Next Article in Journal
Genome-Wide Approach of Gene–Nutrient Intake Interaction Study for Essential Hypertension in a Large Korean Cohort (KoGES)
Next Article in Special Issue
Biological Effect of Food for Special Medical Purposes (NutramilTM Complex) on Melanoma Cells in In Vitro Study
Previous Article in Journal
Bone Fracture Incidence in Postmenopausal Women: Results of a 10 Year Follow Up in a RAC-OST-POL Study of rs1544410, rs7975232 and rs731236 Polymorphisms
Previous Article in Special Issue
Iodine Bioavailability and Biochemical Effects of Brassica oleracea var. sabellica L. Biofortified with 8-Hydroxy-7-iodo-5-quinolinesulfonic Acid in Wistar Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Polyphenols as Immunomodulators and Epigenetic Modulators: An Analysis of Their Role in the Treatment and Prevention of Breast Cancer

by
Esmanur Eren
1,
Jyotirmoyee Das
1 and
Trygve O. Tollefsbol
1,2,3,4,5,*
1
Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
2
Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL 35294, USA
3
O’Neal Comprehensive Cancer Research, University of Alabama at Birmingham, Birmingham, AL 35294, USA
4
Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
5
Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
*
Author to whom correspondence should be addressed.
Nutrients 2024, 16(23), 4143; https://doi.org/10.3390/nu16234143
Submission received: 5 November 2024 / Revised: 26 November 2024 / Accepted: 27 November 2024 / Published: 29 November 2024
(This article belongs to the Special Issue The Effect of Bioactive Compounds in Anti-inflammation)

Abstract

:
Breast cancer poses a substantial health challenge for women globally. Recently, there has been a notable increase in scholarly attention regarding polyphenols, primarily attributed to not only the adverse effects associated with conventional treatments but also their immune-preventive impacts. Polyphenols, nature-derived substances present in vegetation, including fruits and vegetables, have received considerable attention in various fields of science due to their probable wellness merits, particularly in the treatment and hindrance of cancer. This review focuses on the immunomodulatory effects of polyphenols in breast cancer, emphasizing their capacity to influence the reaction of adaptive and innate immune cells within the tumor-associated environment. Polyphenols are implicated in the modulation of inflammation, the enhancement of antioxidant defenses, the promotion of epigenetic modifications, and the support of immune functions. Additionally, these compounds have been shown to influence the activity of critical immune cells, including macrophages and T cells. By targeting pathways involved in immune evasion, polyphenols may augment the capacity of the defensive system to detect and eliminate tumors. The findings suggest that incorporating polyphenol-rich foods into the diet could offer a promising, collaborative (integrative) approach to classical breast cancer remedial procedures by regulating how the defense mechanism interacts with the disease.

1. Introduction

Breast cancer ranks among the top causes of cancer fatalities globally, with increasing incidence rates despite advancements in treatment [1,2]. This shift in focus toward prevention highlights the potential role of dietary interventions [3]. One such promising avenue is polyphenols, present in vegetable-derived products, which are associated with a myriad of health advantages, particularly in reducing cancer risk, owing to their antioxidant and inflammatory-suppressive qualities [3,4,5]. Beyond these well-established features, polyphenols can also influence the human body’s defense system and epigenetic regulation [6]. Polyphenols offer the potential for primary prevention by reducing the risk before the cancer develops [7]. They achieve this by modulating the molecular pathways associated with carcinogenesis and reversing epigenetic alterations, such as histone methylation and acetylation, DNA methylation, and microRNA expression [5,6,7,8,9].
Polyphenols, by modulating key modifications that influence tumor development, offer a potential approach for preventing breast cancer initiation and progression [10]. For instance, epigallocatechin gallate (EGCG), resveratrol, and curcumin have been thoroughly investigated for their remarkable potential in combating cancer. EGCG has been indicated to alleviate the development of breast cancer, induce apoptosis, and inhibit tumorigenesis [7,9,11,12]. Similarly, the influence of resveratrol and curcumin on epigenetic mechanisms and various signaling pathways, including those responsible for inflammation, cell survival, and metastasis implicated in the pathogenesis of breast cancer, has been extensively examined in the scientific literature [13]. Additionally, the immunomodulatory effects of polyphenols may enhance the body’s natural immune defenses, creating an environment that hinders tumor formation and progression [3,14]. In recent years, research has increasingly focused on understanding the connection between diet and cancer prevention, with particular interest in plant-derived compounds [13]. Polyphenols, among the most abundant classes of phytochemicals in human diets, have drawn significant attention due to their potential restorative impact across distinct types of cancers, including breast cancer [13]. These compounds are categorized into flavonoids, phenolic acids, lignans, and stilbenes, with notable examples being EGCG, resveratrol, curcumin, and quercetin [3,9,10,11]. The ability of these polyphenols to modulate both tumor cells and the tumor milieu, mainly through the defense system, opens new avenues for their integration into cancer therapy.
This review aims to fill a critical gap in understanding how polyphenols may contribute to breast cancer prevention through their immunomodulatory and epigenetic effects. By synthesizing available preclinical and clinical evidence, this analysis will provide insights that could inform future research and therapeutic strategies focused on dietary polyphenols as preventive agents.

2. Materials and Methods

Identification of multiple empirical and review articles was conducted through PubMed and Google Scholar databases. The search utilized a range of keywords, including “breast cancer”, “immunomodulatory effects of dietary polyphenols”, “dietary polyphenols”, “epigenetic effects of dietary polyphenols”, “dietary polyphenols in breast cancer”, and “breast cancer therapies”. Comprehensive searches for relevant research and review references were performed on PubMed. Descriptive statistical analyses and meta-analyses were not conducted.

3. Breast Cancer

Breast cancer is a widespread form of cancer in women worldwide, with both its occurrence and mortality rates recently increasing [15]. In accordance with the latest estimates released by the International Agency for Research on Cancer (IARC), the year 2022 witnessed reports of approximately 20 million new instances of cancer, along with 9.7 million fatalities [16]. Breast cancer ranked as the second predominant type of cancer, representing 11.6% of all new cancer cases globally [16]. Breast cancer can be classified into different subtypes based on molecular features, hormone receptor status, and histological patterns (Figure 1). The molecular subtypes of breast cancers are Luminal, Her2-enriched, basal-like, and Claudin-low breast cancers [17,18]. Luminal breast cancers account for 60–70% of all breast cancer types in economically advanced countries [19]. Luminal breast cancer is divided into two subtypes: Luminal A, which has a slower growth rate and better prognosis, and Luminal B, which has a faster growth rate and worse prognosis [15,20,21,22]. Luminal A breast cancers are characterized by the absence of HER2 and the presence of either an ER or a PR [23]. These cancers typically manifest in the mammary duct epithelium. In comparison, Luminal B breast cancers are estrogen receptor positive and may be PR negative and/or HER2 positive [15,17,20,22].

4. Immune System Regulation in Breast Cancer

The immune system is a sophisticated network that aids the body in defending against diseases. The management of the defense mechanism in breast cancer is also a complicated progression impacted by the defensive system’s cells and cancer cells and the surrounding microenvironment, which encompasses cancer and stromal cells, an extracellular matrix (ECM), signaling molecules, blood vessels, and hypoxic regions [26,37,38,39]. Cancer-associated fibroblasts are the primary cell type in the microenvironment [37,38,40]. In addition, it comprises a diverse array of immune cells (Figure 2), including lymphocytes, macrophages, myeloid-derived stromal cells, and cytokines [37,41,42].

5. Breast Cancer Treatment Types

Breast cancer (BC) treatment necessitates a multidisciplinary approach due to its various subtypes. There are numerous conventional and innovative treatment methods available, including surgery, radiation therapy, chemotherapy, hormone therapy, and immunotherapies [72]. These treatments can be used individually or in combination to address breast cancer. For early-stage invasive breast cancer, surgery and mastectomy are often preferred as they boast an 85–95% success rate in preventing tumor recurrence [72,73]. Radiotherapy plays a crucial role in the comprehensive treatment of breast cancer, and it is frequently utilized in conjunction with surgical intervention and chemotherapy to diminish the likelihood of disease recurrence, particularly subsequent to breast-conserving surgery or mastectomy [74,75]. Chemotherapy is a standard treatment method for breast cancer, incorporating the administration of drugs to eliminate cancer cells [72,76]. Its applicability across various stages of breast cancer underscores its diverse therapeutic purposes, tailored to individual cases, and it reduces the risk of recurrence of breast cancer to only 30% [72,76,77]. Hormone therapy serves as a fundamental treatment for hormone receptor-positive breast cancers, where cancer cells proliferate in response to estrogen or progesterone [78,79,80]. This therapeutic approach operates by either obstructing these hormones or diminishing their levels to decelerate or halt cancer progression [79,80,81]. Immunotherapy has garnered significant attention due to its potential to harness the body’s defense system to develop innovative therapeutic strategies [82,83]. Adoptive cell therapies (e.g., CAR-T cell therapy), vaccines (e.g., Personalized Peptide Vaccine), cytokine therapies (e.g., anti-IL-1β), monoclonal antibodies (e.g., Herceptin and Pertuzumab), and immune checkpoint (e.g., PD-1 and CTLA4) blockade represent foundational forms of immunotherapy [72,82,83,84,85,86,87,88,89].
Different breast cancer treatment modalities have distinct advantages and limitations depending on the breast cancer subtype. However, immunotherapy has been favored over traditional therapies in recent years for several reasons. Unlike chemotherapy or radiotherapy, which targets cancerous and healthy cells, immunotherapy harnesses the body’s immune system to selectively attack cancer cells, resulting in fewer side effects overall [84,90]. Although immunotherapy may have defense system-related side effects, they are generally less severe than those associated with chemotherapy or radiation [89,90]. Immunotherapy has also shown significant success in treating aggressive forms of breast cancer, such as TNBC, which is typically less responsive to hormonal or targeted therapies [91]. The efficacy of immunotherapy in the treatment of various subtypes of TNBC is known to vary, highlighting the ongoing need for safe and effective methods for both prevention and treatment [90,92,93,94,95]. This challenge has sparked growing interest in complementary approaches, such as dietary interventions rich in polyphenols, to enhance treatment outcomes [94,95].

6. Polyphenols and Their Importance

Polyphenols represent a wide range of natural substances found in plant-derived foods. They are known for their antioxidant impacts and have garnered significant attention due to their potential health advantages [96,97,98]. Structurally, polyphenols contain multiple phenol units, which play a key role in their biological activity [96,99]. These compounds are abundant in fruits, vegetables, tea, coffee, wine, spices, and whole grains [99]. Due to their wide availability in plant-based diets, polyphenols have become a central focus in nutritional research, particularly concerning disease prevention and health promotion [99]. Polyphenols (Figure 3) can be classified into four categories according to their chemical structure.
Flavonoids: This is the largest group of polyphenols, accounting for over half of all dietary polyphenols. The basic framework is made up of two aromatic rings connected by three carbons. Berries, citrus, oranges, onions, tea, and red wines are all rich in flavonoids [100,101]. Subclasses of flavonoids include flavanols (e.g., quercetin and catechins in green tea) and anthocyanins (e.g., pigments in berries) [102].
Phenolic Acids: These compounds contain a benzene ring with hydroxyl and carboxyl groups and are prevalent in foods like coffee, whole grains, and certain fruits [103]. They include caffeic acid and ferulic acid, both famous for their anti-cancer and inflammatory properties [103].
Stilbenes: While less common than flavonoids with two interconnected benzene rings, stilbenes such as resveratrol (found in grapes, red wine, and berries) are highlighted for the potential significance of their role in promoting longevity and cancer prevention [104,105].
Lignans: Found in seeds, particularly flaxseeds, as well as whole grains and vegetables, lignans are phytoestrogens that are believed to contribute to hormone-related cancer prevention [106].

7. Dietary Polyphenols and Their Impact on Immune System

Dietary polyphenols have been shown to impact multiple aspects of immune function, providing a means to restore or enhance anti-tumor immunity [107,108,109]. For instance, EGCG, a green tea polyphenol, is highlighted for its ability to constrain the rapid growth of cancer cells and promote apoptosis [110]. Beyond its direct effects on cancer cells, EGCG modulates immune responses by boosting the activity of cytotoxic T cells and natural killer cells, key players in targeting and killing cancer cells [109,111,112]. Additionally, EGCG has been shown to downregulate immunosuppressive regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) within the tumor microenvironment, thereby restoring an environment conducive to immune-mediated tumor destruction [112,113].
Curcumin, another well-known polyphenol from turmeric, exerts immunomodulatory effects by targeting a variety of immune pathways [112,114]. It can inhibit inflammation-stimulating immune modulators such as IL-6 and TNF-α, which are often elevated in patients with cancer and contribute to a chronic inflammatory state that promotes tumor growth [112,115,116]. Curcumin also elevates NK and dendritic cells’ activity, facilitating a more robust anti-tumor immune response. Furthermore, its ability to modulate immune checkpoints, such as programmed cell death protein 1 (PD-1), positions it as a favorable phytochemical for potential synergistic application in conjunction with immune checkpoint inhibitors in treating breast cancer [117,118].
Resveratrol, found in grapes and red wine, has also shown significant immunomodulatory potential. It has been reported to suppress the expression of programmed death-ligand 1 (PD-L1) in breast cancer cells, thereby reducing T cell exhaustion and promoting protection against tumor formation [119]. Resveratrol also influences the polarization of TAMs from a tumor-promoting M2 phenotype to a tumoricidal M1 phenotype, creating a more hostile environment for breast cancer cells [120,121].
The anti-inflammatory effects of quercetin, a flavonoid found in many fruits and vegetables, contribute to its immunomodulatory potential in breast cancer [122]. Quercetin can inhibit the formation of immune mediators that induce inflammation, like IL-1β and IL-8, which contribute to a tumor-promoting inflammatory environment [123]. By reducing this inflammation, quercetin may help to tip the balance in favor of an anti-tumor immune response [124].

8. Role of Polyphenols in Regulating the Immune System

Polyphenols are well known for their antioxidant properties, but their role in immune system regulation has gained increasing attention in recent years [3,125]. Polyphenols modulate immune responses through several mechanisms, affecting diverse aspects of immune function by interacting with specific receptors on various defense system cells [3,109]. Their immunomodulatory properties can contribute to the prevention and treatment of a range of diseases, including breast cancer [126].

8.1. Modulation of Immune Cells

Polyphenols exert their effects by influencing assorted defensive cells, such as T cells, B cells, NK cells, macrophages, and dendritic cells [3]. These immune cells possess specific recognition units that interact with polyphenols. For example, EGCG binds to the ZAP-70 receptor, whereas resveratrol links to the Sp1 receptor on different immune cell types. This receptor–ligand correspondence initiates signaling cascades that promote apoptosis and immune responses, such as increasing the activity of cytotoxic T cells and NK cells [3,121,127,128,129]. In addition, polyphenols help balance the immune system by stimulating the diversification of M1 macrophages (pro-inflammatory and anti-tumor) over M2 macrophages (associated with tumor growth and immune suppression) [130]. This shift in macrophage activity can enhance immunological monitoring and the body’s capacity to fight tumors.

