Next Article in Journal
Lactoferrins in Their Interactions with Molecular Targets: A Structure-Based Overview
Previous Article in Journal
Metformin and Breast Cancer: Current Findings and Future Perspectives from Preclinical and Clinical Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Adenosine Increases the Immunosuppressive Capacity of Cervical Cancer Cells by Increasing PD-L1 Expression and TGF-β Production through Its Interaction with A2AR/A2BR

by
Rosario García-Rocha
1,2,
Alberto Monroy-García
1,2,
Ana Luisa Vázquez-Cruz
1,
Luis Antonio Marín-Aquino
2,3,
Benny Weiss-Steider
1,
Jorge Hernández-Montes
1,
Christian Azucena Don-López
1,
Gabriela Molina-Castillo
1,4 and
María de Lourdes Mora-García
1,*
1
Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer-UMIEZ, FES-Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
2
Laboratorio de Inmunología y Cáncer, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México 06720, Mexico
3
Doctorado en Ciencias en Inmunología, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico
4
Programa de Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(3), 397; https://doi.org/10.3390/ph17030397
Submission received: 25 January 2024 / Revised: 10 March 2024 / Accepted: 16 March 2024 / Published: 19 March 2024
(This article belongs to the Special Issue Small Molecules in Cancer Immunotherapy)

Abstract

:
The present study provides evidence showing that adenosine (Ado) increases the expression of programmed death ligand 1 (PD-L1) in cervical cancer (CeCa) cells by interacting with A2AR/A2BR and that TGF-β1 acts in an autocrine manner to induce PD-L1 expression, enhancing the immunosuppressive effects of CeCa cells on activated T lymphocytes (ATLs) and CD8+ cytotoxic T lymphocytes (CTLs) specific for antigenic peptides derived from E6 and E7 proteins of HPV-16. Interestingly, the addition of the antagonists ZM241385 and MRS1754, which are specific for A2AR and A2BR, respectively, or SB-505124, which is a selective TGF-β1 receptor inhibitor, to CeCa cell cultures significantly inhibited PD-L1 expression. In addition, supernatants from CeCa cells that were treated with Ado (CeCa-Ado Sup) increased the expression of PD-1, TGF-β1, and IL-10 and decreased the expression of IFN-γ in ATLs. Interestingly, the addition of an anti-TGF-β neutralizing antibody strongly reversed the effect of CeCa-Ado Sup on PD-1 expression in ATLs. These results strongly suggest the presence of a feedback mechanism that involves the adenosinergic pathway, the production of TGF-β1, and the upregulation of PD-L1 expression in CeCa cells that suppresses the antitumor response of CTLs. The findings of this study suggest that this pathway may be clinically important and may be a therapeutic target.

1. Introduction

Cervical cancer (CeCa) ranks fourth in incidence and mortality among all types of cancer worldwide. In 2020, approximately 600,000 new cases of CeCa and more than 340,000 deaths due to CeCa were recorded. According to data from the WHO, approximately 90% of deaths associated with CeCa occur in developing countries [1,2]. Persistent infection caused by high-risk human papillomaviruses (HR-HPV), such as the HPV-16 and HPV-18 genotypes, which are present in approximately 50% and 20% of CeCa patients, respectively, can trigger progression from low-grade cervical intraepithelial neoplasia (CIN I) to high-grade neoplasia intraepithelial cervical (CIN II-III) and eventually to CeCa [3]. The immune response against HPV antigens can eliminate most precursor infections and lesions; however, some women infected with HR-HPV genotypes develop cancer, suggesting that other risk factors may be involved [4].
Several studies have proposed that immunosuppression mechanisms may play a relevant role in the development of CeCa. In this context, several findings have shown that programmed death ligand 1 (PD-L1), which is expressed on the surface of different types of cancer cells, can induce immunosuppressive mechanisms to enable tumors to evade the host’s anticancer immune responses [5,6]. Additionally, studies have shown that the immune checkpoint receptor of PD-L1, namely, programmed cell death 1 (PD-1), which is primarily expressed on T cells, monocytes, macrophages, and natural killer cells, could be a critical negative regulator of cancer biology and may have the ability to support cancer development, growth, invasion, and metastasis [7]. PD-L1 expression on the surface of tumor cells can attenuate the ability of the immune system to successfully eliminate tumor cells because the binding of the PD-1 receptor to its ligands, namely, PD-L1 and/or PD-L2, negatively regulates T-cell receptor (TCR) signaling, resulting in the inhibition of cytotoxic T lymphocytes [8]. In this sense, blocking the PD-L1/PD-1 pathway through the use of therapeutic antibodies has shown important antitumor effects in patients with advanced cancers [9]. However, the response rates to anti-PD-L1 therapy in several solid tumors have been limited, with a response ranging between 10% and 40% [10]. Therefore, understanding the regulation of PD-L1 expression is relevant for improving the efficacy of immune checkpoint blockade-based immunotherapy. Recent studies have reported that the expression of PD-L1 is intricately regulated by various processes, such as gene transcription, posttranscriptional and posttranslational modifications (PTMs), and exosomal transport [11]. Among the mechanisms that regulate PD-L1 expression, the production of cytokines, such as IL-1a, IL-27, IL-10, and IL-32g, in the tumor microenvironment (TME) can upregulate the expression of PD-L1 on monocytes and tumor cells [12,13]. Similarly, some studies have shown that TGF-β also induces the expression of PD-L1 in different tumors [14,15,16], as well as in human and murine fibroblasts in a Smad2/3- and YAP/TAZ-dependent manner [17]. In addition, TGF-β plays a crucial role in the development of CeCa by establishing a local immunosuppressive microenvironment in the cervix during HR-HPV infection and inhibiting the proliferation and activation of T lymphocytes with antitumor activity [18]. In the particular case of CeCa, a tendency to increase the expression of PD-L1 has been observed in CIN or CeCa patients with HR-HPV infection [19,20,21], a previous history of chemotherapy [20], metastatic tumors [21], a history of multiple parity and abortion [22], an advanced stage [23], poorly differentiated tumors [24], and large tumors [24]. In addition, studies have revealed that the expression of PD-L1 and PD-1 in cervical DCs and T cells, respectively, is associated with HR-HPV positivity and with the degree of CeCa development [25].
Moreover, recent reports indicate that the adenosinergic pathway plays an important role in the pathogenesis of gynecological cancers, including CeCa [26,27]. Through this process, ATP/ADP nucleotides, which are present at high concentrations (>100 μM) in the TME [28], are converted to adenosine monophosphate (AMP) via the hydrolytic activity of the ectoenzyme CD39 (ectonucleoside triphosphate diphosphohydrolase-1, ENTPD1; EC 3.6.1.5) and later to Ado by 5′-ectonucleotidase (CD73, EC 3.1.3.5) [29,30]. The main signaling effects of Ado are mediated by specific adenosine receptors (ARs) that are localized in the membranes of target cells, where they are coupled to G proteins. These ARs are divided into four subtypes: A1R, A2AR, A2BR, and A3R [31]. In the TME, Ado exerts immunosuppressive effects on CTLs, dendritic cells, NK cells, and B cells through interactions with the high-affinity receptor A2AR [32]. In addition, through interaction with A1R, A2AR, and A2BR, Ado can also promote tumor growth by inducing the proliferation, invasion, and metastasis of tumor cells [33,34]. For these reasons, adenosine signaling has emerged as a target in cancer immunotherapy due to its key role in the metabolic pathway that regulates tumor immunity [34]. In previous studies, we reported that Ado induces the production of TGF-β1 by CeCa cells by interacting with ARs, specifically A2AR and A2BR [35], and that this cytokine strongly contributes to enhancing protumoral characteristics related to tumor progression, such as migration and invasion, immune evasion, immunosuppression, and chemoresistance [36]. Considering these findings, in this study, we analyzed the role of Ado in the induction of PD-L1 expression in CeCa cells as well as the ability of these cells to inhibit the activation and cytotoxic activity of CD8+ T lymphocytes. We found that Ado induced PD-L1 expression in CeCa cells through its interaction with A2AR/A2BR and the production of TGF-β. In addition, supernatants derived from CeCa cells that were treated with Ado increased the expression of PD-1, TGF-β1, and IL-10 and decreased the expression of IFN-γ in activated CD8+ T cells, suggesting that this pathway may be clinically important and may be a therapeutic target in CeCa.

