Toll-like receptors and cutaneous melanoma (Review)

  • Authors:
    • Ilaria Coati
    • Serena Miotto
    • Irene Zanetti
    • Mauro Alaibac
  • View Affiliations

  • Published online on: September 21, 2016     https://doi.org/10.3892/ol.2016.5166
  • Pages: 3655-3661
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Innate immune cells recognize highly conserved pathogen-associated molecular patterns (PAMPs) via pattern recognition receptors (PRRs). Previous studies have demonstrated that PRRs also recognize endogenous molecules, termed damage‑associated molecular patterns (DAMPs) that are derived from damaged cells. PRRs include Toll‑like receptors (TLRs), scavenger receptors, C-type lectin receptors and nucleotide oligomerization domain‑like receptors. To date, 10 TLRs have been identified in humans and each receptor responds to a different ligand. The recognition of PAMPS or DAMPs by TLRs leads to the activation of signaling pathways and cellular responses with subsequent pro‑inflammatory cytokine release, phagocytosis and antigen presentation. In the human skin, TLRs are expressed by keratinocytes and melanocytes: The main cells from which skin cancers arise. TLRs 1‑6 and 9 are expressed in keratinocytes, while TLRs 2‑5, 7, 9 and 10 have been identified in melanocytes. It is hypothesized that TLRs may present a target for melanoma therapies. In this review, the involvement of TLRs in the pathogenesis and treatment of melanoma was discussed.

Toll-like receptors and the skin

The skin, which is the largest organ of the human body, represents the interface between the environment and the host. It provides the first line of defense against physical, chemical and biological stressors. The skin is predominantly composed of three cell types: Melanocytes, Langerhans cells and keratinocytes. Keratinocytes are the most common type of skin cell, which serve as a protective physical barrier for the human body and present a fundamental element of the innate immune response (1).

The immune system is classified into two types: Innate and adaptive. Innate immunity refers to nonspecific defense mechanisms that are activated immediately following the identification of an antigen in the body. It provides the initial defense against invading pathogens and aids adaptive responses via antigen presentation. By contrast, adaptive immunity provides antigen-specific responses and immunological memory (2).

The innate immune system is composed of numerous cell types, including neutrophils, eosinophils, basophils, mast cells, monocytes, macrophages, dendritic cells, natural killer (NK) cells and γδ T cells. Innate immune cells recognize highly conserved pathogen-associated molecular patterns (PAMPs) via pattern recognition receptors (PRRs). A recent study demonstrated that PRRs also recognize endogenous molecules, termed damage-associated molecular patterns (DAMPs) that are derived from damaged cells (3).

PRRs include Toll-like receptors (TLRs), scavenger receptors, C-type lectin receptors and nucleotide oligomerization domain-like receptors. The recognition of PAMPS or DAMPs by PRRs leads to the activation of signaling pathways and cellular responses with subsequent pro-inflammatory cytokine release, phagocytosis and antigen presentation (4).

The TLR receptor family consists of >10 members in humans and mice, collectively (2,5).

The Toll gene was originally identified as a regulator gene of dorsal-ventral polarity in Drosophila embryos in 1985 (6,7). Subsequent studies revealed that the protein exhibits a key function in Drosophila responses to fungal infections (8). Further studies, based on database searches, identified homologs of Toll in mammals and humans, thus the name ‘TLRs’ was selected (9,10).

TLR1, −2, −4, −5 and 6 are membrane receptors, whereas TLR3, −7, −8 and −9 are intracellular receptors that are localized to the endoplasmic reticulum, endosomes and lysosomes (Table I). TLRs are type I transmembrane proteins composed of 3 domains: An extracellular domain consisting of leucine-rich repeats, a transmembrane domain and an intracellular Toll-interleukin (IL)-1 receptor (TIR) domain (3,11).

Table I.

Overview of Toll-like receptors and their ligands.

Table I.

Overview of Toll-like receptors and their ligands.

ReceptorLocationLigand(s)Signaling pathwayEffect(s)
TLR1Cell membraneGram negative bacteriaMyD88/TIRAP, IRAK/TRAF6Forms heterodimer with TLR2, activates NF-κB
TLR2Cell membraneTLR1 and TLR6 PeptidoglycansMyD88/TIRAP, IRAK/TRAF6Forms heterodimer with TLR1 and −6, activates NF-κB
TLR3EndosomedsRNATRIF, IRF3Induces IFN
TLR4Cell membrane PhagosomeLPS EndocytosisMyD88/TIRAP, IRAK/TRAF6 TRIF, IRF3Activates NF-κB Induces IFN
TLR5Cell membraneFlagellinMyD88, IRAK/TRAF6Activates NF-κB
TLR6Cell membraneGram positive bacteriaMyD88/TIRAP, IRAK/TRAF6Forms heterodimer with TLR2, activates NF-κB
TLR7EndosomessRNAMyD88, IRAK/TRAF6Activates NF-κB, induces IFN
TLR8EndosomessRNAMyD88, IRAK/TRAF6Activates NF-κB
TLR9EndosomeUnmethylated CpG motifs in DNAMyD88, IRAK/TRAF6Activates NF-κB, induces IFN
TLR10UnknownUnknownUnknownPossibly forms heterodimer with TLR1/2

[i] TLR, Toll-like receptor; MyD88, myeloid differentiation primary-response 88; TIRAP, TIR domain containing adaptor protein; TRAF, tumor necrosis factor receptor-associated factor; IRAK, IL-1 receptor-associated kinase; NF-κB, nuclear factor-κB; TRIF, TIR domain-containing adaptor-inducing interferon-β; ds, double-stranded; LPS, lipopolysaccharide; IRF, interferon regulatory factor; IFN, interferon; ss, single-stranded.

