Abstract
Melanoma, a deadly form of skin cancer, poses significant challenges to the host immune system, allowing tumor cells to evade immune surveillance and persist. This complex interplay between melanoma and the immune system involves a multitude of mechanisms that impair immune recognition and promote tumor progression. This review summarizes the intricate strategies employed by melanoma cells to evade the immune response, including defective immune recognition, immune checkpoint activation, and the role of regulatory T-cells, myeloid-derived suppressor cells, and exosomes in suppressing anti-tumor immunity. Additionally, we discuss potential therapeutic targets aimed at reversing immune evasion in melanoma, highlighting the importance of understanding these mechanisms for developing more effective immunotherapies. Improved insights into the interactions between melanoma and the immune system will aid in the development of novel treatment strategies to enhance anti-tumor immune responses and improve patient outcomes.
Keywords: Melanoma , skin cancer , immune response
Introduction
The skin is the body's largest organ, constantly exposed to various mechanical, thermal, and chemical aggressors.
While any of these factors can damage the skin's integrity, UVR have been linked to premalignant and malignant skin lesions by triggering a cascade of biological effects induced by the presence of photons [1, 2].
This is because the chromophores typically found in human skin, such as melanin, capture UVR-photon energy through a process known as light absorption, causing their electrons to become excited when exposed to light [1].
While the excitation is limited, the process is not harmful, and slight, short-lived inflammation is essential for skin repair, regeneration, and remodeling [3].
However, if the excitation lasts long enough, preventing the chromophore from returning to the basal state (reached by dissipating energy in the form of heat or photon emission), a pro-photooxidative state of the skin is reached, leading to the appearance of reactive oxygen species (ROS), chronic inflammatory signaling, and immunosuppression, resulting in tissue homeostasis alteration [4, 5].
While the immune response to melanoma was considered essential in the treatment of the condition, the survival rates of patients diagnosed with metastatic melanoma remained stagnant as no real progress was made, especially for patients with metastatic progression [6].
However, the emergence of advanced molecular diagnostic methods has led to the identification of many genetic mutations, amplifications, and deletions that seem to play a crucial role in promoting tumor development and survival signaling [7].
This is crucial because melanoma cells are continually changing to adapt to host defenses [7].
In this brief communication, we will briefly review the main cellular and molecular mechanisms through which melanoma cells outsmart and evade the host immune response.
These mechanisms include sustaining proliferative signaling, inducing angiogenesis, evading growth suppressors to activate and sustain invasion/metastasis, enabling replicative immortality, and avoiding apoptosis [8].
Immune response to UVR
While both UVRA and UVRB reach the skin, UVRB can recruit macrophages and neutrophils into the skin [9].
This is because the energy from light induces cell infiltration of melanomas by upregulating Ccr2 [10] and ATF2 [11].
UVRB also induces ligands for the chemokine receptor Ccr2 (only Ccl8, while the expression of other Ccr2 ligands returned to baseline), causing macrophages to produce INF-γ and melanocyte activation, characterized by aberrant growth and migration [10].
An inflammatory positive feedback loop that strengthens macrophage-melanocyte interactions appears.
Surprisingly, IFN-γ (and not type-I interferons) from recruited monocytic cells stimulates melanocyte proliferation, migration, as well as the expression of genes involved in evasion of the immune defense barriers[10].
Anti-IFN-γ antibodies exhibited significantly reduced UVRB-mediated activation of melanocytes [12].
The IFN-γ signaling pathway is involved in the initiation, survival, and/or outgrowth of UVRB-induced melanoma cells, mediating pro-tumorigenic effects [10].
After exposure to UVRB radiation, melanocytes play a role in controlling the skin's immune responses in neonatal mice, with A TF2 being involved in this process.
Additionally, ATF2 mutant animals displayed a reduction in the infiltration of macrophages into their skin [11].