2. Results

2.1. Ado Induces PD-L1 Expression in CeCa Cells through Its Interaction with A2AR/A2BR and the Autocrine Production of TGF-β1

Our research group previously reported that CeCa cells inhibit the effector functions of CD8+ cytotoxic T lymphocytes through the production of high amounts of Ado [37] and that this nucleoside induces the expression and production of TGF-β1 in CeCa cells by interacting with A2AR/A2BR [35]. Because TGF-β1 is associated with the expression of PD-L1 in different tumors [14,15,16], in this study, we analyzed the effect of Ado on the production of TGF-β1 and on the induction of PD-L1 expression in CeCa cells. To this end, 1 × 105 CaSki and HeLa cells were cultured in the presence of 0.1 mM or 1 mM Ado for 72 h. The baseline expression of PD-L1 in CeCa cells served as the Ctl (Figure 1a,b). Ado increased the expression of PD-L1 in both cell lines, and this effect depended on the duration of culture with Ado and the concentration of this nucleoside. The highest expression of this molecule was detected after culture with 1 mM Ado for 72 h (Figure 1a,b). Therefore, these cell culture conditions were used in subsequent experiments.
To analyze whether A2AR or A2BR participates in the Ado signaling-induced expression of PD-L1, CeCa cells were cultured for 72 h in the presence or absence of 10 µM ZM241385 and MRS1754, which are selective antagonists for A2AR and A2BR, respectively, and in the presence or absence of 1 mM Ado. Blocking A2AR significantly reduced the expression of PD-L1 in CaSki and HeLa cells; however, blocking A2BR significantly reduced the expression of PD-L1 in only HeLa cells (Figure 2a and Figure S1). To confirm the participation of A2AR and A2BR in the signaling-induced expression of PD-L1, CeCa cells were cultured for 72 h in the presence of 10 μM CGS21680, a selective A2 adenosine receptor agonist. Interestingly, CGS21680 induced a significant increase in PD-L1 expression in CaSki and HeLa cells, and this increase was comparable to that observed in CeCa cells cultured in the presence of 1 mM Ado (Figure S2).
In contrast, CaSki and HeLa cells cultured in the presence of 1 mM Ado exhibited increased TGF-β1 production. In fact, after 24 h of culture, the TGF-β1 concentrations in the supernatants of both CeCa cell lines were significantly greater than those in the supernatants of cells that were cultured without Ado (black bars), which were used as a Ctl. However, although the levels of TGF-β1 in the CaSKi cell supernatants remained high during 72 h of culture, the levels of this cytokine in the HeLa cell supernatants decreased significantly after 24 h of culture (Figure 2b).
To determine whether TGF-β1 induces the expression of PD-L1, CeCa cells were cultured for 24, 48, and 72 h in the presence of 20 ng/mL recombinant human TGF-β1 (rHuTGF-β1). Initially, the expression of the TGF-β1 type 2 receptor (TGF-β1RII)) in CeCa cells was analyzed. The baseline expression of TGF-β1RII in HeLa cells was 4 times greater than that in CaSki cells, and the mean fluorescence intensity (MFI) values obtained for these cells were 3631 and 892, respectively. However, the culture of CaSki and HeLa cells for 72 h in the presence of 1 mM Ado doubled the expression of TGF-β1RII, as revealed by MFI values of 6313 and 2130, respectively (Figure S3). After 24 h of culture, the expression of PD-L1 was significantly increased in both cell lines; however, over time, the expression of PD-L1 gradually decreased (Figure 2c). To determine whether TGF-β1 acts in an autocrine manner to induce PD-L1 expression, CeCa cells were cultured in the presence or absence of 1 mM Ado and in the presence or absence of 25 μM SB-505124, which is a selective inhibitor of TGF-β1R. The addition of SB-505124 to the cell cultures reversed the Ado-induced expression of PD-L1 by more than 80% in CaSki cells and by approximately 40% in HeLa cells (Figure 2d). These results suggest that the production of TGF-β1 after the interaction of Ado with A2AR and A2BR is important for inducing PD-L1 expression in CeCa cells.

2.2. Ado-Treated CeCa Cells Inhibit the Proliferation of CD8+ T Lymphocytes through PD-L1 Expression

To analyze the PD-L1-mediated immunosuppressive effect of CeCa cells, CaSki and HeLa cells were cultured in the presence or absence of 1 mM Ado (CaSki-Ado and HeLa-Ado) for 72 h. The cells were subsequently fixed with paraformaldehyde, washed, and cocultured with ATLs at a CeCa cell:ATL ratio of 1:5 for 96 h. The level of proliferation exhibited by ATLs that were cultured alone was considered the positive control level. The proliferation indices (PIs) of the ATLs that were determined at the beginning of the culture were set to 1. After 96 h of culture, the PIs of the ATLs were 2.25–2.5 (Figure 3a,b, black line). The addition of CaSki or HeLa cells to the cultures blocked the proliferation of ATLs; in fact, after 96 h of cocultivation, the PIs of the ATLs were 1.75 and 1, respectively (Figure 3a,b, red line). The addition of CaSki-Ado or HeLa-Ado to ATL cultures resulted in greater inhibition of ATL proliferation; after 96 h of coculture, the PIs were 0.9 and 0.7, respectively (Figure 3a,b, blue line). Interestingly, the addition of anti-PD-L1 antibodies to the ATL cultures significantly reversed the CaSki-Ado- and HeLa-Ado-mediated inhibition of ATL proliferation, with PIs of 2.25 and 1.5, respectively (Figure 3a,b, green line). Importantly, CeCa cells that were cultured in the presence or absence of Ado and then cultured with ATLs did not proliferate when they were cultured alone for 96 h. These results suggest that Ado strongly increases the immunosuppressive capacity of CeCa cells through the induction of PD-L1 expression. To corroborate this effect, CaSki cells or CaSki-Ado cells, previously fixed with paraformaldehyde, were cocultured for 48 h at a ratio of 1:5 with CTLs that were previously stimulated with antigenic peptides derived from the E6 and E7 proteins of HPV-16, which specifically bind to the HLA-A2 allele and which we previously analyzed in CaSki cells [38,39,40]. A neutralizing anti-PD-L1 antibody was added to some cocultures. CTLs cultured in the absence of CaSki cells were used as a control (Ctl). Subsequently, CaSki cells labeled with CFSE and 7-AAD were challenged with effector cells at a ratio of 1:2. Interestingly, the coculture with CaSki or CaSki-Ado reduced the cytotoxic activity of CTLs on CaSki cells by approximately 50% compared with the Ctl; however, the addition of anti-PD-L1 antibodies to the cocultures blocked the suppressive effect of CaSki or CaSki-Ado cells (Figure 4).

2.3. Supernatants Derived from CeCa/Ado Cells Strongly Suppress the Activation of CD8+ T Lymphocytes

Increased expression of PD-1/PD-L1 pathway components is correlated with impaired cell-mediated immunity in patients with HR-HPV/related cervical intraepithelial neoplasia [25]. In fact, the proportion of Th1/Th2 cytokines in cervical exudates is correlated with HR-HPV positivity and the CIN grade [41]. The induction of PD-1/PD-L1 can also be regulated by TGF-β [25,42], and Ado induces TGF-β production by CeCa cells [35]. In this study, we also analyzed the effect of supernatants (Sup) from CeCa and CeCa-Ado cells on CD8+ T lymphocyte activation. For this purpose, 2.5 × 105 CD8+ T cells were cultured for 48 h with beads coated with anti-CD2/CD3/CD28 antibodies at a ratio of 2:1 with or without 30% Sup from CeCa cells or CeCa-Ado cells and with or without 1 μg/mL neutralizing anti-TGF-β1 antibody. The expression level of PD-1 in ATLs that were cultured without CeCa or CeCa-Ado Sup was considered the Ctl level. Compared with the Ctl treatment, treatment with Sup derived from CeCa cells increased PD-1 expression in ATLs by 30–60%, whereas treatment with Sup derived from CeCa-Ado cells increased PD-1 expression by approximately 100%. Interestingly, the addition of an anti-TGF-β neutralizing antibody strongly reversed (more than 70%) the CeCa-Ado Sup-induced increase in PD-1 expression by ATLs (Figure 5a).
Additionally, the effects of Sup derived from CeCa or CeCa-Ado cells on IFN-γ, TGF-β, and IL-10 production by ATLs were analyzed. Interestingly, Sup from CeCa and CeCa-Ado cells strongly inhibited the production of IFN-γ by ATLs compared with the IFN-γ production by ATLs that were cultured without Sup from CeCa or CeCa-Ado cells (Ctl). However, the addition of an anti-TGF-β neutralizing antibody did not reverse the inhibitory effect of Sup on IFN-γ production by ATLs (Figure 5b). In contrast, Sup from CeCa cells increased the expression of TGF-β (Figure 5c) and IL-10 (Figure 5d) in ATLs by 20–30%, whereas Sup from CeCa-Ado cells increased the expression of TGF-β (Figure 5c) and IL-10 (Figure 5d) in ATLs by 40–80%. The addition of an anti-TGF-β neutralizing antibody reversed the CeCa-Ado cell-derived Sup-induced increase in TGF-β and IL-10 expression by more than 50% (Figure 5c,d). To confirm the effects of TGF-β on the expression of PD-1, IFN-γ, IL-10, and TGF-β in ATLs, CD8+ T lymphocytes were cultured for 48 h in the presence of beads coated with anti-CD2/CD3/CD28 antibodies at a ratio of 2:1 and in the presence or absence of 20 ng/mL rHuTGF-β. We observed an increase in the expression of PD-1 of more than 60% (Figure 6a), a reduction in the expression of IFN-γ of more than 20% (Figure 6b), and increases in the expression of both TGF-β by more than 50% (Figure 6c) and IL-10 by more than 40% (Figure 6d) in ATLs that were cultured in the presence of rHuTGF-β1 compared with the levels in ATLs that were cultured without this cytokine. Our results suggest that adenosine increases the immunosuppressive capacity of CeCa cells via the production of TGF-β, which acts in an autocrine manner to induce the expression of PD-L1 on CeCa cells and acts in a paracrine manner to inhibit the activation of CD8+ lymphocytes through the induction of PD-1 expression, resulting in decreased production of IFN-γ and increased production of TGF-β and IL-10.

