Minireviews Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Exp Med. Sep 20, 2024; 14(3): 96269
Published online Sep 20, 2024. doi: 10.5493/wjem.v14.i3.96269
Aryl hydrocarbon receptor dynamics in esophageal squamous cell carcinoma: From immune modulation to therapeutic opportunities
Mina Rahmati, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Tehran, Iran
Hassan Moghtaderi, Saeed Mohammadi, Ahmed Al-Harrasi, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Ad Dakhiliyah, Oman
ORCID number: Mina Rahmati (0000-0002-1151-2437); Hassan Moghtaderi (0000-0002-1804-8251); Saeed Mohammadi (0000-0001-9895-8468); Ahmed Al-Harrasi (0000-0003-0317-8448).
Co-first authors: Mina Rahmati and Hassan Moghtaderi.
Co-corresponding authors: Saeed Mohammadi and Ahmed Al-Harrasi.
Author contributions: Rahmati M and Moghtaderi H reviewed the literature, prepared information, and drafted the manuscript; Mohammadi S and Al-Harrasi A conceptualized the review, proposed the title, and provided critical reviews; All authors have read and approved the final manuscript. Rahmati M and Moghtaderi H reviewed relevant literature, gathered essential information, and drafted the initial version of the manuscript. Rahmati M organized the data and ensured the integration of comprehensive and relevant studies into the review. Moghtaderi H synthesized the gathered information and contributed significantly to the initial manuscript draft. Mohammadi S and Al-Harrasi A conceptualized the overall scope of the review article and proposed the title. Mohammadi S offered essential perspectives that maintained the manuscript's logical flow and improved its scientific precision during the revision stages. Al-Harrasi A conducted extensive reviews and contributed to revising the content to meet high academic standards. Both Rahmati M and Moghtaderi H made crucial contributions towards the completion of the review, qualifying as co-first authors. Both Mohammadi S and Al-Harrasi A played important and essential roles in the conceptual design, critical review, and manuscript preparation as co-corresponding authors. Mohammadi S supervised the entire review process, provided continuous oversight, and ensured the manuscript's coherence. Al-Harrasi A contributed significantly to the critical review, enhancing the manuscript's quality and accuracy. This collaboration between Mohammadi S and Al-Harrasi A was essential for the successful completion and publication of this manuscript. Their combined efforts in conceptual design, critical review, and supervision were crucial for maintaining the manuscript's high standards and academic integrity.
Conflict-of-interest statement: All the authors declare that there are no conflicts of interest to disclose.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ahmed Al-Harrasi, PhD, Full Professor, Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al Mauz, PO Box 33, Postal Code 616, Nizwa, Oman. aharrasi@unizwa.edu.om
Received: May 1, 2024
Revised: May 26, 2024
Accepted: June 14, 2024
Published online: September 20, 2024
Processing time: 119 Days and 18.3 Hours

Abstract

Esophageal squamous cell carcinoma (ESCC) is a substantial global health burden. Immune escape mechanisms are important in ESCC progression, enabling cancer cells to escape the surveillance of the host immune system. One key player in this process is the Aryl Hydrocarbon Receptor (AhR), which influences multiple cellular processes, including proliferation, differentiation, metabolism, and immune regulation. Dysregulated AhR signaling participates in ESCC development by stimulating carcinogenesis, epithelial-mesenchymal transition, and immune escape. Targeting AhR signaling is a potential therapeutic approach for ESCC, with AhR ligands showing efficacy in preclinical studies. Additionally, modification of AhR ligands and combination therapies present new opportunities for therapeutic intervention. This review aims to address the knowledge gap related to the role of AhR signaling in ESCC pathogenesis and immune escape.

Key Words: Esophageal squamous cell carcinoma; Aryl hydrocarbon receptor; Immune escape; Tumor microenvironment; Immunosuppression; Therapeutic targeting

Core Tip: Esophageal squamous cell carcinoma (ESCC) utilizes immune escape mechanisms, including major histocompatibility complex downregulation and immune checkpoint manipulation, enhancing tumor progression. The aryl hydrocarbon receptor (AhR), crucial in health and disease, significantly influences ESCC development and immune evasion through carcinogenic pathways. AhR activation triggers proliferation, inhibits apoptosis, and induces epithelial-mesenchymal transition. Moreover, AhR suppresses effector T cells and enrolls immunosuppressive myeloid cells. Therapeutic AhR targeting with AhR ligands showed promise in inhibiting tumor growth, controlling the immune microenvironment, and enhancing treatment efficacy. Combining AhR-targeted therapies with conventional treatments or dietary interventions has the potential to improve ESCC patient outcomes.



INTRODUCTION

Esophageal squamous cell carcinoma (ESCC), a severe malignancy of the esophagus, shows a significant global health burden (ESCC is the eighth most common cancer globally)[1]. It shows a notable geographical variation, with particularly high incidence rates in East Asia, Central Asia, and certain regions of Africa[2].

