Next Article in Journal
Essential Oils, Pituranthos chloranthus and Teucrium ramosissimum, Chemosensitize Resistant Human Uterine Sarcoma MES-SA/Dx5 Cells to Doxorubicin by Inducing Apoptosis and Targeting P-Glycoprotein
Previous Article in Journal
Effects of Feeding Time on Markers of Muscle Metabolic Flexibility Following Acute Aerobic Exercise in Trained Mice Undergoing Time Restricted Feeding
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Polyphenols Modulating Effects of PD-L1/PD-1 Checkpoint and EMT-Mediated PD-L1 Overexpression in Breast Cancer

1
Division of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health Florida A&M University, Tallahassee, FL 32307, USA
2
Faculty of Medical Technology-Misrata, Libyan National Board for Technical & Vocational Education, Misrata LY72, Libya
*
Author to whom correspondence should be addressed.
Submission received: 12 April 2021 / Revised: 11 May 2021 / Accepted: 17 May 2021 / Published: 19 May 2021
(This article belongs to the Section Phytochemicals and Human Health)

Abstract

:
Investigating dietary polyphenolic compounds as antitumor agents are rising due to the growing evidence of the close association between immunity and cancer. Cancer cells elude immune surveillance for enhancing their progression and metastasis utilizing various mechanisms. These mechanisms include the upregulation of programmed death-ligand 1 (PD-L1) expression and Epithelial-to-Mesenchymal Transition (EMT) cell phenotype activation. In addition to its role in stimulating normal embryonic development, EMT has been identified as a critical driver in various aspects of cancer pathology, including carcinogenesis, metastasis, and drug resistance. Furthermore, EMT conversion to another phenotype, Mesenchymal-to-Epithelial Transition (MET), is crucial in developing cancer metastasis. A central mechanism in the upregulation of PD-L1 expression in various cancer types is EMT signaling activation. In breast cancer (BC) cells, the upregulated level of PD-L1 has become a critical target in cancer therapy. Various signal transduction pathways are involved in EMT-mediated PD-L1 checkpoint overexpression. Three main groups are considered potential targets in EMT development; the effectors (E-cadherin and Vimentin), the regulators (Zeb, Twist, and Snail), and the inducers that include members of the transforming growth factor-beta (TGF-β). Meanwhile, the correlation between consuming flavonoid-rich food and the lower risk of cancers has been demonstrated. In BC, polyphenols were found to downregulate PD-L1 expression. This review highlights the effects of polyphenols on the EMT process by inhibiting mesenchymal proteins and upregulating the epithelial phenotype. This multifunctional mechanism could hold promises in the prevention and treating breast cancer.

Graphical Abstract

1. Introduction

The association between metastasis and immunity is considered a hallmark of cancer [1]. Cancer cell metastasis and invasion of vital organs are implicated in poor prognosis and cancer-related deaths [2]. As the first line of defense, the anticancer immune system can distinguish and remove these cancer cells in patients with malignancy. This mechanism initiates T-cell activation, controlled by T-cell receptor (TCR) mediated-signaling pathways, and maintains the immune system homeostasis [3]. However, it has become evident that tumor cells elude immune surveillance for enhancing their progression and metastasis. Tumor utilizes various molecular mechanisms; one of them is the typical immune-suppressive tumor microenvironments that weaken the immune response, allowing an uncontrollable proliferation of cancer cells. More importantly, cancer cells acquire mesenchymal phenotypes that can induce immunosuppression via Epithelial-to-Mesenchymal Transition (EMT).
For epithelial cells, the process of EMT is essential for driving various progressive aspects such as embryonic development and wound healing. However, EMT is also playing a crucial role in immunosuppression and the development of tissue fibrosis, carcinogenesis, metastasis, and drug resistance [4,5]. EMT enhances the migration of epithelial cells to new locations by promoting new organized characters under normal conditions. In contrast, the activated EMT cells in cancer undergo Mesenchymal-to-Epithelial Transition (MET), the crucial phenotype in developing cancer metastasis [6]. In the stage of EMT, the tumor utilizes various molecular mechanisms to escape the immune surveillance; one of them is the upregulation of programmed death-ligand 1 (PD-L1) expression [7]. In various types of cancer, the activation of EMT signaling seems to be a central oncogenic mechanism that upregulates PD-L1 expression [8]. A recent study has cited the close association between EMT and PD-L1, suggesting a bidirectional regulation between EMT status and PD-L1 expression, which leads to tumor immune escape [9,10]. During this process, the cells lose essential epithelial proteins (such as E-cadherin, claudins, cytokeratin, occludins, mucin-1, desmoplakin, and γ-catenin) while express mesenchymal phenotype characteristics with Vimentin, N-cadherin, fibronectin, fibroblast-specific protein 1 (FSP-1), Vitronectin, and smooth-muscle actin which cause immunosuppression and enhance tumor dissemination and migration [11]. Based on that, Pasquier and others have classified the potential therapeutic targets in EMT development into three main groups; the effectors (such as E-cadherin and vimentin), the regulators (such as Zeb, Twist, and Snail transcription factors), and the third group is the inducers that include members of the transforming growth factor-beta (TGF-β) [12].
Immunotherapy has become a novel approach for cancer therapy [13,14]. In advanced cancer, various immune checkpoint inhibitors, including programmed cell death 1 (PD-1), its ligand PD-L1, and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), have achieved promising oncological improvements [15,16,17,18]. The ligand PD-L1 (also known as B7-H1/CD274) is a transmembrane glycoprotein encoded by the CD274 gene [19]. PD-L1 has limited expression on a wide variety of normal cells, including B cells, vascular endothelial cells, epithelial cells, macrophages, and myeloid dendritic cells [7,20,21]. However, cancer cells may possess elevated levels of PD-L1, which indicates the significance of inhibiting this ligand. In a previous study, it was noted that BC cells arising from epithelial carcinoma expressed low levels of PD-L1. The opposite was found in its counterpart, which arises from mesenchymal carcinoma cell models that demonstrating high levels of PD-L1 [22].
It was also known from previous studies using BC cell lines that polyphenols have the potential to impair BC metastasis through numerous mechanisms such as activating the tissue inhibitors of metalloproteinases (TIMPs) expression while inhibiting the matrix metalloproteinase (MMPs) expression [23,24,25], interfering with various signaling pathways, including phosphoinositide 3-kinases/protein kinase B/mammalian target of rapamycin (PI3Ks/AKT/mTOR) [26,27], mitogen-activated protein kinase (MAPK) [28,29], Vascular endothelial growth factor (VEGF) [30], nuclear factor kappa light chain enhancer of activated B cells (NF-κB) [31,32,33] pathways, and modulating EMT process. Extensive studies have shown the impact of different polyphenols on EMT signaling pathways [34,35]. However, meager studies have examined polyphenols’ role in inhibiting PD-L1 to modulate breast cancer (BC) cells’ dissemination and metastasis. Therefore, in this review, we emphasized the polyphenol ability to inhibit EMT and PD-L1 activation to identify new options targeting BC metastasis.

2. PD-1/PD-L1 Checkpoint in Cancer

Cancer cells have direct mechanisms to suppress anticancer immune signaling. However, another indirect mechanism was also protecting the tumor from immune cell-lined death [3]. This mechanism is orchestrated by the CD28 family of receptors that include the PD-1 receptor, in addition to CD28, cytotoxic T-lymphocyte–associated antigen 4 (CTLA-4), inducible co-stimulator (ICOS), and B- and T-lymphocyte attenuator (BTLA) receptors [36,37,38,39]. Normally, the surface protein PD-1 is expressed on various cells, including monocytes, T cells, B cells, dendritic cells (DCs), and natural killer (NK) cells, and its persistent expression is speculated to maintain the functional silence of T cells through delivering inhibitory signals [40]. The bond between PD-1 and its two ligands PD-L1 and PD-L2 generates either a co-stimulatory immunological synapse or inhibitory signals that inhibit T cell response [41]. Although PD-L2 possesses a higher affinity to PD-1 than its counterpart PD-L1, its relatively low expression leads to non-significant interaction with PD-1, which assigning PD-L1 as the primary contributor of PD-1’ suppressive function [17,19,42]. Hence, PD-1/PD-L1 binding is the crucial mechanism in sustaining the immune-suppressive cancer microenvironment. Although these two proteins can also be expressed under normal physiologic conditions, PD-1 and PD-L1 are considered markers of a compromised immune stimulation as their expression is an indicator of T cell dysfunction [43]. Thus, as the main function, their interaction inhibits cytokine production and T cell activation to retain a consistent immune response [19,44].
In cancer cells, the transcription upregulation of PD-L1 is influenced by various elements. Some of them are summarized in Figure 1. Many cytokines were found to induce PD-L1 expression; however, interferon-gamma (IFN-γ) is the main stimulator along with its IFN-γ and toll-like receptor (TLR) ligands [45,46], which also impair the immunity of effector tumor cells [47]. The cytokine IFN-γ is secreted by various types of cells such as activated lymphocytes [48], T cells [49], B cells [50], macrophages [51], monocytes [52], and dendritic cells [53]. As an immunomodulatory agent, IFN-γ acts as a critical coordinator of the immune response with an anticancer effect [54]. A previous study suggested the close association between the loss of IFN-γ pathway genes—Janus kinases (JAK)1 and JAK2—and the increased resistance to PD-1 blockade immunotherapy [55]. Also, it was shown that the prolonged signaling of IFN-γ coordinates the resistance to immune checkpoint blockade, both PD-L1-dependent and independent [56].

3. Oncogenic Signaling Pathways Regulating PD-L1 Expression

3.1. MAPK Pathway

The signaling pathway MAPK—also known as extracellular signal-regulated kinases (ERK) and includes rat sarcoma (Ras), rapidly accelerated fibrosarcoma (Raf), mitogen-activated protein kinase kinase (MEK)-MAPK proteins—is a crucial regulator for vital cellular functions such as cell survival, proliferation, and apoptosis [57]. However, aberrant activation of this pathway is detected in about 50% of cancer patients and was associated with cancer initiation and progression [58]. Also, in triple-negative breast cancer (TNBC) patients, MAPK pathway activation endorses immune evasion, leading to chemotherapeutic drug resistance and poor survival rate [59,60]. The MAPK pathway has been demonstrated to control PD-L1 expression in many cancer cells [61]. Remarkably, in TNBC cells, inhibition of this signaling pathway was found to upregulate IFN-γ–induced PD-L1 expression, both in vivo and in vitro studies, whereas inhibiting MAPK and PD-1/PD-L1 was found to synergize the immune checkpoint inhibitors [62]. On the contrary, in BC cells, the interaction of PD-L1/PD-1 stimulates phosphorylation of MAPK, leading to the activation of MAPK pathways and increases the expression of multidrug resistance protein 1 (MDR1) (also known as permeability glycoprotein, P-gp) [63]. Indeed, the MDRI protein is a member of the adenosine triphosphate (ATP)-binding cassette transporter protein superfamily encoded by the ATP binding cassette subfamily B member 1 (ABCB1) gene [64]. In normal tissues, MDRI is usually disseminated to protect the susceptible organs from toxic substances. However, in multidrug-resistant cancer cells, MDRI is upregulated as a challenging mechanism to decrease these drugs’ intracellular concentration. PD-L1 upregulation is closely associated with MDR1 expression in BC cells, and it is mediated by the activation of PI3K/AKT and MAPK signaling pathways [65].

3.2. PI3K/PTEN/Akt/mTOR Pathway

Various signaling pathways are involved in IFN-γ-mediated PD-L1 induction [66,67,68,69,70]. However, the process is mainly controlled by the loss of phosphatase and tensin homolog (PTEN) tumor suppressor protein and the consequential oncogenic activation of PI3Ks/AKT/mTOR) pathway [71,72]. The fact that interpreted the decrease in PD-L1 expression after using the AKT inhibitors [73]. In BC, abnormalities in the PI3K/AKT/mTOR pathway are the most frequent genomic defects that affect immune surveillance through the regulation of PD-L1. In TNBC, PTEN loss is associated with estrogen receptor (ER)/progesterone receptor (PR)–negative BC cells [74] and explains the increase of PD-L1 in their MDA-MB-157 cell line model. Also, in the MDA-MB-231 cell line, PTEN knockdown resulted in more significant upregulation in PD-L1 expression than the addition of IFN-γ, the common inducer of PD-L1 expression.

4. Transcriptional Control of PD-L1 Expression

4.1. The JAK/STAT Pathway

In TNBC, the activation of the signaling pathway JAK/STAT is proportional to the phosphorylated signal transducer and activator of transcription 1/3 (pSTAT1/3), the key transcription factors that significantly regulate cancer cell survival, proliferation, invasiveness, metastasis, and immunosurveillance [75,76,77,78,79,80]. Notably, STATs modify the immune response through various mechanisms, including regulation of PD-L1 expression [18]—when binding to PD-L1 promoter—as indicated by the abolished PD-L1 expression upon their silencing [81]. Moreover, sole inhibition of STAT1 or STAT3 induces a partial downregulation in PD-L1 expression, while a complete downregulation was achieved upon combined inhibition of these transcription factors [82]. Thus, inhibition of JAK/STAT signaling could be a promising therapeutic target in TNBC [83,84].

4.2. Hypoxia-Inducible Factor 1α (HIF-1α)

The hypoxic feature is well known in BC and other types of cancer as an adaptive mechanism in the reduced oxygen microenvironment. In response to hypoxia, the activated HIF-1α and HIF-2α [85,86,87] lead to poor prognosis and antiestrogen resistance in BC [88]. Once binding to its hypoxia response elements (HRE) promoter, HIF-1α stimulates the transcription of PD-L1 [89]. Indeed, previous studies revealed the co-existence of upregulated HIF-1α, increased PD-L1 expression, and the attenuation of T-cell function [90,91,92]. In TNBC in-vivo model, the PD-L1 expression level was serving as a biomarker in detecting the level of hypoxia [93]. This finding has advocated inhibitors of HIF-1α/PD-1/PD-L1 as a potential therapeutic target to combat the immune suppression behavior of tumors [94,95,96,97].

4.3. NF-ƙB Pathway

The transcriptional factor NF-ƙB has been previously shown to promote and mediate inflammation-cancer pathways, inhibit apoptosis, and enhance tumorigenesis and cancer immune evasion [98,99]. Also, NF-ƙB has the potential to induce PD-L1, either directly through binding to PD-L1 promotor or indirectly, by enhancing the stability of its protein that supports the tumor immune evasion [100]. Notably, the involvement of NF-ƙB in IFN-γ-induced PD-L1 expression has been evidenced by PD-L1 repression in the presence of NF-ƙB inhibitors [98]. Another mechanism that has been previously found to prevent PD-L1 degradation is TNF-α-mediated NF-ƙB activation through enhancing the fifth element of the constitutive photomorphogenesis 9 (COP9) signalosome5 (CSN5) protein [101]. Furthermore, a study on BC demonstrated the ability of natural compounds to induce PD-L1 expression through histone deacetylase 3 (HDAC3)/p300)-mediated NF-ƙB signaling pathway [102]. NF-ƙB-mediated PD-L1 induction is also impacted by aberrant expression of some oncogenes such as B cell lymphoma 3 (Bcl3) [103] and Mucin1 (MUC1) [99] that integrate a variety of signaling pathways. Indeed, Bcl3 promotes IFN-γ-stimulated PD-L1 expression through NF-ƙB p65 acetylation [104]. In TNBC cells, PD-L1 upregulation was revealed to be MUC1-dependent [40]; meanwhile, MUC1 drives PD-L1 overexpression involves MYC proto-oncogene, bHLH transcription factor (Myc), and NF-ƙB-dependent mechanisms [99].
In immune and cancer cells, the Toll-like receptor (TLR)-mediated signaling pathway is a well-known mechanism that upregulates PD-L1 [67] through increasing NF-ƙB activation, which in turn leads to PD-L1 upregulation [105]. Meanwhile, IFNs have been demonstrated to regulate PD-L1 expression on both tumor and non-tumor cells; IFN-γ stands out as the most inducer [45,69]. Also, IFN-γ stimulates nuclear translocation of NF-ƙB signaling pathway, thus upregulating PD-L1’s promoter activity [106].

5. PD-L1 Expression in Breast Cancer

It has become evident that overexpression of PD-L1 protects malignant cells from immune detection in various types of cancers, including BC, an event that leads to the increase of tumor aggressiveness and poor disease prognosis [107,108,109,110,111,112,113,114]. In the highly metastatic TNBC subtype, PD-L1 expression is strongly linked to various adverse aspects of aggressiveness, such as advanced cancer grade, lack of ER, and increased infiltration with T-regulatory cells [73,114]. Notably, the significant overexpression of PD-L1 in MDA-MB-231, the typical TNBC cell model [73], leads to tumor escape from the immune system and the worse outcome [9,10]. PD-L1 expression is induced upon EMT activation, and it is closely associated with the mesenchymal features, as clearly manifested in the claudin-low BC, the subtype that is highly associated with poor prognosis and enriched in EMT features [10,115]. More investigations demonstrated the mechanism underlying EMT-mediated PD-L1 upregulation and attributed this aggressive mechanism to the surface markers as shown by PD-L1 concomitant parallel association with CD44 upregulation and CD24 downregulation [10]. In TNBC cells, the PTEN/PI3K pathway is significant in regulating PD-L1 expression. As mentioned earlier, PTEN loss is a mechanism that promotes PD-L1 expression through the PI3K/AKT/mTOR pathway, and it is correlated with ER/PR–negative tumor [74,116]. Moreover, glycosylation inhibitors were significantly linked to the repressed PD-L1 expression in BC cells [117] and momentous purge of TNBC cells [118]. For example, in TNBC cells, the upregulation of PD-L1 expression and the activation of NF-ƙB is transmembrane glycoprotein MUC1-dependent [99]. MUC1 is overexpressed in various types of cancer and implicated in multiple signaling pathways that enhancing cancer growth and maintenance [119]. The enforced TGF-β1 upregulation was found to induce PD-L1 expression in normal breast cells. However, the mechanism was mainly driven by the induced EMT, not TGF-β1 itself [10]. Overall, these findings support the rationale for applying therapeutic approaches targeting the PD-1/PD-L1 via PI3K pathway in TNBC metastatic subtype [73].