IL-23 inhibits the incidence, growth, and melanoma progression by preventing nevus initiation and growth, reducing metastasis to lymph nodes, and prolonging the survival of transformed melanocytic cells, inhibiting proliferation of melanocytic cell lines, blocking tumor-promoting IFNγ, and inducing melanocyte DNA repair.
Normal melanocytes and nevus cells express IL-12 and IL-23 receptors [13].
Interleukin-6 (IL-6) is a multifunctional immunomodulatory cytokine that is synthesized by a variety of cell types, including melanoma cells.
IL-6 is known to exert a significant influence on the pathophysiology and progression of malignancies.
The suppression of apoptosis and the induction of tumor angiogenesis are mechanisms through which it facilitates the growth of tumors [14].
IL-2, a well-described stimulator of CD8+T cells (CD8 T) and natural killer (NK) cells [15], with minimal modifications in T-regulatory cells, proved useful in metastatic melanoma and renal cell carcinoma.
Despite the low-affinity IL2 receptor beta gamma subunits (IL2Rβγ) and, therefore, the need for high doses, the molecule offered up to 25% durable responses for melanoma treatment [16].
IL-7, IL-15, IL-18 seem to be key supporters of T-cell expansion and function in vivo [17, 18].
UVR is well acknowledged as a prominent risk factor in the pathogenesis of cutaneous melanoma.
The accumulation of stochastic mutations in melanocytes, generated by UVR, results in the transformation of these cells and the creation of tumors.
UVR possesses the ability to elicit both localized and systemic immunological responses that are specific to antigens. Consequently, this allows converted melanocytes to evade immune monitoring [19].
Tumors that were highly antigenic and produced by UVR were allografted into syngeneic mice, and these mice were also subjected to UVR. When UVR was not present, the tumors that were inoculated experienced rejection.
Comparable findings in mice with compromised immune systems were reported, indicating that immunological suppression is facilitated by exposure to UVR [20, 21, 22, 23
Multiple processes have been suggested to be involved in the immune suppression generated by UVR.
These mechanisms encompass impaired antigen presentation, the secretion of immunosuppressive cytokines, and the death of immune cells [24].
Exposure of the skin to UVR has been observed to result in a decrease in the number of Langerhans cells, a specific type of dendritic cell located in the dermis, at the location of exposure [19, 22].
Langerhans cells that have been exposed to UVR undergo migration to the lymph nodes, where they exhibit an impaired ability to activate Th1 cells, a crucial component in the initiation of an efficient immune response.
In contrast, the activation of Th2 cells induces immunological suppression through the activation of regulatory T cells [24, 25, 26].
Furthermore, it has been observed that Langerhans cells, which have been exposed to UVR, exhibit impairments in their ability to present antigens in the lymph nodes and experience death when subjected to greater levels of UVR.
This indicates that UVR exposure has the potential to diminish the presentation of tumor antigens to the immune system [19].
During the transformation of immune cells into a state of immunosuppression, there was also observed an elevation in the levels of immunosuppressive cytokines, namely IL-10, IL-4, and TNF-α, which were detected both at the site of action and throughout the entire system[24,26, 27, 28].
The exposure to UVR results in a decrease in the cytokine IL-12, causing an alteration in the ratio of Th1 and Th2 cells, with a subsequent increase in the latter population [29,30].
Also, stimulation of Th2 cells by UVR may have a role in the elevated presence of M2-like tumor-associated macrophages (TAMs) observed in individuals with melanoma (Figure 1).
Figure 1.
An overview of the immune system dysfunctions that support the onset and spread of melanoma. UV causes systemic and local immune suppression, which lowers immune cell production and function. Cancer cells exploit the immune checkpoint axis through interactions with immune checkpoints expressed on T cells. The cargo carried by exosomes derived from tumors leads to immune suppression, both locally and systemically. Melanoma cells elevate the presence of immune suppressive cells, such as Tregs and MDSCs (M1 and M2-like TAMs)
Collectively, these findings indicate that it is advisable to restrict sun exposure when administering immunotherapeutic agents, substances that restore the immune system.