ESCC emerges through a multistep process involving progressive genetic and molecular alterations within the esophageal epithelium[3]. Chronic exposure to established carcinogens, such as tobacco smoke and excessive alcohol consumption, triggers cellular damage and initiates a cascade of events leading to uncontrolled proliferation and malignant transformation[4]. These factors can lead to point mutations in tumor suppressor genes such as p53, which regulate cell cycle progression and DNA repair[5]. Moreover, amplification of oncogenes, such as cyclin D1, promote cell cycle dysregulation and uncontrolled growth[6]. Furthermore, epigenetic modifications, such as DNA methylation and histone acetylation configurations, can inhibit tumor suppressor genes and provide a pro-tumorigenic microenvironment[7]. These genetic and molecular aberrations interfere with normal cellular processes, leading to the development of pre-cancerous lesions characterized by squamous epithelial hyperplasia and dysplasia. If left unchecked, these pre-cancerous lesions may progress to invasive ESCC.

Despite advancements in treatment procedures, ESCC prognosis is still poor, emphasizing the need for a better understanding of the main processes that stimulate tumorigenesis and immune escape.

IMMUNE ESCAPE: A CANCEROUS DEFENSE MECHANISM

ESCC development is counterbalanced by the host immune system, which employs a two-edged plan for immune surveillance, including innate and adaptive immunity[8]. The ability of cancer cells to evade the detection of immune system and elimination is a hallmark of tumor progression[9]. In ESCC, the immune escape demonstrates through a sophisticated connection of various mechanisms utilized by tumor cells to create a microenvironment that protects them from immune attack[10].

To escape the strong immune surveillance system, ESCC cells organize a multi-dimensional immune escape program (Figure 1). One of the key mechanisms includes downregulation of major histocompatibility complex (MHC) molecules on the cell surface[11]. MHC molecules function as antigen presentation platforms, allowing T cells to recognize and eliminate foreign or abnormal cells. By reducing MHC expression, ESCC cells become undetectable to the immune system, making them resistant to T cell-mediated cytotoxicity[12].

Figure 1
Figure 1 Aryl hydrocarbon receptor signaling in esophageal squamous cell carcinoma development and immune escape. This figure illustrates the multifaceted influence of aryl hydrocarbon receptor (AhR) signaling on esophageal squamous cell carcinoma (ESCC) development and immune escape. Activation of AhR by environmental carcinogens or other ligands initiates a cascade of cellular processes that contribute to tumorigenesis. EMT: Epithelial-mesenchymal transition; NH3: Ammonia; PD-L1: Programmed death-ligand 1; ROS: Reactive oxygen species; TEPA: Triethylenetetramine.

Furthermore, ESCC cells actively modulate the tumor microenvironment to create an immunosuppressive state[13]. They achieve this by secreting chemokines and cytokines that recruit and activate regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs)[10]. The suppressive cell populations directly inhibit the T lymphocytes (CTLs) and suppress the immune response[8].

Immune escape in ESCC also involves utilization of immune checkpoint pathways. These pathways, mediated by molecules such as PD-L1 and CTLA-4, act as a regulatory brake on the immune system to prevent excessive immune activation and autoimmunity[13,14]. ESCC cells can upregulate the expression of these checkpoint ligands, allowing them to bind to their corresponding receptors on T cells and effectively deactivate the anti-tumor T cell response[15].

In addition to the previously mentioned mechanisms, ESCC cells show a flexible immune escape repertoire. They can express ligands that bind to inhibitory receptors on T cells, leading to T cell exhaustion and dysfunction[13]. ESCC cells can also reprogram their metabolism to escape immune recognition and resist immune-mediated cytotoxicity[10]. Furthermore, they can produce immunosuppressive enzymes, such as indoleamine 2,3-dioxygenase, which reduces essential amino acids required for T cell activation and function, disabling the anti-tumor immune response[16].

By using these diverse immune escape methods, ESCC cells create a microenvironment that protects them from immune attack and promotes tumor progression. Understanding these mechanisms is crucial for developing novel therapeutic strategies that can enhance the anti-tumor immune response and improve patient outcomes.

ARYL HYDROCARBON RECEPTOR SIGNALING: A MASTER REGULATOR WITH DIVERSE FUNCTIONS IN HEALTH AND DISEASE

Aryl hydrocarbon receptor (AhR) is a ubiquitous and complicated ligand-activated transcription factor that has significant influences on various physiological processes[17]. AhR, which is primarily found in the cytoplasm with chaperone proteins, goes through significant changes upon binding to ligands[18]. These ligands are categorized into two main groups: Exogenous, which includes environmental pollutants such as polycyclic aromatic hydrocarbons (PAHs), and endogenous, which comprises metabolites from tryptophan breakdown, such as tryptophan and kynurenine[19]. When a ligand binds, AhR alters its shape, releases from its chaperone complex, and moves to the nucleus[20]. It pairs with ARNT and attaches to specific DNA sequences called xenobiotic response elements in the regulatory regions of the target genes. This interaction initiates the transcription of various genes, influencing several cellular processes via AhR signaling[21]. Beyond its well-recognized role in xenobiotic metabolism, AhR signaling applies an excessive influence on numerous biological processes, including cellular proliferation and differentiation[22], angiogenesis[23], cellular metabolism[24], oxidative stress response[25], immune regulation[26], and inflammation[27].

