You are on page 1of 11

Available online at www.sciencedirect.

com

ScienceDirect
Journal of Nutritional Biochemistry 85 (2020) 108428

REVIEWS: CURRENT TOPICS

The immunoregulatory function of polyphenols: implications in cancer immunity

José Tarcísio Giffoni de Carvalho, Debora Da Silva Baldivia, David Tsuyoshi Hiramatsu de Castro,
Helder Freitas dos Santos, Cintia Miranda dos Santos, Alex Santos Oliveira, Tamaeh Monteiro Alfredo,
Kellen Natalice Vilharva, Kely de Picoli Souza, Edson Lucas dos Santos⁎
Research Group on Biotechnology and Bioprospecting Applied to Metabolism, Federal University of Grande Dourados, Dourados, Brazil

Received 20 November 2019; received in revised form 14 May 2020; accepted 18 May 2020

Abstract

Polyphenols have demonstrated several potential biological activities, notably antitumoral activity dependent on immune function. In the present review, we
describe studies that investigated antitumor immune responses influenced by polyphenols and the mechanisms by which polyphenols improve the immune
response. We also discuss the limitations in related areas, especially unexplored areas of research, and next steps required to develop a therapeutic approach
utilizing polyphenols in oncology.
© 2020 Published by Elsevier Inc.

Keywords: Polyphenols; Antitumor immune response; Cancer therapy; Natural compounds; Immuneregulatory properties

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Immune response against tumor cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1. Role of NK cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.2. T cell response against tumor cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.3. Innate cell signaling to activate T cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.4. Therapeutic or prophylactic vaccination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.4.1. Immunogenic cell death . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.4.2. Tumor-based vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.5. Immune checkpoints and Treg cells. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.6. Tumor immune escape . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3. Immune regulation by polyphenols in the cancer microenvironment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.1. Polyphenols promote NK cell activation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.2. Polyphenols as therapeutic or prophylactic vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.3. Polyphenol activation of Th1/CD8+ T cell-mediated immunity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.4. Polyphenols suppress Treg activation and function. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.5. PD/PD-L1 checkpoint modulation by polyphenols . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.6. Polyphenols suppress TAMs. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4. Bioavailability of polyphenols . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Conflicts of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9

⁎ Corresponding author.
E-mail address: edsonsantosphd@gmail.com (E.L. dos Santos).

https://doi.org/10.1016/j.jnutbio.2020.108428
0955-2863/© 2020 Published by Elsevier Inc.
2 J.T.G. de Carvalho et al. / Journal of Nutritional Biochemistry 85 (2020) 108428

1. Introduction NK cells express receptors from three major families: (a) killer cell
immunoglobulin (Ig)-like receptors (KIRs), (b) natural cytotoxicity
Polyphenols are compounds synthesized by plants for protection receptors (NCRs) [23] and (c) Lectin-like Receptors. NK cell activation
against radiation, mechanical damage and microbial infection [1]. depends on the balance between positive and negative signals from
Polyphenols have structural variability and are commonly classified as members of these families. For example, NKG2D (a positive-signaling
flavonoids, phenolic acids, tannins and stilbenes. The main differences member of the Lectin-like Receptor family) is activated by sequences A
among polyphenols are in the number of phenol rings together with and B (MICA and MICB), which are not present on the cell surface in
one or more hydroxyl radicals [2,3]. most healthy tissues but can be expressed on transformed cells and
Several potential biological activities have been described for tumor cells [24,25].
polyphenols, such as antitumor activity [4,5], and these molecules
have great potential for immune regulation [6,7]. In recent years, there 2.2. T cell response against tumor cells
has been an increasing emphasis on the importance of the immune
response against cancer and the search for therapies that can interact CD8+ cytotoxic T cells are primarily responsible for the recognition
with the immune system to eliminate tumor cells [8–10]. In this context, of cancer through molecules expressed on the surface of tumor cells,
phenolic compounds may emerge as adjuvant therapies, as indicated by including MHC class I [26], and initiate apoptotic processes mediated
the studies that are already available, and new therapeutic alternatives. by members of the tumor necrosis factor (TNF) cytokine family [27,28]
Realizing these potentials still depends on the acquisition of more data, and the release of granules.
and many questions remain to be answered. Perforin forms a pore in the membrane of a target cell that allows
In this review, we describe and discuss recent studies that show molecules to enter malignant cells. Granzymes are serine proteases
that polyphenols have antitumor activity dependent on adaptive that cleave the proteins inside a cell, inducing apoptosis by caspase-
immunity and focus on the mechanism through which they act, as well independent (Granzyme B) [29,30] or caspase-dependent pathways
as the next steps for the progression of this field to develop future (Granzyme A) [27,31]. The binding of a ligand in the TNF family to the
therapeutic approaches. corresponding death receptor is an important trigger for apoptosis
[32–35]. Active T cells can secrete TNF or tumor necrosis factor-related
2. Immune response against tumor cells apoptosis-inducing ligand (TRAIL) or overexpress CD95 ligand
(CD95L) on their cell surface, and these molecules bind to “death
CD8+ T cells are the major weapons used by the immune system to receptors” on target cells to initiate apoptosis by interacting with the
eliminate tumor cells, but other cell types also perform important roles corresponding receptors, TNFR, Fas and the death receptors DR3, DR4
in the antitumor immune response, including natural killer (NK) cells and DR5, respectively [27,28].
and macrophages [11–14] (See Fig. 1). CD4+ T cells play an important role in the tumor cell elimination
An adequate CD8+ T cell response against tumor cells depends on T mediated by CD8+ cytotoxic T cells by supporting the activation and
cell activation and involves some key points: first, phagocytosis by expansion of CD8+ T cells and the generation and maintenance of
antigen-presenting cells (APCs) and the presentation of tumor antigens CD8+ T cell memory. Following activation, CD4+ T cells are further
via major histocompatibility complex class I (MHC-I); second, CD8+ T differentiated into T helper 1 (Th1) cells that act by producing
cell differentiation into cytotoxic T lymphocytes (CTLs); and third, cytokines, such as interferon-gamma (IFN-γ) and interleukin (IL)-2, to
recognition of MHC-I on target cells by CTLs, resulting in the elimination promote the CD8+ T cell response [26,36]. In addition, CD4+ T cell help
of tumor cells through the caspase cascade [15,16]. is also dependent on CD40-CD40 L cell–cell interactions. CD40
There has been great progress in understanding the regulation of signaling triggers the up-regulation of the expression of costimulatory
these stages, particularly in regard to the concepts of immunogenic death molecules, such as CD86 and OX40 ligand, in APCs, which provides a
and immune checkpoints, which has allowed the elucidation of new signal via the corresponding receptors on CD8+ T cells, resulting in
antitumor mechanisms that can be targeted by new therapeutic agents. strong activation and effector function [37]. Finally, Th1 cytokines play
important roles in macrophage activation and tumor elimination [38].
2.1. Role of NK cells Accordingly, both the number and function of tumor-specific CTLs
are significantly enhanced in the presence of tumor-specific CD4+ T
NK cells are one of the main effectors against cancer and function cell responses, whereas depletion of CD4+ T cells facilitates tumor
via direct cytotoxicity and/or cytokine and granule secretion to progression and reduces the survival of tumor-bearing hosts,
eliminate cancer cells, like CD8+ T cells (see below) [17–19]. NK indicating the importance of tumor-specific CD4+ T cell help in
cells are characterized phenotypically by absence in expression of the maintaining tumor-reactive CTL functions in vivo [39]. In addition, the
cluster of differentiation (CD)3 of T cells and by expression of CD56 in efficacy of antitumor responses induced by the combined adminis-
humans (CD3 −CD56 +), and expression of NK1.1 + in mice or tration of human CD4+ and CD8+ T cells is notably better than that
expression of NKp46+ in both species [20]. Human NK cells can be induced by applying CD4+ or CD8+ T cells alone [40,41].
further divided into two distinct subsets on the basis of their level of CD4+ T helper cells are the central cells in antitumor immunity that
CD56 expression and the presence or absence of CD16; human NK cells further differentiate into Th1 cells, which act by producing cytokines,
can be identified as positive to CD16 marker and by lower expression mainly IFN-γ and IL-2, to promote CD8+ T cell immunity and
of CD56 cells (CD16+CD56low), which are the most numerous NK cells macrophage activation. When properly activated by IFN-γ, macro-
in the blood, or as negative for CD16 with higher expression of CD56 phages are able to directly eliminate cancer cells [41]. They can also
(CD16−CD56high cells), which are characterized as a less mature support the adaptive immune response through the presentation of
population [21]. tumor antigens and the production of chemokines and cytokines to
NK cells detect and eliminate tumor cells that are deficient MHC-I recruit and activate cytotoxic CD8+ T cells and NK cells [42].
molecules (missing self), exhibit up-regulated expression of stress
ligands recognized by activating NK cell receptors (induced self) or are 2.3. Innate cell signaling to activate T cells
coated with antibodies specific for tumor antigens, which enable
recognition by NK cells expressing the fragment crystallizable (Fc) To engage adaptive immunity in a manner that results in diversity,
receptor and trigger antibody-dependent cell-mediated cytotoxicity specificity and memory, antigens must be processed and presented to
[17,18,22]. T cells [43]. Presentation is mediated by MHC class I and II molecules
J.T.G. de Carvalho et al. / Journal of Nutritional Biochemistry 85 (2020) 108428 3

