (9) (2016) 5126–5138,
cine, Molecules 21 (1) (2016) E108, https://doi.org/10.3390/ https://doi.org/10.1039/c5nr08966b.
molecules21010108. [53] R. Baksi, D.P. Singh, S.P. Borse, R. Rana, V. Sharma, M. Nivsarkar, In vitro and in
[31] F.H. Sarkar, Y. Li, Z. Wang, S. Padhye, Lesson learned from nature for the de- vivo anticancer efficacy potential of Quercetin loaded polymeric nanoparticles,
velopment of novel anti-cancer agents: implication of isoflavone, curcumin, and Biomed. Pharmacother. 106 (2018) 1513–1526, https://doi.org/10.1016/j.
their synthetic analogs, Curr. Pharm. Des. 16 (16) (2010) 1801–1812. biopha.2018.07.106.
[32] N. Tyagi, R. De, J. Begun, A. Popat, Cancer therapeutics with epigallocatechin-3- [54] G. Wang, J.J. Wang, X.L. Chen, L. Du, F. Li, Quercetin-loaded freeze-dried nano-
gallate encapsulated in biopolymeric nanoparticles, Int. J. Pharm. 518 (1–2) micelles: improving absorption and anti-glioma efficiency in vitro and in vivo, J.
(2017) 220–227, https://doi.org/10.1016/j.ijpharm.2016.12.030. Control. Release 235 (2016) 276–290, https://doi.org/10.1016/j.jconrel.2016.05.
[33] O. Krupkova, S.J. Ferguson, K. Wuertz-Kozak, Stability of (-)-epigallocatechin 045.
gallate and its activity in liquid formulations and delivery systems, J. Nutr. [55] A.S. Jain, S.M. Shah, M.S. Nagarsenker, Y. Nikam, R.P. Gude, F. Steiniger,
Biochem. 37 (2016) 1–12, https://doi.org/10.1016/j.jnutbio.2016.01.002. J. Thamm, A. Fahr, Lipid colloidal carriers for improvement of anticancer activity
[34] A. Aras, A.R. Khokhar, M.Z. Qureshi, M.F. Silva, A. Sobczak-Kupiec, E.A. Pineda, of orally delivered quercetin: formulation, characterization and establishing in
A.A. Hechenleitner, A.A. Farooqi, Targeting cancer with nano-bullets: curcumin, vitro-in vivo advantage, J. Biomed. Nanotechnol. 9 (7) (2013) 1230–1240.
EGCG, resveratrol and quercetin on flying carpets, Asian Pac. J. Cancer Prev. 15 [56] C.L. Luo, Y.Q. Liu, P. Wang, C.H. Song, K.J. Wang, L.P. Dai, J.Y. Zhang, H. Ye, The
(9) (2014) 3865–3871. effect of quercetin nanoparticle on cervical cancer progression by inducing
[35] X. Cai, Z. Fang, J. Dou, A. Yu, G. Zhai, Bioavailability of quercetin: problems and apoptosis, autophagy and anti-proliferation via JAK2 suppression, Biomed.
promises, Curr. Med. Chem. 20 (20) (2013) 2572–2582. Pharmacother. 82 (2016) 595–605, https://doi.org/10.1016/j.biopha.2016.05.
[36] U. Ernest, H.Y. Chen, M.J. Xu, Y.D. Taghipour, M. Asad, R. Rahimi, G. Murtaza, 029.
Anti-cancerous potential of polyphenol-loaded polymeric nanotherapeutics, [57] K.W. Ren, Y.H. Li, G. Wu, J.Z. Ren, H.B. Lu, Z.M. Li, X.W. Han, Quercetin nano-
Molecules 23 (11) (2018), https://doi.org/10.3390/molecules23112787. particles display antitumor activity via proliferation inhibition and apoptosis in-
[37] I.A. Siddiqui, V.M. Adhami, N. Ahmad, H. Mukhtar, Nanochemoprevention: sus- duction in liver cancer cells, Int. J. Oncol. 50 (4) (2017) 1299–1311, https://doi.
tained release of bioactive food components for cancer prevention, Nutr. Cancer org/10.3892/ijo.2017.3886.
62 (7) (2010) 883–890, https://doi.org/10.1080/01635581.2010.509537. [58] M. Lou, L.N. Zhang, P.G. Ji, F.Q. Feng, J.H. Liu, C. Yang, B.F. Li, L. Wang,
[38] Y. Davatgaran-Taghipour, S. Masoomzadeh, M.H. Farzaei, R. Bahramsoltani, Quercetin nanoparticles induced autophagy and apoptosis through AKT/ERK/
Z. Karimi-Soureh, R. Rahimi, M. Abdollahi, Polyphenol nanoformulations for Caspase-3 signaling pathway in human neuroglioma cells: in vitro and in vivo,
cancer therapy: experimental evidence and clinical perspective, Int. J. Biomed. Pharmacother. 84 (2016) 1–9, https://doi.org/10.1016/j.biopha.2016.
Nanomedicine 12 (2017) 2689–2702, https://doi.org/10.2147/IJN.S131973. 08.055.
[39] G. Testa, G. Leonarduzzi, B. Sottero, P. Gamba, G. Poli, Design and development of [59] G. Sharma, J. Park, A.R. Sharma, J.S. Jung, H. Kim, C. Chakraborty, D.K. Song,
nanovehicle-based delivery systems for preventive or therapeutic supplementation S.S. Lee, J.S. Nam, Methoxy poly(ethylene glycol)-poly(lactide) nanoparticles
with flavonoids, Curr. Med. Chem. 17 (1) (2010) 74–95. encapsulating quercetin act as an effective anticancer agent by inducing apoptosis
[40] N. Khan, D.J. Bharali, V.M. Adhami, I.A. Siddiqui, H. Cui, S.M. Shabana, in breast cancer, Pharm. Res. 32 (2) (2015) 723–735, https://doi.org/10.1007/
S.A. Mousa, H. Mukhtar, Oral administration of naturally occurring chitosan-based s11095-014-1504-2.
nanoformulated green tea polyphenol EGCG effectively inhibits prostate cancer [60] G. Xu, H. Shi, L. Ren, H. Gou, D. Gong, X. Gao, N. Huang, Enhancing the anti-colon
cell growth in a xenograft model, Carcinogenesis 35 (2) (2014) 415–423, https:// cancer activity of quercetin by self-assembled micelles, Int. J. Nanomedicine 10
doi.org/10.1093/carcin/bgt321. (2015) 2051–2063, https://doi.org/10.2147/IJN.S75550.