Studies have demonstrated that AhR signaling affects the proliferation and differentiation of epithelial cells, lymphocytes, and hematopoietic progenitor cells[22]. Moreover, AhR may influence the expression of pro-angiogenic factors such as vascular endothelial growth factor, and impact both physiological and pathological angiogenesis[23]. AhR activation also influences insulin sensitivity, glucose uptake, and lipogenesis. Dysregulation of these pathways may contribute to the development of metabolic disorders, such as obesity and type 2 diabetes[24]. AhR signaling can influence cellular metabolism and, promote the Warburg effect. This metabolic shift not only enhances tumor cell proliferation, but can also create an immunosuppressive microenvironment by changing the levels of metabolites and signaling molecules that modulate immune cells[28]. The cellular environment is consistently influenced by the reactive oxygen species produced during normal metabolic processes. AhR can modulate the antioxidant defense system by targeting the expression of enzymes such as heme oxygenase-1, NAD, and quinine oxidoreductase 1, which are important in protecting cells from oxidative damage[25].

AhR signaling exerts a detailed and context-specific influence on the immune system. It modulates the function of several immune cell populations, including T cells, B cells, and antigen-presenting cells (APCs)[26]. AhR activation can promote the differentiation of Tregs with immunosuppressive properties, while simultaneously inhibiting the proliferation and function of effector T cells[29]. AhR can influence the expression of MHC, and co-stimulatory molecules on APCs, and impact the efficiency of antigen presentation and T cell activation[30]. The tumor microenvironment is often characterized by chronic inflammation, which can promote tumorigenesis. AhR signaling can influence the expression of inflammatory mediators, such as cytokines and chemokines, forming an inflammatory milieu within the tumor microenvironment. While some inflammatory responses can promote anti-tumor immunity, chronic inflammation initiated by aberrant AhR activation can create a pro-tumorigenic microenvironment[27]. The multi-dimensional nature of the AhR highlights its potential as a double-edged sword. While it contributes to essential physiological processes, including xenobiotic metabolism, immune homeostasis, and tissue development, its dysregulation can be involved in various disease pathologies, including cancer. Understanding the important role of AhR signaling in ESCC, especially its influence on the tumor immune microenvironment and immune escape mechanisms, could be beneficial in the development of novel therapeutic strategies.

AHR AND ESCC DEVELOPMENT

Exposure to environmental carcinogens, such as PAHs found in cigarette smoke and certain dietary components is a well-established risk factor for ESCC development[31]. These carcinogens can activate AhR signaling via direct ligand binding[32]. The activated AhR pathway influences multiple cellular processes that contribute to ESCC tumorigenesis. Studies have demonstrated that AhR activation can promote the proliferation of esophageal epithelial cells by modulating cell cycle regulatory proteins such as cyclin D1 and p21[33]. Moreover, AhR signaling can inhibit apoptosis and, the programmed cell death pathway, by influencing the expression of anti-apoptotic factors such as Bcl-2[34]. These effects create conditions favorable for deregulated cell proliferation, which is a characteristic of cancer. Chronic exposure to carcinogens can induce DNA damage in esophageal epithelial cells[35]. However, AhR activation can further exacerbate this issue by downregulating the expression of key DNA repair enzymes[36]. This impaired repair system increases the accumulation of mutations and, eventually increases the risk of malignant transformation. Cancer cells exhibit a distinct metabolic profile compared to healthy cells. ESCC development and progression may be initiated by a subpopulation of cancer stem cells with self-renewal and differentiation capabilities[37,38]. AhR activation has been studied in promoting the stemness properties of ESCC cells[39].

Epithelial-mesenchymal transition (EMT) is a critical process by which epithelial cells lose their polarized phenotype and acquire mesenchymal characteristics, gaining migratory and invasive potential[40]. AhR activation has been shown to induce EMT in various cell types, including esophageal epithelial cells[41]. Studies have demonstrated that AhR ligands can upregulate the expression of EMT-inducing transcription factors such as Twist1 and Snail, promoting the acquisition of an invasive phenotype by cancer cells[42]. The EMT process is crucial for ESCC progression and metastasis, by enabling cancer cells to detach from the primary tumor, migrate through the basement membrane, and invade the surrounding tissues and lymphatic vessels. EMT can also enhance the resistance of ESCC cells to anoikis, a type of cell death triggered by detachment from the extracellular matrix[43]. This combined effect of increased motility and resistance to anoikis promotes the dissemination of ESCC cells to other organs, resulting in the formation of metastases.

AHR AND IMMUNE ESCAPE IN ESCC

While certain studies have indicated that activating AhR might enhance anti-cancer immune responses, other studies suggest its possible involvement in immune evasion mechanisms in ESCC. AhR activation can suppress effector T cells, the main attacking cells of the adaptive immune response[44]. Studies have shown that some AhR ligands can inhibit the proliferation and cytokine production of CD8+ CTLs and weaken the immune response against ESCC cells[45]. Additionally, AhR activation can promote the differentiation of Tregs, a population of immunosuppressive T cells that dampens the anti-tumor immune response[46]. The tumor microenvironment in ESCC is often infiltrated by immunosuppressive myeloid cells, including MDSCs[13]. AhR activation promotes the recruitment and expansion of MDSCs, which contributes to the suppression of anti-tumor immunity[47]. Immune checkpoint molecules, such as PD-L1 and CTLA-4, act as inhibitory agents in the immune system to prevent autoimmunity. However, ESCC cells can use these checkpoints to escape immune response[48]. Some studies suggested that AhR activation upregulates the expression of PD-L1 on ESCC cells, potentially participating in immune escape by inhibiting T cell function[49]. Table 1 summarizes the influence of AhR signaling on ESCC development, progression, and immune escape.