[44]. MHC class I and II molecules are similar in function: they display patterns (DAMPs), such as heat shock proteins (HSP) 70 and 90,
short peptides to the cell surface, allowing these peptides to be calreticulin (CRT), high-mobility group box 1 (HMGB1) and adenosine
recognized by CD8+ and CD4+ T cells, respectively, and subsequently triphosphate (ATP) [52,53]. These DAMPs are recognized by toll-like
initiate T cell activation. Both MHC I and II molecules participate in T receptors expressed by APCs, resulting in MHC presentation together
cell activation, but MHC-I also participates in the recognition of target with B7 membrane expression for costimulation [54].
cells (tumor) by CD8+ T cells during cytotoxic elimination [45]. Importantly, interest in the induction of ICD has been increasing as
Dendritic cells (DCs) are the most effective APCs in the innate a potential emergent therapy since ICD, like vaccines, has the ability to
immune system [46,47]. These cells act as sentinels with the greatest induce immunological memory and, in subsequent carcinogenic
ability to capture and deliver antigens with adequate signals to induce processes, to raise a fast and strong immune response [55,56].
T cell activation and the T cell response [47].
During activation and differentiation into effector T cells (T eff), 2.4.2. Tumor-based vaccines
naïve lymphocytes receive two signals when they interact with antigen- In the context of emerging therapies for cancer, tumor-based
presenting cells [48,49]. The first signal is the antigen signal provided vaccines have shown great potential. Basically, a tumor-based vaccine
through the MHC molecule to the T cell receptor. For the second signal, a relies on delivering tumor antigens directly to APCs, after which, when
costimulatory signal is delivered when members of the B7 family (CD80 the APCs home to secondary lymphoid organs, DC activity leads to an
or CD86) interact with the receptor CD28 on T cells. enhanced tumor-specific T cell-mediated immune response and
enhanced antitumor effects [57,58]. To achieve this purpose, different
2.4. Therapeutic or prophylactic vaccination types of cancer vaccines have been developed, i.e., cell-based vaccines
that involve autologous DC activation ex vivo and injection into
In the context of emerging therapies for cancer, two different patients [59], protein/peptide derivates for intratumoral administra-
vaccination approaches, specifically therapeutic and prophylactic tion [60] and gene-based vaccines, including RNA and DNA vaccines
vaccines, have shown great potential. Preventive vaccines induce that transfect normal tissue to express tumor antigens [58].
pathogen-specific T cells to establish immune memory, preventing
tumor recurrence, while therapeutic vaccines raise a specific immune 2.5. Immune checkpoints and Treg cells
response against an established tumor. Despite working by different
mechanisms, immunogenic cell death and tumor-based vaccines both The immune system uses many checkpoints to ensure that CD8+ T
target DCs for modulation, resulting in T cell activation and memory. cells react specifically to tumor cells without causing injury to healthy
cells [61,62]. Two well-described checkpoint molecules, cytotoxic T
2.4.1. Immunogenic cell death lymphocyte antigen 4 (CTLA-4) and programmed cell death-1 (PD-1),
Recent advances describe that cell death, under certain circum- act as negative regulators of T cell function and have been associated
stances, can subsequently result in T cell activation and the with immune evasion in cancer [63]. CTLA-4 is a CD28 homolog that
development of adaptative immunity, which is defined as immuno- has much higher binding affinity for B7 than does CD28 [64]; however,
genic cell death (ICD) [50,51]. The signals that mediate the unlike that of CD28, the binding of CTLA-4 to B7 does not produce a
immunogenicity of tumor cells involve endogenous molecules stimulatory signal. As such, this competitive binding can prevent the
released from dying cells, which are danger-associated molecular transduction of the costimulatory signal normally provided by CD28:B7

Imunogenic
Cell
Death
T CELLS ACTIVATION
DAMPs
1° Signal
A
B T CD8+ MHC TCR
T CELLS
Tumor Eliminaon APC
APC 2° Signal
CD80/86 CD28

M1 cells NK T CD4+
D

T CD8 x TARGET CELLS


C
CELL DEATH SIGNAL
CD28
E MHC-
I TCR
TUMOR

T CD8
EFFECTOR SIGNAL
CELLS
IFN-γ FAS FAS-L

Granules

T CD8/CD4+ cells
Fig. 1. Cancer immunity: (A) NK cells represent the first line of defense against a tumor cell. (B) Immunogenic cell death results in DAMP release, which increases the expression of
costimulatory molecules (CD80/86) by APCs. (C) Tumor peptide presentation to T cells leads to subsequent activation. (D) CD8+ T cell-mediated elimination of tumor cells occurs via
MHC-I recognition. (E) Cytokines from T helper cells enhance macrophage activity.
4 J.T.G. de Carvalho et al. / Journal of Nutritional Biochemistry 85 (2020) 108428

binding [48]. Furthermore, the engagement of PD-1 with its coreceptor, such as anti-PD1 (pembrolizumab and nivolumab), anti-PD-L1
programmed death-ligand 1 (PD-L1), which is expressed by cells in the (MPDL3280A) and anti-CTLA4 (ipilimumab) antibodies, to overcome
absence of stress, results in the down-regulation of T cell activity, the ability of cancer cells to resist immune responses and to stimulate
including antitumor activities, thereby restricting cell killing [65]. T cell activation. These inhibitors have shown remarkable success in
To prevent overactive inflammatory responses, the immune enhancing the effector antitumor response in different malignancies,
system generates regulatory T (Treg) cells [66,67]. The differentiation especially in melanoma [65] and lung cancer [73], but the cost of
of this subset is driven by the transcription factor Foxp3 [68], and these treatment per patient is very expensive.
cells exert suppressive effects via high expression of CTLA-4 [48] and For the second approach, cancer cell secretion of suppressive
secretion of key anti-inflammatory cytokines, notably transforming molecules such as IL-10, TGF-β and vascular endothelial growth factor
growth factor-β (TGFβ) and IL-10 [68]. (VEGF) skews the T cell population toward an inadequate profile of T
cell subsets, such as increased Treg or T helper (Th) 2 activation
2.6. Tumor immune escape instead Th1 activation, or induce alternatively activated macrophage
polarization [74,75].
Historically, the first mechanism described for evasion of the Substantial clinical and experimental evidence indicates that there is
immune system, especially CD8 + T cells, was a lack of MHC-I an abundance of macrophages in most tumor types [76,77]. Macro-
expression, and more recently, the immunoediting of tumor cells by phages may be activated in a way dependent on the microenvironment
NK cells has been shown to lead to tumor variants that can evade NK as “classically activated M1” macrophages via the proinflammatory
cell-mediated responses [25,69]. These mechanisms include the molecule IFN-γ or lipopolysaccharide or “alternatively activated M2”
secretion of immunosuppressive factors, shedding of ligands for macrophages induced by IL-4 or IL-13 [38,78]. While M1 macrophages
activating NK cell receptors and induction of ligands for activating exhibit antitumor activity, M2 macrophages can be used by tumors to
NK cell receptors on healthy cells that can serve as decoys [25,69]. promote tumor progression and escape and are commonly referred to
Finally, evidence has shown resistance to apoptosis mediated by as tumor-associated macrophages (TAMs) [79]. TAM-induced immune
overexpression of decoy receptors (DcRs; DcR1, DcR2 and DcR3), suppression is mediated by the expression of T cell immune checkpoint
which is useful for tumors since these molecules compete with death ligands, such as PDL1, and the secretion of several cytokines, such as IL-
receptors for death ligand binding, thus inhibiting apoptosis [25,69] 10 and TGFβ, and important growth factors essential in metastasis,
(See Fig. 2). especially VEGF [76].
In recent years, other mechanisms have been proposed, especially
mechanisms by which tumor cells evade the CD8+ T cell response,
suggesting two main strategies. The first strategy is decreased 3. Immune regulation by polyphenols in the cancer microenvironment
checkpoint activation or recognition by T cells [70]. For example,
triple-negative breast cancer, the most aggressive type of all breast Recent findings indicate that polyphenol antitumor effects are at
tumors, is characterized by a lack of expression of hormone receptors least partially dependent on the immune response, especially on T
and human epidermal growth factor receptor 2 (HER2), was reported cell modulation [80–82]. The mechanism by which polyphenols
by an increase in the expression of PD-L1 [71,72]. Over the past few improve the immune response has been investigated in a few
years, a great advance in cancer treatment has been the development publications. The immunoregulatory effects of polyphenols are
of monoclonal antibodies that serve as immune checkpoint inhibitors, summarized in Fig. 3 and Table 1. Although the topics presented

Tumor Progression, angiogenesis, Tumor escape


and resistance mechanism

VEGF
IL10, IL4 CELL DEATH SIGNAL
MHC-I TCR
T CD8
APC STOP SIGNAL
CELLS
B CD80/86
CD28
C CTLA4
A
↓MHC-I
NK E
↑PD-L1
APC

M2 cells M1 cells
CELL DEATH SIGNAL
TCR
TUMOR

T CD8
STOP CD8 CELLS
D
PD-L1 PD1

↑T reg cells

T CD8/CD4+ cells

Fig. 2. The red line indicates the mechanism of tumor escape. (A) Tumor-secreted factors induce alternative activation in macrophages, leading to the secretion of soluble factors
important for tumor growth and metastasis. (B) Tumor cells evade NK cell vigilance by shedding ligands for activating NK cell receptors and inducing the expression of ligands for
activating NK cell receptors on healthy cells. (C) Factors secreted by tumor cells increase CTLA4 expression. (D) This balance influences the activation of Tregs, which inhibit effector T
cells. (E) Targeted tumor cells escape CD8+ T cells by increasing PDL1 expression.
J.T.G. de Carvalho et al. / Journal of Nutritional Biochemistry 85 (2020) 108428 5

Table 1
Immunoregulatory and antitumor effects of polyphenols
Immune effect/mechanism Polyphenol/concentration Model Outcome Ref.