3. Discussion

Several studies have shown that the expression of PD-L1/PD-1 is associated with HPV-related cancer, especially cancers associated with HPV-16 and HPV-18, which account for approximately 70% of cervical cancer cases [19,20,21]. It has been reported that the HPV E5/E6/E7 oncogenes activate multiple signaling pathways, including the hypoxia-inducible factor 1α, STAT3/NF-kB, PI3K/AKT, MAPK, and microRNA pathways, which can regulate PD-L1/PD-1 expression, leading to suppression of the adaptive immune response and allowing progression of the disease [43]. The expression of PD-L1 is increased in CIN1 and CINII-III as well as in HR-HPV-positive CeCa [19,25,44]. In fact, the proportions of CeCa tumors that express PD-L1 and PD-1 are 34.4–96% [25,43] and 46.97–60.82% [19,20], respectively, suggesting that the treatment of CeCa with checkpoint inhibitors may be a promising approach. However, understanding the mechanisms that promote PD-L1 expression in the TME is highly important because this pathway has been successfully targeted in cancer therapy. Hence, the current drug development strategies aim to overcome the failures of many drugs that are designed to block the PD-1/PD-L1 pathway and to address relapses that can occur in cancer patients after initial tumor regression [44,45].
PD-L1 can be expressed by various types of cells that reside in the TME, including tumor cells, immune cells, endothelial cells, and even immunosuppressor cells. Therefore, the expression of this ligand in tumors has been proposed to be a biomarker that can be used to predict the therapeutic effects of anti-PD-1 and anti-PD-L1 drugs [46]. It has been proposed that tumor cells express PD-L1 to mediate innate or adaptive resistance to immunity [47]. Innate resistance refers to the constitutive expression of PD-L1 in tumor cells due to amplification of the PDL1 gene or aberrant activation of oncogenic signaling pathways [48,49,50,51,52]; adaptive resistance to immunity refers to the expression of PD-L1 in tumors or immune cells in response to inflammatory factors that are secreted in the TME during antitumor immune responses, and IFN-γ is one of the main cytokines that is responsible for inducing the adaptive expression of PD-L1 [53]. However, several additional TME-resident cytokines that can upregulate PD-L1 expression, including IL-1a, IL-10, IL-27, and IL-32g [12,13,54,55], have been identified. Similarly, there is evidence showing that TGF-β regulates PD-L1/PD-1 expression in different tumor models [15,16,42] and that this cytokine promotes the enrichment of PD-L1 in exosomes derived from breast cancer tumor cells [56].
In previous studies, we found that CeCa cells infected with HPV-16 or HPV-18 produce greater amounts of Ado than do CeCa cells without HPV infection [37]. We also found that Ado induces the production of TGF-β1 in CeCa cells by interacting with A2AR/A2BR [35]. The present study provides the first lines of evidence showing that TGF-β1 production by CaSki and HeLa cells is induced by the interaction of Ado with A2AR and that A2BR acts in an autocrine manner to increase the expression of PD-L1 on the surface of these cells, promoting their immunosuppressive effects on CD8+ T lymphocytes. In addition, this factor promoted the expression of PD-1, strongly decreased the production of IFN-γ, and increased the production of TGF-β and IL-10 in activated CD8+ T lymphocytes (Figure 7).
Under the culture conditions used in this study, we observed that the increased expression of PD-L1 in CeCa cells was dependent on the presence of TGF-β in the supernatants. In fact, the increased TGF-β1 production that was observed after 24 h of culture corresponded with increased PD-L1 expression in CaSki and HeLa cells, and the significantly decreased PD-L1 expression that was observed after 72 h in both cell lines was associated with decreased TGF-β1 levels in the supernatants. This effect was also related to the increase in the expression of TGF-β1RII in CaSki and HeLa cells that were treated with Ado, which was doubled at 72 h. In addition, further analysis of whether adenosine-mediated signaling also has a feedback effect on the expression of A2AR and A2BR would be interesting, because Ado promotes angiogenesis, proliferation, migration, and metastasis of tumor cells via A2AR and A2BR [57].
In contrast, CaSki-Ado and HeLa-Ado supernatants also promoted the expression of PD-1, strongly decreased the production of IFN-γ, and increased the production of TGF-β and IL-10 in ATLs. However, although decreased expression of PD-1 or increased production of TGF-β and IL-10 in ATLs were largely reversed by the addition of the anti-TGF-β neutralizing antibody to the cultures, the decrease in IFN-γ production was only marginally reversed by the addition of this antibody, suggesting that other factors produced by Ado-treated CeCa cells play a strong role in the inhibition of T-cell activation by reducing the production of IFN-γ, consequently affecting the antitumor immune response. For example, in a recent study, CeCa patients with tumors expressing high PD-L1 or PD-L2 mRNA levels and exhibiting high IFN-γ signaling activity exhibited better overall survival than did CeCa patients with tumors expressing high PD-L1 expression and low IFN-γ activity [58], suggesting that the presence of factors that regulate IFN-γ production in the TME may play individual or synergistic roles in regulating PD-1 and PD-L1/PD-L2 expression [59].
The development of CeCa is strongly associated with the production of TGF-β1, whose expression is directly correlated with the degree of CeCa progression and antitumor immune response suppression [42]. TGF-β1 has been detected in the sera and tissues of patients infected with HPV-AR and in patients with CIN and CeCa [14,15,16,60]. TGF-β1 is an immunoregulatory factor involved in tumor progression and immunosuppression, and the sources of this factor in the TME include several types of stromal cells in addition to cancer cells [61,62]. We recently found that the secretion of TGF-β by Ado-treated CeCa cells maintains the expression of CD73 [35]. In this work, we observed that TGF-β produced by Ado-treated CeCa cells also induces the expression of PD-L1 in these cells, increasing the immunosuppressive effects of CeCa cells on CD8+ T cells. These findings suggest the existence of a feedback loop between adenosinergic activity and PD-L1 expression in cancer. In this feedback loop, the production of Ado and the signaling of this nucleoside in cancer cells can promote the expression of PD-L1 through TGF-β production, consequently favoring immune evasion and tumor progression in CeCa. In this regard, the correlation between PD-L1 and CD73 has been related to cancer prognosis [63,64], and blocking CD73 and PD-L1 has been shown to promote the T-cell response [65,66]; hence, these results offer a promising therapeutic strategy for cancer. In addition, through the design of bispecific recognition proteins, the inhibition of both TGF-β and PD-L1 was recently proposed for the treatment of cancer [67]. In this context, the use of bintrafusp alfa (BA), which is a protein that inhibits TGF-β and PD-L1, has been shown to decrease the size of tumors in a model of colorectal cancer [68]. Furthermore, encouraging results have been obtained in phase I clinical trials investigating the administration of this drug to patients with advanced CeCa [69]. Therefore, blocking the activity of the adenosinergic pathway to inhibit the production of Ado, as well as its signaling to induce the production of TGF-β and the subsequent expression of PD-L1 in the TME, could contribute to improving the design of combinatorial therapies against CeCa and other types of cancer. However, more information about the relationship between PD-L1 and TGF-β in CeCa is needed, such as, the analysis of their expression levels during CeCa progression to be considered as possible biomarkers.

4. Materials and Methods

4.1. CeCa Cell Lines

The CaSki (HPV-16+) and HeLa (HPV-18+) epithelial CeCa cell lines derived from human epidermoid carcinoma and adenocarcinoma, respectively, were obtained from the American Type Culture Collection (ATCC). These cell lines were free of mycoplasma, and their authenticity was verified by short tandem repeat (STR) genetic profiling. In previous studies, we found that these cell lines express CD73 on their surface and are capable of inhibiting the cytotoxic function of CD8+ T cells through the production of high amounts of Ado [37] and via the production of TGF-β [35].

4.2. Cell Culture

To determine the effect of Ado on the induction of PD-L1 expression in CeCa cells, 1 × 105 CeCa cells were cultured in the presence of Ado (0, 0.1 mM, or 1 mM) (Sigma Aldrich, St. Louis, MO, USA) for 24 h, 48 h, 72 h, and 96 h. The expression of PD-L1 on CeCa cell membranes at each culture time point was analyzed using an APC-labeled anti-PD-L1 monoclonal antibody (clone #130021, R&D Systems, Minneapolis, MN, USA). To determine the role of the interaction of Ado with A2Rs in the induction of PD-L1 expression, selective antagonists of A2BR and A2AR, MRS1754 (Sigma-Aldrich, St. Louis, MO, USA) and ZM241385 (Sigma-Aldrich, St. Louis, MO, USA), or the selective A2 adenosine receptor agonist CGS21680 [38] were added at a concentration of 10 μM 30 min before Ado was added to the CeCa cell cultures, as previously reported [35,36]. The cultures were maintained in Opti-MEM culture media (OCM) composed of medium (Gibco, Oakland, CA, USA) supplemented with 1% bovine fetal serum (SFB; Gibco) dialyzed with a 12-kDa cutoff membrane (Sigma-Aldrich), 100 IU/mL penicillin, and 100 μg/mL streptomycin (Gibco) at 37 °C and 5% CO2.
The levels of TGF-β1 in the supernatants of CeCa cell cultures were determined using a Quantikine ELISA kit for human TGF-β1 (R&D Systems, Inc., Minneapolis, MN, USA) according to the manufacturer’s protocol. The role of CeCa cell-derived TGF-β in the autocrine induction of PD-L1 expression was analyzed after the addition of SB505124 (Sigma-Aldrich, USA), which is a selective inhibitor of the TGF-β1 receptor, at a final concentration of 25 µM. As a positive control for the induction of PD-L1 expression, CeCa cells were cultured for 24, 48, and 72 h in the presence of 20 ng/mL recombinant human TGF-β (rHuTGF-β, PeproTech, Rocky Hill, NJ, USA). To analyze the expression of PD-L1 in CeCa cells that were subjected to different treatments, 25,000 events were acquired via a Cytek Aurora Spectral flow cytometer (Cytek Biosciences, Fremont, CA, USA) and analyzed with the FlowJo V10 program (BD Biosciences, San Jose, CA, USA) after cellular debris was removed.