Table 1 Aryl hydrocarbon receptor signaling and its impact on.
Aspect of ESCC
Impact of AhR activation
Mechanism
Ref.
Carcinogenesis
ProliferationIncreasedUpregulates cyclin D1, downregulates p21[31,33]
ApoptosisInhibitedUpregulates anti-apoptotic factors like Bcl-2[34]
DNA repairImpairedDownregulates expression of key DNA repair enzymes[36]
Metabolic reprogrammingPromotes Warburg effectIncreases reliance on aerobic glycolysis[28]
StemnessEnhancedPromotes self-renewal and differentiation of cancer stem cells[37,38]
EMTInducedUpregulates EMT-inducing transcription factors like Twist1 and Snail[41,43]
Immune escape
T cell functionSuppressedInhibits proliferation and cytokine production of CD8+ CTLs, promotes differentiation of Tregs[45,46]
Myeloid cell recruitmentIncreasedPromotes recruitment and expansion of MDSCs[47]
Immune checkpoint regulationPotential upregulation of PD-L1 expressionMay contribute to immune escape by inhibiting T cell activity[49]
THERAPEUTIC POTENTIAL OF TARGETING AHR SIGNALING IN ESCC

The complex influence of AhR signaling on ESCC development, progression, and immune escape presents an interesting opportunity for the development of novel therapeutic strategies. While the aberrant AhR activation can promote tumorigenesis and create an immunosuppressive microenvironment, its proper modulation could be beneficial for disrupting these processes and boosting the anti-tumor immune response.

Preclinical studies investigating AhR ligands in ESCC models have yielded in interesting results[49]. Several antagonists could competitively bind to the ligand-binding domain, and block the activation of AhR signaling pathways and their downstream effects on ESCC cells[49]. Studies have demonstrated that AhR antagonists can suppress ESCC cell proliferation, migration, and invasion, potentially inhibiting tumor progression and metastasis[50]. AhR antagonists exhibit immunomodulatory effects within the tumor microenvironment, by inhibiting AhR signaling. These antagonists may reduce the population of immunosuppressive MDSCs and Tregs, and simultaneously enhancing the function and proliferation of effector T cells, leading to a more powerful anti-tumor immune response[51,52]. Several AhR antagonists are currently under investigation for their therapeutic potential in ESCC, including CH-223191, NH3 (Ammonia), and specific small molecules[53]. Not only AhR antagonists but also some naturally derived AhR agonists, such as curcumin[54] and quercetin[55] have been shown to exhibit regulatory properties on immune response in cancer. This controversy could be due to the structural and metabolic differences of different AhR ligands. Furthermore, synthetic AhR antagonists are being actively developed, with some demonstrating promising preclinical results[51]. Table 2 has summarized some of the most promising candidates, their mechanisms of action, and potential effects on malignancies.

Table 2 Mechanisms of action and potential effects of promising aryl hydrocarbon receptor antagonists on malignancies.
AhR ligands
Mechanism of action
Potential effects on malignancies
Ref.
Natural products
CurcuminCompetitive binding to the AhR ligand-binding domainSuppresses proliferation, migration, and invasion of cancer cells; may enhance anti-tumor immunity[54,62,63]
QuercetinCompetitive binding to the AhR ligand-binding domain; may also inhibit AhR nuclear translocationSuppresses proliferation and induces apoptosis in cancer cells[55]
ResveratrolMay interfere with AhR-DNA binding; may also possess antioxidant and anti-inflammatory propertiesMay inhibit tumor growth and metastasis[55,64]
Synthetic ligands
CH-223191Competitive binding to the AhR ligand-binding domainSuppresses proliferation and migration of cancer cells[65]
NH3 (Ammonia)Competitive binding to the AhR ligand-binding domainShows promise in preclinical models, but may have limitations due to potential toxicity[66]
Small molecule antagonistsCompetitive binding to the AhR ligand-binding domain or interfering with AhR dimerizationSeveral novel small molecules are under development, with preclinical studies ongoing[67,68]

Beyond directly targeting the AhR itself, manipulating the endogenous ligands that activate this pathway offers another potential therapeutic avenue. The gut microbiota plays an important role in metabolizing dietary components into ligands that can activate AhR signaling[56]. Strategies aimed at modulating the gut microbiota composition, potentially through prebiotics or probiotics, could be studied to influence the production of these ligands and their subsequent impact on AhR signaling in ESCC[57]. Moreover, dietary factors can also influence AhR ligand production. Identifying and employing natural AhR modulators, such as indole-3-carbinol found in cruciferous vegetables, into the diet could be a preventive or complementary therapeutic approach for ESCC patients[58,59]. Some AhR agonists such as TCDD bind tightly to the AhR, leading to long-lasting activation that disrupts normal cellular processes and promotes cancer development. However, I3C, and its metabolites transiently activate the AhR. This controlled activation helps in detoxifying carcinogens, inhibiting abnormal cell growth, and promoting cancer cell death, highlighting their potential anticancer effects[58,59]. However, the effectiveness of dietary AhR modulators is not well-established by the lack of comprehensive studies on the metabolic kinetics, permeability, and transport of dietary compounds. Future research should focus on these aspects to better understand and optimize the role of these compounds on ESCC treatment.