Activation of NK cells
Resveratrol (25 μM, 24 h) Promyeloblastic leukemia ↑ NK cell-mediated leukemia cell lysis [83]
↑ NKG2D
Resveratrol (37 μM, 48 h) Molt4, THP1, KG1 and Jurkat cells ↑ NK cell-mediated leukemia cell lysis [84]
↑ NKG2D
↑ ULBP1, ULBP2, ULBP3 and MICA/B
Resveratrol (1 g/day/4 weeks) Leukemic patients ↑ NKG2D+ cells [85]
↑ CD56+ NKG2D+
Polyphenol fraction of C. spinosa (75 mg/kg/s.c/3 Melanoma in vivo NK cells and CD69+ cells activated [86]
days)
Extract from persimmon leaves; 2 g/kg/o.g./10 Liver tumor in vivo ↑ Promoted NK cell cytotoxicity [87]
days) ↑ IL-18

Polyphenols acting as a
therapeutic or preventive
vaccine
Polyphenol fraction of C. spinosa (75 mg/kg/s.c/3 Melanoma cell and mouse models ↑ CRT, ATP and HMGB1 [88]
days) ↑ Central memory (CD62L+CD44+) CD8+ T cells
Polyphenol fraction of C. spinosa (75 mg/kg/s.c/3 Breast cell and melanoma mouse ↑ Calreticulin, HMGB-1, and ATP [89]
days) models ↑ Cytokines
Resveratrol 0 (25 and 50 μM, 48 h) Ovarian carcinoma mouse model ↑ Calreticulin, HMGB-1, and ATP [90]
Pretreatment produced a vaccine-like effect that
significantly inhibited the growth of a subsequent
inoculated xenograft tumor
EGCC (0.5 mg/mL/o.g./ 5 days) Murine skin tumor model Improved DNA vaccination with LAMP [101]
↑ T cell-mediated immune response and antitumor
effects
Curcumin in micelles (40 mg/kg/i.v./5 days) Melanoma in mice ↑ IL-6 and CCL-2 [102]
↓ TNF-α and IFN-γ levels

Polyphenols induction of
Th1/CD8+ T cells
Resveratrol (1, 2.5 and 5 mg/kg/i.i./6 days) Renal carcinoma in vivo ↑ Th1 cytokines with predominance of IFN-γ [100]
↓ Th 2 cytokines, such as IL-4 and IL-10
Activated CD69+ CD8+ T cells
↑ Perforin, granzyme B and FasL
Rutin (6 or 12 mg/kg/o.g./6 weeks) Murine myelomonocytic leukemia cell ↑ T cells (CD3+) and B cells (CD19+) [103]
line (WEHI-3) in BALB/c mice ↑ Macrophage phagocytosis
Caffeic acid 40 or 80 mg/kg Ehrlich ascites tumor ↑ Th1 cytokines: IL-2, IL-12 and IFN-γ [119]
↓ Th 2 cytokines: IL-4 and IL-10
Linalool (227 μM, 48 h) and p-coumaric acid (478 PBMCs+ A549, T-264 47D, SW-620 or ↑ IFN-γ, IL-12p40, IL-1β and TNF-α [104]
μM, 48 h) Hep G2 cells ↑ CD40/CD40L
Resveratrol low (10 and 25 μM, 24 h) high (60 μM, PBMCs+ HT29 or RKO cells Dose-dependent response [33]
24 h) ↑ IL-1β and IL6 at low dose
↓ IL-1β, IL6, IFN-γ and TNF-α at high dose

Polyphenols suppress Treg


function and activation
Curcumin, 3 g/day/2 weeks) Lung cancer patients (n=30) ↓ CD4+CD25+Foxp3+ Treg cells [105]
↑ IFN-γ-producing Th1 cells
Curcumin, 3 g/day/4 weeks Colon carcinoma patients (n=40) ↓ CD4+CD25+Foxp3+ Treg cells [106]
↑ IFN-γ-producing Th1 cells
EGCG-rich extract green tea (4602 mg green tea/ Chronic lymphocytic leukemia (CLL) ↓ Reductions in circulating lymphocytes and Treg [107]
189 mg of EGCG/6 days) patients (n=20) cells
↓ IL-10 and TGF-β
↑ Activity of CD8+ T cells
Polyphenol fraction of C. spinosa, 75 mg/kg Melanoma in BALB/c or C57BL/6 mice ↓ IL-10 levels and FoxP3 expression [86]

Polyphenols modulate
immune
checkpoints
Apigenin (30 μM, 4 h) and curcumin (25 μM, 4 h) Melanoma cells ↑ PD-L1 expression [110]
Resveratrol (50 μM, 24 h) and piceatannol (50 μM, Breast cells ↑ PD-L1 expression [112]
24 h)
Bisdemethoxycurcumin (3 mg/kg/i.v/3 weeks) Bladder cancer mouse model ↓ PD-1 expression [111]
↑ Activity of CD8+ T cells
↑ IFN-γ and granzyme B
Resveratrol (100 mg/kg/s.c) Ovarian carcinoma mouse model ↓ Tumor growth [90]
↑ Xenograft progression after depletion of CD8+ T
cells

Suppression of TAMs
EGCG (10 mg/kg /i.p./3 days) Balb/c tumors induced by the murine ↓ Chemokines: CSF-1 and CCL-2 [115]
breast cancer cell line 4 T1 ↓ Infiltration of macrophages
↑ miR-16

(continued on next page)


6 J.T.G. de Carvalho et al. / Journal of Nutritional Biochemistry 85 (2020) 108428

Table 1 (continued)

Immune effect/mechanism Polyphenol/concentration Model Outcome Ref.

Curcumin (1.28 mM) + epicatechin (320 μM) Mice with tumors induced by GL261 ↓ Tumor growth [116]
gallate + resveratrol (4 mM/ i.p./5 days) mouse glioblastoma cells ↓ M2: ARG1high iNOS low IL-12 low IL-10 high
↑ M1: ARG1 low iNOS high IL-12 high IL-10 low
Curcumin (1.28 mM) + epicatechin (320 μM) Mice with tumors induced by TC-1 ↓ Tumor growth [117]
gallate + resveratrol (4 mM/i.p./5 days) cells infected with HPV ↓ M2: ARG1 high iNOS low IL-12 low IL-10 high
↑ M1: ARG1 low iNOS high IL-12 high IL-10 low
Resveratrol (25 μM, 24 h) Lung cancer cells ↓ M2 polarization induced by lung cancer cells [113]
Resveratrol (5–50 μM, 24 and 48 h) Lymphatic vessels ↓ Lymphangiogenesis ↓ IL10 and VEGF in M2 [114]
macrophages.
Rutin (200 μM/kg/bw/5 day) B16F-10 melanoma cells in C57BL/6 ↓ VEGF and IL-1β [118]
mice ↑ TNF-α

i.p.: intraperitoneal; i.v. intravenously; s.c. subcutaneous injection; o.g. oral gavage.