6. Epithelial-to-Mesenchymal Transition (EMT) Markers Mediating PD-L1 Induction in Breast Cancer

It is well known that the pro-metastatic phase within the tumor microenvironment is linked to inflammation. Indeed, the host’s tumor-infiltrating immune cells secrete various types of cytokines and chemokines such as TGF-β in an endeavor to fight cancer [120]. Unfortunately, this mechanism provokes the EMT process and promotes cancer cell invasion and migration [121,122]. Contrary to the common belief, many studies using in vivo and in vitro BC models have evidenced upregulated expression of PD-L1 along with normal PTEN and the lack of the INF-γ [10]. Hence, the existence of another mechanism underlying the regulation of PD-L1 in BC was suggested [10]. Indeed, in some types of cancer, a poor prognosis was found in PD-L1(+)/EMT (+) compared with the PDL1(+)/EMT (−) one, which indicates the importance of targeting EMT to limit cancer migration and prognosis [123]. A recent study has summarized the involvement of different molecules such as MUC1, TGF-β, and NF-ƙB [10,99] in EMT-mediated PD-L1 upregulation in BC [115]. Other oncological studies cited the opposite, and they revealed the importance of PD-L1 signaling in maintaining EMT status [10,124,125,126] (Figure 2). Nevertheless, both mechanisms will eventually lead to tumor immune escape [10,115]. Thus, EMT status was considered a co-biomarker with PD-L1 to speculate the prognosis and the likelihood of response to PD-1/PD-L1 checkpoint blockade [115].
Meanwhile, PD-L1 can be stimulated directly; another profound indirect mechanism underlying EMT-mediated PD-L1 expression was demonstrated [10]. A significantly upregulated level of PD-L1 was attributed to the tumor cell surface markers as mentioned above [10]. Indeed, EMT -mediated PD-L1 expression is highly suggested in the claudin-low subtype of TNBC, characterized with high EMT features [10], while decisively downregulated PD-L1 reversed EMT process, which strongly suggests the important role for PD-L1 targeted therapy in this subset of the disease [10]. Also, TGF-β cytokine was the primary inducer of EMT [120], which augments the expression of PD-L1 in BC cells [10]. Interestingly, the upregulation of PD-L1 in BC was attributed to EMT activation but not TGF-β itself [10].
The role of EMT transcription factor (EMT-TFs) in controlling PD-L1 expression was also revealed. It was previously suggested that EMT-TFs, including Zeb, Twist, and SNAIL family proteins, mediate EMT regulation and tumor progression [4,127] and bridge the link between inflammation and cancer [127,128]. Moreover, higher expression of Zeb1, Snail, N-cadherin, and Vimentin and low expression of E-cadherin were closely correlated with the upregulated level of PD-L1 [115,129]. In TNBC cells, various transduction signaling pathways were involved in EMT-mediated PD-L1 expression, with the MAPK pathway the most crucial one [130,131]. Various examples also were reported for proteins involved in the EMT process. The overexpression of the insulin-like growth factor 1 receptor (IGF1R) and focal adhesion kinase (FAK) signaling was crucial for EMT and metastasis [130]. These signaling pathways caused an enhancement of the mesenchymal markers’ expression, Zeb1, Snail1, and Vimentin, while a reduction of the epithelial markers claudin-1, E-cadherin, and Zonula occludens-1 (ZO-1) was found. Similarly, adapter molecule Crk (Crk) protein is implicated in various signaling pathways regulating EMT and EMT-stimulate PD-L1. The Crk mechanism for enhancing cancer metastasis was achieved by upregulating the expression of Zeb1 and N-cadherin and repressing E-cadherin levels [129,132,133]. Indeed, targeting signaling pathways and cytokine-induced EMT could hold promises in inhibiting BC cell dissemination and metastasis [134,135,136,137,138,139,140].
On the other hand, a growing body of literature has suggested the implication of upregulated EMT in increasing drug resistance and cancer progression. This resistance behavior was exhibited in patients diagnosed with solid cancers, including BC, presenting a considerable challenge [15,141]. Indeed, EMT was associated with the upregulated expression of many (ATP)—binding cassette (ABC) transporters that ultimately lead to multidrug resistance [120,142,143]. Hence, combining therapeutic agents against EMT-TFs was a promising approach to overcoming these tumors’ resistance mechanisms [144].

7. Breast Cancer Treatment

For decades, cytotoxic chemotherapeutic drugs were the standard medical treatment for BC patients [145]. Various target—directed approaches have evolved to treat and manage the heterogenous BC characterized by diverse molecular subtypes and stages [145]. Chemotherapeutics drugs with cytotoxic effects—doxorubicin and paclitaxel—are typically applied for patients with metastasized BC. Other treatments, including gemcitabine, cisplatin derivatives, 5-fluorouracil, or vinorelbine, are also used. On the other hand, combined treatments with chemotherapy drugs are considered a promising approach for enhancing BC therapy outcomes [146]. For instance, the estrogen antagonists—tamoxifen or fulvestrant—combined with the aromatase inhibitors—anastrozole, letrozole, and exemestane—are used in the hormone-dependent (ER+/PR+) BC cells [147]. Also, bevacizumab, a monoclonal antibody therapeutic, targets vascular endothelial growth factor receptor (VEGFR), hindering the angiogenesis pathway [148,149,150]. Another monoclonal antibody, trastuzumab, could be used in patients overexpressing the HER-2 receptor, combined with therapeutic hormonal drugs such as the selective HER-2 pathway inhibitors lapatinib [147,149,150]. Moreover, various emerging drugs have shown the potential to overcome hormonal therapy resistance when combined with hormonal drugs [145]. These included the cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitors such as abemaciclib, palbociclib, and ribociclib [151]—which impact cell cycle progression—and inhibitors of PI3K/AKT/mTOR pathway such as buparlisib, pictilisib, pilaralisib, and voxtalisib [152,153]. On the contrary, TNBC—the most aggressive cells with abolished biomarkers expression—the classical chemotherapeutic drugs, such as taxanes, anthracyclines, and platinum agents, remained the exclusive therapeutic option [148,149,150], and they are currently used with/without the monoclonal antibody against VEGF bevacizumab [154]. Recently, new targeted drugs were introduced and still undergo clinical trials for optimizing BC therapeutic outcome, including poly adenosine diphosphate (ADP)-ribose polymerase (PARP) inhibitors—olaparib, talazoparib, veliparib, niraparib, and rucaparib—for those with mutated breast cancer type 1/2 susceptibility protein (BRCA1/2) [155,156,157], the antibody-drug conjugate Glembatumumab vedotin, the androgen receptor inhibitor bicalutamide, and the anti-PD-1 monoclonal antibody pembrolizumab.

8. Current Breast Cancer Immunotherapeutic Strategies

While the treatment regimens of BC have greatly improved in recent years, the disease’s emerging subtypes raised a significant challenge that classified BC as the most frequent cancer type affecting women [158]. TNBC cells—a BC subtype lacking the expression of ER, PR, and the overexpression of the humane epidermal receptor (HER)—were further classified into basal-like and claudin-low subtypes [159,160,161]. The lack of hormonal receptors in TNBC urged the need for developing new therapeutic approaches targeting these subtypes [10,162]. Hence, cancer immunotherapy is considered a narrative approach in different types of cancer [13,163]. Various immune checkpoint blockade, mainly PD-1 and its ligand, PD-L1—the most prognostic biomarker—and CTLA-4 inhibitors, have been established in the clinics [164,165]. Fortunately, PD-1 and PD-L1 inhibitors have been promising in treating various kinds of cancer, including BC [166]. From 2011-2017 exhibited the emergence of valuable drugs that inhibit PD-1 (Pembrolizumab and Nivolumab) and PD-L1 (Atezolizumab, Avelumab, and Durvalumab), as well as the monoclonal antibody Ipilimumab that targeting CTLA4 [167,168].
Although the PD-1 and PD-L1 blockade immunotherapy has achieved an incredible clinical outcome in some subsets of BC patients [169], so far, PD-1 blockade works only in PD-1(+)/PD-L(+) but not in PD-1(−) patients [8,18,170]. Meanwhile, not all PD-L1-expressing cancer patients responded to PD-1/PDL1 inhibitors; PD-L1(−) tumors may respond to these agents [171]. Most importantly, using the immunotherapeutic candidates —targeting PD-L1/PD-1 pathway—was found to enhance other antitumor treatment approaches [172]. For instance, in BC tumor, a solely administered doxorubicin, the conventional chemotherapy drug, attenuated PD-L1’s cell surface expression and exhibited apoptotic effect; however, it increased PD-L1 nuclear expression [172]. Furthermore, the co-existence of doxorubicin and PI3K/AKT pathway inhibitor abolished the doxorubicin-induced nuclear up-regulation of PD-L1, suggesting the significant role of the PI3K/AKT pathway in the nuclear upregulation of PD-L1 in BC cells [172,173].

9. Polyphenols and Cancer

Recently, special attention has been directed to the polyphenols found in a wide variety of edible plants, including vegetables, fruits, soy products, in addition to cereal, wine, and tea [174,175]. Myriads of epidemiological studies have cited the uses of polyphenol in treating a diversity of health issues, including infection [176,177], inflammation [178], oxidative stress [179], bone diseases [180], cardiovascular disease [181], and cancer [182]. In cancer research, extensive literature has correlated the consumption of polyphenol-rich food and the lower risk of cancers [183,184,185,186,187,188,189,190]. It has been suggested that polyphenols may inhibit tumors at various stages, including initiation, relapse, progression, and metastasis to other organs [191,192,193]. The well-known antioxidant activities of the polyphenols were found to induce a chemopreventive effect [192], together with their anticancer effect that conveys antioxidant-independent mechanisms [192,194].
Polyphenols have demonstrated a vital role in modulating various signaling pathways and modifying proteins-mediating cancer progression [34,35,195]. Indeed, polyphenols exhibited anti-oncogenic effects in the NF-ƙB transcription factors, Wnt/β-catenin, peroxisome proliferator activator receptor-gamma (PPAR-γ), STAT3, nuclear factor erythroid 2 (Nrf2), sonic hedgehog (Shh), activator protein-1 (AP-1), growth factors receptors (epidermal growth factor receptor, EGFR; Erb-B2 receptor tyrosine kinase 2, ErbB2, VEGFR; insulin like growth factor1 receptor, IGF1-R). Polyphenols also have been shown prospectively to reverse EMT-underlying tumor metastasis by modifying miRNA’s expression [6]. Moreover, these compounds revealed anti-inflammatory effects through modulating the pro-inflammatory mediators, tumor necrosis factor-α (TNF-α), interleukins (ILs), Cyclooxygenase (COX)-2, 5-Lipoxygenases (LOX), and various protein Kinases (PI3K, mTOR, AMPK, Bcr-abl, and Ras/Raf) [34,35,195,196].
Although many dietary polyphenolic compounds have shown various pharmacological effects, there are still challenges that should be considered for many other polyphenols to be effective in clinical practices [197]. When taking orally—since the mouth is the most common route of administration for small molecule drugs and nutraceuticals [198]—these polyphenols might face many obstacles before reaching their site of action. The challenges may include poor aqueous solubility, weak oral absorption, low bioavailability, or fast systemic elimination [197]. To manage the pharmacokinetics profile of such perplexing polyphenols, various formulations could be approached. Many developed formulations have already been pharmaceutically applied to manage these barriers, such as nanogels, nanoparticles, nanospheres, liposomes, complexation, micelles, and solid dispersions [199]. Significantly, interactions with other elements found in food and other drugs might be highly anticipated with some polyphenols [6], even though they could be prevented by specialized formulations, avoiding specific food intake, and managing dosage regimens.
Clinical trials in BC patients evidenced the potential of the dietary polyphenolic compounds to increase apoptosis while decreasing various tumor biomarkers [200,201], including steroid hormones [202,203], carcinoembryonic antigen (CEA), VEGF [204], and radiation dermatitis severity score (RDS) [205], in addition to anti-inflammatory effects [206]. On the other side, none of these studies demonstrated the potential of these polyphenols to modulate the immune response in BC patients.
Nevertheless, there is a continuous interest in investigating dietary flavonoids as antitumor immunity agents [207,208]. Here, we summarized the most-studied compounds and highlighted their potential to target PD-L1 in BC cells, either directly or indirectly, through modulating EMT markers-mediating PD-L1 activation. This summary will also provide a closer look at the polyphenols’ most specific studies that could be used combined with the current use of PD-L1 blockade and anti-PD-1 immunotherapy to enhance their efficacy against BC.

9.1. Curcumin

Curcumin is a natural polyphenol compound extracted from the turmeric roots and used for a long time as a traditional medicine in the Ayurveda [209,210,211]. This compound has demonstrated various pharmacological properties, including antioxidant [212], anti-inflammatory [213], antimicrobial [214], immunomodulatory [215], and hepatoprotective [216] properties. Furthermore, curcumin has shown anti-metastatic effects through targeting various intracellular signaling pathways implicated in PD-L1 upregulation [217], including NF-ƙB [218], MAPK [219], Wnt/β-catenin [220], PI3K/Akt/mTOR [221], hedgehog [222], Notch [223], and block IκB kinase (IKK) activity that consequently inhibits NF-ƙB signaling pathway [224,225] (Figure 3).
In BC, curcumin also was a potent agent in targeting various genes mediating the EMT process. For instance, it targets H19 long non-coding RNA. Upregulated H19 promotes EMT through increasing vimentin expression and repressing E-cadherin expression, and more importantly, contributes to tamoxifen-resistant tumors [226]. The highly expressed H19 also plays a crucial role in various cellular events, including proliferation, chemoresistance, endocrine resistance, migration, invasiveness, and metastasis [227,228]. Furthermore, recent cohort studies have evidenced the close association between the long non-coding RNA and PD-L1 expression [229,230]. Thus, targeting this gene was considered a key in PD-L1 inhibition [231].
Upon curcumin exposure, other target proteins associated with EMT and metastasis—slug, β-catenin, receptor tyrosine kinase (RTK, aka; AXL), CD24, and vimentin—were repressed in the MDA-MB-231 cells model of TNBC [222,232,233]. These proteins are upregulated in both human and murine BC [234,235,236]. Moreover, curcumin was also found to impact TGF-β and PI3K/AKT signaling pathways regulating doxorubicin-stimulated EMT activation [237,238]. The intrinsic β-catenin is a crucial oncogenic protein in driving cancer initiation and progression through modulating the transcription of many genes such as slug -mediating BC metastasis. Thus, the inhibition of β-catenin, hindering the trans-stimulation of slug and, consequently, restores E-cadherin expression of epithelial phenotype [239,240]. The oncogene β-catenin is a well-known regulator of PD-L1–mediated immunosuppression as revealed by the significant abolition in PD-L1 expression upon reducing β-catenin. On the contrary, upregulated β-catenin was positively correlated with the increased level of PDL1′s protein expression [8,241]. Also, upon inhibiting Axl kinase, a significant decrease in tumor growth was found in the mouse models, the effect that was further potentiated when combined with PD-1 blockade [236]. Pharmacological repression of Axl activity was found to decrease the mRNA expressions of PD-L1, the finding that revealed the implication of Axl in regulating PD-L1 protein expression [242]. Moreover, the cytokine TGF-β—as another mediator in EMT development—is involved in many cellular events and upregulating the expression of PD-L1 [243]. TGF-β has a tumor promoter role in the advanced stages of the disease as it enhances EMT and metastasis [244,245,246]. An interesting report on combining TGF-β inhibitors with PD-1/PD-L1 immune checkpoint blockade has revealed a tumor regression [247]. Therefore, the pharmacological modulation of β-catenin, Axl, and TGF-β are considered putative trends in cancer therapy [248]. This information strongly suggests the pivotal role of curcumin in inhibiting PD-L1 directly and indirectly through deactivating EMT markers in BC patients and ultimately limiting metastasis.

9.2. Apigenin

The flavone apigenin is found in various fruits, vegetables, and herbs, such as parsley, onions, grapefruit, oranges, and chamomile [249,250]. Apigenin was previously found to demonstrate various biological activities, including antioxidant [251], anti-inflammatory [252], antibacterial, antiviral [253], and anticancer effects [254]. Fortunately, apigenin is considered a safe compound for normal healthy cells [255]. As an anticancer agent, low concentrations of apigenin were found to inhibit the proliferation, while a significant apoptotic effect was induced at higher concentrations of the compound [255,256,257,258,259]. Moreover, apigenin’s anti-metastatic effect has been revealed in several cancers, including BC [260,261,262]. The compound showed immunomodulatory properties by targeting the PD-1/PD-L1 checkpoint as a promising immunotherapy candidate [17]. The ability of apigenin to inhibit PD-L1 was also investigated in human and mouse mammary carcinoma cells.
Apigenin was found to inhibit IFN-γ-induced PD-L1 upregulation in MDA-MB-468 TNBC cells, HER2+SK-BR-3, human mammary epithelial cells, and 4T1mouse mammary carcinoma cells. In MDA-MB-468 and 4T1 cells, the repression of PD-L1 level was associated with reduced phosphorylation of STAT1 [263] (Figure 4). Luteolin, the major metabolite of apigenin, was also found to inhibit IFN-γ-induced PD-L1 expression in MDA-MB-468 cells. In the MDA-MB-231 TNBC cell, apigenin did not repress PD-L1 expression, and its anti-metastatic effect was not directed to the EMT markers, Vimentin, or N-cadherin. Instead, the compound repressed IL-6-mediated EMT signal-linked N-cadherin expression [264]. Certainly, the positive association between IL-6 and EMT in the tumor microenvironment has been proven in various types of cancer, including BC, leading to cell migration and invasiveness [265,266,267,268]. It is collectively suggested that more investigations are needed to characterize the effects of apigenin on EMT and PD-L1 inhibition as a safe immunotherapeutic candidate drug for specific subsets of BC disease.