The therapeutic potential of AhR-targeted strategies may be further intensified when combined with existing treatment approaches for ESCC. For instance, combining selected AhR ligands (agonist or antagonist) with conventional therapies such as surgery, radiation, or chemotherapy could offer synergistic effects. These traditional therapies can induce DNA damage and cell death in ESCC cells, while AhR ligands may suppress tumor growth and metastasis by inhibiting proliferation and invasion[60]. Additionally, AhR ligands could be particularly effective when combined with immune checkpoint inhibitors[61]. Clinical trials investigating the combination of AhR-targeted therapies with other treatment strategies are suggested to determine their efficacy and safety in ESCC patients.

CONCLUSION

Immune escape mechanisms play a crucial role in ESCC progression, allowing cancer cells to evade the surveillance of the host immune system. The AhR activation, triggered by environmental carcinogens such as PAHs, promotes various hallmarks of cancer, including proliferation, apoptosis inhibition, and metabolic reprogramming. Moreover, AhR signaling contributes to immune escape by suppressing T cell function, recruiting immunosuppressive myeloid cells, and upregulating immune checkpoint molecules like PD-L1. Targeting AhR signaling presents a promising therapeutic approach for ESCC. AhR ligands (agonist or antagonist), both natural and synthetic, have shown potential in preclinical studies by inhibiting tumor growth and modulating the immune microenvironment. Modulating AhR ligands through dietary interventions may offer new therapeutic approaches. Furthermore, combining AhR-targeted therapies with conventional treatments or immune checkpoint inhibitors may result in synergistic effects, enhancing overall therapeutic efficacy.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Immunology