here are all connected, for ease of understanding, our discussion is Furthermore, Lasso et al. [86] observed increased numbers of total
divided into six topics: NK and CD69+ activated NK cells after C. spinosa (P2Et), a plant native
to South America, treatment in B16F10 mouse melanoma cells but did
I. Polyphenols promote NK cell activation not investigate the associated mechanisms. Chen et al. [87] evaluated
II. Polyphenols as therapeutic or prophylactic vaccines the potential to induce NK activity with an extract rich in flavonoids
III. Immune checkpoint modulation by polyphenols from persimmon (traditional medicine in Asia) in liver tumor-bearing
IV. Polyphenol-mediated activation of Th1/CD8+ T cell immunity mice. This extract promoted NK cell cytotoxicity and enhanced IL-18
V. Polyphenols suppress Treg activation and function production, possibly by stimulating macrophages, thereby up-
VI. Polyphenols suppress TAMs regulating the NK cell-mediated antitumor immune response. These
extracts are rich in polyphenols, particularly flavonoids.
NK cells represent the first line of defense against a tumor process,
3.1. Polyphenols promote NK cell activation therefore, emerging therapies that modulate their activity may
represent a new therapy. In this context, polyphenols appear as a
Previous studies have shown that resveratrol sensitizes leukemia new strategy approach in cancer treatment, but further research is
cell lines to NK cell-mediated antitumor effects in cocultures of needed to more properly address this topic, such as to confirm effect of
CD3+CD56+ cells isolated from the peripheral blood mononuclear resveratrol, and evaluate other polyphenols such as quercetin, rutin
cells (PBMCs) of healthy volunteers. Hu et al. [83] found inhibition of and curcumin on the NK function and activity.
the growth of promyeloblastic leukemia KG-1a cells in the presence of
resveratrol. This effect was correlated with an increase in the cell- 3.2. Polyphenols as therapeutic or prophylactic vaccines
surface expression of NKG2D ligands and DR4, coupled with a down-
regulation of the cell-surface expression of DcR1 on the KG-1a cells. In One group evaluated the potential of P2Et to induce immunogenic
addition, these effects were ameliorated by blocking NKG2D ligands or cell death in two different publications. Gomez-Cadena et al. [88]
TRAIL. Espinoza et al. [84] showed that pretreatment of leukemia cells found that P2Et induced apoptosis in B16F10 mouse melanoma cells.
with resveratrol promoted an increase in the sensitivity of leukemic Confirming immunogenicity, the protective effects of P2Et treatment
cell lines (Molt4, THP1, KG1, and Jurkat) to killing by NK cells and were abolished in immunodeficient mice. Moreover, P2Et induced the
induced variable increases in the expression of NKG2D in NK cells and expression of DAMPs, such as ATP, CRT and HMGB1. Finally, P2Et-
that of NKG2D ligands, including MICA, ULBP1, ULBP2 and ULBP3. In treated cells were injected into C57BL/6 mice, showing that these
fact, leukemia cell lysis was abrogated by blocking NKG2D. Impor- cells worked as a cell vaccine to delay further tumor growth.
tantly, these studies showed that resveratrol could induce an Central memory (CD62L+CD44+) CD8 + T cell levels were also
enhancement in NK activities at low concentrations (25-37 μM) but found to be increased. Urueña et al. [89] found a similar effect
a suppressive effect at high concentrations (60 μM). when evaluating the effect of P2Et on breast tumor 4T1 cells.
An in vivo examination of the effects of administered daily Positive results were found for ICD markers, such as calreticulin
resveratrol (1000 mg) for 4 weeks to nine healthy subjects showed expression, HMGB1 and ATP secretion. Primary tumor control was
an increase in NKG2D+ T cell and CD3−CD56+ NKG2D+ NK cell improved by vaccination with P2Et-pretreated 4T1 cells, yielding
numbers, while CD8 + and CD4+ T cell and CD19 + B cell numbers did long-lasting ex vivo multifunctional CD4 + and CD8 + T lymphocytes
not change [85]. The limitations of the study design and the lack of that secreted IL-2, TNF-α, IL-4, IL-5 and IFN-γ after specific 4T1 cell
appropriate number of patients suggest further evaluation to stimulation.
confirm the positive effect of resveratrol in cancer patients by NK In another study, Zhang et al. [90] observed that resveratrol
actions. As mentioned before, in vitro studies showed that resveratrol induced apoptosis in ovarian carcinoma cells by stimulating cell-
could promote a dual effect on NK activities at different concentra- surface exposure of CRT, secretion of HMGB1 and release of ATP.
tion. Thus, a study with a range of dose administrated could help to Moreover, the pretreatment with resveratrol worked a vaccine since it
understand the in vivo effect of resveratrol in NK activities in cancer significantly inhibited the growth of subsequently inoculated xeno-
patients. graft tumors. The authors further characterized increases in both
Together, these findings show that resveratrol increases NK cell mature dendritic cell and cytotoxic T cell numbers in xenograft tumors
activities in vitro and in vivo, is safe in humans and acts through the in response to resveratrol treatment, which also inhibited TGF-β
modulation of the NKG2D receptor/NKG2D-L system and activation of production and stimulated both IL-12p7 and IFN-γ secretion.
the TRAIL pathway. In spite of the increase in NKG2D+ T cell numbers Although several studies have demonstrated the cytotoxic activity
observed after the administration of resveratrol to healthy subjects, of polyphenols in a large number of cancer lines, it is not clear when
this suggests the need for a future clinical study evaluating the polyphenols induce DAMP release and immunogenic cell death. In
relationship between resveratrol intake and the prevention of cancer. fact, in vitro curcumin treatment induces apoptosis in renal cancer
J.T.G. de Carvalho et al. / Journal of Nutritional Biochemistry 85 (2020) 108428 7

Improve
Imunotherapy
Tumor vaccine Immune response
NK adjuvant Checkpoints:
PD-L1
T CD8+

M2 cells Cytotoxic
T CD8+
activity
POLYPHENOLS APC

T CD4+
M1 cells T reg

Imunogenic
Cell DAMPs
Death release Th2

Th1

Fig. 3. The antitumor and immunoregulatory actions of polyphenols: polyphenols as resveratrol and gallotannin-rich fraction obtained from Caesalpinia spinosa (P2Et) act to enhance NK
cell antitumor activity. Curcumin, epicatechin gallate (EGCC), resveratrol and rutin inhibit the protumoral M2 polarized macrophages and enhancement the polarization of
macrophages toward the M1 phenotype, which exerts antitumor effects. EGCC and curcumin improved the response from tumor-based vaccines to induce an in vivo adaptive immune
response against a tumoral antigen. Resveratrol and P2Et induce immunogenic cell death and DAMPs release, which, in turn, induce cellular immunity by activating Th1 cell responses. In
addition, an epigallocatechin-3-gallate (EGCG)-rich extract green tea, curcumin, and P2Et improve cancer immunity by shift from a Th2 or Treg balance to a Th1 response against the
tumoral antigen. Bisdemethoxycurcumin suppresses PD-L1 expression, thus enhancing the TCD8+ cytotoxically activity and immune surveillance. On the contrary, other polyphenols
such as curcumin, apigenin, resveratrol and piceatannol increase PD-L1 expression and might improve immunotherapy against PD-L1-positive cancer cells.

[91], melanoma [92] and lung cancer cells [93], while quercetin 3.3. Polyphenol activation of Th1/CD8+ T cell-mediated immunity
induces apoptosis in cervical carcinoma [94], colon cancer [95] and
lymphoma cells [96]. Similarly, many tannins, such as EGCG, have been Recent studies have reported that polyphenols exert cytotoxic
described to be cytotoxic to ovarian [97] and breast [98] cancer cells, effects by activating antitumor immunity. Chen et al. [100] investi-
and many extracts rich in polyphenols also show cytotoxic activities gated the effects of resveratrol on the renal carcinoma microenviron-
[99,100]. Hence, further studies should be performed to investigate ment and found a switch from the expression of Th2 cytokines, such as
the possible immunogenic potential of a large number of polyphenols. IL-4 and IL-10, to the expression of Th1 cytokines with predominance
In cellular models of cancer, effective concentrations of polyphenols to of IFN-γ and increased infiltration of tissues by activated CD8+ T cells.
shown an antitumoral effect are usually high ranging from 80 to 200 In addition, cytotoxicity was found to be increased via the up-
μM, that concentration to modulate NK activity which are usually regulation of perforin, granzyme B and FasL expression, concomitant
lower ranging from 10 to 60 μM, suggesting a pleiotomism between with the up-regulation of Fas expression on tumor cells. Lin et al. [103]
these actions induced by polyphenols for each cells, which could demonstrated that rutin, a flavonoid compound, resulted in reductions
exhibit implication in clinical approaches. in tumors in the liver and spleen established by injection of a murine
Other studies have reported the optimization of combinations of myelomonocytic leukemia cell line (WEHI-3) into BALB/c mice and
polyphenols to improve the efficacy of cancer vaccines in preclinical increases in the expression of markers of adaptative immunity, CD3
and clinical studies. Kang et al. [101] evaluated the potential of EGCG and CD19, in peripheral cells after treatment. Although CD11b/
to increase DNA vaccination protection in a murine skin tumor model integrin and Mac3 (macrophage surface glycoprotein) expression
established with HPV-16 E7-expressing TC-1 cells. For this cancer was decreased, macrophage phagocytosis increased in the rutin-
vaccine, the gene lysosomal-associated membrane protein 1 (LAMP- treated group. Additionally, Oršolić et al. (77) studied the effect of
1) represented an effective means of delivering DNA directly into APCs caffeic acid by Ehrlich ascites tumors (EAT) cell inoculation in male
in the skin and thus allowing T cell activation. The combination of Swiss albino mice. After caffeic acid administration, increases in the
EGCG and DNA vaccination led to an enhanced tumor-specific T cell- levels of the cytokines IL-2, IL-12 and IFN-γ and decreases in the levels
mediated immune response and improved antitumor effects, resulting of IL-4 and IL-10 were found.
in a higher cure rate than either immunotherapy or EGCG alone. Lu et Chang et al. [104] and Bergman et al. [33] investigated the effects of
al. [102] created a formulation of curcumin in micelles and polyphenols on cocultures of PBMCs and cancer cell lines. In the first
administered it to mice challenged with the B16F10 melanoma cell study, the authors showed that linalool and p-coumaric acid stimulated
line, which resulted in decreases in IL-6 and chemokine C-C motif the production of proinflammatory cytokines, such as IFN-γ, IL-12 p40, IL-
ligand 2 levels and enhancements in TNF-α and IFN-γ levels. More 1β and TNF-α, and cell–cell interaction markers, such as CD40-ligand and
importantly, curcumin enhanced the vaccine activity of a peptide CD40, when used to stimulate lymphocytes from healthy donors in
derived from a tumor-associated antigen (Trp2), resulting in strong cocultures with the tumor cell line A549 (lung), T-47D (breast), SW-620
tumor inhibition compared with either the Trp2 vaccine or curcumin (colon) or Hep G2 (liver). These results suggest the capacity of phenolic
alone, which showed slight tumor inhibition. These studies suggest compounds to induce an adequate Th1 response. In the second study, the
further perspectives for polyphenols combined with cancer vaccina- authors observed that the combination of PBMCs, resveratrol and
tion approaches. colorectal cells resulted in the induction of apoptosis in cancer cell lines
8 J.T.G. de Carvalho et al. / Journal of Nutritional Biochemistry 85 (2020) 108428