The extracellular domain is involved in ligand recognition (PAMPs and DAMPs) and is characterized by the leucine-rich sequence XLXXLXLXX, in which X is an amino acid. The transmembrane region determines the cellular localization of the receptor and exhibits the leucine-rich repeat carboxy-terminal domain. TIR is a conserved protein-protein interaction domain that is required for downstream signaling (1).

Upon recognition of ligands, TLRs dimerize and undergo a conformational change that is required to activate the downstream signaling pathway. Generally, TLRs form homodimers, with the exception of TLR2 and −4, which form heterodimers (3,12).

The TLR signaling cascade involves the recruitment of the following five adaptor molecules to its TIR: Myeloid differentiation primary-response 88 (MyD88) protein, TIR domain-containing adaptor-inducing interferon (IFN)-β (TRIF), TIR domain containing adaptor protein/MyD88-adaptor-like, TRIF-related adaptor molecule and sterile-α and armadillo motif-containing protein (3,12). Two main TLR signaling pathways have been identified: The MyD88-dependent pathway and the TRIF-dependent pathway, and activation depends on whether MyD88 or TRIF is recruited by the TIR domain (3,12). With the exception of TLR3, which signals through the TRIF, all TLRs recruit MyD88. Both pathways lead to the expression of transcription factors, including nuclear factor-κB (NF-κB), c-Jun N-terminal kinase and mitogen-activated protein kinase, which are required for inflammatory gene transcription. This results in the release of a variety of cytokines and inflammatory markers. such as IL-1, −6 and −8, tumor necrosis factor-α (TNF-α), IFN-α and IFN-β (3,11).

Each of the 10 human TLRs respond to a different ligand. For example, TLR2 is involved in the recognition of lipoproteins and peptidoglycans, TLR4 binds bacterial lipopolysaccharide (LPS) and TLR3, −7 and −8, which are located on endosomes, are involved in the recognition of viral and bacterial nucleic acids. Furthermore, flagellin is recognized by TLR5 and TLR9 recognizes unmethylated CpG motifs in DNA (13,14).

TLRs have been identified in a number of cell types, including dendritic cells, neutrophils, lymphocytes, monocytes and NK cells. TLR7 and −9 are expressed on plasmacytoid dendritic cells (pDCs) and B-lymphocytes, while TLR1-6 and 8 are expressed on myeloid-derived DCs. TLRs 1, 2 and 4–10 are expressed by neutrophils and TLR1 is expressed in NK cells. Monocytes express all TLRs, with the exception of TLR3. B-lymphocytes also express TLR1, while TLR2,-8 and −10 may be present on the membrane of T-lymphocytes (13).

In the human skin, TLRs are expressed by keratinocytes and melanocytes, the main cells from which skin cancers arise. TLR1-6 and −9 are expressed in keratinocytes, while TLRs 2–5, −7, −9 and −10 have been identified in melanocytes (1).

TLR-targeted immunotherapies

Immunosuppression allows tumor cells to escape immune-mediated destruction. TLRs are pathogen pattern recognition molecules that identify a variety of pathogens and thus are involved in the regulation of immune responses (15). In addition to exogenous PAMPs, TLRs recognize endogenous ligands, which may alert the innate and adaptive immune systems to the presence of modified tumor cells (1). Therefore, TLR activation of the innate immune response may promote the enhancement of tumor-specific acquired immunity (16).

The involvement of the innate immune response in tumor suppression was first postulated by William B. Coley >100 years ago (17). Coley used heat-killed bacterial cultures of Streptococcus pyogenes and Serratia marcescens (known as Coley's toxin) to successfully treat patients with inoperable soft tissue sarcoma (17). In the early 1990′s, Polly Matzinger hypothesized that tumor antigens are classified as ‘dangerous’ by the immune system in the presence of bacteria that stimulate the immune response (17). Recently, it has been demonstrated that Bacillus Calmette-Guérin induces tumor regression of metastatic melanoma (13). These antitumor effects are associated with TLR activation by LPS and unmethylated bacterial DNA (18).

TLR agonists may present promising drugs for the treatment of malignancies due to their enhancement of the immune response (19). TLR activation induces the release of cytokines involved in cell-mediated immunity and T-regulatory suppression (IL-6 and −12), which shifts the immune response towards Th1 differentiation. This leads to the activation of the type 1 IFN response, which is essential for dendritic cell maturation, antigen cross-presentation and proliferation of NK cells and memory T cells (13).

TLR expression is not confined to immune cells; they have been identified in several cell types, including tumor cells and TLR expression is conserved in these cells. Therefore, TLR agonists are considered as extremely promising drugs for cancer immunotherapy due to their immunostimulatory properties and their pro-apoptotic effects on tumor cells (19).

Notably, epidemiological studies have identified an association between chronic infections and cancer-related mortality in 15% of patients, suggesting that TLR-mediated activation of the innate immune response and the NF-κB pathway in particular, may also promote tumor development due to the types of immune cells and cytokines involved. For example, IL-1, −6, −8 and transforming growth factor-β promote angiogenesis and tumor growth (20). Chronic infectious diseases, such as helicobacter pylori and hepatitis B and C, are associated with the development of cancer, which indicates that TLR-mediated inflammation that is associated with bacteria and viruses may promote carcinogenesis (21).