Country of origin: Oman

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade A

Creativity or Innovation: Grade A

Scientific Significance: Grade B

P-Reviewer: Wang Z S-Editor: Liu JH L-Editor: Filipodia P-Editor: Wang WB

References
1.  Sheikh M, Roshandel G, McCormack V, Malekzadeh R. Current Status and Future Prospects for Esophageal Cancer. Cancers (Basel). 2023;15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 38]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
2.  Tungekar A, Mandarthi S, Mandaviya PR, Gadekar VP, Tantry A, Kotian S, Reddy J, Prabha D, Bhat S, Sahay S, Mascarenhas R, Badkillaya RR, Nagasampige MK, Yelnadu M, Pawar H, Hebbar P, Kashyap MK. ESCC ATLAS: A population wide compendium of biomarkers for Esophageal Squamous Cell Carcinoma. Sci Rep. 2018;8:12715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
3.  Testa U, Castelli G, Pelosi E. Esophageal Cancer: Genomic and Molecular Characterization, Stem Cell Compartment and Clonal Evolution. Medicines (Basel). 2017;4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 58]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
4.  Tarazi M, Chidambaram S, Markar SR. Risk Factors of Esophageal Squamous Cell Carcinoma beyond Alcohol and Smoking. Cancers (Basel). 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 21]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
5.  Mandard AM, Hainaut P, Hollstein M. Genetic steps in the development of squamous cell carcinoma of the esophagus. Mutat Res. 2000;462:335-342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 148]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
6.  Shi Y, Li MY, Wang H, Li C, Liu WY, Gao YM, Wang B, Song JW, Ma YQ. The Relationship between MACC1/c-Met/Cyclin D1 Axis Expression and Prognosis in ESCC. Anal Cell Pathol (Amst). 2022;2022:9651503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
7.  Liu WJ, Zhao Y, Chen X, Miao ML, Zhang RQ. Epigenetic modifications in esophageal cancer: An evolving biomarker. Front Genet. 2022;13:1087479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
8.  Zheng Y, Chen Z, Han Y, Han L, Zou X, Zhou B, Hu R, Hao J, Bai S, Xiao H, Li WV, Bueker A, Ma Y, Xie G, Yang J, Chen S, Li H, Cao J, Shen L. Immune suppressive landscape in the human esophageal squamous cell carcinoma microenvironment. Nat Commun. 2020;11:6268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 215]  [Article Influence: 53.8]  [Reference Citation Analysis (0)]
9.  Tang S, Ning Q, Yang L, Mo Z, Tang S. Mechanisms of immune escape in the cancer immune cycle. Int Immunopharmacol. 2020;86:106700.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 65]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
10.  Li R, Huang B, Tian H, Sun Z. Immune evasion in esophageal squamous cell cancer: From the perspective of tumor microenvironment. Front Oncol. 2022;12:1096717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
11.  Zhang XJ, Yu Y, Zhao HP, Guo L, Dai K, Lv J. Mechanisms of tumor immunosuppressive microenvironment formation in esophageal cancer. World J Gastroenterol. 2024;30:2195-2208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (8)]
12.  Garrido F, Aptsiauri N. Cancer immune escape: MHC expression in primary tumours versus metastases. Immunology. 2019;158:255-266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 97]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
13.  Baba Y, Nomoto D, Okadome K, Ishimoto T, Iwatsuki M, Miyamoto Y, Yoshida N, Baba H. Tumor immune microenvironment and immune checkpoint inhibitors in esophageal squamous cell carcinoma. Cancer Sci. 2020;111:3132-3141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 149]  [Article Influence: 37.3]  [Reference Citation Analysis (0)]
14.  Zhang H, Dai Z, Wu W, Wang Z, Zhang N, Zhang L, Zeng WJ, Liu Z, Cheng Q. Regulatory mechanisms of immune checkpoints PD-L1 and CTLA-4 in cancer. J Exp Clin Cancer Res. 2021;40:184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 212]  [Article Influence: 70.7]  [Reference Citation Analysis (0)]
15.  Mahmoudian RA, Mozhgani S, Abbaszadegan MR, Mokhlessi L, Montazer M, Gholamin M. Correlation between the immune checkpoints and EMT genes proposes potential prognostic and therapeutic targets in ESCC. J Mol Histol. 2021;52:597-609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
16.  Liu J, Lu G, Tang F, Liu Y, Cui G. Localization of indoleamine 2,3-dioxygenase in human esophageal squamous cell carcinomas. Virchows Arch. 2009;455:441-448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
17.  Kou Z, Dai W. Aryl hydrocarbon receptor: Its roles in physiology. Biochem Pharmacol. 2021;185:114428.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 32]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
18.  Denison MS, Pandini A, Nagy SR, Baldwin EP, Bonati L. Ligand binding and activation of the Ah receptor. Chem Biol Interact. 2002;141:3-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 331]  [Cited by in F6Publishing: 320]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
19.  Safe S, Jin UH, Park H, Chapkin RS, Jayaraman A. Aryl Hydrocarbon Receptor (AHR) Ligands as Selective AHR Modulators (SAhRMs). Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 56]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
20.  Safe S, Han H, Goldsby J, Mohankumar K, Chapkin RS. Aryl Hydrocarbon Receptor (AhR) Ligands as Selective AhR Modulators: Genomic Studies. Curr Opin Toxicol. 2018;11-12:10-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 29]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
21.  Kerkvliet NI. AHR-mediated immunomodulation: the role of altered gene transcription. Biochem Pharmacol. 2009;77:746-760.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 134]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
22.  Yin J, Sheng B, Qiu Y, Yang K, Xiao W, Yang H. Role of AhR in positive regulation of cell proliferation and survival. Cell Prolif. 2016;49:554-560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 49]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
23.  Li Y, Wang K, Zou QY, Jiang YZ, Zhou C, Zheng J. ITE Suppresses Angiogenic Responses in Human Artery and Vein Endothelial Cells: Differential Roles of AhR. Reprod Toxicol. 2017;74:181-188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
24.  Bock KW. Aryl hydrocarbon receptor (AHR) functions in NAD(+) metabolism, myelopoiesis and obesity. Biochem Pharmacol. 2019;163:128-132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