but had variable effects on the cytokine response, whereas the Other studies have evaluated the ability of polyphenols to
combination of PBMCs and HT29 cells resulted in decreased cytokine synergize with immunotherapy. Shao et al. [111] found that a
levels in the presence of resveratrol. The effects on cytokine production combination of an anti-PD-L1 antibody and bisdemethoxycurcumin
varied in a dose-dependent manner for RKO cells cocultured with PBMCs. resulted in a reduction in tumor volume and increased survival in an in
In the presence of different resveratrol concentrations, the levels of IL-1β, vivo mouse model of bladder cancer that was established via
IFN-γ and IL-10 generally dropped, but low resveratrol doses stimulated intravenous injection of MB49 tumor cells. Moreover, bisdemethoxy-
TNF-α expression, while higher doses greatly reduced this expression. curcumin increased the antitumoral activity of CD8+ T cells by
These results suggest that the effects of polyphenols depend on the reducing PD-1 expression, which was accompanied by elevated levels
dosage and the cancer type. Therefore, further conclusive details are of IFN-γ and granzyme B release from CD8+ cells. Zhang et al. [90]
required to evaluate the dose–response effects of polyphenols. observed that resveratrol in combination with an anti-PD-1 antibody
in a model of ovarian carcinoma greatly inhibited tumor growth, while
depletion of CD8+ T cells restored tumor progression.
3.4. Polyphenols suppress Treg activation and function
In contrast, Lucas et al. [112] found enhanced expression of PD-L1
induced by treatment with resveratrol and piceatannol in the triple-
Other studies have demonstrated that polyphenols, such as
negative breast cancer cell line Cal51 and in the colon cancer cell line
curcumin, induce cancer immunity, which is related to the suppres-
SW620. The authors hypothesized that the induction of PD-L1
sion of Treg function and activation. Zou et al. [105] evaluated the
expression in tumor cells can sensitize the tumor cells to improve
effect of treatment for 2 weeks on lung cancer patients. After therapy,
the response to PD-L1 blockade. However, whether resveratrol and/or
an increase in IFN-secreting Th1 cells and suppression of Tregs were
piceatannol intake produces positive outcomes in combination with
observed. Xu et al. [106] similarly evaluated the effect of treatment for
PD-L1 blockade remains unclear because an increase in PD-L1
2 weeks on colon carcinoma patients and demonstrated a reduction in
expression may also impair host T cell-mediated immunity. Thus, to
the frequency of Treg cells, while the frequency of T effector (Foxp3−)
address this hypothesis, whether there is an absence of up-regulated
cells was increased. Another clinical study evaluated the effect of an
PD1 expression induced by these polyphenols in T cells needs to be
EGCG-rich extract from green tea on chronic lymphocytic leukemia
investigated.
patients. The extract induced decreases in the circulating lymphocyte
The duality of polyphenols claims special attention in regard to the
and Treg cell levels, usually observed in these patients, and reductions
in vivo intake of dietary supplements (including natural extracts),
in the circulating levels of IL-10 and TGF-β [107].
which are widely used among patients with cancer to reduce side
Interestingly, pleiotropic effects have been observed for phenolic
effects. Whereas some authors have reported down-regulation of PD-
compounds. In a melanoma establishment model, Lasso et al. [86]
L1 expression by polyphenols, indicating that the use of polyphenols
observed decreases in IL-10 production and FoxP3 expression
together with PD-L1 blockade therapy could be antagonistic, others
following treatment with P2Et, but in contrast, under nontumor
have demonstrated the inverse effect, specifically that polyphenols
conditions, an increase in the Treg frequency was seen in vivo in
can enhance the immune escape of tumor cells and thus act as
healthy BALB/c or C57BL/6 mice given the same treatment. In fact,
protumor agents. Hence, further preclinical studies should investigate
Wong et al. [108] demonstrated the induction of regulatory T cells by
the effect of polyphenols on PD-L1 expression in other cell lines to
the green tea-derived polyphenol EGCG. These results suggest that the
confirm the opposing effects of polyphenols before clinical use in
effects of polyphenols also depend on other factors, such as the
patients.
activation state of target cells (e.g., basal versus primed) and their
microenvironment (e.g., healthy versus cancer cells). In addition, it is
3.6. Polyphenols suppress TAMs
important to note that a phenolic compound, such as EGCG, may have
clinical applicability in the treatment of cancer and inflammatory
Recent works have investigated the ability of polyphenols to
disease, which were, until now, believed to be antagonistic target
suppress the M2-like polarization of TAMs. Sun et al. [113]
interactions, and investigations to elucidate these opposing activities
demonstrated that M2 polarization induced by lung cancer cells was
might reflect a new advance in the understanding of the pathophys-
inhibited by resveratrol. Additionally, an inhibitory function was
iology of these diseases.
observed in lung cancer cells cocultured with human macrophages. In
Another important aspect that remains unknown is whether
addition, Kimura et al. [114] found that resveratrol suppressed the
polyphenols can modulate CTLA-4 expression in naïve T cells. To
formation of new lymphatic vessels (similarly to angiogenesis) by
date, there have been no studies that address this question. Based on
lymphatic endothelial cells via inhibition of the production of IL-10,
studies of an inflammatory model, Sharma et al. [109] demonstrated
monocyte chemoattractant protein-1 and VEGF in M2 macrophages.
down-regulation of CD28 and CD80 expression and up-regulation of
Jang et al. [115] investigated the effect of EGCG on Balb/c tumors
CTLA-4 expression by curcumin. However, as described above, the
induced by the murine breast cancer cell line 4T1. There were a
immune regulation mediated by polyphenols is dependent on the
decrease in the level of CCL-2 and a reduction in the infiltration of
microenvironment, and further studies in cancer cell models should be
TAM-like M2 macrophages. These effects were associated with the up-
carried out.
regulation of microRNA-16 (miR-16) expression in tumor cells via
exosomes. Mukhereej et al. recently formulated liposomes with
3.5. PD/PD-L1 checkpoint modulation by polyphenols curcumin, EGCG and resveratrol. They treated mice with tumors
derived from glioblastoma cells (GL261) [116] or TC-1 cells infected
Recent studies have evaluated the ability of polyphenols to affect with HPV [117]. As a result, they observed reduced tumor growth in
the expression of PD-L1 in cancer cells. Xu et al. [110] observed both studies, as well as modification of the phenotype of TAMs, which
suppression of PD-L1 expression in human melanoma cells (A375, switched from the M2 phenotype marked as higher expression of
A2058 and RPMI-7951) treated with curcumin and apigenin. In arginase and IL-10 to the M1 phenotype identified by higher
addition, they found that treatment of cocultures (A375 cells or Jurkat expression of and lower expression of inducible nitric oxide synthase
cells) with these flavonoids potentiated T cell-mediated melanoma (iNOS) and IL-12.
cell killing. Taken together, these results suggest that curcumin and Guruvayoorappan and Kuttan [118] evaluated the antiangiogenic
apigenin may become an alternative therapeutic approach for activity of rutin by monitoring in vivo B16-F10 melanoma cell-induced
immune checkpoint blockade. capillary formation in C57BL/6 mice. The administration of rutin
J.T.G. de Carvalho et al. / Journal of Nutritional Biochemistry 85 (2020) 108428 9