[41] I.A. Siddiqui, D.J. Bharali, M. Nihal, V.M. Adhami, N. Khan, J.C. Chamcheu, [61] X. Guan, M. Gao, H. Xu, C. Zhang, H. Liu, L. Lv, S. Deng, D. Gao, Y. Tian,
M.I. Khan, S. Shabana, S.A. Mousa, H. Mukhtar, Excellent anti-proliferative and Quercetin-loaded poly (lactic-co-glycolic acid)-d-alpha-tocopheryl polyethylene
pro-apoptotic effects of (-)-epigallocatechin-3-gallate encapsulated in chitosan glycol 1000 succinate nanoparticles for the targeted treatment of liver cancer,
nanoparticles on human melanoma cell growth both in vitro and in vivo, Drug Deliv. 23 (9) (2016) 3307–3318, https://doi.org/10.1080/10717544.2016.
Nanomedicine 10 (8) (2014) 1619–1626, https://doi.org/10.1016/j.nano.2014. 1176087.
05.007. [62] L. Xing, J.Y. Lyu, Y. Yang, P.F. Cui, L.Q. Gu, J.B. Qiao, Y.J. He, T.Q. Zhang, M. Sun,
[42] Y.H. Lin, Z.R. Chen, C.H. Lai, C.H. Hsieh, C.L. Feng, Active targeted nanoparticles J.J. Lu, X. Xu, Y. Liu, H.L. Jiang, pH-Responsive de-PEGylated nanoparticles based
for oral administration of gastric Cancer therapy, Biomacromolecules 16 (9) on triphenylphosphine-quercetin self-assemblies for mitochondria-targeted cancer
(2015) 3021–3032, https://doi.org/10.1021/acs.biomac.5b00907. therapy, Chem. Commun. 53 (62) (2017) 8790–8793, https://doi.org/10.1039/
[43] C.C. Chen, D.S. Hsieh, K.J. Huang, Y.L. Chan, P.D. Hong, M.K. Yeh, C.J. Wu, c7cc04058j.
Improving anticancer efficacy of (-)-epigallocatechin-3-gallate gold nanoparticles [63] J. Li, M. Shi, B. Ma, R. Niu, H. Zhang, L. Kun, Antitumor activity and safety
in murine B16F10 melanoma cells, Drug Des. Devel. Ther. 8 (2014) 459–474, evaluation of nanaparticle-based delivery of quercetin through intravenous ad-
https://doi.org/10.2147/DDDT.S58414. ministration in mice, Mater. Sci. Eng. C Mater. Biol. Appl. 77 (2017) 803–810,
[44] D.S. Hsieh, H. Wang, S.W. Tan, Y.H. Huang, C.Y. Tsai, M.K. Yeh, C.J. Wu, The https://doi.org/10.1016/j.msec.2017.03.191.
treatment of bladder cancer in a mouse model by epigallocatechin-3-gallate-gold [64] C.L. Dora, L.F. Silva, L. Mazzarino, J.M. Siqueira, D. Fernandes, L.K. Pacheco,
nanoparticles, Biomaterials 32 (30) (2011) 7633–7640, https://doi.org/10.1016/ M.F. Maioral, M.C. Santos-Silva, A.L. Baischl, J. Assreuy, E. Lemos-Senna, Oral
j.biomaterials.2011.06.073. delivery of a high quercetin payload nanosized emulsion: in vitro and in vivo
[45] D.S. Hsieh, H.C. Lu, C.C. Chen, C.J. Wu, M.K. Yeh, The preparation and char- activity against B16-F10 melanoma, J. Nanosci. Nanotechnol. 16 (2) (2016)
acterization of gold-conjugated polyphenol nanoparticles as a novel delivery 1275–1281.
system, Int. J. Nanomedicine 7 (2012) 1623–1633, https://doi.org/10.2147/IJN. [65] B.J. Tan, Y. Liu, K.L. Chang, B.K. Lim, G.N. Chiu, Perorally active nanomicellar
S30060. formulation of quercetin in the treatment of lung cancer, Int. J. Nanomed. 7
[46] I.A. Siddiqui, V.M. Adhami, D.J. Bharali, B.B. Hafeez, M. Asim, S.I. Khwaja, (2012) 651–661, https://doi.org/10.2147/IJN.S26538.
N. Ahmad, H. Cui, S.A. Mousa, H. Mukhtar, Introducing nanochemoprevention as [66] Q. Wu, Q. Long, Y. Xiel, Y. Huang, H. Zhang, S. Xiong, Y. Liu, L. Chen, Y. Wei,

13
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

X. Zhao, C. Gong, Induction of apoptosis and inhibition of angiogenesis by (-)-epigallocatechin-3-gallate as drug delivery system for breast Cancer therapy in
PEGylated liposomal quercetin in both cisplatin-sensitive and cisplatin-resistant vivo, ACS Appl. Mater. Interfaces 7 (32) (2015) 18145–18155, https://doi.org/10.
ovarian cancers, J. Biomed. Nanotechnol. 9 (6) (2013) 965–975. 1021/acsami.5b05618.
[67] R. Munagala, F. Aqil, J. Jeyabalan, A.K. Agrawal, A.M. Mudd, A.H. Kyakulaga, [87] V. Sanna, C.K. Singh, R. Jashari, V.M. Adhami, J.C. Chamcheu, I. Rady, M. Sechi,
I.P. Singh, M.V. Vadhanam, R.C. Gupta, Exosomal formulation of anthocyanidins H. Mukhtar, I.A. Siddiqui, Targeted nanoparticles encapsulating (-)-epigalloca-
against multiple cancer types, Cancer Lett. 393 (2017) 94–102, https://doi.org/ techin-3-gallate for prostate cancer prevention and therapy, Sci. Rep. 7 (2017)
10.1016/j.canlet.2017.02.004. 41573, https://doi.org/10.1038/srep41573.