This precaution is necessary because excessive UVR during the treatment protocol could potentially diminish the efficacy of the immunotherapeutic agent."
Impaired Functioning of T-Cells
Cancer cells exploit the immune-checkpoint axis, which involves the interaction between programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1), to their advantage (Figure 1).
The PD-1/PD-L1 axis acts as an inhibitor of the immune response and acts as a protective mechanism for the host against autoimmune reactions [31, 32, 33, 34]
When a host is infected by a pathogen, T-lymphocytes are recruited and guided to the infection site, where they initiate their immune response against the invading pathogen.
Meanwhile, cells in the nearby healthy tissues deploy a protective mechanism against T-lymphocytes by generating PD-L1.
This molecule facilitates an interaction with the PD-1 receptor found on T-lymphocytes, thereby hindering the subsequent activation of these T-cells.
In the context of cancer, the interaction between PD-1 expressed on cytotoxic T-lymphocytes and PD-L1 found on tumor cells, tumor-associated macrophages, NK cells, dendritic cells, and other immune cells results in the establishment of a state known as T-cell exhaustion.
This condition impairs the immune system's capacity to recognize and eliminate tumors and is induced by epigenetic alterations that occur within T-cells [35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48].
The importance of PD-1/PD-L1 interactions goes beyond the confines of the tumor microenvironment, as they have also been observed in the lymph nodes that drain from the tumor.
These interactions take place between PD-1-expressing T-cells and PD-L1-expressing dendritic cells, contributing to the emergence of an anergic or exhausted T-cell phenotype [49].
The Programmed Cell Death Protein 2 (PD-L2) acts as the alternate ligand for PD-1 and has been detected on both antigen-presenting cells and melanoma cells. PD-L2 shares functional characteristics with PD-L1, functioning as a negative regulator of cytotoxic T-cell activity.
Elevated levels of PD-L1 expression have been noted in melanoma, with specific subtypes of the disease exhibiting varying degrees of this expression.
Cutaneous melanoma displays the highest PD-L1 expression, with a rate of 62%, followed by mucosal melanoma at 44%, acral melanoma at 31%, and uveal melanoma at 10% [39,50, 51, 52, 53].
PD-L2 is found in higher abundance compared to PD-L1, and it also exhibits a stronger affinity for PD-1, suggesting that there might be a distinct contribution by PD-L1 and PD-L2 in the modulation of immune responses.
Furthermore, prior research has shown that glycosylation of PD-L1 enhances its stability and strengthens its interaction with PD-1, thereby increasing its ability to induce T-cell exhaustion [54, 55, 56, 57, 58].
Cytotoxic T-cells exposed to ongoing tumor antigens become activated and subsequently produce interferon-γ (IFN-γ), then interacts with the IFN-γ receptor found on melanoma cells, initiating the downstream signaling cascade of the JAK/STAT/IRF1 axis.
This cascade activates transcription factors IRF1 and MYC, leading to their binding to the PD-L1 promoter.
Additionally, the involvement of transcription factors STAT3 and IRF1 is essential for the functioning of PD-L2 [59, 60, 61, 62, 63].
Various transcription factors, including HIF-1, AP-1, and NF-қβ, have been identified as regulators of PD-L1 expression in melanomas.
However, the specific mechanisms through which they exert their regulatory effects vary due to differences in the mutational landscape [58,64, 65].
Upon activation of the T-cell receptor (TCR) through engagement with the antigen/MHC complex, the MAPK and PI3K/AKT pathways are activated, leading to various T-cell activation phenotypes (transcriptional activation, cytokine production, T-cell survival, and proliferation) (Figure 1) [66, 67, 68, 69, 70].
In the context of cancer, the interaction between PD-L1/PD-L2 and PD-1 on cytotoxic T-cells has been observed to recruit SHP1/2 to the TCR.
This interaction subsequently affects various phosphorylation activities, resulting in impaired function and metabolism of cytolytic T-cells [36,37,60,69,71, 72, 73, 74, 75, 76, 77, 78, 79, 80].