4.3. Isolation of Activated CD8+ T Lymphocytes (ATLs)

CD8+ T lymphocytes were harvested from normal donor peripheral blood mononuclear cells (PBMCs) by negative selection (EasySep Enrichment Cocktail, Stem Cell Technologies, Vancouver, BC, Canada). CD8+ T lymphocytes were cultured in supplemented ISCOVE’S medium (SIM), which was composed of ISCOVE’S-modified medium (Sigma–Aldrich, USA), 4 mM L-glutamine, 1 mM sodium pyruvate, 20 μM 2-mercaptoethanol, a mixture of nonessential amino acids (Gibco BRL, USA), antibiotics (100 U/mL penicillin and 100 μg/mL streptomycin), and 10% fetal bovine serum (SFB). Subsequently, the CD8+ T lymphocytes were activated by incubation with beads coated with anti-CD2/CD3/CD28 antibodies (Miltenyi Biotec GmbH, Bergisch Gladbach, Germany) at a bead:T-lymphocyte ratio of 2:1 in the presence of 100 U/mL recombinant interleukin-2 (hrIL-2) in a total volume of 200 μL, as previously reported [39].

4.4. Inhibition of ATLs by CeCa Cells

To determine the inhibitory effect of CeCa cells on CD8+ T-lymphocyte proliferation, CeCa cells cultured in the presence or absence of 1 mM Ado for 72 h were fixed with 4% paraformaldehyde (Sigma-Aldrich, USA), washed twice with SIM, and cocultured for 96 h with ATLs at a ratio of 1:5. ATL proliferation was assessed using the CellTiter 96® Aqueous One Solution reagent (Promega, Madison, WI, USA) following the manufacturer’s instructions. The bioreduction of tetrazolium to formazan was analyzed at a wavelength of 490 nm with a GloMax plate reader (Promega, USA). The ATL proliferation index was determined every 24 h and calculated based on the optical density (OD) of the bioreduction of MTS to formazan induced by the ATLs in culture. The OD value that was obtained at the beginning of the culture was normalized to 1. In some cultures, a neutralizing anti-PD-L1 antibody (Novus, Chesterfield, MO, USA) was added according to the Novus protocol to inhibit the effect of PD-L1 on the proliferation of ATLs.

4.5. Cytotoxicity Assays

To analyze the ability of CeCa cells to inhibit the effector function of CTLs through PD-L1 expression, CTLs with specificity for the KLPQLCTEL antigenic peptide (residues 18–26) derived from the E6 protein of HPV-16 or TLHEYMLDL (residues 7–15) and YMLDLQPETT (residues 11–20) derived from the E7 protein of HPV-16, which bind specifically to the HLA-A0201 allele, expressed in CaSki cells [40], were previously generated using a previously reported method [41]. CaSki cells cultured for 72 h in the presence or absence of 1 mM Ado were fixed with 4% paraformaldehyde, washed twice with SIM, and cocultured for 48 h with CTLs at a ratio of 1:5. To block the inhibitory effect of PD-L1 on the cytotoxic activity of CTLs, in some cultures, a neutralizing anti-PD-L1 antibody (Novus, USA) was added according to the Novus protocol. CTLs cultured in the absence of CaSki cells were used as a control (Ctl). CaSki cells were marked with 5(6)-carboxyfluorescein diacetate N-succinimidyl ester (CFSE) (Sigma-Aldrich, St. Louis, MO, USA), and CTLs were labeled with a CD8-APC antibody (R&D Systems, Inc., Minneapolis, MN, USA) following the protocol provided by the supplier. The cell viability solution 7-AAD (Sigma-Aldrich, St. Louis, MO, USA) was used to measure cell lysis. Effector and target cells were incubated for 4 h at a ratio of 2:1. After the removal of cellular debris, viable cells were analyzed using a Cytek Aurora Spectral flow cytometer (Cytek Biosciences, USA), and a minimum of 100,000 events were collected. In some cases, 5% hydrogen peroxide was used for the total lysis of target cells. The percentage of lysis was calculated according to the following formula: % cytotoxicity = 100 × [experimental lysis (CFSE+, 7-AAD+) − basal lysis (CFSE+, 7AAD+)].

4.6. Assessment of PD-1, TGF-β1 and IL-10 Expression in ATLs

A total of 2.5 × 105 ATLs per well were cultured in a 24-well plate (Corning, NY, USA) for 48 h in SIM supplemented with 30% supernatant from CeCa cells (CeCa-Sup) or CeCa cells that were previously stimulated with 1 mM Ado (CeCa/Ado-Sup) for 72 h. To neutralize the effect of TGF-β in the supernatants of CeCa cells that were treated with Ado, 1 μg/mL rabbit anti-human TGF-β1, TGF-β2, and TGF-β3 neutralizing antibodies (anti-TGF-β; R&D Systems, Minneapolis, MN, USA) were added. The expression levels of PD-1, IFN-γ, TGF-β1, and IL-10 in ATLs cultured in the absence of CeCa-Sup or CeCa/Ado-Sup were considered the control (Ctl) levels. During the last 4 h of culture, brefeldin-A (Sigma-Aldrich, St. Louis, MO, USA) was added at a final concentration of 10 μM to measure the intracellular levels of IFN-γ, TGF-β1, and IL-10. Subsequently, the cells were collected, fixed, and permeabilized using a Cytofix/Cytoperm Kit (BD Biosciences, San Jose, CA, USA). T cells were labeled with anti-IFN-γ/FITC and anti-CD8/APC (R&D Systems, Inc., Minneapolis, MN, USA), anti-human IL-10/FITC (Novus Biologicals, USA), and anti-human TGF-β/PE (LifeSpan, Australia) monoclonal antibodies. The cells were incubated for 30 min at 4 °C and washed three times with PBS. To determine the expression of PD-1, ATLs that were cultured under the above-described conditions in the absence of brefeldin-A were labeled with anti-CD8/Alexa Fluor 405 (R&D, USA) and anti-PD-1/PE (Novus, USA) monoclonal antibodies according to the manufacturer’s protocol. To determine the effect of TGF-β on the expression of PD-1, IFN-γ, TGF-β1, and IL-10, ATLs were cultured for 48 h in the presence of 20 ng/mL rHuTGF-β1. To analyze the cells, 25,000 events were acquired using a Cytek Aurora Spectral flow cytometer (Cytek Biosciences, USA) after removing cellular debris, and the data were analyzed with FlowJo 10.4 software.

4.7. Statistical Analysis

All numerical data are shown as the average values ± SEMs of three independent experiments. The statistical significance was calculated using the one-way ANOVA (nonparametric) test to compare the mean rank of problem data with the mean rank of control data using the GraphPad Prism software version 8 (GraphPad Software, San Diego, CA, USA). Differences were considered significant if p < 0.05.

5. Conclusions

The present study provides evidence showing that the interaction of adenosine with A2AR/A2BR increases the immunosuppressive capacity of CeCa cells via the production of TGF-β, which acts in an autocrine manner to induce the expression of PD-L1 on CeCa cells and acts in a paracrine manner to inhibit the activation of CD8+ lymphocytes through the induction of PD-1 expression; these phenomena result in decreased production of IFN-γ and increased production of TGF-β and IL-10. The findings of this study suggest that PD-L1 and TGF-β may be clinically important biomarkers and serve as therapeutic targets.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph17030397/s1, Figure S1: Blockade of A2AR and A2BR reversed the Ado-mediated induction of PD-L1 expression in CeCa cells; Figure S2: The selective A2 adenosine receptor agonist CGS21680 induces PD-L1 expression on CeCa cells; Figure S3: Effect of Ado on the expression of TGF-βRII in CeCa cells.

Author Contributions

Conceptualization, M.d.L.M.-G. and A.M.-G.; methodology, R.G.-R., A.L.V.-C. and G.M.-C.; formal analysis, R.G.-R. and L.A.M.-A.; investigation, J.H.-M. and C.A.D.-L.; resources, M.d.L.M.-G.; data curation: R.G.-R. and L.A.M.-A.; writing—original draft preparation, M.d.L.M.-G. and A.M.-G.; writing—review and editing, A.M.-G. and M.d.L.M.-G. visualization, B.W.-S.; supervision, B.W.-S.; funding acquisition, M.d.L.M.-G. and A.M.-G. All authors have read and agreed to the published version of the manuscript.

Funding

The present study was performed with the following funds: DGAPA-PAPIIT Grant No. IN211822 to MLMG; and Grant to the protocol No R-2023-3602-011 of the Mexican Social Security Institute (Instituto Mexicano del Seguro Social (IMSS)) to A.M.-G. We also appreciate the support given to M. en CQ Luis Antonio Marín Aquino, student of the Doctorado en Ciencias en Inmunología, Instituto Politécnico Nacional, by the Doctorate Scholarship No. 703111 from CONACyT, and M en C. Gabriela Castillo Molina, student of the Programa de Posgrado en Ciencias Biológicas-UNAM, by the Doctorate Scholarship No. 776284 from CONACyT.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available upon reasonable request to the corresponding author.