[68] S. Chaurasia, R.R. Patel, P. Vure, B. Mishra, Oral naringenin nanocarriers: fabri- [88] R. Shukla, N. Chanda, A. Zambre, A. Upendran, K. Katti, R.R. Kulkarni, S.K. Nune,
cation, optimization, pharmacokinetic and chemotherapeutic efficacy assess- S.W. Casteel, C.J. Smith, J. Vimal, E. Boote, J.D. Robertson, P. Kan,
ments, Nanomedicine 12 (11) (2017) 1243–1260, https://doi.org/10.2217/nnm- H. Engelbrecht, L.D. Watkinson, T.L. Carmack, J.R. Lever, C.S. Cutler, C. Caldwell,
2016-0436. R. Kannan, K.V. Katti, Laminin receptor specific therapeutic gold nanoparticles
[69] S. Chaurasia, R.R. Patel, P. Vure, B. Mishra, Potential of cationic-polymeric na- (198AuNP-EGCg) show efficacy in treating prostate cancer, Proc. Natl. Acad. Sci.
noparticles for oral delivery of naringenin: in vitro and in vivo investigations, J. U. S. A. 109 (31) (2012) 12426–12431, https://doi.org/10.1073/pnas.
Pharm. Sci. 107 (2) (2018) 706–716, https://doi.org/10.1016/j.xphs.2017.10. 1121174109.
006. [89] Y. Zhou, Q. Yu, X. Qin, D. Bhavsar, L. Yang, Q. Chen, W. Zheng, L. Chen, J. Liu,
[70] N. Krishnakumar, N.K. Sulfikkarali, S. Manoharan, P. Venkatachalam, Raman Improving the anticancer efficacy of laminin receptor-specific therapeutic ruthe-
spectroscopic investigation of the chemopreventive response of naringenin and its nium nanoparticles (RuBB-loaded EGCG-RuNPs) via ROS-dependent apoptosis in
nanoparticles in DMBA-induced oral carcinogenesis, Spectrochim. Acta A. Mol. SMMC-7721 cells, ACS Appl. Mater. Interfaces 8 (24) (2016) 15000–15012,
Biomol. Spectrosc. 115 (2013) 648–653, https://doi.org/10.1016/j.saa.2013.05. https://doi.org/10.1021/acsami.5b02261.
076. [90] Z. Chen, T. Yu, B. Zhou, J. Wei, Y. Fang, J. Lu, L. Guo, W. Chen, Z.P. Liu, J. Luo, Mg
[71] N. Sulfikkarali, N. Krishnakumar, S. Manoharan, R.M. Nirmal, Chemopreventive (II)-Catechin nanoparticles delivering siRNA targeting EIF5A2 inhibit bladder
efficacy of naringenin-loaded nanoparticles in 7,12-dimethylbenz(a)anthracene cancer cell growth in vitro and in vivo, Biomaterials 81 (2016) 125–134, https://
induced experimental oral carcinogenesis, Pathol. Oncol. Res. 19 (2) (2013) doi.org/10.1016/j.biomaterials.2015.11.022.
287–296, https://doi.org/10.1007/s12253-012-9581-1. [91] J.E. Chung, S. Tan, S.J. Gao, N. Yongvongsoontorn, S.H. Kim, J.H. Lee, H.S. Choi,
[72] D. Majumdar, K.H. Jung, H. Zhang, S. Nannapaneni, X. Wang, A.R. Amin, Z. Chen, H. Yano, L. Zhuo, M. Kurisawa, J.Y. Ying, Self-assembled micellar nanocomplexes
Z.G. Chen, D.M. Shin, Luteolin nanoparticle in chemoprevention: in vitro and in comprising green tea catechin derivatives and protein drugs for cancer therapy,
vivo anticancer activity, Cancer Prev. Res. 7 (1) (2014) 65–73, https://doi.org/10. Nat. Nanotechnol. 9 (11) (2014) 907–912, https://doi.org/10.1038/nnano.2014.
1158/1940-6207.CAPR-13-0230. 208.
[73] S. Majumdar, S. Bhattacharya, L. Mondal, B. Mukherjee, L. Dutta, I. Ehsan, [92] A. Sarkar, S. Ghosh, S. Chowdhury, B. Pandey, P.C. Sil, Targeted delivery of
M.C. Debnath, R.H. Gaonkar, M.M. Pal, Apigenin loaded nanoparticle delayed quercetin loaded mesoporous silica nanoparticles to the breast cancer cells,
development of hepatocellular carcinoma in rats, Nanomedicine 14 (6) (2018) Biochimica et Biophysica Acta 1860 (10) (2016) 2065–2075, https://doi.org/10.
1905–1917, https://doi.org/10.1016/j.nano.2018.05.011. 1016/j.bbagen.2016.07.001.
[74] S. Das, J. Das, A. Samadder, A. Paul, A.R. Khuda-Bukhsh, Efficacy of PLGA-loaded [93] W. Wang, M. Xi, X. Duan, Y. Wang, F. Kong, Delivery of baicalein and paclitaxel
apigenin nanoparticles in Benzo[a]pyrene and ultraviolet-B induced skin cancer of using self-assembled nanoparticles: synergistic antitumor effect in vitro and in
mice: mitochondria mediated apoptotic signalling cascades, Food Chem. Toxicol. vivo, Int. J. Nanomed. 10 (2015) 3737–3750, https://doi.org/10.2147/IJN.
62 (2013) 670–680, https://doi.org/10.1016/j.fct.2013.09.037. S80297.
[75] H. Zhang, G. Liu, X. Zeng, Y. Wu, C. Yang, L. Mei, Z. Wang, L. Huang, Fabrication [94] K.H. Bae, S. Tan, A. Yamashita, W.X. Ang, S.J. Gao, S. Wang, J.E. Chung,
of genistein-loaded biodegradable TPGS-b-PCL nanoparticles for improved ther- M. Kurisawa, Hyaluronic acid-green tea catechin micellar nanocomplexes: fail-safe
apeutic effects in cervical cancer cells, Int. J. Nanomed. 10 (2015) 2461–2473, cisplatin nanomedicine for the treatment of ovarian cancer without off-target
https://doi.org/10.2147/IJN.S78988. toxicity, Biomaterials 148 (2017) 41–53, https://doi.org/10.1016/j.biomaterials.
[76] B. Wu, Y. Liang, Y. Tan, C. Xie, J. Shen, M. Zhang, X. Liu, L. Yang, F. Zhang, L. Liu, 2017.09.027.
