ArticlePDF AvailableLiterature Review

Abstract and Figures

Breast cancer (BC) is the most common malignancy and the leading cause of cancer-related death in adult women worldwide. Over 85% of BC cases are non-hereditary, caused by modifiable extrinsic factors related to lifestyle, including dietary habits, which play a crucial role in cancer prevention. Although many epidemiological and observational studies have inversely correlated the fruit and vegetable consumption with the BC incidence, the involvement of their phenolic content in this correlation remains contradictory. During decades, wrong approaches that did not consider the bioavailability, metabolism, and breast tissue distribution of dietary phenolics persist behind the large currently existing gap between preclinical and clinical research. In the present review, we provide comprehensive preclinical and clinical evidence according to physiologically relevant in vitro and in vivo studies. Some dietary phenolics such as resveratrol (RSV), quercetin, isoflavones, epigallocatechin gallate (EGCG), lignans, and curcumin are gaining attention for their chemopreventive properties in preclinical research. However, the clinical evidence of dietary phenolics as BC chemopreventive compounds is still inconclusive. Therefore, the only way to validate promising preclinical results is to conduct clinical trials in BC patients. In this regard, future perspectives on dietary phenolics and BC research are also critically discussed.
Content may be subject to copyright.
International Journal of
Molecular Sciences
Review
Dietary Phenolics against Breast Cancer. A Critical
Evidence-Based Review and Future Perspectives
MaríaÁngeles Ávila-Gálvez, Juan Antonio Giménez-Bastida , Juan Carlos Espín * and
Antonio González-Sarrías *
Laboratory of Food and Health, Research Group on Quality, Safety and Bioactivity of Plant Foods,
Dept. Food Science and Technology, CEBAS-CSIC, P.O. Box 164, Campus de Espinardo, 30100 Murcia, Spain;
mavila@cebas.csic.es (M.Á.Á.-G.); jgbastida@cebas.csic.es (J.A.G.-B.)
*Correspondence: jcespin@cebas.csic.es (J.C.E.); agsarrias@cebas.csic.es (A.G.-S.)
Received: 21 July 2020; Accepted: 8 August 2020; Published: 10 August 2020


Abstract:
Breast cancer (BC) is the most common malignancy and the leading cause of cancer-related
death in adult women worldwide. Over 85% of BC cases are non-hereditary, caused by modifiable
extrinsic factors related to lifestyle, including dietary habits, which play a crucial role in cancer
prevention. Although many epidemiological and observational studies have inversely correlated
the fruit and vegetable consumption with the BC incidence, the involvement of their phenolic
content in this correlation remains contradictory. During decades, wrong approaches that did not
consider the bioavailability, metabolism, and breast tissue distribution of dietary phenolics persist
behind the large currently existing gap between preclinical and clinical research. In the present
review, we provide comprehensive preclinical and clinical evidence according to physiologically
relevant
in vitro
and
in vivo
studies. Some dietary phenolics such as resveratrol (RSV), quercetin,
isoflavones, epigallocatechin gallate (EGCG), lignans, and curcumin are gaining attention for their
chemopreventive properties in preclinical research. However, the clinical evidence of dietary phenolics
as BC chemopreventive compounds is still inconclusive. Therefore, the only way to validate promising
preclinical results is to conduct clinical trials in BC patients. In this regard, future perspectives on
dietary phenolics and BC research are also critically discussed.
Keywords: breast cancer; polyphenols; in vitro; animal models; clinical trials; dietary phenolics
1. Breast Cancer: General Aspects
Cancer is one of the world’s most substantial health problems and the second leading cause of
death globally, with 9.6 million cancer-related deaths in 2018 [
1
,
2
]. Among women, breast cancer (BC)
is the first cause of cancer-associated death and the most commonly diagnosed malignant tumour
worldwide, accounting for 2.1 million new BC diagnoses in 2018 [
3
]. Although the advances in
diagnosis, treatment and intensive research have significantly increased the survival rates among BC
subjects in the past years, its incidence has increased worldwide, especially in developing countries.
According to U.S. Breast Cancer Statistics, it is estimated that BC impacts nearly 1 in 8 women in their
lifetime [
4
]. Only 10%–15% of all BC cases are hereditary, meaning that the vast majority are caused
by modifiable risk factors, playing a central role in cancer prevention through lifestyle improvement,
including dietary habits [
5
,
6
]. Apart from the dietary and lifestyle habits, other related factors to
BC include age, ethnicity, hormonal status, hormonal therapy, ultraviolet radiation, and circadian
disruption [
7
]. Regarding genetic risk factors, some mutations such as breast cancer susceptibility
gene 1 (BCRA1), BCRA2, and TP53 are generally accepted, although these mutations are rare and only
constitute approximately 5%–10% of all BC rates incidences [5,8,9].
Int. J. Mol. Sci. 2020,21, 5718; doi:10.3390/ijms21165718 www.mdpi.com/journal/ijms
Int. J. Mol. Sci. 2020,21, 5718 2 of 33
BC is a compilation of distinct malignancies that usually begins in the terminal duct lobular unit
of the mammary gland and progresses in a stepwise manner by multiple molecular alterations. BC is
highly heterogeneous, encompassing from some cases with excellent prognosis to very aggressive
tumours, which usually develop over a long time [
10
,
11
]. Clinically, BC is classified into dierent
grades and types based on histological characteristics. The histological grade is a well-established
prognostic tool based on the degree of aggressiveness or dierentiation of the tumour tissue, including a
combined score for several parameters such as the microscopic evaluation of tubule or gland formation,
nuclear pleomorphism, and the mitotic count (i.e., determination of the proliferation marker Ki-67
by immunohistochemistry). Therefore, BCs with a high histological grade are generally large and
associated with metastasis, rather than those with low histological grade [
12
14
]. On the other hand,
the histological type is based on the growth pattern of the tumours including dierent subtypes,
presenting a higher incidence (around 75%), the so-called invasive or infiltrating ductal carcinomas
of no particular type, compared with other dierent subtypes such as invasive lobular carcinoma,
mucinous, tubular, medullary, etc. Thus, BCs with smaller tubular carcinomas are generally associated
with an earlier stage of the tumour, compared with invasive ductal carcinomas [
13
,
14
]. Besides, the TNM
is a well-accepted classification to designate the BC stage at the time of diagnosis refers to the size
and invasiveness of the tumour (T), lymph node involvement (N), and presence of distant metastasis
(M) [15].
Regarding molecular basis, BC is broadly categorized into dierent subtypes based on the
combination (presence or absence) of three receptors for oestrogens (ER), progesterone (PR) and
human epidermal growth factor receptor 2 (HER2). The dierential expression of these receptors
strongly determines the prognosis and therapeutic strategies against BC [
11
,
14
]. In this regard,
the incidence of hormone receptor-positive (ER+PR+) is the most common BC subtype, representing
around 70%. It exhibits a better prognosis with high survival and low recurrence rates, compared
with hormone receptor-negative tumours. Moreover, according to the elevated expression of HER2
and related genes, BC is further categorized into dierent molecular subtypes, such as luminal A
(ER+PR+HER2-) and(or) luminal B (ER+PR+HER2+). HER2+tumours represent a 15%–20% of
BC cases. Interestingly, although this type is more aggressive, associated with an increase in Ki-67
protein, it responds better to the current therapy resulting in high survival rates [
11
,
16
,
17
]. Finally,
the triple-negative BC for these three biomarkers (TNBC) accounts for about 15% of all BCs. It is
characterized by the worst prognosis involving high invasive properties, high relapse rate, and high
resistance to dierent therapies, what make this subtype highly intractable and invasive (mainly
with the presence of metastasis in other organs) [
11
,
18
]. Six molecular subtypes of TNBC have been
reported according to their gene ontologies and dierential gene expression in specific genes involved
in cell cycle, DNA damage response genes, androgen receptors, epidermal growth factor receptor, and
signalling pathways, among others. This highlights the significance of molecular heterogeneity in
BC [19].
Currently, beyond the prevention and early diagnosis, the therapeutic options for BC include
surgical resection, radiation, as well as chemotherapeutic supplementation according to the molecular
subtypes and the BC stage. Thus, for early-stage non-metastatic BC, surgical resection, together with
postoperative adjuvant chemotherapy, radiotherapy, and(or) hormone therapy depending on the
BC subtype, represent the main treatment options. On the other hand, treatments for metastatic BC
(about 20%–45%) generally consist of conventional chemotherapy, including cytotoxic drugs, such as
the anthracycline (doxorubicin), and(or) taxane (paclitaxel) families, combined with cyclophosphamide.
Other treatments include 5-fluorouracil, cisplatin derivatives, gemcitabine, or vinorelbine, as well as
monoclonal antibody therapy, such as bevacizumab that targets the vascular endothelial growth factor
receptor (VEGFR), what in turn inhibits angiogenesis processes [
11
,
20
,
21
]. Fortunately, the knowledge
of predictive biomarkers in the dierent BC subtypes has led to the use of combined treatments
with adjuvant chemotherapy, allowing for a personalized approach in each patient that increases
the eectiveness of the therapy. In this regard, the main treatments for hormone-dependent BC
Int. J. Mol. Sci. 2020,21, 5718 3 of 33
(ER+/PR+) combined with adjuvant chemotherapy are based on hormonal therapy, which is based
on the use of oestrogen antagonists, such as tamoxifen or fulvestrant and aromatase inhibitors like
anastrozole, letrozole, and exemestane [
22
]. In HER2 overexpressing tumours, the primary treatment
involves monoclonal antibodies such as trastuzumab and pertuzumab and specific HER2 pathway
inhibitors like lapatinib. This treatment could be combined with hormonal therapy, depending on
the hormone receptors’ expression [
20
,
21
,
23
]. As mentioned above, the lack of biomarkers expression
in TNBC is addressed through classical chemotherapy (anthracyclines, taxanes, platinum-containing
chemotherapeutic agents, etc.) as the unique therapeutic option [
11
,
20
,
21
]. However, adverse side
eects of these anticancer drugs alone or in combination are relatively frequent. Furthermore, the further
development of drug resistance processes leads to tumour recurrences after long-time administration.
Therefore, beyond the currently approved chemotherapeutics, natural products (including dietary
compounds) exhibit pleiotropic, multi-target activities and are emerging as possible complementary
chemopreventive molecules against BC with fewer side eects than conventional therapy [6,24].
2. Epidemiological Evidence for Dietary (Poly)Phenols
As stated above, it is well-established that lifestyle improvement, including dietary habits, is the
primary prevention strategy against cancer. Indeed, improper dietary habits (high amounts of fat,
sugar, red meat, and alcohol) may be behind 30%–35% of all the cancer types [
25
,
26
]. In contrast,
epidemiological and observational studies have described the inverse association between high intake of
plant foods and low BC risk [
27
,
28
], or high vegetable intake and low ER-BC risk [
29
,
30
]. Nevertheless,
several studies indicate that this inverse correlation for high consumption of fruit and vegetables
remains still inconsistent or contradictory [
6
,
29
,
31
], and limited evidence has been found according to
the recent European Prospective Investigation into Cancer and Nutrition (EPIC) data [31,32].
Over the past decades, meta-analyses, observational, and epidemiological studies have suggested
the potential role of dietary phenolics in the chemoprevention and chemosensitization in BC [
33
35
].
In this regard, significant protective eects against BC have been attributed to some dietary patterns
that involve high intake of phenolics-rich foods, mainly phytoestrogens that can bind to ER, such as
isoflavones in soy-based products in Asian countries [
36
38
]. Mediterranean dietary patterns implicate
higher intake of polyphenols than Western diets, especially in postmenopausal women and against
TNBC [39,40].
Many dietary phenolics have been reported to exert anticancer activities by playing pleiotropic
multi-target activities on BC cells and animal models related to cell viability, proliferation, dierentiation,
and invasion, as well as by modulating signalling pathways, BC markers, gene, and protein
expression, etc. [41,42]. However, human evidence remains still inconclusive and controversial.
3. Bioavailability of Dietary Phenolics and Their Occurrence in Human Breast Tissues
The bioavailability of dietary phenolics is an essential factor to consider, either increasing or limiting
the overall anticancer eects against systemic cancers such as BC. In line with this, understanding the
metabolic/molecular forms and concentrations of those phenolics that can reach the human breast
tissue, is crucial in order to suggest their possible role in the
in vivo
beneficial eects, as well as to
(re)design physiologically relevant
in vitro
studies, thus increasing the knowledge of their possible
chemopreventive mechanisms of action.
Without considering the food matrix and(or) food processing related factors, the rate and extent
of intestinal absorption of each phenolic compound depend on the chemical structure that determines
its solubility, membrane permeability, and its microbial and(or) phase-II metabolism [4345].
Dietary phenolics are usually characterized by their poor bioavailability, limiting their distribution
in systemic tissues in their native form, mainly as glycosides and complex oligomeric structures.
Once ingested, they are transformed into a variety of diverse bioavailable metabolites that can
remain in the systemic circulation for a few days [
46
]. Most phenolics are hydrolyzed and further
metabolized by either the intestinal enzymes or by the gut microbiota. After absorption, the resulting
Int. J. Mol. Sci. 2020,21, 5718 4 of 33
polyphenol-derived metabolites undergo extensive phase-II metabolism inside the enterocytes to form
sulphates, glucuronides, methyl-derivatives, or other conjugates. Therefore, in contrast to their parental
molecules that rarely appear in the systemic circulation, significant concentrations of phenolic-derived
conjugated metabolites can be detected in plasma and target systemic tissues where might trigger
biological eects, including anticancer activity [4648].
However, it was found that conjugated forms of phenolics or microbial metabolites (phase-II
derivatives) display much lower or even no anticancer eects than their corresponding precursor
unconjugated forms [
49
51
]. Therefore, to date, whether dietary phenolics may protect against the
progression of BC (despite the low bioavailability and high metabolism of these compounds) is unclear,
and several fundamental questions about their mechanism of action remain unanswered.
Regarding the occurrence of phenolic-derived metabolites in human breast tissue, the evidence
is limited to a low number of human intervention studies, in which the sample size is also small
(from 3 to 31 volunteers) and the characteristic of the volunteers are dierent between the studies
(Table 1). In contrast, the number of animal studies is higher, but the results reported are contradictory.
The methodologies used for quantifying the metabolites in most of these studies showed low accuracy
and should be considered with caution (Table S1).
Similar to animal studies, the most robust human evidence is reflected in investigations with
isoflavones. Four clinical studies, conducted in women undergoing aesthetic breast, have allowed us to
establish the identity and quantity of the isoflavone metabolites that can occur in human breast tissues
after the intake of soy-based foods. Overall, these studies showed that unconjugated daidzein, genistein,
and the daidzein microbial-derived metabolite, equol, reached concentrations in the low nanomolar
range, reporting higher levels for equol (in those equol-producer individuals) and daidzein than
genistein [
52
,
53
]. However, no significant dierences between daidzein and genistein were observed in
another study in women undergoing breast biopsy or cancer surgery [
54
]. Notwithstanding, it should
be noted that the breast tissues were hydrolyzed by enzymatic treatment in these studies. Thus,
the levels of these phenolics were overestimated, i.e., the unconjugated phenolics did not reach the
tissue as such, but the enzymatic treatment in the tissue generated them.
In this regard, Bolca et al. identified and quantified the distribution of isoflavones in non-hydrolyzed
and enzymatically hydrolyzed adipose and glandular breast tissues after the intake soy milk or
supplement by healthy women undergoing an aesthetic breast reduction [
55
]. As expected, the results
showed overall total glucuronidation of 98% (900–1150 pmol/g total isoflavone glucuronides), mainly
genistein-7-O-glucuronide and daidzein-7-O-glucuronide, whereas only traces of free genistein and
daidzein were observed (20–25 pmol/g), but not for the microbial daidzein-derived metabolites (equol or
O-DMA). However, after enzymatic hydrolysis, equol and dihydrodaidzein were also detected in a
few subjects, in addition to high levels of unconjugated daidzein and genistein.
According to this fact, in those animal studies where non-hydrolyzed samples were also analyzed
compared with hydrolyzed ones, isoflavones-derived metabolites were mostly detected (>60%) in
their physiologically conjugated forms, mostly glucuronides, and the unconjugated forms were even
undetectable in some cases (Table S1) [
56
58
]. Besides, it should be considered that only an animal
study reported the distribution in breast tumour tissues of isoflavones metabolites, and described
higher levels in tumour than in normal (liver and colon) tissues, for both total and free genistein,
and daidzein to a lesser extent [
59
]. Bolca et al., following the same approach and methodology,
also observed extensive glucuronidation (>90%) for prenylflavonoids in breast adipose/glandular
tissues [
60
]. However, low concentrations (picomolar, ranging from 0.8 to 83.7) of total isoxanthohumol
(IX), xanthohumol (XN) and 8-prenylnaringenin (8-PN, only in those 8-PN producers) were quantified
in hydrolyzed tissues from healthy women undergoing an aesthetic breast reduction after hop intake
for 5 days [
60
]. This result was in agreement with those reported in rodent models where 8-PN and
6-PN glucuronides were predominantly found in the mammary gland tissue after intraperitoneal
administration of prenylflavonoids or hop extract (Table S1) [61,62].
Int. J. Mol. Sci. 2020,21, 5718 5 of 33
In recent years, new human studies have further advanced in the identification of metabolites
in human breast tissue, through the combination of the analysis in hydrolyzed and non-hydrolyzed
samples and the evaluation of the distribution in both normal and malignant tissues. This fact is
especially relevant since breast tumour and normal tissues have been reported to show dierences in
phase-II enzymes expression such as lower and higher activities of UDP-glucuronosyltransferases and
glutathione S-transferases, respectively [
63
], as well as increased the
β
-glucuronidase activity when
exposed to inflammatory or hypoxic conditions within the tumour microenvironment [63,64].
Thus, in two pre-surgery non-controlled dietary interventions with silybin and tea extracts
conducted in patients with newly diagnosed BC, higher concentrations of total silybin and EGCG
were detected in tumour tissues than in the adjacent normal tissues. In both studies, similar to other
phenolics, the total concentration of conjugated forms was higher compared with free silybin and
EGCG (4- and 2-fold, respectively), suggesting a high rate of phase-II metabolism, although free EGCG
was not detected in tumour rather than normal tissues. However, the quantification of conjugates
was not performed due to the lack of conjugated standards [
65
,
66
]. Similar results (considering the
limitations of the conjugated forms quantification) related to the identification of methyl and sulphate
of catechins have been observed in two animal studies conducted in mice fed green tea extracts.
However, it is noteworthy that besides the higher quantification of EGCG, other catechins such as
epicatechin and epigallocatechin were also detected in hydrolyzed samples (Table S1) [67,68].
Finally, a recent pre-surgery-controlled intervention study with a higher number of BC patients
that consumed a cocktail of plant extracts (cocoa, pomegranate, lemon, orange, grapeseed, olive,
and RSV) identified a total of 39 and 33 metabolites in normal and tumour tissues, respectively.
However, although the qualitative and quantitative metabolic profiling of phenolics was quite similar
in normal and tumour tissues, higher levels were found for most phenolics in tumour tissues.In both
tissues, phenolic-derived metabolites, including RSV, urolithins, and hesperetin, showed levels in
the range of the low nM, and were mainly glucuronidated and(or) sulphated (>85%). Among these
conjugates, the percentage of sulphated was slightly lower in normal tissues (31%) than in tumour
(42%) [69].
More recently, the same authors tried to overcome one of the main limitations of these human
studies, i.e., the long pre-surgery fasting that could hamper: (i) the detection of other phenolic-derived
metabolites (including dierent metabolic forms), (ii) the occurrence of higher concentrations that
could be reached in the breast tissues without fasting period. In this study, conducted in rats fed
equivalent doses to those used in the human study, the analyses at shorter times described the presence
of conjugated, but not free metabolites, derived from RSV, hesperetin, urolithins. These results are
in agreement with the data described in the previous human study, although some dierences were
observed: higher concentrations (range of low
µ
M), a higher proportion of RSV sulphates than
glucuronides (opposite to what is observed in humans), as well as the occurrence of other metabolites
at shorter times such as hydroxytyrosol glucuronide and sulphate [
70
]. These results corroborate
some limitations of human studies (associated with hospital protocols) and confirm that human and
animal studies carried out under similar conditions contribute to increasing our knowledge on the
bioavailability of dietary phenolics.
Regarding other phenolics with high potential anticancer activity like curcumin (diferuloylmethane)
or lignans, the evidence is even more limited, with insucient studies. Only studies with rodent
models have detected their presence in breast tissues (Table S1). Like other phenolics, the accumulation
of free and conjugated gut microbiota-derived enterolignan metabolites, mainly enterolactone and
enterodiol glucuronides, has been identified in the mammary tissues of rats and pigs (Table S1) [
71
,
72
].