Acknowledgments

The authors thank Jessica Lakshmi Prieto Chávez from the Flow Cytometry Instrument Center of IMSS for support with the flow cytometry analysis.

Conflicts of Interest

The authors report that they have no conflict of interest in this study.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Buskwofie, A.; David-West, G.; Clare, C.A. A review of cervical cancer: Incidence and disparities. J. Natl. Med. Assoc. 2020, 112, 229–232. [Google Scholar] [CrossRef]
  3. Crosbie, E.J.; Einstein, M.H.; Franceschi, S.; Kitchener, H.C. Human papillomavirus and cervical cancer. Lancet 2013, 382, 889–899. [Google Scholar] [CrossRef] [PubMed]
  4. Kobayashi, A.V.; Weinberg, T.; Darragh, K.; Smith-McCune, K. Evolving immunosuppressive microenvironment during human cervical carcinogenesis. Mucosal Immunol. 2008, 1, 412–420. [Google Scholar] [CrossRef] [PubMed]
  5. Ding, Z.C.; Lu, X.Y.; Yu, M.; Lemos, H.; Huang, L.; Chandler, P.; Liu, K.; Walters, M.; Krasinski, A.; Mack, M.; et al. Immunosuppressive myeloid cells induced by chemotherapy attenuate antitumor CD4+ T Cell Responses through the PD-1/PD-L1 Axis. Cancer Res. 2014, 74, 3441–3453. [Google Scholar] [CrossRef] [PubMed]
  6. Medina, P.J.; Adams, V.R. PD-1 Pathway Inhibitors: Immuno-oncology agents for restoring antitumor immune responses. Pharmacotherapy 2016, 36, 317–334. [Google Scholar] [CrossRef]
  7. Liu, Y.; Cao, X.T. Immunosuppressive cells in tumor immune escape and metastasis. J. Mol. Med. 2016, 94, 509–522. [Google Scholar] [CrossRef] [PubMed]
  8. Mellman, I.; Coukos, G.; Dranoff, G. Cancer immunotherapy comes of age. Nature 2011, 480, 480–489. [Google Scholar] [CrossRef] [PubMed]
  9. Gong, J.; Chehrazi-Raffle, A.; Reddi, S.; Salgia, R. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: A comprehensive review of registration trials and future considerations. J. Immunother. Cancer 2018, 6, 8. [Google Scholar] [CrossRef]
  10. Zou, W.; Wolchok, J.D.; Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci. Transl. Med. 2016, 8, 328rv4. [Google Scholar] [CrossRef]
  11. Cha, J.H.; Chan, L.C.; Li, C.W.; Hsu, J.L.; Hung, M.C. Mechanisms Controlling PD-L1 Expression in Cancer. Mol. Cell 2019, 76, 359–370. [Google Scholar] [CrossRef]
  12. Carbotti, G.; Barisione, G.; Airoldi, I.; Mezzanzanica, D.; Bagnoli, M.; Ferrero, S.; Petretto, A.; Fabbi, M.; Ferrini, S. IL-27 induces the expression of IDO and PD-L1 in human cancer cells. Oncotarget 2015, 6, 43267–43280. [Google Scholar] [CrossRef]
  13. Chen, S.; Crabill, G.A.; Pritchard, T.S.; McMiller, T.L.; Wei, P.; Pardoll, D.M.; Pan, F.; Topalian, S.L. Mechanisms regulating PD-L1 expression on tumor and immune cells. Immunother. Cancer 2019, 7, 305. [Google Scholar] [CrossRef]
  14. Lind, H.; Gameiro, S.R.; Jochems, C.; Donahue, R.N.; Strauss, J.; Gulley, J.L.; Palena, C.; Schlom, J. Dual targeting of TGF-β and PD-L1 via a bifunctional anti-PD-L1/TGF-βRII agent: Status of preclinical and clinical advances. J. Immunother. Cancer 2020, 8, e000433. [Google Scholar] [CrossRef]
  15. Mariathasan, S.; Turley, S.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel, E.E., III; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef]
  16. Sow, H.S.; Ren, J.; Camps, M.; Ossendorp, F.; Dijke, P.T. 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]
  17. Kang, J.H.; Jung, M.Y.; Choudhury, M.; Leof, E.B. Transforming growth factor beta induces fibroblasts to express and release the immunomodulatory protein PD-L1 into extracellular vesicles. FASEB J. 2020, 34, 2213–2226. [Google Scholar] [CrossRef] [PubMed]
  18. Torres-Poveda, K.; Bahena-Roman, M.; Madrid-Gonzalez, C.; Burguete-García, A.I.; Bermúdez-Morales, V.H.; Peralta-Zaragoza, O.; Madrid-Marina, V. Role of IL-10 and TGF-beta1 in local immunosuppression in HPV-associated cervical neoplasia. World J. Clin. Oncol. 2014, 5, 753–763. [Google Scholar] [CrossRef]
  19. Mezache, L.; Paniccia, B.; Nyinawabera, A.; Nuovo, G.J. Enhanced expression of PD L1 in cervical intraepithelial neoplasia and cervical cancers. Modern Pathol. 2015, 28, 1594–1602. [Google Scholar] [CrossRef] [PubMed]
  20. Meng, Y.; Liang, H.; Hu, J.; Liu, S.; Hao, X.; Wong, M.S.K.; Li, X.; Hu, L. PD-L1 expression correlates with tumor infiltrating lymphocytes and response to neoadjuvant chemotherapy in cervical cancer. J. Cancer 2018, 9, 2938–2945. [Google Scholar] [CrossRef] [PubMed]
  21. Heeren, A.M.; Punt, S.; Bleeker, M.C.G.; Gaarenstroom, K.N.; van der Velden, J.; Kenter, G.G.; de Gruijl, T.D.; Jordanova, E.S. Prognostic effect of different PD-L1 expression patterns in squamous cell carcinoma and adenocarcinoma of the cervix. Mod. Pathol. 2016, 29, 753–763. [Google Scholar] [CrossRef]
  22. Feng, Y.C.; Ji, W.L.; Yue, N.; Huang, Y.C.; Ma, X.M. The relationship between the PD-1/PD-L1 pathway and DNA mismatch repair in cervical cancer and its clinical significance. Cancer Manag. Res. 2018, 10, 105–113. [Google Scholar] [CrossRef]
  23. Huang, W.; Liu, J.; Xu, K.; Chen, H.; Bian, C. PD-1/PD-L1 inhibitors for advanced or metastatic cervical cancer: From bench to bed. Front. Oncol. 2022, 12, 849352. [Google Scholar] [CrossRef]
  24. Saglam, O.; Zhou, J.; Wang, X.; Conejo-Garcia, J.R. PD-L1 expression correlates with young age and CD8+ TIL density in poorly differentiated cervical squamous cell carcinoma. Int. J. Gynecol. Pathol. 2020, 39, 428–435. [Google Scholar] [CrossRef]
  25. Yang, W.; Song, Y.; Lu, Y.L.; Sun, J.Z.; Wang, H.W. Increased expression of programmed death (PD)-1 and its ligand PD-L1 correlates with impaired cell-mediated immunity in high-risk human papillomavirus-related cervical intraepithelial neoplasia. Immunology 2013, 139, 513–522. [Google Scholar] [CrossRef]
  26. Bahreyni, A.; Samani, S.S.; Ghorbani, E.; Rahmani, F.; Khayami, R.; Toroghian, Y.; Behnam-Rassouli, R.; Khazaei, M.; Ryzhikov, M.; Parizadeh, M.R.; et al. Adenosine: An endogenous mediator in the pathogenesis of gynecological cancer. J. Cell Physiol. 2018, 233, 2715–2722. [Google Scholar] [CrossRef] [PubMed]
  27. Pfaffenzeller, M.S.; Franciosi, M.L.M.; Cardoso, A.M. Purinergic signaling and tumor microenvironment in cervical cancer. Purinergic Signal. 2020, 16, 123–135. [Google Scholar] [CrossRef] [PubMed]
  28. Vaupel, P.; Mayer, A. Hypoxia-driven adenosine accumulation: A crucial microenvironmental factor promoting tumor progression. Adv. Exp. Med. Biol. 2016, 876, 177–183. [Google Scholar] [PubMed]
  29. Robson, S.C.; Sévigny, J.; Zimmermann, H. The ENTPDase family of ectonucleotidases: Structure function relationships and pathophysiological significance. Purinergic Signal. 2006, 2, 409–430. [Google Scholar] [CrossRef] [PubMed]
  30. Zimmermann, H.; Zebisch, M.; Sträer, N. Cellular function and molecular structure of ecto-nucleotidases. Purinergic Signal. 2012, 8, 437–502. [Google Scholar] [CrossRef] [PubMed]
  31. Linden, J. Molecular approach to adenosine receptors: Receptor-mediated mechanisms of tissue protection. Annu. Rev. Pharmacol. Toxicol. 2001, 41, 775–787. [Google Scholar] [CrossRef] [PubMed]
  32. Antonioli, L.; Fornai, M.; Blandizzi, C.; Pacher, P.; Haskó, G. Adenosine signaling and the immune system: When a lot could be too much. Immunol. Lett. 2019, 205, 9–15. [Google Scholar] [CrossRef] [PubMed]
  33. Vijayan, D.; Young, A.; Teng, M.; Smyth, M. Targeting immunosuppressive adenosine in cancer. Nat. Rev. Cancer 2017, 17, 709–724. [Google Scholar] [CrossRef] [PubMed]
  34. Leone, R.D.; Emens, L.A. Targeting adenosine for cancer immunotherapy. J. Immunother. Cancer 2018, 6, 57. [Google Scholar] [CrossRef]
  35. García-Rocha, R.; Monroy-García, A.; Hernandez-Montes, J.; Benny Weiss-Steider, B.; Gutiérrez-Serrano, V.; Fuentes-Castañeda, M.C.; Ávila-Ibarra, L.-R.; Don-López, C.A.; Torres-Pineda, D.B.; Mora-García, M.L. Cervical cancer cells produce TGF-beta1 through the CD73-adenosine pathway and maintain CD73 expression through the autocrine activity of TGF-beta1. Cytokine 2019, 118, 71–79. [Google Scholar] [CrossRef]
  36. García-Rocha, R.; Monroy-García, A.; Carrera-Martínez, M.; Hernández-Montes, J.; Don-López, C.A.; Weiss-Steider, B.; Monroy-Mora, K.A.; Ponce-Chavero, M.L.A.; Montesinos-Montesinos, J.J.; Escobar-Sánchez, M.L.; et al. Evidence that cervical cancer cells cultured as tumorspheres maintain high CD73 expression and increase their protumor characteristics through TGF-β production. Cell Biochem. Funct. 2022, 40, 760–772. [Google Scholar] [CrossRef] [PubMed]
  37. Mora-García, M.L.; Ávila-Ibarra, L.R.; García-Rocha, R.; Weiss-Steider, B.; Hernández-Montes, J.; Don-López, C.A.; Gutiérrez-Serrano, V.; Titla-Vilchis, I.J.; Fuentes-Castañeda, M.C.; Monroy-Mora, A.; et al. Cervical cancer cells suppress effector functions of cytotoxic T cells through the adenosinergic pathway. Cell Immunol. 2017, 320, 46–55. [Google Scholar] [CrossRef]
  38. Mora-García, M.L.; García-Rocha, R.; Morales-Ramirez, O.; Montesinos, J.J.; Weiss-Steider, B.; Hernández-Montes, J.; Ávila-Ibarra, L.A.; Don-López, C.A.; Velasco-Velázquez, M.; Gutiérrez-Srrano, V.; et al. Mesenchymal stromal cells derived from cervical cancer produce high amounts of adenosine to suppress cytotoxic T lymphocyte functions. J. Transl. Med. 2016, 14, 302. [Google Scholar] [CrossRef]
  39. Evans, C.; Bauer, S.; Grubert, T.; Brucker, C.; Baur, S.; Heeg, K.; Wagner, H.; Lipford, G.B. HLA-A2-restricted peripheral blood cytolytic T lymphocyte response to HPV type 16 proteins E6 and E7 from patients with neoplastic cervical lesions. Cancer Immunol. Immunother. 1996, 42, 151–160. [Google Scholar] [CrossRef]
  40. García-Rocha, R.; Moreno-Lafont, M.C.; Mora-García, M.L.; Weiss-Steider, B.; Montesinos, J.J.; Piña-Sánchez, P.; Monroy-García, A. Mesenchymal stromal cells derived from cervical cancer tumors induce TGF-β1 expression and IL-10 expression and secretion in the cervical cancer cells, resulting in protection from cytotoxic T cell activity. Cytokine 2015, 76, 382–390. [Google Scholar] [CrossRef]