S. Cai, Huai, D. Zheng, R. Zhang, C. Zhang, K. Chen, X. Tang, X. Sui, Genistein- [95] Q. Liu, J. Li, G. Pu, F. Zhang, H. Liu, Y. Zhang, Co-delivery of baicalein and
loaded nanoparticles of star-shaped diblock copolymer mannitol-core PLGA-TPGS doxorubicin by hyaluronic acid decorated nanostructured lipid carriers for breast
for the treatment of liver cancer, Mater. Sci. Eng. C Mater. Biol. Appl. 59 (2016) cancer therapy, Drug Deliv. 23 (4) (2016) 1364–1368, https://doi.org/10.3109/
792–800, https://doi.org/10.1016/j.msec.2015.10.087. 10717544.2015.1031295.
[77] D. Jeong, B.C. Bae, S.J. Park, K. Na, Reactive oxygen species responsive drug re- [96] J. Fang, S. Zhang, X. Xue, X. Zhu, S. Song, B. Wang, L. Jiang, M. Qin, H. Liang,
leasing nanoparticle based on chondroitin sulfate-anthocyanin nanocomplex for L. Gao, Quercetin and doxorubicin co-delivery using mesoporous silica nano-
efficient tumor therapy, J. Control. Release 222 (2016) 78–85, https://doi.org/10. particles enhance the efficacy of gastric carcinoma chemotherapy, Int. J.
1016/j.jconrel.2015.12.009. Nanomedicine 13 (2018) 5113–5126, https://doi.org/10.2147/IJN.S170862.
[78] T. Ramasamy, H.B. Ruttala, N. Chitrapriya, B.K. Poudal, J.Y. Choi, S.T. Kim, [97] S. Li, L. Wang, N. Li, Y. Liu, H. Su, Combination lung cancer chemotherapy: design
Y.S. Youn, S.K. Ku, H.G. Choi, C.S. Yong, J.O. Kim, Engineering of cell micro- of a pH-sensitive transferrin-PEG-Hz-lipid conjugate for the co-delivery of doc-
environment-responsive polypeptide nanovehicle co-encapsulating a synergistic etaxel and baicalin, Biomed. Pharmacother. 95 (2017) 548–555, https://doi.org/
combination of small molecules for effective chemotherapy in solid tumors, Acta 10.1016/j.biopha.2017.08.090.
Biomater. 48 (2017) 131–143, https://doi.org/10.1016/j.actbio.2016.10.034. [98] L. Lv, C. Liu, C. Chen, X. Yu, G. Chen, Y. Shi, F. Qin, J. Ou, K. Qiu, G. Li, Quercetin
[79] B. Liao, H. Ying, C. Yu, Z. Fan, W. Zhang, J. Shi, H. Ying, N. Ravichandran, Y. Xu, and doxorubicin co-encapsulated biotin receptor-targeting nanoparticles for
J. Yin, Y. Jiang, Q. Du, (-)-Epigallocatechin gallate (EGCG)-nanoethosomes as a minimizing drug resistance in breast cancer, Oncotarget 7 (22) (2016)
transdermal delivery system for docetaxel to treat implanted human melanoma 32184–32199, https://doi.org/10.18632/oncotarget.8607.
cell tumors in mice, Int. J. Pharm. 512 (1) (2016) 22–31, https://doi.org/10. [99] C. Murugan, K. Rayappan, R. Thangam, R. Bhanumathi, K. Shanthi, R. Vivek,
1016/j.ijpharm.2016.08.038. R. Thirumurugan, A. Bhattacharyya, S. Sivasubramanian, P. Gunasekaran,
[80] L. Meng, X. Xia, Y. Yang, J. Ye, W. Dong, P. Ma, Y. Jin, Y. Liu, Co-encapsulation of S. Kannan, Combinatorial nanocarrier based drug delivery approach for amalga-
paclitaxel and baicalein in nanoemulsions to overcome multidrug resistance via mation of anti-tumor agents in breast cancer cells: an improved nanomedicine
oxidative stress augmentation and P-glycoprotein inhibition, Int. J. Pharm. 513 strategy, Sci. Rep. 6 (2016) 34053, https://doi.org/10.1038/srep34053.
(1–2) (2016) 8–16, https://doi.org/10.1016/j.ijpharm.2016.09.001. [100] R. Radhakrishnan, H. Kulhari, D. Pooja, S. Gudem, S. Bhargava, R. Shukla,
[81] L.P. Mendes, M.P. Gaeti, P.H. De Avila, M. De Sousa Vieira, B. Dos Santos R. Sistla, Encapsulation of biophenolic phytochemical EGCG within lipid nano-
Rodrigues, R.I. De Avila Marcelino, L.C. Dos Santos, M.C. Valadares, E.M. Lima, particles enhances its stability and cytotoxicity against cancer, Chem. Phys. Lipids
Multicompartimental nanoparticles for co-encapsulation and multimodal drug 198 (2016) 51–60, https://doi.org/10.1016/j.chemphyslip.2016.05.006.
delivery to tumor cells and neovasculature, Pharm. Res. 31 (5) (2014) 1106–1119, [101] N. Di Leo, M. Battaglini, L. Berger, M. Giannaccini, L. Dente, S. Hampel,
https://doi.org/10.1007/s11095-013-1234-x. O. Vittorio, G. Cirillo, V. Raffa, A catechin nanoformulation inhibits WM266
[82] P. Tang, Q. Sun, H. Yang, B. Tang, H. Pu, H. Li, Honokiol nanoparticles based on melanoma cell proliferation, migration and associated neo-angiogenesis, Eur. J.
epigallocatechin gallate functionalized chitin to enhance therapeutic effects Pharm. Biopharm. 114 (2017) 1–10, https://doi.org/10.1016/j.ejpb.2016.12.024.
against liver cancer, Int. J. Pharm. 545 (1–2) (2018) 74–83, https://doi.org/10. [102] L. Luo, L. Zeng, J. Yan, M. Ma, H. Zhu, Preparation and characterization of
1016/j.ijpharm.2018.04.060. (-)-Epigallocatechin-3-gallate (EGCG)-loaded nanoparticles and their inhibitory
[83] B. Zhu, L. Yu, Q. Yue, Co-delivery of vincristine and quercetin by nanocarriers for effects on Human breast cancer MCF-7 cells, Sci. Rep. 7 (2017) 45521, https://doi.
lymphoma combination chemotherapy, Biomed. Pharmacother. 91 (2017) org/10.1038/srep45521.