In 1863, Virchow hypothesized that chronic inflammation enhances cell proliferation: Cancer may develop following exposure to certain irritants, which, in addition to the consequent tissue injury and inflammation caused, enhances cell proliferation (22). It has been established that the proliferation of cells alone does not cause cancer, however, it is hypothesized that an environment rich in inflammatory cells, DNA-damage-promoting agents, activated stroma and growth factors promotes and/or potentiates cell proliferation and increases neoplastic risk (17). In malignant tissues, the tumor microenvironment usually contains an excess of inflammatory cells (23). The therapeutic aim for the future is to normalize the host response by reducing the inflammatory network typically observed in neoplastic tissues: Tumor suppression may be achieved by decreasing the high levels of pro-inflammatory cytokines and increasing the levels of anti-inflammatory cytokines (21).

Various TLR agonists have been investigated for skin cancer immunotherapy: Imidazoquinolines (TLR7 and −8 agonists); CpG oligodeoxynucleotides (ODNs) (TLR9 agonists) (13); and polyriboinosinic-polyribocytidynic acid (Poly I:C) (a synthetic analog of double-stranded RNA that activates TLR3) (19).

Imiquimod

Imiquimod is a member of the imidazoquinolone family, which also includes resiquimod. These drugs topically stimulate the immune response. Stimulation of TLR7- or TLR8-mediated signaling pathways, following treatment with imiquimod or other imidazoquinolines, leads to the activation of central transcription factors, such as NF-κB. Under normal conditions, heterodimeric NF-κB remains inactive within the cytoplasm while bound to the inhibitory factor, inhibitor of κB (IκB). However, following receptor-mediated stimulation, IκB is phosphorylated via the IκB kinase complex (24). This phosphorylation results in the release, activation and nuclear translocation of NF-κB and the subsequent transcription of numerous genes that transcribe cytokines, chemokines, adhesion molecules and apoptosis-related proteins (21). Furthermore, when imiquimod binds to dendritic cells, macrophages and monocytes, activation results in the release of the following pro-inflammatory mediators: TNF-α, IFN-α, IL-1, −6, −8, −12 and −10 (25). These cytokines drive the immune response toward the T helper (Th-1) profile, which is important for control of viruses and tumors, and inhibits the Th-2 pathway, which is implicated in the response against helminths and allergens (26). Imiquimod also acts as a TLR8 agonist, however, it activates TLR-7 more potently (27). In addition, imiquimod stimulates the maturation of Langerhans cells and their migration to regional lymph nodes, with increased levels of antigen presentation to naïve T cells (25). In a mouse model of subcutaneous melanoma, it was demonstrated that pDCs accumulate in subcutaneous melanoma metastases following treatment with imiquimod (28). Furthermore, plasmacytoid predendritic cells migrate to the skin following the application of imiquimod (29). In addition to the indirect stimulation of lymphocytes and NK cells via the activation of dendritic cells, Stary et al (30) demonstrated that imiquimod-treated DCs acquire direct antitumoral functions in vivo. Imiquimod has been demonstrated to modulate signal transducer and activator of transcription-1 signaling pathways, and this interaction may contribute to the induction of apoptosis in a number of cell types (31). Furthermore, imiquimod leads to increased expression of the death receptor, cluster of differentiation (CD)95 (32). Imiquimod may exhibit indirect pro-apoptotic effects on the respective apoptosis-related proteins via TLR-dependent regulation (32). However, imiquimod exerts an additional direct pro-apoptotic activity against tumor cells via activation of the Fas pathway (27).

Imiquimod has been approved for the treatment of condylomata acuminata, superficial basal cell carcinomas (BCCs) and actinic keratosis, however, a number of studies have indicated that it may also be an efficacious treatment for lentigo maligna (LM) and metastatic melanoma (26).

Imiquimod in LM

LM is the in situ phase of LM melanoma (LMM), in which malignant cells are confined to the epidermis. LM occurs in sun-damaged skin and thus it is generally identified on the face or neck of middle-aged or elderly patients (32). The gold standard treatment for LM is conventional surgery using a 5–10 mm margin. However, the localization of the disease, which often arises on the face, makes surgical removal difficult and patients may require extensive plastic repair.

In 2000, Ahmed and Berth-Jones reported the first therapeutic use of imiquimod (5%) in an elderly patient with a large LM on the scalp that refused surgery. Following 7 months of intermittent topical imiquimod application (due to localized reactions), the patient exhibited complete clinical and histological remission and no evidence of recurrence was identified during 9 months of follow up (33).

Additional case reports have demonstrated similar results with regard to LM lesions. Particularly noteworthy is the case of a patient with recurrent LM initially treated using a CO2 laser, who underwent treatment with imiquimod (5%) once or twice a day for 3 months. Following treatment in this patient, biopsy revealed no residual LM (34). Other studies have reported similar outcomes (35); Naylor et al (36) demonstrated a clinical and histological resolution rate of 93% in 28 cases of LM 4 weeks after a 12-week treatment regimen. Additionally, 80% of patients exhibited no evidence of relapse after a year of follow up (36). Similar results were also obtained by Craythorne and Lawrence (37), who demonstrated that in 6/8 LM patients treated with imiquimod, the tumor resolved clinically with no evidence of recurrence after a mean follow up period of 34.2 months. A brisk inflammatory reaction was a prerequisite of therapeutic response (37).

Despite the positive evidence regarding the treatment of LM with imiquimod, at present surgery is considered the best approach for LM treatment.