Int. J. Mol. Sci. 2020,21, 5718 6 of 33
Table 1. Dietary phenolics and derived metabolites identified in human breast tissues.
Group and Sample Size Diet/Compound Administration Extraction and Analytical Conditions Identified and(or) Quantified Phenolic Metabolites References
8 women undergoing breast
biopsy or cancer surgery
Intake of four soy-supplemented bread
rolls per day, providing approximately
45 mg of isoflavonoids, for 14 days prior to
surgery (n =4) Placebo group (n =4).
Breast cancer tissues were extracted and
further hydrolyzed by enzymatic
treatment 1. Quantitative analyses were
performed by GC/MS using specific
standards.
The mean daidzein concentration in
non-soy-supplemented patients was 0.021
(range 0.017–0.028) nmol/g compared with 0.145
(range 0.083–0.218) nmol/g in soy-supplemented patients.
[54]
3 healthy women
undergoing breast
reductions
Soy-based food supplement containing
100 mg of genistein/genistin, 37 mg of
daidzein/daidzin, and 15 mg of
glycitein/glycitin (more than 90% as
glycosides (single and triple dose, n =1
each) or a placebo tablet (n =1) for 5 days
before aesthetic breast surgery.
Breast tissues were extracted and further
hydrolyzed by enzymatic treatment 1.
Analyses were performed by HPLC-DAD
and the compounds were identified by
comparison of the retention time with the
respective standards (UV spectra).
Genistein and equol concentrations were 4.16 µg/g and
52.98
µ
g/g, respectively, after soy-based food supplement
(single dose), whereas daidzein was below the limit of
detection.
Genistein, equol and daidzein concentrations were
35.1 µg/g, 681.7 µg/g and 16.0 µg/g, respectively, after
soy-based food supplement (triple dose).
[52]
28 volunteers before
aesthetic breast surgery
Soy-based food supplement containing
100 mg of genistein/genistin, 37 mg of
daidzein/daidzin, and 15 mg of
glycitein/glycitin (more than 90% as
glycosides, for 5 evenings before aesthetic
breast surgery (n =9). Placebo group
(n =19).
Breast tissues were extracted and further
hydrolyzed by enzymatic treatment 1.
Analyses were performed by HPLC-MS
and the compounds were quantified using
specific standards.
The median daidzein and equol concentrations were
7.03 nmol/L and 2.44 nmol/L, respectively,
in soy-supplemented subjects. Genistein was not
detectable.
The median daidzein concentration was 5.44 nmol/L in
the placebo group. Equol and genistein were only
detectable in some subjects of the placebo group,
with equol ten-fold values higher than genistein.
No significant dierences were found between the
2 groups.
[53]
31 healthy women
undergoing an aesthetic
breast reduction
Soy milk (n =11; 250 mL containing
16.98 mg genistein and 5.40 mg daidzein
(per dose)), soy supplement (n =10;
5.27 mg genistein and 17.56 mg daidzein
aglycone (per dose), or control (n =10,
no soy product). 3 doses of soy milk or soy
supplements were taken daily for 5 days
before an aesthetic breast reduction.
Breast tissues were dissected into fractions
(adipose and glandular tissue) and were
both non-hydrolyzed and hydrolyzed by
enzymatic treatment 1. Quantitative
analyses were performed by
HPLC-MS/MS and the compounds were
quantified using specific standards of
aglycones, but not conjugated forms.
Total isoflavones showed a breast adipose/glandular
tissue distribution of 40:60. In hydrolyzed breast adipose
and glandular tissues, total genistein and daidzein
concentrations ranged between 92.33–493.8 pmol/g and
22.15–770.8 pmol/g. Total equol and dihydrodaidzein
were only detected in few subjects (up to 559.4 pmol/g
and up to 368.8 ±171.1 pmol/g.
In non-hydrolyzed breast adipose and glandular tissues
only traces of genistein and daidzein aglycones were
observed (20–25 pmol/g total isoflavone aglycones),
whereas an overall total glucuronidation of 98% was
estimated (900–1150 pmol/g total isoflavone
glucuronides), mainly genistein-7-O-glucuronide and
daidzein-7-O-glucuronide. Neither glucuronides nor
aglycones of microbial daidzein metabolites (equol and
O-DMA) were detected after isoflavone supplementation.
[55]
21 healthy women
undergoing an aesthetic
breast reduction
Hop-supplemented group (containing
2.04 mg xanthohumol (XN), 1.20 mg
isoxanthohumol (IX), and 0.1 mg
8-prenylnaringenin (8-PN) per
supplement) (n =11) or control group
(n =10). Three supplements were taken
daily for 5 days before surgery.
Breast tissues were dissected into fractions
(adipose and glandular tissue) and were
both non-hydrolyzed and hydrolyzed by
enzymatic treatment 1. Qualitative
analyses were performed by
HPLC-MS/MS and the compounds were
quantified using specific standards for
aglycones, but not conjugated forms.
Total prenylflavonoids showed a breast
adipose/glandular tissue distribution of 38:62.
Total XN and IX concentrations ranged between 0.26 and
5.14 pmol/g and 1.16 and 83.67 pmol/g in hydrolyzed
breast tissue, respectively. 8-PN was only detected in
samples of moderate and strong 8-PN producers
(0.78–4.83 pmol/g). An extensive glucuronidation was
observed (90%).
[60]
Int. J. Mol. Sci. 2020,21, 5718 7 of 33
Table 1. Cont.
Group and Sample Size Diet/Compound Administration Extraction and Analytical Conditions Identified and(or) Quantified Phenolic Metabolites References
12 early breast cancer
patients
Orally bioavailable complex of
silybin–phosphatidylcholine (2.8 g/day)
for 4 weeks prior to surgery.
Breast tissues (tumour and normal) were
extracted and further hydrolyzed both
with and without enzymatic treatment 1.
Qualitative analyses were performed by
HPLC-MS/MS and the free and total
silybin were quantified using specific
standard.
The median total and free silybin concentration in breast
cancer tissues were 131 ng/mg (IQR, 35–869) and 33 ng/g
(IQR, 4–158), respectively.
Concentrations were higher in the tumour as compared
with the adjacent normal tissue (total and free silybin
concentration were 11 ng/mL (IQR, 0–34) and 0 ng/g
(IQR, 0–19), respectively).
[65]
12 early breast cancer
patients
Patients received 300 mg of a caeine-free
green tea catechin extract, equivalent to
44.9 mg of epigallocatechin-3-O-gallate
(EGCG) daily, for 4 weeks prior to surgery.
Breast tissues (tumour and normal) were
extracted and further both
non-hydrolyzed and hydrolyzed by
enzymatic treatment 1. Quantitative
analyses were performed by
HPLC-MS/MS and the compounds were
quantified as free (unconjugated) EGCG,
thereafter through enzymatic hydrolysis
as total EGCG (free and conjugated) using
EGCG standard.
Total EGCG was detectable in all tumour tissue samples
(median total EGCG 3.18 ng/g (IQR, 2.76–4.58)) and
higher amount than in adjacent normal tissue (under the
limit of detectability, minimum, 0 ng/g; maximum,
2.85 ng/g).
Free EGCG concentrations were under the limit of
detectability in tumour tissue but present in adjacent
normal breast tissue (median =1.07 ng/g; IQR, 0–1.25).
[66]
27 breast cancer patients
Breast cancer patients consumed a cocktail
of plant extracts (cocoa, pomegranate,
lemon, orange, grapeseed, and olive) plus
resveratrol, providing 37 dierent
phenolics (473.7 mg), theobromine and
caeine (19.7 mg) (n =19) from diagnosis
to surgical resection (6 ±2 days). Control
group did not consume extracts (n =8).
Normal and tumour glandular breast
tissues were extracted and further both
non-hydrolyzed and hydrolyzed by
enzymatic treatment 1. Quantitative
analyses were performed by
UPLC-ESI-QTOF-MS and carried out by
peak area integration of their EIC using
calibration curves of specific (free and
conjugated metabolites) standards.
A total of 39 and 33 metabolites were identified in normal
and tumour tissues, respectively. Some representative
metabolites detected in tumour tissues (median and
range, pmol/g) were urolithin-A-3-O-glucuronide (26.2;
3.2–66.5), 2,5-dihydroxybenzoic acid (40.2; 27.7–52.2),
RSV-3-O-sulphate (86.4; 7.8–224.4),
dihydroRSV-3-O-glucuronide (109.9; 10.3–229.4), and HP
30-O-glucuronide 12.9 (2.7–14.1).
No significantly dierent conjugation profiling was
found in tumour vs. normal tissues. Overall,
all compounds were mostly glucuronidated and(or)
sulphated in tumour and normal tissues, 85.5% and
86.6%, respectively. Among these conjugates, the
percentage of sulphated was slightly higher in tumour
(42%) than in normal tissues (31%). Quantitative analysis
after hydrolysis was only possible for 2,5- and
2,6-dihydroxybenzoic acids, HP, urolithin A, isourolithin
A, and urolithin B.
[69]
1
enzymatic treatment was performed using
β
-glucuronidase/sulfatase enzymes. Abbreviations: DAD, diode array detection; EGCG, epigallocatechin-3-gallate; EIC, extracted
ion chromatogram; GC, gas chromatography; HP, Hesperetin; HPLC, high performance liquid chromatography; IQR, interquartile range; MS, mass spectrometry; O-DMA,
O-desmethylangolensin; RSV, resveratrol; UPLC-ESI-Q-TOF, ultra-performance liquid chromatography coupled to electrospray ionization quadrupole time-of-flight; UV, ultraviolet.
Int. J. Mol. Sci. 2020,21, 5718 8 of 33
Overall, based on the current evidence, the distribution of phenolic-derived metabolites in breast
tissue is similar to that observed in plasma, yet at lower concentrations. However, in contrast to most
phenolics, curcumin, beyond its poor bioavailability, remains a challenge. Thus, despite curcumin is
rapidly metabolized to conjugated forms, mainly glucuronides, recent animal studies have identified
free curcumin at significantly higher concentrations (up to 15-fold) than curcumin glucuronide in breast
tumour tissues from tumour-bearing mice models, after oral and intravenous administration [
73
].
Besides, similar to that observed for other phenolics, curcumin levels in healthy mammary tissues
were significantly lower than tumours tissues [
73
]. This is in agreement with another recent animal
study where the concentration of free curcumin in serum was lower than its glucuronide. However,
the absolute amount and the percentage of total curcumin was significantly increased in bones (up to
3
µ
M), whereas curcumin glucuronide was barely detectable due to the high glucuronidase activity [
74
].
At present, this fact has not been confirmed in human studies hitherto.
On the other hand, it is also important to note that recent preclinical studies have explored
(non-nutritional) strategies to increase polyphenols’ bioavailability and stability to overcome the fast
clearance of most phenolic-derived metabolites [
46
]. Dierent techniques are included within these
alternative approaches, such as encapsulation (micelles, liposomes, nanoparticles, etc.) and phenolic
loaded self-microemulsifying drug delivery system (SMEDDS) or self-nanoemulsifying drug delivery
system (SNEDDS), which are used for dierent phenolics (curcumin, quercetin, RSV, etc.) in both
bioavailability and chemopreventive animal studies (Tables S1 and S2). Similar approaches to increase
cytotoxicity and bioavailability have also been assayed in BC cell models [7577].
4. Evidence of the Chemopreventive Potential of Phenolic Compounds in Physiologically
Relevant Preclinical Studies
Following the statement “First
in vivo
and then
in vitro
” [
78
], we must be cautious with all the
accumulated evidence on
in vitro
polyphenols’ eects against BC. In this regard, innumerable
in vitro
studies have been carried out with doubtful physiological relevance, i.e., regardless of bioavailability,
metabolism, and tissue distribution of dietary phenolics or their derived metabolites. Therefore, in the
present review, we focus only on those physiologically relevant
in vitro
and
in vivo
studies conducted
with dietary phenolics that could elucidate whether they are responsible for the eects attributed to
plant-based foods.
In the last decades,
in vivo
animal studies have been an essential tool in investigating the
biological eects of natural compounds against BC. More than 200
in vivo
studies, included in this
review (Table S2), describe the eects of dierent phenolic compounds on BC. Isoflavones (genistein
and daidzein), as well as lignans (flaxseed and secoisolariciresinol diglucoside), encompass the highest
number of studies (almost 50%), whereas the eects of other classes, including anthocyanins or flavones
have been less investigated so far. In general, diets supplemented with phenolics (anthocyanins,
flavanones, flavones, flavonols, flavan-3-ols, RSV, curcumin, lignans, and hydrolysable tannins) were
associated with reduction of tumour growth, incidence (percentage of animals that develop breast
tumours), multiplicity (tumours per mouse), metastasis (mainly to lungs), and higher tumour latency
period (time between carcinogen exposure and tumour detection), which in turn are related to a higher
survival rate. However, these eects are less clear regarding the consumption of isoflavones.
A significant number of investigations challenges the reported evidence concerning the benefits
of soy products and isoflavones consumption in the prevention or treatment of BC. At least 15 studies
(out of 52 included in Table S2) described the absence of protection or even the increase of tumour
growth, incidence, metastasis, or reduction of tumour latency. These contradictory results have led the
food scientists to search for new approaches (i.e., a combination of dierent phenolic compounds) to
reduce the stimulatory eects of isoflavones on human breast tissue. In this regard, the combination
of flaxseed, enterolactone, and(or) enterodiol with soy protein or genistein has demonstrated to be
eective attenuating tumour growth promotion in animal models [79,80].
Int. J. Mol. Sci. 2020,21, 5718 9 of 33
On the other hand, the combination of phenolics is not limited to lignans-rich foods and isoflavones.
Dierent mixtures such as quercetin-RSV-genistein, curcumin-EGCG, and curcumin-RSV have been
shown to be eective in reducing tumour growth and modulating apoptosis and angiogenesis signalling
pathways [
81
83
]. In line with this, emerging strategies using self-nanoemulsifying drug delivery
systems (SNEDDS) showed higher plasma concentrations of mixtures of phenolics related to more
ecient anti-breast cancer eects [84].
Regarding the mechanism of action, the reduction on tumour growth associated with the
consumption of the dierent dietary phenolics has been reported to be closely related to higher
levels of apoptosis and lower cell proliferation and angiogenesis (together with the modulation of
molecular mechanisms associated). Additional mechanisms described in the numerous preclinical
studies included in Table S2 support the potential protective role of the phenolics against BC.
One of these mechanisms is the modulation of morphological changes in mammary tissue
structures. Isoflavones [
85
,
86
] and resveratrol [
87
] have shown the capacity to reduce the number of
terminal end buds (TEBs). The reduction of the TEBs density is considered a protective mechanism
since these structures are targets of carcinogens like DMBA. Thus, the reduction of tumour growth
in DMBA-treated rats fed grape juice concentrate was related to a decrease in the formation of
DMBA-DNA adducts [
88
]. RSV also exhibited protection through the reduction of lipid peroxidation
and DNA damage related to its antioxidant activity [
89
]. Further mechanisms by which phenolics exert
their eects on breast tumours involve the regulation of oestrogen/progesterone levels and receptors,
including ER, PR, and HER2. The reduction of 17
β
-oestradiol and progesterone levels was described
in breast cancer animal models fed diets containing hesperetin and soy protein isolated [
90
,
91
]
as well as luteolin and genistein in the presence of letrozole and tamoxifen, respectively [
92
,
93
].
Likewise, pomegranate emulsion, flaxseed, and secoisolariciresinol modulated the expression of ER,
PR, and HER2 [9496].
Moreover, the benefits of the phenolic compounds against BC are also linked to their
anti-inflammatory eects. Dual inhibition of 5-LOX and COX-2 [
97
] and the modulation of cytokines
regarding BC [
98
] are interesting strategies in the treatment or prevention of this disease. Inhibition
of 5-LOX (and its product LTB4) and COX-2 via NF-
κ
B inhibition by resveratrol consumption
has been reported to be involved in BC prevention in DMBA-treated rats [
89
,
99
]. Naringenin
and soy protein modulated the expression of genes involved in regulating the inflammatory
response and cytokine signalling, including MCP-1 and IL-6 [90,100], while wogonoside and the mix
resveratrol-quercentin-genistein reduced the level of IL-6, TNF-
α
, and the TNF receptor-associated
factors TRAF2 and TRAF4 [
84
,
101
]. Several studies have highlighted the role of the immune system
regarding the protective eects of phenolic compounds against BC. Thus, quercetin and silybin
failed to protect against tumour development in immunodeficient mice. However, they were able
to regulate the accumulation of immune cells (i.e., T-cells) as well as the biosynthesis of cytokines
(i.e., TNF-
α
, IL-1
β
, IL-2, IL-4, IL-10, and INF-
γ
) in immunocompetent BALB/c mice [
102
,
103
]. Further
evidence is supported by experimental animal models fed EGCG and naringenin-enriched diet showing
accumulation and activation of T-cells in response to tumour-promoting stimuli [104,105].
Regarding
in vitro
studies, Table 2highlights the limited current evidence on the anticancer
eects for relevant phenolic-derived metabolites against dierent BC cell models. All in vitro studies
corroborated that phase-II conjugation reduced the cytotoxic and(or) antiproliferative eect against
dierent BC cell models and oestrogenic/anti-oestrogenic eects in ER+MCF-7 cells. The most robust
evidence has been reported in few studies for phase-II metabolites of RSV, quercetin, and isoflavones
indicating potential eects but lower than their counterpart unconjugated forms. Besides, a few
in vitro
studies made a comparison between BC and non-tumourigenic cells reporting the lack of cytotoxic
or antiproliferative eects in normal cells, unlike BC cell lines, at the tested concentrations and times
(Table 2) [
106
109
]. On the other hand, to date, no eect has been reported for phase-II metabolites to
other phenolic groups such as flavanones, urolithins and catechins, although the number of
in vitro
Int. J. Mol. Sci. 2020,21, 5718 10 of 33
studies conducted is still scarce. However, these results should be interpreted with caution since the
concentrations of conjugates in most in vitro studies are supra-physiological (>10 µM) (Table 2).
Although the conjugated metabolites are less bioactive than their free forms, and it remains
dicult to attribute their role in the chemopreventive
in vivo
eects, new approaches are being
investigated to confirm their possible
in vitro
anticancer activity. For instance, recent results have been
obtained assaying concentrations close to those detected
in vivo
(up to 10
µ
M) of RSV-derived phase-II
conjugates [
109
]. This study attributed the anticancer activity of RSV conjugates to cellular senescence
induction in BC cells and, therefore, deserves further investigation.
To the best of our knowledge, there are no
in vitro
studies conducted with phase-II curcumin
and enterolignans metabolites despite the large number of studies conducted with their unconjugated
compounds or enriched extracts. The lack of availability of these relevant metabolites may be behind
this gap.
On the other hand, it should not be discarded the potential role of phase-II metabolites (mainly
glucuronides and sulfates) as a source of bioactive aglycones in breast tumour tissues. This process
might occur via deconjugation under specific stimuli such as inflammation within the tumour
microenvironment, where the ratio glucuronyltransferase/glucuronidase is lower than in normal
tissues [
64
]. However, results that support this hypothesis are still limited to a few
in vitro
and animal
studies [74,110].
Int. J. Mol. Sci. 2020,21, 5718 11 of 33
Table 2. In vitro studies conducted with relevant phenolic-derived metabolites on breast cancer (BC) models.
Breast Cellular Model Compound Assayed Dose/Duration Main Outcomes References
Resveratrol
MCF-7, ZR-75-1, MDA-MB-231
(breast cancer cell lines) and
MCF-10A (normal cell line)
RSV, RSV 3-sulph, RSV 4
0
-sulph,
RSV 3,40-disulph. 1–350 µM; 48 h
RSV: IC50 =67.6–82.2 µM against all breast cancer cells;
IC50 =20 µM against MCF-10A.
RSV 3-sulph: IC50 =189–258 µM against MCF-7 and
MDA-MB-231; IC50 =228.3 µM against MCF-10A.
RSV 40-sulph and RSV 3,40-disulph: no cytotoxic eect
against breast cancer cells; IC50 =202.4–202.9 µM
against MCF-10A.
[111]
MCF-7
RSV, RSV 3-sulph, RSV 4
0
-sulph,
RSV 3,5 disulph, RSV
3,40-disulph, RSV
3,40,5-trisulph.
340 µM; 72 h Cytotoxic eect (only RSV and RSV 3-sulph). [112]
Saccharomyces cerevisiae strain
Y190 co-transformed with ERα
LBD and hTif2 coactivator and
MCF-7
RSV, RSV 3-gluc, RSV 3-glur,
RSV 3-sulph, RSV 40-sulph. 0.05–100 µM; 18–24 h
RSV 3-sulph showed anti-oestrogenic activity at 10 and
50
µ
M (with a marked preference for ER
α
at 10–100
µ
M),
and weak oestrogenic activity.