287–294, https://doi.org/10.1016/j.biopha.2017.02.112. [103] T. Zou, S.S. Percival, Q. Cheng, Z. Li, C.A. Rowe, L. Gu, Preparation, character-
[84] A.K. Jain, K. Thanki, S. Jain, Co-encapsulation of tamoxifen and quercetin in ization, and induction of cell apoptosis of cocoa procyanidins-gelatin-chitosan
polymeric nanoparticles: implications on oral bioavailability, antitumor efficacy, nanoparticles, Eur. J. Pharm. Biopharm. 82 (1) (2012) 36–42, https://doi.org/10.
and drug-induced toxicity, Mol. Pharm. 10 (9) (2013) 3459–3474, https://doi. 1016/j.ejpb.2012.05.006.
org/10.1021/mp400311j. [104] R.C. De Pace, X. Liu, M. Sun, S. Nie, J. Zhang, Q. Cai, W. Gao, X. Pan, Z. Fan,
[85] P.S. Sandhu, R. Kumar, S. Beg, S. Jain, V. Kushwah, O.P. Katare, B. Singh, Natural S. Wang, Anticancer activities of (-)-epigallocatechin-3-gallate encapsulated na-
lipids enriched self-nano-emulsifying systems for effective co-delivery of tamox- noliposomes in MCF7 breast cancer cells, J. Liposome Res. 23 (3) (2013) 187–196,
ifen and naringenin: systematic approach for improved breast cancer therapeutics, https://doi.org/10.3109/08982104.2013.788023.
Nanomedicine 13 (5) (2017) 1703–1713, https://doi.org/10.1016/j.nano.2017. [105] S. Rocha, R. Generalov, C. Pereira Mdo, I. Peres, P. Juzenas, M.A. Coelho,
03.003. Epigallocatechin gallate-loaded polysaccharide nanoparticles for prostate cancer
[86] J. Xue, J. Ding, J. Yao, R. Li, B. Bao, L. Jiang, J.J. Zhu, Z. He, Tumor-homing cell- chemoprevention, Nanomedicine 6 (1) (2011) 79–87, https://doi.org/10.2217/
penetrating peptide linked to colloidal mesoporous silica encapsulated nnm.10.101.

14
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

[106] J. Liang, F. Li, Y. Fang, W. Yang, X. An, L. Zhao, Z. Xin, L. Cao, Q. Hu, Cytotoxicity 1016/j.jcis.2014.08.064.
and apoptotic effects of tea polyphenol-loaded chitosan nanoparticles on human [128] J. Varshosaz, A. Jafarian, G. Salehi, B. Zolfaghari, Comparing different sterol
hepatoma HepG2 cells, Mater. Sci. Eng. C Mater. Biol. Appl. 36 (2014) 7–13, containing solid lipid nanoparticles for targeted delivery of quercetin in hepato-
https://doi.org/10.1016/j.msec.2013.11.039. cellular carcinoma, J. Liposome Res. 24 (3) (2014) 191–203, https://doi.org/10.
[107] A. Dube, J.A. Nicolazzo, I. Larson, Chitosan nanoparticles enhance the intestinal 3109/08982104.2013.868476.
absorption of the green tea catechins (+)-catechin and (-)-epigallocatechin gal- [129] M.K. Abdelhalim, H.A. Qaid, Y. Al-Mohy, M.S. Al-Ayed, Effects of quercetin and
late, Eur. J. Pharm. Sci. 41 (2) (2010) 219–225, https://doi.org/10.1016/j.ejps. arginine on the nephrotoxicity and lipid peroxidation induced by gold nano-
2010.06.010. particles in vivo, Int. J. Nanomed. 13 (2018) 7765–7770, https://doi.org/10.
[108] A. Dube, J.A. Nicolazzo, I. Larson, Chitosan nanoparticles enhance the plasma 2147/IJN.S183281.
exposure of (-)-epigallocatechin gallate in mice through an enhancement in in- [130] X. Shi, X. Zhu, X. Zeng, X. Zhang, W. Cao, Y. Wang, H. Chen, T. Wang, H.I. Tsai,
testinal stability, Eur. J. Pharm. Sci. 44 (3) (2011) 422–426, https://doi.org/10. R. Zhang, D. Chang, S. He, L. Mei, The effects of quercetin-loaded PLGA-TPGS
1016/j.ejps.2011.09.004. nanoparticles on ultraviolet B-induced skin damages in vivo, Nanomedicine 12 (3)
[109] J. Cao, J. Hu, D. Webster, A. Shao, The safety of green tea and green tea extract (2016) 623–632, https://doi.org/10.1016/j.nano.2015.10.016.
consumption in adults – results of a systematic review, Regul. Toxicol. Pharmacol. [131] W. Nan, L. Ding, H. Chen, F.U. Khan, L. Yu, X. Sui, X. Shi, Topical use of quercetin-
95 (2018) 412–433, https://doi.org/10.1016/j.yrtph.2018.03.019. loaded chitosan nanoparticles against ultraviolet B radiation, Front. Pharmacol. 9
[110] A.M. Dostal, H. Samavat, S. Bedell, C. Torkelson, R. Wang, K. Swenson, C. Le, (2018) 826, https://doi.org/10.3389/fphar.2018.00826.
A.H. Wu, G. Ursin, J.M. Yuan, M.S. Kurzer, The safety of green tea extract sup- [132] E.M. Shehata, Y.S. Elnaggar, S. Galal, O.Y. Abdallah, Self-emulsifying phospho-
plementation in postmenopausal women at risk for breast cancer: results of the lipid pre-concentrates (SEPPs) for improved oral delivery of the anti-cancer gen-
Minnesota Green Tea Trial, Food Chem. Toxicol. 83 (2015) 26–35, https://doi. istein: development, appraisal and ex-vivo intestinal permeation, Int. J. Pharm.
org/10.1016/j.fct.2015.05.019. 511 (2) (2016) 745–756, https://doi.org/10.1016/j.ijpharm.2016.07.078.
[111] A. Hassan, R.J. Fontana, Liver injury associated with sporting activities, Semin. [133] J.F. Qiu, X. Gao, B.L. Wang, X.W. Wei, M.L. Gou, K. Men, X.Y. Liu, G. Guo,
Liver Dis. 38 (4) (2018) 357–365, https://doi.org/10.1055/s-0038-1670656. Z.Y. Qian, M.J. Huang, Preparation and characterization of monomethoxy poly
[112] Z. Bedrood, M. Rameshrad, H. Hosseinzadeh, Toxicological effects of Camellia (ethylene glycol)-poly(epsilon-caprolactone) micelles for the solubilization and in
sinensis (green tea): a review, Phytother. Res. 32 (7) (2018) 1163–1180, https:// vivo delivery of luteolin, Int. J. Nanomed. 8 (2013) 3061–3069, https://doi.org/
doi.org/10.1002/ptr.6063. 10.2147/IJN.S45062.