8.2. Regulation of Inflammatory Responses

Chronic inflammation is a major driver of many diseases, including cancer [131]. Polyphenols play a crucial role in modulating the inflammatory response, primarily by affecting the production of cytokines—small signaling proteins that regulate immune and inflammatory responses [128]. Polyphenols like curcumin (from turmeric) and quercetin (from fruits and vegetables) reduce the secretion of cytokines driving inflammation, such as TNF-α, IL-6, and IL-1β, related to prolonged inflammation response and tumor progression [132,133].
At the same time, polyphenols promote the generation of interleukin-10, an inflammatory mediator, which helps mitigate excessive immune responses and prevent tissue damage [134,135]. This ability to regulate the balance of inflammatory driving and prevent cytokines helps maintain immune homeostasis and prevents the immune system from becoming more activated, which can lead to autoimmune disorders or chronic inflammation [3,135].

8.3. Immune Checkpoint Regulation

Cancer cells often evade detection and elimination by the immune system by exploiting immune checkpoints, which are regulatory molecules expressed on immune cells that need to be activated or deactivated to start an immune response [136,137]. Polyphenols, such as genistein (from soy) and resveratrol, govern these immune checkpoints [136]. For example, they can inhibit the PD-1/PD-L1 pathway, a mechanism through which cancerous cells subdue T cell activity, thereby helping the body’s defense system recognize and counteract tumor cells more effectively [62].
PD-1 is a transmembrane binding site on immune cells, including helper and cytotoxic T cells and NK cells [138,139,140,141]. The expression of PD-1 on T cells is governed by several pathways, including HIF-1α, NF-κB, PI3K/AKT/mTOR, and JAK/STAT [136,139,142]. Its ligand, PD-L1, is often overproduced in cancer cells, facilitating immune evasion [143]. The presence of PD-L1 in tumors is governed by the MAPK pathway, HIF-1α, and epigenetic mechanisms such as histone deacetylation, along with the JAK/STAT, Wnt, and PI3K/AKT/mTOR pathways [136,139]. Polyphenols have the potential to inhibit these pathways, reducing PD-L1 levels in cancer cells and enhancing T cell activation, which improves the recognition of cancer cells.

8.4. Regulation of T Cells and B Cells

Polyphenols exert significant impacts on the adaptive defense system by modulating lymphocytes’ activity. They are known to enhance the proliferation of B and T cells while concurrently suppressing the function of regulatory T cells and specific subsets of T helper cells, including Th9 and Th17 [3]. For example, resveratrol and curcumin can promote a Th1-type response, which is essential for anti-tumor immunity, while reducing the overactivation of Th2-type responses associated with allergic reactions and immune suppression [144]. Polyphenols also aid in the production of antibodies that are crucial for targeting pathogens and cancer cells by enhancing B cell function [111].

9. Synergistic Potential with Conventional Therapies

The immunomodulatory properties of dietary polyphenols suggest that they could be used in conjunction with conventional cancer therapies [125,145,146,147,148]. Chemotherapy and radiotherapy, while effective at killing tumor cells, often induce immunosuppressive effects, which can limit their long-term efficacy [149]. Polyphenols have the potential to enhance immune function, which could help mitigate the adverse effects associated with various conditions. Although conventional treatments can also impact healthy cells, polyphenols demonstrate the capacity to improve these outcomes due to their antioxidant properties, thereby reducing the side effects that traditional therapies may impose on the defense system. Furthermore, the integration of polyphenols with immunotherapeutic approaches, such as immune checkpoint inhibitors, may yield a synergistic effect by augmenting the activity of immune cells and overcoming the immune resistance mechanisms utilized by breast cancer cells [150,151].

10. Epigenetic Regulation by Polyphenols in Cancer Immunomodulation

Beyond their direct interactions with immune cells, polyphenols can modulate immune responses at the epigenetic level [111,152,153]. Epigenetic modifications, including histone methylation, acetylation, and DNA methylation, are pivotal in regulating gene expression [152,154,155]. Aberrant epigenetic changes are a hallmark of cancer, including in breast cancer, where they can silence tumor suppressor genes and activate oncogenes [9]. Dietary polyphenols have been shown to reverse these modifications, thereby restoring normal cellular functions and immune responses [156,157,158].
For example, EGCG has been reported to inhibit DNA methyltransferases (DNMTs), enzymes that add methyl groups to DNA, and silence gene expression [159,160]. By inhibiting DNMTs, EGCG can reactivate tumor suppressor genes and advocate the immune system’s ability to dispose of breast cancer cells [160,161]. Similarly, curcumin has been shown to modulate histone acetylation, thereby facilitating the reactivation of immune-related genes that had been silenced in the tumor microenvironment. Furthermore, curcumin influences the PD-1/PD-L1 immune checkpoint [9,162]. Moreover, resveratrol has been demonstrated to inhibit histone acetylation while regulating microRNAs, enhancing T cell activation, and reducing tumor progression. Quercetin monitors histone acetylation and modulates the subtype switching of macrophages.

11. Specific Polyphenols Used in Breast Cancer Treatment

As shown in Table 1, EGCG, resveratrol, curcumin, quercetin, genistein, apigenin, and pterostilbene are commonly researched polyphenols for the primary prevention and management of breast cancer [163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192].

11.1. Epigallocatechin-3-Gallate (EGCG)

EGCG (Figure 4) is a natural polyphenol, the most abundant catechin in green tea, and has been extensively studied for its anti-cancer properties, particularly in breast cancer. Its effects on cancer cells include inhibiting cell proliferation, inducing apoptosis, and modulating multiple signaling pathways, such as PI3K/Akt and MAPK [193]. EGCG has been shown to inhibit EGFR or ErBb1 and 2, which are often found to be overexpressed in breast cancer [193]. EGCG has also been shown to inhibit the expression of matrix metalloproteinases (MMPs), which play a crucial role in the progression and dissemination of tumors [194]. Furthermore, it exhibits anti-angiogenic properties by downregulating VEGF expression, thereby repressing new blood vessel generation that supplies tumors [195]. Preclinical studies have demonstrated that EGCG sensitizes breast cancer cells to conventional therapies, such as tamoxifen and trastuzumab, suggesting its potential as an adjunctive treatment [196,197].

11.2. Resveratrol

Resveratrol (Figure 5), a stilbene found in grapes, berries, and peanuts, has gained significant attention for its chemo-preventive and healing impacts in resistance to breast cancer [149,198]. The mechanisms of resveratrol include inducing programmed cell death, inhibiting cell proliferation, and suppressing the growth of estrogen receptor (ER)-positive breast cancer by downregulating ERα signaling [199]. Resveratrol has been shown to inhibit metastasis in TNBC cell lines by altering the TGF-β1-induced epithelial matrix transformation, thus providing promising primary prevention options for these types of cancers that have no cure except for chemotherapies. Moreover, resveratrol influences multiple molecular pathways, including the NF-κB and STAT3 pathways, which are crucial for cancer cell survival and proliferation [200,201]. In TNBC, resveratrol has shown the ability to reduce tumor growth by modulating the expression of oncogenic microRNAs and increasing tumor suppressor genes. Its capacity to penetrate the blood–brain barrier hints at its potential to help prevent the spread of breast cancer to the brain, which is a frequent issue in the advanced stages of the disease [202].

11.3. Curcumin

Curcumin, a key polyphenol (Figure 6) found in turmeric (Curcuma longa), is well recognized for its anti-inflammatory and anti-cancer properties. In breast cancer, it targets multiple signaling pathways, such as PI3K/Akt/mTOR and Wnt/β-catenin, which play pivotal roles in tumor development and progression [203,204]. Curcumin promotes apoptosis in cancer cells by activating caspase enzymes and downregulating anti-apoptotic proteins like Bcl-2 and Bcl-xL [205]. Moreover, it inhibits the epithelial–mesenchymal transition (EMT), a critical process involved in cancer metastasis [206]. Its anti-inflammatory effects are particularly relevant in breast cancer as chronic inflammation is strongly linked to tumor growth [207]. Additionally, curcumin enhances the effectiveness of chemotherapy and helps combat drug resistance in breast cancer cells [207,208].

11.4. Quercetin

Quercetin, a flavonoid (Figure 7) abundantly present in apples, onions, and citrus fruits, exhibits potent oncolytic outcomes in breast cancer via multiple molecular and cellular mechanisms [209]. It promotes apoptosis in breast cancer cells by upregulating pro-apoptotic proteins such as Bax and activating caspases while simultaneously reducing the levels of apoptosis-preventing proteins like Bcl-2 [210]. Beyond its direct anti-cancer activity, quercetin significantly influences immune modulation, contributing to its tumor-suppressive properties [3,211]. It enhances the immune system’s capacity to target and eliminate cancer cells by activating cytotoxic T cells and regulating T-helper cell responses to ensure immune homeostasis. Quercetin also modulates macrophage activity, suppressing the pro-inflammatory M1 phenotype and reducing the secretion of inflammatory cytokines such as TNF-α and IL-6, which are frequently elevated in tumor microenvironments [212]. Concurrently, it promotes a shift towards the M2 macrophage phenotype, fostering tissue repair and anti-inflammatory effects [133,213]. Furthermore, quercetin enhances the cytotoxic function of natural killer (NK) cells, strengthening their ability to directly target and destroy tumor cells [214]. The dual role of quercetin in immune regulation and tumor suppression underscores its potential as a multifaceted agent for the prevention and treatment of breast cancer.

11.5. Genistein

Genistein, a prominent soy-derived isoflavone, has gained considerable attention for its role in breast cancer prevention and treatment [215]. Studies suggest that Asian women consuming soy-rich diets have a lower incidence or occurrence of breast cancer [216]. In ER-positive breast cancer, genistein inhibits cell proliferation by competing with endogenous estrogens for ER binding, thereby reducing estrogen-driven tumor growth [217]. Additionally, genistein inhibits tyrosine kinase activity, affecting key signaling pathways such as PI3K/Akt and MAPK. This includes suppressing protein levels like MEK5 (mitogen-activated protein kinase 5) and ERK5 (extracellular signal-regulated kinase 5), [218,219,220], which are essential for cancer cell survival, consistent with cell growth inhibition and the induction of apoptosis [221,222]. Genistein induces apoptosis primarily through caspase activation, the modulation of endoplasmic reticulum stress regulators, and an increase in the Bax/Bcl-2 ratio. Other proposed mechanisms include proteasome activity inhibition, the downregulation of the anti-apoptotic protein survivin, and the suppression of angiogenesis and tumor progression in breast cancer [223]. Finally, studies also indicate that genistein enhances the efficacy of chemotherapeutic agents like tamoxifen, making it a potential adjunctive therapy in breast cancer treatment [224].

11.6. Apigenin

Apigenin, a flavone present in parsley, celery, and chamomile, has demonstrated promising anti-cancer properties in breast cancer research [225]. It exerts its effects by inducing cell cycle arrest at the G2/M phase and triggering apoptosis through caspase-3 activation while downregulating anti-apoptotic proteins such as Bcl-2 [226]. Beyond its pro-apoptotic activity, apigenin inhibits metastasis by reducing the expression of matrix metalloproteinases (MMPs) and suppressing epithelial–mesenchymal transition (EMT) [227,228]. Additionally, it mitigates inflammation within the tumor microenvironment by inhibiting the NF-κB pathway, a critical regulator of inflammation-driven tumor progression [229,230]. Notably, apigenin enhances the vulnerability of breast cancer cells to radiation and chemotherapy, highlighting its feasibility as a complementary therapeutic agent [231].

11.7. Pterostilbene

Pterostilbene, a naturally occurring demethylated derivative of resveratrol, is found in blueberries and grapes. It has shown potential in breast cancer therapy through its strong cancer-preventive features, including the inhibition of cell proliferation, the induction of apoptosis, and the suppression of metastasis [232,233]. Notably, pterostilbene influences key molecular pathways such as PI3K/Akt and JAK/STAT, which are essential for the survival and growth of cancer cells [234]. In triple-negative breast cancer (TNBC) models, pterostilbene has been shown to impede tumor growth and metastasis by reducing the expression of oncogenic signaling molecules, including MMPs and VEGF [235,236]. Furthermore, its antioxidant properties alleviate oxidative stress in cancer cells, amplifying its therapeutic potential [192].

12. Discussion

Polyphenols constitute an assortment of naturally occurring nutrients common in plant-based foods and have attracted considerable interest due to their multifaceted bioactive properties [3,237]. These compounds are recognized for their antioxidant capabilities, anti-inflammatory effects, the modulation of estrogen activity, the inhibition of carcinogenic pathways, epigenetic modifications, and their roles in inhibiting angiogenesis and cellular migration [3,5,97,238,239,240,241]. Additionally, polyphenols function as immunomodulators and are readily accessible through dietary sources [12,98,242,243,244].
The potential of polyphenols as a primary preventive strategy for breast cancer is particularly noteworthy given their immunomodulatory effects. Breast cancer is regarded as one of the less immune-inducing cancer types; thus, enhancing the immune response within the tumor microenvironment may be advantageous for preventing disease onset [241,242]. Based on our comprehensive review of the existing literature, polyphenols may serve as promising therapeutic agents for the treatment of various subtypes of breast cancer.
Empirical studies indicate that polyphenols can modulate the activity of several immune cell types within the breast cancer tumor microenvironment [134,145]. For instance, epigallocatechin gallate (EGCG) has been demonstrated to activate natural killer (NK) cells and induce cytotoxic T cells, thereby promoting anti-tumor activity [109,111,112]. Similarly, pterostilbene has been shown to facilitate the transition of macrophages from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype [245]. This phenotypic shift is instrumental in mitigating chronic inflammation and fostering tissue repair, resulting in a more balanced immune response that is less susceptible to excessive inflammatory damage. Moreover, polyphenols generally downregulate regulatory T cell (Treg) expression within the tumor microenvironment, thereby counteracting immune suppression [3]. Cancer cells frequently evade immune surveillance through mechanisms that involve checkpoint inhibitors such as PD-1 [137]. However, polyphenols, including genistein (derived from soy) and resveratrol, have been found to modulate these immune checkpoints by inhibiting the PD-1/PD-L1 pathway [136,139,246]. This modulation enhances the immune system’s ability to recognize and effectively target tumor cells.
Current therapeutic modalities for breast cancer, including immunotherapy and chemotherapy, primarily focus on the treatment or reduction in cancerous cells. These approaches often lead to numerous undesirable outcomes, such as significant autoimmune reactions impacting the skin, liver, lungs, and other organs [137,247]. Additionally, the high costs associated with immunotherapy and the requirement for frequent clinical administration constrain its applicability as a widespread preventive measure. Notably, these conventional treatments do not emphasize cancer prevention at the initial stages. In contrast, polyphenols are naturally present in various dietary sources and can be consumed regularly without necessitating medical supervision [248]. The long-term intake of polyphenol-rich foods has been linked to minimal side effects, thus rendering them suitable for strategies aimed at the primary prevention of breast cancer [249].