A recent literature review (38) postulated that for the treatment of LM surgical intervention remains the most widely used and recommended available treatment, however, no randomized controlled trials have demonstrated that surgery is the best therapeutic modality for LM. The use of non-surgical interventions, such as imiquimod as a monotherapy, may be effective and may be considered in selected cases whereby surgical procedures are contraindicated.

The same hypothesis was supported by Kallini et al (39), which considered topical imiquimod as a second line therapy for LM, with surgery as the primary therapeutic option. LM usually occurs in elderly patients, often with concomitant conditions that make surgery difficult to perform; in these conditions imiquimod may represent an alternative treatment choice (40).

Disagreements between certain authors may be due to the absence of shared guidelines for the treatment of LM with imiquimod. In previous studies, dosage (3 times daily or weekly) and treatment duration (2 weeks-7 months) has varied and reported follow-up periods were short, with a median follow up time of <24 months, as reported by Erickson et al in 2010 (41).

A small number of cases that exhibited progression from LM to LMM during treatment with imiquimod have been reported (42,43). We postulate that imiquimod acts by increasing the production of TNF-α, which stimulates the production of metalloproteinase 9, a factor that contributes to the invasive capacity of melanoma, thereby inducing recurrence. However, the treatment of LM that already exhibits an unknown invasive component presents a problem: The application of imiquimod in theses cases represents a significant risk for tumor progression (43).

Data regarding imiquimod use for amelanotic lesions is limited. A recent study demonstrated the histologically-confirmed resolution of an amelanotic LM treated with imiquimod 7 times a week for 8 weeks (44). By contrast, another study reported the accidental use of imiquimod for an achromic superficial spreading melanoma due to incorrect diagnosis, which resulted in a poor response to topical treatment and an increase in lesion size (45).

Imiquimod use in metastatic melanoma

The immune system is essential for the restriction of melanocyte proliferation. This may explain why the eruptive melanocytic nevi phenomenon is observed in organ transplant recipients with clear immunosuppressive conditions, as the immune system of these patients is no longer able to inhibit melanocyte proliferation (46). Furthermore, restoration of complete immune responsiveness leads to regression of melanocytic nevi (47). These findings indicate an association between melanocytic proliferation and the immune system. However, the mechanism by which immunosuppression induces melanoma remains unclear. Notably, the incidence of melanoma in organ transplant recipients is only 2–3 folds higher than that in the general population (47).

In halo nevi and atopic dermatitis, melanocyte proliferation is inhibited due to the pro-inflammatory response (48,49). On the basis of this finding, it has been postulated that imiquimod application, as a result of the pro-inflammatory responses exhibited, may stimulate immune recognition of atypical melanocytes leading to complete or partial elimination of melanocytes within atypical nevi (50). However, in a study by Somani et al (50) no resolution of atypical nevi was observed after twelve weeks of imiquimod treatment, suggesting that melanocytic neoplasms, such as dysplastic nevi, were resistant to imiquimod therapy (50).

By contrast, several studies have demonstrated that topical administration of imiquimod induces regression in melanoma lesions (5154). In 2000, Steinmann et al (54) suggested that topical treatment of cutaneous melanoma metastasis with imiquimod may stimulate melanoma-specific cytotoxic T cells as a consequence of the cross-presentation of melanoma antigens by dendritic cells. Following this, Bong et al (51) investigated the efficacy of imiquimod in the treatment of cutaneous metastasis of melanoma in 3 patients with >15 cutaneous in-transit metastases with unilateral localization in the leg. Imiquimod (5%) was applied topically under occlusion twice a day for 21–28 weeks. Two patients exhibited >90% regression, however, the third patient only responded following the administration of intralesional IL-2 for 2 weeks (51).

Wolf et al (52) observed complete clinical and histopathological remission of melanoma skin metastases in 2 patients following 4 and 8 months treatment with imiquimod (5% cream) on cutaneous lesions, respectively, whereby imiquimod was applied with a 1-cm surrounding margin 3 times a week.

However, poor drug penetration following topical application may limit imiquimod efficacy (55). Turza et al (56) reported that after treatment with imiquimod, a number of dermal melanomas showed clinical regression, but exhibited histopathologically-proven persistence of subcutaneous disease. This suggests that subcutaneous melanomas are resistant to imiquimod as a monotherapy (56,57). In addition, melanomas with high constitutive B cell lymphoma 2 expression appear to be imiquimod-resistant (58).

Therefore, at present imiquimod is not suitable for use as a first line therapy for metastases of cutaneous melanoma (55).

Resiquimod

Resiquimod is a TLR7/8 agonist that is chemically similar to imiquimod. The two drugs are synthetic low molecular weight imidazoquinolinamines. These drugs are immune response modifiers, which exhibit antiviral and antitumor activity by enhancing the production of cytokine and antigen-specific antibodies leading to a shift in immunity towards a Th1 response (59,60). Resiquimod is 10-fold more potent than imiquimod in the stimulation of the Th1 response (61). Furthermore, by contrast to imiquimod, resiquimod may be orally administered (60). Resiquimod may be considered as a potential cancer vaccine adjuvant due to its ability to increase antigen presentation via the direct activation of dendritic cells (DCs), determine local activation of immune cells and enhance the production of pro-inflammatory cytokines and the subsequent transcription of NF-κB and type I IFN (62). A previous study revealed that combined treatment with NY-ESO-1 antigens (a frequently expressed tumor-specific antigen that stimulate humoral and cellular immune responses in cancer patients), Montanide (an immune adjuvant that ensures the slow release of antigens and the recruitment of antigen-presenting cells to the injection site) and topical resiquimod results in high immunogenicity in melanoma patients. This combination increased the number of antibodies and CD4+ T cells, however, no consistent CD8+ T-cell response was identified (62). Conversely, another study demonstrated that topical resiquimod exhibits potent cytotoxic T lymphocyte responses to parenteral antigens in mice (63).