RSV showed oestrogenic activity in MCF-7 (5–10 µM).
[113]
MCF-7 and MDA-MB-231
RSV, RSV 3-glur, RSV 3-sulph,
RSV 40-sulph, DH-RSV,
DH-RSV 3-glur.
10 and 50 µM; 3 and 7 days
Proliferation in MDA-MB-231 cells (only RSV and
DH-RSV).
Oestrogenic/anti-oestrogenic eect in MCF-7 (only RSV
and DH-RSV).
[51]
MCF-7, MDA-MB-231 and
MCF-10A (normal cell line)
RSV, RSV 3-glur, RSV 3-sulph,
RSV 40-sulph, DH-RSV,
DH-RSV 3-glur.
0.5, 1, and 10 µM;
1–14 days
The eects were only observed in MCF-7 cells for all
compounds: Clonogenicity; cell cycle arrest;
senescence induction; modulation of p53/p21 and
p16/Rb pathways.
In MDA-MB-231 cells (only RSV at 10 µM):
clonogenicity.
[109]
Flavanones
MCF-7 and normal mammary
epithelial cells H184B5F5/M10
Baicalein, baicalein 7-O-sulph,
and baicalein-8-sodium
sulphonate.
50, 100, and 200 µM; 24 h
Eects in MCF-7 cells: cell viability; LDH release;
induction of cell cycle arrest; induction of morphological
changes; apoptosis; ROS; caspase-3, -9 activity.
Eects on H184B5F5/M10 cells: No cytotoxic.
[114]
Int. J. Mol. Sci. 2020,21, 5718 12 of 33
Table 2. Cont.
Breast Cellular Model Compound Assayed Dose/Duration Main Outcomes References
MCF-7 and MDA-MB-231 HP, HP 30-glur, HP 7-glur, HP
30-sulph, HP 7-sulph. 10 and 50 µM; 3 and 7 days
Proliferation in MDA-MB-231 cells (only HP at 50
µ
M).
Oestrogenic/anti-oestrogenic eect in MCF-7 (only HP at
10 and 50 µM).
[51]
Quercetin
MCF-7
Quer, Quer-3-O-gluc, Quer
3-O-glur, Quer 40-O-sulph,
Tamarixetin, Isorhamnetin.
50 µM; 48 h
Cell proliferation (no eect of Quer-3-O-gluc and Quer
3-O-glur). [115]
Saccharomyces cerevisiae
expressing ERα-Tif2 or ERβ-Tif2
and MCF-7
Quer, rutin, isoquercitrin, Quer
3-O-glur, Quer 3-O-sulph.
0.1–100 µM;
18–24 h
Quer 3-O-glur (IC50 =2.1 ±0.38 µM) and Quer (IC50 =
2.4 ±0.93 µM) showed oestrogenic activity in MCF-7
cells.
Quer 3-O-glur showed ERα-Tif2 (IC50 =103 ±2.24 µM)
and ERβ-Tif2 (IC50 =96 ±1.2 µM) agonistic activity.
Isoquercitrin showed similar ER
β
-Tif2 agonistic activity
than Quer 3-O-glur.
Quer showed weak ERβ-Tif2 (IC50 =5.3 ±0.9 µM)
agonistic activity.
[116]
MDA-MB-231 Quer 3-O-glur (alone or
together with A and NA).
1010–104M (binding
assay) and 0.01–1 µM (cell
assay); 1–24 h
Quer 3-O-glur showed competitive binding of [3H]-NA
to β2-AR (10-4–102µM). ROS formation; HMOX1,
MMP-2, and MMP-9 gene expression; intracellular
cAMP level; p-ERK 1/2 and p-P38; RAS activation;
invasive capacity of MDA-MB-231; MMP-9 activity.
[117]
MCF-10A (normal cell line)
Quer and Quer 3-O-glur (alone
or together with 4-OHE2and
NA).
1010–104M (binding
assay) and 0.05–10 µM (cell
assay); 2 h
Quer and Quer 3-O-glur showed competitive binding of
[3H]-NA to α2-AR (104–102µM).
γ-H2AX and AP sites activation.
[118]
MCF-7 and normal mammary
epithelial cells H184B5F5/M10
Quer, Isorhamnetin, and
Isorhamnetin 3-O-glucuronide. 25, 50, and 100 µM; 24–48 h
Eects in MCF-7 cells:
Cell viability; LDH release: induction of cell cycle
arrest; induction of morphological changes; apoptosis;
ROS.
Eects on H184B5F5/M10 cells: No cytotoxic eects.
[107]
MCF-7 and normal mammary
epithelial cells H184B5F5/M10
Quer, Quer 3-O-glur, and Quer
3-O-sulph. 25, 50 and 100 µM; 24–48 h
Eects in MCF-7 cells: cell growth; LDH release;
ROS; apoptosis; induction of cell cycle arrest;
induction of morphological changes.
Eects on H184B5F5/M10 cells: No cytotoxic eects.
[108]
Int. J. Mol. Sci. 2020,21, 5718 13 of 33
Table 2. Cont.
Breast Cellular Model Compound Assayed Dose/Duration Main Outcomes References
Urolithins
MCF-7 and MDA-MB-231
Uro-A, Uro-A 3-glur, Uro-A
8-glur, Uro-A 3-sulph, IsoUro-A,
IsoUro-A 3-glur, IsoUro-A
9-glur, Uro-B, Uro-B 3-glur,
Uro-B 3-sulph.
10 and 50 µM; 3 and 7 days
Proliferation in MCF-7 (Uro-A and IsoUro-A) and
MDA-MB-231 (free forms and conjugates at 50 µM).
Oestrogenic activity in MCF-7 (only free forms at 10 and
50 µM). Anti-oestrogenic activity in MCF-7 (Uro-A and
its conjugates, IsoUro-A and its conjugates, and Uro-B at
10 and 50 µM).
[51]
Catechin and epicatechin
MCF-7
Epi, Epi-30-O-sulph,
30-O-methyl-Epi,
40-O-methyl-Epi, catechin,
30-O-methyl-catechin.
100 µM; 48 h Cell proliferation (only 40-O-methyl-Epi). [115]
Isoflavones
MCF-7
GEN, GEN 40-O-glur, GEN
7-O-glur, GEN
40-O-sulph-7-O-glur, GEN
40-O-sulph, GEN 7-O-sulph,
GEN 40,7-di-O-sulph, DAZ,
DAZ 40-O-glur, DAZ 7-O-glur,
DAZ 40-O-sulph-7-O-glur, DAZ
40-O-sulph, DAZ 7-O-sulph,
DAZ 40,7-di-O-sulph, Glycitein,
Glycitein 7-O-glur, DH-DAZ,
DH-DAZ 7-O-glur, O-DMA,
O-DMA 7-O-glur.
10–1000 µM; 24 h
Low stimulatory MCF-7 growth, β-gal induction, and
binding to ERs: sulphates of GEN and glucuronides of
glycitein and DH-DAZ.
Moderate binding, but low (or lack) MCF-7 growth and
β-gal induction: glucuronides of GEN, DAZ, and
O-DMA.
Moderate MCF-7 growth and β-gal induction, but low
(or lack) binding: DAZ 40-O-sulph-7-O-glur.
O-DMA was the most active compound stimulating
MCF-7 growth, binding to hERβ, and inducing β-gal.
[119]
MCF-7
GEN, GEN 7-O-glur, DAZ, DAZ
7-O-glur. 16 µM; 72 h Cytotoxicity (only DAZ). [120]
Int. J. Mol. Sci. 2020,21, 5718 14 of 33
Table 2. Cont.
Breast Cellular Model Compound Assayed Dose/Duration Main Outcomes References
Isoflavones
MCF-7
GEN, GEN 7-O-sulph, GEN
40-O-sulph, DAZ, DAZ
7-O-sulph, DAZ 40-O-sulph,
equol, equol-7-sulph (in the
presence of 4 ×1010 M
[2,4,6,7-3H] oestradiol).
107–104M; 18–24 h
(binding and gene
expression assay) and 7 or
14 days (proliferation assay)
The sulphation in position 7 reduced the oestrogen
capacity of GEN and equol in all assays.
GEN 40-O-sulph showed lower proliferative activity (at
1 and 10 µM), and similar or even higher binding
anity to ER (compared to GEN).
DAZ 40-O-sulph showed lower binding anity to ER
and increased proliferative activity (at 10 µM). DAZ
7-O-sulph showed similar or even higher anity to ER
and similar proliferative activity (at 1 and 10 µM)
(compared to DAZ).
[121]
HS578T, MDA-MB-231, and
MCF-7
Puerarin, GEN, DAZ, and mix
of DAZ glucuronides/sulphates.
12.5–100 µM
(free compounds) and 2.35
µM the mix of DAZ
conjugates; 24–72 h
Eect of only with free compounds:
(IC50 =29–71 µM) cell viability; induction of cell cycle
arrest; apoptosis; caspase-3 activity.
Eect of mix of DAZ conjugates: (IC50 =2.35 µM) cell
proliferation; induction of cell cycle arrest; apoptosis;
caspase-9, p53, p21, and Bax.
[122]
MCF-7, T47D, and MCF-10A
(normal cell line)
GEN, GEN 7-O-sulph, GEN
40-O-sulph, GEN 7-O-glur.
5.12 ×103–80 µM for GEN
and 2.2–4.5 µM for
conjugates; 3 days
Dissimilar eects of GEN cell proliferation: at low
concentrations and at higher concentrations. GEN
7-O-glur stimulated cells growth. This eect was related
to deconjugation. GEN 7-O-sulph, GEN 40-O-sulph
exerted no eects.
[106]
T47D and T47D-ERβ
(tetracycline dependent ERβ
expression)
GEN, GEN 7-O-glur, DAZ, DAZ
7-O-glur. 105–1000 µM; 48 h
Dissimilar eects on proliferation: at low
concentrations and at higher concentrations.
Lower proliferative potency than 17β-oestradiol:
PC
50
in T47D-wt: 17
β
-oestradiol =4.2
×
10
6µ
M; GEN =
0.19 µM; GEN 7-O-glur =21.4 µM; DAZ =0.186 µM;
DAZ 7-O-glur =107 µM. PC50 in T47ERβ:
17β-oestradiol =2.45×107µM; GEN =0.2 µM; GEN
7-O-glur =7.8 µM; DAZ =0.035 µM; DAZ 7-O-glur =
400 µM.
[123]
Int. J. Mol. Sci. 2020,21, 5718 15 of 33
Table 2. Cont.
Breast Cellular Model Compound Assayed Dose/Duration Main Outcomes References
Isoflavones
MCF-7 and T47D
DAZ, DAZ 40-O-sulph, DAZ
7-O-sulph, DAZ 4
0
,7-di-O-sulph,
equol, O-DMA.
0.1, 1, and 10 µM; 1–24 h
Dierent eects on NGB: equol, O-DMA, DAZ
7-O-sulph, DAZ 40,7-di-O-sulph and DAZ and DAZ
40-O-sulph. Eects of DAZ, DAZ 40-O-sulph and equol
on protein phosphorylation: p-ERα/ERα
(all compounds);
p-Akt/Akt (only equol; 1 h treatment);
p-p38/p38 (all molecules; 1 and 24 h treatment). DAZ,
DAZ 40-O-sulph and equol preserve PAX activity in
MCF-7 cells (NGB, PARP-1 cleavage, and cell
number) in the presence of 17β-oestradiol.
Eects of the mix of metabolites:
1
Mix of sulphates metabolites:
NGB by in the presence
or absence of 1 µM DAZ; the mix of sulphates together
with 1 µM DAZ preserve PAX eects (PARP-1
cleavage) in the presence of 17β-oestradiol.
2Mix of gut metabolites: NGB by equol +O-DMA
(1 µM each) in the presence or absence of 1 µM DAZ;
PAX eects (PARP-1 cleavage) in the presence of
17β-oestradiol.
[124]
1
Mix of sulphate metabolites composition: DAZ 4
0
-O-sulph +DAZ 7-O-sulph +DAZ 4
0
,7-di-O-sulph (1
µ
M each);
2
Mix of gut metabolites composition: equol +O-DMA (1
µ
M each).
Abbreviations: A, adrenaline;
α2
-AR,
α2
-adrenergic receptor;
β2
-AR,
β2
-adrenergic receptor; cAMP, cyclic adenosine monophosphate; DAZ, daidzein; DH-DAZ, dihydrodaidzein; DH-RSV,
dihydroresveratrol; Epi, epicatechin; ER+, oestrogen receptor+; ERE-CAT, oestrogen response element chloramphenicol acetyl transferase;
γ
-H2AX, H2A histone family member X
phosphorylated on Ser139; GEN, genistein; gluc, glucoside; glur, glucuronide; hER
α
, human oestrogen receptor alpha; HMOX1, heme oxygenase 1; HP, hesperetin; hTif2, coactivator
receptor-interacting domain; LBD, ligand binding domain; LDH, lactate deshydrogenase; NA, noradrenaline; NGB, neuroglobin; O-DMA, O-desmethylangolensin; PAX, paclitaxel; PC,
potency concentration; PARP-1, poly [ADP-ribose] polymerase 1; Quer, quercetin; ROS, reactive oxygen species; RSV, resveratrol; 4-OHE2, 4-hydroxy-oestradiol; sulph, sulphate.
Int. J. Mol. Sci. 2020,21, 5718 16 of 33
5. Dietary (Poly)Phenols and Clinical Studies in Breast Cancer Patients
Table 3summarizes the current evidence regarding the eects of several dietary (poly)phenols or
phenolic-containing products in breast cancer patients. The main search criteria used were as follows:
(i) clinical trials dealing with BC patients (either with active disease or cured patients to prevent
recurrence), (ii) evaluation of the eects of orally administered (poly)phenols or phenolic-containing
products (pure standards, plant extracts or foodstus) on tumour biomarkers or other clinically-relevant
outcomes related to BC, including the decrease of side-eects associated with chemo- or radiotherapy
(i.e., intravenous administration or articles dealing with only ‘hot flashes’, ‘wellness’ or other general
determinations were excluded). Articles dealing with either healthy or at-risk volunteers (including
postmenopausal women) were omitted, as were those studies conducted with non-dietary sources.
Trials, where metabolites disposition was evaluated in malignant breast tissue from patients, were also
included only when some eects on tumour-related biomarkers were evaluated.
As expected, the evidence on the anticancer eects of dietary phenolics in BC patients is limited,
fragmented, and inconclusive. In this context, the most studied polyphenols are EGCG, as a component
of green tea extracts, and curcuminoids present in curcumin extracts.
The pioneer pilot study of Thompson et al. revealed the increase of apoptosis, and the reduction of
both Ki-67 (a cell proliferation marker) and c-erB2 (also known as HER-2) expression in 19 BC patients
that consumed 25 g/day flaxseed vs. 13 patients that consumed placebo for approximately 37 days
from the diagnosis until the resection of the tumour [
125
]. Despite the promising results, it took almost
10 years for a new study on flaxseed to be published by
McCann et al
. [
126
]. This new study was also a
pre-surgery pilot study in which postmenopausal women diagnosed with ER+BC consumed flaxseed
(25 g/day) alone or combined with the aromatase inhibitor anastrozole or placebo, between tumour
biopsy and resection (mean of 19 days). The authors found a reduction of endogenous oestrogen
production (androstenedione in the flaxseed +anastrozole group, and dehydroepiandrosterone in
the flaxseed group) (Table 3) [
126
]. Aromatase inhibitors block the enzyme aromatase that catalyzes
the conversion of androgens into oestrogens, the primary source of endogenous oestrogens in
postmenopausal women. This is relevant since anti-estrogen therapy aims to prevent access of the
tumour to oestrogen and has been considered the standard of care in ER+tumours [127].
Green tea extracts are the most widely tested polyphenol-containing products to date in BC patients
(Table 3). The main active component is supposed to be EGCG, but the specific association between
EGCG and the reported outcomes has not been unequivocally proven so far. Green tea extracts have
been assayed to explore the eects on cancer biomarkers after short treatments (pre-surgery) [
66
,
128
]
as well as after longer treatments (6 months) to assess the maximum tolerability dose (MTD) [
129
] and
eects on some cancer-related biomarkers [
129
,
130
]. The MTD was established at 1200 mg/day for
6 months in patients with ER- and PR- breast tumours, but no significant changes were observed on ER,
Ki-67, IGF-1, IGFBP-3, and mammographic density vs. placebo after 6 months [
129
]. The same authors
explored the eects on more biomarkers in the same group of patients, but no eects were observed
on VEGF, oxidative damage, and inflammatory markers, among others, but only a transient decrease
of serum HGF in green tea consumers vs. placebo [
130
]. The other pre-surgery studies reported a
significant decrease of Ki-67 (only in healthy breast tissue, but not in the malignant one), while no eects
were observed on apoptosis (caspase-3) and angiogenesis (CD34) in both healthy and malignant breast
tissues after consuming green tea extract (2.2 g/day for 35 days) [
128
]. More recently,
Lazzeroni et al
.
evaluated the disposition of EGCG in normal and malignant tissues from 12 patients with ER+
tumours, but the authors did not find any eect on cell proliferation, angiogenesis, oxidative stress,
inflammation or other markers [
66
]. However, they found a significant increase (36%) in testosterone
levels. Although testosterone can be aromatized to oestradiol, which increases proliferation and hence,
BC risk, testosterone has also been associated with BC prevention [131,132].
Curcumin extracts, also containing the related curcuminoids, demethoxycurcumin and
bisdemethoxycurcumin, have also been assayed in BC patients. The first trial was conducted by
Bayet-Robert et al. in 14 patients with metastatic breast cancer to evaluate the MTD or oral curcumin
Int. J. Mol. Sci. 2020,21, 5718 17 of 33
in combination with intravenous administration of docetaxel (Table 3) [
133
], a chemotherapeutic drug
used in metastatic BC. The authors found a high MTD of oral curcumin (8 g/day) in combination with
the drug (100 mg/m
2
), administered every 3 weeks for six cycles, with a total of 63 cycles, and observed
a significant decrease of plasmatic CEA and VEGF from the 3rd cycle. However, no clear association
was found between markers decrease and the occurrence of curcumin in plasma [
133
]. Later,
Ryan et al
.
observed the protection of oral curcumin (6 g/day) administration vs. placebo after 6 months against
radiation dermatitis in BC patients (n =30) with prescribed radiation therapy (RT) [
134
]. The same
authors tried to confirm their previous exploratory results in an ambitious trial that enrolled 686 patients
with prescribed RT [
135
]. Nevertheless, curcumin did not reduce radiation dermatitis severity at
the end of RT compared to placebo after 6 months (30 RT sessions). Overall, this is a warning to
consider the minimum number of patients necessary to conclude the possible eects associated with
the consumption of phenolics.
Other trials with BC patients and phenolics or phenolic-containing products, included the assay
of grapeseed [
136
], milk thistle [
65
], and red clover [
137
] extracts, walnuts [
138
], and also a product
containing a mixture of EPA, DHA hydroxytyrosol and curcumin [
139
]. In one of the pioneer studies
dealing with polyphenols and BC, Brooker et al. did not find any eect on tissue hardness due to RT in
69 BC patients after consuming a grapeseed proanthocyanidins extract (300 mg/day) for 12 months vs.
placebo [
136
]. Lazzeroni et al. described the disposition of some metabolites in breast tissue. However,
they did not observe any eect on NO, IGF-1, and Ki-67 in 12 patients with ER+breast cancer after
consuming 2.8 g/day of a silybin-containing milk thistle extract in combination with phosphatidylcholine
for 4 weeks between biopsy diagnosis and resection [
65
]. The only study dealing with food and BC has
been conducted so far by Hardman et al. [
138
]. These authors conducted a pre-surgery trial in 10 BC
patients that consumed around 30 g walnuts/day for 15 days (between tumour biopsy and resection).