9.3. Hesperidin

Hesperidin is a flavonoid found in various Rutaceae family members [269] and was used in China as traditional herbal medicine. In pharmacological studies, the compound exhibited various anticancer effects with anti-proliferative [270], anti-inflammatory [271], and apoptotic [269,272] properties. Previously reported research indicated the safety of the compound against normal cells. Remarkably, the properties of hesperidin endorsed the compound’s use as an anticancer candidate against BC and other cancer types. The significant effect of hesperidin in inhibiting the expression of EMT markers was recently exposed [273]. Also, one report investigating the effect of hesperidin in MDA-MB-231 cells has cited its ability to inhibit the levels of PD-L1 at both the protein and the transcriptional level through inhibiting PI3K/Akt and NF-ƙB signaling pathway [274] (Figure 5). These previous findings support our hypothesis that this polyphenol compound has the potential to ultimately inhibit PD-L1, directly or indirectly, through impacting EMT markers. Still, more emphasis on hesperidin and its mechanism against EMT markers and PD-1/PD-L1 checkpoints are highly encouraged.

9.4. Resveratrol

Resveratrol is a polyphenol component found in peanuts and grapes as well as other plants [275]. The compound has shown significant biological activities and may hold promises as a therapeutic agent against cancer [276]. Notably, resveratrol showed a potency to inhibit various tumors’ initiation and progress [276,277]. The impact of this polyphenol in BC is multidisciplinary as extensive studies revealed resveratrol’s ability to utilize different mechanisms in targeting epigenetic response, cell proliferation, apoptosis, EMT/metastasis, and most appreciably, increased sensitivity to chemotherapy [278]. In BC, resveratrol mediates cellular aging and inhibits the EMT process by inducing the tumor suppressor Rad9-dependent mechanism [279]. The adaptor protein Rad9 is crucial for the DNA damage response (DDR) protein [280]. A reduction of Rad9 expression was detected in the highly invasive MDA-MB-231 cells [279]. Most importantly, Rad9 protein has shown a selective mechanism in controlling genes linked to EMT, such as p21 [281], Neil1 [282], and slug [280]. Also, the compound inhibited cell migration through PI3K/Akt and Wnt/β-catenin signaling pathways [283] (Figure 6)—the pivotal elements in regulating PD-L1 protein expression—in BC cells [284]. Remarkably, the compound demonstrated a potential to overcome chemotherapy resistance in BC. Resveratrol sensitized the cells to tamoxifen through TGF-β/Smad-driven EMT [285] and promotes cell sensitization to doxorubicin by inhibiting EMT and modulating SIRT1/β-catenin signaling pathway [285,286]. Similarly, recent studies using MDA-MB-231 cells indicated resveratrol’s ability to inhibit cell migration by reversing TGF-β1-induced EMT [140] and inducing a significant suppression of PD-L1 through targeting PD-L1 glycosylation enzymes [287]. Therefore, resveratrol’s unique properties should be highlighted in the field of BC immunotherapy and drug resistance management.

9.5. Sativan

The compound (−)-sativan is a natural isoflavone found in Spatholobus suberectus. The traditional Chinese herb Spatholobus suberectus is commonly used in China for treating many diseases, including anemia, rheumatism, and menoxenia [288]. This herb has been found to possess antioxidant and anti-inflammatory properties [289]. Several recent studies indicated the anticancer effects of Spatholobus suberectus in BC with the potential to trigger apoptosis, cell cycle arrest, lactate dehydrogenase inhibition [290], and preventing cancer cell migration through the MAPK PI3K/AKT pathway [291] (Figure 7). Further, a recent study has demonstrated the potential of the compound sativan to induce an anticancer effect in TNBC cells through inhibiting both of EMT process and PD-L1 mRNA expression [292]. Sativan impacted various oncogenic transcription regulators mediated EMT activation [292] and showed the ability to stimulate E-cadherin while decreasing N-cadherin and vimentin. Moreover, Snail and slug were significantly inhibited by the compound [292]. As with any other novel therapeutic agent, further investigations should be considered to shed light on the possible therapeutic mechanisms that can be disclosed for the BC immunotherapy approach.

10. Conclusions

Cancer metastasis to vital organs is the leading cause of poor prognosis and cancer-related death, and it is accomplished by the immune evasion strategy. Indeed, tumor suppresses the anticancer immune signaling, either directly or indirectly. One mechanism in inhibiting these signals is the upregulation of the CD28 family of the receptor, in particular PD-1. Indeed, the association between PD-1 and its ligand PD-L1 provokes immune inhibitory signals. In BC cells, overexpression of PD-L1 protects malignant cells from immune devastation, and it is strongly linked to tumor aggressiveness, poor prognosis, and drug resistance. In tumors, the EMT defend process drives various aspects of carcinogenesis, metastasis, immunosuppression, and drug resistance. Notably, there is a strong association between activated EMT and PD-L1 expression.
On the other hand, polyphenols have shown a significant effect as elements of anticancer immunity. Indeed, polyphenols can inhibit the PD-L1 expression directly. However, in this review, we highlighted the indirect mechanism of polyphenol in inhibiting EMT-mediate PD-L1 expression through inhibiting the mesenchymal protein and upregulating the epithelial counterpart. Indeed, apigenin and its major metabolite, luteolin, were able to inhibit IFN-γ-induced PD-L1 expression, in addition to repressing IL-6 mediated EMT process. Also, hesperidin was found to impact EMT markers and targeting various signaling pathways such as PI3K/Akt and NF-ƙB signaling pathway. Resveratrol also showed a potential to inhibit the EMT process by stimulating the tumor suppressor Rad9-dependent mechanism, reversing TGF-β1-induced EMT, as well as targeting PD-L1 glycosylation enzymes. Furthermore, sativan has been shown to impact various oncogenic transcription regulators mediated EMT activation through stimulating E-cadherin while inhibiting N-cadherin, Vimentin, Snail, and Slug. In conclusion, having a direct/indirect/or both mechanisms in targeting PD-L1 expression holds promise in limiting metastasis and treating patients suffering from BC disease. Various polyphenolic compounds have been used in BC clinical trials. These compounds have demonstrated promising anticancer effects in patients with various stages of BC. These effects include anti-inflammatory, pro-apoptosis induction, and suppression of various tumor biomarkers such as CEA, VEGF, and RDS. On the other side, few limited studies have proved the potential of these compounds to impact EMT-underlying tumor metastasis through modifying miRNA’s expression. Hence, further comprehensive investigations are suggested to highlight and focus on the potential of these dietary polyphenolics to reverse or inhibit the challenged EMT process.