13. Conclusions

To enhance the effectiveness of immunotherapy and polyphenols for specific breast cancer subtypes, particularly triple-negative breast cancer (TNBC), researchers are examining combination treatments that pair immunotherapy drugs with polyphenols or integrate various polyphenolic compounds. However, polyphenols can interact with certain medications and foods due to their distinct composition, potentially leading to adverse effects [134]. Moreover, their low absorption and fast elimination from the body present challenges for their use as therapeutic agents in clinical practice, underscoring the need for further research in this area [98,134]. Future investigations should continue to explore how polyphenols contribute to cancer prevention and their potential synergistic effects alongside other preventive strategies, such as lifestyle modifications and targeted therapies [125,145,146,147,148].

Author Contributions

E.E. conceived of the review paper with the guidance of T.O.T. The manuscript was drafted and edited by E.E., J.D. and T.O.T. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by a grant from the National Institute of Health (NCI R01CA178441).

Acknowledgments

We thank the members of Tollefsbol lab for their supportive comments.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AktProtein Kinase B
BaxBCL2-Associated X (Apoptosis Regulator)
BCBreast Cancer
Bcl-2B Cell Lymphoma Protein 2
Bcl-xlB Cell Lymphoma-Extra Large
BL1Basal-Like 1
BL2Basal-Like 2
CAFsCancer-Associated Fibroblasts
CAR-TChimeric Antigen Receptor T cells
CDK4Cyclin-Dependent Kinase 4
CLBCClaudin-Low Breast Cancer
CTLsCytotoxic T lymphocytes
CTLA4Cytotoxic T Lymphocyte-Associated Protein 4
DCDendritic Cells
DCISDuctal Carcinoma In Situ
ECMExtracellular Matrix
EGCGEpigallocatechin Gallate
EREstrogen Receptor
ER+Estrogen Receptor-Positive
EMTEpithelial–Mesenchymal Transition
HER2Human Epidermal Growth Factor 2
HER2+Human Epidermal Growth Factor 2-Positive
HRHormone Receptor
HR+Hormone Receptor-Positive
HIF-1αHypoxia-Inducible Factor 1-Alpha
JAK/STATJanus Kinase–Signal Transducer and Activator of Transcription
IDCInvasive Ductal Carcinoma
ILCInvasive Lobular Carcinoma
IMImmunomodulatory
LARLuminal Androgen Receptor
LCISLobular Carcinoma In Situ
MMesenchymal
M1M1-Type Macrophage
M2M2-Type Macrophage
MAPKMitogen-Activated Protein Kinase
MDSCsMyeloid-Derived Suppressor Cells
MEK5Mitogen-Activated Protein Kinase 5
MSLMesenchymal Stem-Like
MMPMatrix Metalloproteinase
mTORMammalian Target of Rapamycin
NFκBNuclear Factor Kappa B
NK CellsNatural Killer Cells
PD-1Programmed Cell Death Protein 1
PD-L1Programmed Death-Ligand 1
PI3KPhosphoinositide 3-Kinase
PRProgesterone Receptor
PR+Progesterone Receptor-Positive
Sp1Transcription Factor-Specific Protein 1
STAT3Signal Transducer and Activator of Transcription 3
TAMsTumor-Associated Macrophages
Th1Type 1 T Helper
Th2Type 2 T Helper
Th9Type 9 T Helper
Th17Type 17 T Helper
TMETumor Microenvironment
TNBCTriple-Negative Breast Cancer
TregsRegulatory T Cells
VEGFVascular Endothelial Growth Factor
ZAP-70Zeta Chain-Associated 70kDa Protein Receptor