Surprisingly, with only 10 patients, and taking into account the huge inter-individual variability
in gene expression and the previously reported artefacts [
140
,
141
], they observed the activation of
genes participating in pathways that promote apoptosis and cell adhesion and inhibition of pathways
that promote cell proliferation and migration (involving 456 genes in the tumour) due to walnut
consumption [
138
]. Recently, Mart
í
nez et al. described the anti-inflammatory eect (decrease of
plasma CRP vs. placebo) in BC patients with no active disease, and receiving adjuvant hormonal
therapy, after consuming a mixture containing 460 mg fish oil (EPA, DHA), olive extract (125 mg
hydroxytyrosol) and 50 mg curcumin extract for 30 days [
139
]. Although the authors described a
relevant decreased of plasma CRP from 8.2 mg/L to 5.3 mg/L, unfortunately, the final CRP levels
were in the boundary of those associated with patient’s survival [
142
] and still associated with high
cardiovascular risk [
143
]. Besides, the responsible compound for the anti-inflammatory eect observed
was not identified from such a complex mix. Finally, Ferraris et al. recently evaluated the eect of a
red clover extract containing isoflavones (80 mg/day) to reduce side-eects of tamoxifen treatment in
patients with ER+BC under tamoxifen treatment for 2 years. Although the consumption of red clover
extract was safe, no eects were observed in the targets related to tamoxifen treatment (menopausal
rating score, sex hormone levels, endometrial thickness, and breast density, among others). Only BMI
and waist circumference were significantly reduced in the red clover group vs. placebo [137].
Int. J. Mol. Sci. 2020,21, 5718 18 of 33
Table 3. Clinical trials with dietary (poly)phenols or phenolic-containing foodstus in patients with breast cancer.
Cohort and Sample Size Trial Design Objective Outcomes References
Newly biopsy-diagnosed
breast cancer patients (n =32),
with no hormone therapy
Design: Pre-surgery, randomized double-blind,
placebo-controlled clinical trial.
Product and dose: Patients (n =19) consumed
flaxseed (25 g/d) or placebo (n =13).
Follow-up: 37 and 39 days in the flaxseed and
placebo groups, respectively.
Eect of flaxseed on tumour
biomarkers.
Significant reductions in Ki-67 labelling
index (34.2%) and in c-erbB2
expression, (71%), and an increase in
apoptosis (30.7%) were observed in the
flaxseed, but not in the placebo group.
[125]
Patients (n =66) with tissue
hardness due to radiotherapy
for early breast cancer (at least
24 months prior to trial entry),
with no active disease
Design: Phase II, placebo-controlled,
randomized trial.
Product and dose: Capsules containing a grape
seed proanthocyanidin extract 100 mg three
times a day orally, or placebo.
Follow-up: 6 months.
Eect on the surface area of
palpable breast induration after
12 months. Secondary
endpoints: change in
photographic breast appearance
and patient self-assessment of
breast hardness, pain and
tenderness.
No significant dierence between
treatment and control groups in terms
of external assessments of tissue
hardness, breast appearance or patient
self-assessments of breast hardness,
pain or tenderness.
[136]
Patients (n =14) with
metastatic breast cancer
Design: Open label, phase I, non-controlled
trial.
Product and dose: Intravenous docetaxel plus
oral curcumin (escalated dose until toxicity
limit is reached).
Follow-up: Docetaxel 100 mg/m2was
administered every 3 weeks (w) for six cycles.
Curcumin was given orally for 7 consecutive
days (d) (from d-4 to d+2) for six cycles (a total
of 63 cycles).
Establishment of the maximal
tolerated dose (MTD) of oral
curcumin plus intravenous
docetaxel.
MTD of curcumin was 8 g/day, in
combination with docetaxel 100 mg/m
2
administered every 3 w for six cycles.
Recommended curcumin dose: 6 g/d
for 7 consecutive d every 3 w in
combination with a standard dose of
docetaxel.
Decrease of plasmatic CEA and VEGF.
[133]
Patients (n =40) with resected
stage I-III ER- and PR- breast
cancer with no active disease
Design: Randomized, phase IB,
double-blinded, placebo-controlled,
and dose-escalation study.
Product and dose: Capsules (green tea extract)
containing EGCG (treated group, n =30).
Daily oral dose was 800 mg (n =16), 1200 mg
(n =11), and 1600 mg (n =3) EGCG. Control
group (n =10): placebo.
Follow-up: 6 months.
Establishment of the MTD for
EGCG.
MTD was 1200 mg/d EGCG for
6 months.
No significant change in SHBG, IGF-1,
IGFBP-3, ER, Ki-67 proliferation index
or mammographic density
[129]
Int. J. Mol. Sci. 2020,21, 5718 19 of 33
Table 3. Cont.
Cohort and Sample Size Trial Design Objective Outcomes References
Patients (n =28) with ductal
carcinoma in situ or primary
invasive stage I or II breast
cancer
Design: Pre-surgery, controlled study.
Product and dose: 3 capsules/d (green tea
extract) containing EGCG (treated group,
n=13). Daily oral dose was 940 mg EGCG.
Control group (n =15): no capsules.
Follow-up: Average duration of green tea
intake was 35 days.
Short-term eects of green tea
supplementation on cancer
biomarkers.
Significant decrease of Ki-67 in the tea
group, but only in normal tissue.
No eects on apoptosis (caspase-3),
and angiogenesis (CD34) markers in
benign or malignant tissue.
[128]
Patients (n =30) with
non-inflammatory breast
cancer or carcinoma in situ
and prescribed RT without
concurrent chemotherapy
Design: Randomized, double-blind,
placebo-controlled clinical trial.
Product and dose: Patients (n =14) consumed
6 g/d curcumin extract (a daily dose of 4.7 g
curcumin, 0.9 g demethoxycurcumin, and
0.15 g bisdemethoxycurcumin) or placebo
(n =16).
Follow-up: 6 months (30 RT sessions)
Eect of curcumin to reduce
RDS.
Significant reduction of RDS and moist
desquamation at the end of treatment
vs. placebo (mean RDS =2.6 vs. 3.4;
and 28.6% vs. 87.5%; respectively).
[134]
Postmenopausal women (n =
24) with newly-diagnosed,
and resectable, ER+breast
cancer
Design: Pre-surgery, 2 ×2 factorial,
randomized, placebo-controlled trial.
Product and dose: 25 g/d ground flaxseed +
1/d placebo pill (n =6); 1 mg/d anastrozole
(aromatase inhibitor) (n =7); 25 g/d ground
flaxseed +1 mg/d anastrozole (n =6); or 1/d
placebo pill control (n =5).
Follow-up: Mean of 18.8 days.
Eects of flaxseed and the
aromatase inhibitor, anastrozole,
on steroid hormones and
tumour-related biomarkers.
Mean ERβexpression was
approximately 40% lower from pre- to
post-intervention in the flaxseed (FS) +
anastrozole (AI) group only. Significant
negative association for
androstenedione in the FS +AI group
vs. placebo, and for
dehydroepiandrosterone with AI
treatment.
[126]
Patients (n =34) with resected
stage I-III ER- and PR- breast
cancer with no active disease.
(Ancillary study to that of
Crew et al. [129]
Design: Randomized, phase IB,
double-blinded, placebo-controlled, and
dose-escalation study.
Product and dose: Capsules (green tea extract)
containing EGCG (treated group, n =26).
Daily oral dose was 800 mg (n =14), 1200 mg
(n =11), and 1600 mg (n =1) EGCG. Control
group (n =8): placebo.
Follow-up: 6 months
Eect of EGCG on cancer
biomarkers risk.
Significant transient decrease of serum
HGF (only after 2 months of treatment)
in EGCG consumers. No significant
eects on VEGF, serum cholesterol and
triglycerides, oxidative damage,
and inflammatory biomarkers.
[130]
Int. J. Mol. Sci. 2020,21, 5718 20 of 33
Table 3. Cont.
Cohort and Sample Size Trial Design Objective Outcomes References
Patients (n =12) with newly
diagnosed breast cancer, ER+,
not eligible for neoadjuvant
treatment
Design: Pre-surgery (non-controlled) dietary
intervention.
Product and dose: Silybin-phosphatidylcholine
complex (2.8 g/d) given orally.
Follow-up: 4 w before surgery.
Eects on NO, IGF-1 and Ki-67.
No eects on NO, IGF-1 and Ki-67 were
observed. [65]
Patients (n =12) with newly
diagnosed breast cancer, not
eligible for neoadjuvant
treatment.
Design: Pre-surgery (non-controlled) dietary
intervention.
Product and dose: Catechin (65.1 mg/d) and
EGCG (44.9 mg/d) given orally (300 mg tea
extract).
Follow-up: 4 w before surgery.
Eect on cell proliferation,
angiogenesis, oxidative stress,
chronic inflammation, and
adiposity-related endocrine
mechanism.
Significant increase of testosterone.
No eect in the rest of markers. [66]
Patients (n =578) with
non-inflammatory breast
cancer or carcinoma in situ,
and prescribed fractionated
radiation therapy (RT)
without concurrent
chemotherapy
Design: Phase II, randomized, double-blind,
placebo-controlled clinical trial.
Product and dose: Patients (n =283) consumed
either 6 g/d curcumin extract (a daily dose of
5.4 g curcumin, 0.48 g dimethoxy curcumin,
and 0.12 g bisdemethoxy curcumin), or placebo
(n =295).
Follow-up: 6 months (30 RT sessions).
Confirmatory study on the
eect of curcumin to reduce
radiation dermatitis severity.
Curcumin did not reduce radiation
dermatitis severity at the end of RT
compared to placebo.
[135]
ER+and(or) PR+
postmenopausal women
(n =45) with resected breast
cancer at early stage (with no
active disease), and receiving
adjuvant hormonal therapy
Design: Open-label, single-arm
(no placebo-controlled).
Product and dose: Three daily capsules,
containing 460 mg of fish oil (EPA and DHA),
125 mg of olive extract (12.5 mg
hydroxytyrosol), and 50 mg extract of
curcumin (47.5 mg curcuminoids)
Follow-up: 30 days.
Eect on inflammation and
pain.
Significant decrease of plasma CRP
(from 8.2 ±6.4 mg/L at baseline to
5.3 ±3.2 mg/L), and pain (21.5%) after
30 days.
[139]
Int. J. Mol. Sci. 2020,21, 5718 21 of 33
Table 3. Cont.
Cohort and Sample Size Trial Design Objective Outcomes References
Breast cancer patients (n =10)
Design: Pre-surgery, two arms, controlled
study.
Product and dose: Patients (n =5) consumed
walnuts (around 60 g/d) or not (i.e., controls,
n=5).
Follow-up: About 15 days.
Eect of walnut consumption
on gene expression in breast
cancer tissue.
Significant change of 456 genes in the
tumour due to walnut consumption.
Activation of pathways that promote
apoptosis and cell adhesion, and
inhibition of pathways that promote
cell proliferation and migration.
[138]
Patients (n =81) with
histologically confirmed
operable ER+breast cancer,
with no distant metastasis,
and receiving tamoxifen.
Design: Randomized, double-blind, and
placebo-controlled trial.
Product and dose: Patients (n =42) consumed
a red clover extract (80 mg isoflavones/d) or
placebo (n =39).
Follow-up: 2 years
Eect of isoflavones from red
clover extract and lifestyle
change to reduce side-eects of
tamoxifen treatment.
The reductions in BMI and waist
circumference were significantly
greater in the treatment than placebo
group. No dierences between groups
in the rest of determinations: MRS,
HDLc, insulin, total cholesterol, LDLc,
triglycerides, insulin resistance, sex
hormone levels, endometrial thickness
and breast density.
[137]
Abbreviations: CEA, carcinoembryonic antigen; c-erbB2 (or HER2), Humanized epidermal growth factor receptor 2; DHA, docosahexanoic acid; EGCG, epigallocatechin gallate; EPA,
eicosapentaenoic acid; ER: oestrogen receptor; HGF, hepatocyte growth factor; IGF-1, insulin-like growth factor 1; IGFBP-3, Insulin-like growth factor binding protein-3; MRS, menopausal
rating score; MT, malignant tissue; NT, normal tissue; PR: progesterone receptor; RDS, Radiation Dermatitis Severity score; RT, radiation therapy; SHBG, Sex hormone binding globulin;
VEGF, vascular endothelial growth factor.
Int. J. Mol. Sci. 2020,21, 5718 22 of 33
6. Identifying the Existing Gaps to Address Future Preclinical and Clinical Research
As thoroughly detailed in the present review, despite substantial progress made during the
last decades in advancing our knowledge of how dietary phenolics exert their biologic eects and,
consequently, how they may be involved in the prevention and treatment of BC, to date, minimal
success has been achieved.
Overall, several reasons have led to the vast gap between
in vitro
and
in vivo
evidence. Among the
main reasons, it should be highlighted the low number of well-conducted human studies reported so
far. Furthermore, it is essential to point on several central issues found in clinical trials, and pending
aspects that should be addressed in future clinical studies:
(i) There is a need for multidisciplinary teams beyond researchers (oncologists, surgeons, pathologists,
etc.) to conduct dietary interventions in BC patients. Indeed, this issue is inherent in every clinical
trial dealing with cancer patients. In the case of ‘dietary interventions’, oncologists usually oppose
the assay of high concentrations of dietary phenolics to patients undergoing chemotherapy due to
the limited knowledge regarding the possible interactions between dietary compounds, including
phenolics, and chemotherapy drugs. Overall, this challenging logistic yields a low number of
recruited BC patients. Consequently, a restricted number of samples to explore or validate results,
which is even more challenging when assaying molecules with relatively low anticancer activity
compared to standard chemotherapy drugs.
(ii)
The high heterogeneity of the results obtained. Inter-individual variability is perhaps one
of the most critical aspects to establish definite conclusions in clinical studies. The low
number of volunteers, as well as the specific selection criteria of the volunteers selected
in the dierent studies, are key factors to understand the high variability reported. Genetic
polymorphisms or dierences in the composition or functionality of the gut microbiota will
also contribute to the dierent response of individuals to interventions with polyphenol-rich
foods [48,144146]
. In this regard, a highly variable metabolism between individuals has been
described for some phenolic compounds. This allows for grouping the population according
to the excretion of the metabolized compound into “high or low producers”, as it occurs in
the metabolism of the phenolic precursor-derived metabolite pairs hesperidin-hesperetin [
147
],
lignans-enterolignans [
148
], ellagic acid-urolithins [
149
] and procyanidins-valerolactones [
150
,
151
].
Besides, specific metabotypes, attributed to the dierent composition and functionality of the gut
microbiota, have been described in the case of urolithins [
146
,
152
] or isoflavones [
153
], allowing
the stratification of individuals based on the production of specific metabolites associated with a
particular gut microbiota ecology. Thus, the inter-individual variability related to bioavailability
and metabolism of dietary phenolics is essential to comprehend the results in clinical studies and
to identify whether the possible beneficial eects can be extrapolated to the whole population
or only to certain specific individuals [
146
]. Future research should be conducted to evaluate
associations between specific phenolic-related gut microbiota metabotypes and protective eects
against BC.
(iii)
The diculty of attributing chemopreventive eects to phenolics. Most human studies have
been conducted with phenolic-containing products or extracts, which makes it challenging to
identify whether single phenolics are (or not) actually responsible for the possible anti-tumour
eect. Along this line, it is important to consider the dierences in phenolic composition between
plant foods, the diculty in the estimation of the dietary intake of phenolics modulated by
several factors such as food matrix, food processing-related factors, as well as the interaction
between phenolics and the gut microbiota, and also other food components, such as proteins
and carbohydrates that will undoubtedly influence the bioavailability and subsequent potential
chemopreventive eects [45,154].
Another element that can help establish causality between eect and polyphenols is the
determination of the derived metabolites in the target breast tissue. However, the characterization of the
Int. J. Mol. Sci. 2020,21, 5718 23 of 33
metabolic profiling of phenolic-derived metabolites in breast tissues remains poorly explored. Several
reasons contribute to this gap: the lack of pure standards (mostly phase-II conjugates) that prevent their
accurate quantification, and the accessibility to enough mammary biopsies to perform the analyses.
Besides, the lack of standardized analytical protocols contributes to reporting incomplete or even
misleading identifications/quantifications (heterogeneity of samples, hydrolysis vs. non-hydrolysis
approaches, etc.).
Finally, it should be taken into account that phenolics generally exert relatively low biological
activity, which makes dicult to discern their possible anticancer eects. In this regard, clinical studies
should be focused on looking for eects upon long-term consumption of phenolic-rich foods. Therefore,
in the coming years, there is a need to improve and re-design clinical trials to demonstrate the ecacy
of dietary phenolics against BC unequivocally.
(iv)
There is not enough evidence regarding the possible interaction between dietary phenolics and
conventional chemotherapy drugs. Despite the preclinical evidence that supports the potential
benefits of dietary phenolics as possible adjuvants in BC management [
155
,
156
], to date, only three
clinical studies have been conducted using a combination of phenolics and chemotherapeutic
drugs [
126
,
137
,
139
]. Therefore, there is a need to evaluate the potential interactions between
phenolics and chemotherapeutic drugs to support phenolics’ usefulness as adjuvants in BC
treatment and further follow-up of patients to prevent relapses.
As far as chemoprevention is concerned, the assayed conditions in animal models that cannot
be extrapolated to humans, and the non-physiological
in vitro
approaches were predominant in the
past. Nowadays, the trend is to perform physiologically relevant studies, as highlighted in recent
guidelines [
78
,
157
]. Regarding BC, attention should be paid to the following gaps and pitfalls performed
in the past strategies for phenolics’ chemoprevention determination in animal and cellular BC models:
(i)
The diculty of extrapolating the results obtained from animal research to humans. As in the
case of human studies, several problems can be found in the BC animal models to evaluate the
chemopreventive eect of phenolics: the methodology issues (low number of animals, and lack
of controls, etc.), the heterogeneity of data, the diculty of attributing chemopreventive eects to
phenolics (use of whole-foods or extracts instead of single phenolics, determination of phenolics
in breast tissues, etc.).
Besides, other intrinsic aspects should be considered and improved in animal research: any breast
tumour-induction process (chemically-induced BC, murine mutant models, etc.) can reflect the
heterogeneity of human BCs. It is habitual to use inadequate doses and exposure times for single
phenolics, food, or extracts. Instead, doses should be equivalent to those achievable in humans
after the regular intake of the phenolic-containing foods. The systemic administration of phenolics
or food extracts (intravenous, intraperitoneal, etc.) is not representative of a dietary context and
omits the significant handicaps of bioavailability and metabolism. Finally, the suitability of the
specific animal model, according to the capability of each species to metabolize phenolic compounds,
is of particular interest, especially in polyphenol-related gut microbiota metabotypes (urolithin and
isoflavone metabotypes).
Besides, we must add to all the above logical inter-species dierences (rats, mice, pigs, etc.),
and even within strains from the same animal model, which contribute to the great challenge of
extrapolating
in vivo
results to BC patients [
158
]. Nevertheless, animal research remains an essential
step before clinical trials, mainly in evaluating the interaction with chemotherapeutics.
(ii)
In vitro
studies must have physiological relevance to establishing potential eects and conclusions.
Dierent aspects of
in vitro
research should be avoided: the use of both unrealistic concentration
and metabolic forms of phenolics; the unsuitability of BC cell models using a single cell line instead
of considering the heterogeneity of cell lines using multiple cell lines with dierent mutations and
other genetic characteristics (ER/PR positive or negative, etc.), or even more realistic advanced
Int. J. Mol. Sci. 2020,21, 5718 24 of 33
models such as primary cell cultures, organoids, etc.; and testing single phenolic or derived
metabolite without considering the real mixture of compounds present
in vivo
, as well as the
food matrix eect, avoiding the possible synergistic, antagonistic, or additive eects among
them. Therefore, following these principles, transferring the information on the pharmacokinetic
properties and bioavailability of phytochemicals in breast tissues plays one of the critical roles for
a better evaluation of their mechanism of action involved in their BC chemoprevention.
7. Concluding Remarks
According to the detailed literature survey, including only physiologically relevant preclinical
studies and clinical studies, we can conclude that some dietary phenolics, especially RSV, quercetin,
isoflavones, EGCG, lignans, and curcumin, seem to be the most promising candidates. However,
the clinical evidence is still minimal when compared with that obtained in preclinical studies.
As stated above, the increase and development of highly robust, well-designed clinical studies
in BC patients and the quantitative and qualitative determination of phenolic-derived metabolites
that reach breast tissues will make possible the continuous progress of the preclinical research.
This approach will allow a more in-depth understanding of the molecular mechanisms involved in
their chemopreventive and(or) chemotherapeutic properties and, perhaps, allowing the establishment
of preventive nutritional strategies.
In the “omics” era, complementary approaches, including in silico and molecular docking studies,
as well as high-throughput “omics” technologies, such as (epi)genomics, proteomics, and metabolomics
are rapidly gaining interest to identify new ligands for specific cancer molecular targets in order to
improve the therapeutic potential of both phenolics and their metabolic derivatives [
159
,
160
]. Besides,
the analysis of the composition and functionality of the intra-tumour microbiota in breast cancer
has been reported to be useful to predict dierent responses between tumour types and subtypes
against immunotherapy [
161
]. Whether the microbiota of breast cancer, particularly rich and diverse,
could modulate dietary phenolics’ eects deserves further research.