Author Contributions

Conceptualization, S.S.M.; writing—original draft preparation, S.S.M.; review and editing, S.S.M., N.O.Z., and K.F.A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by NIH grants from the National Institute on Minority Health and Health Disparities (NIMHD), grant number G12 MD007582.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Janssen, L.M.E.; Ramsay, E.E.; Logsdon, C.D.; Overwijk, W.W. The immune system in cancer metastasis: Friend or foe? J. Immunother. Cancer 2017, 5, 79. [Google Scholar] [CrossRef]
  2. Vincent, C.T.; Fuxe, J. EMT, inflammation and metastasis. Semin. Cancer Biol. 2017, 47, 168–169. [Google Scholar] [CrossRef]
  3. Ahmad, S.M.; Borch, T.H.; Hansen, M.; Andersen, M.H. PD-L1-specific T cells. Cancer Immunol. Immunother. 2016, 65, 797–804. [Google Scholar] [CrossRef]
  4. Brabletz, T.; Kalluri, R.; Nieto, M.A.; Weinberg, R.A. EMT in cancer. Nat. Rev. Cancer 2018, 18, 128–134. [Google Scholar] [CrossRef]
  5. Nieto, M.A.; Huang, R.Y.; Jackson, R.A.; Thiery, J.P. EMT: 2016. Cell 2016, 166, 21–45. [Google Scholar] [CrossRef] [Green Version]
  6. Amawi, H.; Ashby, C.R.; Samuel, T.; Peraman, R.; Tiwari, A.K. Polyphenolic Nutrients in Cancer Chemoprevention and Metastasis: Role of the Epithelial-to-Mesenchymal (EMT) Pathway. Nutrients 2017, 9, 911. [Google Scholar] [CrossRef] [Green Version]
  7. Soliman, H.; Khalil, F.; Antonia, S. PD-L1 expression is increased in a subset of basal type breast cancer cells. PLoS ONE 2014, 9, e88557. [Google Scholar] [CrossRef] [Green Version]
  8. Tuo, Z.; Zong, Y.; Li, J.; Xiao, G.; Zhang, F.; Li, G.; Wang, S.; Lv, Y.; Xia, J.; Liu, J. PD-L1 regulation by SDH5 via β-catenin/ZEB1 signaling. Oncoimmunology 2019, 8, 1655361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Afreen, S.; Dermime, S. The immunoinhibitory B7-H1 molecule as a potential target in cancer: Killing many birds with one stone. Hematol. Oncol. Stem Cell Ther. 2014, 7, 1–17. [Google Scholar] [CrossRef] [Green Version]
  10. Alsuliman, A.; Colak, D.; Al-Harazi, O.; Fitwi, H.; Tulbah, A.; Al-Tweigeri, T.; Al-Alwan, M.; Ghebeh, H. Bidirectional crosstalk between PD-L1 expression and epithelial to mesenchymal transition: Significance in claudin-low breast cancer cells. Mol. Cancer 2015, 14, 149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Moreno-Bueno, G.; Portillo, F.; Cano, A. Transcriptional regulation of cell polarity in EMT and cancer. Oncogene 2008, 27, 6958–6969. [Google Scholar] [CrossRef] [Green Version]
  12. Pasquier, J.; Abu-Kaoud, N.; Al Thani, H.; Rafii, A. Epithelial to Mesenchymal Transition in a Clinical Perspective. J. Oncol. 2015, 2015, 792182. [Google Scholar] [CrossRef] [Green Version]
  13. Xu, J.W.; Wang, L.; Cheng, Y.G.; Zhang, G.Y.; Hu, S.Y.; Zhou, B.; Zhan, H.X. Immunotherapy for pancreatic cancer: A long and hopeful journey. Cancer Lett. 2018, 425, 143–151. [Google Scholar] [CrossRef]
  14. Mellman, I.; Coukos, G.; Dranoff, G. Cancer immunotherapy comes of age. Nature 2011, 480, 480–489. [Google Scholar] [CrossRef] [PubMed]
  15. Terry, S.; Savagner, P.; Ortiz-Cuaran, S.; Mahjoubi, L.; Saintigny, P.; Thiery, J.P.; Chouaib, S. New insights into the role of EMT in tumor immune escape. Mol. Oncol. 2017, 11, 824–846. [Google Scholar] [CrossRef] [Green Version]
  16. Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [Green Version]
  17. Topalian, S.L.; Drake, C.G.; Pardoll, D.M. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr. Opin. Immunol. 2012, 24, 207–212. [Google Scholar] [CrossRef] [Green Version]
  18. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [PubMed]
  19. Dong, H.; Zhu, G.; Tamada, K.; Chen, L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat. Med. 1999, 5, 1365–1369. [Google Scholar] [CrossRef] [PubMed]
  20. Blank, C.; Brown, I.; Peterson, A.C.; Spiotto, M.; Iwai, Y.; Honjo, T.; Gajewski, T.F. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res. 2004, 64, 1140–1145. [Google Scholar] [CrossRef] [Green Version]
  21. Butte, M.J.; Peña-Cruz, V.; Kim, M.J.; Freeman, G.J.; Sharpe, A.H. Interaction of human PD-L1 and B7-1. Mol. Immunol. 2008, 45, 3567–3572. [Google Scholar] [CrossRef] [Green Version]
  22. Dongre, A.; Rashidian, M.; Reinhardt, F.; Bagnato, A.; Keckesova, Z.; Ploegh, H.L.; Weinberg, R.A. Epithelial-to-Mesenchymal Transition Contributes to Immunosuppression in Breast Carcinomas. Cancer Res. 2017, 77, 3982–3989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Deb, G.; Thakur, V.S.; Limaye, A.M.; Gupta, S. Epigenetic induction of tissue inhibitor of matrix metalloproteinase-3 by green tea polyphenols in breast cancer cells. Mol. Carcinog. 2015, 54, 485–499. [Google Scholar] [CrossRef]
  24. Hassan, Z.K.; Elamin, M.H.; Daghestani, M.H.; Omer, S.A.; Al-Olayan, E.M.; Elobeid, M.A.; Virk, P.; Mohammed, O.B. Oleuropein induces anti-metastatic effects in breast cancer. Asian Pac. J. Cancer Prev. 2012, 13, 4555–4559. [Google Scholar] [CrossRef] [Green Version]
  25. Lewandowska, U.; Szewczyk, K.; Owczarek, K.; Hrabec, Z.; Podsędek, A.; Koziołkiewicz, M.; Hrabec, E. Flavanols from Japanese quince (Chaenomeles japonica) fruit inhibit human prostate and breast cancer cell line invasiveness and cause favorable changes in Bax/Bcl-2 mRNA ratio. Nutr. Cancer 2013, 65, 273–285. [Google Scholar] [CrossRef]
  26. Zhou, H.; Huang, S. mTOR signaling in cancer cell motility and tumor metastasis. Crit. Rev. Eukaryot. Gene Expr. 2010, 20, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Dihlmann, S.; Kloor, M.; Fallsehr, C.; von Knebel Doeberitz, M. Regulation of AKT1 expression by beta-catenin/Tcf/Lef signaling in colorectal cancer cells. Carcinogenesis 2005, 26, 1503–1512. [Google Scholar] [CrossRef]
  28. Tang, F.Y.; Chiang, E.P.; Sun, Y.C. Resveratrol inhibits heregulin-beta1-mediated matrix metalloproteinase-9 expression and cell invasion in human breast cancer cells. J. Nutr. Biochem. 2008, 19, 287–294. [Google Scholar] [CrossRef]
  29. Wang, L.; Ling, Y.; Chen, Y.; Li, C.L.; Feng, F.; You, Q.D.; Lu, N.; Guo, Q.L. Flavonoid baicalein suppresses adhesion, migration and invasion of MDA-MB-231 human breast cancer cells. Cancer Lett. 2010, 297, 42–48. [Google Scholar] [CrossRef] [PubMed]
  30. Song, M.; Ramaswamy, S.; Ramachandran, S.; Flowers, L.C.; Horowitz, I.R.; Rock, J.A.; Parthasarathy, S. Angiogenic role for glycodelin in tumorigenesis. Proc. Natl. Acad. Sci. USA 2001, 98, 9265–9270. [Google Scholar] [CrossRef] [Green Version]
  31. Yeap, S.K.; Abu, N.; Mohamad, N.E.; Beh, B.K.; Ho, W.Y.; Ebrahimi, S.; Yusof, H.M.; Ky, H.; Tan, S.W.; Alitheen, N.B. Chemopreventive and immunomodulatory effects of Murraya koenigii aqueous extract on 4T1 breast cancer cell-challenged mice. BMC Complement. Altern. Med. 2015, 15, 306. [Google Scholar] [CrossRef] [Green Version]
  32. Farhangi, B.; Alizadeh, A.M.; Khodayari, H.; Khodayari, S.; Dehghan, M.J.; Khori, V.; Heidarzadeh, A.; Khaniki, M.; Sadeghiezadeh, M.; Najafi, F. Protective effects of dendrosomal curcumin on an animal metastatic breast tumor. Eur. J. Pharmacol. 2015, 758, 188–196. [Google Scholar] [CrossRef]
  33. Bachmeier, B.; Nerlich, A.G.; Iancu, C.M.; Cilli, M.; Schleicher, E.; Vené, R.; Dell’Eva, R.; Jochum, M.; Albini, A.; Pfeffer, U. The chemopreventive polyphenol Curcumin prevents hematogenous breast cancer metastases in immunodeficient mice. Cell. Physiol. Biochem. 2007, 19, 137–152. [Google Scholar] [CrossRef] [Green Version]
  34. Benvenuto, M.; Fantini, M.; Masuelli, L.; De Smaele, E.; Zazzeroni, F.; Tresoldi, I.; Calabrese, G.; Galvano, F.; Modesti, A.; Bei, R. Inhibition of ErbB receptors, Hedgehog and NF-kappaB signaling by polyphenols in cancer. Front. Biosci. 2013, 18, 1290–1310. [Google Scholar] [CrossRef]
  35. Fantini, M.; Benvenuto, M.; Masuelli, L.; Frajese, G.V.; Tresoldi, I.; Modesti, A.; Bei, R. In vitro and in vivo antitumoral effects of combinations of polyphenols, or polyphenols and anticancer drugs: Perspectives on cancer treatment. Int. J. Mol. Sci. 2015, 16, 9236–9282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Ostrand-Rosenberg, S.; Horn, L.A.; Alvarez, J.A. Novel strategies for inhibiting PD-1 pathway-mediated immune suppression while simultaneously delivering activating signals to tumor-reactive T cells. Cancer Immunol. Immunother. 2015, 64, 1287–1293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Durgan, K.; Ali, M.; Warner, P.; Latchman, Y.E. Targeting NKT cells and PD-L1 pathway results in augmented anti-tumor responses in a melanoma model. Cancer Immunol. Immunother. 2011, 60, 547–558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Blank, C.; Mackensen, A. Contribution of the PD-L1/PD-1 pathway to T-cell exhaustion: An update on implications for chronic infections and tumor evasion. Cancer Immunol. Immunother. 2007, 56, 739–745. [Google Scholar] [CrossRef]
  39. Blank, C.; Gajewski, T.F.; Mackensen, A. Interaction of PD-L1 on tumor cells with PD-1 on tumor-specific T cells as a mechanism of immune evasion: Implications for tumor immunotherapy. Cancer Immunol. Immunother. 2005, 54, 307–314. [Google Scholar] [CrossRef]
  40. Wherry, E.J. T cell exhaustion. Nat. Immunol. 2011, 12, 492–499. [Google Scholar] [CrossRef]
  41. Hornig, N.; Reinhardt, K.; Kermer, V.; Kontermann, R.E.; Müller, D. Evaluating combinations of costimulatory antibody-ligand fusion proteins for targeted cancer immunotherapy. Cancer Immunol. Immunother. 2013, 62, 1369–1380. [Google Scholar] [CrossRef] [PubMed]
  42. Freeman, G.J.; Long, A.J.; Iwai, Y.; Bourque, K.; Chernova, T.; Nishimura, H.; Fitz, L.J.; Malenkovich, N.; Okazaki, T.; Byrne, M.C.; et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 2000, 192, 1027–1034. [Google Scholar] [CrossRef] [Green Version]
  43. Xu-Monette, Z.Y.; Zhang, M.; Li, J.; Young, K.H. PD-1/PD-L1 Blockade: Have We Found the Key to Unleash the Antitumor Immune Response? Front. Immunol. 2017, 8, 1597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Sheppard, K.A.; Fitz, L.J.; Lee, J.M.; Benander, C.; George, J.A.; Wooters, J.; Qiu, Y.; Jussif, J.M.; Carter, L.L.; Wood, C.R.; et al. PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3zeta signalosome and downstream signaling to PKCtheta. FEBS Lett. 2004, 574, 37–41. [Google Scholar] [CrossRef] [Green Version]
  45. Dong, H.; Strome, S.E.; Salomao, D.R.; Tamura, H.; Hirano, F.; Flies, D.B.; Roche, P.C.; Lu, J.; Zhu, G.; Tamada, K.; et al. Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nat. Med. 2002, 8, 793–800. [Google Scholar] [CrossRef]
  46. Brown, J.A.; Dorfman, D.M.; Ma, F.R.; Sullivan, E.L.; Munoz, O.; Wood, C.R.; Greenfield, E.A.; Freeman, G.J. Blockade of programmed death-1 ligands on dendritic cells enhances T cell activation and cytokine production. J. Immunol. 2003, 170, 1257–1266. [Google Scholar] [CrossRef] [PubMed]
  47. Abiko, K.; Matsumura, N.; Hamanishi, J.; Horikawa, N.; Murakami, R.; Yamaguchi, K.; Yoshioka, Y.; Baba, T.; Konishi, I.; Mandai, M. IFN-γ from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer. Br. J. Cancer 2015, 112, 1501–1509. [Google Scholar] [CrossRef] [Green Version]
  48. Kasahara, T.; Hooks, J.J.; Dougherty, S.F.; Oppenheim, J.J. Interleukin 2-mediated immune interferon (IFN-gamma) production by human T cells and T cell subsets. J. Immunol. 1983, 130, 1784–1789. [Google Scholar]
  49. Gao, Y.; Yang, W.; Pan, M.; Scully, E.; Girardi, M.; Augenlicht, L.H.; Craft, J.; Yin, Z. Gamma delta T cells provide an early source of interferon gamma in tumor immunity. J. Exp. Med. 2003, 198, 433–442. [Google Scholar] [CrossRef] [Green Version]
  50. Harris, D.P.; Goodrich, S.; Gerth, A.J.; Peng, S.L.; Lund, F.E. Regulation of IFN-gamma production by B effector 1 cells: Essential roles for T-bet and the IFN-gamma receptor. J. Immunol. 2005, 174, 6781–6790. [Google Scholar] [CrossRef] [Green Version]
  51. Robinson, C.M.; O’Dee, D.; Hamilton, T.; Nau, G.J. Cytokines involved in interferon-gamma production by human macrophages. J. Innate Immun. 2010, 2, 56–65. [Google Scholar] [CrossRef]
  52. Kraaij, M.D.; Vereyken, E.J.; Leenen, P.J.; van den Bosch, T.P.; Rezaee, F.; Betjes, M.G.; Baan, C.C.; Rowshani, A.T. Human monocytes produce interferon-gamma upon stimulation with LPS. Cytokine 2014, 67, 7–12. [Google Scholar] [CrossRef] [PubMed]
  53. Ohteki, T.; Fukao, T.; Suzue, K.; Maki, C.; Ito, M.; Nakamura, M.; Koyasu, S. Interleukin 12-dependent interferon gamma production by CD8alpha+ lymphoid dendritic cells. J. Exp. Med. 1999, 189, 1981–1986. [Google Scholar] [CrossRef] [Green Version]
  54. Gresser, I. Biologic effects of interferons. J. Investig. Dermatol 1990, 95, 66s–71s. [Google Scholar] [CrossRef] [Green Version]
  55. Zaretsky, J.M.; Garcia-Diaz, A.; Shin, D.S.; Escuin-Ordinas, H.; Hugo, W.; Hu-Lieskovan, S.; Torrejon, D.Y.; Abril-Rodriguez, G.; Sandoval, S.; Barthly, L.; et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N. Engl. J. Med. 2016, 375, 819–829. [Google Scholar] [CrossRef] [PubMed]
  56. Benci, J.L.; Xu, B.; Qiu, Y.; Wu, T.J.; Dada, H.; Twyman-Saint Victor, C.; Cucolo, L.; Lee, D.S.M.; Pauken, K.E.; Huang, A.C.; et al. Tumor Interferon Signaling Regulates a Multigenic Resistance Program to Immune Checkpoint Blockade. Cell 2016, 167, 1540–1554.e1512. [Google Scholar] [CrossRef] [Green Version]
  57. Kolch, W. Coordinating ERK/MAPK signalling through scaffolds and inhibitors. Nat. Rev. Mol. Cell Biol. 2005, 6, 827–837. [Google Scholar] [CrossRef] [PubMed]
  58. Herrero, A.; Pinto, A.; Colón-Bolea, P.; Casar, B.; Jones, M.; Agudo-Ibáñez, L.; Vidal, R.; Tenbaum, S.P.; Nuciforo, P.; Valdizán, E.M.; et al. Small Molecule Inhibition of ERK Dimerization Prevents Tumorigenesis by RAS-ERK Pathway Oncogenes. Cancer Cell 2015, 28, 170–182. [Google Scholar] [CrossRef] [Green Version]
  59. Bartholomeusz, C.; Gonzalez-Angulo, A.M.; Liu, P.; Hayashi, N.; Lluch, A.; Ferrer-Lozano, J.; Hortobágyi, G.N. High ERK protein expression levels correlate with shorter survival in triple-negative breast cancer patients. Oncologist 2012, 17, 766–774. [Google Scholar] [CrossRef] [Green Version]
  60. McCubrey, J.A.; Steelman, L.S.; Chappell, W.H.; Abrams, S.L.; Wong, E.W.; Chang, F.; Lehmann, B.; Terrian, D.M.; Milella, M.; Tafuri, A.; et al. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim. Biophys. Acta 2007, 1773, 1263–1284. [Google Scholar] [CrossRef] [Green Version]
  61. Zerdes, I.; Matikas, A.; Bergh, J.; Rassidakis, G.Z.; Foukakis, T. Genetic, transcriptional and post-translational regulation of the programmed death protein ligand 1 in cancer: Biology and clinical correlations. Oncogene 2018, 37, 4639–4661. [Google Scholar] [CrossRef] [Green Version]
  62. Loi, S.; Dushyanthen, S.; Beavis, P.A.; Salgado, R.; Denkert, C.; Savas, P.; Combs, S.; Rimm, D.L.; Giltnane, J.M.; Estrada, M.V.; et al. Correction: RAS/MAPK Activation Is Associated with Reduced Tumor-Infiltrating Lymphocytes in Triple-Negative Breast Cancer: Therapeutic Cooperation Between MEK and PD-1/PD-L1 Immune Checkpoint Inhibitors. Clin. Cancer Res. 2019, 25, 1437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Liu, S.; Chen, S.; Yuan, W.; Wang, H.; Chen, K.; Li, D.; Li, D. PD-1/PD-L1 interaction up-regulates MDR1/P-gp expression in breast cancer cells via PI3K/AKT and MAPK/ERK pathways. Oncotarget 2017, 8, 99901–99912. [Google Scholar] [CrossRef] [Green Version]
  64. Dizdarevic, S.; Peters, A.M. Imaging of multidrug resistance in cancer. Cancer Imaging 2011, 11, 1–8. [Google Scholar] [CrossRef] [Green Version]
  65. Kobori, T.; Harada, S.; Nakamoto, K.; Tokuyama, S. Mechanisms of P-glycoprotein alteration during anticancer treatment: Role in the pharmacokinetic and pharmacological effects of various substrate drugs. J. Pharmacol. Sci. 2014, 125, 242–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Lee, S.J.; Jang, B.C.; Lee, S.W.; Yang, Y.I.; Suh, S.I.; Park, Y.M.; Oh, S.; Shin, J.G.; Yao, S.; Chen, L.; et al. Interferon regulatory factor-1 is prerequisite to the constitutive expression and IFN-gamma-induced upregulation of B7-H1 (CD274). FEBS Lett. 2006, 580, 755–762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Liu, J.; Hamrouni, A.; Wolowiec, D.; Coiteux, V.; Kuliczkowski, K.; Hetuin, D.; Saudemont, A.; Quesnel, B. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-{gamma} and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood 2007, 110, 296–304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Hao, Y.; Chapuy, B.; Monti, S.; Sun, H.H.; Rodig, S.J.; Shipp, M.A. Selective JAK2 inhibition specifically decreases Hodgkin lymphoma and mediastinal large B-cell lymphoma growth in vitro and in vivo. Clin. Cancer Res. 2014, 20, 2674–2683. [Google Scholar] [CrossRef] [Green Version]