Apart from PD-1, several other immune suppressive checkpoint molecules exist that decrease the activity of cytotoxic T-cells against cancer cells: neuropilin-1 (NRP-1), cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), T-cell immunoglobulin, lymphocyte-activation gene 3 (LAG-3), mucin domain-containing protein 3 (TIM-3), and V-domain Ig suppressor of T-cell activation (VISTA) [81, 82, 83, 84, 85, 86, 87, 88, 89].
Multiple trials suggest that administering anti-PD1/anti-PD-L1 treatment for early-stage melanoma may hinder the establishment of durable immune responses.
An alternative strategy to activate suppressed immune cells is the sequential administration of cancer vaccines in addition to anti-PD-1/anti-PD-L1 therapy.
CTLA-4 is recognized as the second most prominent immune suppressive checkpoint regulator (Figure 1).
The significance of CTLA-4 as a negative regulator of the immune system is evidenced by the occurrence of severe autoimmune disorders in mice lacking CTLA-4, which arises from uncontrolled T-cell activation. CTLA-4 expression on T-cells is responsible for its immunosuppressive effects, achieved by suppressing T-cell activation through competitive binding with CD28 for the ligands CD80/CD86, T process known as “T cell anergy” [82,83,90, 91, 92, 93, 94].
The actions of PD-1, PD-L1, and CTLA-4 may have broader implications beyond their established roles as negative regulators of the immune system (Figure 1).
The signaling pathways involving PD-1, PD-L1, and CTLA-4 in melanomas have been shown to potentially impact various aspects of tumor biology (proliferation, growth, metastatic signaling, survival, and the establishment of an immunosuppressive microenvironment around the tumor cells) [95, 96, 97, 98, 99, 100].
Exosomes
Both normal and cancer cells release exosomes, but cancer cells release larger quantities compared to normal cells [101].
The cargo carried by tumor-derived exosomes plays a crucial role in activating the pre-metastatic environment and suppressing anti-tumor immune responses, thereby facilitating cancer cell metastasis (Figure 1) [101].
Melanoma-derived exosomes have been observed to migrate to lymph nodes, inducing tumor tolerance and preparing the lymph nodes for the arrival of melanoma cells [102, 103].
Exosomes transfer melanoma-derived MHC I towards antigen-presenting cells, decrease the expression of molecules like CD80/CD86, and increase the expression of immunosuppressive cytokines such as IL-6 and TGF-β, resulting in the altered function of antigen-presenting cells and decreased T-cell proliferation [104].
Tumor-derived exosomes interact with T-cells, inhibiting their activation and inducing apoptosis [105, 106, 107].
Melanoma exosomes impairs cytolytic function of NK cells and contribute to the formation of myeloid-derived suppressor cells, leading to their accumulation within the tumor microenvironment and suppression of the immune response [107, 108, 109].
Melanoma exosomes can activate a mixed population of M1-and M2-like TAMs, with the tumor microenvironment favorizing the M2-like TAM phenotype [110, 111, 112, 113].
Also, melanoma cancer cells-derived exosomes have been demonstrated to express the immune control molecule PD-L1 on their membranes, leading to the suppression of the immune response, both locally and generalized [114, 115].
Spontaneous Melanoma-Prone Mouse Model
These models accurately replicate the immunological profiles observed in human melanoma patients and display an increase in immunosuppressive myeloid-derived suppressor cells (MDSCs) within the tumor microenvironment, correlating with the presence of anergic gp100-expressing melanoma-specific CD8+T-cells, in the same line with the melanoma patient data.
These models exhibit an inverse relationship between dendritic cell (DC) levels and tumor burden, and restoring DC populations by Flt3L, a growth factor, can enhance cytotoxic cytokine production by T-cells [116, 117, 118].
Another study demonstrated an increase in regulatory T-cells (Tregs) (Figure 1), a decrease in CD8+T-cells from the tumor tissues, together with an increase of immunosuppressive cytokines IL-10 and TGF-β, correlating with increasing tumor mass in these mice, and paralleling melanoma patient data.