References

  1. Wu, H.; Van Der Pol, W.J.; Dubois, L.G.; Morrow, C.D.; Tollefsbol, T.O. Dietary Supplementation of Inulin Contributes to the Prevention of Estrogen Receptor-Negative Mammary Cancer by Alteration of Gut Microbial Communities and Epigenetic Regulations. Int. J. Mol. Sci. 2023, 24, 9015. [Google Scholar] [CrossRef] [PubMed]
  2. Rahman, M.M.; Wu, H.; Tollefsbol, T.O. A novel combinatorial approach using sulforaphane- and withaferin A-rich extracts for prevention of estrogen receptor-negative breast cancer through epigenetic and gut microbial mechanisms. Sci. Rep. 2024, 14, 12091. [Google Scholar] [CrossRef] [PubMed]
  3. Shakoor, H.; Feehan, J.; Apostolopoulos, V.; Platat, C.; Al Dhaheri, A.S.; Ali, H.I.; Ismail, L.C.; Bosevski, M.; Stojanovska, L. Immunomodulatory Effects of Dietary Polyphenols. Nutrients 2021, 13, 728. [Google Scholar] [CrossRef] [PubMed]
  4. Duffy, S.W.; Tabár, L.; Yen, A.M.; Dean, P.B.; Smith, R.A.; Jonsson, H.; Törnberg, S.; Chen, S.L.; Chiu, S.Y.; Fann, J.C.; et al. Mammography screening reduces rates of advanced and fatal breast cancers: Results in 549,091 women. Cancer 2020, 126, 2971–2979. [Google Scholar] [CrossRef]
  5. Kim, H.S.; Quon, M.J.; Kim, J.A. New insights into the mechanisms of polyphenols beyond antioxidant properties; lessons from the green tea polyphenol, epigallocatechin 3-gallate. Redox Biol. 2014, 2, 187–195. [Google Scholar] [CrossRef]
  6. Murakami, A.; Ohnishi, K. Target molecules of food phytochemicals: Food science bound for the next dimension. Food Funct. 2012, 3, 462–476. [Google Scholar] [CrossRef]
  7. 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 effects against chronic diseases. Antioxid. Redox Signal. 2013, 18, 1818–1892. [Google Scholar] [CrossRef]
  8. Maleki Dana, P.; Sadoughi, F.; Asemi, Z.; Yousefi, B. The role of polyphenols in overcoming cancer drug resistance: A comprehensive review. Cell Mol. Biol. Lett. 2022, 27, 1. [Google Scholar] [CrossRef]
  9. Rajendran, P.; Abdelsalam, S.A.; Renu, K.; Veeraraghavan, V.; Ben Ammar, R.; Ahmed, E.A. Polyphenols as Potent Epigenetics Agents for Cancer. Int. J. Mol. Sci. 2022, 23, 11712. [Google Scholar] [CrossRef]
  10. Briguglio, G. Polyphenols in cancer prevention: New insights. Int. J. Funct. Nutr. 2020, 1, 9. [Google Scholar] [CrossRef]
  11. Kawabata, K.; Mukai, R.; Ishisaka, A. Quercetin and related polyphenols: New insights and implications for their bioactivity and bioavailability. Food Funct. 2015, 6, 1399–1417. [Google Scholar] [CrossRef] [PubMed]
  12. Calder, P.C.; Ahluwalia, N.; Brouns, F.; Buetler, T.; Clement, K.; Cunningham, K.; Esposito, K.; Jönsson, L.S.; Kolb, H.; Lansink, M.; et al. Dietary factors and low-grade inflammation in relation to overweight and obesity. Br. J. Nutr. 2011, 106 (Suppl. S3), S5–S78. [Google Scholar] [CrossRef]
  13. Farghadani, R.; Naidu, R. The anticancer mechanism of action of selected polyphenols in triple-negative breast cancer (TNBC). Biomed. Pharmacother. 2023, 165, 115170. [Google Scholar] [CrossRef] [PubMed]
  14. Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef] [PubMed]
  15. Lukasiewicz, S.; Czeczelewski, M.; Forma, A.; Baj, J.; Sitarz, R.; Stanisławek, A. Breast Cancer-Epidemiology, Risk Factors, Classification, Prognostic Markers, and Current Treatment Strategies—An Updated Review. Cancers 2021, 13, 4287. [Google Scholar] [CrossRef]
  16. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  17. Perou, C.M.; Sørlie, T.; Eisen, M.B.; van de Rijn, M.; Jeffrey, S.S.; Rees, C.A.; Pollack, J.R.; Ross, D.T.; Johnsen, H.; Akslen, L.A.; et al. Molecular portraits of human breast tumours. Nature 2000, 406, 747–752. [Google Scholar] [CrossRef]
  18. Prat, A.; Perou, C.M. Deconstructing the molecular portraits of breast cancer. Mol. Oncol. 2011, 5, 5–23. [Google Scholar] [CrossRef]
  19. Howlader, N.; Altekruse, S.F.; Li, C.I.; Chen, V.W.; Clarke, C.A.; Ries, L.A.; Cronin, K.A. US incidence of breast cancer subtypes defined by joint hormone receptor and HER2 status. J. Natl. Cancer Inst. 2014, 106, dju055. [Google Scholar] [CrossRef]
  20. Ignatiadis, M.; Sotiriou, C. Luminal breast cancer: From biology to treatment. Nat. Rev. Clin. Oncol. 2013, 10, 494–506. [Google Scholar] [CrossRef]
  21. Higgins, M.J.; Stearns, V. Understanding resistance to tamoxifen in hormone receptor-positive breast cancer. Clin. Chem. 2009, 55, 1453–1455. [Google Scholar] [CrossRef]
  22. Inic, Z.; Zegarac, M.; Inic, M.; Markovic, I.; Kozomara, Z.; Djurisic, I.; Inic, I.; Pupic, G.; Jancic, S. Difference between Luminal A and Luminal B Subtypes According to Ki-67, Tumor Size, and Progesterone Receptor Negativity Providing Prognostic Information. Clin. Med. Insights Oncol. 2014, 8, 107–111. [Google Scholar] [CrossRef]
  23. Kaneko, K.; Ishigami, S.; Kijima, Y.; Funasako, Y.; Hirata, M.; Okumura, H.; Shinchi, H.; Koriyama, C.; Ueno, S.; Yoshinaka, H.; et al. Clinical implication of HLA class I expression in breast cancer. BMC Cancer 2011, 11, 454. [Google Scholar] [CrossRef] [PubMed]
  24. Yersal, O.; Barutca, S. Biological subtypes of breast cancer: Prognostic and therapeutic implications. World J. Clin. Oncol. 2014, 5, 412–424. [Google Scholar] [CrossRef] [PubMed]
  25. Kavarthapu, R.; Anbazhagan, R.; Dufau, M.L. Crosstalk between PRLR and EGFR/HER2 Signaling Pathways in Breast Cancer. Cancers 2021, 13, 4685. [Google Scholar] [CrossRef] [PubMed]
  26. Harbeck, N.; Penault-Llorca, F.; Cortes, J.; Gnant, M.; Houssami, N.; Poortmans, P.; Ruddy, K.; Tsang, J.; Cardoso, F. Breast cancer. Nat. Rev. Dis. Primers 2019, 5, 66. [Google Scholar] [CrossRef]
  27. Fragomeni, S.M.; Sciallis, A.; Jeruss, J.S. Molecular Subtypes and Local-Regional Control of Breast Cancer. Surg. Oncol. Clin. N. Am. 2018, 27, 95–120. [Google Scholar] [CrossRef]
  28. Shaath, H.; Elango, R.; Alajez, N.M. Molecular Classification of Breast Cancer Utilizing Long Non-Coding RNA (lncRNA) Transcriptomes Identifies Novel Diagnostic lncRNA Panel for Triple-Negative Breast Cancer. Cancers 2021, 13, 5350. [Google Scholar] [CrossRef]
  29. Plasilova, M.L.; Hayse, B.; Killelea, B.K.; Horowitz, N.R.; Chagpar, A.B.; Lannin, D.R. Features of triple-negative breast cancer: Analysis of 38,813 cases from the national cancer database. Medicine 2016, 95, e4614. [Google Scholar] [CrossRef]
  30. Orrantia-Borunda, E.; Anchondo-Nuñez, P.; Acuña-Aguilar, L.E.; Gómez-Valles, F.O.; Ramírez-Valdespino, C.A. Subtypes of Breast Cancer. In Breast Cancer; Mayrovitz, H.N., Ed.; Exon publications: Brisbane, Australia, 2022. [Google Scholar]
  31. 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. [Google Scholar] [CrossRef]
  32. Wang, D.Y.; Jiang, Z.; Ben-David, Y.; Woodgett, J.R.; Zacksenhaus, E. Molecular stratification within triple-negative breast cancer subtypes. Sci. Rep. 2019, 9, 19107. [Google Scholar] [CrossRef] [PubMed]
  33. Yin, L.; Duan, J.J.; Bian, X.W.; Yu, S.C. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 2020, 22, 61. [Google Scholar] [CrossRef] [PubMed]
  34. Pan, C.; Xu, A.; Ma, X.; Yao, Y.; Zhao, Y.; Wang, C.; Chen, C. Research progress of Claudin-low breast cancer. Front. Oncol. 2023, 13, 1226118. [Google Scholar] [CrossRef] [PubMed]
  35. Weigelt, B.; Geyer, F.C.; Reis-Filho, J.S. Histological types of breast cancer: How special are they? Mol. Oncol. 2010, 4, 192–208. [Google Scholar] [CrossRef]
  36. Russnes, H.G.; Lingjærde, O.C.; Børresen-Dale, A.L.; Caldas, C. Breast Cancer Molecular Stratification: From Intrinsic Subtypes to Integrative Clusters. Am. J. Pathol. 2017, 187, 2152–2162. [Google Scholar] [CrossRef]
  37. Ruffell, B.; Au, A.; Rugo, H.S.; Esserman, L.J.; Hwang, E.S.; Coussens, L.M. Leukocyte composition of human breast cancer. Proc. Natl. Acad. Sci. USA 2012, 109, 2796–2801. [Google Scholar] [CrossRef]
  38. Hanahan, D.; Coussens, L.M. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef]
  39. Comba, A.; Faisal, S.M.; Varela, M.L.; Hollon, T.; Al-Holou, W.N.; Umemura, Y.; Nunez, F.J.; Motsch, S.; Castro, M.G.; Lowenstein, P.R. Uncovering Spatiotemporal Heterogeneity of High-Grade Gliomas: From Disease Biology to Therapeutic Implications. Front. Oncol. 2021, 11, 703764. [Google Scholar] [CrossRef]
  40. Goff, S.L.; Danforth, D.N. The Role of Immune Cells in Breast Tissue and Immunotherapy for the Treatment of Breast Cancer. Clin. Breast Cancer 2021, 21, e63–e73. [Google Scholar] [CrossRef]
  41. Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef]
  42. Polyak, K. Heterogeneity in breast cancer. J. Clin. Investig. 2011, 121, 3786–3788. [Google Scholar] [CrossRef] [PubMed]
  43. Guc, E.; Pollard, J.W. Redefining macrophage and neutrophil biology in the metastatic cascade. Immunity 2021, 54, 885–902. [Google Scholar] [CrossRef] [PubMed]
  44. DeNardo, D.G.; Ruffell, B. Macrophages as regulators of tumour immunity and immunotherapy. Nat. Rev. Immunol. 2019, 19, 369–382. [Google Scholar] [CrossRef] [PubMed]
  45. de Visser, K.E.; Joyce, J.A. The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell 2023, 41, 374–403. [Google Scholar] [CrossRef]
  46. Jaillon, S.; Ponzetta, A.; Di Mitri, D.; Santoni, A.; Bonecchi, R.; Mantovani, A. Neutrophil diversity and plasticity in tumour progression and therapy. Nat. Rev. Cancer 2020, 20, 485–503. [Google Scholar] [CrossRef]
  47. Wu, L.; Saxena, S.; Singh, R.K. Neutrophils in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1224, 1–20. [Google Scholar]
  48. Amer, H.T.; Stein, U.; El Tayebi, H.M. The Monocyte, a Maestro in the Tumor Microenvironment (TME) of Breast Cancer. Cancers 2022, 14, 5460. [Google Scholar] [CrossRef]
  49. Olingy, C.E.; Dinh, H.Q.; Hedrick, C.C. Monocyte heterogeneity and functions in cancer. J. Leukoc. Biol. 2019, 106, 309–322. [Google Scholar] [CrossRef]
  50. Gerhard, G.M.; Bill, R.; Messemaker, M.; Klein, A.M.; Pittet, M.J. Tumor-infiltrating dendritic cell states are conserved across solid human cancers. J. Exp. Med. 2021, 218, e20200264. [Google Scholar] [CrossRef]
  51. Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [Google Scholar] [CrossRef]
  52. Broz, M.L.; Binnewies, M.; Boldajipour, B.; Nelson, A.E.; Pollack, J.L.; Erle, D.J.; Barczak, A.; Rosenblum, M.D.; Daud, A.; Barber, D.L.; et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 2014, 26, 638–652. [Google Scholar] [CrossRef] [PubMed]
  53. Engelhardt, J.J.; Boldajipour, B.; Beemiller, P.; Pandurangi, P.; Sorensen, C.; Werb, Z.; Egeblad, M.; Krummel, M.F. Marginating dendritic cells of the tumor microenvironment cross-present tumor antigens and stably engage tumor-specific T cells. Cancer Cell 2012, 21, 402–417. [Google Scholar] [CrossRef] [PubMed]
  54. Gupta, Y.H.; Khanom, A.; Acton, S.E. Control of Dendritic Cell Function Within the Tumour Microenvironment. Front. Immunol. 2022, 13, 733800. [Google Scholar] [CrossRef] [PubMed]
  55. Majorini, M.T.; Colombo, M.P.; Lecis, D. Few, but Efficient: The Role of Mast Cells in Breast Cancer and Other Solid Tumors. Cancer Res. 2022, 82, 1439–1447. [Google Scholar] [CrossRef]
  56. Grisaru-Tal, S.; Rothenberg, M.E.; Munitz, A. Eosinophil-lymphocyte interactions in the tumor microenvironment and cancer immunotherapy. Nat. Immunol. 2022, 23, 1309–1316. [Google Scholar] [CrossRef]
  57. Blomberg, O.S.; Spagnuolo, L.; Garner, H.; Voorwerk, L.; Isaeva, O.I.; van Dyk, E.; Bakker, N.; Chalabi, M.; Klaver, C.; Duijst, M.; et al. IL-5-producing CD4(+) T cells and eosinophils cooperate to enhance response to immune checkpoint blockade in breast cancer. Cancer Cell 2023, 41, 106–123.e10. [Google Scholar] [CrossRef]
  58. Ghaffari, S.; Rezaei, N. Eosinophils in the tumor microenvironment: Implications for cancer immunotherapy. J. Transl. Med. 2023, 21, 551. [Google Scholar] [CrossRef]
  59. Gambardella, A.R.; Antonucci, C.; Zanetti, C.; Noto, F.; Andreone, S.; Vacca, D.; Pellerito, V.; Sicignano, C.; Parrottino, G.; Tirelli, V.; et al. IL-33 stimulates the anticancer activities of eosinophils through extracellular vesicle-driven reprogramming of tumor cells. J. Exp. Clin. Cancer Res. 2024, 43, 209. [Google Scholar] [CrossRef] [PubMed]
  60. Veglia, F.; Sanseviero, E.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol. 2021, 21, 485–498. [Google Scholar] [CrossRef]
  61. Nepal, M.R.; Shah, S.; Kang, K.T. Dual roles of myeloid-derived suppressor cells in various diseases: A review. Arch. Pharm. Res. 2024, 47, 597–616. [Google Scholar] [CrossRef]
  62. Zhou, Y.; Cheng, L.; Liu, L.; Li, X. NK cells are never alone: Crosstalk and communication in tumour microenvironments. Mol. Cancer 2023, 22, 34. [Google Scholar] [CrossRef] [PubMed]
  63. Chan, I.S.; Ewald, A.J. The changing role of natural killer cells in cancer metastasis. J. Clin. Investig. 2022, 132, e143762. [Google Scholar] [CrossRef] [PubMed]
  64. Yuen, G.J.; Demissie, E.; Pillai, S. B lymphocytes and cancer: A love-hate relationship. Trends Cancer 2016, 2, 747–757. [Google Scholar] [CrossRef]
  65. Laumont, C.M.; Banville, A.C.; Gilardi, M.; Hollern, D.P.; Nelson, B.H. Tumour-infiltrating B cells: Immunological mechanisms, clinical impact and therapeutic opportunities. Nat. Rev. Cancer 2022, 22, 414–430. [Google Scholar] [CrossRef]
  66. Zhang, E.; Ding, C.; Li, S.; Zhou, X.; Aikemu, B.; Fan, X.; Sun, J.; Zheng, M.; Yang, X. Roles and mechanisms of tumour-infiltrating B cells in human cancer: A new force in immunotherapy. Biomark. Res. 2023, 11, 28. [Google Scholar] [CrossRef]
  67. Philip, M.; Schietinger, A. CD8(+) T cell differentiation and dysfunction in cancer. Nat. Rev. Immunol. 2022, 22, 209–223. [Google Scholar] [CrossRef] [PubMed]
  68. van der Leun, A.M.; Thommen, D.S.; Schumacher, T.N. CD8(+) T cell states in human cancer: Insights from single-cell analysis. Nat. Rev. Cancer 2020, 20, 218–232. [Google Scholar] [CrossRef]
  69. DeNardo, D.G.; Barreto, J.B.; Andreu, P.; Vasquez, L.; Tawfik, D.; Kolhatkar, N.; Coussens, L.M. CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell 2009, 16, 91–102. [Google Scholar] [CrossRef]