Supplementary Materials:
Supplementary materials can be found at http://www.mdpi.com/1422-0067/21/16/
5718/s1. Table S1: Dietary phenolic-derived metabolites identified in breast tissues in animal model studies. Table
S2: Animal studies conducted with dietary phenolics and/or phenolic-rich extracts on BC models.
Author Contributions:
J.C.E. and A.G.-S. conceived and designed the study; M.
Á
.
Á
.-G., J.A.G.-B., J.C.E. and
A.G.-S. contributed to the discussions and preparation of the manuscript; J.A.G.-B., J.C.E. and A.G.-S. wrote the
article. All authors have read and agreed to the published version of the manuscript.
Funding:
This research was funded by the projects PID2019-103914RB-I00 (MICINN, Spain), 19900/GERM/15
(Fundaci
ó
n S
é
neca de la Regi
ó
n de Murcia, Spain), and 201870E014 and 201770E081 (CSIC, Spain). J.A.G.B.
was supported by a Juan de la Cierva contract (IJCI-2016-27633) from the Ministry of Science, Innovation and
Universities (Spain) and a Standard European Marie Curie Fellowship from the European Commission. This project
has received funding from the European Union’s Horizon 2020 research and innovation programme under the
Marie Sklodowska-Curie grant agreement No 838991.
Conflicts of Interest: The authors declare no conflict of interest.
Abbreviations
BC Breast Cancer
DMBA 7,12-Dimethylbenz[a]anthracene
EGCG Epigallocatechin Gallate
ER Oestrogen Receptor
HER2 Human Epidermal growth factor Receptor 2
MTD Maximum Tolerability Dose
PR Progesterone Receptor
RSV Resveratrol
TNBC Triple Negative Breast Cancer
Int. J. Mol. Sci. 2020,21, 5718 25 of 33
References
1.
WHO Media Centre (Cancer—Fact Sheets). Available online: http://www.who.int/mediacentre/factsheets/
fs297/en/(accessed on 3 May 2020).
2.
Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin.
2020
,70, 7–30. [CrossRef]
[PubMed]
3.
Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018:
GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer
J. Clin. 2018,68, 394. [CrossRef] [PubMed]
4.
U.S. Breast Cancer Statistics. Available online: https://www.breastcancer.org/symptoms/understand_bc/
statistics (accessed on 13 February 2020).
5.
Campeau, P.M.; Foulkes, W.D.; Tischkowitz, M.D. Hereditary breast cancer: New genetic developments,
new therapeutic avenues. Hum. Genet. 2008,124, 31–42. [CrossRef] [PubMed]
6.
Li, Y.; Li, S.; Meng, X.; Gan, R.Y.; Zhang, J.J.; Li, H.B. Dietary natural products for prevention and treatment
of breast cancer. Nutrients 2017,9, 728. [CrossRef]
7.
Tamimi, R.M.; Spiegelman, D.; Smith-Warner, S.A.; Wang, M.; Pazaris, M.; Willett, W.C.; Eliassen, A.H.;
Hunter, D.J. Population Attributable Risk of Modifiable and Nonmodifiable Breast Cancer Risk Factors in
Postmenopausal Breast Cancer. Am. J. Epidemiol. 2016,184, 884–893. [CrossRef] [PubMed]
8.
Bodai, B.I.; Tuso, P. Breast cancer survivorship: A comprehensive review of long-term medical issues and
lifestyle recommendations. Perm. J. 2015,19, 48–79. [CrossRef]
9.
Timms, K.M.; Abkevich, V.; Hughes, E.; Ne, C.; Reid, J.; Morris, B.; Kalva, S.; Potter, J.; Tran, T.V.;
Chenet, J.; et al.
Association of BRCA1/2 defects with genomic scores predictive of DNA damage repair
deficiency among breast cancer subtypes. Breast Cancer Res. 2014,16, 475. [CrossRef]
10.
Rivenbark, A.G.; O’Connor, S.M.; Coleman, W.B. Molecular and cellular heterogeneity in breast cancer:
Challenges for personalized medicine. Am. J. Pathol. 2013,183, 1113–1124. [CrossRef]
11. Waks, A.G.; Winer, E.P. Breast Cancer Treatment: A Review. JAMA 2019,321, 288–300. [CrossRef]
12.
Elston, C.W.; Ellis, I.O. Pathological prognostic factors in breast cancer. I. The value of histological grade in
breast cancer: Experience from a large study with long-term follow-up. Histopathology
1991
,19, 403–410.
[CrossRef]
13.
Weigelt, B.; Geyer, F.C.; Reis-Filho, J.S. Histological types of breast cancer: How special are they? Mol. Oncol.
2010,4, 192–208. [CrossRef] [PubMed]
14.
Tao, Z.; Shi, A.; Lu, C.; Song, T.; Zhang, Z.; Zhao, J. Breast Cancer: Epidemiology and Etiology.
Cell Biochem. Biophys. 2015,72, 333–338. [CrossRef] [PubMed]
15.
Sawaki, M.; Shien, T.; Iwata, H. TNM classification of malignant tumors (Breast Cancer Study Group). Jpn. J.
Clin. Oncol. 2019,49, 228–231. [CrossRef]
16.
Engstrøm, M.J.; Opdahl, S.; Hagen, A.I.; Romundstad, P.R.; Akslen, L.A.; Haugen, O.A.; Vatten, L.J.;
Bofin, A.M. Molecular subtypes, histopathological grade and survival in a historic cohort of breast cancer
patients. Breast Cancer Res. Treat. 2013,140, 463–473. [CrossRef]
17.
Cadoo, K.A.; Fornier, M.N.; Morris, P.G. Biological subtypes of breast cancer: Current concepts and
implications for recurrence patterns. Q J. Nucl. Med. Mol. Imaging 2013,57, 312–321.
18.
Marotti, J.D.; de Abreu, F.B.; Wells, W.A.; Tsongalis, G.J. Triple-negative breast cancer. Am. J. Pathol.
2017
,
187, 2133–2138. [CrossRef]
19.
Lehmann, B.D.; Bauer, J.A.; Chen, X.; Sanders, M.E.; Chakravarthy, A.B.; Shyr, Y.; Pietenpol, J.A. Identification
of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies.
J. Clin. Investig. 2011,121, 2750–2767. [CrossRef]
20.
Di Cosimo, S.; Baselga, J. Management of breast cancer with targeted agents: Importance of heterogeneity.
Nat. Rev. Clin. Oncol. 2010,7, 139–147. [CrossRef]
21.
Balduzzi, S.; Mantarro, S.; Guarneri, V.; Tagliabue, L.; Pistotti, V.; Moja, L.; D’Amico, R. Trastuzumab
containing regimens for metastatic breast cancer. Cochrane Database Syst. Rev.
2014
,12, CD006242. [CrossRef]
22.
Tang, Y.; Wang, Y.; Kiani, M.F.; Wang, B. Classification, treatment strategy, and associated drug resistance in
breast cancer. Clin. Breast Cancer 2016,16, 335–343. [CrossRef]
23.
Guarneri, V.; Conte, P. Metastatic breast cancer: Therapeutic options according to molecular subtypes and
prior adjuvant therapy. Oncologist 2009,14, 645–656. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2020,21, 5718 26 of 33
24.
Surh, Y.J. Cancer chemoprevention with dietary phytochemicals. Nat. Rev. Cancer
2003
,3, 768–780. [CrossRef]
[PubMed]
25.
Irigaray, P.; Newby, J.A.; Clapp, R.; Hardell, L.; Howard, V.; Montagnier, L.; Epstein, S.; Belpomme, D.
Lifestyle-related factors and environmental agents causing cancer: An overview. Biomed. Pharmacother.
2007
,
61, 640–658. [CrossRef] [PubMed]
26.
Catsburg, C.; Kim, R.S.; Kirsh, V.A.; Soskolne, C.L.; Kreiger, N.; Rohan, T.E. Dietary patterns and breast
cancer risk: A study in 2 cohorts. Am. J. Clin. Nutr. 2015,101, 817–823. [CrossRef]
27.
Aune, D.; Chan, D.S.; Vieira, A.R.; Rosenblatt, D.A.; Vieira, R.; Greenwood, D.C.; Norat, T. Fruits, vegetables
and breast cancer risk: A systematic review and meta-analysis of prospective studies. Breast Cancer Res. Treat.
2012,134, 479–493. [CrossRef] [PubMed]
28.
Farvid, M.S.; Chen, W.Y.; Rosner, B.A.; Tamimi, R.M.; Willett, W.C.; Eliassen, A.H. Fruit and vegetable
consumption and breast cancer incidence: Repeated measures over 30 years of follow-up. Int. J. Cancer
2019
,
144, 1496–1510. [CrossRef]
29.
Emaus, M.J.; Peeter, P.H.; Bakker, M.F.; Overvad, K.; Tjonneland, A.; Olsen, A.; Romieu, I.; Ferrari, P.; Dossus, L.;
Boutron-Ruault, M.C.; et al. Vegetable and fruit consumption and the risk of hormone receptor-defined
breast cancer in the EPIC cohort. Am. J. Clin. Nutr. 2016,103, 168–177. [CrossRef] [PubMed]
30.
Jung, S.; Spiegelman, D.; Baglietto, L.; Bernstein, L.; Boggs, D.A.; van den Brandt, P.A.; Buring, J.E.;
Cerhan, J.R.; Gaudet, M.M.; Giles, G.G.; et al. Fruit and vegetable intake and risk of breast cancer by hormone
receptor status. J. Natl. Cancer Inst. 2013,105, 219–236. [CrossRef]
31.
Heath, A.K.; Muller, D.C.; van den Brandt, P.A.; Papadimitriou, N.; Critselis, E.; Gunter, M.; Vineis, P.;
Weiderpass, E.; Fagherazzi, G.; Boeing, H.; et al. Nutrient-wide association study of 92 foods and nutrients
and breast cancer risk. Breast Cancer Res. 2020,22, 5. [CrossRef]
32.
Karavasiloglou, N.; Hüsing, A.; Masala, G.; van Gils, C.H.; Turzanski Fortner, R.; Chang-Claude, J.;
Huybrechts, I.; Weiderpass, E.; Gunter, M.; Arveux, P.; et al. Adherence to the World Cancer Research
Fund/American Institute for Cancer Research cancer prevention recommendations and risk of in situ breast
cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. BMC Med.
2019
,
17, 221. [CrossRef]
33.
Farvid, M.S.; Chen, W.Y.; Michels, K.B.; Cho, E.; Willett, W.C.; Eliassen, A.H. Fruit and vegetable consumption
in adolescence and early adulthood and risk of breast cancer: Population based cohort study. BMJ
2016
,
353, i2343. [CrossRef] [PubMed]
34.
Rothwell, J.A.; Knaze, V.; Zamora-Ros, R. Polyphenols: Dietary assessment and role in the prevention of
cancers. Curr. Opin. Clin. Nutr. Metab. Care 2017,20, 512–521. [CrossRef] [PubMed]
35.
Grosso, G.; Bella, F.; Godos, J.; Sciacca, S.; Del Rio, D.; Ray, S.; Giovannucci, E.L. Possible role of diet in cancer:
Systematic review and multiple meta-analyses of dietary patterns, lifestyle factors, and cancer risk. Nutr. Res.
2017,75, 405–419. [CrossRef] [PubMed]
36.
Dong, J.Y.; Qin, L.Q. Soy isoflavones consumption and risk of breast cancer incidence or recurrence.
A meta-analysis of prospective studies. Breast Cancer Res. Treat. 2011,125, 315–323. [CrossRef]
37.
Chen, M.; Rao, Y.; Zheng, Y.; Wei, S.; Li, Y.; Guo, T.; Yin, P. Association between soy isoflavone intake
and breast cancer risk for pre- and post-menopausal women: A meta-analysis of epidemiological studies.
PLoS ONE 2014,9, e89288. [CrossRef] [PubMed]
38.
Micek, A.; Godos, J.; Brzostek, T.; Gniadek, A.; Favari, C.; Mena, P.; Libra, M.; Del Rio, D.; Galvano, F.;
Grosso, G. Dietary phytoestrogens and biomarkers of their intake in relation to cancer survival and recurrence:
A comprehensive systematic review with meta-analysis. Nutr. Rev.
2020
, nuaa043, (published online ahead
of print, 6 July 2020). [CrossRef]
39.
Castell
ó
, A.; Poll
á
n, M.; Buijsse, B.; Ruiz, A.; Casas, A.M.; Baena-Cañada, J.M.; Lope, V.; Antol
í
n, S.; Ramos, M.;
Muñoz, M.; et al. Spanish Mediterranean diet and other dietary patterns and breast cancer risk: Case-control
EpiGEICAM study. Br. J. Cancer 2014,111, 1454–1462. [CrossRef]
40.
Castell
ó
, A.; Boldo, E.; P
é
rez-G
ó
mez, B.; Lope, V.; Altzibar, J.M.; Mart
í
n, V.; Castaño-Vinyals, G.; Guevara, M.;
Dierssen-Sotos, T.; Tard
ó
n, A.; et al. Adherence to the Western, Prudent and Mediterranean dietary patterns
and breast cancer risk: MCC-Spain study. Maturitas 2017,103, 8–15. [CrossRef]
41.
Pan, M.H.; Chiou, Y.S.; Chen, L.H.; Ho, C.T. Breast cancer chemoprevention by dietary natural phenolic
compounds: Specific epigenetic related molecular targets. Mol. Nutr. Food Res. 2015,59, 21–35. [CrossRef]
Int. J. Mol. Sci. 2020,21, 5718 27 of 33
42.
Losada-Echeberr
í
a, M.; Herranz-L
ó
pez, M.; Micol, V.; Barraj
ó
n-Catal
á
n, E. Polyphenols as Promising Drugs
Against Main Breast Cancer Signatures. Antioxidants 2017,6, 88. [CrossRef]
43.
Manach, C.; Scalbert, A.; Morand, C.; R
é
m
é
sy, C.; Jim
é
nez, L. Polyphenols: Food sources and bioavailability.
Am. J. Clin. Nutr. 2004,79, 727–747. [CrossRef]
44.
Del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.; Tognolini, M.; Borges, G.; Crozier, A. Dietary (poly)phenolics
in human health: Structures, bioavailability, and evidence of protective eects against chronic diseases.
Antioxid. Redox Signal. 2013,18, 1818–1892. [CrossRef] [PubMed]
45.
Tom
á
s-Barber
á
n, F.A.; Esp
í
n, J.C. Eect of Food Structure and Processing on (Poly)phenol-Gut Microbiota
Interactions and the Eects on Human Health. Annu. Rev. Food Sci. Technol.
2019
,10, 221–238. [CrossRef]
[PubMed]
46.
Gonz
á
lez-Sarr
í
as, A.; Esp
í
n, J.C.; Tom
á
s-Barber
á
n, F.A. Non-extractable polyphenols produce gut microbiota
metabolites that persist in circulation and show anti-inflammatory and free radical-scavenging eects.
Trends Food Sci. Technol. 2017,69, 281–288. [CrossRef]
47.
Selma, M.V.; Esp
í
n, J.C.; Tom
á
s-Barber
á
n, F.A. Interaction between phenolics and gut microbiota: Role in
human health. J. Agric. Food Chem. 2009,57, 6485–6501. [CrossRef] [PubMed]
48.
Esp
í
n, J.C.; Gonz
á
lez-Sarr
í
as, A.; Tom
á
s-Barber
á
n, F.A. The gut microbiota: A key factor in the therapeutic
eects of (poly) phenols. Biochem. Pharmacol. 2017,139, 82–93. [CrossRef]
49.
Aires, V.; Limagne, E.; Cotte, A.K.; Latrue, N.; Ghiringhelli, F.; Delmas, D. Resveratrol metabolites inhibit
human metastatic colon cancer cells progression and synergize with chemotherapeutic drugs to induce cell
death. Mol. Nutr. Food Res. 2013,57, 1170–1181. [CrossRef]
50.
Gonz
á
lez-Sarr
í
as, A.; Gim
é
nez-Bastida, J.A.; N
ú
ñez-S
á
nchez, M.
Á
.; Larrosa, M.; Garc
í
a-Conesa, M.T.;
Tom
á
s-Barber
á
n, F.A.; Esp
í
n, J.C. Phase-II metabolism limits the antiproliferative activity of urolithins in
human colon cancer cells. Eur. J. Nutr. 2014,53, 853–864. [CrossRef]
51. Á
vila-G
á
lvez, M.
Á
.; Esp
í
n, J.C.; Gonz
á
lez-Sarr
í
as, A. Physiological Relevance of the Antiproliferative and
Estrogenic Eects of Dietary Polyphenol Aglycones versus Their Phase-II Metabolites on Breast Cancer Cells:
A Call of Caution. J. Agric. Food Chem. 2018,66, 8547–8555. [CrossRef]
52.
Maubach, J.; Bracke, M.E.; Heyerick, A.; Depypere, H.T.; Serreyn, R.F.; Mareel, M.M.; De Keukeleire, D.
Quantitation of soy-derived phytoestrogens in human breast tissue and biological fluids by high-performance
liquid chromatography. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2003,784, 137–144. [CrossRef]
53.
Maubach, J.; Depypere, H.T.; Goeman, J.; Van Der Eycken, J.; Heyerick, A.; Bracke, M.E.; Blondeel, P.;
De Keukeleire, D. Distribution of soy-derived phytoestrogens in human breast tissue and biological fluids.
Obstet. Gynecol. 2004,103, 892–898. [CrossRef] [PubMed]
54.
Pumford, S.; Morton, M.; Turkes, A.; Griths, K. Determination of the isoflavonoids genistein and daidzein
in biological samples by gas chromatography–mass spectrometry. Ann. Clin. Biochem.
2002
,39, 281–292.
[CrossRef] [PubMed]
55.
Bolca, S.; Urpi-Sarda, M.; Blondeel, P.; Roche, N.; Vanhaecke, L.; Possemiers, S.; Al-Maharik, N.; Botting, N.;
De Keukeleire, D.; Bracke, M.; et al. Disposition of soy isoflavones in normal human breast tissue. Am. J.
Clin. Nutr. 2010,91, 976–984. [CrossRef] [PubMed]
56.
Chang, H.C.; Churchwell, M.I.; Delclos, K.B.; Newbold, R.R.; Doerge, D.R. Mass spectrometric determination
of genistein tissue distribution in diet-exposed Sprague-Dawley rats. J. Nutr.
2000
,130, 1963–1970. [CrossRef]
[PubMed]
57.
Lamartiniere, C.A.; Wang, J.; Smith-Johnson, M.; Eltoum, I.E. Daidzein: Bioavailability, potential for
reproductive toxicity, and breast cancer chemoprevention in female rats. Toxicol. Sci.
2002
,65, 228–238.
[CrossRef]
58.
Urpi-Sarda, M.; Morand, C.; Besson, C.; Kraft, G.; Viala, D.; Scalbert, A.; Besle, J.M.; Manach, C. Tissue
distribution of isoflavones in ewes after consumption of red clover silage. Arch. Biochem. Biophys.
2008
,476,
205–210. [CrossRef]
59.
Yuan, L.; Wagatsuma, C.; Yoshida, M.; Miura, T.; Mukoda, T.; Fujii, H.; Sun, B.; Kim, J.H.; Surh, Y.J. Inhibition
of human breast cancer growth by GCP (genistein combined polysaccharide) in xenogeneic athymic mice:
Involvement of genistein biotransformation by beta-glucuronidase from tumor tissues. Mutat. Res.
2003
,
523, 55–62. [CrossRef]
Int. J. Mol. Sci. 2020,21, 5718 28 of 33
60.
Bolca, S.; Li, J.; Nikolic, D.; Roche, N.; Blondeel, P.; Possemiers, S.; De Keukeleire, D.; Bracke, M.; Heyerick, A.;
van Breemen, R.B.; et al. Disposition of hop prenylflavonoids in human breast tissue. Mol. Nutr. Food Res.
2010,54, S284–S294. [CrossRef]
61.
Overk, C.R.; Guo, J.; Chadwick, L.R.; Lantvit, D.D.; Minassi, A.; Appendino, G.; Chen, S.N.; Lankin, D.C.;
Farnsworth, N.R.; Pauli, G.F.; et al.
In vivo
estrogenic comparisons of Trifolium pratense (red clover), Humulus
lupulus (hops), and the pure compounds isoxanthohumol and 8-prenylnaringenin.
Chem. Biol. Interact. 2008
,
176, 30–39. [CrossRef]
62.