As tumor burden rises, CD8+T-cell activation markers and lymphocyte proliferative capacity decrease in these models [116].
Defective Immune Recognition of Melanomas by the Immune System
During melanoma progression, there is a process called immune editing, characterized by the selection of subclones based on their ability to evade immune detection, and includes three phases: elimination, equilibrium, and escape [119, 120].
In the elimination phase, dendritic cells detect antigenic melanoma clones, capture these melanoma antigens, process them, and present them on their Major Histocompatibility Complex II (MHC II) to naïve T-cells in the lymph nodes, leading to the activation of melanoma-specific cytotoxic CD8+T-cells.
The equilibrium phase involves immune responses eliminating highly antigenic melanoma clones, while some clones escape anti-tumor immune responses.
During the escape phase, low immunogenic melanoma clones proliferate and disseminate rapidly [119].
As melanoma progresses, tumor cells and immune cells that suppress the immune system produce different soluble molecules that interfere with dendritic cells' ability to activate naïve T-cells into effector CD8+T-cells in lymph nodes.
Immunosuppressive cytokines, such as IL-10 can lead to defective antigen presentation by dendritic cells, reducing T-cell activation.
Dendritic cells downregulate membranous expression of MHC II and co-stimulatory molecules CD80/CD86, which are crucial for T-cell activation [121, 122, 123].
The expression of CTLA-4, PD-1, PD-L1, and PD-L2 (immune checkpoint molecules), on dendritic cells disrupts their innate immune functions and affects T-cell activation [120,124, 125, 126, 127, 128, 129].
Melanoma subclones, during genetic and epigenetic changes, can successfully downregulate essential elements of their MHC I antigen presentation pathways, efficiently evading immune surveillance.
Native melanoma-associated antigens (MART-1/Melan-A, gp100, tyrosinase), are variably expressed in melanoma cells.
As melanoma subclones successfully downregulate MHC I or another melanoma antigen's expression, they can establish an immune-refractory tumor.
Interestingly, melanoma cells express MHC II on their surface, attracting tumor-specific CD4+T-cells that suppress anti-tumoral activity of cytotoxic T-cell by counteracting the IFN-γ pathway of the immune responses [130].
Myeloid-derived suppressor cells (MDSCs) have the ability to undergo differentiation into tumor-associated macrophages (TAMs) and exhibit plasticity by transitioning between M1-and M2-like macrophage phenotypes within the microenvironment of the tumor (Figure 1).
Hypoxic regions present within the tumor have a tendency to induce a shift in TAMs towards an M2-like phenotype, whereas adequately oxygenated conditions promote an M1-like phenotype [3,111, 112, 131, 132, 133].
The progression of melanoma is accompanied by a heightened infiltration of TAMs, with a specific increase in M2-like TAMs.
The ratio of M1 to M2 TAMs has been identified as a significant prognostic indicator [131, 134, 135, 136, 137].
M1 TAMs have been found to exhibit anti-tumor actions, whereas M2 TAMs have been observed to promote tumor growth. those with melanomas that have a greater abundance of M1 gene signatures seem to have a more favorable prognosis in comparison to those with elevated M2 gene signatures [138, 139].
The inhibition of M-CSF receptors on MDSCs has been observed to induce a shift in TAMs towards an M1 phenotype, which is further reinforced by GM-CSF signaling [140].
Furthermore, the inhibition of the macrophage receptor with collagenous structure (MARCO) through the use of an antibody has been shown to facilitate the differentiation of TAMs into an M1 phenotype [141].
The MARCO, which is a scavenger receptor involved in pattern recognition, has been found to be linked to a gene expression profile that resembles the phenotype of M2-like tumor-associated macrophages [141].
The aforementioned findings indicate that the M-CSF and MARCO potentially have a role in regulating the PI3K/AKT/mTOR axis and the polarization of TAMs into M1 or M2 phenotypes [140, 141, 142, 143, 144, 145, 146].