  70. Borst, J.; Ahrends, T.; Bąbała, N.; Melief, C.J.M.; Kastenmüller, W. CD4(+) T cell help in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2018, 18, 635–647. [Google Scholar] [CrossRef]
  71. Togashi, Y.; Shitara, K.; Nishikawa, H. Regulatory T cells in cancer immunosuppression—Implications for anticancer therapy. Nat. Rev. Clin. Oncol. 2019, 16, 356–371. [Google Scholar] [CrossRef]
  72. Wang, J.; Wu, S.G. Breast Cancer: An Overview of Current Therapeutic Strategies, Challenge, and Perspectives. Breast Cancer 2023, 15, 721–730. [Google Scholar] [CrossRef] [PubMed]
  73. Wilson, R.E. Surgical management of locally advanced and recurrent breast cancer. Cancer 1984, 53 (Suppl. S3), 752–757. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, Y.; Shen, J.; Gu, P.; Wang, Z. Recent advances progress in radiotherapy for breast cancer after breast-conserving surgery: A review. Front. Oncol. 2023, 13, 1195266. [Google Scholar] [CrossRef] [PubMed]
  75. Kolarova, I.; Melichar, B.; Sirák, I.; Vaňásek, J.; Petera, J.; Horáčková, K.; Pohanková, D.; Ďatelinka, F.; Šinkorová, Z.; Vošmik, M. The Role of Adjuvant Radiotherapy in the Treatment of Breast Cancer. Curr. Oncol. 2024, 31, 1207–1220. [Google Scholar] [CrossRef] [PubMed]
  76. Anampa, J.; Makower, D.; Sparano, J.A. Progress in adjuvant chemotherapy for breast cancer: An overview. BMC Med. 2015, 13, 195. [Google Scholar] [CrossRef]
  77. Mano, M.S.; Awada, A. Primary chemotherapy for breast cancer: The evidence and the future. Ann. Oncol. 2004, 15, 1161–1171. [Google Scholar] [CrossRef]
  78. Puhalla, S.; Bhattacharya, S.; Davidson, N.E. Hormonal therapy in breast cancer: A model disease for the personalization of cancer care. Mol. Oncol. 2012, 6, 222–236. [Google Scholar] [CrossRef]
  79. Ramadan, W.S.; Alseksek, R.K.; Mouffak, S.; Talaat, I.M.; Saber-Ayad, M.M.; Menon, V.; Ilce, B.Y.; El-Awady, R. Impact of HDAC6-mediated progesterone receptor expression on the response of breast cancer cells to hormonal therapy. Eur. J. Pharmacol. 2024, 983, 177001. [Google Scholar] [CrossRef]
  80. Du, L.; Yau, C.; Brown-Swigart, L.; Gould, R.; Krings, G.; Hirst, G.L.; Bedrosian, I.; Layman, R.M.; Carter, J.M.; Klein, M.; et al. Predicted sensitivity to endocrine therapy for stage II-III hormone receptor-positive and HER2-negative (HR+/HER2-) breast cancer before chemo-endocrine therapy. Ann. Oncol. 2021, 32, 642–651. [Google Scholar] [CrossRef]
  81. Mancuso, M.R.; Massarweh, S.A. Endocrine therapy and strategies to overcome therapeutic resistance in breast cancer. Curr. Probl. Cancer 2016, 40, 95–105. [Google Scholar] [CrossRef]
  82. Adams, S.; Gatti-Mays, M.E.; Kalinsky, K.; Korde, L.A.; Sharon, E.; Amiri-Kordestani, L.; Bear, H.; McArthur, H.L.; Frank, E.; Perlmutter, J.; et al. Current Landscape of Immunotherapy in Breast Cancer: A Review. JAMA Oncol. 2019, 5, 1205–1214. [Google Scholar] [CrossRef] [PubMed]
  83. Barzaman, K.; Moradi-Kalbolandi, S.; Hosseinzadeh, A.; Kazemi, M.H.; Khorramdelazad, H.; Safari, E.; Farahmand, L. Breast cancer immunotherapy: Current and novel approaches. Int. Immunopharmacol. 2021, 98, 107886. [Google Scholar]
  84. Vasileiou, M. Current Advancements and Future Perspectives of Immunotherapy in Breast Cancer Treatment. Immuno 2023, 3, 195–216. [Google Scholar] [CrossRef]
  85. Vu, T.; Claret, F.X. Trastuzumab: Updated mechanisms of action and resistance in breast cancer. Front. Oncol. 2012, 2, 62. [Google Scholar] [CrossRef] [PubMed]
  86. Capelan, M.; Pugliano, L.; De Azambuja, E.; Bozovic, I.; Saini, K.S.; Sotiriou, C.; Loi, S.; Piccart-Gebhart, M.J. Pertuzumab: New hope for patients with HER2-positive breast cancer. Ann. Oncol. 2013, 24, 273–282. [Google Scholar] [CrossRef]
  87. June, C.H.; Riddell, S.R.; Schumacher, T.N. Adoptive cellular therapy: A race to the finish line. Sci. Transl. Med. 2015, 7, 280ps7. [Google Scholar] [CrossRef]
  88. Alaluf, E.; Shalamov, M.M.; Sonnenblick, A. Update on current and new potential immunotherapies in breast cancer, from bench to bedside. Front. Immunol. 2024, 15, 1287824. [Google Scholar] [CrossRef]
  89. Semiglazov, V.; Tseluiko, A.; Kudaybergenova, A.; Artemyeva, A.; Krivorotko, P.; Donskih, R. Immunology and immunotherapy in breast cancer. Cancer Biol. Med. 2022, 19, 609–618. [Google Scholar] [CrossRef]
  90. Tan, S.; Li, D.; Zhu, X. Cancer immunotherapy: Pros, cons and beyond. Biomed. Pharmacother. 2020, 124, 109821. [Google Scholar] [CrossRef]
  91. Debien, V.; De Caluwé, A.; Wang, X.; Piccart-Gebhart, M.; Tuohy, V.K.; Romano, E.; Buisseret, L. Immunotherapy in breast cancer: An overview of current strategies and perspectives. NPJ Breast Cancer 2023, 9, 7. [Google Scholar] [CrossRef]
  92. Li, S.; Wu, H.; Chen, M.; Tollefsbol, T.O. Paternal Combined Botanicals Contribute to the Prevention of Estrogen Receptor-Negative Mammary Cancer in Transgenic Mice. J. Nutr. 2023, 153, 1959–1973. [Google Scholar] [CrossRef]
  93. Putti, T.C.; El-Rehim, D.M.; Rakha, E.A.; Paish, C.E.; Lee, A.H.; Pinder, S.E.; Ellis, I.O. Estrogen receptor-negative breast carcinomas: A review of morphology and immunophenotypical analysis. Mod. Pathol. 2005, 18, 26–35. [Google Scholar] [CrossRef]
  94. Shaikh, A.A.; Braakhuis, A.J.; Bishop, K.S. The Mediterranean Diet and Breast Cancer: A Personalised Approach. Healthcare 2019, 7, 104. [Google Scholar] [CrossRef] [PubMed]
  95. Sharma, M.; Arora, I.; Stoll, M.L.; Li, Y.; Morrow, C.D.; Barnes, S.; Berryhill, T.F.; Li, S.; Tollefsbol, T.O. Nutritional combinatorial impact on the gut microbiota and plasma short-chain fatty acids levels in the prevention of mammary cancer in Her2/neu estrogen receptor-negative transgenic mice. PLoS ONE 2020, 15, e0234893. [Google Scholar] [CrossRef] [PubMed]
  96. Milani, A.; Basirnejad, M.; Shahbazi, S.; Bolhassani, A. Carotenoids: Biochemistry, pharmacology and treatment. Br. J. Pharmacol. 2017, 174, 1290–1324. [Google Scholar] [CrossRef] [PubMed]
  97. Hollman, P.C.; Geelen, A.; Kromhout, D. Dietary flavonol intake may lower stroke risk in men and women. J. Nutr. 2010, 140, 600–604. [Google Scholar] [CrossRef]
  98. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef]
  99. Scalbert, A.; Johnson, I.T.; Saltmarsh, M. Polyphenols: Antioxidants and beyond. Am. J. Clin. Nutr. 2005, 81 (Suppl. S1), 215S–217S. [Google Scholar] [CrossRef]
  100. 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. [Google Scholar] [CrossRef]
  101. Serafini, M.; Peluso, I.; Raguzzini, A. Flavonoids as anti-inflammatory agents. Proc. Nutr. Soc. 2010, 69, 273–278. [Google Scholar] [CrossRef]
  102. Williamson, G.; Kay, C.D.; Crozier, A. The Bioavailability, Transport, and Bioactivity of Dietary Flavonoids: A Review from a Historical Perspective. Compr. Rev. Food Sci. Food Saf. 2018, 17, 1054–1112. [Google Scholar] [CrossRef]
  103. Huang, W.Y.; Cai, Y.Z.; Zhang, Y. Natural phenolic compounds from medicinal herbs and dietary plants: Potential use for cancer prevention. Nutr. Cancer 2010, 62, 1–20. [Google Scholar] [CrossRef] [PubMed]
  104. Ganguly, S.; Arora, I.; Tollefsbol, T.O. Impact of Stilbenes as Epigenetic Modulators of Breast Cancer Risk and Associated Biomarkers. Int. J. Mol. Sci. 2021, 22, 10033. [Google Scholar] [CrossRef] [PubMed]
  105. Gonzalez, R.; Ballester, I.; López-Posadas, R.; Suárez, M.D.; Zarzuelo, A.; Martínez-Augustin, O.; Sánchez de Medina, F. Effects of flavonoids and other polyphenols on inflammation. Crit. Rev. Food Sci. Nutr. 2011, 51, 331–362. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, Z.; Fei, Y.J.; Cao, X.H.; Xu, D.; Tang, W.J.; Yang, K.; Xu, W.X.; Tang, J.H. Lignans intake and enterolactone concentration and prognosis of breast cancer: A systematic review and meta-analysis. J. Cancer 2021, 12, 2787–2796. [Google Scholar] [CrossRef]
  107. Mileo, A.M.; Nistico, P.; Miccadei, S. Polyphenols: Immunomodulatory and Therapeutic Implication in Colorectal Cancer. Front. Immunol. 2019, 10, 729. [Google Scholar] [CrossRef]
  108. Szliszka, E.; Krol, W. The role of dietary polyphenols in tumor necrosis factor-related apoptosis inducing ligand (TRAIL)-induced apoptosis for cancer chemoprevention. Eur. J. Cancer Prev. 2011, 20, 63–69. [Google Scholar] [CrossRef]
  109. Ding, S.; Jiang, H.; Fang, J. Regulation of Immune Function by Polyphenols. J. Immunol. Res. 2018, 2018, 1264074. [Google Scholar] [CrossRef]
  110. Yang, C.C.; Wu, C.J.; Chien, C.Y.; Chien, C.T. Green Tea Polyphenol Catechins Inhibit Coronavirus Replication and Potentiate the Adaptive Immunity and Autophagy-Dependent Protective Mechanism to Improve Acute Lung Injury in Mice. Antioxidants 2021, 10, 928. [Google Scholar] [CrossRef]
  111. Ferreira, C.; Vieira, P.; Sá, H.; Malva, J.; Castelo-Branco, M.; Reis, F.; Viana, S. Polyphenols: Immunonutrients tipping the balance of immunometabolism in chronic diseases. Front. Immunol. 2024, 15, 1360065. [Google Scholar] [CrossRef]
  112. Huang, X.; Wang, Y.; Yang, W.; Dong, J.; Li, L. Regulation of dietary polyphenols on cancer cell pyroptosis and the tumor immune microenvironment. Front. Nutr. 2022, 9, 974896. [Google Scholar] [CrossRef]
  113. 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. [Google Scholar] [CrossRef] [PubMed]
  114. Ros, M.; Riesco-Llach, G.; Polonio-Alcalá, E.; Morla-Barcelo, P.M.; Ruiz-Martínez, S.; Feliu, L.; Planas, M.; Puig, T. Inhibition of Cancer Stem-like Cells by Curcumin and Other Polyphenol Derivatives in MDA-MB-231 TNBC Cells. Int. J. Mol. Sci. 2024, 25, 7446. [Google Scholar] [CrossRef] [PubMed]
  115. Milano, F.; Mari, L.; van de Luijtgaarden, W.; Parikh, K.; Calpe, S.; Krishnadath, K.K. Nano-curcumin inhibits proliferation of esophageal adenocarcinoma cells and enhances the T cell mediated immune response. Front. Oncol. 2013, 3, 137. [Google Scholar] [CrossRef]
  116. Hong, E.H.; Heo, E.Y.; Song, J.H.; Kwon, B.E.; Lee, J.Y.; Park, Y.; Kim, J.; Chang, S.Y.; Chin, Y.W.; Jeon, S.M.; et al. Trans-scirpusin A showed antitumor effects via autophagy activation and apoptosis induction of colorectal cancer cells. Oncotarget 2017, 8, 41401–41411. [Google Scholar] [CrossRef]
  117. Hayakawa, T.; Yaguchi, T.; Kawakami, Y. Enhanced anti-tumor effects of the PD-1 blockade combined with a highly absorptive form of curcumin targeting STAT3. Cancer Sci. 2020, 111, 4326–4335. [Google Scholar] [CrossRef]
  118. Shafabakhsh, R.; Pourhanifeh, M.H.; Mirzaei, H.R.; Sahebkar, A.; Asemi, Z.; Mirzaei, H. Targeting regulatory T cells by curcumin: A potential for cancer immunotherapy. Pharmacol. Res. 2019, 147, 104353. [Google Scholar] [CrossRef] [PubMed]
  119. Yang, M.; Li, Z.; Tao, J.; Hu, H.; Li, Z.; Zhang, Z.; Cheng, F.; Sun, Y.; Zhang, Y.; Yang, J.; et al. Resveratrol induces PD-L1 expression through snail-driven activation of Wnt pathway in lung cancer cells. J. Cancer Res. Clin. Oncol. 2021, 147, 1101–1113. [Google Scholar] [CrossRef]
  120. Zhang, Q.; Huang, H.; Zheng, F.; Liu, H.; Qiu, F.; Chen, Y.; Liang, C.L.; Dai, Z. Resveratrol exerts antitumor effects by downregulating CD8(+)CD122(+) Tregs in murine hepatocellular carcinoma. Oncoimmunology 2020, 9, 1829346. [Google Scholar] [CrossRef]
  121. Malaguarnera, L. Influence of Resveratrol on the Immune Response. Nutrients 2019, 11, 946. [Google Scholar] [CrossRef]
  122. Ezzati, M.; Yousefi, B.; Velaei, K.; Safa, A. A review on anti-cancer properties of Quercetin in breast cancer. Life Sci. 2020, 248, 117463. [Google Scholar] [CrossRef]
  123. Sato, M.; Miyazaki, T.; Kambe, F.; Maeda, K.; Seo, H. Quercetin, a bioflavonoid, inhibits the induction of interleukin 8 and monocyte chemoattractant protein-1 expression by tumor necrosis factor-alpha in cultured human synovial cells. J. Rheumatol. 1997, 24, 1680–1684. [Google Scholar] [PubMed]
  124. Li, Y.; Yao, J.; Han, C.; Yang, J.; Chaudhry, M.T.; Wang, S.; Liu, H.; Yin, Y. Quercetin, Inflammation and Immunity. Nutrients 2016, 8, 167. [Google Scholar] [CrossRef] [PubMed]
  125. Kotecha, R.; Takami, A.; Espinoza, J.L. Dietary phytochemicals and cancer chemoprevention: A review of the clinical evidence. Oncotarget 2016, 7, 52517–52529. [Google Scholar] [CrossRef]
  126. Garcia-Lafuente, A.; Guillamón, E.; Villares, A.; Rostagno, M.A.; Martínez, J.A. Flavonoids as anti-inflammatory agents: Implications in cancer and cardiovascular disease. Inflamm. Res. 2009, 58, 537–552. [Google Scholar] [CrossRef] [PubMed]
  127. Falchetti, R.; Fuggetta, M.P.; Lanzilli, G.; Tricarico, M.; Ravagnan, G. Effects of resveratrol on human immune cell function. Life Sci. 2001, 70, 81–96. [Google Scholar] [CrossRef]
  128. Li, D.; Cao, D.; Sun, Y.; Cui, Y.; Zhang, Y.; Jiang, J.; Cao, X. The roles of epigallocatechin gallate in the tumor microenvironment, metabolic reprogramming, and immunotherapy. Front. Immunol. 2024, 15, 1331641. [Google Scholar] [CrossRef]
  129. Pan, P.; Huang, Y.W.; Oshima, K.; Yearsley, M.; Zhang, J.; Arnold, M.; Yu, J.; Wang, L.S. The immunomodulatory potential of natural compounds in tumor-bearing mice and humans. Crit. Rev. Food Sci. Nutr. 2019, 59, 992–1007. [Google Scholar] [CrossRef]
  130. Jang, J.Y.; Lee, J.K.; Jeon, Y.K.; Kim, C.W. Exosome derived from epigallocatechin gallate treated breast cancer cells suppresses tumor growth by inhibiting tumor-associated macrophage infiltration and M2 polarization. BMC Cancer 2013, 13, 421. [Google Scholar] [CrossRef]
  131. Singh, N.; Baby, D.; Rajguru, J.P.; Patil, P.B.; Thakkannavar, S.S.; Pujari, V.B. Inflammation and cancer. Ann. Afr. Med. 2019, 18, 121–126. [Google Scholar] [CrossRef]
  132. Sahebkar, A.; Cicero, A.F.G.; Simental-Mendía, L.E.; Aggarwal, B.B.; Gupta, S.C. Curcumin downregulates human tumor necrosis factor-alpha levels: A systematic review and meta-analysis ofrandomized controlled trials. Pharmacol. Res. 2016, 107, 234–242. [Google Scholar] [CrossRef]
  133. Cheng, S.C.; Huang, W.C.; S Pang, J.H.; Wu, Y.H.; Cheng, C.Y. Quercetin Inhibits the Production of IL-1beta-Induced Inflammatory Cytokines and Chemokines in ARPE-19 Cells via the MAPK and NF-kappaB Signaling Pathways. Int. J. Mol. Sci. 2019, 20, 2957. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, Q.; Yang, B.; Wang, N.; Gu, J. Tumor immunomodulatory effects of polyphenols. Front. Immunol. 2022, 13, 1041138. [Google Scholar] [CrossRef] [PubMed]