  41. Rekik, R.; Hmida, N.B.; Hmid, A.B.; Zamali, I.; Kammoun, N.; Ahmed, M.B. PD-1 induction through TCR activation is partially regulated by endogenous TGF-β. Cell Mol. Immunol. 2015, 12, 648–649. [Google Scholar] [CrossRef]
  42. Zhang, L.; Zhao, Y.; Tu, Q.; Xue, X.; Zhu, X.; Zhao, K.N. The Roles of Programmed Cell Death Ligand-1/Programmed Cell Death-1 (PD-L1/PD-1) in HPV-induced Cervical Cancer and Potential for their Use in Blockade Therapy. Curr. Med. Chem. 2021, 28, 893–909. [Google Scholar] [CrossRef]
  43. Reddy, O.L.; Shintaku, P.I.; Moatamed, N.A. Programmed death-ligand 1 (PD-L1) is expressed in a significant number of the uterine cervical carcinomas. Diagn. Pathol. 2017, 12, 45. [Google Scholar] [CrossRef]
  44. Enwere, E.K.; Kornaga, E.N.; Dean, M.; Koulis, T.A.; Phan, T.; Kalantarian, M.; Köbel, M.; Ghatage, P.; Magliocco, A.M.; Lees-Miller, S.P.; et al. Expression of PD-L1 and presence of CD8-positive T cells in pre-treatment specimens of locally advanced cervical cancer. Mod. Pathol. 2017, 30, 577–586. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, X.; Si, F.; Bagley, D.; Ma, F.; Zhang, Y.; Tao, Y.; Shaw, E.; Peng, G. Blockades of effector T cell senescence and exhaustion synergistically enhance antitumor immunity and immunotherapy. Immunother. Cancer 2022, 10, e005020. [Google Scholar] [CrossRef]
  46. Chow, A.; Perica, K.; Klebanoff, C.A.; Wolchok, J.D. Clinical implications of T cell exhaustion for cancer immunotherapy. Nat. Rev. Clin. Oncol. 2022, 19, 775–790. [Google Scholar] [CrossRef] [PubMed]
  47. Cottrell, T.R.; Taube, J.M. PD-L1 and emerging biomarkers in immune checkpoint blockade therapy. Cancer J. 2018, 24, 416. [Google Scholar] [CrossRef]
  48. Wang, S.; Xie, K.; Liu, T. Cancer Immunotherapies: From Efficacy to Resistance Mechanisms –Not Only Checkpoint Matters. Front. Immunol. 2021, 12, 690112. [Google Scholar] [CrossRef] [PubMed]
  49. 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]
  50. Green, M.R.; Rodig, S.; Juszczynski, P.; Ouyang, J.; Sinha, P.; O’Donneli, E.; Neuberg, D.; Shipp, M.A. Constitutive AP-1 activity and EBV infection induce PD-L1 in Hodgkin lymphomas and posttransplant lymphoproliferative disorders: Implications for targeted therapy. Clin. Cancer Res. 2012, 18, 1611–1618. [Google Scholar] [CrossRef]
  51. Chen, L.; Gibbons, D.L.; Goswami, S.; Cortez, M.A.; Ahn, Y.H.; Byers, L.A.; Zhang, X.; Yi, X.; Dwyer, D.; Lin, W.; et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat. Commun. 2014, 5, 5241. [Google Scholar] [CrossRef]
  52. Jiang, X.; Zhou, J.; Giobbie-Hurder, A.; Wargo, J.; Hodi, F.S. The activation of MAPK in melanoma cells resistant to BRAF inhibition promotes PD-L1 expression that is reversible by MEK and PI3K inhibition. Clin. Cancer Res. 2013, 19, 598–609. [Google Scholar] [CrossRef]
  53. Crane, C.; Panner, A.; Pieper, R.O.; Arbiser, J.; Parsa, A.T. Honokiol-mediated inhibition of PI3K/mTOR pathway: A potential strategy to overcome immunoresistance in glioma, breast, and prostate carcinoma without impacting T cell function. J. Immunother. 2009, 32, 585–592. [Google Scholar] [CrossRef]
  54. Bellucci, R.; Martin, A.; Bommarito, D.; Wang, K.; Hansen, S.H.; Freeman, G.J.; Ritz, J. Interferon-gamma-induced activation of JAK1 and JAK2 suppresses tumor cell susceptibility to NK cells through upregulation of PD-L1 expression. Oncoimmunology 2015, 4, e1008824. [Google Scholar] [CrossRef] [PubMed]
  55. Hirahara, K.; Ghoreschi, K.; Yang, X.P.; Takahashi, H.; Laurence, A.; Vahedi, G.; Sciumé, G.; Hall, A.O.; Dupont, C.D.; Francisco, L.M.; et al. Interleukin-27 priming of T cells controls IL-17 production in trans via induction of the ligand PD-L1. Immunity 2012, 36, 1017–1030. [Google Scholar] [CrossRef]
  56. Hirayama, A.; Tanaka, K.; Tsutsumi, H.; Nakanishi, T.; Yamashita, S.; Mizusaki, S.; Ishii, Y.; Ota, K.; Yoneshima, Y.; Iwama, E.; et al. Regulation of PD-L1 expression in non-small cell lung cancer by interleukin-1β. Front. Immunol. 2023, 14, 1192861. [Google Scholar] [CrossRef] [PubMed]
  57. Chatterjee, S.; Chatterjee, A.; Jana, S.; Dey, S.; Roy, H.; Das, M.K.; Alam, J.; Adhikary, A.; Chowdhury, A.; Biswas, A.; et al. Transforming growth factor beta orchestrates PD-L1 enrichment in tumor-derived exosomes and mediates CD8 T-cell dysfunction regulating early phosphorylation of TCR signalome in breast cancer. Carcinogenesis 2021, 42, 38–47. [Google Scholar] [CrossRef] [PubMed]
  58. Kazemi, M.H.; Mohseni, S.R.; Hojjat-Farsangi, M.; Anvari, E.; Ghalamfarsa, G.; Mohammadi, H.; Jadidi-Niaragh, F. Adenosine and adenosine receptors in the immunopathogenesis and treatment of cancer. J. Cell Physiol. 2018, 233, 2032–2057. [Google Scholar] [CrossRef]
  59. Rotman, J.; den Otter, L.A.S.; Bleeker, M.C.G.; Samuels, S.S.; Heeren, A.M.; Roemer, M.G.M.; Kenter, G.G.; Zijlmans, H.J.M.A.A.; van Trommel, N.E.; de Gruijl, T.D.; et al. PD-L1 and PD-L2 Expression in Cervical Cancer: Regulation and Biomarker Potential. Front. Immunol. 2020, 11, 596825. [Google Scholar] [CrossRef] [PubMed]
  60. 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] [PubMed]
  61. Lin, H.; Huang, C.C.; Ou, Y.C.; Huang, E.Y.; Changchien, C.C.; Tseng, C.W.; Fu, H.C.; Li, C.J.; Ma, Y.Y. High immunohistochemical expression of TGF-beta1 predicts a poor prognosis incervical cancer patients who harbor enriched endoglin microvessel density. Int. J. Gynecol. Pathol. 2012, 31, 482–489. [Google Scholar] [CrossRef] [PubMed]
  62. Akhurst, R.J.; Derynck, R. TGF-beta signaling in cancer a double-edged sword. Trends Cell Biol. 2001, 11, S44–S51. [Google Scholar] [PubMed]
  63. Yetişir, A.E.; Paydaş, S.; Gönlüşen, G.; Erdoğan, K.E.; Büyükşimşek, M.; Oğul, A.; Tohumcuoğlu, M.; Mirili, C.; Yetişir, A. Sarcoma-correlation between CD73 and PD-L1 and their relationship with prognosis. Pol. J. Pathol. 2022, 73, 338–342. [Google Scholar] [CrossRef] [PubMed]
  64. Tahkola, K.; Ahtiainen, M.; Kellokumpu, I.; Mecklin, J.P.; Laukkarinen, J.; Laakkonen, J.; Kenessey, I.; Jalkanen, S.; Salmi, M.; Böhm, J. Prognostic impact of CD73 expression and its relationship to PD-L1 in patients with radically treated pancreatic cancer. Virchows Arch. 2021, 478, 209–217. [Google Scholar] [CrossRef] [PubMed]
  65. Tu, E.; McGlinchey, K.; Wang, J.; Martin, P.; Ching, S.L.; Floc’h, N.; Kurasawa, J.; Starrett, J.H.; Lazdun, Y.; Wetzel, L.; et al. Anti-PD-L1 and anti-CD73 combination therapy promotes T cell response to EGFR-mutated NSCLC. JCI Insight 2022, 7, e142843. [Google Scholar] [CrossRef] [PubMed]
  66. Ye, J.; Gavras, N.W.; Keeley, D.C.; Hughson, A.L.; Hannon, G.; Vrooman, T.G.; Lesch, M.L.; Johnston, C.J.; Lord, E.M.; Belt, B.A.; et al. CD73 and PD-L1 dual blockade amplifies antitumor efficacy of SBRT in murine PDAC models. J. Immunother. Cancer 2023, 11, e006842. [Google Scholar] [CrossRef] [PubMed]
  67. Redman, J.M.; Friedman, J.; Robbins, Y.; Sievers, C.; Yang, X.; Lassoued, W.; Sinkoe, A.; Papanicolau-Sengos, A.; Lee, C.C.; Marte, J.L.; et al. Enhanced neoepitope-specific immunity following neoadjuvant PD-L1 and TGF-β blockade in HPV-unrelated head and neck cancer. J. Clin. Investig. 2022, 132, e161400. [Google Scholar] [CrossRef]
  68. Lan, Y.; Yeung, T.L.; Huang, H.; Wegener, A.A.; Saha, S.; Toister-Achituv, M.; Jenkins, M.H.; Chiu, L.Y.; Lazorrchack, A.; Tarcic, O.; et al. Colocalized targeting of TGF-β and PD-L1 by bintrafusp alfa elicits distinct antitumor responses. J. Immunother. Cancer 2022, 10, e004122. [Google Scholar] [CrossRef]
  69. Birrer, M.J.; Fujiwara, K.; Oaknin, A.; Randall, l.; Ojalvo, L.S.; Valencia, C.; Ray-Coquard, I. The Changing Landscape of Systemic Treatment for Cervical Cancer: Rationale for Inhibition of the TGF-β and PD-L1 Pathways. Front. Oncol. 2022, 12, 814169. [Google Scholar] [CrossRef]
Figure 1. Effect of Ado on the expression of PD-L1 in CeCa cells. CaSki (a) and HeLa (b) cells (1 × 105 of each) were cultured in the presence of 0.1 mM or 1 mM Ado for 96 h. The baseline expression level of PD-L1 in CeCa cells was considered the control (Ctl) level. PD-L1 expression was analyzed via flow cytometry every 24 h. Representative data from three independent experiments are shown. MFI, mean fluorescence intensity.
Figure 1. Effect of Ado on the expression of PD-L1 in CeCa cells. CaSki (a) and HeLa (b) cells (1 × 105 of each) were cultured in the presence of 0.1 mM or 1 mM Ado for 96 h. The baseline expression level of PD-L1 in CeCa cells was considered the control (Ctl) level. PD-L1 expression was analyzed via flow cytometry every 24 h. Representative data from three independent experiments are shown. MFI, mean fluorescence intensity.
Pharmaceuticals 17 00397 g001
Figure 2. Ado increases PD-L1 expression on CeCa cells by interacting with A2AR and A2BR to induce the production of TGF-β1. (a) CaSki and HeLa cells (1 × 105 each) were first incubated for 30 min in the presence of 10 µM ZM241385 and MRS1754, which are selective antagonists for A2AR and A2BR, respectively. The cells were then cultured for 72 h in the presence or absence of 1 mM Ado. Cells that were cultured in the absence of both Ado and AR antagonists were used as controls (black bars). (b) The levels of TGF-β in the supernatants of CeCa cells cultured with 1 mM Ado for 24 h, 48 h and 72 h were determined via ELISA. The TGF-β levels in the supernatants of CeCa cells that were cultured in the absence of Ado were considered control levels (black bars). (c) CeCa cells were cultured in the presence of 20 ng of rHuTGF-β1. PDL-1 expression in CeCa cells was determined at 24 h, 48 h, and 72 h. CeCa cells that were cultured in the absence of rHuTGF-β1 were used as a control (black bars). (d) CeCa cells were cultured for 72 h in the presence or absence of 1 mM Ado and in the presence or absence (black bars) of 25 μM SB-505124, which is a selective TGF-β1 receptor inhibitor. The statistical significance was calculated using the one-way ANOVA (nonparametric) test. Representative data from three independent experiments (means ± SEMs) are shown. * p < 0.05; ** p < 0.001. +, presence. -, absence.