Th (Helper T cells) have a crucial function in the regulation of adaptive immune responses.
They accomplish this by stimulating cytotoxic T cells and exerting an influence on the phagocytic and digesting characteristics of macrophages.
The polarization of M1 and M2 macrophages is attributed to two subclasses of Th, namely Th1 and Th2 [138,147].
A Th1 bias has been observed in both healthy individuals and patients who have undergone surgical resection of melanomas.
Nevertheless, it has been observed that in individuals with melanoma, there is a prevalence of Th2 cells, which contributes to the development of chronic inflammation, which further facilitates the evolution of melanoma by causing a shift in the polarization of TAMs towards M2 phenotypes [148, 149, 150].
M1 TAMs demonstrate anti-tumor characteristics through the release of proinflammatory cytokines, ROS, nitric oxide, and their ability to function as proficient antigen-presenting cells, hence facilitating adaptive anti-tumor immune responses.
On the other hand, it has been observed that M2 macrophages have a role in promoting the growth of melanoma by facilitating tumor angiogenesis, promoting the function of regulatory T cells to suppress the activity of cytolytic T cells, and secreting soluble substances that inhibit immune responses against the tumor [131,151, 152, 153, 154].
In the progression of melanoma, there is a notable tendency for M1 TAMs to undergo a phenotypic transition towards an M2.
This shift in phenotype is known to facilitate tumor growth and enable evasion of the immune system.
The enhancement of melanoma treatment results could be achieved by the development of medicines capable of inducing a transition of M2 to M1 TAMs [138,141].
Role of Regulatory T-Cells in Melanoma Immune Evasion
CD4+Tregs have a significant role in the regulation of an exaggerated immune response, thereby mitigating potential harm to the host.
Nevertheless, cancer cells manipulate the identical defensive mechanisms utilized by Tregs in order to evade the immune system. In the context of melanoma, Tregs exhibit an upregulation in their presence within the peripheral blood, lymph nodes, and tumor microenvironment.
This increase in Tregs has been associated with a decrease in the cytolytic activity of immune cells that are responsible for targeting and eliminating tumor cells.
Melanomas employ the strategy of recruiting and stimulating Tregs by the secretion of H-ferratin and chemoattractant cytokines/ chemokines.
This process effectively influences the functionality of Tregs within the localized tumor microenvironment.
Tregs utilize a variety of strategies to inhibit the immune system, which can be categorized into four distinct mechanisms.
These mechanisms involve the secretion of immune suppressive cytokines, namely IL-10, IL-35, and TGF-β, the induction of cytolysis in immune cells, the targeting of dendritic cells, and the disruption of immune cell function through metabolic processes [155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166].
Role of MDSCs in Melanoma Immune Evasion
Myeloid cells play a crucial role in the innate immune system, serving as vital constituents that safeguard the host against pathogens through the processes of phagocytosis and the initiation of inflammatory responses, which in turn recruit other immune cells.
It has been observed that cancer cells have the ability to induce the transformation of myeloid cells located in the bone marrow into MDSCs [167, 168].
MDSCs have been identified as key contributors to the advancement of cancer, as they facilitate the spread of tumor cells and impede the functioning of T-cells.
The presence of MDSCs in both the peripheral blood and tumor microenvironment has been observed to be correlated with disease progression, diminished T-cell activity, and prognostic significance in melanoma [117,168, 169, 170, 171, 172, 173].
Conclusions
UVR is a well-established risk factor for the development of cutaneous melanoma.
It triggers a cascade of biological effects in the skin, particularly in melanocytes, which can lead to DNA damage and the formation of melanoma lesions.
Melanin and other chromophores in the skin absorb UVR energy, leading to electron excitation.
Prolonged excitation can result in the generation of ROS, chronic inflammation, and immunosuppression, which can alter tissue homeostasis and promote melanoma progression.
Advanced molecular diagnostic methods have identified various genetic mutations, amplifications, and deletions that play a crucial role in melanoma development and survival signaling.