  135. Singh, A.; Yau, Y.F.; Leung, K.S.; El-Nezami, H.; Lee, J.C. Interaction of Polyphenols as Antioxidant and Anti-Inflammatory Compounds in Brain-Liver-Gut Axis. Antioxidants 2020, 9, 669. [Google Scholar] [CrossRef]
  136. Messeha, S.S.; Zarmouh, N.O.; Soliman, K.F.A. Polyphenols Modulating Effects of PD-L1/PD-1 Checkpoint and EMT-Mediated PD-L1 Overexpression in Breast Cancer. Nutrients 2021, 13, 1718. [Google Scholar] [CrossRef]
  137. Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018, 8, 1069–1086. [Google Scholar] [CrossRef]
  138. Frebel, H.; Nindl, V.; Schuepbach, R.A.; Braunschweiler, T.; Richter, K.; Vogel, J.; Wagner, C.A.; Loffing-Cueni, D.; Kurrer, M.; Ludewig, B.; et al. Programmed death 1 protects from fatal circulatory failure during systemic virus infection of mice. J. Exp. Med. 2012, 209, 2485–2499. [Google Scholar] [CrossRef]
  139. Delmas, D.; Hermetet, F.; Aires, V. PD-1/PD-L1 Checkpoints and Resveratrol: A Controversial New Way for a Therapeutic Strategy. Cancers 2021, 13, 4509. [Google Scholar] [CrossRef] [PubMed]
  140. Sun, C.; Mezzadra, R.; Schumacher, T.N. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018, 48, 434–452. [Google Scholar] [CrossRef]
  141. Lin, D.Y.; Tanaka, Y.; Iwasaki, M.; Gittis, A.G.; Su, H.P.; Mikami, B.; Okazaki, T.; Honjo, T.; Minato, N.; Garboczi, D.N. The PD-1/PD-L1 complex resembles the antigen-binding Fv domains of antibodies and T cell receptors. Proc. Natl. Acad. Sci. USA 2008, 105, 3011–3016. [Google Scholar] [CrossRef]
  142. Okazaki, T.; Chikuma, S.; Iwai, Y.; Fagarasan, S.; Honjo, T. A rheostat for immune responses: The unique properties of PD-1 and their advantages for clinical application. Nat. Immunol. 2013, 14, 1212–1218. [Google Scholar] [CrossRef]
  143. Juneja, V.R.; McGuire, K.A.; Manguso, R.T.; LaFleur, M.W.; Collins, N.; Haining, W.N.; Freeman, G.J.; Sharpe, A.H. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J. Exp. Med. 2017, 214, 895–904. [Google Scholar] [CrossRef] [PubMed]
  144. Khalil, A.; Al-Massarani, G.; Aljapawe, A.; Ekhtiar, A.; Bakir, M.A. Resveratrol Modulates the Inflammatory Profile of Immune Responses and Circulating Endothelial Cells’ (CECs’) Population During Acute Whole Body Gamma Irradiation. Front. Pharmacol. 2020, 11, 528400. [Google Scholar] [CrossRef] [PubMed]
  145. Surh, Y.J. Cancer chemoprevention with dietary phytochemicals. Nat. Rev. Cancer 2003, 3, 768–780. [Google Scholar] [CrossRef] [PubMed]
  146. Aggarwal, B.B.; Shishodia, S. Molecular targets of dietary agents for prevention and therapy of cancer. Biochem. Pharmacol. 2006, 71, 1397–1421. [Google Scholar] [CrossRef] [PubMed]
  147. Lee, K.W.; Bode, A.M.; Dong, Z. Molecular targets of phytochemicals for cancer prevention. Nat. Rev. Cancer 2011, 11, 211–218. [Google Scholar] [CrossRef]
  148. Lee, K.W.; Lee, H.J. The roles of polyphenols in cancer chemoprevention. Biofactors 2006, 26, 105–121. [Google Scholar] [CrossRef]
  149. Almatroodi, S.A.; A Alsahli, M.; S M Aljohani, A.; Alhumaydhi, F.A.; Babiker, A.Y.; Khan, A.A.; Rahmani, A.H. Potential Therapeutic Targets of Resveratrol, a Plant Polyphenol, and Its Role in the Therapy of Various Types of Cancer. Molecules 2022, 27, 2665. [Google Scholar] [CrossRef]
  150. Vladu, A.F.; Ficai, D.; Ene, A.G.; Ficai, A. Combination Therapy Using Polyphenols: An Efficient Way to Improve Antitumoral Activity and Reduce Resistance. Int. J. Mol. Sci. 2022, 23, 2665. [Google Scholar] [CrossRef]
  151. Patra, S.; Pradhan, B.; Nayak, R.; Behera, C.; Das, S.; Patra, S.K.; Efferth, T.; Jena, M.; Bhutia, S.K. Dietary polyphenols in chemoprevention and synergistic effect in cancer: Clinical evidences and molecular mechanisms of action. Phytomedicine 2021, 90, 153554. [Google Scholar] [CrossRef]
  152. Cuevas, A.; Saavedra, N.; Salazar, L.A.; Abdalla, D.S. Modulation of immune function by polyphenols: Possible contribution of epigenetic factors. Nutrients 2013, 5, 2314–2332. [Google Scholar] [CrossRef]
  153. Ciz, M.; Dvořáková, A.; Skočková, V.; Kubala, L. The Role of Dietary Phenolic Compounds in Epigenetic Modulation Involved in Inflammatory Processes. Antioxidants 2020, 9, 691. [Google Scholar] [CrossRef] [PubMed]
  154. He, L.; Hannon, G.J. MicroRNAs: Small RNAs with a big role in gene regulation. Nat. Rev. Genet. 2004, 5, 522–531. [Google Scholar] [CrossRef] [PubMed]
  155. Jaenisch, R.; Bird, A. Epigenetic regulation of gene expression: How the genome integrates intrinsic and environmental signals. Nat. Genet. 2003, 33, 245–254. [Google Scholar] [CrossRef]
  156. Borsoi, F.T.; Neri-Numa, I.A.; de Oliveira, W.Q.; de Araújo, F.F.; Pastore, G.M. Dietary polyphenols and their relationship to the modulation of non-communicable chronic diseases and epigenetic mechanisms: A mini-review. Food Chem. 2023, 6, 100155. [Google Scholar] [CrossRef]
  157. Yaskolka Meir, A.; Keller, M.; Hoffmann, A.; Rinott, E.; Tsaban, G.; Kaplan, A.; Zelicha, H.; Hagemann, T.; Ceglarek, U.; Isermann, B.; et al. The effect of polyphenols on DNA methylation-assessed biological age attenuation: The DIRECT PLUS randomized controlled trial. BMC Med. 2023, 21, 364. [Google Scholar] [CrossRef]
  158. Pan, M.H.; Lai, C.S.; Wu, J.C.; Ho, C.T. Epigenetic and disease targets by polyphenols. Curr. Pharm. Des. 2013, 19, 6156–6185. [Google Scholar] [CrossRef] [PubMed]
  159. Li, Y.; Yuan, Y.Y.; Meeran, S.M.; Tollefsbol, T.O. Synergistic epigenetic reactivation of estrogen receptor-alpha (ERalpha) by combined green tea polyphenol and histone deacetylase inhibitor in ERalpha-negative breast cancer cells. Mol. Cancer 2010, 9, 274. [Google Scholar] [CrossRef]
  160. Henning, S.M.; Wang, P.; Carpenter, C.L.; Heber, D. Epigenetic effects of green tea polyphenols in cancer. Epigenomics 2013, 5, 729–741. [Google Scholar] [CrossRef]
  161. Akhavan-Niaki, H.; Samadani, A.A. DNA methylation and cancer development: Molecular mechanism. Cell Biochem. Biophys. 2013, 67, 501–513. [Google Scholar] [CrossRef]
  162. Yuan, Z.; Syed, M.A.; Panchal, D.; Rogers, D.; Joo, M.; Sadikot, R.T. Curcumin mediated epigenetic modulation inhibits TREM-1 expression in response to lipopolysaccharide. Int. J. Biochem. Cell Biol. 2012, 44, 2032–2043. [Google Scholar] [CrossRef]
  163. Abdal Dayem, A.; Choi, H.Y.; Yang, G.M.; Kim, K.; Saha, S.K.; Cho, S.G. The Anti-Cancer Effect of Polyphenols against Breast Cancer and Cancer Stem Cells: Molecular Mechanisms. Nutrients 2016, 8, 581. [Google Scholar] [CrossRef] [PubMed]
  164. Wahyudi, S.; Sargowo, D. Green tea polyphenols inhibit oxidized LDL-induced NF-KB activation in human umbilical vein endothelial cells. Acta Med. Indones. 2007, 39, 66–70. [Google Scholar]
  165. Yang, L.; Zhang, W.; Chopra, S.; Kaur, D.; Wang, H.; Li, M.; Chen, P.; Zhang, W. The Epigenetic Modification of Epigallocatechin Gallate (EGCG) on Cancer. Curr. Drug Targets 2020, 21, 1099–1104. [Google Scholar] [CrossRef] [PubMed]
  166. Kuo, C.L.; Chen, T.S.; Liou, S.Y.; Hsieh, C.C. Immunomodulatory effects of EGCG fraction of green tea extract in innate and adaptive immunity via T regulatory cells in murine model. Immunopharmacol. Immunotoxicol. 2014, 36, 364–370. [Google Scholar] [CrossRef] [PubMed]
  167. Abiri, B.; Amini, S.; Hejazi, M.; Hosseinpanah, F.; Zarghi, A.; Abbaspour, F.; Valizadeh, M. Tea’s anti-obesity properties, cardiometabolic health-promoting potentials, bioactive compounds, and adverse effects: A review focusing on white and green teas. Food Sci. Nutr. 2023, 11, 5818–5836. [Google Scholar] [CrossRef] [PubMed]
  168. Riba, A.; Deres, L.; Sumegi, B.; Toth, K.; Szabados, E.; Halmosi, R. Cardioprotective Effect of Resveratrol in a Postinfarction Heart Failure Model. Oxid. Med. Cell Longev. 2017, 2017, 6819281. [Google Scholar] [CrossRef]
  169. Rauf, A.; Imran, M.; Butt, M.S.; Nadeem, M.; Peters, D.G.; Mubarak, M.S. Resveratrol as an anti-cancer agent: A review. Crit. Rev. Food Sci. Nutr. 2018, 58, 1428–1447. [Google Scholar] [CrossRef]
  170. Zhang, S.; Kiarasi, F. Therapeutic effects of resveratrol on epigenetic mechanisms in age-related diseases: A comprehensive review. Phytother. Res. 2024, 38, 2347–2360. [Google Scholar] [CrossRef]
  171. Fernandes, G.F.S.; Silva GD, B.; Pavan, A.R.; Chiba, D.E.; Chin, C.M.; Dos Santos, J.L. Epigenetic Regulatory Mechanisms Induced by Resveratrol. Nutrients 2017, 9, 1201. [Google Scholar] [CrossRef]
  172. Alesci, A.; Nicosia, N.; Fumia, A.; Giorgianni, F.; Santini, A.; Cicero, N. Resveratrol and Immune Cells: A Link to Improve Human Health. Molecules 2022, 27, 424. [Google Scholar] [CrossRef]
  173. Kursvietiene, L.; Stanevičienė, I.; Mongirdienė, A.; Bernatonienė, J. Multiplicity of effects and health benefits of resveratrol. Medicina 2016, 52, 148–155. [Google Scholar] [CrossRef] [PubMed]
  174. Sharifi-Rad, J.; Rayess, Y.E.; Rizk, A.A.; Sadaka, C.; Zgheib, R.; Zam, W.; Sestito, S.; Rapposelli, S.; Neffe-Skocińska, K.; Zielińska, D.; et al. Turmeric and Its Major Compound Curcumin on Health: Bioactive Effects and Safety Profiles for Food, Pharmaceutical, Biotechnological and Medicinal Applications. Front. Pharmacol. 2020, 11, 01021. [Google Scholar] [CrossRef] [PubMed]
  175. Hassan, F.U.; Rehman, M.S.; Khan, M.S.; Ali, M.A.; Javed, A.; Nawaz, A.; Yang, C. Curcumin as an Alternative Epigenetic Modulator: Mechanism of Action and Potential Effects. Front. Genet. 2019, 10, 514. [Google Scholar] [CrossRef] [PubMed]
  176. Reuter, S.; Gupta, S.C.; Park, B.; Goel, A.; Aggarwal, B.B. Epigenetic changes induced by curcumin and other natural compounds. Genes. Nutr. 2011, 6, 93–108. [Google Scholar] [CrossRef] [PubMed]
  177. Yadav, V.S.; Mishra, K.P.; Singh, D.P.; Mehrotra, S.; Singh, V.K. Immunomodulatory effects of curcumin. Immunopharmacol. Immunotoxicol. 2005, 27, 485–497. [Google Scholar] [CrossRef]
  178. Tayyem, R.F.; Heath, D.D.; Al-Delaimy, W.K.; Rock, C.L. Curcumin content of turmeric and curry powders. Nutr. Cancer 2006, 55, 126–131. [Google Scholar] [CrossRef]
  179. Shabir, I.; Kumar Pandey, V.; Shams, R.; Dar, A.H.; Dash, K.K.; Khan, S.A.; Bashir, I.; Jeevarathinam, G.; Rusu, A.V.; Esatbeyoglu, T.; et al. Promising bioactive properties of quercetin for potential food applications and health benefits: A review. Front. Nutr. 2022, 9, 999752. [Google Scholar] [CrossRef]
  180. Zhu, J.; Cheng, X.; Naumovski, N.; Hu, L.; Wang, K. Epigenetic regulation by quercetin: A comprehensive review focused on its biological mechanisms. Crit. Rev. Food Sci. Nutr. 2023, 1–20. [Google Scholar] [CrossRef]
  181. Manjunath, S.H.; Thimmulappa, R.K. Antiviral, immunomodulatory, and anticoagulant effects of quercetin and its derivatives: Potential role in prevention and management of COVID-19. J. Pharm. Anal. 2022, 12, 29–34. [Google Scholar] [CrossRef]
  182. Sutrisno, S.; Aprina, H.; Simanungkalit, H.M.; Andriyani, A.; Barlianto, W.; Sujuti, H.; Santoso, S.; Dwijayasa, P.M.; Wahyuni, E.S.; Mustofa, E. Genistein modulates the estrogen receptor and suppresses angiogenesis and inflammation in the murine model of peritoneal endometriosis. J. Tradit. Complement. Med. 2018, 8, 278–281. [Google Scholar] [CrossRef]
  183. Sundaram, M.K.; Unni, S.; Somvanshi, P.; Bhardwaj, T.; Mandal, R.K.; Hussain, A.; Haque, S. Genistein Modulates Signaling Pathways and Targets Several Epigenetic Markers in HeLa Cells. Genes 2019, 10, 955. [Google Scholar] [CrossRef] [PubMed]
  184. Guo, T.L.; McCay, J.A.; Zhang, L.X.; Brown, R.D.; You, L.; Karrow, N.A.; Germolec, D.R.; White, K.L., Jr. Genistein modulates immune responses and increases host resistance to B16F10 tumor in adult female B6C3F1 mice. J. Nutr. 2001, 131, 3251–3258. [Google Scholar] [CrossRef]
  185. Yu, L.; Rios, E.; Castro, L.; Liu, J.; Yan, Y.; Dixon, D. Genistein: Dual Role in Women’s Health. Nutrients 2021, 13, 3048. [Google Scholar] [CrossRef] [PubMed]
  186. Shukla, S.; Gupta, S. Apigenin: A promising molecule for cancer prevention. Pharm. Res. 2010, 27, 962–978. [Google Scholar] [CrossRef]
  187. Paredes-Gonzalez, X.; Fuentes, F.; Su, Z.Y.; Kong, A.N. Apigenin reactivates Nrf2 anti-oxidative stress signaling in mouse skin epidermal JB6 P + cells through epigenetics modifications. AAPS J. 2014, 16, 727–735. [Google Scholar] [CrossRef]
  188. Feng, Y.B.; Chen, L.; Chen, F.X.; Yang, Y.; Chen, G.H.; Zhou, Z.H.; Xu, C.F. Immunopotentiation effects of apigenin on NK cell proliferation and killing pancreatic cancer cells. Int. J. Immunopathol. Pharmacol. 2023, 37, 3946320231161174. [Google Scholar] [CrossRef]
  189. McKay, D.L.; Blumberg, J.B. A review of the bioactivity and potential health benefits of chamomile tea (Matricaria recutita L.). Phytother. Res. 2006, 20, 519–530. [Google Scholar] [CrossRef] [PubMed]
  190. Kong, Y.; Chen, G.; Xu, Z.; Yang, G.; Li, B.; Wu, X.; Xiao, W.; Xie, B.; Hu, L.; Sun, X.; et al. Pterostilbene induces apoptosis and cell cycle arrest in diffuse large B-cell lymphoma cells. Sci. Rep. 2016, 6, 37417. [Google Scholar] [CrossRef] [PubMed]
  191. Harandi-Zadeh, S.; Boycott, C.; Beetch, M.; Yang, T.; Martin, B.J.E.; Ren, K.; Kwasniak, A.; Dupuis, J.H.; Lubecka, K.; Yada, R.Y.; et al. Pterostilbene Changes Epigenetic Marks at Enhancer Regions of Oncogenes in Breast Cancer Cells. Antioxidants 2021, 10, 1232. [Google Scholar] [CrossRef]
  192. Liu, P.; Tang, W.; Xiang, K.; Li, G. Pterostilbene in the treatment of inflammatory and oncological diseases. Front. Pharmacol. 2023, 14, 1323377. [Google Scholar] [CrossRef]
  193. Joo, S.Y.; Song, Y.A.; Park, Y.L.; Myung, E.; Chung, C.Y.; Park, K.J.; Cho, S.B.; Lee, W.S.; Kim, H.S.; Rew, J.S.; et al. Epigallocatechin-3-gallate Inhibits LPS-Induced NF-kappaB and MAPK Signaling Pathways in Bone Marrow-Derived Macrophages. Gut Liver 2012, 6, 188–196. [Google Scholar] [CrossRef] [PubMed]