Figure 2. Ado increases PD-L1 expression on CeCa cells by interacting with A2AR and A2BR to induce the production of TGF-β1. (a) CaSki and HeLa cells (1 × 105 each) were first incubated for 30 min in the presence of 10 µM ZM241385 and MRS1754, which are selective antagonists for A2AR and A2BR, respectively. The cells were then cultured for 72 h in the presence or absence of 1 mM Ado. Cells that were cultured in the absence of both Ado and AR antagonists were used as controls (black bars). (b) The levels of TGF-β in the supernatants of CeCa cells cultured with 1 mM Ado for 24 h, 48 h and 72 h were determined via ELISA. The TGF-β levels in the supernatants of CeCa cells that were cultured in the absence of Ado were considered control levels (black bars). (c) CeCa cells were cultured in the presence of 20 ng of rHuTGF-β1. PDL-1 expression in CeCa cells was determined at 24 h, 48 h, and 72 h. CeCa cells that were cultured in the absence of rHuTGF-β1 were used as a control (black bars). (d) CeCa cells were cultured for 72 h in the presence or absence of 1 mM Ado and in the presence or absence (black bars) of 25 μM SB-505124, which is a selective TGF-β1 receptor inhibitor. The statistical significance was calculated using the one-way ANOVA (nonparametric) test. Representative data from three independent experiments (means ± SEMs) are shown. * p < 0.05; ** p < 0.001. +, presence. -, absence.
Pharmaceuticals 17 00397 g002
Figure 3. Ado-stimulated CeCa cells inhibit the proliferation of activated CD8+ T cells by expressing PD-L1. The proliferation indices (PIs) of activated CD8+ T lymphocytes (ATLs) that were cultured for 24, 48, 72, and 96 h in the presence (red lines) or absence (black lines) of CaSki (a) and HeLa (b) cells that were first cultured in either the presence (blue lines) or the absence (red lines) of 1 mM Ado are shown. In some cultures, a neutralizing anti-PD-L1 antibody was added to inhibit the effect of PD-L1 on the proliferation of ATLs cultured with CeCa cells that were previously incubated in the presence (green lines) of 1 mM Ado. The PIs of the ATLs at the beginning of the culture were set to 1. The statistical significance was calculated using the one-way ANOVA (nonparametric) test. Representative data from three independent experiments (means ± SEMs) are shown. * p < 0.05, ** p < 0.01, *** p < 0.001.
Figure 3. Ado-stimulated CeCa cells inhibit the proliferation of activated CD8+ T cells by expressing PD-L1. The proliferation indices (PIs) of activated CD8+ T lymphocytes (ATLs) that were cultured for 24, 48, 72, and 96 h in the presence (red lines) or absence (black lines) of CaSki (a) and HeLa (b) cells that were first cultured in either the presence (blue lines) or the absence (red lines) of 1 mM Ado are shown. In some cultures, a neutralizing anti-PD-L1 antibody was added to inhibit the effect of PD-L1 on the proliferation of ATLs cultured with CeCa cells that were previously incubated in the presence (green lines) of 1 mM Ado. The PIs of the ATLs at the beginning of the culture were set to 1. The statistical significance was calculated using the one-way ANOVA (nonparametric) test. Representative data from three independent experiments (means ± SEMs) are shown. * p < 0.05, ** p < 0.01, *** p < 0.001.
Pharmaceuticals 17 00397 g003
Figure 4. Ado-stimulated CeCa cells inhibit the effect of cytotoxic T lymphocytes by expressing PD-L1. CaSki cells or CaSki-Ado cells were fixed with paraformaldehyde and cocultured for 48 h at a ratio of 1:5 with CTLs (CD8+ T lymphocytes) specific for the KLPQLCTEL antigenic peptide (residues 18–26) derived from the E6 protein of HPV-16 or TLHEYMLDL (residues 7–15) and YMLDLQPETT (residues 11–20) derived from the E7 protein of HPV-16. To block the inhibitory effect of PD-L1 on the cytotoxic activity of CTLs, in some cultures, a neutralizing anti-PD-L1 antibody was added. CTLs cultured in the absence of CaSki cells were used as a control Ctl (Black bar). CTL activity was evaluated by measuring the viability of cells stained with CFSE and 7-AAD, as indicated in the Methods section. The statistical significance was calculated using the one-way ANOVA (nonparametric) test. The data represent three independent experiments; the means ± SEMs are shown. * p < 0.05; ** p < 0.001. +, presence. -, absence.
Figure 4. Ado-stimulated CeCa cells inhibit the effect of cytotoxic T lymphocytes by expressing PD-L1. CaSki cells or CaSki-Ado cells were fixed with paraformaldehyde and cocultured for 48 h at a ratio of 1:5 with CTLs (CD8+ T lymphocytes) specific for the KLPQLCTEL antigenic peptide (residues 18–26) derived from the E6 protein of HPV-16 or TLHEYMLDL (residues 7–15) and YMLDLQPETT (residues 11–20) derived from the E7 protein of HPV-16. To block the inhibitory effect of PD-L1 on the cytotoxic activity of CTLs, in some cultures, a neutralizing anti-PD-L1 antibody was added. CTLs cultured in the absence of CaSki cells were used as a control Ctl (Black bar). CTL activity was evaluated by measuring the viability of cells stained with CFSE and 7-AAD, as indicated in the Methods section. The statistical significance was calculated using the one-way ANOVA (nonparametric) test. The data represent three independent experiments; the means ± SEMs are shown. * p < 0.05; ** p < 0.001. +, presence. -, absence.
Pharmaceuticals 17 00397 g004
Figure 5. Effect of CeCa-Ado supernatants on the activation of CD8+ T cells. CD8+ T cells (2.5 × 105) were cultured for 48 h in the presence of beads coated with anti-CD2/CD3/CD28 antibodies at a ratio of 2:1 with or without 30% supernatant (Sup) from CeCa cells or CeCa-Ado cells and with or without 1 μg/mL anti-TGF-β1 neutralizing antibody. The expression of PD-1 (a), IFN-γ (b), TGF-β (c), and IL-10 (d) in ATLs was analyzed as described in the Materials and Methods section. ATLs that were cultured without CeCa or CeCa-Ado Sup were considered controls (Ctls). Representative data from three independent experiments are shown. MFI, mean fluorescence intensity.
Figure 5. Effect of CeCa-Ado supernatants on the activation of CD8+ T cells. CD8+ T cells (2.5 × 105) were cultured for 48 h in the presence of beads coated with anti-CD2/CD3/CD28 antibodies at a ratio of 2:1 with or without 30% supernatant (Sup) from CeCa cells or CeCa-Ado cells and with or without 1 μg/mL anti-TGF-β1 neutralizing antibody. The expression of PD-1 (a), IFN-γ (b), TGF-β (c), and IL-10 (d) in ATLs was analyzed as described in the Materials and Methods section. ATLs that were cultured without CeCa or CeCa-Ado Sup were considered controls (Ctls). Representative data from three independent experiments are shown. MFI, mean fluorescence intensity.
Pharmaceuticals 17 00397 g005
Figure 6. Effect of TGF-β on the activation of CD8+ T cells. CD8+ T cells (2.5 × 105) were cultured for 48 h in the presence of beads coated with anti-CD2/CD3/CD28 antibodies at a ratio of 2:1 in the presence or absence of 20 ng/mL rHuTGF-β. The expression of PD-1 (a), IFN-γ (b), TGF-β (c) and IL-10 (d) in ATLs was analyzed as described in the Materials and Methods section. The expression levels of PD-1, IFN-γ, TGF-β, and IL-10 in ATLs that were cultured without rHuTGF-β were considered control levels (black bars) and were set to 1. Representative data from three independent experiments (means ± SEMs) are shown. The statistical significance was calculated using the one-way ANOVA (nonparametric) test. * p < 0.05 compared to the control.
Figure 6. Effect of TGF-β on the activation of CD8+ T cells. CD8+ T cells (2.5 × 105) were cultured for 48 h in the presence of beads coated with anti-CD2/CD3/CD28 antibodies at a ratio of 2:1 in the presence or absence of 20 ng/mL rHuTGF-β. The expression of PD-1 (a), IFN-γ (b), TGF-β (c) and IL-10 (d) in ATLs was analyzed as described in the Materials and Methods section. The expression levels of PD-1, IFN-γ, TGF-β, and IL-10 in ATLs that were cultured without rHuTGF-β were considered control levels (black bars) and were set to 1. Representative data from three independent experiments (means ± SEMs) are shown. The statistical significance was calculated using the one-way ANOVA (nonparametric) test. * p < 0.05 compared to the control.
Pharmaceuticals 17 00397 g006
Figure 7. Proposed mechanism by which Ado increases the immunosuppressive effects of CeCa cells by increasing PD-L1 expression and TGF-β production. The interaction of Ado with A2AR and A2BR (1) induces the production of TGF-β1 by CeCa cells (2). This cytokine acts in an autocrine manner, increasing the expression of PD-L1 on the membrane (3); as a result, the immunosuppressive effect of CeCa cells on activated CD8+ T lymphocytes (ATLs) is increased. Furthermore, TGF-β inhibits ATLs by decreasing the production of IFN-γ (4) and increasing the expression of PD-1 and the production of TGF-β and IL-10 (5).
Figure 7. Proposed mechanism by which Ado increases the immunosuppressive effects of CeCa cells by increasing PD-L1 expression and TGF-β production. The interaction of Ado with A2AR and A2BR (1) induces the production of TGF-β1 by CeCa cells (2). This cytokine acts in an autocrine manner, increasing the expression of PD-L1 on the membrane (3); as a result, the immunosuppressive effect of CeCa cells on activated CD8+ T lymphocytes (ATLs) is increased. Furthermore, TGF-β inhibits ATLs by decreasing the production of IFN-γ (4) and increasing the expression of PD-1 and the production of TGF-β and IL-10 (5).
Pharmaceuticals 17 00397 g007
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