  69. Garcia-Diaz, A.; Shin, D.S.; Moreno, B.H.; Saco, J.; Escuin-Ordinas, H.; Rodriguez, G.A.; Zaretsky, J.M.; Sun, L.; Hugo, W.; Wang, X.; et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep. 2017, 19, 1189–1201, Erratum in 2019, 29, 3766. [Google Scholar] [CrossRef] [Green Version]
  70. Yamamoto, R.; Nishikori, M.; Tashima, M.; Sakai, T.; Ichinohe, T.; Takaori-Kondo, A.; Ohmori, K.; Uchiyama, T. B7-H1 expression is regulated by MEK/ERK signaling pathway in anaplastic large cell lymphoma and Hodgkin lymphoma. Cancer Sci. 2009, 100, 2093–2100. [Google Scholar] [CrossRef]
  71. Parsa, A.T.; Waldron, J.S.; Panner, A.; Crane, C.A.; Parney, I.F.; Barry, J.J.; Cachola, K.E.; Murray, J.C.; Tihan, T.; Jensen, M.C.; et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat. Med. 2007, 13, 84–88. [Google Scholar] [CrossRef]
  72. Lastwika, K.J.; Wilson, W.; Li, Q.K.; Norris, J.; Xu, H.; Ghazarian, S.R.; Kitagawa, H.; Kawabata, S.; Taube, J.M.; Yao, S.; et al. Control of PD-L1 Expression by Oncogenic Activation of the AKT-mTOR Pathway in Non-Small Cell Lung Cancer. Cancer Res. 2016, 76, 227–238. [Google Scholar] [CrossRef] [Green Version]
  73. Mittendorf, E.A.; Philips, A.V.; Meric-Bernstam, F.; Qiao, N.; Wu, Y.; Harrington, S.; Su, X.; Wang, Y.; Gonzalez-Angulo, A.M.; Akcakanat, A.; et al. PD-L1 expression in triple-negative breast cancer. Cancer Immunol. Res. 2014, 2, 361–370. [Google Scholar] [CrossRef] [Green Version]
  74. Saal, L.H.; Holm, K.; Maurer, M.; Memeo, L.; Su, T.; Wang, X.; Yu, J.S.; Malmström, P.O.; Mansukhani, M.; Enoksson, J.; et al. PIK3CA mutations correlate with hormone receptors, node metastasis, and ERBB2, and are mutually exclusive with PTEN loss in human breast carcinoma. Cancer Res. 2005, 65, 2554–2559. [Google Scholar] [CrossRef] [Green Version]
  75. Marotta, L.L.; Almendro, V.; Marusyk, A.; Shipitsin, M.; Schemme, J.; Walker, S.R.; Bloushtain-Qimron, N.; Kim, J.J.; Choudhury, S.A.; Maruyama, R.; et al. The JAK2/STAT3 signaling pathway is required for growth of CD44⁺CD24⁻ stem cell-like breast cancer cells in human tumors. J. Clin. Investig. 2011, 121, 2723–2735. [Google Scholar] [CrossRef] [PubMed]
  76. Zerdes, I.; Wallerius, M.; Sifakis, E.G.; Wallmann, T.; Betts, S.; Bartish, M.; Tsesmetzis, N.; Tobin, N.P.; Coucoravas, C.; Bergh, J.; et al. STAT3 Activity Promotes Programmed-Death Ligand 1 Expression and Suppresses Immune Responses in Breast Cancer. Cancers 2019, 11, 1479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Frank, D.A. Transcription factor STAT3 as a prognostic marker and therapeutic target in cancer. J. Clin. Oncol. 2013, 31, 4560–4561. [Google Scholar] [CrossRef] [PubMed]
  78. Yu, H.; Jove, R. The STATs of cancer--new molecular targets come of age. Nat. Rev. Cancer 2004, 4, 97–105. [Google Scholar] [CrossRef]
  79. Schindler, C.; Levy, D.E.; Decker, T. JAK-STAT signaling: From interferons to cytokines. J. Biol. Chem. 2007, 282, 20059–20063. [Google Scholar] [CrossRef] [Green Version]
  80. Koromilas, A.E.; Sexl, V. The tumor suppressor function of STAT1 in breast cancer. Jak-Stat 2013, 2, e23353. [Google Scholar] [CrossRef]
  81. Marzec, M.; Zhang, Q.; Goradia, A.; Raghunath, P.N.; Liu, X.; Paessler, M.; Wang, H.Y.; Wysocka, M.; Cheng, M.; Ruggeri, B.A.; et al. Oncogenic kinase NPM/ALK induces through STAT3 expression of immunosuppressive protein CD274 (PD-L1, B7-H1). Proc. Natl. Acad. Sci. USA 2008, 105, 20852–20857. [Google Scholar] [CrossRef] [Green Version]
  82. Sasidharan Nair, V.; Toor, S.M.; Ali, B.R.; Elkord, E. Dual inhibition of STAT1 and STAT3 activation downregulates expression of PD-L1 in human breast cancer cells. Expert Opin. Ther. Targets 2018, 22, 547–557. [Google Scholar] [CrossRef] [PubMed]
  83. Jing, N.; Tweardy, D.J. Targeting Stat3 in cancer therapy. Anticancer Drugs 2005, 16, 601–607. [Google Scholar] [CrossRef] [PubMed]
  84. Chen, M.; Pockaj, B.; Andreozzi, M.; Barrett, M.T.; Krishna, S.; Eaton, S.; Niu, R.; Anderson, K.S. JAK2 and PD-L1 Amplification Enhance the Dynamic Expression of PD-L1 in Triple-negative Breast Cancer. Clin. Breast Cancer 2018, 18, e1205–e1215. [Google Scholar] [CrossRef] [Green Version]
  85. Jögi, A.; Ehinger, A.; Hartman, L.; Alkner, S. Expression of HIF-1α is related to a poor prognosis and tamoxifen resistance in contralateral breast cancer. PLoS ONE 2019, 14, e0226150. [Google Scholar] [CrossRef] [Green Version]
  86. Ortmann, B.; Druker, J.; Rocha, S. Cell cycle progression in response to oxygen levels. Cell. Mol. Life Sci. 2014, 71, 3569–3582. [Google Scholar] [CrossRef] [Green Version]
  87. Semenza, G.L. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene 2010, 29, 625–634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Bos, R.; van der Groep, P.; Greijer, A.E.; Shvarts, A.; Meijer, S.; Pinedo, H.M.; Semenza, G.L.; van Diest, P.J.; van der Wall, E. Levels of hypoxia-inducible factor-1alpha independently predict prognosis in patients with lymph node negative breast carcinoma. Cancer 2003, 97, 1573–1581. [Google Scholar] [CrossRef]
  89. Noman, M.Z.; Chouaib, S. Targeting hypoxia at the forefront of anticancer immune responses. Oncoimmunology 2014, 3, e954463. [Google Scholar] [CrossRef] [Green Version]
  90. Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014, 211, 781–790. [Google Scholar] [CrossRef]
  91. Pollizzi, K.N.; Powell, J.D. Integrating canonical and metabolic signalling programmes in the regulation of T cell responses. Nat. Rev. Immunol. 2014, 14, 435–446. [Google Scholar] [CrossRef] [Green Version]
  92. Shehade, H.; Oldenhove, G.; Moser, M. Hypoxia in the intestine or solid tumors: A beneficial or deleterious alarm signal? Eur. J. Immunol. 2014, 44, 2550–2557. [Google Scholar] [CrossRef]
  93. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef] [PubMed]
  94. Brown, J.M.; Wilson, W.R. Exploiting tumour hypoxia in cancer treatment. Nat. Rev. Cancer 2004, 4, 437–447. [Google Scholar] [CrossRef]
  95. Vaupel, P.; Mayer, A. Hypoxia in cancer: Significance and impact on clinical outcome. Cancer Metastasis Rev. 2007, 26, 225–239. [Google Scholar] [CrossRef]
  96. Jing, X.; Yang, F.; Shao, C.; Wei, K.; Xie, M.; Shen, H.; Shu, Y. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol. Cancer 2019, 18, 157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Wang, Y.; Wang, H.; Yao, H.; Li, C.; Fang, J.Y.; Xu, J. Regulation of PD-L1: Emerging Routes for Targeting Tumor Immune Evasion. Front. Pharmacol. 2018, 9, 536. [Google Scholar] [CrossRef] [PubMed]
  98. Gowrishankar, K.; Gunatilake, D.; Gallagher, S.J.; Tiffen, J.; Rizos, H.; Hersey, P. Inducible but not constitutive expression of PD-L1 in human melanoma cells is dependent on activation of NF-κB. PLoS ONE 2015, 10, e0123410. [Google Scholar] [CrossRef] [Green Version]
  99. Maeda, T.; Hiraki, M.; Jin, C.; Rajabi, H.; Tagde, A.; Alam, M.; Bouillez, A.; Hu, X.; Suzuki, Y.; Miyo, M.; et al. MUC1-C Induces PD-L1 and Immune Evasion in Triple-Negative Breast Cancer. Cancer Res. 2018, 78, 205–215. [Google Scholar] [CrossRef] [Green Version]
  100. Betzler, A.C.; Theodoraki, M.N.; Schuler, P.J.; Döscher, J.; Laban, S.; Hoffmann, T.K.; Brunner, C. NF-κB and Its Role in Checkpoint Control. Int. J. Mol. Sci. 2020, 21, 3949. [Google Scholar] [CrossRef]
  101. Lim, S.O.; Li, C.W.; Xia, W.; Cha, J.H.; Chan, L.C.; Wu, Y.; Chang, S.S.; Lin, W.C.; Hsu, J.M.; Hsu, Y.H.; et al. Deubiquitination and Stabilization of PD-L1 by CSN5. Cancer Cell 2016, 30, 925–939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Lucas, J.; Hsieh, T.C.; Halicka, H.D.; Darzynkiewicz, Z.; Wu, J.M. Upregulation of PD-L1 expression by resveratrol and piceatannol in breast and colorectal cancer cells occurs via HDAC3/p300-mediated NF-κB signaling. Int. J. Oncol. 2018, 53, 1469–1480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Puvvada, S.D.; Funkhouser, W.K.; Greene, K.; Deal, A.; Chu, H.; Baldwin, A.S.; Tepper, J.E.; O’Neil, B.H. NF-kB and Bcl-3 activation are prognostic in metastatic colorectal cancer. Oncology 2010, 78, 181–188. [Google Scholar] [CrossRef] [Green Version]
  104. Zou, Y.; Uddin, M.M.; Padmanabhan, S.; Zhu, Y.; Bu, P.; Vancura, A.; Vancurova, I. The proto-oncogene Bcl3 induces immune checkpoint PD-L1 expression, mediating proliferation of ovarian cancer cells. J. Biol. Chem. 2018, 293, 15483–15496. [Google Scholar] [CrossRef] [Green Version]
  105. Li, H.; Xia, J.Q.; Zhu, F.S.; Xi, Z.H.; Pan, C.Y.; Gu, L.M.; Tian, Y.Z. LPS promotes the expression of PD-L1 in gastric cancer cells through NF-κB activation. J. Cell. Biochem. 2018, 119, 9997–10004. [Google Scholar] [CrossRef] [PubMed]
  106. Lee, S.K.; Seo, S.H.; Kim, B.S.; Kim, C.D.; Lee, J.H.; Kang, J.S.; Maeng, P.J.; Lim, J.S. IFN-gamma regulates the expression of B7-H1 in dermal fibroblast cells. J. Dermatol. Sci. 2005, 40, 95–103. [Google Scholar] [CrossRef]
  107. Kozako, T.; Yoshimitsu, M.; Fujiwara, H.; Masamoto, I.; Horai, S.; White, Y.; Akimoto, M.; Suzuki, S.; Matsushita, K.; Uozumi, K.; et al. PD-1/PD-L1 expression in human T-cell leukemia virus type 1 carriers and adult T-cell leukemia/lymphoma patients. Leukemia 2009, 23, 375–382. [Google Scholar] [CrossRef]
  108. Kozako, T.; Yoshimitsu, M.; Akimoto, M.; White, Y.; Matsushita, K.; Soeda, S.; Shimeno, H.; Kubota, R.; Izumo, S.; Arima, N. Programmed death-1 (PD-1)/PD-1 ligand pathway-mediated immune responses against human T-lymphotropic virus type 1 (HTLV-1) in HTLV-1-associated myelopathy/tropical spastic paraparesis and carriers with autoimmune disorders. Hum. Immunol. 2011, 72, 1001–1006. [Google Scholar] [CrossRef]
  109. Droeser, R.A.; Hirt, C.; Viehl, C.T.; Frey, D.M.; Nebiker, C.; Huber, X.; Zlobec, I.; Eppenberger-Castori, S.; Tzankov, A.; Rosso, R.; et al. Clinical impact of programmed cell death ligand 1 expression in colorectal cancer. Eur. J. Cancer 2013, 49, 2233–2242. [Google Scholar] [CrossRef]
  110. Thompson, R.H.; Gillett, M.D.; Cheville, J.C.; Lohse, C.M.; Dong, H.; Webster, W.S.; Krejci, K.G.; Lobo, J.R.; Sengupta, S.; Chen, L.; et al. Costimulatory B7-H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. Proc. Natl. Acad. Sci. USA 2004, 101, 17174–17179. [Google Scholar] [CrossRef] [Green Version]
  111. Chen, Y.B.; Mu, C.Y.; Huang, J.A. Clinical significance of programmed death-1 ligand-1 expression in patients with non-small cell lung cancer: A 5-year-follow-up study. Tumori 2012, 98, 751–755. [Google Scholar] [CrossRef]
  112. Hamanishi, J.; Mandai, M.; Iwasaki, M.; Okazaki, T.; Tanaka, Y.; Yamaguchi, K.; Higuchi, T.; Yagi, H.; Takakura, K.; Minato, N.; et al. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc. Natl. Acad. Sci. USA 2007, 104, 3360–3365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Nomi, T.; Sho, M.; Akahori, T.; Hamada, K.; Kubo, A.; Kanehiro, H.; Nakamura, S.; Enomoto, K.; Yagita, H.; Azuma, M.; et al. Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clin. Cancer Res. 2007, 13, 2151–2157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Ghebeh, H.; Mohammed, S.; Al-Omair, A.; Qattan, A.; Lehe, C.; Al-Qudaihi, G.; Elkum, N.; Alshabanah, M.; Bin Amer, S.; Tulbah, A.; et al. The B7-H1 (PD-L1) T lymphocyte-inhibitory molecule is expressed in breast cancer patients with infiltrating ductal carcinoma: Correlation with important high-risk prognostic factors. Neoplasia 2006, 8, 190–198. [Google Scholar] [CrossRef] [Green Version]
  115. Jiang, Y.; Zhan, H. Communication between EMT and PD-L1 signaling: New insights into tumor immune evasion. Cancer Lett. 2020, 468, 72–81. [Google Scholar] [CrossRef] [PubMed]
  116. Gonzalez-Angulo, A.M.; Ferrer-Lozano, J.; Stemke-Hale, K.; Sahin, A.; Liu, S.; Barrera, J.A.; Burgues, O.; Lluch, A.M.; Chen, H.; Hortobagyi, G.N.; et al. PI3K pathway mutations and PTEN levels in primary and metastatic breast cancer. Mol. Cancer Ther. 2011, 10, 1093–1101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Li, C.W.; Lim, S.O.; Xia, W.; Lee, H.H.; Chan, L.C.; Kuo, C.W.; Khoo, K.H.; Chang, S.S.; Cha, J.H.; Kim, T.; et al. Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity. Nat. Commun. 2016, 7, 12632. [Google Scholar] [CrossRef] [Green Version]
  118. Li, C.W.; Lim, S.O.; Chung, E.M.; Kim, Y.S.; Park, A.H.; Yao, J.; Cha, J.H.; Xia, W.; Chan, L.C.; Kim, T.; et al. Eradication of Triple-Negative Breast Cancer Cells by Targeting Glycosylated PD-L1. Cancer Cell 2018, 33, 187–201.e110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Farahmand, L.; Merikhian, P.; Jalili, N.; Darvishi, B.; Majidzadeh, A.K. Significant Role of MUC1 in Development of Resistance to Currently Existing Anti-cancer Therapeutic Agents. Curr. Cancer Drug Targets 2018, 18, 737–748. [Google Scholar] [CrossRef]
  120. Huang, J.; Li, H.; Ren, G. Epithelial-mesenchymal transition and drug resistance in breast cancer (Review). Int. J. Oncol. 2015, 47, 840–848. [Google Scholar] [CrossRef] [Green Version]
  121. Palucka, A.K.; Coussens, L.M. The Basis of Oncoimmunology. Cell 2016, 164, 1233–1247. [Google Scholar] [CrossRef] [Green Version]
  122. Fuxe, J.; Karlsson, M.C. TGF-β-induced epithelial-mesenchymal transition: A link between cancer and inflammation. Semin. Cancer Biol. 2012, 22, 455–461. [Google Scholar] [CrossRef] [PubMed]
  123. Ock, C.Y.; Kim, S.; Keam, B.; Kim, M.; Kim, T.M.; Kim, J.H.; Jeon, Y.K.; Lee, J.S.; Kwon, S.K.; Hah, J.H.; et al. PD-L1 expression is associated with epithelial-mesenchymal transition in head and neck squamous cell carcinoma. Oncotarget 2016, 7, 15901–15914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Qiu, X.Y.; Hu, D.X.; Chen, W.Q.; Chen, R.Q.; Qian, S.R.; Li, C.Y.; Li, Y.J.; Xiong, X.X.; Liu, D.; Pan, F.; et al. PD-L1 confers glioblastoma multiforme malignancy via Ras binding and Ras/Erk/EMT activation. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 1754–1769. [Google Scholar] [CrossRef]
  125. Wang, Y.; Wang, H.; Zhao, Q.; Xia, Y.; Hu, X.; Guo, J. PD-L1 induces epithelial-to-mesenchymal transition via activating SREBP-1c in renal cell carcinoma. Med. Oncol. 2015, 32, 212. [Google Scholar] [CrossRef] [PubMed]
  126. Xu, G.L.; Ni, C.F.; Liang, H.S.; Xu, Y.H.; Wang, W.S.; Shen, J.; Li, M.M.; Zhu, X.L. Upregulation of PD-L1 expression promotes epithelial-to-mesenchymal transition in sorafenib-resistant hepatocellular carcinoma cells. Gastroenterol. Rep. 2020, 8, 390–398. [Google Scholar] [CrossRef] [PubMed]
  127. Goossens, S.; Vandamme, N.; Van Vlierberghe, P.; Berx, G. EMT transcription factors in cancer development re-evaluated: Beyond EMT and MET. Biochim. Biophys. Acta Rev. Cancer 2017, 1868, 584–591. [Google Scholar] [CrossRef] [PubMed]
  128. López-Novoa, J.M.; Nieto, M.A. Inflammation and EMT: An alliance towards organ fibrosis and cancer progression. EMBO Mol. Med. 2009, 1, 303–314. [Google Scholar] [CrossRef] [Green Version]
  129. Ueno, T.; Tsuchikawa, T.; Hatanaka, K.C.; Hatanaka, Y.; Mitsuhashi, T.; Nakanishi, Y.; Noji, T.; Nakamura, T.; Okamura, K.; Matsuno, Y.; et al. Prognostic impact of programmed cell death ligand 1 (PD-L1) expression and its association with epithelial-mesenchymal transition in extrahepatic cholangiocarcinoma. Oncotarget 2018, 9, 20034–20047. [Google Scholar] [CrossRef] [Green Version]
  130. Taliaferro-Smith, L.; Oberlick, E.; Liu, T.; McGlothen, T.; Alcaide, T.; Tobin, R.; Donnelly, S.; Commander, R.; Kline, E.; Nagaraju, G.P.; et al. FAK activation is required for IGF1R-mediated regulation of EMT, migration, and invasion in mesenchymal triple negative breast cancer cells. Oncotarget 2015, 6, 4757–4772. [Google Scholar] [CrossRef] [Green Version]
  131. Cevenini, A.; Orrù, S.; Mancini, A.; Alfieri, A.; Buono, P.; Imperlini, E. Molecular Signatures of the Insulin-like Growth Factor 1-mediated Epithelial-Mesenchymal Transition in Breast, Lung and Gastric Cancers. Int. J. Mol. Sci. 2018, 19, 2411. [Google Scholar] [CrossRef] [Green Version]