  194. Tanabe, H.; Suzuki, T.; Ohishi, T.; Isemura, M.; Nakamura, Y.; Unno, K. Effects of Epigallocatechin-3-Gallate on Matrix Metalloproteinases in Terms of Its Anticancer Activity. Molecules 2023, 28, 525. [Google Scholar] [CrossRef] [PubMed]
  195. Rodriguez, S.K.; Guo, W.; Liu, L.; Band, M.A.; Paulson, E.K.; Meydani, M. Green tea catechin, epigallocatechin-3-gallate, inhibits vascular endothelial growth factor angiogenic signaling by disrupting the formation of a receptor complex. Int. J. Cancer 2006, 118, 1635–1644. [Google Scholar] [CrossRef] [PubMed]
  196. Eddy, S.F.; Kane, S.E.; Sonenshein, G.E. Trastuzumab-resistant HER2-driven breast cancer cells are sensitive to epigallocatechin-3 gallate. Cancer Res. 2007, 67, 9018–9023. [Google Scholar] [CrossRef] [PubMed]
  197. 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 effective against breast cancer. Carcinogenesis 2006, 27, 2424–2433. [Google Scholar] [CrossRef]
  198. Delmas, D.; Lançon, A.; Colin, D.; Jannin, B.; Latruffe, N. Resveratrol as a chemopreventive agent: A promising molecule for fighting cancer. Curr. Drug Targets 2006, 7, 423–442. [Google Scholar] [CrossRef]
  199. Hu, C.; Liu, Y.; Teng, M.; Jiao, K.; Zhen, J.; Wu, M.; Li, Z. Resveratrol inhibits the proliferation of estrogen receptor-positive breast cancer cells by suppressing EZH2 through the modulation of ERK1/2 signaling. Cell Biol. Toxicol. 2019, 35, 445–456. [Google Scholar] [CrossRef]
  200. Ren, Z.; Wang, L.; Cui, J.; Huoc, Z.; Xue, J.; Cui, H.; Mao, Q.; Yang, R. Resveratrol inhibits NF-kB signaling through suppression of p65 and IkappaB kinase activities. Pharmazie 2013, 68, 689–694. [Google Scholar]
  201. Baek, S.H.; Ko, J.H.; Lee, H.; Jung, J.; Kong, M.; Lee, J.W.; Lee, J.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; et al. Resveratrol inhibits STAT3 signaling pathway through the induction of SOCS-1: Role in apoptosis induction and radiosensitization in head and neck tumor cells. Phytomedicine 2016, 23, 566–577. [Google Scholar] [CrossRef]
  202. Otsuka, K.; Yamamoto, Y.; Ochiya, T. Regulatory role of resveratrol, a microRNA-controlling compound, in HNRNPA1 expression, which is associated with poor prognosis in breast cancer. Oncotarget 2018, 9, 24718–24730. [Google Scholar] [CrossRef]
  203. Ghasemi, F.; Shafiee, M.; Banikazemi, Z.; Pourhanifeh, M.H.; Khanbabaei, H.; Shamshirian, A.; Amiri Moghadam, S.; ArefNezhad, R.; Sahebkar, A.; Avan, A.; et al. Curcumin inhibits NF-kB and Wnt/beta-catenin pathways in cervical cancer cells. Pathol. Res. Pract. 2019, 215, 152556. [Google Scholar] [CrossRef] [PubMed]
  204. Zoi, V.; Kyritsis, A.P.; Galani, V.; Lazari, D.; Sioka, C.; Voulgaris, S.; Alexiou, G.A. The Role of Curcumin in Cancer: A Focus on the PI3K/Akt Pathway. Cancers 2024, 16, 1554. [Google Scholar] [CrossRef] [PubMed]
  205. Singh, S.; Barnes, C.A.; D’Souza, J.S.; Hosur, R.V.; Mishra, P. Curcumin, a potential initiator of apoptosis via direct interactions with Bcl-xL and Bid. Proteins 2022, 90, 455–464. [Google Scholar] [CrossRef] [PubMed]
  206. Huang, T.; Chen, Z.; Fang, L. Curcumin inhibits LPS-induced EMT through downregulation of NF-kappaB-Snail signaling in breast cancer cells. Oncol. Rep. 2013, 29, 117–124. [Google Scholar] [CrossRef]
  207. Peng, Y.; Ao, M.; Dong, B.; Jiang, Y.; Yu, L.; Chen, Z.; Hu, C.; Xu, R. Anti-Inflammatory Effects of Curcumin in the Inflammatory Diseases: Status, Limitations and Countermeasures. Drug Des. Devel Ther. 2021, 15, 4503–4525. [Google Scholar] [CrossRef]
  208. Farghadani, R.; Naidu, R. Curcumin as an Enhancer of Therapeutic Efficiency of Chemotherapy Drugs in Breast Cancer. Int. J. Mol. Sci. 2022, 23, 2144. [Google Scholar] [CrossRef]
  209. Kasiri, N.; Rahmati, M.; Ahmadi, L.; Eskandari, N.; Motedayyen, H. Therapeutic potential of quercetin on human breast cancer in different dimensions. Inflammopharmacology 2020, 28, 39–62. [Google Scholar] [CrossRef] [PubMed]
  210. Kiyga, E.; Şengelen, A.; Adıgüzel, Z.; Önay Uçar, E. Investigation of the role of quercetin as a heat shock protein inhibitor on apoptosis in human breast cancer cells. Mol. Biol. Rep. 2020, 47, 4957–4967. [Google Scholar] [CrossRef]
  211. Michalski, J.; Deinzer, A.; Stich, L.; Zinser, E.; Steinkasserer, A.; Knippertz, I. Quercetin induces an immunoregulatory phenotype in maturing human dendritic cells. Immunobiology 2020, 225, 151929. [Google Scholar] [CrossRef]
  212. Kobori, M.; Takahashi, Y.; Sakurai, M.; Akimoto, Y.; Tsushida, T.; Oike, H.; Ippoushi, K. Quercetin suppresses immune cell accumulation and improves mitochondrial gene expression in adipose tissue of diet-induced obese mice. Mol. Nutr. Food Res. 2016, 60, 300–312. [Google Scholar] [CrossRef]
  213. Ge, Y.W.; Feng, K.; Liu, X.L.; Zhu, Z.A.; Chen, H.F.; Chang, Y.Y.; Sun, Z.Y.; Wang, H.W.; Zhang, J.W.; Yu, D.G.; et al. Quercetin inhibits macrophage polarization through the p-38alpha/beta signalling pathway and regulates OPG/RANKL balance in a mouse skull model. J. Cell Mol. Med. 2020, 24, 3203–3216. [Google Scholar] [CrossRef] [PubMed]
  214. Su, T.; Shen, H.; He, M.; Yang, S.; Gong, X.; Huang, C.; Guo, L.; Wang, H.; Feng, S.; Mi, T.; et al. Quercetin promotes the proportion and maturation of NK cells by binding to MYH9 and improves cognitive functions in aged mice. Immun. Ageing 2024, 21, 29. [Google Scholar] [CrossRef] [PubMed]
  215. Wu, A.H.; Yu, M.C.; Tseng, C.C.; Pike, M.C. Epidemiology of soy exposures and breast cancer risk. Br. J. Cancer 2008, 98, 9–14. [Google Scholar] [CrossRef]
  216. Wu, A.H.; Koh, W.P.; Wang, R.; Lee, H.P.; Yu, M.C. Soy intake and breast cancer risk in Singapore Chinese Health Study. Br. J. Cancer 2008, 99, 196–200. [Google Scholar] [CrossRef]
  217. Spagnuolo, C.; Russo, G.L.; Orhan, I.E.; Habtemariam, S.; Daglia, M.; Sureda, A.; Nabavi, S.F.; Devi, K.P.; Loizzo, M.R.; Tundis, R.; et al. Genistein and cancer: Current status, challenges, and future directions. Adv. Nutr. 2015, 6, 408–419. [Google Scholar] [CrossRef] [PubMed]
  218. Li, Y.; Ahmed, F.; Ali, S.; Philip, P.A.; Kucuk, O.; Sarkar, F.H. Inactivation of nuclear factor kappaB by soy isoflavone genistein contributes to increased apoptosis induced by chemotherapeutic agents in human cancer cells. Cancer Res. 2005, 65, 6934–6942. [Google Scholar] [CrossRef]
  219. Prietsch, R.F.; Monte, L.G.; da Silva, F.A.; Beira, F.T.; Del Pino, F.A.; Campos, V.F.; Collares, T.; Pinto, L.S.; Spanevello, R.M.; Gamaro, G.D.; et al. Genistein induces apoptosis and autophagy in human breast MCF-7 cells by modulating the expression of proapoptotic factors and oxidative stress enzymes. Mol. Cell. Biochem. 2014, 390, 235–342. [Google Scholar] [CrossRef] [PubMed]
  220. Bhat, S.S.; Prasad, S.K.; Shivamallu, C.; Prasad, K.S.; Syed, A.; Reddy, P.; Cull, C.A.; Amachawadi, R.G. Genistein: A Potent Anti-Breast Cancer Agent. Curr. Issues Mol. Biol. 2021, 43, 1502–1517. [Google Scholar] [CrossRef]
  221. Konstantinou, E.K.; Gioxari, A.; Dimitriou, M.; Panoutsopoulos, G.I.; Panagiotopoulos, A.A. Molecular Pathways of Genistein Activity in Breast Cancer Cells. Int. J. Mol. Sci. 2024, 25, 5556. [Google Scholar] [CrossRef]
  222. Sohel, M. Genistein, a Potential Phytochemical against Breast Cancer Treatment-Insight into the Molecular Mechanisms. Processes 2022, 10, 415. [Google Scholar] [CrossRef]
  223. Yu, X.; Zhu, J.; Mi, M.; Chen, W.; Pan, Q.; Wei, M. Anti-angiogenic genistein inhibits VEGF-induced endothelial cell activation by decreasing PTK activity and MAPK activation. Med. Oncol. 2012, 29, 349–357. [Google Scholar] [CrossRef] [PubMed]
  224. Mai, Z.; Blackburn, G.L.; Zhou, J.R. Genistein sensitizes inhibitory effect of tamoxifen on the growth of estrogen receptor-positive and HER2-overexpressing human breast cancer cells. Mol. Carcinog. 2007, 46, 534–542. [Google Scholar] [CrossRef]
  225. Imran, M.; Aslam Gondal, T.; Atif, M.; Shahbaz, M.; Batool Qaisarani, T.; Hanif Mughal, M.; Salehi, B.; Martorell, M.; Sharifi-Rad, J. Apigenin as an anticancer agent. Phytother. Res. 2020, 34, 1812–1828. [Google Scholar] [CrossRef]
  226. Zhu, Y.; Mao, Y.; Chen, H.; Lin, Y.; Hu, Z.; Wu, J.; Xu, X.; Xu, X.; Qin, J.; Xie, L. Apigenin promotes apoptosis, inhibits invasion and induces cell cycle arrest of T24 human bladder cancer cells. Cancer Cell Int. 2013, 13, 54. [Google Scholar] [CrossRef]
  227. Liang, Y.-C. Apigenin inhibits migration and induces apoptosis of human endometrial carcinoma Ishikawa cells via PI3K-AKT-GSK-3β pathway and endoplasmic reticulum stress. J. Funct. Foods 2022, 94, 105116. [Google Scholar] [CrossRef]
  228. Tong, J.; Shen, Y.; Zhang, Z.; Hu, Y.; Zhang, X.; Han, L. Apigenin inhibits epithelial-mesenchymal transition of human colon cancer cells through NF-kappaB/Snail signaling pathway. Biosci. Rep. 2019, 39, BSR20190452. [Google Scholar] [CrossRef] [PubMed]
  229. Seo, H.S.; Choi, H.S.; Kim, S.R.; Choi, Y.K.; Woo, S.M.; Shin, I.; Woo, J.K.; Park, S.Y.; Shin, Y.C.; Ko, S.G. Apigenin induces apoptosis via extrinsic pathway, inducing p53 and inhibiting STAT3 and NFkappaB signaling in HER2-overexpressing breast cancer cells. Mol. Cell. Biochem. 2012, 366, 319–334. [Google Scholar] [CrossRef]
  230. Shukla, S.; Shankar, E.; Fu, P.; MacLennan, G.; Gupta, S. Suppression of NF-kappaB and NF-kappaB-Regulated Gene Expression by Apigenin through IkappaBalpha and IKK Pathway in TRAMP Mice. PLoS ONE 2015, 10, e0138710. [Google Scholar] [CrossRef] [PubMed]
  231. Mahbub, A.A.; Le Maitre, C.L.; Cross, N.A.; Jordan-Mahy, N. The effect of apigenin and chemotherapy combination treatments on apoptosis-related genes and proteins in acute leukaemia cell lines. Sci. Rep. 2022, 12, 8858. [Google Scholar] [CrossRef]
  232. Obrador, E.; Salvador-Palmer, R.; Jihad-Jebbar, A.; López-Blanch, R.; Dellinger, T.H.; Dellinger, R.W.; Estrela, J.M. Pterostilbene in Cancer Therapy. Antioxidants 2021, 10, 492. [Google Scholar] [CrossRef]
  233. Elsherbini, A.M.; Sheweita, S.A.; Sultan, A.S. Pterostilbene as a Phytochemical Compound Induces Signaling Pathways Involved in the Apoptosis and Death of Mutant P53-Breast Cancer Cell Lines. Nutr. Cancer 2021, 73, 1976–1984. [Google Scholar] [CrossRef] [PubMed]
  234. Surien, O.; Masre, S.F.; Basri, D.F.; Ghazali, A.R. Potential Chemopreventive Role of Pterostilbene in Its Modulation of the Apoptosis Pathway. Int. J. Mol. Sci. 2023, 24, 9707. [Google Scholar] [CrossRef] [PubMed]
  235. Ma, K. Pterostilbene inhibits the metastasis of TNBC via suppression of β-catenin-mediated epithelial to mesenchymal transition and stemness. J. Funct. Foods 2022, 96, 105219. [Google Scholar] [CrossRef]
  236. Su, C.M.; Lee, W.H.; Wu, A.T.; Lin, Y.K.; Wang, L.S.; Wu, C.H.; Yeh, C.T. Pterostilbene inhibits triple-negative breast cancer metastasis via inducing microRNA-205 expression and negatively modulates epithelial-to-mesenchymal transition. J. Nutr. Biochem. 2015, 26, 675–685. [Google Scholar] [CrossRef]
  237. Link, A.; Balaguer, F.; Goel, A. Cancer chemoprevention by dietary polyphenols: Promising role for epigenetics. Biochem. Pharmacol. 2010, 80, 1771–1792. [Google Scholar] [CrossRef]
  238. Li, M.J.; Lin, Y.C.; Wang, J.; Jiang, Y.F. Green tea compounds in breast cancer prevention and treatment. World J. Clin. Oncol. 2014, 5, 520–528. [Google Scholar] [CrossRef]
  239. Asensi, M.; Mena, S.; Feddi, F.; Estrela, J.M. Natural polyphenols in cancer therapy. Crit. Rev. Clin. Lab. Sci. 2011, 48, 197–216. [Google Scholar] [CrossRef]
  240. Pan, M.H.; Ho, C.T. Chemopreventive effects of natural dietary compounds on cancer development. Chem. Soc. Rev. 2008, 37, 2558–2574. [Google Scholar] [CrossRef]
  241. Mocanu, M.M.; Nagy, P.; Szollosi, J. Chemoprevention of Breast Cancer by Dietary Polyphenols. Molecules 2015, 20, 22578–22620. [Google Scholar] [CrossRef]
  242. Higdon, J.V.; Frei, B. Tea catechins and polyphenols: Health effects, metabolism, and antioxidant functions. Crit. Rev. Food Sci. Nutr. 2003, 43, 89–143. [Google Scholar] [CrossRef]
  243. Liu, R.H. Potential synergy of phytochemicals in cancer prevention: Mechanism of action. J. Nutr. 2004, 134 (Suppl. S12), 3479S–3485S. [Google Scholar] [CrossRef] [PubMed]
  244. Rudrapal, M.; Khairnar, S.J.; Khan, J.; Dukhyil, A.B.; Ansari, M.A.; Alomary, M.N.; Alshabrmi, F.M.; Palai, S.; Deb, P.K.; Devi, R. Dietary Polyphenols and Their Role in Oxidative Stress-Induced Human Diseases: Insights Into Protective Effects, Antioxidant Potentials and Mechanism(s) of Action. Front. Pharmacol. 2022, 13, 806470. [Google Scholar]
  245. V Ganesh, G.; Ramkumar, K.M. Pterostilbene attenuates hemin-induced dysregulation of macrophage M2 polarization via Nrf2 activation in experimental hyperglycemia. Inflammopharmacology 2023, 31, 2133–2145. [Google Scholar] [CrossRef] [PubMed]
  246. Marzocchella, L.; Fantini, M.; Benvenuto, M.; Masuelli, L.; Tresoldi, I.; Modesti, A.; Bei, R. Dietary flavonoids: Molecular mechanisms of action as anti- inflammatory agents. Recent. Pat. Inflamm. Allergy Drug Discov. 2011, 5, 200–220. [Google Scholar] [CrossRef] [PubMed]
  247. Ostios-Garcia, L.; Villamayor, J.; Garcia-Lorenzo, E.; Vinal, D.; Feliu, J. Understanding the immune response and the current landscape of immunotherapy in pancreatic cancer. World J. Gastroenterol. 2021, 27, 6775–6793. [Google Scholar] [CrossRef]
  248. Aherne, S.A.; O’Brien, N.M. Dietary flavonols: Chemistry, food content, and metabolism. Nutrition 2002, 18, 75–81. [Google Scholar] [CrossRef]
  249. Cimino, S.; Sortino, G.; Favilla, V.; Castelli, T.; Madonia, M.; Sansalone, S.; Russo, G.I.; Morgia, G. Polyphenols: Key issues involved in chemoprevention of prostate cancer. Oxid. Med. Cell Longev. 2012, 2012, 632959. [Google Scholar] [CrossRef]