[113] G. Mazzanti, A. Di Sotto, A. Vitalone, Hepatotoxicity of green tea: an update, Arch. [134] S. Das, J. Das, A. Samadder, A. Paul, A.R. Khuda-Bukhsh, Strategic formulation of
Toxicol. 89 (8) (2015) 1175–1191, https://doi.org/10.1007/s00204-015-1521-x. apigenin-loaded PLGA nanoparticles for intracellular trafficking, DNA targeting
[114] K. Kulandaivelu, A.K. Mandal, Positive regulation of biochemical parameters by and improved therapeutic effects in skin melanoma in vitro, Toxicol. Lett. 223 (2)
tea polyphenol encapsulated solid lipid nanoparticles at in vitro and in vivo (2013) 124–138, https://doi.org/10.1016/j.toxlet.2013.09.012.
conditions, IET Nanobiotechnol. 10 (6) (2016) 419–424, https://doi.org/10.1049/ [135] L. Mary Lazer, B. Sadhasivam, K. Palaniyandi, T. Muthuswamy, I. Ramachandran,
iet-nbt.2015.0113. A. Balakrishnan, S. Pathak, S. Narayan, S. Ramalingam, Chitosan-based nano-
[115] S. Kumar, R. Meena, P. Rajamani, Fabrication of BSA-green tea polyphenols- formulation enhances the anticancer efficacy of hesperetin, Int. J. Biol. Macromol.
chitosan nanoparticles and their role in radioprotection: a molecular and bio- 107 (Pt B) (2018) 1988–1998, https://doi.org/10.1016/j.ijbiomac.2017.10.064.
chemical approach, J. Agric. Food Chem. 64 (30) (2016) 6024–6034, https://doi. [136] H.Y. Si, D.P. Li, T.M. Wang, H.L. Zhang, F.Y. Ren, Z.G. Xu, Y.Y. Zhao, Improving
org/10.1021/acs.jafc.6b02068. the anti-tumor effect of genistein with a biocompatible superparamagnetic drug
[116] L. Xiao, M. Mertens, L. Wortmann, S. Kremer, M. Valldor, T. Lammers, F. Kiessling, delivery system, J. Nanosci. Nanotechnol. 10 (4) (2010) 2325–2331.
S. Mathur, Enhanced in vitro and in vivo cellular imaging with green tea coated [137] J. Pham, O. Grundmann, T. Elbayoumi, Mitochondriotropic nanoemulsified gen-
water-soluble iron oxide nanocrystals, ACS Appl. Mater. Interfaces 7 (12) (2015) istein-loaded vehicles for cancer therapy, Methods Mol. Biol. 1265 (2015) 85–101,
6530–6540, https://doi.org/10.1021/am508404t. https://doi.org/10.1007/978-1-4939-2288-8_7.
[117] X. Yuan, Y. He, G. Zhou, X. Li, A. Feng, W. Zheng, Target challenging-cancer drug [138] E.U. Stolarczyk, K. Stolarczyk, M. Laszcz, M. Kubiszewski, W. Maruszak,
delivery to gastric cancer tissues with a fucose graft epigallocatechin-3-gallate- W. Olejarz, D. Bryk, Synthesis and characterization of genistein conjugated with
gold particles nanocomposite approach, J. Photochem. Photobiol. B Biol. 183 gold nanoparticles and the study of their cytotoxic properties, Eur. J. Pharm. Sci.
(2018) 147–153, https://doi.org/10.1016/j.jphotobiol.2018.04.026. 96 (2017) 176–185, https://doi.org/10.1016/j.ejps.2016.09.019.
[118] S. Kondath, B. Srinivas Raghavan, R. Anantanarayanan, R. Rajaram, Synthesis and [139] Y.S. Elnaggar, M.A. Elsheikh, O.Y. Abdallah, Phytochylomicron as a dual nano-
characterisation of morin reduced gold nanoparticles and its cytotoxicity in MCF-7 carrier for liver cancer targeting of luteolin: in vitro appraisal and pharmacody-
cells, Chem. Biol. Interact. 224 (2014) 78–88, https://doi.org/10.1016/j.cbi.2014. namics, Nanomedicine 13 (2) (2018) 209–232, https://doi.org/10.2217/nnm-
09.025. 2017-0220.
[119] M. Colombo, F. Figueiro, A. De Fraga Dias, H.F. Teixeira, A.M.O. Battastini, [140] O. Yesil-Celiktas, C. Pala, E.O. Cetin-Uyanikgil, C. Sevimli-Gur, Synthesis of silica-
L.S. Koester, Kaempferol-loaded mucoadhesive nanoemulsion for intranasal ad- PAMAM dendrimer nanoparticles as promising carriers in Neuro blastoma cells,
ministration reduces glioma growth in vitro, Int. J. Pharm. 543 (1–2) (2018) Anal. Biochem. 519 (2017) 1–7, https://doi.org/10.1016/j.ab.2016.12.004.
214–223, https://doi.org/10.1016/j.ijpharm.2018.03.055. [141] T. Liang, R. Guan, H. Shen, Q. Xia, M. Liu, Optimization of conditions for cyanidin-
[120] H. Luo, B. Jiang, B. Li, Z. Li, B.H. Jiang, Y.C. Chen, Kaempferol nanoparticles 3-OGlucoside (C3G) nanoliposome production by response surface methodology
achieve strong and selective inhibition of ovarian cancer cell viability, Int. J. and cellular uptake studies in Caco-2 cells, Molecules 22 (3) (2017), https://doi.
Nanomedicine 7 (2012) 3951–3959, https://doi.org/10.2147/IJN.S33670. org/10.3390/molecules22030457.
[121] Q. Wang, Y. Bao, J. Ahire, Y. Chao, Co-encapsulation of biodegradable nano- [142] S.P. Kumar, K. Birundha, K. Kaveri, K.T. Devi, Antioxidant studies of chitosan
particles with silicon quantum dots and quercetin for monitored delivery, Adv. nanoparticles containing naringenin and their cytotoxicity effects in lung cancer
Healthc. Mater. 2 (3) (2013) 459–466, https://doi.org/10.1002/adhm. cells, Int. J. Biol. Macromol. 78 (2015) 87–95, https://doi.org/10.1016/j.