significantly inhibited (43.35%) the number of tumor-directed point of view, the data are still inconclusive, and further preclinical
capillaries, which was accompanied by reductions in the levels of studies should be performed before beginning clinical use in patients.
VEGF and IL-1β and an increase in the level of TNF-α. We propose that future studies investigate PD-1 expression in T cells,
Oršolić et al. [119] studied the effect of caffeic acid on Ehrlich CTLA-4 expression in naïve T cells, interactions with immunotherapy,
ascites tumors induced by EAT cell inoculation in male Swiss albino the mechanism underlying pleiotropism (cancer versus inflamma-
mice. Caffeic acid administration produced a significant decrease in tion) and dose responsiveness in different cancer lines and immune
tumor growth and increased the life span of the mice by 28.39%. The cells with a special focus on resveratrol, curcumin and EGCC since
effect on macrophage polarization was evidenced by decreased Arg1 more data are available for those compounds. In addition, further
activity in splenic and peritoneal macrophages and an increased studies for improve the bioavailability profile of polyphenols com-
percentage of macrophages in the peritoneum. In addition, there was pounds are essentials to clinical use. Together, these data could clarify
significant inhibition of neovascularization and the VEGF level in the the role of polyphenols in cancer immunity to more precisely define
peritoneal fluid after caffeic acid administration. These studies suggest the potential of the clinical application of polyphenols in immuno-
that some polyphenols suppress M2 polarization, inhibiting tumor oncology.
progression and metastasis.
Conflicts of interest
4. Bioavailability of polyphenols
The authors declare that they have no conflicts of interest.
Large numbers of studies mentioned above have demonstrated
that polyphenols have immunoregulatory effects on cancer microen- Acknowledgments
vironment. Most of these biologic studies have been conducted in
vitro, i.e., utilizing cultured cells, which could not reflect the same This work was supported by grants from Universidade Federal da
effect when expanded to clinical use in human patients. Even though a Grande Dourados (UFGD); Fundação de Apoio ao Desenvolvimento do
compound has strong activities in vitro, it would have little biological Ensino Ciência e Tecnologia do Estado de Mato Grosso do Sul (FUNDECT);
activity in vivo if little or none of the compound gets to the target Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES);
tissues [120]. A major reason for the in vitro/in vivo discrepancies may Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq)
be the lack of bioavailability of polyphenols in vivo, in particular after and Financiadora de Estudos e Projetos (FINEP).
oral administration [121,122]. Large studies follow the general
directions for improving bioavailability of polyphenols by generation
References
of analogs using nanostructure-based drug delivery system, lipid-
based carriers and others [120]. [1] Di Ferdinando M, Brunetti C, Agati G, Tattini M. Multiple functions of
Another parameter that should be taken into account in in vitro polyphenols in plants inhabiting unfavorable Mediterranean areas. Environ
Exp Bot. 2014;103:107–16.
studies is if the test substance is appropriate. The most amounts of [2] González S, Fernández M, Cuervo A, Lasheras C. Dietary intake of polyphenols
dietary polyphenols exist in food as aglycones or glycosides. In and major food sources in an institutionalised elderly population. J Hum Nutr
glycosides forms, the polyphenols cannot be absorbed and must be Diet Off J Br Diet Assoc. 2014;27:176–83.
[3] Tsao R. Chemistry and biochemistry of dietary polyphenols. Nutrients. 2010;2:
hydrolyzed by the intestinal enzymes or by the colonic microflora 1231–46.
before absorption [121,123]. After absorption, they reach the liver [4] Niedzwiecki A, Roomi MW, Kalinovsky T, Rath M. Anticancer efficacy of
where they may be further subjected to extensive first-pass phase II polyphenols and their combinations. Nutrients. 2016:8.
[5] Abdal Dayem A, Choi HY, Yang G-M, Kim K, Saha SK, Cho S-G. The anti-cancer
metabolism (including glucuronidation, methylation, sulfation or a effect of polyphenols against breast cancer and cancer stem cells: molecular
combination of these), resulting in a series of metabolites derivatives mechanisms. Nutrients. 2016:8.
[121,122]. All these modifications deeply affect the biological activity of [6] Ding S, Jiang H, Fang J. 2018 Regulation of immune function by polyphenols J.
Immunol Res. 2018:1264074.
polyphenols. Consequently, the compounds that reach cells and tissues
[7] Mileo AM, Nisticò P, Miccadei S. Polyphenols: Immunomodulatory and
are chemically, biologically and, in many instances, functionally therapeutic implication in colorectal Cancer. Front Immunol. 2019;10:729.
different from the original dietary form [124]. Often, the compounds [8] Mougel A, Terme M, Tanchot C. Therapeutic cancer vaccine and combinations
tested were polyphenol aglycones or their sugar conjugates rather than with antiangiogenic therapies and immune checkpoint blockade. Front Immu-
nol. 2019;10:467.
their active metabolites. Thus, many investigators tested the wrong [9] Kalbasi A, Ribas A. Tumour-intrinsic resistance to immune checkpoint blockade.
compound when trying to unravel which physiological mechanisms Nat Rev Immunol. 2019:1–15.
were involved in the health effects of the polyphenols. [10] Wieder T, Eigentler T, Brenner E, Röcken M. Immune checkpoint blockade
therapy. J Allergy Clin Immunol. 2018;142:1403–14.
To avoid these problems, the best choice to do is on in vivo [11] Whiteside TL. The role of immune cells in the tumor microenvironment. Cancer
experiments. As far as animal studies are concerned, they provide Treat Res. 2006;130:103–24.
extremely valuable information and overcome some of the difficulties [12] Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al.
Understanding the tumor immune microenvironment (TIME) for effective
associated with in vitro studies utilizing cell culture. However, therapy. Nat Med. 2018;24:541–50.
differences between the human and animal may also lead to potential [13] Gajewski TF, Schreiber H, Fu Y-X. Innate and adaptive immune cells in the tumor
problems of extrapolation. For example, some metabolites generated microenvironment. Nat Immunol. 2013;14:1014.
[14] Gun SY, Lee SWL, Sieow JL, Wong SC. Targeting immune cells for cancer therapy.
in rodents were not present in humans [125]. Redox Biol. 2019:101174.
Thus, future preclinical studies should be conducted considering all [15] Maimela NR, Liu S, Zhang Y. Fates of CD8+ T cells in tumor microenvironment.
these factors, such as bioactive metabolite interference, the use of Comput Struct Biotechnol J. 2018;17:1–13.
[16] Tsukumo S-I, Yasutomo K. Regulation of CD8+ T cells and antitumor immunity
analogues with better bioavailability profile or formulations with
by Notch signaling. Front Immunol. 2018;9:101.
better bioavailability in conjunction with the use of animal models. [17] Smyth MJ, Cretney E, Kelly JM, Westwood JA, Street SEA, Yagita H, et al.
Activation of NK cell cytotoxicity. Mol Immunol. 2005;42:501–10.
[18] Chan CJ, Smyth MJ, Martinet L. Molecular mechanisms of natural killer cell
5. Conclusion
activation in response to cellular stress. Cell Death Differ. 2014;21:5–14.
[19] Morvan MG, Lanier LL. NK cells and cancer: you can teach innate cells new tricks.
Different mechanisms support the antitumor and immunoregula- Nat Rev Cancer. 2016;16:7–19.
tory effects of polyphenols, including the suppression of immune [20] Anon NK cells and cancer: you can teach innate cells new tricks | Nature Reviews
Cancer
escape mechanisms (Tregs and PD-L1), TAM suppression and [21] Zwirner NW, Ziblat A. Regulation of NK cell activation and effector functions by
adaptative immune activation (memory). From an efficacy and safety the IL-12 family of cytokines: the case of IL-27. Front Immunol. 2017;8:25.
10 J.T.G. de Carvalho et al. / Journal of Nutritional Biochemistry 85 (2020) 108428