  132. Kumar, S.; Davra, V.; Obr, A.E.; Geng, K.; Wood, T.L.; De Lorenzo, M.S.; Birge, R.B. Crk adaptor protein promotes PD-L1 expression, EMT and immune evasion in a murine model of triple-negative breast cancer. Oncoimmunology 2017, 7, e1376155. [Google Scholar] [CrossRef]
  133. Noman, M.Z.; Janji, B.; Abdou, A.; Hasmim, M.; Terry, S.; Tan, T.Z.; Mami-Chouaib, F.; Thiery, J.P.; Chouaib, S. The immune checkpoint ligand PD-L1 is upregulated in EMT-activated human breast cancer cells by a mechanism involving ZEB-1 and miR-200. Oncoimmunology 2017, 6, e1263412. [Google Scholar] [CrossRef] [PubMed]
  134. Gao, J.; Yan, Q.; Wang, J.; Liu, S.; Yang, X. Epithelial-to-mesenchymal transition induced by TGF-β1 is mediated by AP1-dependent EpCAM expression in MCF-7 cells. J. Cell. Physiol. 2015, 230, 775–782. [Google Scholar] [CrossRef]
  135. Johansson, J.; Tabor, V.; Wikell, A.; Jalkanen, S.; Fuxe, J. TGF-β1-Induced Epithelial-Mesenchymal Transition Promotes Monocyte/Macrophage Properties in Breast Cancer Cells. Front. Oncol. 2015, 5, 3. [Google Scholar] [CrossRef] [Green Version]
  136. Lee, Y.J.; Park, J.H.; Oh, S.M. TOPK promotes epithelial-mesenchymal transition and invasion of breast cancer cells through upregulation of TBX3 in TGF-β1/Smad signaling. Biochem. Biophys. Res. Commun. 2020, 522, 270–277. [Google Scholar] [CrossRef]
  137. Ma, F.; Li, W.; Liu, C.; Li, W.; Yu, H.; Lei, B.; Ren, Y.; Li, Z.; Pang, D.; Qian, C. MiR-23a promotes TGF-β1-induced EMT and tumor metastasis in breast cancer cells by directly targeting CDH1 and activating Wnt/β-catenin signaling. Oncotarget 2017, 8, 69538–69550. [Google Scholar] [CrossRef] [Green Version]
  138. Ma, M.; He, M.; Jiang, Q.; Yan, Y.; Guan, S.; Zhang, J.; Yu, Z.; Chen, Q.; Sun, M.; Yao, W.; et al. MiR-487a Promotes TGF-β1-induced EMT, the Migration and Invasion of Breast Cancer Cells by Directly Targeting MAGI2. Int. J. Biol. Sci. 2016, 12, 397–408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Pang, M.F.; Georgoudaki, A.M.; Lambut, L.; Johansson, J.; Tabor, V.; Hagikura, K.; Jin, Y.; Jansson, M.; Alexander, J.S.; Nelson, C.M.; et al. TGF-β1-induced EMT promotes targeted migration of breast cancer cells through the lymphatic system by the activation of CCR7/CCL21-mediated chemotaxis. Oncogene 2016, 35, 748–760. [Google Scholar] [CrossRef] [PubMed]
  140. Sun, Y.; Zhou, Q.M.; Lu, Y.Y.; Zhang, H.; Chen, Q.L.; Zhao, M.; Su, S.B. Resveratrol Inhibits the Migration and Metastasis of MDA-MB-231 Human Breast Cancer by Reversing TGF-β1-Induced Epithelial-Mesenchymal Transition. Molecules 2019, 24, 1131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. O’Donnell, J.S.; Long, G.V.; Scolyer, R.A.; Teng, M.W.; Smyth, M.J. Resistance to PD1/PDL1 checkpoint inhibition. Cancer Treat. Rev. 2017, 52, 71–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Dave, B.; Mittal, V.; Tan, N.M.; Chang, J.C. Epithelial-mesenchymal transition, cancer stem cells and treatment resistance. Breast Cancer Res. 2012, 14, 202. [Google Scholar] [CrossRef] [Green Version]
  143. Mallini, P.; Lennard, T.; Kirby, J.; Meeson, A. Epithelial-to-mesenchymal transition: What is the impact on breast cancer stem cells and drug resistance. Cancer Treat. Rev. 2014, 40, 341–348. [Google Scholar] [CrossRef]
  144. Yoshida, R.; Niki, M.; Jyotaki, M.; Sanematsu, K.; Shigemura, N.; Ninomiya, Y. Modulation of sweet responses of taste receptor cells. Semin. Cell Dev. Biol. 2013, 24, 226–231. [Google Scholar] [CrossRef] [PubMed]
  145. Tinoco, G.; Warsch, S.; Glück, S.; Avancha, K.; Montero, A.J. Treating breast cancer in the 21st century: Emerging biological therapies. J. Cancer 2013, 4, 117–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Ávila-Gálvez, M.; Giménez-Bastida, J.A.; Espín, J.C.; González-Sarrías, A. Dietary Phenolics against Breast Cancer. A Critical Evidence-Based Review and Future Perspectives. Int. J. Mol. Sci. 2020, 21, 5718. [Google Scholar] [CrossRef]
  147. Tang, Y.; Wang, Y.; Kiani, M.F.; Wang, B. Classification, Treatment Strategy, and Associated Drug Resistance in Breast Cancer. Clin. Breast Cancer 2016, 16, 335–343. [Google Scholar] [CrossRef] [PubMed]
  148. Waks, A.G.; Winer, E.P. Breast Cancer Treatment. JAMA 2019, 321, 316. [Google Scholar] [CrossRef] [Green Version]
  149. Di Cosimo, S.; Baselga, J. Management of breast cancer with targeted agents: Importance of heterogeneity. [corrected]. Nat. Rev. Clin. Oncol. 2010, 7, 139–147. [Google Scholar] [CrossRef] [PubMed]
  150. Balduzzi, S.; Mantarro, S.; Guarneri, V.; Tagliabue, L.; Pistotti, V.; Moja, L.; D’Amico, R. Trastuzumab-containing regimens for metastatic breast cancer. Cochrane Database Syst. Rev. 2014, 2014, Cd006242. [Google Scholar] [CrossRef] [Green Version]
  151. Shah, A.N.; Cristofanilli, M. The Growing Role of CDK4/6 Inhibitors in Treating Hormone Receptor-Positive Advanced Breast Cancer. Curr. Treat. Options Oncol. 2017, 18, 6. [Google Scholar] [CrossRef] [PubMed]
  152. Krop, I.E.; Mayer, I.A.; Ganju, V.; Dickler, M.; Johnston, S.; Morales, S.; Yardley, D.A.; Melichar, B.; Forero-Torres, A.; Lee, S.C.; et al. Pictilisib for oestrogen receptor-positive, aromatase inhibitor-resistant, advanced or metastatic breast cancer (FERGI): A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 2016, 17, 811–821. [Google Scholar] [CrossRef] [Green Version]
  153. Blackwell, K.; Burris, H.; Gomez, P.; Lynn Henry, N.; Isakoff, S.; Campana, F.; Gao, L.; Jiang, J.; Macé, S.; Tolaney, S.M. Phase I/II dose-escalation study of PI3K inhibitors pilaralisib or voxtalisib in combination with letrozole in patients with hormone-receptor-positive and HER2-negative metastatic breast cancer refractory to a non-steroidal aromatase inhibitor. Breast Cancer Res. Treat. 2015, 154, 287–297. [Google Scholar] [CrossRef]
  154. Berrada, N.; Delaloge, S.; André, F. Treatment of triple-negative metastatic breast cancer: Toward individualized targeted treatments or chemosensitization? Ann. Oncol. 2010, 21, vii30–vii35. [Google Scholar] [CrossRef]
  155. Robson, M.; Im, S.A.; Senkus, E.; Xu, B.; Domchek, S.M.; Masuda, N.; Delaloge, S.; Li, W.; Tung, N.; Armstrong, A.; et al. Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. N. Engl. J. Med. 2017, 377, 1700. [Google Scholar] [CrossRef]
  156. Brown, J.S.; Kaye, S.B.; Yap, T.A. PARP inhibitors: The race is on. Br. J. Cancer 2016, 114, 713–715. [Google Scholar] [CrossRef] [Green Version]
  157. Hurvitz, S.A.; Quek, R.G.W.; Turner, N.C.; Telli, M.L.; Rugo, H.S.; Mailliez, A.; Ettl, J.; Grischke, E.; Mina, L.A.; Balmaña, J.; et al. Quality of life with talazoparib after platinum or multiple cytotoxic non-platinum regimens in patients with advanced breast cancer and germline BRCA1/2 mutations: Patient-reported outcomes from the ABRAZO phase 2 trial. Eur. J. Cancer 2018, 104, 160–168. [Google Scholar] [CrossRef] [PubMed]
  158. Bresalier, R.S.; Kopetz, S.; Brenner, D.E. Blood-based tests for colorectal cancer screening: Do they threaten the survival of the FIT test? Dig. Dis. Sci. 2015, 60, 664–671. [Google Scholar] [CrossRef] [PubMed]
  159. Chlebowski, R.T. Current concepts in breast cancer chemoprevention. Pol. Arch. Med. Wewnętrznej 2014, 124, 191–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Denkert, C.; Liedtke, C.; Tutt, A.; von Minckwitz, G. Molecular alterations in triple-negative breast cancer-the road to new treatment strategies. Lancet 2017, 389, 2430–2442. [Google Scholar] [CrossRef] [Green Version]
  161. 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] [PubMed] [Green Version]
  162. Wein, L.; Loi, S. Mechanisms of resistance of chemotherapy in early-stage triple negative breast cancer (TNBC). Breast 2017, 34, S27–S30. [Google Scholar] [CrossRef]
  163. Finck, A.; Gill, S.I.; June, C.H. Cancer immunotherapy comes of age and looks for maturity. Nat. Commun. 2020, 11, 3325. [Google Scholar] [CrossRef] [PubMed]
  164. Buder-Bakhaya, K.; Hassel, J.C. Biomarkers for Clinical Benefit of Immune Checkpoint Inhibitor Treatment-A Review From the Melanoma Perspective and Beyond. Front. Immunol. 2018, 9, 1474. [Google Scholar] [CrossRef] [PubMed]
  165. Golay, J.; Andrea, A.E. Combined Anti-Cancer Strategies Based on Anti-Checkpoint Inhibitor Antibodies. Antibodies 2020, 9, 17. [Google Scholar] [CrossRef]
  166. Force, J.; Leal, J.H.S.; McArthur, H.L. Checkpoint Blockade Strategies in the Treatment of Breast Cancer: Where We Are and Where We Are Heading. Curr. Treat. Options Oncol. 2019, 20, 35. [Google Scholar] [CrossRef] [PubMed]
  167. Bensch, F.; van der Veen, E.L.; Lub-de Hooge, M.N.; Jorritsma-Smit, A.; Boellaard, R.; Kok, I.C.; Oosting, S.F.; Schröder, C.P.; Hiltermann, T.J.N.; van der Wekken, A.J.; et al. (89)Zr-atezolizumab imaging as a non-invasive approach to assess clinical response to PD-L1 blockade in cancer. Nat. Med. 2018, 24, 1852–1858. [Google Scholar] [CrossRef]
  168. Rosenberg, J.E.; Hoffman-Censits, J.; Powles, T.; van der Heijden, M.S.; Balar, A.V.; Necchi, A.; Dawson, N.; O’Donnell, P.H.; Balmanoukian, A.; Loriot, Y.; et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: A single-arm, multicentre, phase 2 trial. Lancet 2016, 387, 1909–1920. [Google Scholar] [CrossRef] [Green Version]
  169. Bertucci, F.; Gonçalves, A. Immunotherapy in Breast Cancer: The Emerging Role of PD-1 and PD-L1. Curr. Oncol. Rep. 2017, 19, 64. [Google Scholar] [CrossRef]
  170. Daud, A.I.; Wolchok, J.D.; Robert, C.; Hwu, W.J.; Weber, J.S.; Ribas, A.; Hodi, F.S.; Joshua, A.M.; Kefford, R.; Hersey, P.; et al. Programmed Death-Ligand 1 Expression and Response to the Anti-Programmed Death 1 Antibody Pembrolizumab in Melanoma. J. Clin. Oncol. 2016, 34, 4102–4109. [Google Scholar] [CrossRef]
  171. Alsaab, H.O.; Sau, S.; Alzhrani, R.; Tatiparti, K.; Bhise, K.; Kashaw, S.K.; Iyer, A.K. PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer Immunotherapy: Mechanism, Combinations, and Clinical Outcome. Front. Pharmacol. 2017, 8, 561. [Google Scholar] [CrossRef] [PubMed]
  172. Ghebeh, H.; Lehe, C.; Barhoush, E.; Al-Romaih, K.; Tulbah, A.; Al-Alwan, M.; Hendrayani, S.F.; Manogaran, P.; Alaiya, A.; Al-Tweigeri, T.; et al. Doxorubicin downregulates cell surface B7-H1 expression and upregulates its nuclear expression in breast cancer cells: Role of B7-H1 as an anti-apoptotic molecule. Breast Cancer Res. 2010, 12, R48. [Google Scholar] [CrossRef] [Green Version]
  173. Hasan, A.; Ghebeh, H.; Lehe, C.; Ahmad, R.; Dermime, S. Therapeutic targeting of B7-H1 in breast cancer. Expert Opin. Ther. Targets 2011, 15, 1211–1225. [Google Scholar] [CrossRef]
  174. 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]
  175. Mattera, R.; Benvenuto, M.; Giganti, M.G.; Tresoldi, I.; Pluchinotta, F.R.; Bergante, S.; Tettamanti, G.; Masuelli, L.; Manzari, V.; Modesti, A.; et al. Effects of Polyphenols on Oxidative Stress-Mediated Injury in Cardiomyocytes. Nutrients 2017, 9, 523. [Google Scholar] [CrossRef] [Green Version]
  176. 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] [Green Version]
  177. Cho, Y.S.; Schiller, N.L.; Kahng, H.Y.; Oh, K.H. Cellular responses and proteomic analysis of Escherichia coli exposed to green tea polyphenols. Curr. Microbiol. 2007, 55, 501–506. [Google Scholar] [CrossRef] [PubMed]
  178. Sies, H.; Schewe, T.; Heiss, C.; Kelm, M. Cocoa polyphenols and inflammatory mediators. Am. J. Clin. Nutr. 2005, 81, 304s–312s. [Google Scholar] [CrossRef] [Green Version]
  179. Scalbert, A.; Johnson, I.T.; Saltmarsh, M. Polyphenols: Antioxidants and beyond. Am. J. Clin. Nutr. 2005, 81, 215s–217s. [Google Scholar] [CrossRef]
  180. Hubert, P.A.; Lee, S.G.; Lee, S.K.; Chun, O.K. Dietary Polyphenols, Berries, and Age-Related Bone Loss: A Review Based on Human, Animal, and Cell Studies. Antioxidants 2014, 3, 144–158. [Google Scholar] [CrossRef] [Green Version]
  181. Vita, J.A. Polyphenols and cardiovascular disease: Effects on endothelial and platelet function. Am. J. Clin. Nutr. 2005, 81, 292s–297s. [Google Scholar] [CrossRef]
  182. Lambert, J.D.; Hong, J.; Yang, G.Y.; Liao, J.; Yang, C.S. Inhibition of carcinogenesis by polyphenols: Evidence from laboratory investigations. Am. J. Clin. Nutr. 2005, 81, 284s–291s. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Adhami, V.M.; Syed, D.N.; Khan, N.; Mukhtar, H. Dietary flavonoid fisetin: A novel dual inhibitor of PI3K/Akt and mTOR for prostate cancer management. Biochem. Pharmacol. 2012, 84, 1277–1281. [Google Scholar] [CrossRef] [Green Version]
  184. Christensen, K.Y.; Naidu, A.; Parent, M.; Pintos, J.; Abrahamowicz, M.; Siemiatycki, J.; Koushik, A. The risk of lung cancer related to dietary intake of flavonoids. Nutr. Cancer 2012, 64, 964–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Knekt, P.; Järvinen, R.; Seppänen, R.; Hellövaara, M.; Teppo, L.; Pukkala, E.; Aromaa, A. Dietary flavonoids and the risk of lung cancer and other malignant neoplasms. Am. J. Epidemiol. 1997, 146, 223–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Le Marchand, L.; Murphy, S.P.; Hankin, J.H.; Wilkens, L.R.; Kolonel, L.N. Intake of flavonoids and lung cancer. J. Natl. Cancer Inst. 2000, 92, 154–160. [Google Scholar] [CrossRef] [Green Version]
  187. Bosetti, C.; Spertini, L.; Parpinel, M.; Gnagnarella, P.; Lagiou, P.; Negri, E.; Franceschi, S.; Montella, M.; Peterson, J.; Dwyer, J.; et al. Flavonoids and breast cancer risk in Italy. Cancer Epidemiol. Biomark. Prev. 2005, 14, 805–808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Rossi, M.; Bosetti, C.; Negri, E.; Lagiou, P.; La Vecchia, C. Flavonoids, proanthocyanidins, and cancer risk: A network of case-control studies from Italy. Nutr. Cancer 2010, 62, 871–877. [Google Scholar] [CrossRef]
  189. Rossi, M.; Negri, E.; Lagiou, P.; Talamini, R.; Dal Maso, L.; Montella, M.; Franceschi, S.; La Vecchia, C. Flavonoids and ovarian cancer risk: A case-control study in Italy. Int. J. Cancer 2008, 123, 895–898. [Google Scholar] [CrossRef]
  190. Rossi, R.E.; Pericleous, M.; Mandair, D.; Whyand, T.; Caplin, M.E. The role of dietary factors in prevention and progression of breast cancer. Anticancer Res. 2014, 34, 6861–6875. [Google Scholar]
  191. Surh, Y.J. Cancer chemoprevention with dietary phytochemicals. Nat. Rev. Cancer 2003, 3, 768–780. [Google Scholar] [CrossRef]
  192. Weng, C.J.; Yen, G.C. Chemopreventive effects of dietary phytochemicals against cancer invasion and metastasis: Phenolic acids, monophenol, polyphenol, and their derivatives. Cancer Treat. Rev. 2012, 38, 76–87. [Google Scholar] [CrossRef] [PubMed]
  193. Chen, D.; Daniel, K.G.; Kuhn, D.J.; Kazi, A.; Bhuiyan, M.; Li, L.; Wang, Z.; Wan, S.B.; Lam, W.H.; Chan, T.H.; et al. Green tea and tea polyphenols in cancer prevention. Front. Biosci. 2004, 9, 2618–2631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Hadi, S.M.; Asad, S.F.; Singh, S.; Ahmad, A. Putative mechanism for anticancer and apoptosis-inducing properties of plant-derived polyphenolic compounds. IUBMB Life 2000, 50, 167–171. [Google Scholar] [CrossRef] [PubMed]
  195. Rice-Evans, C. Flavonoid antioxidants. Curr. Med. Chem. 2001, 8, 797–807. [Google Scholar] [CrossRef] [PubMed]
  196. Pandey, M.K.; Gupta, S.C.; Nabavizadeh, A.; Aggarwal, B.B. Regulation of cell signaling pathways by dietary agents for cancer prevention and treatment. Semin. Cancer Biol. 2017, 46, 158–181. [Google Scholar] [CrossRef] [PubMed]
  197. Feng, T.; Wei, Y.; Lee, R.J.; Zhao, L. Liposomal curcumin and its application in cancer. Int. J. Nanomed. 2017, 12, 6027–6044. [Google Scholar] [CrossRef] [Green Version]
  198. Anselmo, A.C.; Mitragotri, S. An overview of clinical and commercial impact of drug delivery systems. J. Control. Release 2014, 190, 15–28. [Google Scholar] [CrossRef] [Green Version]
  199. Heenatigala Palliyage, G.; Singh, S.; Ashby, C.R., Jr.; Tiwari, A.K.; Chauhan, H. Pharmaceutical Topical Delivery of Poorly Soluble Polyphenols: Potential Role in Prevention and Treatment of Melanoma. AAPS PharmSciTech 2019, 20, 250. [Google Scholar] [CrossRef]
  200. Thompson, L.U.; Chen, J.M.; Li, T.; Strasser-Weippl, K.; Goss, P.E. Dietary flaxseed alters tumor biological markers in postmenopausal breast cancer. Clin. Cancer Res. 2005, 11, 3828–3835. [Google Scholar] [CrossRef] [Green Version]