Clinical studies have demonstrated the safety and the efficacy of topical application of resiquimod and its analogs in activating the local immune response (6467). However, resiquimod injection may induce systemic cytokine release and thus, must only be formulated to cause local immune activation, preventing systemic effects (68). Parenteral resiquimod has been associated with transient peripheral blood leukopenia and lymphopenia due to general endothelial cell activation with consequent transiently reduced availability of peripheral-blood leukocytes (69).

CpG ODNs (TLR9 agonists)

CpG are short single-strand DNA cytosine and guanine-rich sequences or ODNs (13,18). CpGs may be classified into 3 types according to their effect on immune cells: CpG-A, a stimulator of NK cells due to its marked IFN-α-producing effect on pDCs; CpG-B, a moderate IFN-α inducer that enhances antigen-specific immune responses; and CpG-C, which combines the properties of CpG-A and CpG-B (13).

CpGs are highly potent immune activators that trigger TLR9 and activate pDCs. Activated pDCs subsequently, release IFN-α, which augments T and NK cell responses and activates conventional myeloid DCs (mDCs) (70).

PF-3512676 is a synthetic CpG-B sequence and TLR9 agonist that has been studied in a variety of tumor types, including renal cell carcinoma, glioblastoma, non-Hodgkin's lymphoma, melanoma and mycosis fungoides (13).

A phase II trial of CpG administered by subcutaneous injections demonstrated a response in 10% of melanoma patients, in addition to evidence of immune system activation (71). A phase I study of intralesional treatment with PF-3512676 in BCC and melanoma patients demonstrated local tumor regression and immune cell activation (72).

Intradermal CpG injections surrounding primary melanoma excisions have been demonstrated to activate pDC and mDCs, reduce the number of regulatory T cells in the draining lymph node and increase the number of melanoma-specific CD8+ T cells, as well as NK cell responses (70,73).

These data suggest potential for investigation of CpG intratumoral injections to induce immunomodulatory reponses in melanoma patients.

Poly I:C

Poly I:C and its more stable derivative poly I:C-poly-L-lysine (Poly ICLC) are synthetic double-stranded RNA sequences that induce IFN production. Their biological effects are mediated by two major double-stranded RNA receptors: TLR3 in the endosome and melanoma differentiation associated gene 5 (MDA5) in the cytosol. After binding poly I:C or poly ICLC, TLR3 and MDA5 initiate downstream signaling that leads to the activation of transcription factors, including IFN regulatory factor (IRF)-3, IRF-7 and NF-κB, resulting in the increased production of type I IFNs and pro-inflammatory cytokines (74).

Previously, poly I:C and poly ICLC were evaluated as single agents in metastatic melanoma, anaplastic glioma and renal cell carcinoma, however, no antitumor efficacy was observed (13). However, recently, they have been used as adjuvants in cancer vaccines. A clinical study demonstrated that the co-administration of poly-ICLC with dendritic cell vaccines decreased the recurrence of malignant glioma (75). These data indicate that poly I:C and poly ICLC may potentially be used as immunological adjuvants to enhance the efficiency of therapeutic cancer vaccines (74).

Conclusion

Development of successful immunotherapies against melanoma has been hindered due to the complex interactions that occur between melanoma and the immune system. In particular, TLRs are expressed by a number of distinct cell types and thus may trigger different responses depending on the cell and the environment. The diverse cell- and stimulus-specific patterns of TLR expression and the distinct actions of TLR agonists indicate the requirement for a more complete understanding of their function in melanoma therapies. The application of TLR agonists presents a novel immunotherapeutic approach for the treatment of melanoma.

References

1 

Burns EM and Yusuf N: Toll-like receptors and skin cancer. Front Immunol. 5:1352014. View Article : Google Scholar : PubMed/NCBI

2 

Akira S, Takeda K and Kaisho T: Toll-like receptors: Critical proteins linking innate and acquired immunity. Nat Immunol. 2:675–680. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Seneviratne AN, Sivagurunathan B and Monaco C: Toll-like receptors and macrophage activation in atherosclerosis. Clin Chim Acta. 413:3–14. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Portou MJ, Baker D, Abraham D and Tsui J: The innate immune system, toll-like receptors and dermal wound healing: A review. Vascul Pharmacol. 71:31–36. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Medzhitov R: Toll-like receptors and innate immunity. Nat Rev Immunol. 1:135–145. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Anderson KV, Bokla L and Nüsslein-Volhard C: Establishment of dorsal-ventral polarity in the Drosophila embryo: The induction of polarity by the Toll gene product. Cell. 42:791–798. 1985. View Article : Google Scholar : PubMed/NCBI

7 

Hashimoto C, Hudson KL and Anderson KV: The toll gene of Drosophila, required for dorsal-ventral embryonic polarity, appears to encode a transmembrane protein. Cell. 52:269–279. 1988. View Article : Google Scholar : PubMed/NCBI

8 

Lemaitre B, Nicolas E, Michaut L, Reichhart JM and Hoffmann JA: The dorsoventral regulatory gene cassette spätzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell. 86:973–983. 1996. View Article : Google Scholar : PubMed/NCBI

9 

Medzhitov R, Preston-Hurlburt P and Janeway CA Jr: A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 388:394–397. 1997. View Article : Google Scholar : PubMed/NCBI