[22] Barrow AD, Martin CJ, Colonna M. The natural cytotoxicity receptors in health [55] Montico B, Nigro A, Casolaro V, Dal Col J. Immunogenic apoptosis as a novel tool
and disease. Front Immunol. 2019;10. for anticancer vaccine development. Int J Mol Sci. 2018;19.
[23] Anon Frontiers | The natural cytotoxicity receptors in health and disease | [56] Nigro A, Montico B, Casolaro V, Dal Col J. A novel dendritic cell-based vaccination
Immunology protocol to stimulate immunosurveillance of aggressive cancers. Methods Mol
[24] Geary CD, Sun JC. Memory responses of natural killer cells. Semin Immunol. Biol Clifton NJ. 2019;1884:317–33.
2017;31:11–9. [57] Lopes A, Vandermeulen G, Préat V. Cancer DNA vaccines: current preclinical and
[25] Min-Oo G, Kamimura Y, Hendricks DW, Nabekura T, Lanier LL. Natural killer clinical developments and future perspectives. J Exp Clin Cancer Res. 2019;38:146.
cells: walking three paths down memory lane. Trends Immunol. 2013;34:251–8. [58] Yang B, Jeang J, Yang A, Wu TC, Hung C-F. DNA vaccine for cancer
[26] Lai Y-P, Jeng C-J, Che S-C. The Roles of CD4+ T Cells in Tumor Immunity. Int Sch immunotherapy. Hum Vaccines Immunother. 2014;10:3153–64.
Res. 2011. https://doi.org/10.5402/2011/497397 497397. [59] Shang N, Figini M, Shangguan J, Wang B, Sun C, Pan L, et al. Dendritic cells based
[27] Farhood B, Najafi M, Mortezaee K. CD8+ cytotoxic T lymphocytes in cancer immunotherapy. Am J Cancer Res. 2017;7:2091–102.
immunotherapy: a review J. Cell Physiol. 2019;234:8509–21. [60] Parmiani G, Russo V, Maccalli C, Parolini D, Rizzo N, Maio M. Peptide-based
[28] Durgeau A, Virk Y, Corgnac S, Mami-Chouaib F. Recent advances in targeting CD8 vaccines for cancer therapy. Hum Vaccines Immunother. 2014;10:3175–8.
T-cell immunity for more effective cancer immunotherapy. Front Immunol. [61] Darvin P, Toor SM, Nair VS, Elkord E. Immune checkpoint inhibitors: recent
2018;9:14. progress and potential biomarkers. Exp Mol Med. 2018;50:1–11.
[29] Goping IS, Barry M, Liston P, Sawchuk T, Constantinescu G, Michalak KM, et al. [62] Chang X, Lu X, Guo J, Teng G-J. Interventional therapy combined with immune
Granzyme B-induced apoptosis requires both direct caspase activation and relief checkpoint inhibitors: emerging opportunities for cancer treatment in the era of
of caspase inhibition. Immunity. 2003;18:355–65. immunotherapy. Cancer Treat Rev. 2019;74:49–60.
[30] Sutton VR, Wowk ME, Cancilla M, Trapani JA. Caspase activation by Granzyme B [63] Broglie L, Gershan J, Burke MJ. Checkpoint inhibition of PD-L1 and CTLA-4 in a
is indirect, and caspase autoprocessing requires the release of proapoptotic child with refractory acute leukemia. Int J Hematol Oncol. 2019;8.
mitochondrial factors. Immunity. 2003;18:319–29. [64] Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and
[31] Loh J, Thomas D A, Revell P A, Ley T J, Virgin H W and IV 2004 Granzymes and implications of their inhibition. Am J Clin Oncol. 2016;39:98–106.
caspase 3 play important roles in control of Gammaherpesvirus latency. J Virol [65] Mahoney KM, Freeman GJ, McDermott DF. The next immune-checkpoint
78 12519 inhibitors: PD-1/PD-L1 blockade in melanoma. Clin Ther. 2015;37:764–82.
[32] Yolcu ES, Shirwan H, Askenasy N. Fas/Fas-ligand interaction as a mechanism of [66] Kondĕlková K, Vokurková D, Krejsek J, Borská L, Fiala Z, Ctirad A. Regulatory T
immune homeostasis and β-cell cytotoxicity: enforcement rather than neutral- cells (TREG) and their roles in immune system with respect to immunopath-
ization for treatment of type 1 diabetes. Front Immunol. 2017;8. ological disorders. Acta Med (Hradec Kralove). 2010;53:73–7.
[33] Bergmann-Leitner ES, Abrams SI. Differential role of Fas/Fas ligand interactions [67] Zaini RG, Al-Rehaili AA. The therapeutic strategies of regulatory T cells in
in cytolysis of primary and metastatic colon carcinoma cell lines by human malignancies and stem cell transplantations. J Oncologia. 2019;2019:5981054.
antigen-specific CD8+ CTL. J Immunol. 2000;164:4941–54. [68] Romano M, Fanelli G, Albany CJ, Giganti G, Lombardi G. Past, present, and future
[34] Malyshkina A, Littwitz-Salomon E, Sutter K, Zelinskyy G, Windmann S, of regulatory T cell therapy in transplantation and autoimmunity. Front
Schimmer S, et al. Fas ligand-mediated cytotoxicity of CD4+ T cells during Immunol. 2019;10.
chronic retrovirus infection. Sci Rep. 2017;7:1–10. [69] Hu W, Wang G, Huang D, Sui M, Xu Y. Cancer immunotherapy based on natural
[35] Zhang S, Liu H, Yu H, Cooper GJS. Fas-associated death receptor signaling evoked killer cells: current progress and new opportunities. Front Immunol. 2019;10.
by human amylin in islet beta-cells. Diabetes. 2008;57:348–56. [70] Wellenstein MD, de Visser KE. Cancer-cell-intrinsic mechanisms shaping the
[36] Ostroumov D, Fekete-Drimusz N, Saborowski M, Kühnel F, Woller N. CD4 and tumor immune landscape. Immunity. 2018;48:399–416.
CD8 T lymphocyte interplay in controlling tumor growth. Cell Mol Life Sci. 2018; [71] Planes-Laine G, Rochigneux P, Bertucci F, Chrétien A-S, Viens P, Sabatier R, et al.
75:689–713. PD-1/PD-L1 targeting in breast cancer: the first clinical evidences are emerging—
[37] Kawabe T, Matsushima M, Hashimoto N, Imaizumi K, Hasegawa Y. CD40/CD40 a literature review. Cancers. 2019;11:1033.
ligand interactions in immune responses and pulmonary immunity. Nagoya J [72] Marra A, Viale G, Curigliano G. Recent advances in triple negative breast cancer:
Med Sci. 2011;73:69–78. the immunotherapy era. BMC Med. 2019:17.
[38] Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili S-A, [73] Corrales L, Scilla K, Caglevic C, Miller K, Oliveira J, Rolfo C. Immunotherapy in
Mardani F, et al. Macrophage plasticity, polarization, and function in health and lung cancer: a new age in cancer treatment. Adv Exp Med Biol. 2018;995:65–95.
disease. J Cell Physiol. 2018;233:6425–40. [74] Beatty GL, Gladney WL. Immune escape mechanisms as a guide for cancer
[39] Ganss R, Ryschich E, Klar E, Arnold B, Hämmerling GJ. Combination of T-cell immunotherapy. Clin Cancer Res. 2015;21:687–92.
therapy and trigger of inflammation induces remodeling of the vasculature and [75] Bryan Iorgulescu J, Braun D, Oliveira G, Keskin DB, Wu CJ. Acquired mechanisms
tumor eradication. Cancer Res. 2002;62:1462–70. of immune escape in cancer following immunotherapy. Genome Med. 2018:10.
[40] Gyobu H, Tsuji T, Suzuki Y, Ohkuri T, Chamoto K, Kuroki M, et al. Generation and [76] Lin Y, Xu J, Lan H. Tumor-associated macrophages in tumor metastasis: biological
targeting of human tumor-specific Tc1 and Th1 cells transduced with a lentivirus roles and clinical therapeutic applications. J Hematol OncolJ Hematol Oncol.
containing a chimeric immunoglobulin T-cell receptor. Cancer Res. 2004;64: 2019;12:76.
1490–5. [77] Yahaya M a. F, Lila M a. M, Ismail S, Zainol M and Afizan N A R N M 2019 Tumour-
[41] Tang X, Mo C, Wang Y, Wei D, Xiao H. Anti-tumour strategies aiming to target associated macrophages (TAMs) in colon cancer and how to reeducate them. J
tumour-associated macrophages. Immunology. 2013;138:93–104. Immunol Res
[42] Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P. Tumour-associated [78] Zhou D, Huang C, Lin Z, Zhan S, Kong L, Fang C, et al. Macrophage polarization and
macrophages as treatment targets in oncology Nat. Rev Clin Oncol. 2017;14: function with emphasis on the evolving roles of coordinated regulation of
399–416. cellular signaling pathways. Cell Signal. 2014;26:192–7.
[43] Blum JS, Wearsch PA, Cresswell P. Pathways of antigen processing. Annu Rev [79] van Dalen FJ, van Stevendaal MHME, Fennemann FL, Verdoes M, Ilina O. Molecular
Immunol. 2013;31:443–73. repolarisation of tumour-associated macrophages. Molecules. 2018:24.
[44] Sloan JM, Kershaw MH, Touloukian CE, Lapointe R, Robbins PF, Restifo NP, et al. [80] Mileo AM, Nisticò P, Miccadei S. Polyphenols: immunomodulatory and
MHC class I and class II presentation of tumor antigen in retrovirally and therapeutic implication in colorectal cancer. Front Immunol. 2019;10.
adenovirally transduced dendritic cells. Cancer Gene Ther. 2002;9:946–50. [81] Focaccetti C, Izzi V, Benvenuto M, Fazi S, Ciuffa S, Giganti MG, et al. Polyphenols
[45] Leone P, Shin E-C, Perosa F, Vacca A, Dammacco F, Racanelli V. MHC class I as immunomodulatory compounds in the tumor microenvironment: friends or
antigen processing and presenting machinery: organization, function, and foes? Int J Mol Sci. 2019;20.
defects in tumor cells. J Natl Cancer Inst. 2013;105:1172–87. [82] Cuevas A, Saavedra N, Salazar LA, Abdalla DSP. Modulation of immune function
[46] Kushwah R, Hu J. Complexity of dendritic cell subsets and their function in the by polyphenols: possible contribution of epigenetic factors. Nutrients. 2013;5:
host immune system. Immunology. 2011;133:409–19. 2314–32.
[47] Fu C, Jiang A. Dendritic cells and CD8 T cell immunity in tumor microenviron- [83] Hu L, Cao D, Li Y, He Y, Guo K. Resveratrol sensitized leukemia stem cell-like KG-
ment. Front Immunol. 2018;9. 1a cells to cytokine-induced killer cells-mediated cytolysis through NKG2D
[48] Holt MP, Punkosdy GA, Glass DD, Shevach EM. 2017 TCR signaling and CD28/ ligands and TRAIL receptors. Cancer Biol Ther. 2012;13:516–26.
CTLA-4 signaling cooperatively modulate T regulatory cell homeostasis. J [84] Luis Espinoza J, Takami A, Trung LQ, Nakao S. Ataxia-telangiectasia mutated
Immunol Baltim Md. 1950;198:1503–11. kinase-mediated upregulation of NKG2D ligands on leukemia cells by resveratrol
[49] Croft M, Dubey C. Accessory molecule and costimulation requirements for CD4 T results in enhanced natural killer cell susceptibility. Cancer Sci. 2013;104:
cell response. Crit Rev Immunol. 2017;37:261–90. 657–62.
[50] Zhou J, Wang G, Chen Y, Wang H, Hua Y, Cai Z. Immunogenic cell death in [85] Espinoza JL, Trung LQ, Inaoka PT, Yamada K, An DT, Mizuno S, et al. The repeated
cancer therapy: present and emerging inducers. J Cell Mol Med. 2019;23: administration of resveratrol has measurable effects on circulating T-cell subsets
4854–65. in humans. Oxidative Med Cell Longev. 2017;2017:6781872.
[51] Dudek AM, Garg AD, Krysko DV, De Ruysscher D, Agostinis P. Inducers of [86] Lasso P, Gomez-Cadena A, Urueña C, Donda A, Martinez-Usatorre A, Barreto A,
immunogenic cancer cell death. Cytokine Growth Factor Rev. 2013;24:319–33. et al. Prophylactic vs. therapeutic treatment with P2Et polyphenol-rich extract
[52] Garg AD, Martin S, Golab J, Agostinis P. Danger signalling during cancer cell has opposite effects on tumor growth. Front Oncologia. 2018;8.
death: origins, plasticity and regulation. Cell Death Differ. 2014;21:26–38. [87] Chen L, Wei Y, Zhao S, Zhang M, Yan X, Gao X, et al. Antitumor and
[53] Radogna F, Diederich M. Stress-induced cellular responses in immunogenic cell immunomodulatory activities of total flavonoids extract from persimmon leaves
death: implications for cancer immunotherapy. Biochem Pharmacol. 2018;153: in H22 liver tumor-bearing mice. Sci Rep. 2018;8:10523.
12–23. [88] Gomez-Cadena A, Urueña C, Prieto K, Martinez-Usatorre A, Donda A, Barreto A, et al.
[54] Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in Immune-system-dependent anti-tumor activity of a plant-derived polyphenol rich
cancer and infectious disease. Nat Rev Immunol. 2017;17:97–111. fraction in a melanoma mouse model. Cell Death Dis. 2016;7:e2243.
J.T.G. de Carvalho et al. / Journal of Nutritional Biochemistry 85 (2020) 108428 11