García-Rocha, R.; Monroy-García, A.; Vázquez-Cruz, A.L.; Marín-Aquino, L.A.; Weiss-Steider, B.; Hernández-Montes, J.; Don-López, C.A.; Molina-Castillo, G.; Mora-García, M.d.L. Adenosine Increases the Immunosuppressive Capacity of Cervical Cancer Cells by Increasing PD-L1 Expression and TGF-β Production through Its Interaction with A2AR/A2BR. Pharmaceuticals 2024, 17, 397. https://doi.org/10.3390/ph17030397

AMA Style

García-Rocha R, Monroy-García A, Vázquez-Cruz AL, Marín-Aquino LA, Weiss-Steider B, Hernández-Montes J, Don-López CA, Molina-Castillo G, Mora-García MdL. Adenosine Increases the Immunosuppressive Capacity of Cervical Cancer Cells by Increasing PD-L1 Expression and TGF-β Production through Its Interaction with A2AR/A2BR. Pharmaceuticals. 2024; 17(3):397. https://doi.org/10.3390/ph17030397

Chicago/Turabian Style

García-Rocha, Rosario, Alberto Monroy-García, Ana Luisa Vázquez-Cruz, Luis Antonio Marín-Aquino, Benny Weiss-Steider, Jorge Hernández-Montes, Christian Azucena Don-López, Gabriela Molina-Castillo, and María de Lourdes Mora-García. 2024. "Adenosine Increases the Immunosuppressive Capacity of Cervical Cancer Cells by Increasing PD-L1 Expression and TGF-β Production through Its Interaction with A2AR/A2BR" Pharmaceuticals 17, no. 3: 397. https://doi.org/10.3390/ph17030397

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