10 

Rock FL, Hardiman G, Timans JC, Kastelein RA and Bazan JF: A family of human receptors structurally related to Drosophila Toll. Proc Natl Acad Sci USA. 95:588–593. 1998. View Article : Google Scholar : PubMed/NCBI

11 

Navi A, Patel H, Shaw S, Baker D and Tsui J: Therapeutic role of toll-like receptor modification in cardiovascular dysfunction. Vascul Pharmacol. 58:231–239. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Akira S and Takeda K: Toll-like receptor signaling. Nat Rev Immunol. 4:499–511. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Adams S: Toll-like receptor agonists in cancer therapy. Immunotherapy. 1:949–964. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Kawai T and Akira S: TLR signaling. Semin Immunol. 19:24–32. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Wang RF, Miyahara Y and Wang HY: Toll-like receptors and immune regulation: Implications for cancer therapy. Oncogene. 27:181–189. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Hoebe K, Janssen E and Beutler B: The interface between innate and adaptive immunity. Nat Immunol. 5:971–974. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Paulos CM, Kaiser A, Wrzesinski C, Hinrichs CS, Cassard L, Boni A, Muranski P, Sanchez-Perez L, Palmer DC, Yu Z, et al: Toll-like receptors in tumor immunotherapy. Clin Cancer Res. 13:5280–5289. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Huen AO and Rook AH: Toll receptor agonist therapy of skin cancer and cutaneous T-cell lymphoma. Curr Opin Oncol. 26:237–244. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Salaun B, Lebecque S, Matikainen S, Rimoldi D and Romero P: Toll-like receptor 3 expressed by melanoma cells as a target for therapy? Clin Cancer Res. 13:4565–4574. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Karin M and Greten FR: NF-kappaB: Linking inflammation and immunity to cancer development and progression. Nat Rev Immunol. 5:749–759. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Coussens LM and Werb Z: Inflammation and cancer. Nature. 420:860–867. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Balkwill F and Mantovani A: Inflammation and cancer: Back to Virchow? Lancet. 357:539–545. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Shacter E and Weitzman SA: Chronic inflammation and cancer. Oncology (Williston Park). 16:217–232. 2002.PubMed/NCBI

24 

Karin M: Nuclear factor-kappaB in cancer development and progression. Nature. 441:431–436. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Stanley MA: Imiquimod and the imidazoquinolones: Mechanism of action and therapeutic potential. Clin Exp Dermatol. 27:571–577. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Navi D and Huntley A: Imiquimod 5 percent cream and the treatment of cutaneous malignancy. Dermatol Online J. 10:42004.PubMed/NCBI

27 

Lee J, Chuang TH, Redecke V, She L, Pitha PM, Carson DA, Raz E and Cottam HB: Molecular basis for the immunostimulatory activity of guanine nucleoside analogs: Activation of Toll-like receptor 7. Proc Natl Acad Sci USA. 100:6646–6651. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Palamara F, Meindl S, Holcmann M, Lührs P, Stingl G and Sibilia M: Identification and characterization of pDC-like cells in normal mouse skin and melanomas treated with imiquimod. J Immunol. 173:3051–3061. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Urosevic M, Dummer R, Conrad C, Beyeler M, Laine E, Burg G and Gilliet M: Disease-independent skin recruitment and activation of plasmacytoid predendritic cells following imiquimod treatment. J Natl Cancer Inst. 97:1143–1153. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Stary G, Bangert C, Tauber M, Strohal R, Kopp T and Stingl G: Tumoricidal activity of TLR7/8-activated inflammatory dendritic cells. J Exp Med. 204:1441–1451. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Stephanou A and Latchman DS: Opposing actions of STAT-1 and STAT-3. Growth Factors. 23:177–182. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Schön MP and Schön M: TLR7 and TLR8 as targets in cancer therapy. Oncogene. 27:190–199. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Ahmed I and Berth-Jones J: Imiquimod: A novel treatment for lentigo maligna. Br J Dermatol. 143:843–845. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Chapman MS, Spencer SK and Brennick JB: Histologic resolution of melanoma in situ (lentigo maligna) with 5% imiquimod cream. Arch Dermatol. 139:943–944. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Powell AM, Russell-Jones R and Barlow RJ: Topical imiquimod immunotherapy in the management of lentigo maligna. Clin Exp Dermatol. 29:15–21. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Naylor MF, Crowson N, Kuwahara R, Teague K, Garcia C, Mackinnis C, Haque R, Odom C, Jankey C and Cornelison RL: Treatment of lentigo maligna with topical imiquimod. Br J Dermatol. 149(Suppl 66): S66–S70. 2003. View Article : Google Scholar

37 

Craythorne EE and Lawrence CM: Observational study of topical imiquimod immunotherapy in the treatment of difficult lentigo maligna. Clin Med Oncol. 2:551–554. 2008.PubMed/NCBI

38 

Tzellos T, Kyrgidis A, Mocellin S, Chan AW, Pilati P and Apalla Z: Interventions for melanoma in situ, including lentigo maligna. Cochrane Database Syst Rev. 12:CD0103082014.PubMed/NCBI

39 

Kallini JR, Jain SK and Khachemoune A: Lentigo maligna: Review of salient characteristics and management. Am J Clin Dermatol. 14:473–480. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Nagore E and Botella-Estrada R: Imiquimod in the treatment of lentigo maligna. Actas Dermosifiliogr. 102:559–562. 2011.(In Spanish). View Article : Google Scholar : PubMed/NCBI