25.  Huang Y, Zhang J, Tao Y, Ji C, Aniagu S, Jiang Y, Chen T. AHR/ROS-mediated mitochondria apoptosis contributes to benzo[a]pyrene-induced heart defects and the protective effects of resveratrol. Toxicology. 2021;462:152965.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 34]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
26.  Hao N, Whitelaw ML. The emerging roles of AhR in physiology and immunity. Biochem Pharmacol. 2013;86:561-570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 155]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
27.  Neavin DR, Liu D, Ray B, Weinshilboum RM. The Role of the Aryl Hydrocarbon Receptor (AHR) in Immune and Inflammatory Diseases. Int J Mol Sci. 2018;19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 156]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
28.  Gabriely G, Wheeler MA, Takenaka MC, Quintana FJ. Role of AHR and HIF-1α in Glioblastoma Metabolism. Trends Endocrinol Metab. 2017;28:428-436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 80]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
29.  Liu X, Hu H, Fan H, Zuo D, Shou Z, Liao Y, Nan Z, Tang Q. The role of STAT3 and AhR in the differentiation of CD4+ T cells into Th17 and Treg cells. Medicine (Baltimore). 2017;96:e6615.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 29]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
30.  Mohammadi S, Memarian A, Sedighi S, Behnampour N, Yazdani Y. Immunoregulatory effects of indole-3-carbinol on monocyte-derived macrophages in systemic lupus erythematosus: A crucial role for aryl hydrocarbon receptor. Autoimmunity. 2018;51:199-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 47]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
31.  Abedi-Ardekani B, Kamangar F, Hewitt SM, Hainaut P, Sotoudeh M, Abnet CC, Taylor PR, Boffetta P, Malekzadeh R, Dawsey SM. Polycyclic aromatic hydrocarbon exposure in oesophageal tissue and risk of oesophageal squamous cell carcinoma in north-eastern Iran. Gut. 2010;59:1178-1183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 70]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
32.  Wang Z, Snyder M, Kenison JE, Yang K, Lara B, Lydell E, Bennani K, Novikov O, Federico A, Monti S, Sherr DH. How the AHR Became Important in Cancer: The Role of Chronically Active AHR in Cancer Aggression. Int J Mol Sci. 2020;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 51]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
33.  Liu J, Min L, Zhu S, Guo Q, Li H, Zhang Z, Zhao Y, Xu C, Zhang S. Cyclin-Dependent Kinase Inhibitor 3 Promoted Cell Proliferation by Driving Cell Cycle from G1 to S Phase in Esophageal Squamous Cell Carcinoma. J Cancer. 2019;10:1915-1922.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
34.  Al-Dhfyan A, Alaiya A, Al-Mohanna F, Attwa MW, AlAsmari AF, Bakheet SA, Korashy HM. Crosstalk between aryl hydrocarbon receptor (AhR) and BCL-2 pathways suggests the use of AhR antagonist to maintain normal differentiation state of mammary epithelial cells during BCL-2 inhibition therapy. J Adv Res. 2023;50:177-192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
35.  Bai H, Wu M, Zhang H, Tang G. Chronic polycyclic aromatic hydrocarbon exposure causes DNA damage and genomic instability in lung epithelial cells. Oncotarget. 2017;8:79034-79045.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
36.  Pollet M, Shaik S, Mescher M, Frauenstein K, Tigges J, Braun SA, Sondenheimer K, Kaveh M, Bruhs A, Meller S, Homey B, Schwarz A, Esser C, Douki T, Vogel CFA, Krutmann J, Haarmann-Stemmann T. The AHR represses nucleotide excision repair and apoptosis and contributes to UV-induced skin carcinogenesis. Cell Death Differ. 2018;25:1823-1836.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 52]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
37.  Wu Q, Wu Z, Bao C, Li W, He H, Sun Y, Chen Z, Zhang H, Ning Z. Cancer stem cells in esophageal squamous cell cancer. Oncol Lett. 2019;18:5022-5032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
38.  Khosravi A, Jafari SM, Asadi J. Knockdown of TAZ decrease the cancer stem properties of ESCC cell line YM-1 by modulation of Nanog, OCT-4 and SOX2. Gene. 2021;769:145207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
39.  Wu C, Yu S, Tan Q, Guo P, Liu H. Role of AhR in regulating cancer stem cell-like characteristics in choriocarcinoma. Cell Cycle. 2018;17:2309-2320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
40.  Brabletz T, Kalluri R, Nieto MA, Weinberg RA. EMT in cancer. Nat Rev Cancer. 2018;18:128-134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1104]  [Cited by in F6Publishing: 1317]  [Article Influence: 219.5]  [Reference Citation Analysis (0)]
41.  Zhu P, Yu H, Zhou K, Bai Y, Qi R, Zhang S. 3,3'-Diindolylmethane modulates aryl hydrocarbon receptor of esophageal squamous cell carcinoma to reverse epithelial-mesenchymal transition through repressing RhoA/ROCK1-mediated COX2/PGE(2) pathway. J Exp Clin Cancer Res. 2020;39:113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 27]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
42.  Chuang KT, Chiou SS, Hsu SH. Recent Advances in Transcription Factors Biomarkers and Targeted Therapies Focusing on Epithelial-Mesenchymal Transition. Cancers (Basel). 2023;15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
43.  Liao C, Wang Q, An J, Long Q, Wang H, Xiang M, Xiang M, Zhao Y, Liu Y, Liu J, Guan X. Partial EMT in Squamous Cell Carcinoma: A Snapshot. Int J Biol Sci. 2021;17:3036-3047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 28]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
44.  Zhang H, Yang Z, Yuan W, Liu J, Luo X, Zhang Q, Li Y, Chen J, Zhou Y, Lv J, Zhou N, Ma J, Tang K, Huang B. Sustained AhR activity programs memory fate of early effector CD8(+) T cells. Proc Natl Acad Sci U S A. 2024;121:e2317658121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
45.  Wang G, Pan C, Cao K, Zhang J, Geng H, Wu K, Wen J, Liu C. Impacts of Cigarette Smoking on the Tumor Immune Microenvironment in Esophageal Squamous Cell Carcinoma. J Cancer. 2022;13:413-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
46.  Leclerc D, Staats Pires AC, Guillemin GJ, Gilot D. Detrimental activation of AhR pathway in cancer: an overview of therapeutic strategies. Curr Opin Immunol. 2021;70:15-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 41]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
47.  Neamah WH, Singh NP, Alghetaa H, Abdulla OA, Chatterjee S, Busbee PB, Nagarkatti M, Nagarkatti P. AhR Activation Leads to Massive Mobilization of Myeloid-Derived Suppressor Cells with Immunosuppressive Activity through Regulation of CXCR2 and MicroRNA miR-150-5p and miR-543-3p That Target Anti-Inflammatory Genes. J Immunol. 2019;203:1830-1844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 62]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
48.  Banday AH, Abdalla M. Immune Checkpoint Inhibitors: Recent Clinical Advances and Future Prospects. Curr Med Chem. 2023;30:3215-3237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
49.  Zhu P, Zhou K, Lu S, Bai Y, Qi R, Zhang S. Modulation of aryl hydrocarbon receptor inhibits esophageal squamous cell carcinoma progression by repressing COX2/PGE2/STAT3 axis. J Cell Commun Signal. 2020;14:175-192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
50.  To KK, Yu L, Liu S, Fu J, Cho CH. Constitutive AhR activation leads to concomitant ABCG2-mediated multidrug resistance in cisplatin-resistant esophageal carcinoma cells. Mol Carcinog. 2012;51:449-464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
51.  Baker JR, Sakoff JA, McCluskey A. The aryl hydrocarbon receptor (AhR) as a breast cancer drug target. Med Res Rev. 2020;40:972-1001.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 39]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
52.  Sun L. Recent advances in the development of AHR antagonists in immuno-oncology. RSC Med Chem. 2021;12:902-914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
53.  Safe S, Zhang L. The Role of the Aryl Hydrocarbon Receptor (AhR) and Its Ligands in Breast Cancer. Cancers (Basel). 2022;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 13]  [Reference Citation Analysis (0)]
54.  Nakai R, Fukuda S, Kawase M, Yamashita Y, Ashida H. Curcumin and its derivatives inhibit 2,3,7,8,-tetrachloro-dibenzo-p-dioxin-induced expression of drug metabolizing enzymes through aryl hydrocarbon receptor-mediated pathway. Biosci Biotechnol Biochem. 2018;82:616-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
55.  Mohammadi-Bardbori A, Bengtsson J, Rannug U, Rannug A, Wincent E. Quercetin, resveratrol, and curcumin are indirect activators of the aryl hydrocarbon receptor (AHR). Chem Res Toxicol. 2012;25:1878-1884.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 124]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
56.  Dai G, Chen X, He Y. The Gut Microbiota Activates AhR Through the Tryptophan Metabolite Kyn to Mediate Renal Cell Carcinoma Metastasis. Front Nutr. 2021;8:712327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
57.  Yang F, DeLuca JAA, Menon R, Garcia-Vilarato E, Callaway E, Landrock KK, Lee K, Safe SH, Chapkin RS, Allred CD, Jayaraman A. Effect of diet and intestinal AhR expression on fecal microbiome and metabolomic profiles. Microb Cell Fact. 2020;19:219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
58.  Peng C, Wu C, Xu X, Pan L, Lou Z, Zhao Y, Jiang H, He Z, Ruan B. Indole-3-carbinol ameliorates necroptosis and inflammation of intestinal epithelial cells in mice with ulcerative colitis by activating aryl hydrocarbon receptor. Exp Cell Res. 2021;404:112638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
59.  Mohammadi S, Seyedhosseini FS, Behnampour N, Yazdani Y. Indole-3-carbinol induces G1 cell cycle arrest and apoptosis through aryl hydrocarbon receptor in THP-1 monocytic cell line. J Recept Signal Transduct Res. 2017;37:506-514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 28]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
60.  Bayat Mokhtari R, Homayouni TS, Baluch N, Morgatskaya E, Kumar S, Das B, Yeger H. Combination therapy in combating cancer. Oncotarget. 2017;8:38022-38043.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 890]  [Cited by in F6Publishing: 1287]  [Article Influence: 214.5]  [Reference Citation Analysis (0)]
61.  Tan N, Zhao W, Wang Y, Li P, Liu J, Sun Z, Pan J, Song S, Li S, Liu Z, Bian Y. AHR, a novel inhibitory immune checkpoint receptor, is a potential therapeutic target for chemoresistant glioblastoma. J Cancer Res Clin Oncol. 2023;149:9705-9720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
62.  Bahrami A, Majeed M, Sahebkar A. Curcumin: a potent agent to reverse epithelial-to-mesenchymal transition. Cell Oncol (Dordr). 2019;42:405-421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 50]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
63.  Ang HL, Mohan CD, Shanmugam MK, Leong HC, Makvandi P, Rangappa KS, Bishayee A, Kumar AP, Sethi G. Mechanism of epithelial-mesenchymal transition in cancer and its regulation by natural compounds. Med Res Rev. 2023;43:1141-1200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 49]  [Reference Citation Analysis (0)]
64.  Jin Z, Feng W, Ji Y, Jin L. Resveratrol mediates cell cycle arrest and cell death in human esophageal squamous cell carcinoma by directly targeting the EGFR signaling pathway. Oncol Lett. 2017;13:347-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 9]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
65.  Choi EY, Lee H, Dingle RW, Kim KB, Swanson HI. Development of novel CH223191-based antagonists of the aryl hydrocarbon receptor. Mol Pharmacol. 2012;81:3-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
66.  Wu H, Ma W, Wang Y, Wang Y, Sun X, Zheng Q. Gut microbiome-metabolites axis: A friend or foe to colorectal cancer progression. Biomed Pharmacother. 2024;173:116410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
67.  Kober C, Roewe J, Schmees N, Roese L, Roehn U, Bader B, Stoeckigt D, Prinz F, Gorjánácz M, Roider HG, Olesch C, Leder G, Irlbacher H, Lesche R, Lefranc J, Oezcan-Wahlbrink M, Batra AS, Elmadany N, Carretero R, Sahm K, Oezen I, Cichon F, Baumann D, Sadik A, Opitz CA, Weinmann H, Hartung IV, Kreft B, Offringa R, Platten M, Gutcher I. Targeting the aryl hydrocarbon receptor (AhR) with BAY 2416964: a selective small molecule inhibitor for cancer immunotherapy. J Immunother Cancer. 2023;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
68.  Joseph J, Gonzalez-lopez M, Galang C, Garcia C, Lemar H, Lu J, Vaidya K, Fischer M, Frey C, Alam M, Yao B, Dillon M, Hager JH, Venetsanakos E, Aswad F. Abstract 4719: Small-molecule antagonists of the Aryl Hydrocarbon Receptor (AhR) promote activation of human PBMCs in vitro and demonstrate significant impact on tumor growth and immune modulation in vivo. Cancer Res. 2018;78:4719-4719.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]