  201. Yu, S.S.; Spicer, D.V.; Hawes, D.; Tseng, C.C.; Yang, C.S.; Pike, M.C.; Wu, A.H. Biological effects of green tea capsule supplementation in pre-surgery postmenopausal breast cancer patients. Front. Oncol. 2013, 3, 298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Lazzeroni, M.; Guerrieri-Gonzaga, A.; Gandini, S.; Johansson, H.; Serrano, D.; Cazzaniga, M.; Aristarco, V.; Macis, D.; Mora, S.; Caldarella, P.; et al. A Presurgical Study of Lecithin Formulation of Green Tea Extract in Women with Early Breast Cancer. Cancer Prev. Res. 2017, 10, 363–370. [Google Scholar] [CrossRef] [Green Version]
  203. McCann, S.E.; Edge, S.B.; Hicks, D.G.; Thompson, L.U.; Morrison, C.D.; Fetterly, G.; Andrews, C.; Clark, K.; Wilton, J.; Kulkarni, S. A pilot study comparing the effect of flaxseed, aromatase inhibitor, and the combination on breast tumor biomarkers. Nutr. Cancer 2014, 66, 566–575. [Google Scholar] [CrossRef] [Green Version]
  204. Bayet-Robert, M.; Kwiatkowski, F.; Leheurteur, M.; Gachon, F.; Planchat, E.; Abrial, C.; Mouret-Reynier, M.A.; Durando, X.; Barthomeuf, C.; Chollet, P. Phase I dose escalation trial of docetaxel plus curcumin in patients with advanced and metastatic breast cancer. Cancer Biol. Ther. 2010, 9, 8–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Ryan, J.L.; Heckler, C.E.; Ling, M.; Katz, A.; Williams, J.P.; Pentland, A.P.; Morrow, G.R. Curcumin for radiation dermatitis: A randomized, double-blind, placebo-controlled clinical trial of thirty breast cancer patients. Radiat. Res. 2013, 180, 34–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Martínez, N.; Herrera, M.; Frías, L.; Provencio, M.; Pérez-Carrión, R.; Díaz, V.; Morse, M.; Crespo, M.C. A combination of hydroxytyrosol, omega-3 fatty acids and curcumin improves pain and inflammation among early stage breast cancer patients receiving adjuvant hormonal therapy: Results of a pilot study. Clin. Transl. Oncol. 2019, 21, 489–498. [Google Scholar] [CrossRef] [PubMed]
  207. Lewandowska, H.; Kalinowska, M.; Lewandowski, W.; Stępkowski, T.M.; Brzóska, K. The role of natural polyphenols in cell signaling and cytoprotection against cancer development. J. Nutr. Biochem. 2016, 32, 1–19. [Google Scholar] [CrossRef]
  208. Takac, P.; Kello, M.; Pilatova, M.B.; Kudlickova, Z.; Vilkova, M.; Slepcikova, P.; Petik, P.; Mojzis, J. New chalcone derivative exhibits antiproliferative potential by inducing G2/M cell cycle arrest, mitochondrial-mediated apoptosis and modulation of MAPK signalling pathway. Chem. Biol. Interact. 2018, 292, 37–49. [Google Scholar] [CrossRef]
  209. Grover, A.K.; Samson, S.E. Benefits of antioxidant supplements for knee osteoarthritis: Rationale and reality. Nutr. J. 2016, 15, 1. [Google Scholar] [CrossRef] [Green Version]
  210. Nayak, A.P.; Mills, T.; Norton, I. Lipid Based Nanosystems for Curcumin: Past, Present and Future. Curr. Pharm. Des. 2016, 22, 4247–4256. [Google Scholar] [CrossRef]
  211. Radomska-Leśniewska, D.M.; Osiecka-Iwan, A.; Hyc, A.; Góźdź, A.; Dąbrowska, A.M.; Skopiński, P. Therapeutic potential of curcumin in eye diseases. Cent. Eur J. Immunol. 2019, 44, 181–189. [Google Scholar] [CrossRef]
  212. Sahin Kavaklı, H.; Koca, C.; Alıcı, O. Antioxidant effects of curcumin in spinal cord injury in rats. Ulus Travma Acil Cerrahi Derg 2011, 17, 14–18. [Google Scholar] [CrossRef] [Green Version]
  213. Zhang, N.; Li, H.; Jia, J.; He, M. Anti-inflammatory effect of curcumin on mast cell-mediated allergic responses in ovalbumin-induced allergic rhinitis mouse. Cell. Immunol. 2015, 298, 88–95. [Google Scholar] [CrossRef]
  214. Zhu, L.; Ding, X.; Zhang, D.; Yuan, C.; Wang, J.; Ndegwa, E.; Zhu, G. Curcumin inhibits bovine herpesvirus type 1 entry into MDBK cells. Acta Virol. 2015, 59, 221–227. [Google Scholar] [CrossRef] [Green Version]
  215. Mollazadeh, H.; Cicero, A.F.G.; Blesso, C.N.; Pirro, M.; Majeed, M.; Sahebkar, A. Immune modulation by curcumin: The role of interleukin-10. Crit. Rev. Food Sci. Nutr. 2019, 59, 89–101. [Google Scholar] [CrossRef]
  216. Fujiwara, H.; Hosokawa, M.; Zhou, X.; Fujimoto, S.; Fukuda, K.; Toyoda, K.; Nishi, Y.; Fujita, Y.; Yamada, K.; Yamada, Y.; et al. Curcumin inhibits glucose production in isolated mice hepatocytes. Diabetes Res. Clin. Pract. 2008, 80, 185–191. [Google Scholar] [CrossRef] [Green Version]
  217. Varghese, E.; Samuel, S.M.; Abotaleb, M.; Cheema, S.; Mamtani, R.; Büsselberg, D. The "Yin and Yang" of Natural Compounds in Anticancer Therapy of Triple-Negative Breast Cancers. Cancers 2018, 10, 346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Poma, P.; Labbozzetta, M.; D’Alessandro, N.; Notarbartolo, M. NF-κB Is a Potential Molecular Drug Target in Triple-Negative Breast Cancers. Omics 2017, 21, 225–231. [Google Scholar] [CrossRef] [PubMed]
  219. Giltnane, J.M.; Balko, J.M. Rationale for targeting the Ras/MAPK pathway in triple-negative breast cancer. Discov. Med. 2014, 17, 275–283. [Google Scholar] [PubMed]
  220. King, T.D.; Suto, M.J.; Li, Y. The Wnt/β-catenin signaling pathway: A potential therapeutic target in the treatment of triple negative breast cancer. J. Cell. Biochem. 2012, 113, 13–18. [Google Scholar] [CrossRef] [PubMed]
  221. Gordon, V.; Banerji, S. Molecular pathways: PI3K pathway targets in triple-negative breast cancers. Clin. Cancer Res. 2013, 19, 3738–3744. [Google Scholar] [CrossRef] [Green Version]
  222. Gallardo, M.; Calaf, G.M. Curcumin inhibits invasive capabilities through epithelial mesenchymal transition in breast cancer cell lines. Int. J. Oncol. 2016, 49, 1019–1027. [Google Scholar] [CrossRef] [PubMed]
  223. Speiser, J.J.; Erşahin, C.; Osipo, C. The functional role of Notch signaling in triple-negative breast cancer. Vitam. Horm. 2013, 93, 277–306. [Google Scholar] [CrossRef]
  224. Jobin, C.; Bradham, C.A.; Russo, M.P.; Juma, B.; Narula, A.S.; Brenner, D.A.; Sartor, R.B. Curcumin blocks cytokine-mediated NF-kappa B activation and proinflammatory gene expression by inhibiting inhibitory factor I-kappa B kinase activity. J. Immunol. 1999, 163, 3474–3483. [Google Scholar] [PubMed]
  225. Shishodia, S.; Koul, D.; Aggarwal, B.B. Cyclooxygenase (COX)-2 inhibitor celecoxib abrogates TNF-induced NF-kappa B activation through inhibition of activation of I kappa B alpha kinase and Akt in human non-small cell lung carcinoma: Correlation with suppression of COX-2 synthesis. J. Immunol. 2004, 173, 2011–2022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Cai, J.; Sun, H.; Zheng, B.; Xie, M.; Xu, C.; Zhang, G.; Huang, X.; Zhuang, J. Curcumin attenuates lncRNA H19-induced epithelial-mesenchymal transition in tamoxifen-resistant breast cancer cells. Mol. Med. Rep. 2021, 23. [Google Scholar] [CrossRef]
  227. Peng, F.; Li, T.T.; Wang, K.L.; Xiao, G.Q.; Wang, J.H.; Zhao, H.D.; Kang, Z.J.; Fan, W.J.; Zhu, L.L.; Li, M.; et al. H19/let-7/LIN28 reciprocal negative regulatory circuit promotes breast cancer stem cell maintenance. Cell Death Dis. 2017, 8, e2569. [Google Scholar] [CrossRef] [Green Version]
  228. Zhu, Q.N.; Wang, G.; Guo, Y.; Peng, Y.; Zhang, R.; Deng, J.L.; Li, Z.X.; Zhu, Y.S. LncRNA H19 is a major mediator of doxorubicin chemoresistance in breast cancer cells through a cullin4A-MDR1 pathway. Oncotarget 2017, 8, 91990–92003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Yu, Y.; Zhang, W.; Li, A.; Chen, Y.; Ou, Q.; He, Z.; Zhang, Y.; Liu, R.; Yao, H.; Song, E. Association of Long Noncoding RNA Biomarkers With Clinical Immune Subtype and Prediction of Immunotherapy Response in Patients with Cancer. JAMA Netw. Open 2020, 3, e202149. [Google Scholar] [CrossRef] [Green Version]
  230. Zhou, Y.; Zhu, Y.; Xie, Y.; Ma, X. The Role of Long Non-coding RNAs in Immunotherapy Resistance. Front. Oncol. 2019, 9, 1292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  231. Shek, D.; Read, S.A.; Akhuba, L.; Qiao, L.; Gao, B.; Nagrial, A.; Carlino, M.S.; Ahlenstiel, G. Non-coding RNA and immune-checkpoint inhibitors: Friends or foes? Immunotherapy 2020, 12, 513–529. [Google Scholar] [CrossRef] [PubMed]
  232. Gallardo, M.; Kemmerling, U.; Aguayo, F.; Bleak, T.C.; Muñoz, J.P.; Calaf, G.M. Curcumin rescues breast cells from epithelial-mesenchymal transition and invasion induced by anti-miR-34a. Int. J. Oncol. 2020, 56, 480–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Barrallo-Gimeno, A.; Nieto, M.A. The Snail genes as inducers of cell movement and survival: Implications in development and cancer. Development 2005, 132, 3151–3161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Asiedu, M.K.; Beauchamp-Perez, F.D.; Ingle, J.N.; Behrens, M.D.; Radisky, D.C.; Knutson, K.L. AXL induces epithelial-to-mesenchymal transition and regulates the function of breast cancer stem cells. Oncogene 2014, 33, 1316–1324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Zhang, Y.X.; Knyazev, P.G.; Cheburkin, Y.V.; Sharma, K.; Knyazev, Y.P.; Orfi, L.; Szabadkai, I.; Daub, H.; Kéri, G.; Ullrich, A. AXL is a potential target for therapeutic intervention in breast cancer progression. Cancer Res. 2008, 68, 1905–1915. [Google Scholar] [CrossRef] [Green Version]
  236. Guo, Z.; Li, Y.; Zhang, D.; Ma, J. Axl inhibition induces the antitumor immune response which can be further potentiated by PD-1 blockade in the mouse cancer models. Oncotarget 2017, 8, 89761–89774. [Google Scholar] [CrossRef] [Green Version]
  237. Chen, W.C.; Lai, Y.A.; Lin, Y.C.; Ma, J.W.; Huang, L.F.; Yang, N.S.; Ho, C.T.; Kuo, S.C.; Way, T.D. Curcumin suppresses doxorubicin-induced epithelial-mesenchymal transition via the inhibition of TGF-β and PI3K/AKT signaling pathways in triple-negative breast cancer cells. J. Agric. Food Chem. 2013, 61, 11817–11824. [Google Scholar] [CrossRef]
  238. Paramita, P.; Wardhani, B.W.; Wanandi, S.I.; Louisa, M. Curcumin for the Prevention of Epithelial-Mesenchymal Transition in Endoxifen-Treated MCF-7 Breast Cancer Cel. Asian Pac. J. Cancer Prev. 2018, 19, 1243–1249. [Google Scholar] [CrossRef] [PubMed]
  239. Mukherjee, S.; Mazumdar, M.; Chakraborty, S.; Manna, A.; Saha, S.; Khan, P.; Bhattacharjee, P.; Guha, D.; Adhikary, A.; Mukhjerjee, S.; et al. Curcumin inhibits breast cancer stem cell migration by amplifying the E-cadherin/β-catenin negative feedback loop. Stem Cell Res. Ther. 2014, 5, 116. [Google Scholar] [CrossRef] [Green Version]
  240. Gallardo, M.; Calaf, G.M. Curcumin and epithelial-mesenchymal transition in breast cancer cells transformed by low doses of radiation and estrogen. Int. J. Oncol. 2016, 48, 2534–2542. [Google Scholar] [CrossRef] [Green Version]
  241. Han, C.; Fu, Y.X. β-Catenin regulates tumor-derived PD-L1. J. Exp. Med. 2020, 217. [Google Scholar] [CrossRef] [PubMed]
  242. Tsukita, Y.; Fujino, N.; Miyauchi, E.; Saito, R.; Fujishima, F.; Itakura, K.; Kyogoku, Y.; Okutomo, K.; Yamada, M.; Okazaki, T.; et al. Axl kinase drives immune checkpoint and chemokine signalling pathways in lung adenocarcinomas. Mol. Cancer 2019, 18, 24. [Google Scholar] [CrossRef] [Green Version]
  243. Song, S.; Yuan, P.; Wu, H.; Chen, J.; Fu, J.; Li, P.; Lu, J.; Wei, W. Dendritic cells with an increased PD-L1 by TGF-β induce T cell anergy for the cytotoxicity of hepatocellular carcinoma cells. Int. Immunopharmacol. 2014, 20, 117–123. [Google Scholar] [CrossRef]
  244. Heldin, C.H.; Landström, M.; Moustakas, A. Mechanism of TGF-beta signaling to growth arrest, apoptosis, and epithelial-mesenchymal transition. Curr. Opin. Cell Biol. 2009, 21, 166–176. [Google Scholar] [CrossRef] [PubMed]
  245. Hao, Y.; Baker, D.; Ten Dijke, P. TGF-β-Mediated Epithelial-Mesenchymal Transition and Cancer Metastasis. Int. J. Mol. Sci. 2019, 20, 2767. [Google Scholar] [CrossRef] [Green Version]
  246. Nakajima, A.; F Shuler, C.; Gulka, A.O.D.; Hanai, J.I. TGF-β Signaling and the Epithelial-Mesenchymal Transition during Palatal Fusion. Int. J. Mol. Sci. 2018, 19, 3638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Sow, H.S.; Ren, J.; Camps, M.; Ossendorp, F.; Ten Dijke, P. Combined Inhibition of TGF-β Signaling and the PD-L1 Immune Checkpoint Is Differentially Effective in Tumor Models. Cells 2019, 8, 320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Thakur, R.; Mishra, D.P. Pharmacological modulation of beta-catenin and its applications in cancer therapy. J. Cell. Mol. Med. 2013, 17, 449–456. [Google Scholar] [CrossRef]
  249. Shukla, S.; Gupta, S. Apigenin: A promising molecule for cancer prevention. Pharm. Res. 2010, 27, 962–978. [Google Scholar] [CrossRef] [PubMed]
  250. Meyer, H.; Bolarinwa, A.; Wolfram, G.; Linseisen, J. Bioavailability of apigenin from apiin-rich parsley in humans. Ann. Nutr. Metab. 2006, 50, 167–172. [Google Scholar] [CrossRef] [PubMed]
  251. Pápay, Z.E.; Kósa, A.; Böddi, B.; Merchant, Z.; Saleem, I.Y.; Zariwala, M.G.; Klebovich, I.; Somavarapu, S.; Antal, I. Study on the Pulmonary Delivery System of Apigenin-Loaded Albumin Nanocarriers with Antioxidant Activity. J. Aerosol Med. Pulm. Drug Deliv. 2017, 30, 274–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Wang, Y.C.; Huang, K.M. In vitro anti-inflammatory effect of apigenin in the Helicobacter pylori-infected gastric adenocarcinoma cells. Food Chem. Toxicol. 2013, 53, 376–383. [Google Scholar] [CrossRef]
  253. Ozçelik, B.; Kartal, M.; Orhan, I. Cytotoxicity, antiviral and antimicrobial activities of alkaloids, flavonoids, and phenolic acids. Pharm. Biol. 2011, 49, 396–402. [Google Scholar] [CrossRef]
  254. Yan, X.; Qi, M.; Li, P.; Zhan, Y.; Shao, H. Apigenin in cancer therapy: Anti-cancer effects and mechanisms of action. Cell Biosci. 2017, 7, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Gupta, S.; Afaq, F.; Mukhtar, H. Selective growth-inhibitory, cell-cycle deregulatory and apoptotic response of apigenin in normal versus human prostate carcinoma cells. Biochem. Biophys. Res. Commun. 2001, 287, 914–920. [Google Scholar] [CrossRef] [PubMed]
  256. Wang, W.; Heideman, L.; Chung, C.S.; Pelling, J.C.; Koehler, K.J.; Birt, D.F. Cell-cycle arrest at G2/M and growth inhibition by apigenin in human colon carcinoma cell lines. Mol. Carcinog. 2000, 28, 102–110. [Google Scholar] [CrossRef]
  257. Ujiki, M.B.; Ding, X.Z.; Salabat, M.R.; Bentrem, D.J.; Golkar, L.; Milam, B.; Talamonti, M.S.; Bell, R.H., Jr.; Iwamura, T.; Adrian, T.E. Apigenin inhibits pancreatic cancer cell proliferation through G2/M cell cycle arrest. Mol. Cancer 2006, 5, 76. [Google Scholar] [CrossRef] [Green Version]
  258. Harrison, M.E.; Power Coombs, M.R.; Delaney, L.M.; Hoskin, D.W. Exposure of breast cancer cells to a subcytotoxic dose of apigenin causes growth inhibition, oxidative stress, and hypophosphorylation of Akt. Exp. Mol. Pathol. 2014, 97, 211–217. [Google Scholar] [CrossRef] [PubMed]
  259. Choi, E.J.; Kim, G.H. Apigenin causes G(2)/M arrest associated with the modulation of p21(Cip1) and Cdc2 and activates p53-dependent apoptosis pathway in human breast cancer SK-BR-3 cells. J. Nutr. Biochem. 2009, 20, 285–290. [Google Scholar] [CrossRef]
  260. Lindenmeyer, F.; Li, H.; Menashi, S.; Soria, C.; Lu, H. Apigenin acts on the tumor cell invasion process and regulates protease production. Nutr. Cancer 2001, 39, 139–147. [Google Scholar] [CrossRef]
  261. Hu, X.W.; Meng, D.; Fang, J. Apigenin inhibited migration and invasion of human ovarian cancer A2780 cells through focal adhesion kinase. Carcinogenesis 2008, 29, 2369–2376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Liu, L.Z.; Fang, J.; Zhou, Q.; Hu, X.; Shi, X.; Jiang, B.H. Apigenin inhibits expression of vascular endothelial growth factor and angiogenesis in human lung cancer cells: Implication of chemoprevention of lung cancer. Mol. Pharmacol. 2005, 68, 635–643. [Google Scholar] [CrossRef] [Green Version]
  263. Coombs, M.R.; Harrison, M.E.; Hoskin, D.W. Apigenin inhibits the inducible expression of programmed death ligand 1 by human and mouse mammary carcinoma cells. Cancer Lett. 2016, 380, 424–433. [Google Scholar] [CrossRef]
  264. Lee, H.H.; Jung, J.; Moon, A.; Kang, H.; Cho, H. Antitumor and Anti-Invasive Effect of Apigenin on Human Breast Carcinoma through Suppression of IL-6 Expression. Int. J. Mol. Sci. 2019, 20, 3143. [Google Scholar] [CrossRef] [Green Version]