41 

Erickson C and Miller SJ: Treatment options in melanoma in situ: Topical and radiation therapy, excision and Mohs surgery. Int J Dermatol. 49:482–491. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Powell AM, Robson AM, Russell-Jones R and Barlow RJ: Imiquimod and lentigo maligna: A search for prognostic features in a clinicopathological study with long-term follow-up. Br J Dermatol. 160:994–998. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Woodmansee CS and McCall MW: Recurrence of lentigo maligna and development of invasive melanoma after treatment of lentigo maligna with imiquimod. Dermatol Surg. 35:1286–1289. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Lapresta A, García-Almagro D and Sejas AG: Amelanotic lentigo maligna managed with topical imiquimod. J Dermatol. 39:503–505. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Zattra E, Salmaso R, Tonin E and Alaibac M: Achromic superficial spreading melanoma accidentally treated with imiquimod. Acta Derm Venereol. 92:107–108. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Zattra E, Fortina AB, Bordignon M, Piaserico S and Alaibac M: Immunosuppression and melanocyte proliferation. Melanoma Res. 19:63–68. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Russo I, Piaserico S, Belloni-Fortina A and Alaibac M: Cutaneous melanoma in solid organ transplant patients. G Ital Dermatol Venereol. 149:389–394. 2014.PubMed/NCBI

48 

Swope VB, Abdel-Malek Z, Kassem LM and Nordlund JJ: Interleukins 1 alpha and 6 and tumor necrosis factor-alpha are paracrine inhibitors of human melanocyte proliferation and melanogenesis. J Invest Dermatol. 96:180–185. 1991. View Article : Google Scholar : PubMed/NCBI

49 

Tokura Y, Yamanaka K, Wakita H, Kurokawa S, Horiguchi D, Usui A, Sayama S and Takigawa M: Halo congenital nevus undergoing spontaneous regression. Involvement of T-cell immunity in involution and presence of circulating anti-nevus cell IgM antibodies. Arch Dermatol. 130:1036–1041. 1994. View Article : Google Scholar : PubMed/NCBI

50 

Somani N, Martinka M, Crawford RI, Dutz JP and Rivers JK: Treatment of atypical nevi with imiquimod 5% cream. Arch Dermatol. 143:379–385. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Bong AB, Bonnekoh B, Franke I, Schön M, Ulrich J and Gollnick H: Imiquimod, a topical immune response modifier, in the treatment of cutaneous metastases of malignant melanoma. Dermatology. 205:135–138. 2002. View Article : Google Scholar : PubMed/NCBI

52 

Wolf IH, Smolle J, Binder B, Cerroni L, Richtig E and Kerl H: Topical imiquimod in the treatment of metastatic melanoma to skin. Arch Dermatol. 139:273–276. 2003. View Article : Google Scholar : PubMed/NCBI

53 

Arbiser JL, Bips M, Seidler A, Bonner MY and Kovach C: Combination therapy of imiquimod and gentian violet for cutaneous melanoma metastases. J Am Acad Dermatol. 67:e81–e83. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Steinmann A, Funk JO, Schuler G and von den Driesch P: Topical imiquimod treatment of a cutaneous melanoma metastasis. J Am Acad Dermatol. 43:555–556. 2000. View Article : Google Scholar : PubMed/NCBI

55 

Maverakis E, Cornelius LA, Bowen GM, Phan T, Patel FB, Fitzmaurice S, He Y, Burrall B, Duong C, Kloxin AM, et al: Metastatic melanoma-a review of current and future treatment options. Acta Derm Venereol. 95:516–524. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Turza K, Dengel LT, Harris RC, Patterson JW, White K, Grosh WW and Slingluff CL Jr: Effectiveness of imiquimod limited to dermal melanoma metastases, with simultaneous resistance of subcutaneous metastasis. J Cutan Pathol. 37:94–98. 2010. View Article : Google Scholar : PubMed/NCBI

57 

Green DS, Bodman-Smith MD, Dalgleish AG and Fischer MD: Phase I/II study of topical imiquimod and intralesional interleukin-2 in the treatment of accessible metastases in malignant melanoma. Br J Dermatol. 156:337–345. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Schön MP, Wienrich BG, Drewniok C, Bong AB, Eberle J, Geilen CC, Gollnick H and Schön M: Death receptor-independent apoptosis in malignant melanoma induced by the small-molecule immune response modifier imiquimod. J Invest Dermatol. 122:1266–1276. 2004. View Article : Google Scholar : PubMed/NCBI

59 

Schön M and Schön MP: The antitumoral mode of action of imiquimod and other imidazoquinolines. Curr Med Chem. 14:681–687. 2007. View Article : Google Scholar : PubMed/NCBI

60 

Dockrell DH and Kinghorn GR: Imiquimod and resiquimod as novel immunomodulators. J Antimicrob Chemother. 48:751–755. 2001. View Article : Google Scholar : PubMed/NCBI

61 

Thomsen LL, Topley P, Daly MG, Brett SJ and Tite JP: Imiquimod and resiquimod in a mouse model: Adjuvants for DNA vaccination by particle-mediated immunotherapeutic delivery. Vaccine. 22:1799–1809. 2004. View Article : Google Scholar : PubMed/NCBI

62 

Sabado RL, Pavlick A, Gnjatic S, Cruz CM, Vengco I, Hasan F, Spadaccia M, Darvishian F, Chiriboga L, Holman RM, et al: Resiquimod as an immunologic adjuvant for NY-ESO-1 protein vaccination in patients with high-risk melanoma. Cancer Immunol Res. 3:278–287. 2015. View Article : Google Scholar : PubMed/NCBI