Dietz, B.M.; Hagos, G.K.; Eskra, J.N.; Wijewickrama, G.T.; Anderson, J.R.; Nikolic, D.; Wright, B.; Chen, S.-N.;
Pauli, G.F.; van Breemen, R.B.; et al. Dierential regulation of detoxification enzymes in hepatic and mammary
tissue by hops (Humulus lupulus)
in vitro
and
in vivo
.Mol. Nutr. Food Res.
2013
,57, 1055–1066. [CrossRef]
63.
Albin, N.; Massaad, L.; Toussaint, C.; Mathieu, M.C.; Morizet, J.; Parise, O.; Gouyette, A.; Chabot, G.G. Main
Drug-Metabolizing Enzyme Systems in Human Breast Tumors and Peritumoral Tissues. Cancer Res.
1993
,
53, 3541. [PubMed]
64.
Shimoi, K.; Nakayama, T. Glucuronidase deconjugation in inflammation. Methods Enzymol.
2005
,400,
263–272. [CrossRef] [PubMed]
65.
Lazzeroni, M.; Guerrieri-Gonzaga, A.; Gandini, S.; Johansson, H.; Serrano, D.; Cazzaniga, M.; Aristarco, V.;
Puccio, A.; Mora, S.; Caldarella, P.; et al. A presurgical study of oral silybin-phosphatidylcholine in patients
with early breast cancer. Cancer Prev. Res. 2016,9, 89–95. [CrossRef] [PubMed]
66.
Lazzeroni, M.; Guerrieri-Gonzaga, A.; Gandini, S.; Johansson, H.; Serrano, D.; Cazzaniga, M.; Aristarco, V.;
Macis, D.; Mora, S.; Caldarella, P.; et al. A Presurgical Study of Lecithin Formulation of Green Tea Extract in
Women with Early Breast Cancer. Cancer Prev. Res. 2017,10, 363–370. [CrossRef]
67.
Sartippour, M.R.; Pietras, R.; Marquez-Garban, D.C.; Chen, H.-W.; Heber, D.; Henning, S.M.; Sartippour, G.;
Zhang, L.; Lu, M.; Weinberg, O.; et al. The combination of green tea and tamoxifen is eective against breast
cancer. Carcinogenesis 2006,27, 2424–2433. [CrossRef]
68.
Kaur, S.; Greaves, P.; Cooke, D.N.; Edwards, R.; Steward, W.P.; Gescher, A.J.; Marczylo, T.H. Breast Cancer
Prevention by Green Tea Catechins and Black Tea Theaflavins in the C3(1) SV40 T,t Antigen Transgenic
Mouse Model Is Accompanied by Increased Apoptosis and a Decrease in Oxidative DNA Adducts. J. Agric.
Food Chem. 2007,55, 3378–3385. [CrossRef]
69. Á
vila-G
á
lvez, M.
Á
.; Garc
í
a-Villalba, R.; Mart
í
nez-D
í
az, F.; Ocaña-Castillo, B.; Monedero-Saiz, T.;
Torrecillas-S
á
nchez, A.; Abell
á
n, B.; Gonz
á
lez-Sarr
í
as, A.; Esp
í
n, J.C. Metabolic Profiling of Dietary
Polyphenols and Methylxanthines in Normal and Malignant Mammary Tissues from Breast Cancer Patients.
Mol. Nutr. Food Res. 2019,63, e1801239. [CrossRef]
70. Á
vila-G
á
lvez, M.
Á
.; Romo-Vaquero, M.; Gonz
á
lez-Sarr
í
as, A.; Esp
í
n, J.C. Kinetic disposition of dietary
polyphenols and methylxanthines in the rat mammary tissue. J. Funct. Foods 2019,61, 103516. [CrossRef]
71.
Lærke, H.N.; Mortensen, M.A.; Hedemann, M.S.; Bach-Knudsen, K.E.; Penalvo, J.L.; Adlercreutz, H.
Quantitative aspects of the metabolism of lignans in pigs fed fibre-enriched rye and wheat bread. Br. J. Nutr.
2009,102, 985–994. [CrossRef]
72.
Garc
í
a-Mateos, D.; Garc
í
a-Villalba, R.; Marañ
ó
n, J.A.; Esp
í
n, J.C.; Merino, G.;
Á
lvarez, A.I. The Breast Cancer
Resistance Protein (BCRP/ABCG2) influences the levels of enterolignans and their metabolites in plasma,
milk and mammary gland. J. Funct. Foods 2017,35, 648–654. [CrossRef]
73.
Liu, G.; Khanna, V.; Kirtane, A.; Grill, A.; Panyam, J. Chemopreventive ecacy of oral curcumin: A prodrug
hypothesis. FASEB J. 2019,33, 9453–9465. [CrossRef] [PubMed]
74.
Kunihiro, A.G.; Brickey, J.A.; Frye, J.B.; Luis, P.B.; Schneider, C.; Funk, J.L. Curcumin, but not curcumin-
glucuronide, inhibits Smad signaling in TGF
β
-dependent bone metastatic breast cancer cells and is enriched
in bone compared to other tissues. J. Nutr. Biochem. 2019,63, 150–156. [CrossRef] [PubMed]
75.
De Pace, R.C.; Liu, X.; Sun, M.; Nie, S.; Zhang, J.; Cai, Q.; Gao, W.; Pan, X.; Fan, Z.; Wang, S. Anticancer activities
of (-)-epigallocatechin-3-gallate encapsulated nanoliposomes in MCF7 breast cancer cells.
J. Liposome Res.
2013,23, 187–196. [CrossRef] [PubMed]
76.
Phan, V.; Walters, J.; Brownlow, B.; Elbayoumi, T. Enhanced cytotoxicity of optimized liposomal genistein via
specific induction of apoptosis in breast, ovarian and prostate carcinomas. J. Drug Target
2013
,21, 1001–1011.
[CrossRef]
Int. J. Mol. Sci. 2020,21, 5718 29 of 33
77.
Khan, M.N.; Haggag, Y.A.; Lane, M.E.; McCarron, P.A.; Tambuwala, M.M. Polymeric Nano-Encapsulation of
Curcumin Enhances its Anti-Cancer Activity in Breast (MDA-MB231) and Lung (A549) Cancer Cells Through
Reduction in Expression of HIF-1
α
and Nuclear p65 (Rel A). Curr. Drug Deliv.
2018
,15, 286–295. [CrossRef]
78. Á
vila-G
á
lvez, M.
Á
.; Gonz
á
lez-Sarr
í
as, A.; Esp
í
n, J.C. In Vitro Research on Dietary Polyphenols and Health:
A Call of Caution and a Guide on How To Proceed. J. Agric. Food Chem. 2018,66, 7857–7858. [CrossRef]
79.
Power, K.A.; Saarinen, N.M.; Chen, J.M.; Thompson, L.U. Mammalian lignans enterolactone and enterodiol,
alone and in combination with the isoflavone genistein, do not promote the growth of MCF-7 xenografts in
ovariectomized athymic nude mice. Int. J. Cancer 2018,118, 1316–1320. [CrossRef]
80.
Saarinen, N.M.; Power, K.; Chen, J.; Thompson, L.U. Flaxseed attenuates the tumor growth stimulating eect
of soy protein in ovariectomized athymic mice with MCF-7 human breast cancer xenografts. Int. J. Cancer
2006,119, 925–931. [CrossRef]
81.
Somers-Edgar, T.J.; Scandlyn, M.J.; Stuart, E.C.; Le Nedelec, M.J.; Valentine, S.P.; Rosengren, R.J.
The combination of epigallocatechin gallate and curcumin suppresses ER
α
-breast cancer cell growth
in vitro and in vivo. Int. J. Cancer 2007,122, 1966–1971. [CrossRef]
82.
Schlachterman, A.; Valle, F.; Wall, K.M.; Azios, N.G.; Castillo, L.; Morell, L.; Washington, A.V.; Cubano, L.A.;
Dharmawardhane, S.F. Combined Resveratrol, Quercetin, and Catechin Treatment Reduces Breast Tumor
Growth in a Nude Mouse Model. Transl. Oncol. 2008,1, 19–27. [CrossRef]
83.
Mohapatra, P.; Satapathy, S.R.; Siddharth, S.; Das, D.; Nayak, A.; Kundu, C.N. Resveratrol and curcumin
synergistically induces apoptosis in cigarette smoke condensate transformed breast epithelial cells through a
p21Waf1/Cip1 mediated inhibition of Hh-Gli signaling. Int. J. Biochem. Cell Biol.
2015
,66, 75–84. [CrossRef]
[PubMed]
84.
Tripathi, S.; Kushwah, V.; Thanki, K.; Jain, S. Triple antioxidant SNEDDS formulation with enhanced oral
bioavailability: Implication of chemoprevention of breast cancer. Nanomed. Nanotechnol. Biol. Med.
2016
,12,
1431–1443. [CrossRef] [PubMed]
85.
Fritz, W. Dietary genistein: Perinatal mammary cancer prevention, bioavailability and toxicity testing in the
rat. Carcinogenesis 1998,19, 2151–2158. [CrossRef] [PubMed]
86.
Rowlands, J.C.; He, L.; Hakkak, R.; Ronis, M.J.J.; Badger, T.M. Soy and Whey Proteins Downregulate
DMBA-Induced Liver and Mammary Gland CYP1 Expression in Female Rats. J. Nutr.
2001
,131, 3281–3287.
[CrossRef]
87.
Papoutsis, A.J.; Selmin, O.I.; Borg, J.L.; Romagnolo, D.F. Gestational exposure to the AhR agonist
2,3,7,8-tetrachlorodibenzo-p-dioxin induces BRCA-1 promoter hypermethylation and reduces BRCA-1
expression in mammary tissue of rat ospring: Preventive eects of resveratrol: GESTATIONAL EXPOSURE
TO THE AhR AGONIST 2,3,7,8-TETRACHLORODIBENZO-p-DIOXIN. Mol. Carcinog.
2015
,54, 261–269.
[CrossRef]
88.
Jung, K.-J.; Wallig, M.A.; Singletary, K.W. Purple grape juice inhibits 7,12-dimethylbenz[a]anthracene
(DMBA)-induced rat mammary tumorigenesis and
in vivo
DMBA-DNA adduct formation. Cancer Lett.
2006
,
233, 279–288. [CrossRef]
89.
Chatterjee, M.; Das, S.; Janarthan, M.; Ramachandran, H.K.; Chatterjee, M. Role of 5-lipoxygenase in
resveratrol mediated suppression of 7,12-dimethylbenz(
α
)anthracene-induced mammary carcinogenesis in
rats. Eur. J. Pharmacol. 2011,668, 99–106. [CrossRef]
90.
Rahal, O.M.; Machado, H.L.; Montales, M.T.E.; Pabona, J.M.P.; Heard, M.E.; Nagarajan, S.; Simmen, R.C.M.
Dietary suppression of the mammary CD29hiCD24+epithelial subpopulation and its cytokine/chemokine
transcriptional signatures modifies mammary tumor risk in MMTV-Wnt1 transgenic mice. Stem Cell Res.
2013,11, 1149–1162. [CrossRef]
91.
Ye, L.; Chan, F.L.; Chen, S.; Leung, L.K. The citrus flavonone hesperetin inhibits growth of aromatase-
expressing MCF-7 tumor in ovariectomized athymic mice. J. Nutr. Biochem.
2012
,23, 1230–1237. [CrossRef]
92.
Li, F.; Wong, T.Y.; Lin, S.; Chow, S.; Cheung, W.; Chan, F.L.; Chen, S.; Leung, L.K. Coadministrating Luteolin
Minimizes the Side Eects of the Aromatase Inhibitor Letrozole. J. Pharmacol. Exp. Ther.
2014
,351, 270–277.
[CrossRef]
93.
Mai, Z.; Blackburn, G.L.; Zhou, J.-R. Soy phytochemicals synergistically enhance the preventive eect of
tamoxifen on the growth of estrogen-dependent human breast carcinoma in mice. Carcinogenesis
2007
,28,
1217–1223. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2020,21, 5718 30 of 33
94.
Mandal, A.; Bishayee, A. Mechanism of Breast Cancer Preventive Action of Pomegranate: Disruption of
Estrogen Receptor and Wnt/
β
-Catenin Signaling Pathways. Molecules
2015
,20, 22315–22328. [CrossRef]
[PubMed]
95.
Ezzat, S.M.; Shouman, S.A.; Elkhoely, A.; Attia, Y.M.; Elsesy, M.S.; El Senousy, A.S.; Choucry, M.A.;
El Gayed, S.H.; El Sayed, A.A.; Sattar, E.A.; et al. Anticancer potentiality of lignan rich fraction of six Flaxseed
cultivars. Sci. Rep. 2018,8, 544. [CrossRef] [PubMed]
96.
Chen, J.; Saggar, J.K.; Corey, P.; Thompson, L.U. Flaxseed and Pure Secoisolariciresinol Diglucoside, but Not
Flaxseed Hull, Reduce Human Breast Tumor Growth (MCF-7) in Athymic Mice. J. Nutr.
2009
,139, 2061–2066.
[CrossRef]
97.
Harris, R.; Beebe, J.; Schwartzbaum, J. Chemoprevention of breast cancer by cyclooxygenase and lipoxygenase
inhibitors. World Acad. Sci. J. 2020,2, 14–18. [CrossRef]
98.
Esquivel-Vel
á
zquez, M.; Ostoa-Saloma, P.; Palacios-Arreola, M.I.; Nava-Castro, K.E.; Castro, J.I.;
Morales-Montor, J. The Role of Cytokines in Breast Cancer Development and Progression. J. Interferon
Cytokine Res. 2015,35, 1–16. [CrossRef]
99.
Banerjee, S.; Bueso-Ramos, C.; Aggarwal, B.B. Suppression of 7,12-dimethylbenz(a)anthracene-induced
mammary carcinogenesis in rats by resveratrol: Role of nuclear factor-kappaB, cyclooxygenase 2, and matrix
metalloprotease 9. Cancer Res. 2002,62, 4945–4954.
100.
Ke, J.-Y.; Banh, T.; Hsiao, Y.-H.; Cole, R.M.; Straka, S.R.; Yee, L.D.; Belury, M.A. Citrus flavonoid naringenin
reduces mammary tumor cell viability, adipose mass, and adipose inflammation in obese ovariectomized
mice. Mol. Nutr. Food Res. 2017,61, 1600934. [CrossRef]
101.
Yao, Y.; Zhao, K.; Yu, Z.; Ren, H.; Zhao, L.; Li, Z.; Guo, Q.; Lu, N. Wogonoside inhibits invasion and migration
through suppressing TRAF2/4 expression in breast cancer. J. Exp. Clin. Cancer Res.
2017
,36, 103. [CrossRef]
102.
Du, G.; Lin, H.; Yang, Y.; Zhang, S.; Wu, X.; Wang, M.; Ji, L.; Lu, L.; Yu, L.; Han, G. Dietary quercetin
combining intratumoral doxorubicin injection synergistically induces rejection of established breast cancer
in mice. Int. Immunopharmacol. 2010,10, 819–826. [CrossRef]
103.
Forghani, P.; Khorramizadeh, M.R.; Waller, E.K. Silibinin inhibits accumulation of myeloid-derived suppressor
cells and tumor growth of murine breast cancer. Cancer Med. 2014,3, 215–224. [CrossRef] [PubMed]
104.
Qin, L.; Jin, L.; Lu, L.; Lu, X.; Zhang, C.; Zhang, F.; Liang, W. Naringenin reduces lung metastasis in a breast
cancer resection model. Protein Cell 2011,2, 507–516. [CrossRef] [PubMed]
105.
Xu, P.; Yan, F.; Zhao, Y.; Chen, X.; Sun, S.; Wang, Y.; Ying, L. Green Tea Polyphenol EGCG Attenuates
MDSCs-mediated Immunosuppression through Canonical and Non-Canonical Pathways in a 4T1 Murine
Breast Cancer Model. Nutrients 2020,12, 1042. [CrossRef] [PubMed]
106.
Yuan, B.; Wang, L.; Jin, Y.; Zhen, H.; Xu, P.; Xu, Y.; Li, C.; Xu, H. Role of Metabolism in the Eects of Genistein
and Its Phase II Conjugates on the Growth of Human Breast Cell Lines. AAPS J.
2012
,14, 329–344. [CrossRef]
[PubMed]
107.
Wu, Q.; Kroon, P.A.; Shao, H.; Needs, P.W.; Yang, X. Dierential Eects of Quercetin and Two of Its Derivatives,
Isorhamnetin and Isorhamnetin-3-glucuronide, in Inhibiting the Proliferation of Human Breast-Cancer
MCF-7 Cells. J. Agric. Food Chem. 2018,66, 7181–7189. [CrossRef]
108.
Wu, Q.; Needs, P.W.; Lu, Y.; Kroon, P.A.; Ren, D.; Yang, X. Dierent antitumor eects of quercetin,
quercetin-3
0
-sulfate and quercetin-3-glucuronide in human breast cancer MCF-7 cells. Food Funct.
2018
,9,
1736–1746. [CrossRef]
109.
Gim
é
nez-Bastida, J.A.;
Á
vila-G
á
lvez, M.
Á
.; Esp
í
n, J.C.; Gonz
á
lez-Sarr
í
as, A. Conjugated Physiological
Resveratrol Metabolites Induce Senescence in Breast Cancer Cells: Role of p53/p21 and p16/Rb Pathways,
and ABC Transporters. Mol. Nutr. Food Res. 2019,63, 1900629. [CrossRef]
110. Á
vila-G
á
lvez, M.
Á
.; Gim
é
nez-Bastida, J.A.; Gonz
á
lez-Sarr
í
as, A.; Esp
í
n, J.C. Tissue deconjugation of urolithin
A glucuronide to free urolithin A in systemic inflammation. Food Funct. 2019,10, 3135–3141. [CrossRef]
111.
Miksits, M.; Wlcek, K.; Svoboda, M.; Kunert, O.; Haslinger, E.; Thalhammer, T.; Szekeres, T.; Jäger, W.
Antitumor Activity of Resveratrol and its Sulfated Metabolites against Human Breast Cancer Cells. Planta Med.
2009,75, 1227–1230. [CrossRef]
112.
Hoshino, J.; Park, E.-J.; Kondratyuk, T.P.; Marler, L.; Pezzuto, J.M.; van Breemen, R.B.; Mo, S.; Li, Y.;
Cushman, M. Selective Synthesis and Biological Evaluation of Sulfate-Conjugated Resveratrol Metabolites.
J. Med. Chem. 2010,53, 5033–5043. [CrossRef]
Int. J. Mol. Sci. 2020,21, 5718 31 of 33
113.
Ruotolo, R.; Calani, L.; Fietta, E.; Brighenti, F.; Crozier, A.; Meda, C.; Maggi, A.; Ottonello, S.; Del Rio, D.
Anti-estrogenic activity of a human resveratrol metabolite. Nutr. Metab. Cardiovasc. Dis.
2013
,23, 1086–1092.
[CrossRef] [PubMed]
114.
Wang, N.; Ren, D.; Deng, S.; Yang, X. Differential effects of baicalein and its sulfated derivatives in inhibiting
proliferation of human breast cancer MCF-7
cells. Chem. Biol. Interact. 2014,
221, 99–108. [CrossRef] [PubMed]
115.
Delgado, L.; Fernandes, I.; Gonz
á
lez-Manzano, S.; de Freitas, V.; Mateus, N.; Santos-Buelga, C. Anti-proliferative
eects of quercetin and catechin metabolites. Food Funct. 2014,5, 797. [CrossRef] [PubMed]
116.
Ruotolo, R.; Calani, L.; Brighenti, F.; Crozier, A.; Ottonello, S.; Del Rio, D. Glucuronidation does not
suppress the estrogenic activity of quercetin in yeast and human breast cancer cell model systems.
Arch. Biochem. Biophys. 2014,559, 62–67. [CrossRef]
117.
Yamazaki, S.; Miyoshi, N.; Kawabata, K.; Yasuda, M.; Shimoi, K. Quercetin-3-O-glucuronide inhibits
noradrenaline-promoted invasion of MDA-MB-231 human breast cancer cells by blocking b2-adrenergic
signaling. Arch. Biochem. Biophys. 2014,557, 18–27. [CrossRef]
118.
Yamazaki, S.; Sakakibara, H.; Takemura, H.; Yasuda, M.; Shimoi, K. Quercetin-3-O-glucronide inhibits
noradrenaline binding to
α
2-adrenergic receptor, thus suppressing DNA damage induced by treatment with
4-hydroxyestradiol and noradrenaline in MCF-10A cells. J. Steroid Biochem. Mol. Biol.
2014
,143, 122–129.
[CrossRef]
119.
Kinjo, J.; Tsuchihashi, R.; Morito, K.; Hirose, T.; Aomori, T.; Nagao, T.; Okabe, H.; Nohara, T.; Masamune, Y.