201200178. ijbiomac.2015.03.045.
[122] G. Wang, J.J. Wang, X.L. Chen, S.M. Du, D.S. Li, Z.J. Pei, H. Lan, L.B. Wu, The [143] K. Gurushankar, S.S. Nazeer, R.S. Jayasree, N. Krishnakumar, Evaluation of anti-
JAK2/STAT3 and mitochondrial pathways are essential for quercetin nanolipo- tumor activity of hesperetin-loaded nanoparticles against DMBA-induced oral
some-induced C6 glioma cell death, Cell Death Dis. 4 (2013) e746, https://doi. carcinogenesis based on tissue autofluorescence spectroscopy and multivariate
org/10.1038/cddis.2013.242. analysis, J. Fluoresc. 25 (4) (2015) 931–939, https://doi.org/10.1007/s10895-
[123] G. Wang, J.J. Wang, G.Y. Yang, S.M. Du, N. Zeng, D.S. Li, R.M. Li, J.Y. Chen, 015-1575-4.
J.B. Feng, S.H. Yuan, F. Ye, Effects of quercetin nanoliposomes on C6 glioma cells [144] N. Krishnakumar, N.K. Sulfikkarali, S. Manoharan, R.M. Nirmal, Screening of
through induction of type III programmed cell death, Int. J. Nanomed. 7 (2012) chemopreventive effect of naringenin-loaded nanoparticles in DMBA-induced
271–280, https://doi.org/10.2147/IJN.S26935. hamster buccal pouch carcinogenesis by FT-IR spectroscopy, Mol. Cell. Biochem.
[124] F. Aghapour, A.A. Moghadamnia, A. Nicolini, S.N.M. Kani, L. Barari, 382 (1–2) (2013) 27–36, https://doi.org/10.1007/s11010-013-1715-6.
P. Morakabati, L. Rezazadeh, S. Kazemi, Quercetin conjugated with silica nano- [145] M. Singh, P. Bhatnagar, S. Mishra, P. Kumar, Y. Shukla, K.C. Gupta, PLGA-en-
particles inhibits tumor growth in MCF-7 breast cancer cell lines, Biochem. capsulated tea polyphenols enhance the chemotherapeutic efficacy of cisplatin
Biophys. Res. Commun. 500 (4) (2018) 860–865, https://doi.org/10.1016/j.bbrc. against human cancer cells and mice bearing Ehrlich ascites carcinoma, Int. J.
2018.04.174. Nanomedicine 10 (2015) 6789–6809, https://doi.org/10.2147/IJN.S79489.
[125] S. Das, S. Balakrishnan, S. Mukherjee, F.A. Bhat, P. Raja Singh, C.R. Patra, [146] D. Jeong, K. Na, Chondroitin sulfate based nanocomplex for enhancing the sta-
J. Arunakaran, Gold nanoparticles-conjugated quercetin induces apoptosis via bility and activity of anthocyanin, Carbohydr. Polym. 90 (1) (2012) 507–515,
inhibition of EGFR/PI3K/Akt-mediated pathway in breast cancer cell lines (MCF-7 https://doi.org/10.1016/j.carbpol.2012.05.072.
and MDA-MB-231), Cell Biochem. Funct. 35 (4) (2017) 217–231, https://doi.org/ [147] T. Cheng, J. Liu, J. Ren, F. Huang, H. Ou, Y. Ding, Y. Zhang, R. Ma, Y. An, J. Liu,
10.1002/cbf.3266. L. Shi, Green tea catechin-based complex micelles combined with doxorubicin to
[126] J. Suksiriworapong, K. Phoca, S. Ngamsom, K. Sripha, P. Moongkarndi, overcome cardiotoxicity and multidrug resistance, Theranostics 6 (9) (2016)
V.B. Junyaprasert, Comparison of poly(epsilon-caprolactone) chain lengths of poly 1277–1292, https://doi.org/10.7150/thno.15133.
(epsilon-caprolactone)-co-d-alpha-tocopheryl-poly(ethylene glycol) 1000 succi- [148] K. Liu, W. Chen, T. Yang, B. Wen, D. Ding, M. Keidar, J. Tang, W. Zhang, Paclitaxel
nate nanoparticles for enhancement of quercetin delivery to SKBR3 breast cancer and quercetin nanoparticles co-loaded in microspheres to prolong retention time
cells, Eur. J. Pharm. Biopharm. 101 (2016) 15–24, https://doi.org/10.1016/j.ejpb. for pulmonary drug delivery, Int. J. Nanomed. 12 (2017) 8239–8255, https://doi.
2016.01.008. org/10.2147/IJN.S147028.
[127] S.R. Kumar, S. Priyatharshni, V.N. Babu, D. Mangalaraj, C. Viswanathan, [149] S. Fatma, S. Talegaonkar, Z. Iqbal, A.K. Panda, L.M. Negi, D.G. Goswami, M. Tariq,
S. Kannan, N. Ponpandian, Quercetin conjugated superparamagnetic magnetite Novel flavonoid-based biodegradable nanoparticles for effective oral delivery of
nanoparticles for in-vitro analysis of breast cancer cell lines for chemotherapy etoposide by P-glycoprotein modulation: an in vitro, ex vivo and in vivo in-
applications, J. Colloid Interface Sci. 436 (2014) 234–242, https://doi.org/10. vestigations, Drug Deliv. 23 (2) (2016) 500–511, https://doi.org/10.3109/

15
P. Aiello, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

10717544.2014.923956. nanoparticles for imaging and drug delivery in colorectal cancer, Semin. Cancer
[150] M. Kumar, G. Sharma, C. Misra, R. Kumar, B. Singh, O.P. Katare, K. Raza, N- Biol. (2019), https://doi.org/10.1016/j.semcancer.2019.06.017.