[89] Urueña C, Gomez A, Sandoval T, Hernandez J, Li S, Barreto A, et al. Multifunctional [108] Wong CP, Nguyen LP, Noh SK, Bray TM, Bruno RS, Ho E. Induction of regulatory T
T lymphocytes generated after therapy with an antitumor gallotanin-rich cells by green tea polyphenol EGCG. Immunol Lett. 2011;139:7–13.
normalized fraction are related to primary tumor size reduction in a breast [109] Sharma S, Chopra K, Kulkarni SK, Agrewala JN. Resveratrol and curcumin
cancer model. Integr Cancer Ther. 2015;14:468–83. suppress immune response through CD28/CTLA-4 and CD80 co-stimulatory
[90] Zhang Y, Yang S, Yang Y, Liu T. Resveratrol induces immunogenic cell death of pathway. Clin Exp Immunol. 2007;147:155–63.
human and murine ovarian carcinoma cells. Infect Agent Cancer. 2019;14:27. [110] Xu L, Zhang Y, Tian K, Chen X, Zhang R, Mu X, et al. Apigenin suppresses PD-L1
[91] Seo BR, Min K-J, Cho IJ, Kim SC, Kwon TK. Curcumin significantly enhances dual expression in melanoma and host dendritic cells to elicit synergistic therapeutic
PI3K/Akt and mTOR inhibitor NVP-BEZ235-induced apoptosis in human renal effects. J Exp Clin Cancer Res CR. 2018;37:261.
carcinoma Caki cells through down-regulation of p53-dependent Bcl-2 expres- [111] Shao Y, Zhu W, Da J, Xu M, Wang Y, Zhou J, et al. Bisdemethoxycurcumin in
sion and inhibition of Mcl-1 protein stability. PloS One. 2014;9:e95588. combination with α-PD-L1 antibody boosts immune response against bladder
[92] Zhao G, Han X, Zheng S, Li Z, Sha Y, Ni J, et al. Curcumin induces autophagy, cancer. OncoTargets Ther. 2017;10:2675–83.
inhibits proliferation and invasion by downregulating AKT/mTOR signaling [112] Lucas J, Hsieh T-C, Halicka HD, Darzynkiewicz Z, Wu JM. Upregulation of PD-L1
pathway in human melanoma cells. Oncol Rep. 2016;35:1065–74. expression by resveratrol and piceatannol in breast and colorectal cancer cells
[93] Wu S-H, Hang L-W, Yang J-S, Chen H-Y, Lin H-Y, Chiang J-H, et al. Curcumin occurs via HDAC3/p300-mediated NF-κB signaling. Int J Oncol. 2018;53:1469–80.
induces apoptosis in human non-small cell lung cancer NCI-H460 cells through [113] Sun L, Chen B, Jiang R, Li J, Wang B. Resveratrol inhibits lung cancer growth by
ER stress and caspase cascade- and mitochondria-dependent pathways. suppressing M2-like polarization of tumor associated macrophages. Cell
Anticancer Res. 2010;30:2125–33. Immunol. 2017;311:86–93.
[94] Bishayee K, Ghosh S, Mukherjee A, Sadhukhan R, Mondal J, Khuda-Bukhsh AR. [114] Kimura Y, Sumiyoshi M. Resveratrol prevents tumor growth and metastasis by
Quercetin induces cytochrome-c release and ROS accumulation to promote inhibiting Lymphangiogenesis and M2 macrophage activation and differentia-
apoptosis and arrest the cell cycle in G2/M, in cervical carcinoma: signal cascade tion in tumor-associated macrophages. Nutr Cancer. 2016;68:667–78.
and drug-DNA interaction. Cell Prolif. 2013;46:153–63. [115] Jang J-Y, Lee J-K, Jeon Y-K, Kim C-W. Exosome derived from epigallocatechin
[95] Kim GT, Lee SH, Kim YM. Quercetin regulates Sestrin 2-AMPK-mTOR signaling gallate treated breast cancer cells suppresses tumor growth by inhibiting tumor-
pathway and induces apoptosis via increased intracellular ROS in HCT116 colon associated macrophage infiltration and M2 polarization. BMC Cancer. 2013;13:
cancer cells. J Cancer Prev. 2013;18:264–70. 421.
[96] Granato M, Rizzello C, Gilardini Montani MS, Cuomo L, Vitillo M, Santarelli R, [116] Mukherjee S, Baidoo JNE, Sampat S, Mancuso A, David L, Cohen LS, et al.
et al. Quercetin induces apoptosis and autophagy in primary effusion lymphoma Liposomal TriCurin, a synergistic combination of curcumin, epicatechin
cells by inhibiting PI3K/AKT/mTOR and STAT3 signaling pathways. J Nutr gallate and resveratrol, repolarizes tumor-associated microglia/macro-
Biochem. 2017;41:124–36. phages, and eliminates glioblastoma (GBM) and GBM stem cells. Mol Basel
[97] Rao SD, Pagidas K. Epigallocatechin-3-gallate, a natural polyphenol, inhibits cell Switz. 2018;23.
proliferation and induces apoptosis in human ovarian cancer cells. Anticancer [117] Mukherjee S, Hussaini R, White R, Atwi D, Fried A, Sampat S, et al. TriCurin, a
Res. 2010;30:2519–23. synergistic formulation of curcumin, resveratrol, and epicatechin gallate,
[98] Braicu C, Gherman CD, Irimie A, Berindan-Neagoe I. Epigallocatechin-3-Gallate repolarizes tumor-associated macrophages and triggers an immune re-
(EGCG) inhibits cell proliferation and migratory behaviour of triple negative sponse to cause suppression of HPV+ tumors. Cancer Immunol Immunother
breast cancer cells. J Nanosci Nanotechnol. 2013;13:632–7. CII. 2018;67:761–74.
[99] Kwon Y. Food-derived polyphenols inhibit the growth of ovarian cancer cells [118] Girija Kuttan CG. Antiangiogenic effect of rutin and its regulatory effect on the
irrespective of their ability to induce antioxidant responses. Heliyon. 2018:4. production of VEGF, IL-1β and TNF-α in tumor associated macrophages —
[100] Chen L, Yang S, Liao W, Xiong Y. Modification of antitumor immunity and tumor SciAlert responsive version. J Biol Sci. 2007;1511–9.
microenvironment by resveratrol in mouse renal tumor model. Cell Biochem [119] Oršolić N, Kunštić M, Kukolj M, Gračan R, Nemrava J. Oxidative stress,
Biophys. 2015;72:617–25. polarization of macrophages and tumour angiogenesis: efficacy of caffeic acid.
[101] Kang TH, Lee JH, Song CK, Han HD, Shin BC, Pai SI, et al. Epigallocatechin-3- Chem Biol Interact. 2016;256:111–24.
gallate enhances CD8+ T cell-mediated antitumor immunity induced by DNA [120] D'Archivio M, Filesi C, Varì R, Scazzocchio B, Masella R. Bioavailability of the
vaccination. Cancer Res. 2007;67:802–11. polyphenols: status and controversies. Int. J Mol Sci. 2010;11:1321–42.
[102] Lu Y, Miao L, Wang Y, Xu Z, Zhao Y, Shen Y, et al. Curcumin micelles remodel [121] Cardona F, Andrés-Lacueva C, Tulipani S, Tinahones FJ, Queipo-Ortuño MI.
tumor microenvironment and enhance vaccine activity in an advanced Benefits of polyphenols on gut microbiota and implications in human health. J
melanoma model. Mol Ther J Am Soc Gene Ther. 2016;24:364–74. Nutr Biochem. 2013;24:1415–22.
[103] Lin J-P, Yang J-S, Lu C-C, Chiang J-H, Wu C-L, Lin J-J, et al. Rutin inhibits the [122] Chen L, Cao H and Xiao J 2018 2-Polyphenols: absorption, bioavailability, and
proliferation of murine leukemia WEHI-3 cells in vivo and promotes immune metabolomics polyphenols: properties, recovery, and applications ed C M
response in vivo. Leuk Res. 2009;33:823–8. Galanakis (Woodhead Publishing) pp 45–67
[104] Chang M-Y, Shen Y-L. Linalool exhibits cytotoxic effects by activating antitumor [123] Rubió L, Macià A, Motilva M-J. Impact of various factors on pharmacokinetics of
immunity. Mol Basel Switz. 2014;19:6694–706. bioactive polyphenols: an overview. Curr Drug Metab. 2014;15:62–76.
[105] Zou JY, Su CH, Luo HH, Lei YY, Zeng B, Zhu HS, et al. Curcumin converts Foxp3+ [124] Lee M-J, Maliakal P, Chen L, Meng X, Bondoc FY, Prabhu S, et al. Pharmacokinetics
regulatory T cells to T helper 1 cells in patients with lung cancer. J Cell Biochem. of tea catechins after ingestion of green tea and (−)-epigallocatechin-3-gallate
2018;119:1420–8. by humans: formation of different metabolites and individual variability. Cancer
[106] Xu B, Yu L, Zhao L-Z. Curcumin up regulates T helper 1 cells in patients with colon Epidemiol Biomark Prev Publ Am Assoc Cancer Res Cosponsored Am Soc Prev
cancer. Am J Transl Res. 2017;9:1866–75. Oncol. 2002;11:1025–32.
[107] D'Arena G, Simeon V, De Martino L, Statuto T, D'Auria F, Volpe S, et al. Regulatory [125] Mullen W, Edwards CA, Crozier A. Absorption, excretion and metabolite profiling
T-cell modulation by green tea in chronic lymphocytic leukemia. Int J of methyl-, glucuronyl-, glucosyl- and sulpho-conjugates of quercetin in human
Immunopathol Pharmacol. 2013;26:117–25. plasma and urine after ingestion of onions. Br J Nutr. 2006;96:107–16.

You might also like