Interactions of phytoestrogens with estrogen receptors alpha and beta (III). Estrogenic activities of soy
isoflavone aglycones and their metabolites isolated from human urine. Biol. Pharm. Bull. 2004,27, 185–188.
[CrossRef]
120.
Pritchett, L.; Atherton, K.; Mutch, E.; Ford, D. Glucuronidation of the soyabean isoflavones genistein and
daidzein by human liver is related to levels of UGT1A1 and UGT1A9 activity and alters isoflavone response
in the MCF-7 human breast cancer cell line. J. Nutr. Biochem. 2008,19, 739–745. [CrossRef]
121.
Pugazhendhi, D.; Watson, K.A.; Mills, S.; Botting, N.; Pope, G.S.; Darbre, P.D. Eect of sulphation on the
oestrogen agonist activity of the phytoestrogens genistein and daidzein in MCF-7 human breast cancer cells.
J. Endocrinol. 2008,197, 503–515. [CrossRef]
122.
Lin, Y.-J.; Hou, Y.C.; Lin, C.-H.; Hsu, Y.-A.; Sheu, J.J.C.; Lai, C.-H.; Chen, B.-H.; Lee Chao, P.-D.; Wan, L.;
Tsai, F.-J. Puerariae radix isoflavones and their metabolites inhibit growth and induce apoptosis in breast
cancer cells. Biochem. Biophys. Res. Commun. 2009,378, 683–688. [CrossRef]
123.
Islam, M.A.; Bekele, R.; vanden Berg, J.H.J.; Kuswanti, Y.; Thapa, O.; Soltani, S.; van Leeuwen, F.X.R.;
Rietjens, I.M.C.M.; Murk, A.J. Deconjugation of soy isoflavone glucuronides needed for estrogenic activity.
Toxicol. In Vitro 2015,29, 706–715. [CrossRef] [PubMed]
124.
Montalesi, E.; Cipolletti, M.; Cracco, P.; Fiocchetti, M.; Marino, M. Divergent Eects of Daidzein and Its
Metabolites on Estrogen-Induced Survival of Breast Cancer Cells. Cancers
2020
,12, 167. [CrossRef] [PubMed]
125.
Thompson, L.U.; Chen, J.M.; Li, T.; Strasser-Weippl, K.; Goss, P.E. Dietary flaxseed alters tumor biological
markers in postmenopausal breast cancer. Clin. Cancer Res. 2005,11, 3828–3835. [CrossRef] [PubMed]
126.
McCann, S.E.; Edge, S.B.; Hicks, D.G.; Thompson, L.U.; Morrison, C.D.; Fetterly, G.; Andrews, C.; Clark, K.;
Wilton, J.; Kulkarni, S. A pilot study comparing the eect of flaxseed, aromatase inhibitor, and the combination
on breast tumor biomarkers. Nutr. Cancer. 2014,66, 566–575. [CrossRef]
127.
Ferreira Almeida, C.; Oliveira, A.; Jo
ã
o Ramos, M.; Fernandes, P.A.; Teixeira, N.; Amaral, C. Estrogen
receptor-positive (ER
+
) breast cancer treatment: Are multi-target compounds the next promising approach?
Biochem. Pharmacol. 2020,177, 113989. [CrossRef]
128.
Yu, S.S.; Spicer, D.V.; Hawes, D.; Tseng, C.C.; Yang, C.S.; Pike, M.C.; Wu, A.H. Biological eects of green tea
capsule supplementation in pre-surgery postmenopausal breast cancer patients. Front. Oncol.
2013
,3, 298.
[CrossRef]
129.
Crew, K.D.; Brown, P.; Greenlee, H.; Bevers, T.B.; Arun, B.; Hudis, C.; McArthur, H.L.; Chang, J.; Rimawi, M.;
Vornik, L.; et al. Phase IB randomized, double-blinded, placebo-controlled, dose escalation study of
Ppolyphenon E in women with hormone receptor-negative breast cancer. Cancer Prev. Res. (Phila)
2012
,5,
1144–1154. [CrossRef]
130.
Crew, K.D.; Ho, K.A.; Brown, P.; Greenlee, H.; Bevers, T.B.; Arun, B.; Sneige, N.; Hudis, C.; McArthur, H.L.;
Chang, J.; et al
. Effects of a green tea extract, Polyphenon E, on systemic biomarkers of growth factor signalling
in women with hormone receptor-negative breast cancer. J. Hum. Nutr. Diet 2015,28, 272–282. [CrossRef]
Int. J. Mol. Sci. 2020,21, 5718 32 of 33
131.
Davis, S.R.; Wahlin-Jacobsen, S. Testosterone in women–the clinical significance. Lancet Diabetes Endocrinol.
2015,3, 980–992. [CrossRef]
132.
Glaser, R.; Dimitrakakis, C. Testosterone and breast cancer prevention. Maturitas
2015
,82, 291–295. [CrossRef]
133.
Bayet-Robert, M.; Kwiatkowski, F.; Leheurteur, M.; Gachon, F.; Planchat, E.; Abrial, C.; Mouret-Reynier, M.A.;
Durando, X.; Barthomeuf, C.; Chollet, P. Phase I dose escalation trial of docetaxel plus curcumin in patients
with advanced and metastatic breast cancer. Cancer Biol. Ther. 2010,9, 8–14. [CrossRef] [PubMed]
134.
Ryan, J.L.; Heckler, C.E.; Ling, M.; Katz, A.; Williams, J.P.; Pentland, A.P.; Morrow, G.R. Curcumin for
radiation dermatitis: A randomized, double-blind, placebo-controlled clinical trial of thirty breast cancer
patients. Radiat. Res. 2013,180, 34–43. [CrossRef] [PubMed]
135.
Ryan Wolf, J.; Heckler, C.E.; Guido, J.J.; Peoples, A.R.; Gewandter, J.S.; Ling, M.; Vinciguerra, V.P.; Anderson, T.;
Evans, L.; Wade, J.; et al. Oral curcumin for radiation dermatitis: A URCC NCORP study of 686 breast cancer
patients. Support. Care Cancer 2018,6, 1543–1552. [CrossRef] [PubMed]
136.
Brooker, S.; Martin, S.; Pearson, A.; Bagchi, D.; Earl, J.; Gothard, L.; Hall, E.; Porter, L.; Yarnold, J. Double-blind,
placebo-controlled, randomised phase II trial of IH636 grape seed proanthocyanidin extract (GSPE) in
patients with radiation-induced breast induration. Radiother. Oncol. 2006,79, 45–51. [CrossRef] [PubMed]
137.
Ferraris, C.; Ballestra, B.; Listorti, C.; Cappelletti, V.; Reduzzi, C.; Scaperrotta, G.P.; Pulice, I.; Ferrari, E.G.A.;
Folli, S.; Mariani, L.; et al. Red clover and lifestyle changes to contrast menopausal symptoms in
premenopausal patients with hormone-sensitive breast cancer receiving tamoxifen. Breast Cancer Res. Treat.
2020,180, 157–165. [CrossRef]
138.
Hardman, W.E.; Primerano, D.A.; Legenza, M.T.; Morgan, J.; Fan, J.; Denvir, J. Dietary walnut altered gene
expressions related to tumor growth, survival, and metastasis in breast cancer patients: A pilot clinical trial.
Nutr. Res. 2019,66, 82–94. [CrossRef]
139.
Mart
í
nez, N.; Herrera, M.; Fr
í
as, L.; Provencio, M.; P
é
rez-Carri
ó
n, R.; D
í
az, V.; Morse, M.; Crespo, M.C.
A combination of hydroxytyrosol, omega-3 fatty acids and curcumin improves pain and inflammation
among early stage breast cancer patients receiving adjuvant hormonal therapy: Results of a pilot study.
Clin. Transl. Oncol. 2019,21, 489–498. [CrossRef]
140.
N
ú
ñez-S
á
nchez, M.A.; Gonz
á
lez-Sarr
í
as, A.; Romo-Vaquero, M.; Garc
í
a-Villalba, R.; Selma, M.V.;
Tom
á
s-Barber
á
n, F.A.; Garc
í
a-Conesa, M.T.; Esp
í
n, J.C. Dietary phenolics against colorectal cancer—From
promising preclinical results to poor translation into clinical trials: Pitfalls and future needs. Mol. Nutr.
Food Res. 2015,59, 1274–1291. [CrossRef]
141.
Nuñez-S
á
nchez, M.A.; Gonz
á
lez-Sarr
í
as, A.; Garc
í
a-Villalba, R.; Monedero-Saiz, T.; Garc
í
a-Talavera, N.V.;
G
ó
mez-S
á
nchez, M.B.; S
á
nchez-
Á
lvarez, C.; Garc
í
a-Albert, A.M.; Rodr
í
guez-Gil, F.J.;
Ruiz-Marín, M.; et al
.
Gene expression changes in colon tissues from colorectal cancer patients following the intake of an
ellagitannin-containing pomegranate extract: A randomized clinical trial. J. Nutr. Biochem.
2017
,42, 126–133.
[CrossRef]
142.
Pierce, B.L.; Ballard-Barbash, R.; Bernstein, L.; Baumgartner, R.N.; Neuhouser, M.L.; Wener, M.H.;
Baumgartner, K.B.; Gilliland, F.D.; Sorensen, B.E.; McTiernan, A.; et al. Elevated biomarkers of inflammation
are associated with reduced survival among breast cancer patients. J. Clin. Oncol.
2009
,27, 3437–3444. [CrossRef]
143.
Tom
é
-Carneiro, J.; Gonz
á
lvez, M.; Larrosa, M.; Y
á
ñez-Gasc
ó
n, M.J.; Garc
í
a-Almagro, F.J.; Ruiz-Ros, J.A.;
Garc
í
a-Conesa, M.T.; Tom
á
s-Barber
á
n, F.A.; Esp
í
n, J.C. One-year consumption of a grape nutraceutical
containing resveratrol improves the inflammatory and fibrinolytic status of patients in primary prevention
of cardiovascular disease. Am. J. Cardiol. 2012,110, 356–363. [CrossRef] [PubMed]
144.
Manach, C.; Milenkovic, D.; Van de Wiele, T.; Rodriguez-Mateos, A.; de Roos, B.; Garcia-Conesa, M.T.;
Landberg, R.; Gibney, E.R.; Heinonen, M.; Tom
á
s-Barber
á
n, F.; et al. Addressing the inter-individual variation
in response to consumption of plant food bioactives: Towards a better understanding of their role in healthy
aging and cardiometabolic risk reduction. Mol. Nutr. Food Res. 2017,61, 1600557. [CrossRef] [PubMed]
145.
Gibney, E.R.; Milenkovic, D.; Combet, E.; Ruskovska, T.; Greyling, A.; Gonz
á
lez-Sarr
í
as, A.; de Roos, B.;
Tom
á
s-Barber
á
n, F.; Morand, C.; Rodriguez-Mateos, A. Factors influencing the cardiometabolic response to
(poly)phenols and phytosterols: A review of the COST Action POSITIVe activities. Eur. J. Nutr.
2019
,58
(Suppl. 2), 37–47. [CrossRef] [PubMed]
146.
Cort
é
s-Mart
í
n, A.; Selma, M.V.; Tom
á
s-Barber
á
n, F.A.; Gonz
á
lez-Sarr
í
as, A.; Esp
í
n, J.C. Where to Look
into the Puzzle of Polyphenols and Health? The Postbiotics and Gut Microbiota Associated with Human
Metabotypes. Mol. Nutr. Food Res. 2020,64, e1900952. [CrossRef]
Int. J. Mol. Sci. 2020,21, 5718 33 of 33
147.
Vallejo, F.; Larrosa, M.; Escudero, E.; Zafrilla, M.P.; Cerd
á
, B.; Boza, J.; Garc
í
a-Conesa, M.T.; Esp
í
n, J.C.;
Tom
á
s-Barber
á
n, F.A. Concentration and solubility of flavanones in orange beverages aect their
bioavailability in humans. J. Agric. Food Chem. 2010,58, 6516–6524. [CrossRef]
148.
Quartieri, A.; Garc
í
a-Villalba, R.; Amaretti, A.; Raimondi, S.; Leonardi, A.; Rossi, M.; Tom
à
s-Barber
à
n, F.
Detection of novel metabolites of flaxseed lignans
in vitro
and
in vivo
. Detection of novel metabolites of
flaxseed lignans in vitro and in vivo. Mol. Nutr. Food Res. 2016,60, 1590–1601. [CrossRef]
149.
Tom
á
s-Barber
á
n, F.A.; Gonz
á
lez-Sarr
í
as, A.; Garc
í
a-Villalba, R.; N
ú
ñez-S
á
nchez, M.A.; Selma, M.V.;
Garc
í
a-Conesa, M.T.; Esp
í
n, J.C. Urolithins, the rescue of “old” metabolites to understand a “new” concept:
Metabotypes as a nexus among phenolic metabolism, microbiota dysbiosis, and host health status. Mol. Nutr.
Food Res. 2017,61. [CrossRef]
150.
Cort
é
s-Mart
í
n, A.; Selma, M.V.; Esp
í
n, J.C.; Garc
í
a-Villalba, R. The Human Metabolism of Nuts
Proanthocyanidins does not Reveal Urinary Metabolites Consistent with Distinctive Gut Microbiota
Metabotypes. Mol. Nutr. Food Res. 2019,63, e1800819. [CrossRef]
151.
Mena, P.; Ludwig, I.A.; Tomatis, V.B.; Acharjee, A.; Calani, L.; Rosi, A.; Brighenti, F.; Ray, S.; Grin, J.L.;
Bluck, L.J.; et al. Inter-individual variability in the production of flavan-3-ol colonic metabolites: Preliminary
elucidation of urinary metabotypes. Eur. J. Nutr. 2019,58, 1529–1543. [CrossRef]
152.
Romo-Vaquero, M.; Cort
é
s-Mart
í
n, A.; Loria-Kohen, V.; Ram
í
rez-de-Molina, A.; Garc
í
a-Mantrana, I.;
Collado, M.C.; Esp
í
n, J.C.; Selma, M.V. Deciphering the Human Gut Microbiome of Urolithin Metabotypes:
Association with Enterotypes and Potential Cardiometabolic Health Implications. Mol. Nutr. Food Res.
2019
,
63, e1800958. [CrossRef]
153.
Frankenfeld, C.L. Cardiometabolic risk and gut microbial phytoestrogen metabolite phenotypes. Mol. Nutr.
Food Res. 2017,61. [CrossRef] [PubMed]
154.
D’Archivio, M.; Filesi, C.; Di Benedetto, R.; Gargiulo, R.; Giovannini, C.; Masella, R. Polyphenols, dietary
sources and bioavailability. Ann.-Ist. Super Sanita. 2007,43, 348–361.
155.
Basu, P.; Maier, C. Phytoestrogens and breast cancer:
In vitro
anticancer activities of isoflavones, lignans,
coumestans, stilbenes and their analogs and
derivatives. Biomed. Pharmacother. 2018
,107, 1648–1666.
[CrossRef]
156.
Mbese, Z.; Khwaza, V.; Aderibigbe, B.A. Curcumin and Its Derivatives as Potential Therapeutic Agents in
Prostate, Colon and Breast Cancers. Molecules 2019,24, 4386. [CrossRef] [PubMed]
157.
Mena, P.; Del Rio, D. Gold Standards for Realistic (Poly) phenol Research. J. Agric. Food Chem.
2018
,66,
8221–8223. [CrossRef] [PubMed]
158.
Singh, V.P.; Pratap, K.; Sinha, J.; Desiraju, K.; Bahal, D.; Kukreti, R. Critical evaluation of challenges and
future use of animals in experimentation for biomedical research. Int. J. Immunopathol. Pharmacol.
2016
,29,
551–561. [CrossRef] [PubMed]
159.
Del Rio, A.; Da Costa, F. Molecular Approaches to Explore Natural and Food-Compound Modulators in
Cancer Epigenetics and Metabolism. In Foodinformatics; Martinez-Mayorga, K., Medina-Franco, J., Eds.;
Springer: Cham, Switzerland, 2014; pp. 131–149.
160.
Bayram, B.; Gonz
á
lez-Sarr
í
as, A.; Istas, G.; Garcia-Aloy, M.; Morand, C.; Tuohy, K.; Garc
í
a-Villalba, R.;
Mena, P. Breakthroughs in the Health Eects of Plant Food Bioactives: A Perspective on Microbiomics,
Nutri(epi)genomics, and Metabolomics. J. Agric. Food Chem. 2018,66, 10686–10692. [CrossRef]
161.
Nejman, D.; Livyatan, I.; Fuks, G.; Gavert, N.; Zwang, Y.; Geller, L.T.; Rotter-Maskowitz, A.; Weiser, R.;
Mallel, G.; Gigi, E.; et al. The human tumor microbiome is composed of tumor type-specific intracellular
bacteria. Science 2020,368, 973–980. [CrossRef]
©
2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... Furthermore, it was discovered to reduce the development of tumors in women with postmenopausal breast cancer. 15,16 Flaxseeds are rich in αlinolenic acid oil and a significant source of phytoestrogens lignan, secoisolariciresinol diglycoside (SDG). In the human or animal gut, SDG is metabolized into enterolactone (ENL) and enterodiol (END), which are mammalian lignans. ...
Article
Background: Breast cancer is a multifactorial malignancy with uncertain treatment outcomes that make its challenging treatment. Balanced level of genes expression, including BAX, BCL2L1 (bcl-xl) and BCL-2 are needed to regulate apoptosis pathway in cancers. The aim of this study was to examine the impact of flaxseed extract on the expression of BAX, BCL-2, and BCL2L1 (bcl-xl) gens in BT-474 and MCF-7 breast cancer cells. Method: This in vitro experimental study investigated the impact of flaxseed extract on apoptosis and the proliferation of BT-474 and MCF-7 breast cancer cells. MTT assay was conducted on flaxseed extract treated BT-474 and MCF-7 cells at concentrations 3.90628, 7.8125, 156.125, 312.5, 125, 2, 1000, and 2000 µg/ml for 24, 48 and 72h. IC50 (μg/ml) was calculated for flaxseed extract in both cells. The expression of BCL2L1 (bcl-xl), bcl-2 and Bax genes were evaluated at these concentrations using real time Polymerase Chain Reaction (PCR). The data from different groups were analyzed using the Student's t-test. Results: Flaxseed extract reduced the multiplication and growth of BT-474 and MCF-7 cells in a concentration-dependent manner over 24, 48, and 72 hours of incubation. In both flaxseed-extract exposed BT-474 and MCF-7 cell lines, the BAX gene expression increased (P < 0.05); however, the gene expression of BCL2L1 (bcl-xl) and BCL-2 genes decreased (P < 0.05). Conclusion: Incubating BT-474 and MCF-7 with flaxseed extract repressed their growth and induced apoptosis. These results may provide precious information for developing a plant-based agent to prevent or treat breast cancer.
... CUR can selectively affect the cancer cells with no to limited toxicity towards the normal cells [41]. However, its medicinal advantages are limited because of its low ability to be absorbed into the bloodstream when administered orally [21,42]. Despite the substantial number of studies conducted, the low solubility of CUR in water continues to be a significant obstacle in terms of its capacity to be absorbed by the body and its effectiveness in therapeutic settings [21]. ...
Article
Full-text available
Cancer remains a leading cause of mortality, with non-small cell lung cancer (NSCLC) being a primary contributor to cancer-related deaths. Traditional treatment strategies such as chemotherapy, radiation, and hormone therapy often present challenges, including severe side effects, drug resistance, and toxicity. Recent advancements in nanotechnology aim to enhance the effectiveness of cancer therapies by targeting drugs selectively and specifically to tumor cells. Among these innovations, exosomes, or small extracellular vesicles (EVs), have emerged as promising carriers for drug delivery due to their natural origin and ability to encapsulate both small molecules and biologics. This study explores the use of exosomes derived from camel milk in Hail, Saudi Arabia, as a vehicle for delivering curcumin (CUR), a polyphenol with known chemopreventive properties but limited bioavailability. Camel milk was processed to isolate exosomes through differential centrifugation, followed by characterization using dynamic light scattering, zeta potential measurements, and Western blot analysis to confirm exosomal markers. The encapsulation of CUR into camel milk-derived exosomes demonstrated a 20% loading efficiency as analyzed by UPLC. In vitro antiproliferative assays revealed that the exosomal formulation of CUR (ExoCUR) significantly enhanced cytotoxicity against drug -sensitive (A549) and taxol-resistant (A549TR) lung cancer cells compared to free CUR. Molecular docking studies and molecular dynamics simulations indicated that CUR has a strong binding affinity for the epidermal growth factor receptor (EGFR), comparable to the established drug gefitinib. Furthermore, CUR effectively downregulated EGFR and STAT3 expression in cancer cells, suggesting its potential to disrupt key signaling pathways involved in tumor progression. Our findings highlight the potential of camel milk-derived exosomes as an effective and biocompatible delivery system for CUR, offering a promising strategy to overcome the limitations of current cancer therapies and enhance the therapeutic efficacy of chemopreventive agents.