63 

Chang BA, Cross JL, Najar HM and Dutz JP: Topical resiquimod promotes priming of CTL to parenteral antigens. Vaccine. 27:5791–5799. 2009. View Article : Google Scholar : PubMed/NCBI

64 

Craft N, Birnbaum R, Quanquin N, Erfe MC, Quant C, Haskell J and Bruhn KW: Topical resiquimod protects against visceral infection with Leishmania infantum chagasi in mice. Clin Vaccine Immunol. 21:1314–1322. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Mark KE, Spruance S, Kinghorn GR, Sacks SL, Slade HB, Meng TC, Selke S, Magaret A and Wald A: Three phase III randomized controlled trials of topical resiquimod 0.01-percent gel to reduce anogenital herpes recurrences. Antimicrob Agents Chemother. 58:5016–5023. 2014. View Article : Google Scholar : PubMed/NCBI

66 

Meyer T, Surber C, French LE and Stockfleth E: Resiquimod, a topical drug for viral skin lesions and skin cancer. Expert Opin Investig Drugs. 22:149–59. 2013. View Article : Google Scholar : PubMed/NCBI

67 

Rook AH, Gelfand JM, Wysocka M, Troxel AB, Benoit B, Surber C, Elenitsas R, Buchanan MA, Leahy DS, Watanabe R, et al: Topical resiquimod can induce disease regression and enhance T-cell effector functions in cutaneous T-cell lymphoma. Blood. 126:1452–1461. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Tomai MA, Miller RL, Lipson KE, Kieper WC, Zarraga IE and Vasilakos JP: Resiquimod and other immune response modifiers as vaccine adjuvants. Expert Rev Vaccines. 6:835–847. 2007. View Article : Google Scholar : PubMed/NCBI

69 

Gunzer M, Riemann H, Basoglu Y, Hillmer A, Weishaupt C, Balkow S, Benninghoff B, Ernst B, Steinert M, Scholzen T, et al: Systemic administration of a TLR7 ligand leads to transient immune incompetence due to peripheral-blood leukocyte depletion. Blood. 106:2424–2432. 2005. View Article : Google Scholar : PubMed/NCBI

70 

Molenkamp BG, van Leeuwen PA, Meijer S, Sluijter BJ, Wijnands PG, Baars A, van den Eertwegh AJ, Scheper RJ and de Gruijl TD: Intradermal CpG-B activates both plasmacytoid and myeloid dendritic cells in the sentinel lymph node of melanoma patients. Clin Cancer Res. 13:2961–2969. 2007. View Article : Google Scholar : PubMed/NCBI

71 

Pashenkov M, Goëss G, Wagner C, Hörmann M, Jandl T, Moser A, Britten CM, Smolle J, Koller S, Mauch C, et al: Phase II trial of a toll-like receptor 9-activating oligonucleotide in patients with metastatic melanoma. J Clin Oncol. 24:5716–5724. 2006. View Article : Google Scholar : PubMed/NCBI

72 

Hofmann MA, Kors C, Audring H, Walden P, Sterry W and Trefzer U: Phase 1 evaluation of intralesionally injected TLR9-agonist PF-3512676 in patients with basal cell carcinoma or metastatic melanoma. J Immunother. 31:520–527. 2008. View Article : Google Scholar : PubMed/NCBI

73 

Molenkamp BG, Sluijter BJ, van Leeuwen PA, Santegoets SJ, Meijer S, Wijnands PG, Haanen JB, van den Eertwegh AJ, Scheper RJ and de Gruijl TD: Local administration of PF-3512676 CpG-B instigates tumor-specific CD8+ T-cell reactivity in melanoma patients. Clin Cancer Res. 14:4532–4542. 2008. View Article : Google Scholar : PubMed/NCBI

74 

Wang C, Zhuang Y, Zhang Y, Luo Z, Gao N, Li P, Pan H, Cai L and Ma Y: Toll-like receptor 3 agonist complexed with cationic liposome augments vaccine-elicited antitumor immunity by enhancing TLR3-IRF3 signaling and type I interferons in dendritic cells. Vaccine. 30:4790–4799. 2012. View Article : Google Scholar : PubMed/NCBI

75 

Pollack IF, Jakacki RI, Butterfield LH, Hamilton RL, Panigrahy A, Normolle DP, Connelly AK, Dibridge S, Mason G, Whiteside TL and Okada H: Immune responses and outcome after vaccination with glioma-associated antigen peptides and poly-ICLC in a pilot study or pediatric recurrent low-grade gliomas. Neuro Oncol. 18:1157–1168. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 12 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Coati I, Miotto S, Zanetti I and Alaibac M: Toll-like receptors and cutaneous melanoma (Review). Oncol Lett 12: 3655-3661, 2016
APA
Coati, I., Miotto, S., Zanetti, I., & Alaibac, M. (2016). Toll-like receptors and cutaneous melanoma (Review). Oncology Letters, 12, 3655-3661. https://doi.org/10.3892/ol.2016.5166
MLA
Coati, I., Miotto, S., Zanetti, I., Alaibac, M."Toll-like receptors and cutaneous melanoma (Review)". Oncology Letters 12.5 (2016): 3655-3661.
Chicago
Coati, I., Miotto, S., Zanetti, I., Alaibac, M."Toll-like receptors and cutaneous melanoma (Review)". Oncology Letters 12, no. 5 (2016): 3655-3661. https://doi.org/10.3892/ol.2016.5166