Desmethyl tamoxifen and quercetin-loaded multiwalled CNTs: a synergistic ap- [159] N. Merchant, G.P. Nagaraju, B. Rajitha, S. Lammata, K.K. Jella, Z.S. Buchwald,
proach to overcome MDR in cancer cells, Mater. Sci. Eng. C Mater. Biol. Appl. 89 S.S. Lakka, A.N. Ali, Matrix metalloproteinases: their functional role in lung
(2018) 274–282, https://doi.org/10.1016/j.msec.2018.03.033. cancer, Carcinogenesis 38 (8) (2017) 766–780, https://doi.org/10.1093/carcin/
[151] V. Sanna, G. Pintus, A.M. Roggio, S. Punzoni, A.M. Posadino, A. Arca, bgx063.
S. Marceddu, P. Bandiera, S. Uzzau, M. Sechi, Targeted biocompatible nano- [160] R. De Oliveira Pedro, F.M. Goycoolea, S. Pereira, C.C. Schmitt, M.G. Neumann,
particles for the delivery of (-)-epigallocatechin 3-gallate to prostate cancer cells, Synergistic effect of quercetin and pH-responsive DEAE-chitosan carriers as drug
J. Med. Chem. 54 (5) (2011) 1321–1332, https://doi.org/10.1021/jm1013715. delivery system for breast cancer treatment, Int. J. Biol. Macromol. 106 (2018)
[152] S.M. Chiavenna, J.P. Jaworski, A. Vendrell, State of the art in anti-cancer mAbs, J. 579–586, https://doi.org/10.1016/j.ijbiomac.2017.08.056.
Biomed. Sci. 24 (1) (2017) 15, https://doi.org/10.1186/s12929-016-0311-y. [161] W. Li, M. Yalcin, Q. Lin, M.M. Ardawi, S.A. Mousa, Self-assembly of green tea
[153] R.I. El-Gogary, N. Rubio, J.T. Wang, W.T. Al-Jamal, M. Bourgognon, H. Kafa, catechin derivatives in nanoparticles for oral lycopene delivery, J. Control. Release
M. Naeem, R. Klippstein, V. Abbate, F. Leroux, S. Bals, G. Van Tendeloo, 248 (2017) 117–124, https://doi.org/10.1016/j.jconrel.2017.01.009.
A.O. Kamel, G.A. Awad, N.D. Mortada, K.T. Al-Jamal, Polyethylene glycol con- [162] P. Kuppusamy, M.M. Yusoff, G.P. Maniam, N. Govindan, Biosynthesis of metallic
jugated polymeric nanocapsules for targeted delivery of quercetin to folate-ex- nanoparticles using plant derivatives and their new avenues in pharmacological
pressing cancer cells in vitro and in vivo, ACS Nano 8 (2) (2014) 1384–1401, applications – an updated report, Saudi Pharm. J. 24 (4) (2016) 473–484, https://
https://doi.org/10.1021/nn405155b. doi.org/10.1016/j.jsps.2014.11.013.
[154] J. Li, J. Zhang, Y. Wang, X. Liang, Z. Wusiman, Y. Yin, Q. Shen, Synergistic in- [163] J.S. Park, E.Y. Ahn, Y. Park, Asymmetric dumbbell-shaped silver nanoparticles and
hibition of migration and invasion of breast cancer cells by dual docetaxel/quer- spherical gold nanoparticles green-synthesized by mangosteen (Garcinia man-
cetin-loaded nanoparticles via Akt/MMP-9 pathway, Int. J. Pharm. 523 (1) (2017) gostana) pericarp waste extracts, Int. J. Nanomed. 12 (2017) 6895–6908, https://
300–309, https://doi.org/10.1016/j.ijpharm.2017.03.040. doi.org/10.2147/IJN.S140190.
[155] L. Ray, P. Kumar, K.C. Gupta, The activity against Ehrlich’s ascites tumors of [164] S. Jain, T. Garg, V. Kushwah, K. Thanki, A.K. Agrawal, C.P. Dora, alpha-
doxorubicin contained in self assembled, cell receptor targeted nanoparticle with Tocopherol as functional excipient for resveratrol and coenzyme Q10-loaded
simultaneous oral delivery of the green tea polyphenol epigallocatechin-3-gallate, SNEDDS for improved bioavailability and prophylaxis of breast cancer, J. Drug
Biomaterials 34 (12) (2013) 3064–3076, https://doi.org/10.1016/j.biomaterials. Target. 25 (6) (2017) 554–565, https://doi.org/10.1080/1061186X.2017.
2012.12.044. 1298603.
[156] J. Li, S. Wu, C. Wu, L. Qiu, G. Zhu, C. Cui, Y. Liu, W. Hou, Y. Wang, L. Zhang, [165] I. Peluso, N.S. Yarla, R. Ambra, G. Pastore, G. Perry, MAPK signalling pathway in
I.T. Teng, H.H. Yang, W. Tan, Versatile surface engineering of porous nanoma- cancers: olive products as cancer preventive and therapeutic agents, Semin.
terials with bioinspired polyphenol coatings for targeted and controlled drug de- Cancer Biol. 56 (2019) 185–195, https://doi.org/10.1016/j.semcancer.2017.09.
livery, Nanoscale 8 (16) (2016) 8600–8606, https://doi.org/10.1039/ 002.
c6nr00600k. [166] G. Seeta Rama Raju, E. Pavitra, G.P. Nagaraju, K. Ramesh, B.F. El-Rayes, J.S. Yu,
[157] B. Rajitha, R.R. Malla, R. Vadde, P. Kasa, G.L.V. Prasad, B. Farran, S. Kumari, Imaging and curcumin delivery in pancreatic cancer cell lines using PEGylated
E. Pavitra, M.A. Kamal, G.S.R. Raju, S. Peela, G.P. Nagaraju, Horizons of nano- alpha-Gd2(MoO4)3 mesoporous particles, Dalton Trans. 43 (8) (2014)
technology applications in female specific cancers, Semin. Cancer Biol. (2019), 3330–3338, https://doi.org/10.1039/c3dt52692e.
https://doi.org/10.1016/j.semcancer.2019.07.005. [167] G.P. Nagaraju, L. Benton, S.R. Bethi, M. Shoji, B.F. El-Rayes, Curcumin analogs:
[158] E. Pavitra, B. Dariya, G. Srivani, S.M. Kang, A. Alam, P.R. Sudhir, M.A. Kamal, their roles in pancreatic cancer growth and metastasis, Int. J. Cancer 145 (1)
G.S.R. Raju, Y.K. Han, B. Lakkakula, G.P. Nagaraju, Y.S. Huh, Engineered (2019) 10–19, https://doi.org/10.1002/ijc.31867.

16

You might also like