  265. Castellana, B.; Aasen, T.; Moreno-Bueno, G.; Dunn, S.E.; Ramón y Cajal, S. Interplay between YB-1 and IL-6 promotes the metastatic phenotype in breast cancer cells. Oncotarget 2015, 6, 38239–38256. [Google Scholar] [CrossRef] [PubMed]
  266. Rokavec, M.; Öner, M.G.; Li, H.; Jackstadt, R.; Jiang, L.; Lodygin, D.; Kaller, M.; Horst, D.; Ziegler, P.K.; Schwitalla, S.; et al. IL-6R/STAT3/miR-34a feedback loop promotes EMT-mediated colorectal cancer invasion and metastasis. J. Clin. Investig. 2014, 124, 1853–1867. [Google Scholar] [CrossRef] [Green Version]
  267. Sullivan, N.J.; Sasser, A.K.; Axel, A.E.; Vesuna, F.; Raman, V.; Ramirez, N.; Oberyszyn, T.M.; Hall, B.M. Interleukin-6 induces an epithelial-mesenchymal transition phenotype in human breast cancer cells. Oncogene 2009, 28, 2940–2947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  268. Xie, G.; Yao, Q.; Liu, Y.; Du, S.; Liu, A.; Guo, Z.; Sun, A.; Ruan, J.; Chen, L.; Ye, C.; et al. IL-6-induced epithelial-mesenchymal transition promotes the generation of breast cancer stem-like cells analogous to mammosphere cultures. Int. J. Oncol. 2012, 40, 1171–1179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  269. Banjerdpongchai, R.; Wudtiwai, B.; Khaw-On, P.; Rachakhom, W.; Duangnil, N.; Kongtawelert, P. Hesperidin from Citrus seed induces human hepatocellular carcinoma HepG2 cell apoptosis via both mitochondrial and death receptor pathways. Tumour Biol. 2016, 37, 227–237. [Google Scholar] [CrossRef] [Green Version]
  270. Lee, C.J.; Wilson, L.; Jordan, M.A.; Nguyen, V.; Tang, J.; Smiyun, G. Hesperidin suppressed proliferations of both human breast cancer and androgen-dependent prostate cancer cells. Phytother. Res. 2010, 24, S15–S19. [Google Scholar] [CrossRef]
  271. Haidari, F.; Heybar, H.; Jalali, M.T.; Ahmadi Engali, K.; Helli, B.; Shirbeigi, E. Hesperidin supplementation modulates inflammatory responses following myocardial infarction. J. Am. Coll. Nutr. 2015, 34, 205–211. [Google Scholar] [CrossRef] [PubMed]
  272. Xia, R.; Sheng, X.; Xu, X.; Yu, C.; Lu, H. Hesperidin induces apoptosis and G0/G1 arrest in human non-small cell lung cancer A549 cells. Int. J. Mol. Med. 2018, 41, 464–472. [Google Scholar] [CrossRef]
  273. Xia, R.; Xu, G.; Huang, Y.; Sheng, X.; Xu, X.; Lu, H. Hesperidin suppresses the migration and invasion of non-small cell lung cancer cells by inhibiting the SDF-1/CXCR-4 pathway. Life Sci. 2018, 201, 111–120. [Google Scholar] [CrossRef] [PubMed]
  274. Kongtawelert, P.; Wudtiwai, B.; Shwe, T.H.; Pothacharoen, P.; Phitak, T. Inhibitory Effect of Hesperidin on the Expression of Programmed Death Ligand (PD-L1) in Breast Cancer. Molecules 2020, 25, 252. [Google Scholar] [CrossRef] [Green Version]
  275. Huang, X.; Zhu, H.L. Resveratrol and its analogues: Promising antitumor agents. Anti-Cancer Agents Med. Chem. 2011, 11, 479–490. [Google Scholar] [CrossRef]
  276. Rauf, A.; Imran, M.; Suleria, H.A.R.; Ahmad, B.; Peters, D.G.; Mubarak, M.S. A comprehensive review of the health perspectives of resveratrol. Food Funct. 2017, 8, 4284–4305. [Google Scholar] [CrossRef]
  277. Yousef, M.; Vlachogiannis, I.A.; Tsiani, E. Effects of Resveratrol against Lung Cancer: In Vitro and In Vivo Studies. Nutrients 2017, 9, 1231. [Google Scholar] [CrossRef] [Green Version]
  278. Sinha, D.; Sarkar, N.; Biswas, J.; Bishayee, A. Resveratrol for breast cancer prevention and therapy: Preclinical evidence and molecular mechanisms. Semin. Cancer Biol. 2016, 40–41, 209–232. [Google Scholar] [CrossRef] [PubMed]
  279. Chen, K.Y.; Chen, C.C.; Chang, Y.C.; Chang, M.C. Resveratrol induced premature senescence and inhibited epithelial-mesenchymal transition of cancer cells via induction of tumor suppressor Rad9. PLoS ONE 2019, 14, e0219317. [Google Scholar] [CrossRef] [Green Version]
  280. Wen, F.C.; Chang, T.W.; Tseng, Y.L.; Lee, J.C.; Chang, M.C. hRAD9 functions as a tumor suppressor by inducing p21-dependent senescence and suppressing epithelial-mesenchymal transition through inhibition of Slug transcription. Carcinogenesis 2014, 35, 1481–1490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  281. Yin, Y.; Zhu, A.; Jin, Y.J.; Liu, Y.X.; Zhang, X.; Hopkins, K.M.; Lieberman, H.B. Human RAD9 checkpoint control/proapoptotic protein can activate transcription of p21. Proc. Natl. Acad. Sci. USA 2004, 101, 8864–8869. [Google Scholar] [CrossRef] [Green Version]
  282. Panigrahi, S.K.; Hopkins, K.M.; Lieberman, H.B. Regulation of NEIL1 protein abundance by RAD9 is important for efficient base excision repair. Nucleic Acids Res. 2015, 43, 4531–4546. [Google Scholar] [CrossRef] [Green Version]
  283. Tsai, J.H.; Hsu, L.S.; Lin, C.L.; Hong, H.M.; Pan, M.H.; Way, T.D.; Chen, W.J. 3,5,4’-Trimethoxystilbene, a natural methoxylated analog of resveratrol, inhibits breast cancer cell invasiveness by downregulation of PI3K/Akt and Wnt/β-catenin signaling cascades and reversal of epithelial-mesenchymal transition. Toxicol. Appl. Pharmacol. 2013, 272, 746–756. [Google Scholar] [CrossRef] [PubMed]
  284. Du, Z.; Yan, D.; Li, Z.; Gu, J.; Tian, Y.; Cao, J.; Tang, Z. Genes Involved in the PD-L1 Pathway Might Associate with Radiosensitivity of Patients with Gastric Cancer. J. Oncol. 2020, 2020, 7314195. [Google Scholar] [CrossRef]
  285. Shi, X.P.; Miao, S.; Wu, Y.; Zhang, W.; Zhang, X.F.; Ma, H.Z.; Xin, H.L.; Feng, J.; Wen, A.D.; Li, Y. Resveratrol sensitizes tamoxifen in antiestrogen-resistant breast cancer cells with epithelial-mesenchymal transition features. Int. J. Mol. Sci. 2013, 14, 15655–15668. [Google Scholar] [CrossRef] [PubMed]
  286. Jin, X.; Wei, Y.; Liu, Y.; Lu, X.; Ding, F.; Wang, J.; Yang, S. Resveratrol promotes sensitization to Doxorubicin by inhibiting epithelial-mesenchymal transition and modulating SIRT1/β-catenin signaling pathway in breast cancer. Cancer Med. 2019, 8, 1246–1257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  287. Verdura, S.; Cuyàs, E.; Cortada, E.; Brunet, J.; Lopez-Bonet, E.; Martin-Castillo, B.; Bosch-Barrera, J.; Encinar, J.A.; Menendez, J.A. Resveratrol targets PD-L1 glycosylation and dimerization to enhance antitumor T-cell immunity. Aging 2020, 12, 8–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  288. Huang, Y.; Chen, L.; Feng, L.; Guo, F.; Li, Y. Characterization of total phenolic constituents from the stems of Spatholobus suberectus using LC-DAD-MS(n) and their inhibitory effect on human neutrophil elastase activity. Molecules 2013, 18, 7549–7556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  289. Ha, H.; Shim, K.S.; An, H.; Kim, T.; Ma, J.Y. Water extract of Spatholobus suberectus inhibits osteoclast differentiation and bone resorption. BMC Complement. Altern. Med. 2013, 13, 112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  290. Wang, Z.; Wang, D.; Han, S.; Wang, N.; Mo, F.; Loo, T.Y.; Shen, J.; Huang, H.; Chen, J. Bioactivity-guided identification and cell signaling technology to delineate the lactate dehydrogenase A inhibition effects of Spatholobus suberectus on breast cancer. PLoS ONE 2013, 8, e56631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  291. Sun, J.Q.; Zhang, G.L.; Zhang, Y.; Nan, N.; Sun, X.; Yu, M.W.; Wang, H.; Li, J.P.; Wang, X.M. Spatholobus suberectus Column Extract Inhibits Estrogen Receptor-Positive Breast Cancer via Suppressing ER MAPK PI3K/AKT Pathway. Evid. Based Complement. Altern. Med. 2016, 2016, 2934340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Peng, F.; Xiong, L.; Peng, C. (-)-Sativan Inhibits Tumor Development and Regulates miR-200c/PD-L1 in Triple-Negative Breast Cancer Cells. Front. Pharmacol. 2020, 11, 251. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Tumor-intrinsic PD-L1 signaling in cancer initiation and development. The diagram highlights downstream signaling of PD-L1 activation in cancer. Hypoxia-inducible factors, HIF; interferon regulatory factor1, IRF1; MYC proto-oncogene, bHLH transcription factor, Myc; Janus kinase, JAK; signal transducer and activator of transcription (STAT)1/3; nuclear factor-kappa B, NF-ƙB; bromodomain-containing protein 4, BRD4; interferon-gamma, IFN-γ; IFN-γ receptor 1/2, IFNGR1/2; phosphoinositide 3-kinase, PI3K; protein kinase B, AKT; mammalian target of rapamycin, mTOR; extracellular-signal-regulated kinase, ERK; mitogen-activated protein kinase, MEK; B-Raf Serine/Threonine-Protein, BRAF; rat sarcoma, Ras; epidermal growth factor, EGF; hepatocyte growth factor HGF; programmed death-ligand 1, PD-L1.
Figure 1. Tumor-intrinsic PD-L1 signaling in cancer initiation and development. The diagram highlights downstream signaling of PD-L1 activation in cancer. Hypoxia-inducible factors, HIF; interferon regulatory factor1, IRF1; MYC proto-oncogene, bHLH transcription factor, Myc; Janus kinase, JAK; signal transducer and activator of transcription (STAT)1/3; nuclear factor-kappa B, NF-ƙB; bromodomain-containing protein 4, BRD4; interferon-gamma, IFN-γ; IFN-γ receptor 1/2, IFNGR1/2; phosphoinositide 3-kinase, PI3K; protein kinase B, AKT; mammalian target of rapamycin, mTOR; extracellular-signal-regulated kinase, ERK; mitogen-activated protein kinase, MEK; B-Raf Serine/Threonine-Protein, BRAF; rat sarcoma, Ras; epidermal growth factor, EGF; hepatocyte growth factor HGF; programmed death-ligand 1, PD-L1.
Nutrients 13 01718 g001
Figure 2. PD-L1-mediated EMT stimulation. The diagram highlights the downstream signaling of EMT in cancer. Interferon-gamma, IFN-γ; IFN-γ receptor 1/2, IFNGR1/2; epidermal growth factor, EGF; hepatocyte growth factor HGF; Janus kinase, JAK; signal transducer and activator of tran-scription3, STAT3; nuclear factor-kappa B, NF-ƙB; phosphoinositide 3-kinase, PI3K; protein kinase B, AKT; mammalian target of rapamycin, mTOR; zinc finger E-box binding homeobox 1/2, Zeb1/2; Snail family transcriptional repressor 1, Snail 1; extracellular-signal-regulated kinase, ERK; mitogen-activated protein kinase, MEK; B-Raf Serine/Threonine-Protein, BRAF; rat sarcoma, Ras; programmed death-ligand 1, PD-L1; transforming growth factor-beta, TGF-β; mothers against decapentaplegic, Smad; epithelial-mesenchymal transition, EMT.
Figure 2. PD-L1-mediated EMT stimulation. The diagram highlights the downstream signaling of EMT in cancer. Interferon-gamma, IFN-γ; IFN-γ receptor 1/2, IFNGR1/2; epidermal growth factor, EGF; hepatocyte growth factor HGF; Janus kinase, JAK; signal transducer and activator of tran-scription3, STAT3; nuclear factor-kappa B, NF-ƙB; phosphoinositide 3-kinase, PI3K; protein kinase B, AKT; mammalian target of rapamycin, mTOR; zinc finger E-box binding homeobox 1/2, Zeb1/2; Snail family transcriptional repressor 1, Snail 1; extracellular-signal-regulated kinase, ERK; mitogen-activated protein kinase, MEK; B-Raf Serine/Threonine-Protein, BRAF; rat sarcoma, Ras; programmed death-ligand 1, PD-L1; transforming growth factor-beta, TGF-β; mothers against decapentaplegic, Smad; epithelial-mesenchymal transition, EMT.
Nutrients 13 01718 g002
Figure 3. The mechanism of curcumin-mediated programmed death-ligand 1 (PD-L1) inhibition in breast cancer cells. Signal transducer and activator of transcription, STAT; Janus kinase, JAK; nuclear factor-kappa β, NF-ƙB; smoothened, frizzled class receptor, Smo; interferon-gamma, IFN-γ; Phosphoinositide 3-kinase, PI3K; protein kinase β, AKT; mammalian target of rapamycin, mTOR; mitogen-activated protein kinase, MAPK; B-raf serine/threonine-protein, Braf; rat sarcoma, Ras; transforming growth factor-β, TGF-β; mothers against decapentaplegic, Smad; wingless-related integration site, Wnt; zinc finger E-box binding homeobox, ZEB; snail family transcriptional repressor, Snail; inhibitor of kappa light polypeptide gene enhancer in β-cells, kinase beta, IKKβ; programmed death-ligand 1, PD-L1; AXL receptor tyrosine kinase, AXL.
Figure 3. The mechanism of curcumin-mediated programmed death-ligand 1 (PD-L1) inhibition in breast cancer cells. Signal transducer and activator of transcription, STAT; Janus kinase, JAK; nuclear factor-kappa β, NF-ƙB; smoothened, frizzled class receptor, Smo; interferon-gamma, IFN-γ; Phosphoinositide 3-kinase, PI3K; protein kinase β, AKT; mammalian target of rapamycin, mTOR; mitogen-activated protein kinase, MAPK; B-raf serine/threonine-protein, Braf; rat sarcoma, Ras; transforming growth factor-β, TGF-β; mothers against decapentaplegic, Smad; wingless-related integration site, Wnt; zinc finger E-box binding homeobox, ZEB; snail family transcriptional repressor, Snail; inhibitor of kappa light polypeptide gene enhancer in β-cells, kinase beta, IKKβ; programmed death-ligand 1, PD-L1; AXL receptor tyrosine kinase, AXL.
Nutrients 13 01718 g003
Figure 4. The mechanisms of Apigenin-mediated programmed death-ligand 1 (PD-L1) inhibition in breast cancer cells. Interferon-gamma, IFN-γ; Interleukin 6, IL-6; Janus kinase, JAK; signal transducer and activator of transcription1, STAT1; major histocompatibility complex, MHC; T-cell receptor, TCR; programmed cell death protein 1, PD-1.
Figure 4. The mechanisms of Apigenin-mediated programmed death-ligand 1 (PD-L1) inhibition in breast cancer cells. Interferon-gamma, IFN-γ; Interleukin 6, IL-6; Janus kinase, JAK; signal transducer and activator of transcription1, STAT1; major histocompatibility complex, MHC; T-cell receptor, TCR; programmed cell death protein 1, PD-1.
Nutrients 13 01718 g004
Figure 5. The mechanisms of hesperidin-mediated programmed death-ligand 1 (PD-L1) inhibition in breast cancer cells. Protein kinase B, AKT; nuclear factor-kappa B, NF-ƙB.
Figure 5. The mechanisms of hesperidin-mediated programmed death-ligand 1 (PD-L1) inhibition in breast cancer cells. Protein kinase B, AKT; nuclear factor-kappa B, NF-ƙB.
Nutrients 13 01718 g005
Figure 6. The mechanisms of resveratrol-mediated programmed death-ligand 1 (PD-L1) inhibition in breast cancer cells. Interferon-gamma, IFN-γ; IFN-γ receptor 1/2, IFNGR1/2; transforming growth factor-beta, TGF-β; Janus kinase, JAK; signal transducer and activator of transcription, STAT; phosphoinositide 3-kinase, PI3K; protein kinase B, AKT; mitogen-activated protein kinase, MAPK.
Figure 6. The mechanisms of resveratrol-mediated programmed death-ligand 1 (PD-L1) inhibition in breast cancer cells. Interferon-gamma, IFN-γ; IFN-γ receptor 1/2, IFNGR1/2; transforming growth factor-beta, TGF-β; Janus kinase, JAK; signal transducer and activator of transcription, STAT; phosphoinositide 3-kinase, PI3K; protein kinase B, AKT; mitogen-activated protein kinase, MAPK.
Nutrients 13 01718 g006
Figure 7. The mechanisms of sativan-mediated programmed death-ligand 1 (PD-L1) inhibition in breast cancer cells. Signal transducer and activator of transcription, STAT; Janus kinase, JAK; Phosphoinositide 3-kinase, PI3K; protein kinase B, AKT; mammalian target of rapamycin, mTOR; mitogen-activated protein kinase, MAPK; B-Raf Serine/Threonine-Protein, BRAF; rat sarcoma, Ras; interferon-gamma, IFN-γ; IFN-γ receptor 1/2, IFNGR1/2; Snail family transcriptional repressor, Snail; Epithelial-to-Mesenchymal Transition, EMT.
Figure 7. The mechanisms of sativan-mediated programmed death-ligand 1 (PD-L1) inhibition in breast cancer cells. Signal transducer and activator of transcription, STAT; Janus kinase, JAK; Phosphoinositide 3-kinase, PI3K; protein kinase B, AKT; mammalian target of rapamycin, mTOR; mitogen-activated protein kinase, MAPK; B-Raf Serine/Threonine-Protein, BRAF; rat sarcoma, Ras; interferon-gamma, IFN-γ; IFN-γ receptor 1/2, IFNGR1/2; Snail family transcriptional repressor, Snail; Epithelial-to-Mesenchymal Transition, EMT.
Nutrients 13 01718 g007
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

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. https://0-doi-org.brum.beds.ac.uk/10.3390/nu13051718

AMA Style

Messeha SS, Zarmouh NO, Soliman KFA. Polyphenols Modulating Effects of PD-L1/PD-1 Checkpoint and EMT-Mediated PD-L1 Overexpression in Breast Cancer. Nutrients. 2021; 13(5):1718. https://0-doi-org.brum.beds.ac.uk/10.3390/nu13051718

Chicago/Turabian Style

Messeha, Samia S., Najla O. Zarmouh, and Karam F. A. Soliman. 2021. "Polyphenols Modulating Effects of PD-L1/PD-1 Checkpoint and EMT-Mediated PD-L1 Overexpression in Breast Cancer" Nutrients 13, no. 5: 1718. https://0-doi-org.brum.beds.ac.uk/10.3390/nu13051718

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