Figure 1. Breast cancer and its subtypes. (A) Molecular classification. (B) Hormone receptor status classification. (C) Histological classification. Created in BioRender. Lab, T. (2024) https://BioRender.com/u39p454 (accessed on 4 November 2024). HER2-enriched breast cancer carries a high risk of recurrence and neoplastic dissemination [24,25,26,27]. Approximately no more than three out of twenty breast cancer subtypes are classified as HER2 positive [24,26]. HER2-enriched breast cancers have HER2 expression while lacking ER and PR [26,27]. Basal-like breast cancer constitutes around one-fifteenth to one-fifth of all breast cancer incidences and lacks the expression of any HR [13,26,27,28]. This results in TNBC being the most aggressive and metastatic subtype, with a higher likelihood of early relapse and poor prognosis [13,27,28]. TNBC comprises six well-defined subtypes: basal-like 1 (BL1), basal-like 2 (BL2), immunomodulatory (IM), luminal androgen receptor (LAR), mesenchymal (M), and mesenchymal stem-like (MSL) [15,29,30,31,32,33]. Claudin-low breast cancer (CLBC) is distinguished by reduced tight junction proteins such as Claudins 3 and 7 and adhesion proteins [15,34,35]. CLBC represents 7–14% of all aggressive breast cancers and is connected to a low survival rate, as these tumors typically test negative for ER, PR, and HER2 [15,34,35]. The hormone receptor status can further categorize breast cancers based on hormone receptor expression (HR+), such as ER- and PR-positive breast cancers [26,28,36]. Ductal carcinoma in situ (DCIS), lobular carcinoma in situ (LCIS), Invasive Ductal Carcinoma (IDC), and Invasive Lobular Carcinoma (ILC) are histological characteristics of breast cancer subtypes [23,26,34,35,36].
Figure 1. Breast cancer and its subtypes. (A) Molecular classification. (B) Hormone receptor status classification. (C) Histological classification. Created in BioRender. Lab, T. (2024) https://BioRender.com/u39p454 (accessed on 4 November 2024). HER2-enriched breast cancer carries a high risk of recurrence and neoplastic dissemination [24,25,26,27]. Approximately no more than three out of twenty breast cancer subtypes are classified as HER2 positive [24,26]. HER2-enriched breast cancers have HER2 expression while lacking ER and PR [26,27]. Basal-like breast cancer constitutes around one-fifteenth to one-fifth of all breast cancer incidences and lacks the expression of any HR [13,26,27,28]. This results in TNBC being the most aggressive and metastatic subtype, with a higher likelihood of early relapse and poor prognosis [13,27,28]. TNBC comprises six well-defined subtypes: basal-like 1 (BL1), basal-like 2 (BL2), immunomodulatory (IM), luminal androgen receptor (LAR), mesenchymal (M), and mesenchymal stem-like (MSL) [15,29,30,31,32,33]. Claudin-low breast cancer (CLBC) is distinguished by reduced tight junction proteins such as Claudins 3 and 7 and adhesion proteins [15,34,35]. CLBC represents 7–14% of all aggressive breast cancers and is connected to a low survival rate, as these tumors typically test negative for ER, PR, and HER2 [15,34,35]. The hormone receptor status can further categorize breast cancers based on hormone receptor expression (HR+), such as ER- and PR-positive breast cancers [26,28,36]. Ductal carcinoma in situ (DCIS), lobular carcinoma in situ (LCIS), Invasive Ductal Carcinoma (IDC), and Invasive Lobular Carcinoma (ILC) are histological characteristics of breast cancer subtypes [23,26,34,35,36].
Nutrients 16 04143 g001
Figure 2. The diversity of immune system cells in the cancer-associated environment of breast cancer [43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71]. Created in BioRender. Lab, T. (2024) https://BioRender.com/s56n309 (accessed on 4 November 2024).
Figure 2. The diversity of immune system cells in the cancer-associated environment of breast cancer [43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71]. Created in BioRender. Lab, T. (2024) https://BioRender.com/s56n309 (accessed on 4 November 2024).
Nutrients 16 04143 g002
Figure 3. General subtypes of polyphenols. Created in BioRender. Lab, T. (2024) https://BioRender.com/q91l231 (accessed on 4 November 2024).
Figure 3. General subtypes of polyphenols. Created in BioRender. Lab, T. (2024) https://BioRender.com/q91l231 (accessed on 4 November 2024).
Nutrients 16 04143 g003
Figure 4. The common impacts of EGCG (epigallocatechin gallate) in breast cancer. EGCG influences multiple pathways, reducing cell proliferation and promoting apoptosis by inhibiting the PI3K/AKT pathway and activating tumor suppressor proteins like p53, leading to Bax activation and BCL-2 suppression. EGCG blocks the Wnt/β-catenin and MAPK pathways, reducing COX-2 levels and consequently lowering inflammation. It also suppresses matrix metalloproteinases (MMPs), preventing extracellular matrix degradation and angiogenesis. Created in BioRender. Lab, T. (2024) https://BioRender.com/e47a401 (accessed on 4 November 2024).
Figure 4. The common impacts of EGCG (epigallocatechin gallate) in breast cancer. EGCG influences multiple pathways, reducing cell proliferation and promoting apoptosis by inhibiting the PI3K/AKT pathway and activating tumor suppressor proteins like p53, leading to Bax activation and BCL-2 suppression. EGCG blocks the Wnt/β-catenin and MAPK pathways, reducing COX-2 levels and consequently lowering inflammation. It also suppresses matrix metalloproteinases (MMPs), preventing extracellular matrix degradation and angiogenesis. Created in BioRender. Lab, T. (2024) https://BioRender.com/e47a401 (accessed on 4 November 2024).
Nutrients 16 04143 g004
Figure 5. The immunomodulatory effects of resveratrol on cancer and inflammation. Resveratrol impacts several signaling pathways and immune cells, contributing to anti-cancer and anti-inflammatory actions. Resveratrol alters cancer hallmarks by suppressing inflammatory mediators while promoting IL-10 production through the inhibition of the PI3K/AKT pathway. This influences tumor-infiltrating immune cells, such as cytotoxic T cells, dendritic cells, Tregs, and tumor-associated macrophages, enhancing immune response against tumors. It reduces TNF-α, impacting IL-2 and IL-10 levels, thus modifying immune and inflammatory responses within the tumor microenvironment. Created in BioRender. Lab, T. (2024) https://BioRender.com/e44e698 (accessed on 4 November 2024).
Figure 5. The immunomodulatory effects of resveratrol on cancer and inflammation. Resveratrol impacts several signaling pathways and immune cells, contributing to anti-cancer and anti-inflammatory actions. Resveratrol alters cancer hallmarks by suppressing inflammatory mediators while promoting IL-10 production through the inhibition of the PI3K/AKT pathway. This influences tumor-infiltrating immune cells, such as cytotoxic T cells, dendritic cells, Tregs, and tumor-associated macrophages, enhancing immune response against tumors. It reduces TNF-α, impacting IL-2 and IL-10 levels, thus modifying immune and inflammatory responses within the tumor microenvironment. Created in BioRender. Lab, T. (2024) https://BioRender.com/e44e698 (accessed on 4 November 2024).
Nutrients 16 04143 g005
Figure 6. Important effects of curcumin on breast cancer. Curcumin suppresses Wnt and MAPK pathways, along with epithelial–mesenchymal transition (EMT) in the tumor microenvironment, histone acetylation, angiogenesis, and T-regulatory cells, which play significant roles in cancer development. In contrast, curcumin activates dendritic cells, which promotes the secretion of IL-12 and activates cytotoxic T cells. Furthermore, curcumin stimulates macrophages, cytotoxic T cells, and natural killer (NK) cells, leading to the activation of T helper cells and the increased apoptosis of tumor cells. Notably, curcumin also reduces PD-1 expression on T cells, thereby enhancing the immune response against tumor cells. Created in BioRender. Lab, T. (2024) https://BioRender.com/w77n343 (accessed on 4 November 2024).
Figure 6. Important effects of curcumin on breast cancer. Curcumin suppresses Wnt and MAPK pathways, along with epithelial–mesenchymal transition (EMT) in the tumor microenvironment, histone acetylation, angiogenesis, and T-regulatory cells, which play significant roles in cancer development. In contrast, curcumin activates dendritic cells, which promotes the secretion of IL-12 and activates cytotoxic T cells. Furthermore, curcumin stimulates macrophages, cytotoxic T cells, and natural killer (NK) cells, leading to the activation of T helper cells and the increased apoptosis of tumor cells. Notably, curcumin also reduces PD-1 expression on T cells, thereby enhancing the immune response against tumor cells. Created in BioRender. Lab, T. (2024) https://BioRender.com/w77n343 (accessed on 4 November 2024).
Nutrients 16 04143 g006
Figure 7. Diverse effects of quercetin on breast cancer. Quercetin effectively inhibits the PI3K pathway, angiogenesis, DNA methylation, and the activity of myeloid-derived suppressor cells (MDSCs), which typically suppress the functions of dendritic cells (DCs), macrophages, and T-regulatory cells. Additionally, quercetin promotes the secretion of IL-10 and enhances histone acetylation. It also activates T helper cells, natural killer (NK) cells, and monocytes, contributing to a more robust immune response. Created in BioRender. Lab, T. (2024) https://BioRender.com/i98p170 (accessed on 4 November 2024).
Figure 7. Diverse effects of quercetin on breast cancer. Quercetin effectively inhibits the PI3K pathway, angiogenesis, DNA methylation, and the activity of myeloid-derived suppressor cells (MDSCs), which typically suppress the functions of dendritic cells (DCs), macrophages, and T-regulatory cells. Additionally, quercetin promotes the secretion of IL-10 and enhances histone acetylation. It also activates T helper cells, natural killer (NK) cells, and monocytes, contributing to a more robust immune response. Created in BioRender. Lab, T. (2024) https://BioRender.com/i98p170 (accessed on 4 November 2024).
Nutrients 16 04143 g007
Table 1. Common polyphenols used in breast cancer treatment.
Table 1. Common polyphenols used in breast cancer treatment.
NameTheir ImportanceEpigenetic EffectsImmunomodulatory EffectsFound In
Epigallocatechin gallate (EGCG)Strong antioxidant and anti-inflammatory properties and potential anti-cancer properties; inhibition of tumor cell proliferation [163,164]DNA methylation inhibition, histone modification, miRNA expression changes [165]It enhances cytotoxic T cell function, inhibits Tregs, and increases macrophages. It also reduces inflammation and suppresses NF-kB signaling [166].Green tea, white tea [167]Nutrients 16 04143 i001
ResveratrolCardioprotective and anti-inflammatory properties, potential cancer prevention; promotes cell death in cancer cells [168,169]Promotes histone acetylation, inhibits DNA methyltransferases, modifies miRNA [170,171]It modulates T helper cell activity, particularly by promoting a shift from a pro-inflammatory Th1 response (which could contribute to chronic inflammation) to a more balanced Th2 or regulatory T cell (Treg) response, reducing inflammation while maintaining immune surveillance against cancer [172].Grapes, red wine, berries, peanuts [173]Nutrients 16 04143 i002
CurcuminAnti-inflammatory and antioxidant properties; modulates multiple cell signaling pathways; potential anti-cancer agent [174]Histone acetylation inhibition [175,176]It modulates the differentiation of T helper cells, reduces the expression of immune suppressive Tregs, and modulates macrophages and NK cells [177].Turmeric (Curcuma longa) [178] Nutrients 16 04143 i003
QuercetinAnti-inflammatory, antioxidant, and antiviral properties; may enhance immunity and reduce cancer risk [179] Demethylation of tumor suppressor genes and histone acetylation, modifies miRNA [180]Monocyte activation leads to the synthesis of interleukin-10 and TGF-β, inducing M1 macrophages while inhibiting M2 macrophages, which are associated with pro-tumor activity [181].Apples, onions, berries, leafy greens [179]Nutrients 16 04143 i004
GenisteinModulates estrogen receptors; potentially prevents breast cancer; antioxidant properties [182]Inhibits DNA methylation, induces histone acetylation, and alters miRNA expression [183]It enhances the activity of cytotoxic T cells. It modulates the function of T helper cells and leads to the activation of B cells and macrophages [184].Soybeans, tofu, soy products [185]Nutrients 16 04143 i005
ApigeninAnti-inflammatory and antioxidant properties; may induce apoptosis in cancer cells; neuroprotective effects [186]Inhibits histone deacetylases, promotes DNA demethylation [187]Inducing NK cell activity, boosting their ability to destroy tumor cells [188].Parsley, chamomile, celery, citrus fruits [186,189]Nutrients 16 04143 i006
PterostilbeneAntioxidant and anti-inflammatory properties; potential anti-cancer properties; modulates lipid metabolism [190]Modulates histone acetylation and DNA methylation, affects miRNA profiles [191]It suppresses macrophage activity and promotes dendritic cells’ maturation and has an antigen-presenting function [192].Blueberries, grapes, heartwood of Pterocarpus marsupium [190]Nutrients 16 04143 i007
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Eren, E.; Das, J.; Tollefsbol, T.O. Polyphenols as Immunomodulators and Epigenetic Modulators: An Analysis of Their Role in the Treatment and Prevention of Breast Cancer. Nutrients 2024, 16, 4143. https://doi.org/10.3390/nu16234143

AMA Style

Eren E, Das J, Tollefsbol TO. Polyphenols as Immunomodulators and Epigenetic Modulators: An Analysis of Their Role in the Treatment and Prevention of Breast Cancer. Nutrients. 2024; 16(23):4143. https://doi.org/10.3390/nu16234143

Chicago/Turabian Style

Eren, Esmanur, Jyotirmoyee Das, and Trygve O. Tollefsbol. 2024. "Polyphenols as Immunomodulators and Epigenetic Modulators: An Analysis of Their Role in the Treatment and Prevention of Breast Cancer" Nutrients 16, no. 23: 4143. https://doi.org/10.3390/nu16234143

APA Style

Eren, E., Das, J., & Tollefsbol, T. O. (2024). Polyphenols as Immunomodulators and Epigenetic Modulators: An Analysis of Their Role in the Treatment and Prevention of Breast Cancer. Nutrients, 16(23), 4143. https://doi.org/10.3390/nu16234143

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop