Next Article in Journal
19th International Symposium on Epstein–Barr Virus and Associated Diseases, 29–30 July 2021, Asahikawa, Japan
Previous Article in Journal
Analysis of Cross-Combinations of Feature Selection and Machine-Learning Classification Methods Based on [18F]F-FDG PET/CT Radiomic Features for Metabolic Response Prediction of Metastatic Breast Cancer Lesions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microbial-Derived Toll-like Receptor Agonism in Cancer Treatment and Progression

1
Division of Hematology, Oncology, and Cell Therapy, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA
2
Division of Translational and Precision Medicine, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA
3
Division of Pediatric Surgery, Department of Surgery, Rush University Medical Center, Chicago, IL 60612, USA
4
Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, IL 60612, USA
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(12), 2923; https://doi.org/10.3390/cancers14122923
Submission received: 18 May 2022 / Revised: 2 June 2022 / Accepted: 13 June 2022 / Published: 14 June 2022
(This article belongs to the Topic Anti-Tumor Immune Responses)

Abstract

:

Simple Summary

Toll like receptors (TLRs) are a group of transmembrane receptors belonging to the class of pattern recognition receptors (PRR), which are involved in recognition of pathogen associated molecular patterns (PAMPs), inducing immune response. During the past decade, a number of preclinical and clinical breakthroughs in the field of TLR agonists has immerged in cancer research and some of these agents have performed exceptionally well in clinical trials. Based on evidence from scientific studies, we draw attention to several microbial based TLR agonists and discuss their relevance in various cancer and explore various microbial based TLR agonists for developing effective immunotherapeutic strategies against cancer.

Abstract

Toll-like receptors (TLRs) are typical transmembrane proteins, which are essential pattern recognition receptors in mediating the effects of innate immunity. TLRs recognize structurally conserved molecules derived from microbes and damage-associated molecular pattern molecules that play an important role in inflammation. Since the first discovery of the Toll receptor by the team of J. Hoffmann in 1996, in Drosophila melanogaster, numerous TLRs have been identified across a wide range of invertebrate and vertebrate species. TLR stimulation leads to NF-κB activation and the subsequent production of pro-inflammatory cytokines and chemokines, growth factors and anti-apoptotic proteins. The expression of TLRs has also been observed in many tumors, and their stimulation results in tumor progression or regression, depending on the TLR and tumor type. The anti-tumoral effects can result from the activation of anti-tumoral immune responses and/or the direct induction of tumor cell death. The pro-tumoral effects may be due to inducing tumor cell survival and proliferation or by acting on suppressive or inflammatory immune cells in the tumor microenvironment. The aim of this review is to draw attention to the effects of TLR stimulation in cancer, the activation of various TLRs by microbes in different types of tumors, and, finally, the role of TLRs in anti-cancer immunity and tumor rejection.

1. Introduction

The Toll receptor was identified in Drosophila melanogaster, a receptor essential for establishing the dorsal–ventral axis during embryonic development [1,2]. Following the discovery of the Toll receptor, several mammalian proteins were found to share structural similarities to the D. melanogaster Toll receptor, and thus they were named Toll-like receptors (TLRs) [3]. In 2011, the Nobel Prize in Physiology/Medicine was awarded to Dr. Jules A. Hoffmann and Dr. Bruce A. Beutler who made significant contributions to the discoveries concerning TLRs and their role in innate immunity [4,5]. It was by this discovery that scientists found innate immunity is essential and critical in immune responses to pathogen infections in connection with the activation of adaptive immunity [6].
TLRs comprise a family of type I transmembrane receptors, which are characterized by an extracellular leucine-rich repeat (LRR) domain and an intracellular Toll/IL-1 receptor (TIR) domain [7,8,9]. TLRs trigger immune responses against various invading pathogens by recognizing specific pathogen-associated molecular patterns (PAMPs), which are highly conserved and derived from potential pathogenic microorganisms, such as bacteria, viruses, fungi, and parasites [10,11].
Microbial sensing through TLRs initiates a signaling cascade that induces pro-inflammatory responses [10]. TLRs recruit TIR-specific domain-containing adaptor proteins for the activation of downstream signaling. The main domains recruited by TLRs are the myeloid differentiation factor-88 (MyD88), the Toll/IL-1 receptor domain adaptor protein, and the TIR-domain-containing adapter-inducing interferon-β (TRIF) [12,13]. Upon activation, various signaling pathways are initiated, which results in a variety of inflammatory cytokines, thus undergoing transcription by the phosphorylation of IkBa to activate NF-κB [14,15]. The multiple signaling pathways contribute to the rapid response of the innate immune system to pathogens. TLRs also regulate adaptive immunity by the activation and maturation of dendritic cells and the production of pro-inflammatory cytokines and chemokines, which induce the proliferation and differentiation of Th1 and Th2 cells [16].
TLR signaling has been well-studied in various diseases, including cancer. TLRs are expressed not only on the surface of immune cells but also on tumor cells [17,18]. TLRs are documented to have pro and anti-tumor responses, though to date, the role of TLR signaling is still not completely understood in cancer progression. This review will describe the usage of bacterial- and viral-derived TLR activation in cancer immunotherapy, TLR expression profile on tumors, and the involvement of TLR signaling in tumor outcomes. We will also discuss the status of research in utilizing TLR agonists as potential therapeutic agents in cancer treatment.

2. TLR Localization and Recognition of Microbial Ligands

TLRs are involved in the recognition of microbial exogenously and endogenously derived molecular patterns. This occurs at the plasma membrane and at intracellular compartments, respectively, and thus TLR ligands can be either exogenous or host-derived. TLRs 1, 2, 4, 5, and 6 are located primarily in the plasma membrane, where they interact with components of microbial pathogens that exogenously encounter the cell. In contrast, TLRs 3, 7, 8, and 9 are situated in the membranes of endosomes and lysosomes, where they interact with components that are endogenous. Figure 1 depicts in detail the location of specific TLRs and their respective, best-characterized ligands.

3. Expression of TLRs on Tumor Cells and Its Clinical Relevance

TLRs are known to be expressed and activated in innate immune cells, such as macrophages and dendritic cells (DCs); however, in recent years, several studies have shown that TLRs are also highly expressed by various tumor cells. Therefore, the study of TLR expression and function in cancer has become a focus for researchers in field of cancer immunotherapy. Table 1 summarizes the TLRs expressed on various tumor cells in different cancer types along with clinical outcomes.

4. Microbial Derived TLR Agonists and Their Role in Cancer Immunotherapy

Harnessing the host response to infection has been utilized to target cancer for centuries [81,82]. Microbial sensing through TLRs initiates a signaling cascade that induces pro-inflammatory responses. The Toll-like receptor signaling pathway plays a crucial role in host immune defenses against numerous diseases and has been identified as an immunotherapeutic target against various types of cancer. New avenues to combat cancers involves the regulation of the host’s innate immune system using agonists, which can bind to a variety of TLRs. These agonists can be used in combination with other cancer therapies, like chemotherapy and radiotherapy, to provide a broader spectrum of protection.

5. Bacterial-Derived TLR Agonists

Many of the pivotal studies in the field of bacterial-based cancer immunotherapy (BBCT) were pioneered by 19th century clinician–scientist Dr. William Coley [81,82]. Dr. Coley utilized a combination of live Streptococcus pyogenes and Serratia marcescens to treat patients with inoperable sarcomas [81]. In the patients with the most dramatic tumor regression, Dr. Coley noted that an erysipelas infection and subsequent fever had been induced in these patients. Dr. Coley’s observations were likely a result of robust immune system activation from Coley’s bacterial-based toxins, thus implicating a link between response to infection and tumor eradication that would serve as the basis for modern cancer immunotherapies. Though unbeknownst to Dr. Coley, a likely mediator in his patients’ responses was innate microbial recognition through Toll-like receptors [82,83].
Molecular triggers responsible for remissions from Coley’s toxins are TLR ligands that target patter recognition receptors (PRRs) [84]. PRRs share the ability to recognize relatively conserved microbial components, which are generally referred to as microbe- or pathogen-associated molecular patterns (MAMPs or PAMPs), as well as endogenous danger signals commonly known as damage-associated molecular patterns (DAMPs). Common TLR-activating MAMPs include viral and bacterial nucleic acids (which can signal through TLR3, TLR7, TLR8, or TLR9), flagellin (a TLR5 agonist), as well as lipopolysaccharide (LPS), lipoteichoic acid, and mannans (which signal through TLR2 or TLR4). Endogenous nucleic acids and the nuclear non-histone protein high mobility group box 1 (HGMB1) are prototypic TLR-activating DAMPs.
Due to their role in self/nonself-differentiation [85] and their ability to induce antigen presenting cell (APC) maturation, TLR agonists are considered promising adjuvant candidates [86]. In fact, a number of TLR agonists, including Pam3CSK4, Pam2CSK4, MPLA (a LPS derivative), CpG, PolyI:C, and flagellin, are currently being tested as cancer agonists [87].
Roberts et al. recently showed that an attenuated strain of Clostridium novyi efficiently decreased tumor size in rat and dog cancer models in addition to one sarcoma patient [88]. This treatment is well-targeted as spores of Clostridium novyi germinate selectively within the hypoxic regions of cancerous tissue and induce immune responses likely via TLR activation [89].

6. Viral-Derived TLR Agonists

Oncolytic viruses (OVs) preferentially target tumor cells and activate antitumor immunity while limiting pathogenicity, thus they have emerged as a promising tool for viral-based cancer therapies [90]. Though additional studies must be conducted to further characterize the TLR contribution to antitumor responses from oncolytic virotherapy, OVs possess several TLR-stimulating moieties that can contribute to activation of host immune response at the tumor site. Intratumoral treatment of murine glioma tumors with oncolytic adenovirus Delta-24-RGD significantly reduced tumor growth compared to PBS-treated tumors [91]. Delta-24-RGD treatment was demonstrated to remodel the tumor microenvironment, predominantly through enhanced CD8+ and CD4+ T cell infiltration in the tumor [91]. Immune-mediated targeting of the tumor is likely a result of TLR9-medated T cell proliferation as well as maturation of APCs, following the TLR9 recognition of double-stranded adenoviral DNA [92,93,94].
Enhancing the intrinsic TLR stimulating properties of oncolytic adenoviruses has been another strategy for targeting TLR-mediated immune responses. Ad5D24-CpG, an oncolytic adenovirus genetically manipulated to express TLR9 stimulating CpG islands, significantly controlled tumor growth compared to CpG unenhanced Ad5D24 treated tumors [95]. Antitumor response from Ad5D24-CpG treatment was determined to be highly reliant on TLR9-mediated NK cell activation, leading to the effective killing of tumor cells [95]. Because stimulation of TLRs expressed on NK cells enhances release of cytotoxic granules and cytokine production, in this context it is suggested that the CpG insertion in Ad5D24 enhances these NK cell properties, facilitating more robust antitumor responses [96,97].
TLRs have also been implicated in the usage of non-oncolytic viruses for viral-based therapeutics. The intratumoral administration of heat-inactivated influenza but not active influenza virus was found to drastically inhibit tumor growth in a B16 melanoma model [98]. This response from heat-inactivated influenza treatment was determined to be dependent on increased cross-presenting CD8+ dendritic cell infiltration in the tumor [98]. Paralleling the in vivo findings, heat-inactivated influenza was observed to more potently stimulate TLR7 compared to active influenza virus, determined by using TLR7 reporter cells [98]. Given that the recognition of TLR ligands can enhance antigen cross presentation in DCs, it is likely that heat-inactivated influenza more potently engaged TLR7, leading to an increase in cross-presenting DCs within the tumor and the activation of CD8+ T cells and thereby reduced tumor progression [99,100,101].

7. TLR–TLR Cross-Talk and the Modulation of Immune Response

Targeting the ability of multiple TLRs to synergize with each other has been another strategy in TLR-focused cancer therapies, as it emulates the multi-TLR activation encompassed by an invading pathogen [102,103,104,105]. Mimicking the TLR recognition profile of influenza virus, the intratumoral administration of a TLR3 agonist and a TLR7 agonist has been shown to dramatically reduce tumor progression through increased granzyme B and perforin expression in CD8+ T cells, as well as an increased M1 to M2 macrophage ratio within murine lung tumors [106]. TLR synergy in a cancer context has been demonstrated in studies where the administration of either TLR agonist individually did not elicit the same robust antitumor response as did the combination [107]. Targeting the same microbial-like TLR activation profile has also been demonstrated to be effective in other tumor models. Nanoparticle complexes comprising TLR3 and TLR7 agonists poly(I:C) and imiquimod, respectively, were demonstrated to achieve complete tumor rejection in a B16 melanoma model. Immune memory from treatment with the TLR agonist complex was further demonstrated through a lack of tumor development following rechallenge with B16 melanoma [108].
Viral-like TLR activation for cancer therapeutic applications extends beyond an influenza-mimicking TLR activation profile. DNA viruses, such as herpes simplex virus and vaccinia virus, are also multi-TLR activators, through TLR2 [109] recognition of viral envelope residing in glycoproteins as well as TLR9 [110] activation from unmethylated CpG motifs found in the viral genome [111,112,113,114]. To utilize the synergistic interactions between TLR2 and TLR9, the cervical cancer tumor antigen E7 was combined with TLR2/TLR9 agonists to reverse the immunosuppressive tumor microenvironment in TC-1 tumors. By diminishing myeloid-derived suppressor cell (MDSC) and regulatory T cell populations, the tumor became permissive to antitumor CD8+ T cell infiltration and the immune-mediated suppression of tumor growth [114,115]. TLR2/TLR9 activation has also been shown to enhance DC maturation, which is essential for the effective processing and presentation of tumor antigens on MHC molecules [116,117,118,119]. Targeting DCs with lipoprotein and CpG ODN TLR agonists combined with tumor antigen decreased the production of immunosuppressive cytokine IL-10 while increasing IL-12 production from DCs. The shift in cytokine production has a dual effect on the tumor microenvironment with the decrease in IL-10 production relieving IL-10-mediated suppression of DC function, and the increase in IL-12 production driving cytotoxic T lymphocyte-activating Th1 responses [118,120].
Bacteria also possess multiple structural motifs recognized by TLRs that amplify host inflammatory responses when activated simultaneously. Therapeutic synergy of bacterial ligand sensing TLRs has been demonstrated in D2F2 tumors, following treatment with both CpG ODN and TLR5 agonist flagellin [121]. Tumors treated with CpG ODN or flagellin as a single therapy did not exhibit inhibition of tumor growth, suggesting that signaling from multiple TLRs is needed to initiate robust antitumor responses. Other combinations of TLR agonists mimicking bacterial recognition have been effective in the formation of cancer vaccines when combined with tumor antigens. Mirroring a TLR activation profile of Gram-negative bacteria [122,123], monophosphoryl6 lipid A and CpG ODN were bound to silicified murine ovarian tumor cells as a source of tumor antigen. Mice vaccinated with this combination of tumor cells and TLR agonists experienced significant tumor regression of established high-grade ovarian tumors [124]. Akin to the enhanced DC maturation following bacterial recognition by TLRs, tumor vaccine-treated DCs also experienced increased tumor antigen uptake and MHC expression, contributing to the induction of tumor-specific T cell immunity [124,125,126].
The usage of the TLR4/TLR9 activation profile in cancer vaccines has been extended to clinical trials [127,128,129,130]. AS15, an immunostimulant containing TLR4 and TLR9 agonists combined with tumor antigen MAGE-A3 and high dose IL-2, was assessed for anticancer responses in metastatic melanoma patients. The combination therapy had a disease control rate of 63%, in which 19% of patients experienced a complete response to the combination of therapy. Peripheral blood regulatory T cells from treatment responders had decreased expression in immune checkpoint proteins, such as CTLA-4 and 4-1BB, while expression was increased in non-responders [130].

8. TLR Signaling in Cancer

8.1. Effects of Tumor-Promoting TLR Signaling

TLRs have been associated with tumorigenesis, as they can activate multiple cancer-associated signaling pathways. To date, TLRs have been recognized to transduce signals through NF-κB, PI3k-Akt, and MAPK-ERK to advance cancer. NF-κB is recognized as the canonical signaling target upon TLR activation. When activated, NF-κB plays a role in several cellular functions, including cell proliferation, pro-inflammatory cytokine production, and cell survival/apoptosis, as described in Figure 2 [131]. As a result of regulating many cellular processes, the NF-κB pathway is an optimal target for aberrant, pro-tumorigenic signaling [132]. Through the LPS stimulation of TLR4, NF-κB activation enhanced the proliferation of gastric cancer cell lines BGC-823 and SGC-7901 [131,133]. In similar form, NF-κB activation via TLR4 resulted in apoptosis evasion in lung and head and neck cancer, with the addition of TNF-related apoptosis-inducing ligand (TRAIL) [134,135].
Tumorigenic TLR signaling extends beyond the canonical NF-κB pathway. Adaptor protein BCAP has been recognized as a link to TLR activation and downstream PI3k-Akt signaling [136,137]. PI3K-Akt signaling regulates cell proliferation, cell growth, and survival, thus is frequently upregulated in many cancers [138] (Figure 3). TLR7 activation was found to perpetuate pancreatic cancer progression through upregulated PI3K/Akt signaling. Consequently, an upregulation of downstream signaling targets was also observed, including antiapoptotic and pro-proliferative genes Bcl-xL and c-Myc [64,134]. Apoptosis resistance in PCI-30 cells has been shown to be mediated by TLR4-PI3K/Akt signaling [135]. Enhanced angiogenesis has also been described as a product of PI3K-Akt signaling, particularly in a cancer context. TLR4 stimulation in PANC-1 cells has increased vasculature formation and proliferation through PI3K/Akt dependent signaling [138].
Aberrant MAPK/ERK signaling has been heavily implicated in approximately a third of all human cancers [139]. Through activation of TAK1, TLR stimulation can activate the MAPK/ERK pathway to regulate growth, cell survival, and metastasis [140] (Figure 4). LPS-induced TLR4 stimulation in A549 and H1299 cells was found to promote secretion of pro-angiogenic factors VEGF and IL-8 in a p38 MAPK dependent manner [141]. MAPK/ERK signaling utilizes transcription regulation to promote tumoral immune evasion [140]. Endogenous activation of TLR2 on glioma-associated microglia was observed to downregulate MHC class II expression and impede antigen presentation in a TLR2-ERK1/2-dependent manner [142,143]. As a result, proliferation and activation of glioma targeting CD4+ T cells was hindered.

8.2. Effects of Anti-Tumor TLR Signaling

The fate of pro- or anti-tumor TLR signaling seems to be largely context-dependent, with considerations being the cancer type, the ligand activating the TLR, and the TLR itself. Tumor-rejecting TLR signaling utilizes several of the same pathways that perpetuate tumor growth, including NF-κB. TLR-mediated NF-κB signaling has been shown to induce IL-1β, IL-6, and TNF-α production in breast and bladder cancer models [26,144]. Targeting MAPK/ERK signaling has also been reported to promote anti-tumor responses. Dendritic cells stimulated with TLR3 and TLR7 agonists were found to upregulate ERK signaling, likely contributing to the enhanced dendritic cell activation and anti-tumor T-cell responses observed in CT26 tumors [145].
Apoptosis-induction in cancer cells appears to be a primary target for anti-tumor TLR signaling. Unsurprisingly, modulating NF-κB signaling has been of interest to promote cancer cell apoptosis [131]. The poly(I:C) stimulation of TLR3 on PCI-15B cells was reported to enhance apoptosis through sustained NF-κB inactivation [146]. As a result, inactive NF-κB is likely unable to activate downstream anti-apoptotic genes, such as BCL-2 [131]. In contrast, TLR3-mediated NF-κB activation was found to be required for apoptosis in poly(I:C)-treated CAMA-1 cells [22]. It appears that the activation or inactivation of NF-κB to promote apoptosis is dependent on the cancer type and additional factors that may be driving the cancer.
Beyond NF-κB, the stimulation of TLRs can activate a type I interferon (IFN) signaling pathway to promote anti-tumor responses (Figure 5). Because IFN signaling can occur to induce apoptosis in infected cells, it has been recognized as a beneficial tool for initiating apoptosis in malignant cells. Treating LNCaP cells with a TLR3 agonist was found to promote apoptosis in an IRF-3 signaling-dependent manner. Through TLR–IFN signaling, pro-apoptotic protein Noxa was subsequently upregulated downstream, a likely contributor to the increased cancer cell death [147]. Apoptosis mediated by IFN signaling extends to other cancer types. In a non-muscle invasive bladder cancer model, TLR4 activation with P-MAPA was shown to enhance IFN signaling. TLR4-mediated IFN signaling resulted in increased apoptosis and iNOS expression, an enzyme responsible for nitric oxide (NO) synthesis [144]. Increased intracellular NO concentrations have been shown to induce apoptosis [148], thus it is likely that the apoptosis of bladder cancer tumors occurs through a TLR4–IFN–iNOS axis [149].

9. Conclusions and Future Directions

Significant advances are being made in cancer biology, which includes deciphering some specifics on how TLRs play a key role in anti-cancer immunity and cancer rejection. As exogenous and endogenous TLR ligands are crucial for anti-cancer immunity, there is an urgent need to develop novel TLR-stimulating therapies. Meaningful research is needed to assuredly elucidate the roles of TLRs in modulating cancer immunotherapy and their clinical outcomes. In this regard, the majority of the currently investigated TLR agonists as anti-cancer targets are non-protein microbial components, such as lipopolysaccharides, oligonucleotides, and lipopeptides. However, a growing number of studies reveal that TLR signaling and subsequent immune responses can be activated by numerous microbial proteins and antigens.
While we still need to understand the TLR–TLR ligand pairs to produce desired oncological outcomes, there is growing evidence to suggest that TLR-modulating therapies will prove to be a safe treatment for some types of cancers. Since cancers are heterogeneous in nature, multi-treatment regimens may be useful, in some cases, in addition to known immunotherapies. TLRs are known to play a complex functional role in tumor biology and, at times, act as a double-edged sword in immunotherapy.
Recently it has become evident that TLRs do not differ from other immune receptors in their compliment to launch both host defenses and cell death. Our understanding of the signaling cascades starting from TLR activation down to cell activation has significantly progressed during the last decade. However, the molecular pathways leading to TLR-induced apoptosis are yet to be addressed.
Extensive research is required to determine the combinatorial use of TLR ligands that will prove to be smarter therapies with less toxicity and improved potency. Further studies on the roles of TLRs and their functions as anti-cancer immunity and cancer rejection will nobly advance the development of therapeutic interventions and will benefit patients undergoing immunotherapy.

Author Contributions

Writing—original draft preparation, E.F.G. and K.H.G.; Review and editing, E.F.G., M.B.M., A.Z. and K.H.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Imler, J.L.; Hoffmann, J.A. Toll receptors in Drosophila: A family of molecules regulating development and immunity. Curr. Top. Microbiol. Immunol. 2002, 270, 63–79. [Google Scholar] [CrossRef] [PubMed]
  2. Hashimoto, C.; Hudson, K.L.; Anderson, K.V. The Toll gene of Drosophila, required for dorsal-ventral embryonic polarity, appears to encode a transmembrane protein. Cell 1988, 52, 269–279. [Google Scholar] [CrossRef]
  3. Vogel, S.N. How Discovery of Toll-Mediated Innate Immunity in Drosophila Impacted Our Understanding of TLR Signaling (and Vice Versa). J. Immunol. 2012, 188, 5207–5209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Beutler, B. Toll-like receptors: How they work and what they do. Curr. Opin. Hematol. 2002, 9, 2–10. [Google Scholar] [CrossRef] [PubMed]
  5. O’Neill, L.A.; Golenbock, D.; Bowie, A.G. The history of Toll-like receptors—Redefining innate immunity. Nat. Rev. Immunol. 2013, 13, 453–460. [Google Scholar] [CrossRef]
  6. Medzhitov, R. Toll-like receptors and innate immunity. Nat. Rev. Immunol. 2001, 1, 135–145. [Google Scholar] [CrossRef]
  7. Kobe, B.; Deisenhofer, J. Proteins with leucine-rich repeats. Curr. Opin. Struct. Biol. 1995, 5, 409–416. [Google Scholar] [CrossRef]
  8. Rock, F.L.; Hardiman, G.; Timans, J.C.; Kastelein, R.A.; Bazan, J.F. A family of human receptors structurally related to Drosophila Toll. Proc. Natl. Acad. Sci. USA 1998, 95, 588–593. [Google Scholar] [CrossRef] [Green Version]
  9. El-Zayat, S.R.; Sibaii, H.; Mannaa, F.A. Toll-like receptors activation, signaling, and targeting: An overview. Bull. Natl. Res. Cent. 2019, 43, 187. [Google Scholar] [CrossRef] [Green Version]
  10. Takeda, K.; Akira, S. Toll-like receptors in innate immunity. Int. Immunol. 2005, 17, 1–14. [Google Scholar] [CrossRef]
  11. Takeda, K.; Kaisho, T.; Akira, S. Toll-like receptors. Annu. Rev. Immunol. 2003, 21, 335–376. [Google Scholar] [CrossRef] [PubMed]
  12. O’Neill, L.A.; Bowie, A.G. The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nat. Rev. Immunol. 2007, 7, 353–364. [Google Scholar] [CrossRef] [PubMed]
  13. Kaisho, T.; Takeuchi, O.; Kawai, T.; Hoshino, K.; Akira, S. Endotoxin-induced maturation of MyD88-deficient dendritic cells. J. Immunol. 2001, 166, 5688–5694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Arbibe, L.; Mira, J.P.; Teusch, N.; Kline, L.; Guha, M.; Mackman, N.; Godowski, P.J.; Ulevitch, R.J.; Knaus, U.G. Toll-like receptor 2-mediated NF-kappa B activation requires a Rac1-dependent pathway. Nat. Immunol. 2000, 1, 533–540. [Google Scholar] [CrossRef]
  15. Santoni, M.; Andrikou, K.; Sotte, V.; Bittoni, A.; Lanese, A.; Pellei, C.; Piva, F.; Conti, A.; Nabissi, M.; Santoni, G.; et al. Toll like receptors and pancreatic diseases: From a pathogenetic mechanism to a therapeutic target. Cancer Treat. Rev. 2015, 41, 569–576. [Google Scholar] [CrossRef]
  16. Medzhitov, R.; Janeway, C.A., Jr. Innate immunity: Impact on the adaptive immune response. Curr. Opin. Immunol. 1997, 9, 4–9. [Google Scholar] [CrossRef]
  17. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef]
  18. Szekely, B.; Bossuyt, V.; Li, X.; Wali, V.B.; Patwardhan, G.A.; Frederick, C.; Silber, A.; Park, T.; Harigopal, M.; Pelekanou, V.; et al. Immunological differences between primary and metastatic breast cancer. Ann. Oncol. 2018, 29, 2232–2239. [Google Scholar] [CrossRef]
  19. Mehmeti, M.; Allaoui, R.; Bergenfelz, C.; Saal, L.H.; Ethier, S.P.; Johansson, M.E.; Jirström, K.; Leandersson, K. Expression of functional toll like receptor 4 in estrogen receptor/progesterone receptor-negative breast cancer. Breast Cancer Res. 2015, 17, 130. [Google Scholar] [CrossRef] [Green Version]
  20. Wang, Y.; Liu, S.; Zhang, Y.; Yang, J. Dysregulation of TLR2 Serves as a Prognostic Biomarker in Breast Cancer and Predicts Resistance to Endocrine Therapy in the Luminal B Subtype. Front. Oncol. 2020, 10, 547. [Google Scholar] [CrossRef]
  21. González-Reyes, S.; Marín, L.; González, L.; González, L.O.; del Casar, J.M.; Lamelas, M.L.; González-Quintana, J.M.; Vizoso, F.J. Study of TLR3, TLR4 and TLR9 in breast carcinomas and their association with metastasis. BMC Cancer 2010, 10, 665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Salaun, B.; Coste, I.; Rissoan, M.C.; Lebecque, S.J.; Renno, T. TLR3 can directly trigger apoptosis in human cancer cells. J. Immunol. 2006, 176, 4894–4901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. González-Reyes, S.; Fernández, J.M.; González, L.O.; Aguirre, A.; Suárez, A.; González, J.M.; Escaff, S.; Vizoso, F.J. Study of TLR3, TLR4, and TLR9 in prostate carcinomas and their association with biochemical recurrence. Cancer Immunol. Immunother. 2011, 60, 217–226. [Google Scholar] [CrossRef] [PubMed]
  24. Haricharan, S.; Brown, P. TLR4 has a TP53-dependent dual role in regulating breast cancer cell growth. Proc. Natl. Acad. Sci. USA 2015, 112, E3216–E3225. [Google Scholar] [CrossRef] [Green Version]
  25. Shi, D.; Zhao, S.; Jiang, W.; Zhang, C.; Liang, T.; Hou, G. TLR5: A prognostic and monitoring indicator for triple-negative breast cancer. Cell Death Dis. 2019, 10, 954. [Google Scholar] [CrossRef] [Green Version]
  26. Cai, Z.; Sanchez, A.; Shi, Z.; Zhang, T.; Liu, M.; Zhang, D. Activation of Toll-like receptor 5 on breast cancer cells by flagellin suppresses cell proliferation and tumor growth. Cancer Res. 2011, 71, 2466–2475. [Google Scholar] [CrossRef] [Green Version]
  27. Jiang, W.; Han, Y.; Liang, T.; Zhang, C.; Gao, F.; Hou, G. Down-Regulation of Toll-Like Receptor 5 (TLR5) Increased VEGFR Expression in Triple Negative Breast Cancer (TNBC) Based on Radionuclide Imaging. Front. Oncol. 2021, 11, 708047. [Google Scholar] [CrossRef]
  28. Diao, Y.; Wang, X.; Wan, Y.; Zhong, J.; Gao, D.; Liu, Y.; Gao, N.; Li, W.; Liu, B.; Huang, X.; et al. Antitumor activity of a novel small molecule TLR7 agonist via immune response induction and tumor microenvironment modulation. Oncol. Rep. 2016, 35, 793–800. [Google Scholar] [CrossRef] [Green Version]
  29. Adams, S.F.; Grimm, A.J.; Chiang, C.L.; Mookerjee, A.; Flies, D.; Jean, S.; McCann, G.A.; Michaux, J.; Pak, H.; Huber, F.; et al. Rapid tumor vaccine using Toll-like receptor-activated ovarian cancer ascites monocytes. J. Immunother. Cancer 2020, 8, e000875. [Google Scholar] [CrossRef]
  30. Tuomela, J.; Sandholm, J.; Karihtala, P.; Ilvesaro, J.; Vuopala, K.S.; Kauppila, J.H.; Kauppila, S.; Chen, D.; Pressey, C.; Härkönen, P.; et al. Low TLR9 expression defines an aggressive subtype of triple-negative breast cancer. Breast Cancer Res. Treat. 2012, 135, 481–493. [Google Scholar] [CrossRef]
  31. Droemann, D.; Albrecht, D.; Gerdes, J.; Ulmer, A.J.; Branscheid, D.; Vollmer, E.; Dalhoff, K.; Zabel, P.; Goldmann, T. Human lung cancer cells express functionally active Toll-like receptor 9. Respir. Res. 2005, 6, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Chatterjee, S.; Crozet, L.; Damotte, D.; Iribarren, K.; Schramm, C.; Alifano, M.; Lupo, A.; Cherfils-Vicini, J.; Goc, J.; Katsahian, S.; et al. TLR7 promotes tumor progression, chemotherapy resistance, and poor clinical outcomes in non-small cell lung cancer. Cancer Res. 2014, 74, 5008–5018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bianchi, F.; Milione, M.; Casalini, P.; Centonze, G.; Le Noci, V.M.; Storti, C.; Alexiadis, S.; Truini, M.; Sozzi, G.; Pastorino, U.; et al. Toll-like receptor 3 as a new marker to detect high risk early stage Non-Small-Cell Lung Cancer patients. Sci. Rep. 2019, 9, 14288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Deng, Y.; Yang, J.; Qian, J.; Liu, R.; Huang, E.; Wang, Y.; Luo, F.; Chu, Y. TLR1/TLR2 signaling blocks the suppression of monocytic myeloid-derived suppressor cell by promoting its differentiation into M1-type macrophage. Mol. Immunol. 2019, 112, 266–273. [Google Scholar] [CrossRef] [PubMed]
  35. Liu, Y.; Gu, Y.; Han, Y.; Zhang, Q.; Jiang, Z.; Zhang, X.; Huang, B.; Xu, X.; Zheng, J.; Cao, X. Tumor Exosomal RNAs Promote Lung Pre-metastatic Niche Formation by Activating Alveolar Epithelial TLR3 to Recruit Neutrophils. Cancer Cell 2016, 30, 243–256. [Google Scholar] [CrossRef] [Green Version]
  36. Zhou, H.; Chen, J.H.; Hu, J.; Luo, Y.Z.; Li, F.; Xiao, L.; Zhong, M.Z. High expression of Toll-like receptor 5 correlates with better prognosis in non-small-cell lung cancer: An anti-tumor effect of TLR5 signaling in non-small cell lung cancer. J. Cancer Res. Clin. Oncol. 2014, 140, 633–643. [Google Scholar] [CrossRef]
  37. Bianchi, F.; Alexiadis, S.; Camisaschi, C.; Truini, M.; Centonze, G.; Milione, M.; Balsari, A.; Tagliabue, E.; Sfondrini, L. TLR3 Expression Induces Apoptosis in Human Non-Small-Cell Lung Cancer. Int. J. Mol. Sci. 2020, 21, 1440. [Google Scholar] [CrossRef] [Green Version]
  38. Vinod, N.; Hwang, D.; Azam, S.H.; Van Swearingen, A.E.D.; Wayne, E.; Fussell, S.C.; Sokolsky-Papkov, M.; Pecot, C.V.; Kabanov, A.V. High-capacity poly(2-oxazoline) formulation of TLR 7/8 agonist extends survival in a chemo-insensitive, metastatic model of lung adenocarcinoma. Sci. Adv. 2020, 6, eaba5542. [Google Scholar] [CrossRef]
  39. Cherfils-Vicini, J.; Platonova, S.; Gillard, M.; Laurans, L.; Validire, P.; Caliandro, R.; Magdeleinat, P.; Mami-Chouaib, F.; Dieu-Nosjean, M.C.; Fridman, W.H.; et al. Triggering of TLR7 and TLR8 expressed by human lung cancer cells induces cell survival and chemoresistance. J. Clin. Investig. 2010, 120, 1285–1297. [Google Scholar] [CrossRef]
  40. Wang, K.; Wang, J.; Wei, F.; Zhao, N.; Yang, F.; Ren, X. Expression of TLR4 in Non-Small Cell Lung Cancer Is Associated with PD-L1 and Poor Prognosis in Patients Receiving Pulmonectomy. Front. Immunol. 2017, 8, 456. [Google Scholar] [CrossRef] [Green Version]
  41. Waldmannová, E.; Caisová, V.; Fáberová, J.; Sváčková, P.; Kovářová, M.; Sváčková, D.; Kumžáková, Z.; Jačková, A.; Vácová, N.; Nedbalová, P.; et al. The use of Zymosan A and bacteria anchored to tumor cells for effective cancer immunotherapy: B16-F10 murine melanoma model. Int. Immunopharmacol. 2016, 39, 295–306. [Google Scholar] [CrossRef] [PubMed]
  42. Goto, Y.; Arigami, T.; Kitago, M.; Nguyen, S.L.; Narita, N.; Ferrone, S.; Morton, D.L.; Irie, R.F.; Hoon, D.S. Activation of Toll-like receptors 2, 3, and 4 on human melanoma cells induces inflammatory factors. Mol. Cancer Ther. 2008, 7, 3642–3653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Saint-Jean, M.; Knol, A.C.; Nguyen, J.M.; Khammari, A.; Dréno, B. TLR expression in human melanoma cells. Eur. J. Dermatol. 2011, 21, 899–905. [Google Scholar] [CrossRef] [PubMed]
  44. Eiró, N.; Ovies, C.; Fernandez-Garcia, B.; Álvarez-Cuesta, C.C.; González, L.; González, L.O.; Vizoso, F.J. Expression of TLR3, 4, 7 and 9 in cutaneous malignant melanoma: Relationship with clinicopathological characteristics and prognosis. Arch. Dermatol. Res. 2013, 305, 59–67. [Google Scholar] [CrossRef] [PubMed]
  45. Fu, X.Q.; Liu, B.; Wang, Y.P.; Li, J.K.; Zhu, P.L.; Li, T.; Tse, K.W.; Chou, J.Y.; Yin, C.L.; Bai, J.X.; et al. Activation of STAT3 is a key event in TLR4 signaling-mediated melanoma progression. Cell Death Dis. 2020, 11, 246. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, M.; Yan, Z.; Wang, J.; Yao, X. Toll-like receptors 7 and 8 expression correlates with the expression of immune biomarkers and positively predicts the clinical outcome of patients with melanoma. OncoTargets Ther. 2017, 10, 4339–4346. [Google Scholar] [CrossRef] [Green Version]
  47. Aspord, C.; Tramcourt, L.; Leloup, C.; Molens, J.P.; Leccia, M.T.; Charles, J.; Plumas, J. Imiquimod inhibits melanoma development by promoting pDC cytotoxic functions and impeding tumor vascularization. J. Investig. Dermatol. 2014, 134, 2551–2561. [Google Scholar] [CrossRef] [Green Version]
  48. Kapp, K.; Volz, B.; Curran, M.A.; Oswald, D.; Wittig, B.; Schmidt, M. EnanDIM—A novel family of L-nucleotide-protected TLR9 agonists for cancer immunotherapy. J. Immunother. Cancer 2019, 7, 5. [Google Scholar] [CrossRef] [Green Version]
  49. Nihon-Yanagi, Y.; Terai, K.; Murano, T.; Matsumoto, T.; Okazumi, S. Tissue expression of Toll-like receptors 2 and 4 in sporadic human colorectal cancer. Cancer Immunol. Immunother. 2012, 61, 71–77. [Google Scholar] [CrossRef] [Green Version]
  50. Beilmann-Lehtonen, I.; Hagström, J.; Kaprio, T.; Stenman, U.H.; Strigård, K.; Palmqvist, R.; Gunnarsson, U.; Böckelman, C.; Haglund, C. The Relationship between the Tissue Expression of TLR2, TLR4, TLR5, and TLR7 and Systemic Inflammatory Responses in Colorectal Cancer Patients. Oncology 2021, 99, 790–801. [Google Scholar] [CrossRef]
  51. Lu, C.C.; Kuo, H.C.; Wang, F.S.; Jou, M.H.; Lee, K.C.; Chuang, J.H. Upregulation of TLRs and IL-6 as a marker in human colorectal cancer. Int. J. Mol. Sci. 2014, 16, 159–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Yoshida, T.; Miura, T.; Matsumiya, T.; Yoshida, H.; Morohashi, H.; Sakamoto, Y.; Kurose, A.; Imaizumi, T.; Hakamada, K. Toll-Like Receptor 3 as a Recurrence Risk Factor and a Potential Molecular Therapeutic Target in Colorectal Cancer. Clin. Exp. Gastroenterol. 2020, 13, 427–438. [Google Scholar] [CrossRef] [PubMed]
  53. Bugge, M.; Bergstrom, B.; Eide, O.K.; Solli, H.; Kjønstad, I.F.; Stenvik, J.; Espevik, T.; Nilsen, N.J. Surface Toll-like receptor 3 expression in metastatic intestinal epithelial cells induces inflammatory cytokine production and promotes invasiveness. J. Biol. Chem. 2017, 292, 15408–15425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Cammarota, R.; Bertolini, V.; Pennesi, G.; Bucci, E.O.; Gottardi, O.; Garlanda, C.; Laghi, L.; Barberis, M.C.; Sessa, F.; Noonan, D.M.; et al. The tumor microenvironment of colorectal cancer: Stromal TLR-4 expression as a potential prognostic marker. J. Transl. Med. 2010, 8, 112. [Google Scholar] [CrossRef] [Green Version]
  55. Rhee, S.H.; Im, E.; Pothoulakis, C. Toll-like receptor 5 engagement modulates tumor development and growth in a mouse xenograft model of human colon cancer. Gastroenterology 2008, 135, 518–528. [Google Scholar] [CrossRef] [Green Version]
  56. Liu, Z.; Xie, Y.; Xiong, Y.; Liu, S.; Qiu, C.; Zhu, Z.; Mao, H.; Yu, M.; Wang, X. TLR 7/8 agonist reverses oxaliplatin resistance in colorectal cancer via directing the myeloid-derived suppressor cells to tumoricidal M1-macrophages. Cancer Lett. 2020, 469, 173–185. [Google Scholar] [CrossRef]
  57. Gao, C.; Qiao, T.; Zhang, B.; Yuan, S.; Zhuang, X.; Luo, Y. TLR9 signaling activation at different stages in colorectal cancer and NF-kappaB expression. OncoTargets Ther. 2018, 11, 5963–5971. [Google Scholar] [CrossRef] [Green Version]
  58. Lundy, J.; Gearing, L.J.; Gao, H.; West, A.C.; McLeod, L.; Deswaerte, V.; Yu, L.; Porazinski, S.; Pajic, M.; Hertzog, P.J.; et al. TLR2 activation promotes tumour growth and associates with patient survival and chemotherapy response in pancreatic ductal adenocarcinoma. Oncogene 2021, 40, 6007–6022. [Google Scholar] [CrossRef]
  59. Grimmig, T.; Moench, R.; Kreckel, J.; Haack, S.; Rueckert, F.; Rehder, R.; Tripathi, S.; Ribas, C.; Chandraker, A.; Germer, C.T.; et al. Toll Like Receptor 2, 4, and 9 Signaling Promotes Autoregulative Tumor Cell Growth and VEGF/PDGF Expression in Human Pancreatic Cancer. Int. J. Mol. Sci. 2016, 17, 2060. [Google Scholar] [CrossRef] [Green Version]
  60. Schwartz, A.L.; Malgor, R.; Dickerson, E.; Weeraratna, A.T.; Slominski, A.; Wortsman, J.; Harii, N.; Kohn, A.D.; Moon, R.T.; Schwartz, F.L.; et al. Phenylmethimazole decreases Toll-like receptor 3 and noncanonical Wnt5a expression in pancreatic cancer and melanoma together with tumor cell growth and migration. Clin. Cancer Res. 2009, 15, 4114–4122. [Google Scholar] [CrossRef] [Green Version]
  61. Shojaei, H.; Oberg, H.H.; Juricke, M.; Marischen, L.; Kunz, M.; Mundhenke, C.; Gieseler, F.; Kabelitz, D.; Wesch, D. Toll-like receptors 3 and 7 agonists enhance tumor cell lysis by human gammadelta T cells. Cancer Res. 2009, 69, 8710–8717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Ochi, A.; Nguyen, A.H.; Bedrosian, A.S.; Mushlin, H.M.; Zarbakhsh, S.; Barilla, R.; Zambirinis, C.P.; Fallon, N.C.; Rehman, A.; Pylayeva-Gupta, Y.; et al. MyD88 inhibition amplifies dendritic cell capacity to promote pancreatic carcinogenesis via Th2 cells. J. Exp. Med. 2012, 209, 1671–1687. [Google Scholar] [CrossRef] [PubMed]
  63. Grimmig, T.; Matthes, N.; Hoeland, K.; Tripathi, S.; Chandraker, A.; Grimm, M.; Moench, R.; Moll, E.M.; Friess, H.; Tsaur, I.; et al. TLR7 and TLR8 expression increases tumor cell proliferation and promotes chemoresistance in human pancreatic cancer. Int. J. Oncol. 2015, 47, 857–866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Ochi, A.; Graffeo, C.S.; Zambirinis, C.P.; Rehman, A.; Hackman, M.; Fallon, N.; Barilla, R.M.; Henning, J.R.; Jamal, M.; Rao, R.; et al. Toll-like receptor 7 regulates pancreatic carcinogenesis in mice and humans. J. Clin. Investig. 2012, 122, 4118–4129. [Google Scholar] [CrossRef]
  65. Zambirinis, C.P.; Levie, E.; Nguy, S.; Avanzi, A.; Barilla, R.; Xu, Y.; Seifert, L.; Daley, D.; Greco, S.H.; Deutsch, M.; et al. TLR9 ligation in pancreatic stellate cells promotes tumorigenesis. J. Exp. Med. 2015, 212, 2077–2094. [Google Scholar] [CrossRef]
  66. Volk-Draper, L.; Hall, K.; Griggs, C.; Rajput, S.; Kohio, P.; DeNardo, D.; Ran, S. Paclitaxel therapy promotes breast cancer metastasis in a TLR4-dependent manner. Cancer Res. 2014, 74, 5421–5434. [Google Scholar] [CrossRef] [Green Version]
  67. Zhou, M.; McFarland-Mancini, M.M.; Funk, H.M.; Husseinzadeh, N.; Mounajjed, T.; Drew, A.F. Toll-like receptor expression in normal ovary and ovarian tumors. Cancer Immunol. Immunother. 2009, 58, 1375–1385. [Google Scholar] [CrossRef] [PubMed]
  68. Kelly, M.G.; Alvero, A.B.; Chen, R.; Silasi, D.A.; Abrahams, V.M.; Chan, S.; Visintin, I.; Rutherford, T.; Mor, G. TLR-4 signaling promotes tumor growth and paclitaxel chemoresistance in ovarian cancer. Cancer Res. 2006, 66, 3859–3868. [Google Scholar] [CrossRef] [Green Version]
  69. Szajnik, M.; Szczepanski, M.J.; Czystowska, M.; Elishaev, E.; Mandapathil, M.; Nowak-Markwitz, E.; Spaczynski, M.; Whiteside, T.L. TLR4 signaling induced by lipopolysaccharide or paclitaxel regulates tumor survival and chemoresistance in ovarian cancer. Oncogene 2009, 28, 4353–4363. [Google Scholar] [CrossRef] [Green Version]
  70. Wang, A.C.; Su, Q.B.; Wu, F.X.; Zhang, X.L.; Liu, P.S. Role of TLR4 for paclitaxel chemotherapy in human epithelial ovarian cancer cells. Eur. J. Clin. Investig. 2009, 39, 157–164. [Google Scholar] [CrossRef]
  71. Block, M.S.; Vierkant, R.A.; Rambau, P.F.; Winham, S.J.; Wagner, P.; Traficante, N.; Tołoczko, A.; Tiezzi, D.G.; Taran, F.A.; Sinn, P.; et al. MyD88 and TLR4 Expression in Epithelial Ovarian Cancer. Mayo Clin. Proc. 2018, 93, 307–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Park, G.B.; Kim, D. TLR5/7-mediated PI3K activation triggers epithelial-mesenchymal transition of ovarian cancer cells through WAVE3-dependent mesothelin or OCT4/SOX2 expression. Oncol. Rep. 2017, 38, 3167–3176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Rutkowski, M.R.; Stephen, T.L.; Svoronos, N.; Allegrezza, M.J.; Tesone, A.J.; Perales-Puchalt, A.; Brencicova, E.; Escovar-Fadul, X.; Nguyen, J.M.; Cadungog, M.G.; et al. Microbially driven TLR5-dependent signaling governs distal malignant progression through tumor-promoting inflammation. Cancer Cell 2015, 27, 27–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Xu, G.; Feng, D.; Yao, Y.; Li, P.; Sun, H.; Yang, H.; Li, C.; Jiang, R.; Sun, B.; Chen, Y. Listeria-based hepatocellular carcinoma vaccine facilitates anti-PD-1 therapy by regulating macrophage polarization. Oncogene 2020, 39, 1429–1444. [Google Scholar] [CrossRef]
  75. Berger, R.; Fiegl, H.; Goebel, G.; Obexer, P.; Ausserlechner, M.; Doppler, W.; Hauser-Kronberger, C.; Reitsamer, R.; Egle, D.; Reimer, D.; et al. Toll-like receptor 9 expression in breast and ovarian cancer is associated with poorly differentiated tumors. Cancer Sci. 2010, 101, 1059–1066. [Google Scholar] [CrossRef] [Green Version]
  76. Galli, R.; Starace, D.; Busà, R.; Angelini, D.F.; Paone, A.; De Cesaris, P.; Filippini, A.; Sette, C.; Battistini, L.; Ziparo, E.; et al. TLR stimulation of prostate tumor cells induces chemokine-mediated recruitment of specific immune cell types. J. Immunol. 2010, 184, 6658–6669. [Google Scholar] [CrossRef]
  77. Rezania, S.; Amirmozaffari, N.; Rashidi, N.; Mirzadegan, E.; Zarei, S.; Ghasemi, J.; Zarei, O.; Katouzian, L.; Zarnani, A.H. The same and not the same: Heterogeneous functional activation of prostate tumor cells by TLR ligation. Cancer Cell Int. 2014, 14, 54. [Google Scholar] [CrossRef] [Green Version]
  78. Paone, A.; Starace, D.; Galli, R.; Padula, F.; De Cesaris, P.; Filippini, A.; Ziparo, E.; Riccioli, A. Toll-like receptor 3 triggers apoptosis of human prostate cancer cells through a PKC-alpha-dependent mechanism. Carcinogenesis 2008, 29, 1334–1342. [Google Scholar] [CrossRef] [Green Version]
  79. Hua, D.; Liu, M.Y.; Cheng, Z.D.; Qin, X.J.; Zhang, H.M.; Chen, Y.; Qin, G.J.; Liang, G.; Li, J.N.; Han, X.F.; et al. Small interfering RNA-directed targeting of Toll-like receptor 4 inhibits human prostate cancer cell invasion, survival, and tumorigenicity. Mol. Immunol. 2009, 46, 2876–2884. [Google Scholar] [CrossRef]
  80. Väisänen, M.R.; Väisänen, T.; Jukkola-Vuorinen, A.; Vuopala, K.S.; Desmond, R.; Selander, K.S.; Vaarala, M.H. Expression of toll-like receptor-9 is increased in poorly differentiated prostate tumors. Prostate 2010, 70, 817–824. [Google Scholar] [CrossRef]
  81. Coley, W.B. The Treatment of Inoperable Sarcoma by Bacterial Toxins (the Mixed Toxins of the Streptococcus erysipelas and the Bacillus prodigiosus). Proc. R. Soc. Med. 1910, 3, 1–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Coley, W.B. The treatment of malignant tumors by repeated inoculations of erysipelas. With a report of ten original cases. Am. J. Med. Sci. 1893, 105, 487. [Google Scholar] [CrossRef]
  83. Gupta, K.H.; Nowicki, C.; Giurini, E.F.; Marzo, A.L.; Zloza, A. Bacterial-Based Cancer Therapy (BBCT): Recent Advances, Current Challenges, and Future Prospects for Cancer Immunotherapy. Vaccines 2021, 9, 1497. [Google Scholar] [CrossRef] [PubMed]
  84. Orange, M.; Reuter, U.; Hobohm, U. Coley’s Lessons Remembered: Augmenting Mistletoe Therapy. Integr. Cancer Ther. 2016, 15, 502–511. [Google Scholar] [CrossRef] [Green Version]
  85. Hobohm, U.; Stanford, J.L.; Grange, J.M. Pathogen-associated molecular pattern in cancer immunotherapy. Crit. Rev. Immunol. 2008, 28, 95–107. [Google Scholar] [CrossRef]
  86. Janeway, C.A., Jr.; Medzhitov, R. Innate immune recognition. Annu. Rev. Immunol. 2002, 20, 197–216. [Google Scholar] [CrossRef] [Green Version]
  87. Bendelac, A.; Medzhitov, R. Adjuvants of immunity: Harnessing innate immunity to promote adaptive immunity. J. Exp. Med. 2002, 195, F19–F23. [Google Scholar] [CrossRef]
  88. Roberts, N.J.; Zhang, L.; Janku, F.; Collins, A.; Bai, R.Y.; Staedtke, V.; Rusk, A.W.; Tung, D.; Miller, M.; Roix, J.; et al. Intratumoral injection of Clostridium novyi-NT spores induces antitumor responses. Sci. Transl. Med. 2014, 6, 249ra111. [Google Scholar] [CrossRef] [Green Version]
  89. Agrawal, N.; Bettegowda, C.; Cheong, I.; Geschwind, J.F.; Drake, C.G.; Hipkiss, E.L.; Tatsumi, M.; Dang, L.H.; Diaz, L.A., Jr.; Pomper, M.; et al. Bacteriolytic therapy can generate a potent immune response against experimental tumors. Proc. Natl. Acad. Sci. USA 2004, 101, 15172–15177. [Google Scholar] [CrossRef] [Green Version]
  90. Macedo, N.; Miller, D.M.; Haq, R.; Kaufman, H.L. Clinical landscape of oncolytic virus research in 2020. J. Immunother. Cancer 2020, 8, e001486. [Google Scholar] [CrossRef]
  91. Martínez-Vélez, N.; Garcia-Moure, M.; Marigil, M.; González-Huarriz, M.; Puigdelloses, M.; Gallego Pérez-Larraya, J.; Zalacaín, M.; Marrodán, L.; Varela-Guruceaga, M.; Laspidea, V.; et al. The oncolytic virus Delta-24-RGD elicits an antitumor effect in pediatric glioma and DIPG mouse models. Nat. Commun. 2019, 10, 2235. [Google Scholar] [CrossRef] [PubMed]
  92. Krieg, A.M. Toll-like receptor 9 (TLR9) agonists in the treatment of cancer. Oncogene 2008, 27, 161–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Rahman, A.H.; Taylor, D.K.; Turka, L.A. The contribution of direct TLR signaling to T cell responses. Immunol. Res. 2009, 45, 25–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Kabelitz, D. Expression and function of Toll-like receptors in T lymphocytes. Curr. Opin. Immunol. 2007, 19, 39–45. [Google Scholar] [CrossRef]
  95. Cerullo, V.; Diaconu, I.; Romano, V.; Hirvinen, M.; Ugolini, M.; Escutenaire, S.; Holm, S.L.; Kipar, A.; Kanerva, A.; Hemminki, A. An oncolytic adenovirus enhanced for toll-like receptor 9 stimulation increases antitumor immune responses and tumor clearance. Mol. Ther. 2012, 20, 2076–2086. [Google Scholar] [CrossRef] [Green Version]
  96. Noh, J.Y.; Yoon, S.R.; Kim, T.D.; Choi, I.; Jung, H. Toll-Like Receptors in Natural Killer Cells and Their Application for Immunotherapy. J. Immunol. Res. 2020, 2020, 2045860. [Google Scholar] [CrossRef]
  97. Adib-Conquy, M.; Scott-Algara, D.; Cavaillon, J.M.; Souza-Fonseca-Guimaraes, F. TLR-mediated activation of NK cells and their role in bacterial/viral immune responses in mammals. Immunol. Cell Biol. 2014, 92, 256–262. [Google Scholar] [CrossRef]
  98. Newman, J.H.; Chesson, C.B.; Herzog, N.L.; Bommareddy, P.K.; Aspromonte, S.M.; Pepe, R.; Estupinian, R.; Aboelatta, M.M.; Buddhadev, S.; Tarabichi, S.; et al. Intratumoral injection of the seasonal flu shot converts immunologically cold tumors to hot and serves as an immunotherapy for cancer. Proc. Natl. Acad. Sci. USA 2020, 117, 1119–1128. [Google Scholar] [CrossRef] [Green Version]
  99. Datta, S.K.; Redecke, V.; Prilliman, K.R.; Takabayashi, K.; Corr, M.; Tallant, T.; DiDonato, J.; Dziarski, R.; Akira, S.; Schoenberger, S.P.; et al. A subset of Toll-like receptor ligands induces cross-presentation by bone marrow-derived dendritic cells. J. Immunol. 2003, 170, 4102–4110. [Google Scholar] [CrossRef] [Green Version]
  100. Crespo, M.I.; Zacca, E.R.; Núñez, N.G.; Ranocchia, R.P.; Maccioni, M.; Maletto, B.A.; Pistoresi-Palencia, M.C.; Morón, G. TLR7 triggering with polyuridylic acid promotes cross-presentation in CD8α+ conventional dendritic cells by enhancing antigen preservation and MHC class I antigen permanence on the dendritic cell surface. J. Immunol. 2013, 190, 948–960. [Google Scholar] [CrossRef] [Green Version]
  101. Oh, J.Z.; Kurche, J.S.; Burchill, M.A.; Kedl, R.M. TLR7 enables cross-presentation by multiple dendritic cell subsets through a type I IFN-dependent pathway. Blood 2011, 118, 3028–3038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Lebel, M.; Chartrand, K.; Tarrab, E.; Savard, P.; Leclerc, D.; Lamarre, A. Potentiating Cancer Immunotherapy Using Papaya Mosaic Virus-Derived Nanoparticles. Nano Lett. 2016, 16, 1826–1832. [Google Scholar] [CrossRef] [PubMed]
  103. Cai, H.; Shukla, S.; Wang, C.; Masarapu, H.; Steinmetz, N.F. Heterologous Prime-Boost Enhances the Antitumor Immune Response Elicited by Plant-Virus-Based Cancer Vaccine. J. Am. Chem. Soc. 2019, 141, 6509–6518. [Google Scholar] [CrossRef] [PubMed]
  104. Kines, R.C.; Thompson, C.D.; Spring, S.; Li, Z.; de Los Pinos, E.; Monks, S.; Schiller, J.T. Virus-Like Particle-Drug Conjugates Induce Protective, Long-lasting Adaptive Antitumor Immunity in the Absence of Specifically Targeted Tumor Antigens. Cancer Immunol. Res. 2021, 9, 693–706. [Google Scholar] [CrossRef]
  105. Shukla, S.; Wang, C.; Beiss, V.; Steinmetz, N.F. Antibody Response against Cowpea Mosaic Viral Nanoparticles Improves In Situ Vaccine Efficacy in Ovarian Cancer. ACS Nano 2020, 14, 2994–3003. [Google Scholar] [CrossRef]
  106. De Nardo, D.; De Nardo, C.M.; Nguyen, T.; Hamilton, J.A.; Scholz, G.M. Signaling crosstalk during sequential TLR4 and TLR9 activation amplifies the inflammatory response of mouse macrophages. J. Immunol. 2009, 183, 8110–8118. [Google Scholar] [CrossRef]
  107. Sørensen, L.N.; Reinert, L.S.; Malmgaard, L.; Bartholdy, C.; Thomsen, A.R.; Paludan, S.R. TLR2 and TLR9 synergistically control herpes simplex virus infection in the brain. J. Immunol. 2008, 181, 8604–8612. [Google Scholar] [CrossRef]
  108. Sieben, M.; Schäfer, P.; Dinsart, C.; Galle, P.R.; Moehler, M. Activation of the human immune system via toll-like receptors by the oncolytic parvovirus H-1. Int. J. Cancer 2013, 132, 2548–2556. [Google Scholar] [CrossRef]
  109. Zhu, J.; Martinez, J.; Huang, X.; Yang, Y. Innate immunity against vaccinia virus is mediated by TLR2 and requires TLR-independent production of IFN-beta. Blood 2007, 109, 619–625. [Google Scholar] [CrossRef] [Green Version]
  110. Samuelsson, C.; Hausmann, J.; Lauterbach, H.; Schmidt, M.; Akira, S.; Wagner, H.; Chaplin, P.; Suter, M.; O’Keeffe, M.; Hochrein, H. Survival of lethal poxvirus infection in mice depends on TLR9, and therapeutic vaccination provides protection. J. Clin. Investig. 2008, 118, 1776–1784. [Google Scholar] [CrossRef]
  111. Whitmore, M.M.; DeVeer, M.J.; Edling, A.; Oates, R.K.; Simons, B.; Lindner, D.; Williams, B.R. Synergistic activation of innate immunity by double-stranded RNA and CpG DNA promotes enhanced antitumor activity. Cancer Res. 2004, 64, 5850–5860. [Google Scholar] [CrossRef] [Green Version]
  112. Madan-Lala, R.; Pradhan, P.; Roy, K. Combinatorial Delivery of Dual and Triple TLR Agonists via Polymeric Pathogen-like Particles Synergistically Enhances Innate and Adaptive Immune Responses. Sci. Rep. 2017, 7, 2530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Iwasaki, A.; Medzhitov, R. Toll-like receptor control of the adaptive immune responses. Nat. Immunol. 2004, 5, 987–995. [Google Scholar] [CrossRef] [PubMed]
  114. Anfray, C.; Mainini, F.; Digifico, E.; Maeda, A.; Sironi, M.; Erreni, M.; Anselmo, A.; Ummarino, A.; Gandoy, S.; Expósito, F.; et al. Intratumoral combination therapy with poly(I:C) and resiquimod synergistically triggers tumor-associated macrophages for effective systemic antitumoral immunity. J. Immunother. Cancer 2021, 9, e002408. [Google Scholar] [CrossRef] [PubMed]
  115. Chang, L.-S.; Leng, C.-H.; Yeh, Y.-C.; Wu, C.-C.; Chen, H.-W.; Huang, H.-M.; Liu, S.-J. Toll-like receptor 9 agonist enhances anti-tumor immunity and inhibits tumor-associated immunosuppressive cells numbers in a mouse cervical cancer model following recombinant lipoprotein therapy. Mol. Cancer 2014, 13, 60. [Google Scholar] [CrossRef] [Green Version]
  116. Bocanegra Gondan, A.I.; Ruiz-de-Angulo, A.; Zabaleta, A.; Gómez Blanco, N.; Cobaleda-Siles, B.M.; García-Granda, M.J.; Padro, D.; Llop, J.; Arnaiz, B.; Gato, M.; et al. Effective cancer immunotherapy in mice by polyIC-imiquimod complexes and engineered magnetic nanoparticles. Biomaterials 2018, 170, 95–115. [Google Scholar] [CrossRef] [Green Version]
  117. Ma, Y.; He, B. Recognition of herpes simplex viruses: Toll-like receptors and beyond. J. Mol. Biol. 2014, 426, 1133–1147. [Google Scholar] [CrossRef] [Green Version]
  118. Uyangaa, E.; Choi, J.Y.; Patil, A.M.; Hossain, F.M.A.; Park, S.O.; Kim, B.; Kim, K.; Eo, S.K. Dual TLR2/9 Recognition of Herpes Simplex Virus Infection Is Required for Recruitment and Activation of Monocytes and NK Cells and Restriction of Viral Dissemination to the Central Nervous System. Front. Immunol. 2018, 9, 905. [Google Scholar] [CrossRef]
  119. Bafica, A.; Scanga, C.A.; Feng, C.G.; Leifer, C.; Cheever, A.; Sher, A. TLR9 regulates Th1 responses and cooperates with TLR2 in mediating optimal resistance to Mycobacterium tuberculosis. J. Exp. Med. 2005, 202, 1715–1724. [Google Scholar] [CrossRef]
  120. Kumar, P.; John, V.; Marathe, S.; Das, G.; Bhaskar, S. Mycobacterium indicus pranii induces dendritic cell activation, survival, and Th1/Th17 polarization potential in a TLR-dependent manner. J. Leukoc. Biol. 2015, 97, 511–520. [Google Scholar] [CrossRef] [Green Version]
  121. Sfondrini, L.; Rossini, A.; Besusso, D.; Merlo, A.; Tagliabue, E.; Mènard, S.; Balsari, A. Antitumor activity of the TLR-5 ligand flagellin in mouse models of cancer. J. Immunol. 2006, 176, 6624–6630. [Google Scholar] [CrossRef] [PubMed]
  122. Kawai, T.; Akira, S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 2011, 34, 637–650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Bhan, U.; Ballinger, M.N.; Zeng, X.; Newstead, M.J.; Cornicelli, M.D.; Standiford, T.J. Cooperative interactions between TLR4 and TLR9 regulate interleukin 23 and 17 production in a murine model of gram negative bacterial pneumonia. PLoS ONE 2010, 5, e9896. [Google Scholar] [CrossRef] [PubMed]
  124. Guo, J.; De May, H.; Franco, S.; Noureddine, A.; Tang, L.; Brinker, C.J.; Kusewitt, D.F.; Adams, S.F.; Serda, R.E. Cancer vaccines from cryogenically silicified tumour cells functionalized with pathogen-associated molecular patterns. Nat. Biomed. Eng. 2022, 6, 19–31. [Google Scholar] [CrossRef] [PubMed]
  125. Oshikawa, T.; Okamoto, M.; Tano, T.; Sasai, A.; Kan, S.; Moriya, Y.; Ryoma, Y.; Saito, M.; Akira, S.; Sato, M. Antitumor effect of OK-432-derived DNA: One of the active constituents of OK-432, a streptococcal immunotherapeutic agent. J. Immunother. 2006, 29, 143–150. [Google Scholar] [CrossRef]
  126. Koya, T.; Yanagisawa, R.; Higuchi, Y.; Sano, K.; Shimodaira, S. Interferon-α-inducible Dendritic Cells Matured with OK-432 Exhibit TRAIL and Fas Ligand Pathway-mediated Killer Activity. Sci. Rep. 2017, 7, 42145. [Google Scholar] [CrossRef] [Green Version]
  127. Cohen, A.D.; Lendvai, N.; Nataraj, S.; Imai, N.; Jungbluth, A.A.; Tsakos, I.; Rahman, A.; Mei, A.H.; Singh, H.; Zarychta, K.; et al. Autologous Lymphocyte Infusion Supports Tumor Antigen Vaccine-Induced Immunity in Autologous Stem Cell Transplant for Multiple Myeloma. Cancer Immunol. Res. 2019, 7, 658–669. [Google Scholar] [CrossRef]
  128. Hamilton, E.; Blackwell, K.; Hobeika, A.C.; Clay, T.M.; Broadwater, G.; Ren, X.R.; Chen, W.; Castro, H.; Lehmann, F.; Spector, N.; et al. Phase 1 clinical trial of HER2-specific immunotherapy with concomitant HER2 kinase inhibition [corrected]. J. Transl. Med. 2012, 10, 28. [Google Scholar] [CrossRef] [Green Version]
  129. Limentani, S.A.; Campone, M.; Dorval, T.; Curigliano, G.; de Boer, R.; Vogel, C.; White, S.; Bachelot, T.; Canon, J.L.; Disis, M.; et al. A non-randomized dose-escalation Phase I trial of a protein-based immunotherapeutic for the treatment of breast cancer patients with HER2-overexpressing tumors. Breast Cancer Res. Treat. 2016, 156, 319–330. [Google Scholar] [CrossRef]
  130. McQuade, J.L.; Homsi, J.; Torres-Cabala, C.A.; Bassett, R.; Popuri, R.M.; James, M.L.; Vence, L.M.; Hwu, W.J. A phase II trial of recombinant MAGE-A3 protein with immunostimulant AS15 in combination with high-dose Interleukin-2 (HDIL2) induction therapy in metastatic melanoma. BMC Cancer 2018, 18, 1274. [Google Scholar] [CrossRef] [Green Version]
  131. Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Park, M.H.; Hong, J.T. Roles of NF-κB in Cancer and Inflammatory Diseases and Their Therapeutic Approaches. Cells 2016, 5, 15. [Google Scholar] [CrossRef] [PubMed]
  133. Yuan, X.; Zhou, Y.; Wang, W.; Li, J.; Xie, G.; Zhao, Y.; Xu, D.; Shen, L. Activation of TLR4 signaling promotes gastric cancer progression by inducing mitochondrial ROS production. Cell Death Dis. 2013, 4, e794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. He, W.; Liu, Q.; Wang, L.; Chen, W.; Li, N.; Cao, X. Corrigendum to “TLR4 signaling promotes immune escape of human lung cancer cells by inducing immunosuppressive cytokines and apoptosis resistance” [Mol. Immunol. 44 (2007) 2850–2859]. Mol. Immunol. 2020, 122, 232–234. [Google Scholar] [CrossRef]
  135. Szczepanski, M.J.; Czystowska, M.; Szajnik, M.; Harasymczuk, M.; Boyiadzis, M.; Kruk-Zagajewska, A.; Szyfter, W.; Zeromski, J.; Whiteside, T.L. Triggering of Toll-like receptor 4 expressed on human head and neck squamous cell carcinoma promotes tumor development and protects the tumor from immune attack. Cancer Res. 2009, 69, 3105–3113. [Google Scholar] [CrossRef] [Green Version]
  136. Luo, L.; Lucas, R.M.; Liu, L.; Stow, J.L. Signalling, sorting and scaffolding adaptors for Toll-like receptors. J. Cell Sci. 2019, 133, 239194. [Google Scholar] [CrossRef]
  137. Hoxhaj, G.; Manning, B.D. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat. Rev. Cancer 2020, 20, 74–88. [Google Scholar] [CrossRef]
  138. Sun, Y.; Wu, C.; Ma, J.; Yang, Y.; Man, X.; Wu, H.; Li, S. Toll-like receptor 4 promotes angiogenesis in pancreatic cancer via PI3K/AKT signaling. Exp. Cell Res. 2016, 347, 274–282. [Google Scholar] [CrossRef]
  139. Dhillon, A.S.; Hagan, S.; Rath, O.; Kolch, W. MAP kinase signalling pathways in cancer. Oncogene 2007, 26, 3279–3290. [Google Scholar] [CrossRef] [Green Version]
  140. Santarpia, L.; Lippman, S.M.; El-Naggar, A.K. Targeting the MAPK-RAS-RAF signaling pathway in cancer therapy. Expert Opin. Ther. Targets 2012, 16, 103–119. [Google Scholar] [CrossRef] [Green Version]
  141. He, W.; Liu, Q.; Wang, L.; Chen, W.; Li, N.; Cao, X. TLR4 signaling promotes immune escape of human lung cancer cells by inducing immunosuppressive cytokines and apoptosis resistance. Mol. Immunol. 2007, 44, 2850–2859. [Google Scholar] [CrossRef] [PubMed]
  142. Kong, C.; Yan, X.; Zhu, Y.; Zhu, H.; Luo, Y.; Liu, P.; Ferrandon, S.; Kalady, M.F.; Gao, R.; He, J.; et al. Fusobacterium Nucleatum Promotes the Development of Colorectal Cancer by Activating a Cytochrome P450/Epoxyoctadecenoic Acid Axis via TLR4/Keap1/NRF2 Signaling. Cancer Res. 2021, 81, 4485–4498. [Google Scholar] [CrossRef] [PubMed]
  143. Qian, J.; Luo, F.; Yang, J.; Liu, J.; Liu, R.; Wang, L.; Wang, C.; Deng, Y.; Lu, Z.; Wang, Y.; et al. TLR2 Promotes Glioma Immune Evasion by Downregulating MHC Class II Molecules in Microglia. Cancer Immunol. Res. 2018, 6, 1220–1233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Garcia, P.V.; Seiva, F.R.; Carniato, A.P.; de Mello Júnior, W.; Duran, N.; Macedo, A.M.; de Oliveira, A.G.; Romih, R.; Nunes Ida, S.; Nunes Oda, S.; et al. Increased toll-like receptors and p53 levels regulate apoptosis and angiogenesis in non-muscle invasive bladder cancer: Mechanism of action of P-MAPA biological response modifier. BMC Cancer 2016, 16, 422. [Google Scholar] [CrossRef] [Green Version]
  145. Hotz, C.; Treinies, M.; Mottas, I.; Rötzer, L.C.; Oberson, A.; Spagnuolo, L.; Perdicchio, M.; Spinetti, T.; Herbst, T.; Bourquin, C. Reprogramming of TLR7 signaling enhances antitumor NK and cytotoxic T cell responses. Oncoimmunology 2016, 5, e1232219. [Google Scholar] [CrossRef] [Green Version]
  146. Umemura, N.; Zhu, J.; Mburu, Y.K.; Forero, A.; Hsieh, P.N.; Muthuswamy, R.; Kalinski, P.; Ferris, R.L.; Sarkar, S.N. Defective NF-κB signaling in metastatic head and neck cancer cells leads to enhanced apoptosis by double-stranded RNA. Cancer Res. 2012, 72, 45–55. [Google Scholar] [CrossRef] [Green Version]
  147. Gambara, G.; Desideri, M.; Stoppacciaro, A.; Padula, F.; De Cesaris, P.; Starace, D.; Tubaro, A.; Del Bufalo, D.; Filippini, A.; Ziparo, E.; et al. TLR3 engagement induces IRF-3-dependent apoptosis in androgen-sensitive prostate cancer cells and inhibits tumour growth in vivo. J. Cell. Mol. Med. 2015, 19, 327–339. [Google Scholar] [CrossRef]
  148. Umansky, V.; Schirrmacher, V. Nitric oxide-induced apoptosis in tumor cells. Adv. Cancer Res. 2001, 82, 107–131. [Google Scholar] [CrossRef]
  149. Yadav, J.; Dikshit, N.; Ismaeel, S.; Qadri, A. Innate Activation of IFN-γ-iNOS Axis During Infection with Salmonella Represses the Ability of T Cells to Produce IL-2. Front. Immunol. 2020, 11, 514. [Google Scholar] [CrossRef]
Figure 1. Cellular localization of the Toll-like receptor (TLR) family. TLR1, TLR2, TLR4, TLR5, TLR6, and TLR10 are localized to the cell surface to recognize common microbial structural components and endogenous ligands. TLR3, TLR7, TLR8, TLR9, TLR12 and TLR13 are located on endosomes to sense microbial nucleic acids that have entered the cell. TLR11 and TLR12 are localized to endosomes to recognize Toxoplasma gondii derived profilin. Created with Biorender.com.
Figure 1. Cellular localization of the Toll-like receptor (TLR) family. TLR1, TLR2, TLR4, TLR5, TLR6, and TLR10 are localized to the cell surface to recognize common microbial structural components and endogenous ligands. TLR3, TLR7, TLR8, TLR9, TLR12 and TLR13 are located on endosomes to sense microbial nucleic acids that have entered the cell. TLR11 and TLR12 are localized to endosomes to recognize Toxoplasma gondii derived profilin. Created with Biorender.com.
Cancers 14 02923 g001
Figure 2. TLR-NF-κB signaling in progression or regression of cancer. Upon recognition of a ligand, TLRs transduce signals through a MyD88-dependent signaling pathway, or alternatively a MyD88-independent TRIF-activating pathway. Upon activation of the MyD88-dependent pathway, adapter molecule TIRAP transduces signals to MyD88. MyD88 activates IRAK1 and IRAK4. Activated IRAK1 and IRAK4 lead to activation of TRAF6. TRAF6 activates the TAK1 complex. The TAK1 complex activates the IKK complex comprised of NEMO, IKKα, and IKKβ. IKK complex activation leads to phosphorylation of IKβα, a protein responsible for sequestering NF-κB to the cytoplasm. Once activated, NF-κB translocates to the nucleus to activate genes that can promote or inhibit tumorigenesis. NF-κB also can be activated through TRIF, notably through TLR3. TRIF activation results in RIP1 and TRAF6 activation. Through RIP1 and TRAF6, the TAK1 complex is activated. Following TAK1 complex activation, subsequent steps in NF-κB signaling are shared between the two pathways. Created with Biorender.com.
Figure 2. TLR-NF-κB signaling in progression or regression of cancer. Upon recognition of a ligand, TLRs transduce signals through a MyD88-dependent signaling pathway, or alternatively a MyD88-independent TRIF-activating pathway. Upon activation of the MyD88-dependent pathway, adapter molecule TIRAP transduces signals to MyD88. MyD88 activates IRAK1 and IRAK4. Activated IRAK1 and IRAK4 lead to activation of TRAF6. TRAF6 activates the TAK1 complex. The TAK1 complex activates the IKK complex comprised of NEMO, IKKα, and IKKβ. IKK complex activation leads to phosphorylation of IKβα, a protein responsible for sequestering NF-κB to the cytoplasm. Once activated, NF-κB translocates to the nucleus to activate genes that can promote or inhibit tumorigenesis. NF-κB also can be activated through TRIF, notably through TLR3. TRIF activation results in RIP1 and TRAF6 activation. Through RIP1 and TRAF6, the TAK1 complex is activated. Following TAK1 complex activation, subsequent steps in NF-κB signaling are shared between the two pathways. Created with Biorender.com.
Cancers 14 02923 g002
Figure 3. TLR-mediated PI3k-Akt signaling in tumorigenesis. PI3k is activated upon TLR stimulation through MyD88, MAL, and BCAP. Activated PI3k converts phosphatidylinositol 4,5-bisphosphate (PIP2) to phosphatidylinositol (3,4,5)-trisphosphate (PIP3). Akt activation occurs through PIP3-facilitated recruitment to the plasma membrane and phosphorylation by PDK1. Activated Akt promotes tumor progression through anti-inflammatory cytokine production, apoptosis resistance, tumor growth, angiogenesis, and metastasis. Created with Biorender.com.
Figure 3. TLR-mediated PI3k-Akt signaling in tumorigenesis. PI3k is activated upon TLR stimulation through MyD88, MAL, and BCAP. Activated PI3k converts phosphatidylinositol 4,5-bisphosphate (PIP2) to phosphatidylinositol (3,4,5)-trisphosphate (PIP3). Akt activation occurs through PIP3-facilitated recruitment to the plasma membrane and phosphorylation by PDK1. Activated Akt promotes tumor progression through anti-inflammatory cytokine production, apoptosis resistance, tumor growth, angiogenesis, and metastasis. Created with Biorender.com.
Cancers 14 02923 g003
Figure 4. TLR-MAPK/ERK signaling in the progression or regression of cancer. MAPK/ERK pathway activation can occur through MyD88-dependent or MyD88-independent TRIF signaling. The MyD88-dependent and -independent signaling converges in the activation of the TAK1 complex. Following the activation of the TAK1 complex, MEKK1/2, MKK4/7, and MKK3/6 are activated. MEKK1/2, MKK4/7, and MKK3/6 activation leads to the activation of ERK1/2, JNK, and p38 MAPK, respectively. Signaling from ERK1/2, JNK, and p38 MAPK can promote tumor growth or inhibition, APC modulation, angiogenesis, and metastasis. Created with Biorender.com.
Figure 4. TLR-MAPK/ERK signaling in the progression or regression of cancer. MAPK/ERK pathway activation can occur through MyD88-dependent or MyD88-independent TRIF signaling. The MyD88-dependent and -independent signaling converges in the activation of the TAK1 complex. Following the activation of the TAK1 complex, MEKK1/2, MKK4/7, and MKK3/6 are activated. MEKK1/2, MKK4/7, and MKK3/6 activation leads to the activation of ERK1/2, JNK, and p38 MAPK, respectively. Signaling from ERK1/2, JNK, and p38 MAPK can promote tumor growth or inhibition, APC modulation, angiogenesis, and metastasis. Created with Biorender.com.
Cancers 14 02923 g004
Figure 5. TLR–IFN signaling in anti-tumor responses. TLRs use a MyD88-independent pathway for IFN signaling. Upon TLR stimulation, membrane-localized TLRs (i.e., TLR4) activate TRIF through adaptor protein TRAM. TRIF activates TRAF3, which activates the TBK1–IKKε complex. The TBK–IKKε complex phosphorylates IRF3. Endosome-localized TLR3 activates TRIF directly. Following TRIF activation, TRAF3 is activated and shares downstream IFN signaling steps with membrane-localized TLRs. TLR–IFN signaling induces type I IFN production, apoptosis, and activation of immune cells. Created with Biorender.com.
Figure 5. TLR–IFN signaling in anti-tumor responses. TLRs use a MyD88-independent pathway for IFN signaling. Upon TLR stimulation, membrane-localized TLRs (i.e., TLR4) activate TRIF through adaptor protein TRAM. TRIF activates TRAF3, which activates the TBK1–IKKε complex. The TBK–IKKε complex phosphorylates IRF3. Endosome-localized TLR3 activates TRIF directly. Following TRIF activation, TRAF3 is activated and shares downstream IFN signaling steps with membrane-localized TLRs. TLR–IFN signaling induces type I IFN production, apoptosis, and activation of immune cells. Created with Biorender.com.
Cancers 14 02923 g005
Table 1. TLR Expression and Tumor Outcomes.
Table 1. TLR Expression and Tumor Outcomes.
Cancer Type TLR-Cell Line CharacterizationPre-Clinical FindingsTumor Profile and Patient OutcomesReferences
Breast CancerTLR2MDA-MB-231, SUM-149, SUM-159-Expression observed in primary tumors and metastatic tissue; high expression associated with shorter overall survival[18,19,20]
TLR3MDA-MB-231, MDA-MB-468, SUM-149, SUM-159Poly(I:C) stimulation reduces breast cancer cell proliferation and induces apoptosisUpregulated in recurring tumors; associated with lower relapse-free survival[19,21,22]
TLR4MDA-MB-231, SUM-149, SUM-159LPS stimulation induces IL-6 and IL-9 production; activation promotes chemoresistance and apoptosis evasion; downregulation enhances paclitaxel sensitivity; upregulation promotes paclitaxel resistanceExpression observed in primary tumors and metastatic tissue; upregulation associated with tumor recurrence and poor survival in TP53 mutant tumors[18,19,21,23,24]
TLR54T1Downregulation upregulates VEGFR and cell proliferation; upregulation and downregulation of receptor increases lung metastases; flagellin treatment reduces tumor growthHighly expressed in metastatic cancer[18,25,26,27]
TLR7/8--Low expression observed in metastases; imiquimod promotes immune cell infiltration in skin-residing metastases[18,28,29]
TLR9MCF-7, T47D, CAMA, MDA-MB-231, MDA-MB-468, SUM-149, SUM-159Receptor knockdown promotes MDA-MB-231 tumor growthExpression observed on tumors; low expression in metastases; downregulation associated with poor disease-specific survival[18,19,21,30]
Lung CancerTLR2-Treatment with lipoprotein reduces Lewis lung carcinoma tumor growthHigh tumoral TLR2 expression is positively correlated with prolonged overall survival and progression-free survival
TLR3Calu-3; H460Lewis lung carcinoma tumors in TLR3-deficient mice had fewer metastases compared to TLR3 competent mice; stimulation with Poly(I:C) induces apoptosisTLR3 positive tumors have greater overall survival and slower disease progression in early-stage NSCLC[26,31,32,33]
TLR4A549; H1299Stimulation with LPS induces production of TGF-β, VEGF, and IL-8High expression associated with decreased overall survival; expression correlated with tumoral PD-L1 expression[28,34,35]
TLR5SPC-A1; A549; H1975; H1299Stimulation with flagellin induces IL-6 and CXCL5 productionHigh expression associated with improved disease-free survival[36]
TLR7/8A549, H1355, SK-MES; LL/2Stimulation promotes survival and chemotherapy resistance, CL264 treatment enhances Lewis lung carcinoma tumor growth; resiquimod formulation improves overall survival and reduced 344SQ tumor progressionHigh expression associated with poor overall survival in stage I-III NSCLC patients[27,37,38]
TLR9A549, NCI-H727Expressed on human NSCLC cell line A549; synthetic oligonucleotide treatment reduces tumor growth in H520, H358, A549, and H1299 xenograftsHigher expression in tumors compared to non-cancerous tissue[39,40]
MelanomaTLR2ME5, ME9, ME16, ME17, ME19Stimulation promotes cell migration; treatment with Zymosan-A and bacteria reduces B16-F10 tumor growthExpression observed on tumors[41,42,43]
TLR3ME2, ME9, ME16, ME17, ME19, M288, M301, M305, M299, M342Stimulation promotes cell migrationExpression observed on tumors[42,43,44]
TLR4ME2, ME9, ME16, ME17, ME19Stimulation promotes cell migrationHighly expressed on primary and metastatic tumors; expression associated with shortened relapse-free survival[42,43,44,45]
TLR7/8M288, M301, M305, M284, M379, M299, M342, M383, M350, M383, M387Imiquimod stimulation inhibits tumoral angiogenesis in a melanoma-bearing humanized mouse modelUpregulated expression in stage III melanoma patients; high expression associated with longer overall survival time; expression correlated with CD8+ T-cell infiltration; treatment with imiquimod inhibits metastasis[43,44,46,47]
TLR9M288, M301, M305, M350, M387Treatment with L-nucleotide-protected TLR agonists reduce B16-F10 tumor growthExpression observed on tumors[43,44,48]
Colorectal CancerTLR1/2--Upregulated in cancerous tissue; high expression associated with improved disease-specific survival[49,50,51]
TLR3HCT116, HT29, SW620Poly(I:C) stimulation induces CCL2, CCL5, and IL-8 production; CXCL8 production, and invasiveness in CRC cell linesLow expression associated with lymph node metastasis and tumor recurrence[52,53]
TLR4-Upregulated in chemically induced CRC in Tir8 −/− miceExpression upregulated in cancerous tissue; high expression associated with poor disease-free survival[54]
TLR5DLD-1Knockdown promotes DLD-1 tumor growth and inhibits immune cell infiltrationLow expression associated with advanced cancer stage; high expression associated with improved disease-specific survival[50,55]
TLR7/8-R848 treatment of CT26 tumors reverses chemoresistance to oxaliplatinUpregulation observed in tumors, associated with lower cancer stage; high expression associated with improved disease-specific survival[50,51,56]
TLR9-Stimulation reduces CT26 tumor growth, increases CD8+ T-cell infiltration in the tumorHigh expression correlated with invasiveness, metastasis, and advanced-stage CRC[48,57]
Pancreatic CancerTLR2HPAC, MIA PaCa-2, PANC-1, BXPC-3, PaCaDD135Stimulation promotes cell proliferation, VEGF expression, and colony formationHighly expressed in all stages of PDAC; upregulation correlated with poor patient survival[58,59]
TLR3PANC-1, BXPC-3Activation promotes cell proliferation-[60,61]
TLR4MIA PaCa-2, SW1990LPS stimulation mediates tumorigenesis in p48Cre;KrasG12D mice, and promotes cell proliferation and VEGF expressionUpregulated in cancerous tissue[59,62]
TLR7/8PANC-2Stimulation promotes cell proliferation, chemoresistance, and tumorigenesis in p48Cre;KrasG12D mice; inhibition prevents tumor progressionExpression upregulated in early and advanced stages of PDAC[63,64]
TLR9PANC-1, SW1990, PaCaDD185, PAN02Stimulation promotes cell proliferation, VEGF expression, and tumorigenesis in p48Cre;LsL-KrasG12D mice; inhibition improves survival and prevents tumor progressionUpregulated in cancerous tissue[59,65]
Ovarian CancerTLR2SKOV3, CAOV3Expression upregulated upon tumor injury in xenografted mice; activation promotes tumoral repair and persistenceUpregulated in cancerous tissue[66,67]
TLR3ES2, OVCAR3, SKOV3, CAOV3Stimulation induces CCL2 and IL-6 productionUpregulated in cancerous tissue[67]
TLR4R182, CP70, A2780, R179, OVCAR3, SKOV3, AD-10, ES2Stimulation promotes cancer cell viability and cell proliferation and induces CCL2, IL-6, and CXCL1 production; knockdown enhances sensitivity to paclitaxelHigh expression in cancerous tissue; high expression associated with improved survival[67,68,69,70,71]
TLR5OVCAR3TLR5-deficiency reduces tumor growth; stimulation promotes invasionPolymorphism diminishing TLR5 signaling improves long-term survival[72,73]
TLR7/8CaOV3, OVCAR3, OV90, SKOV3Stimulation promotes invasion-[72,74]
TLR9--Increased expression associated with rising tumor grade[75]
Prostate CancerTLR2/6PC3Stimulation promotes cell proliferation and invasiveness and induces IL-6 and IL-8 production-[76,77]
TLR3LNCaP, DU145, PC3Stimulation inhibits cell proliferation and promotes apoptosis and induces IL-8, CCL3, CCL5, and CXCL10 productionUpregulated in cancerous tissue; high expression associated with poor patient survival[23,76,77,78]
TLR4PC3, DU145Stimulation promotes cell proliferation and induces IL-6 and IL-8 production; knockdown diminishes tumorigenesis, reduces cell invasiveness and proliferation, and induces apoptosisUpregulated in cancerous tissue[23,76,77,79]
TLR5DU145, PC3, LNCaPStimulation induces IL-8 and CCL5 production-[76]
TLR9--Upregulated in cancerous tissue; high expression associated with poor patient survival[79,80]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Giurini, E.F.; Madonna, M.B.; Zloza, A.; Gupta, K.H. Microbial-Derived Toll-like Receptor Agonism in Cancer Treatment and Progression. Cancers 2022, 14, 2923. https://doi.org/10.3390/cancers14122923

AMA Style

Giurini EF, Madonna MB, Zloza A, Gupta KH. Microbial-Derived Toll-like Receptor Agonism in Cancer Treatment and Progression. Cancers. 2022; 14(12):2923. https://doi.org/10.3390/cancers14122923

Chicago/Turabian Style

Giurini, Eileena F., Mary Beth Madonna, Andrew Zloza, and Kajal H. Gupta. 2022. "Microbial-Derived Toll-like Receptor Agonism in Cancer Treatment and Progression" Cancers 14, no. 12: 2923. https://doi.org/10.3390/cancers14122923

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