... This action in animal models has been found to be mediated by a variety of pathways, including an increase in caspases activation, endostatin, and p21, and a decrease in angiogenesis, matrix metalloproteinases, K67, and the Bcl-2/Bax ratio. 28 However, only a tiny percentage of the consumed intact (poly)phenolics enter the bloodstream due to their limited bioavailability. 29 Moreover, (poly)phenols are significantly metabolized by phase-II enzymes after absorption, mainly glucuronyl, sulfate, and catechol-methyl transferases. ...
Article
Full-text available
This in-depth review examines the recently emerging field of using exosomes from milk as anticancer medication nanocarriers. It gives a summary of the most current developments, difficulties, and opportunities in this cutting-edge therapeutic strategy. Exosomes are found in many different body fluids and are considered a promising option for precision medicine due to their biocompatibility and innate cell-targeting abilities. These extracellular vesicles are nanoscale. The review commences with a comprehensive synopsis of the composition and biogenesis of exosomes derived from milk, highlighting their distinct membrane properties and capacity to transport cargo. Interestingly, these naturally occurring nanocarriers hold a variety of bioactive substances that can be precisely delivered as anticancer medications. These substances include proteins, lipids, and nucleic acids. Because dietary(poly) phenols are rapidly metabolized, their ability to prevent cancer is limited. Extracellular vesicles called exosomes may shield polyphenols from metabolism. Our objective was to evaluate the anticancer effects of free curcumin and resveratrol in breast cancer cell lines compared to their encapsulation in extracellular matrix derived from milk. Breast tissue was disposed of kinetically using rats. Curcumin and resveratrol were assessed using UPLC-QTOF-MS and GV-MS, respectively. Dietary polyphenols have a limited capacity to prevent cancer due to their rapid metabolism. Extracellular vesicles called exosomes may shield polyphenols from metabolism. Our goal was to assess in breast tissue. UPLC-QTOF-MS and GV-MS were used to evaluate curcumin and resveratrol, respectively. Curcumin and Resveratrol anticancer activity and bioavailability were improved by milk extracellular urea, which served as Trojan horses to get around the ABC-mediated chemoresistance of cancer cells. Exosomes derived from milk are being studied for their potential as carriers of therapeutic and diagnostic agents, emphasizing the potential benefits of personalized and precision medicine approaches to cancer treatment. The review also covers the challenges that the clinical translation of milk-derived exosome-based drug delivery systems currently faces, including scalability, standardization and safety profiles. The article's conclusion presents an optimistic view of how milk-derived exosomes will develop in the future in terms of anticancer medication delivery. The review highlights the revolutionary potential of using milk-derived exosomes, nature's nanocarriers, to advance the field toward more precise and effective cancer treatments, anticipating future advancements and emerging trends.
... NO 3 Ag) to increase its solubility, membrane permeability, and avoid their microbial and (or) phase II metabolism); can act as chemotherapeutic molecules to prevent the breast cancer carcinomas (breast cancer phase 0) and as coadjutant of the traditional breast cancer pharmacological therapies. Their anti-cancer proprieties are generally attributed to their anti-oxidant activity, acting as and exogenous antioxidant preventing the apparition of aberrant mutations due the oxidation damages of DNA and RNA and stimulating the immune system [11]. ...
Preprint
Full-text available
The aim of this study was to investigate the phenolic compound from Pinaropappus roseus , and its human breast anticancer properties. The phenolic molecules were isolated from the aqua-ethanoic extract isolated from the leaves of P. roseus by solid-phase extraction (SPE). The total phenolic content was determined by the Folin-Ciocalteu technique. The profile of phenolic compounds was analyzed by mass spectrometry (LC-ESI-MS ² ). The quantification of phenolic molecules identified by mass spectrometry was carried out by UV-spectrophotometry (LC-PDA). The in vitro cytotoxicity assay was carried on MCF-7 and HMEC cell lines using the MTT assay method. The docking simulation was carried out in anti-apoptotic proteins, Bcl-2 and Bcl-xL. Six phenolic compounds were identified of which the apigenin (37.5 mg CAE g db ⁻¹ ) was the most abundant compound. In the in vitro anti-cancer assay, the IC50 for the MCF-7 cells was of 426.15 µg g db ⁻¹ at 24h and 297.40 µg g db ⁻¹ at 48 h for the maximum evaluated phenolic extract concentration. The rutin and the chlorogenic acid showed the higher binding energies in the docking simulation for the active sites of the Bcl-2 and Bcl-xL proteins respectively. The phenolic compounds of P. roseus have cytotoxic activity against human breast cancer (MCF-7 cell line) and a low cytotoxic activity against normal human epithelial cells (HMEC cell line).These results suggest that the phenolic extract of P. roseus may have therapeutic potential against human cancer pathologies.
Article
Breast cancer is the most common malignancy among women globally, with incidence rates continuing to rise. A comprehensive understanding of its risk factors and the underlying biological mechanisms that drive tumor initiation is essential for developing effective prevention strategies. This review examines key non-modifiable risk factors, such as genetic predisposition, demographic characteristics, family history, mammographic density, and reproductive milestones, as well as modifiable risk factors like exogenous hormone exposure, obesity, diet, and physical inactivity. Importantly, reproductive history plays a dual role, providing long-term protection while temporarily increasing breast cancer risk shortly after pregnancy. Current chemoprevention strategies primarily depend on selective estrogen receptor modulators (SERMs), including tamoxifen and raloxifene, which have demonstrated efficacy in reducing the incidence of estrogen receptor-positive breast cancer but remain underutilized due to adverse effects. Emerging approaches such as aromatase inhibitors, RANKL inhibitors, progesterone antagonists, PI3K inhibitors, and immunoprevention strategies show promise for expanding preventive options. Understanding the interactions between risk factors, hormonal influences, and tumorigenesis is critical for optimizing breast cancer prevention and advancing safer, more targeted chemopreventive interventions
Article
Full-text available
Personalized nutrition” aims to establish nutritional strategies to improve health outcomes for non‐responders. However, it is utopian since most people share similar nutritional requirements. “Precision health,” encompassing lifestyles, may be more fitting. Dietary (poly)phenols are “healthy” but non‐nutritional molecules (thus, we can live without them). The gut microbiota influences (poly)phenol effects, producing metabolites with different activity than their precursors. Furthermore, producing distinctive metabolites, like urolithins, lunularin, and equol, leads to the term “polyphenol‐related gut microbiota metabotypes,” grouping individuals based on a genuine microbial metabolism of ellagic acid, resveratrol, and isoflavones, respectively. Additionally, (poly)phenols exert prebiotic‐like effects through their antimicrobial activities, typically reducing microbial diversity and modulating microbiota functionality by impacting its composition and transcriptomics. Since the gut microbiota perceives (poly)phenols as a threat, (poly)phenol effects are mostly a consequence of microbiota adaptation through differential (poly)phenol metabolism (e.g., distinctive reductions, dehydroxylations, etc.). This viewpoint is less prosaic than considering (poly)phenols as essential nutritional players in human health, yet underscores their health significance in a coevolutionary partnership with the gut microbiota. In the perspective on the gut microbiota and (poly)phenols interplay, microbiota metabotypes could arbiter health effects. An innovative aspect is also emphasized: modulating the interacting microbial networks without altering the composition.
Article
Full-text available
Several studies report that breast cancer survivors (BCS) tend to have a poor diet, as fruit, vegetable, and legume consumption is often reduced, resulting in a decreased intake of nutraceuticals. Moreover, weight gain has been commonly described among BCS during treatment, increasing recurrence rate and mortality. Improving lifestyle and nutrition after the diagnosis of BC may have important benefits on patients' general health and on specific clinical outcomes. The Mediterranean diet (MD), known for its multiple beneficial effects on health, can be considered a nutritional pool comprising several nutraceuticals: bioactive compounds and foods with anti-inflammatory and antioxidant effects. Recent scientific advances have led to the identification of nutraceuticals that could amplify the benefits of the MD and favorably influence gene expression in these patients. Nutraceuticals could have beneficial effects in the postdiagnostic phase of BC, including helping to mitigate the adverse effects of chemotherapy and radiotherapy. Moreover, the MD could be a valid and easy-to-follow option for managing excess weight. The aim of this narrative review is to evaluate the recent scientific literature on the possible beneficial effects of consuming functional and nutraceutical foods in the framework of MD in BCS.
Article
Full-text available
Profiling tumor bacteria Bacteria are well-known residents in human tumors, but whether their presence is advantageous to the tumors or to the bacteria themselves has been unclear. As an initial step toward addressing this question, Nejman et al. produced an exhaustive catalog of the bacteria present in more than 1500 human tumors representing seven different tumor types (see the Perspective by Atreya and Turnbaugh). They found that the bacteria within tumors were localized within both cancer cells and immune cells and that the bacterial composition varied according to tumor type. Certain biologically plausible associations were identified. For example, breast cancer subtypes characterized by increased oxidative stress were enriched in bacteria that produce mycothiol, which can detoxify reactive oxygen species. Science , this issue p. 973 ; see also p. 938
Article
Full-text available
Several studies in the past decades have reported anti-tumor activity of the bioactive compounds extracted from tea leaves, with a focus on the compound epigallocatechin-3-gallate (EGCG). However, further investigations are required to unravel the underlying mechanisms behind the anti-tumor activity of EGCG. In this study, we demonstrate that EGCG significantly inhibits the growth of 4T1 breast cancer cells in vitro and in vivo. EGCG ameliorated immunosuppression by significantly decreasing the accumulation of myeloid-derived suppressor cells (MDSCs) and increasing the proportions of CD4+ and CD8+ T cells in spleen and tumor sites in 4T1 breast tumor-bearing mice. Surprisingly, a low dose of EGCG (0.5–5 μg/mL) effectively reduced the cell viability and increased the apoptosis rate of MDSCs in vitro. EGCG down-regulated the canonical pathways in MDSCs, mainly through the Arg-1/iNOS/Nox2/NF-κB/STAT3 signaling pathway. Moreover, transcriptomic analysis suggested that EGCG also affected the non-canonical pathways in MDSCs, such as ECM–receptor interaction and focal adhesion. qRT-PCR further validated that EGCG restored nine key genes in MDSCs, including Cxcl3, Vcan, Col4a1, Col8a1, Oasl2, Mmp12, Met, Itsnl and Acot1. Our results provide new insight into the mechanism of EGCG-associated key pathways/genes in MDSCs in the murine breast tumor model.
Article
Full-text available
PurposeTo determine whether a red clover preparation plus dietary intervention administered to premenopausal women with breast cancer (BC), improves menopausal symptoms due to anti-oestrogen treatment, and hence promotes compliance with tamoxifen, prevents weight gain and is safe.Methods Surgically-treated premenopausal women with oestrogen receptor (ER) positive disease taking tamoxifen were recruited to a prospective double-blind randomized trial (NCT03844685). The red clover group (N = 42) received one oral tablet/day (Promensil® Forte) containing 80 mg red clover extract for 24 months. The placebo group (N = 39) received one oral tablet/day without active ingredient. All women were encouraged to follow a Mediterranean-type diet and keep active. Outcomes were Menopausal Rating Score (MRS), body mass index (BMI), waist and hip girth, insulin resistance, and levels of cholesterol, triglycerides, and sex hormones. As safety indicators, endometrial thickness, breast density, and effects of patient serum on ER-positive BC cell lines were investigated.ResultsMRS reduced significantly (p < 0.0001) with no between-group difference (p = 0.69). The red clover group had significantly greater reductions in BMI and waist circumference (p < 0.0001 both cases). HDL cholesterol increased significantly in both groups (p = 0.01). Hormone levels and insulin resistance changed little. Endometrial thickness remained constant (p = 0.93). Breast density decreased significantly in both groups (p < 0.0001). Proliferation and oestrogen-regulated gene expression didn’t differ in cell lines treated with serum from each group.Conclusions This is the first trial to assess red clover in BC patients on tamoxifen. The preparation proved safe clinically and in vitro, and was associated with reduced BMI and waist circumference, but the diet-lifestyle intervention probably improved the menopausal symptoms.
Article
Full-text available
Background: Several dietary factors have been reported to be associated with risk of breast cancer, but to date, unequivocal evidence only exists for alcohol consumption. We sought to systematically assess the association between intake of 92 foods and nutrients and breast cancer risk using a nutrient-wide association study. Methods: Using data from 272,098 women participating in the European Prospective Investigation into Cancer and Nutrition (EPIC) study, we assessed dietary intake of 92 foods and nutrients estimated by dietary questionnaires. Cox regression was used to quantify the association between each food/nutrient and risk of breast cancer. A false discovery rate (FDR) of 0.05 was used to select the set of foods and nutrients to be replicated in the independent Netherlands Cohort Study (NLCS). Results: Six foods and nutrients were identified as associated with risk of breast cancer in the EPIC study (10,979 cases). Higher intake of alcohol overall was associated with a higher risk of breast cancer (hazard ratio (HR) for a 1 SD increment in intake = 1.05, 95% CI 1.03-1.07), as was beer/cider intake and wine intake (HRs per 1 SD increment = 1.05, 95% CI 1.03-1.06 and 1.04, 95% CI 1.02-1.06, respectively), whereas higher intakes of fibre, apple/pear, and carbohydrates were associated with a lower risk of breast cancer (HRs per 1 SD increment = 0.96, 95% CI 0.94-0.98; 0.96, 95% CI 0.94-0.99; and 0.96, 95% CI 0.95-0.98, respectively). When evaluated in the NLCS (2368 cases), estimates for each of these foods and nutrients were similar in magnitude and direction, with the exception of beer/cider intake, which was not associated with risk in the NLCS. Conclusions: Our findings confirm a positive association of alcohol consumption and suggest an inverse association of dietary fibre and possibly fruit intake with breast cancer risk.
Article
Full-text available
Although soy consumption is associated with breast cancer prevention, the low bioavailability and the extensive metabolism of soy-active components limit their clinical application. Here, the impact of daidzein (D) and its metabolites on estrogen-dependent anti-apoptotic pathway has been evaluated in breast cancer cells. In estrogen receptor α-positive breast cancer cells treated with D and its metabolites, single or in mixture, ERα activation and Neuroglobin (NGB) levels, an anti-apoptotic estrogen/ERα-inducible protein, were evaluated. Moreover, the apoptotic cascade activation, as well as the cell number after stimulation was assessed in the absence/presence of paclitaxel to determine the compound effects on cell susceptibility to a chemotherapeutic agent. Among the metabolites, only D-4′-sulfate maintains the anti-estrogenic effect of D, reducing the NGB levels and rendering breast cancer cells more prone to the paclitaxel treatment, whereas other metabolites showed estrogen mimetic effects, or even estrogen independent effects. Intriguingly, the co-stimulation of D and gut metabolites strongly reduced D effects. The results highlight the important and complex influence of metabolic transformation on isoflavones physiological effects and demonstrate the need to take biotransformation into account when assessing the potential health benefits of consumption of soy isoflavones in cancer.
Article
Full-text available
Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States and compiles the most recent data on population‐based cancer occurrence. Incidence data (through 2016) were collected by the Surveillance, Epidemiology, and End Results Program; the National Program of Cancer Registries; and the North American Association of Central Cancer Registries. Mortality data (through 2017) were collected by the National Center for Health Statistics. In 2020, 1,806,590 new cancer cases and 606,520 cancer deaths are projected to occur in the United States. The cancer death rate rose until 1991, then fell continuously through 2017, resulting in an overall decline of 29% that translates into an estimated 2.9 million fewer cancer deaths than would have occurred if peak rates had persisted. This progress is driven by long‐term declines in death rates for the 4 leading cancers (lung, colorectal, breast, prostate); however, over the past decade (2008‐2017), reductions slowed for female breast and colorectal cancers, and halted for prostate cancer. In contrast, declines accelerated for lung cancer, from 3% annually during 2008 through 2013 to 5% during 2013 through 2017 in men and from 2% to almost 4% in women, spurring the largest ever single‐year drop in overall cancer mortality of 2.2% from 2016 to 2017. Yet lung cancer still caused more deaths in 2017 than breast, prostate, colorectal, and brain cancers combined. Recent mortality declines were also dramatic for melanoma of the skin in the wake of US Food and Drug Administration approval of new therapies for metastatic disease, escalating to 7% annually during 2013 through 2017 from 1% during 2006 through 2010 in men and women aged 50 to 64 years and from 2% to 3% in those aged 20 to 49 years; annual declines of 5% to 6% in individuals aged 65 years and older are particularly striking because rates in this age group were increasing prior to 2013. It is also notable that long‐term rapid increases in liver cancer mortality have attenuated in women and stabilized in men. In summary, slowing momentum for some cancers amenable to early detection is juxtaposed with notable gains for other common cancers.
Article
Context: Recent studies have outlined the potential role of dietary factors in patients who have survived cancer. Objective: The aim of this study was to summarize the evidence of the relation between dietary intake of phytoestrogens and their blood biomarkers and, overall, cancer-specific mortality and recurrence in patients with cancer. Data sources: A systematic search of PubMed, EMBASE, and Web of Science databases of studies published up to September 2019 was performed. Databases were searched for prospective and retrospective cohort studies reporting on dietary phytoestrogen intake and/or blood biomarkers and the outcomes investigated. Data extraction: Data were extracted from each identified study using a standardized form. Data analysis: Twenty-eight articles on breast, lung, prostate, and colorectal cancer, and glioma were included for systematic review. Given the availability of studies, a quantitative meta-analysis was performed solely for breast cancer outcomes. A significant inverse association among higher dietary isoflavone intake, higher serum/plasma enterolactone concentrations, and overall mortality and cancer recurrence was found. Among other cancer types, 2 studies reported that higher serum enterolactone and higher intake of lignans were associated with cancer-specific survival for colorectal cancer and glioma, respectively. Conclusions: Dietary phytoestrogens may play a role in survival from breast cancer ; evidence regarding other cancers is too limited to draw any conclusions.
Article
Endocrine therapy is currently the main therapeutic approach for estrogen receptor-positive (ER⁺) breast cancer, the most frequent subtype of breast cancer in women worldwide. For this subtype of tumors, the current clinical treatment includes aromatase inhibitors (AIs) and anti-estrogenic compounds, such as Tamoxifen and Fulvestrant, being AIs the first-line treatment option for post-menopausal women. Moreover, the recent guidelines also suggest the use of these compounds by pre-menopausal women after suppressing ovaries function. However, besides its therapeutic efficacy, the prolonged use of these type of therapies may lead to the development of several adverse effects, as well as, endocrine resistance, limiting the effectiveness of such treatments. In order to surpass this issues and clinical concerns, during the last years, several studies have been suggesting alternative therapeutic approaches, considering the function of aromatase, ERα and ERβ. Here, we review the structural and functional features of these three targets and their importance in ER⁺ breast cancer treatment, as well as, the current treatment strategies used in clinic, emphasizing the importance of the development of multi-target compounds able to simultaneously modulate these key targets, as a novel and promising therapeutic strategy for this type of cancer.
Article
The full consensus on the role of dietary polyphenols as human health‐promoting compounds remains elusive. The two‐way interaction between polyphenols and gut microbiota (GM) (i.e., modulation of GM by polyphenols and their catabolism by the GM) is determinant in polyphenols’ effects. The identification of human metabotypes associated with a differential gut microbial metabolism of polyphenols has opened new research scenarios to explain the inter‐individual variability upon polyphenols consumption. The metabotypes unequivocally identified so far are those involved in the metabolism of isoflavones (equol and(or) O‐desmethylangolesin producers vs. non‐producers), and ellagic acid (urolithin metabotypes, including producers of only urolithin‐A (UM‐A), producers of urolithin‐A, isourolithin‐A, and urolithin‐B (UM‐B), and non‐producers (UM‐0)). Besides, the microbial metabolites (phenolic‐derived postbiotics) such as equol, urolithins, valerolactones, enterolactone and enterodiol, and 8‐prenylnaringenin, among others, can exert differential health effects. We update the knowledge and take position here on i) the two‐way interaction between GM and polyphenols, ii) the evidence between phenolic‐derived postbiotics and health, iii) the role of metabotypes as biomarkers of GM and the clustering of individuals depending on their metabotypes (metabotyping) to explain polyphenols’ effects, and iv) the gut microbial metabolism of catecholamines to illustrate the intersection between personalized nutrition and precision medicine. This article is protected by copyright. All rights reserved