Melanoma cells continually adapt to host defenses, making treatment challenging.
UVR exposure can trigger an immune response, recruiting immune cells like macrophages and neutrophils to the skin.
However, the interplay between UVR and immune responses can lead to melanocyte activation and, in some cases, contribute to pro-tumorigenic effects.
Various cytokines, including IL-23, IL-6, IL-2, IL-7, IL-15, and IL-18, have roles in modulating immune responses in melanoma.
They can either inhibit or promote tumor growth and affect the function of T-cells and other immune cells.
Immune checkpoint molecules like PD-1, PD-L1, and CTLA-4 are exploited by melanoma cells to evade the host immune response.
These molecules lead to T-cell exhaustion, impairing the immune system's ability to recognize and eliminate tumors.
Melanoma-derived exosomes play a significant role in suppressing anti-tumor immune responses, priming pre-metastatic niches, and facilitating cancer cell metastasis.
They carry various cargo that can inhibit immune cell function.
Melanoma progression involves a process known as immune editing, where tumor cells selectively evade immune detection.
This process includes phases of elimination, equilibrium, and escape, with immune suppression mechanisms playing a crucial role. Myeloid-derived suppressor cells (MDSCs) can transform into tumor-associated macrophages (TAMs) and contribute to an immunosuppressive microenvironment.
M1-like TAMs have anti-tumor properties, while M2-like TAMs promote tumor growth.
Tregs are recruited and stimulated by melanoma cells, leading to immune suppression.
They employ various mechanisms, including the secretion of immunosuppressive cytokines and the targeting of dendritic cells, to inhibit the immune response.
The main findings of this study are summarized in Table 1.
Table 1.
Key Findings and Concepts
Immune Response to UVR |
- UVR exposure recruits macrophages and neutrophils to the skin. |
- UVR induces melanocyte activation, IFN-γ production, and immune interactions. | |
- Immune responses to UVR can have pro-tumorigenic effects via IFN-γ signaling. | |
- ATF2 plays a role in immune responses to UVR radiation. | |
Cytokines in Melanoma |
- Cytokines such as IL-23, IL-6, IL-2, IL-7, IL-15, and IL-18 modulate immune responses in melanoma. |
- IL-2 is used for treating metastatic melanoma. | |
- IL-23 inhibits melanoma progression by various mechanisms. | |
UVR and Immune Suppression |
- UVR exposure leads to immune suppression through multiple mechanisms. |
- Langerhans cells are affected by UVR, impairing antigen presentation. | |
- Changes in cytokine levels, including IL-10, IL-4, and TNF-α, are observed with UVR exposure. | |
Impaired Function of T-Cells |
- Immune checkpoint molecules such as PD-1, PD-L1, and CTLA-4 play a role in melanoma immune evasion. |
- These molecules lead to T-cell exhaustion and impair immune recognition of tumors. | |
- Other checkpoint molecules like TIM-3 and LAG-3 are also involved. | |
Exosomes |
- Tumor-derived exosomes have a significant role in suppressing anti-tumor immune responses. |
- They carry cargo that interferes with immune cell function and promotes tumor tolerance. | |
- Melanoma-derived exosomes express PD-L1, further suppressing the immune response. | |
Spontaneous Melanoma Models |
- Spontaneous melanoma-prone mouse models mirror immunological profiles seen in human melanoma patients. |
- These models reveal correlations between MDSCs, Tregs, TAMs, and immune activity. | |
- The balance of M1 and M2 TAMs is a prognostic indicator. | |
Defective Immune Recognition |
- Melanoma progression involves immune editing with phases of elimination, equilibrium, and escape. |
- Immunosuppressive mechanisms include reduced antigen presentation and immune checkpoint expression. | |
- Melanoma cells can downregulate MHC I and MHC II expression to evade immune detection. | |
Role of Regulatory T-Cells |
- CD4+ Tregs are upregulated in melanoma and contribute to immune suppression. |
- Tregs employ multiple mechanisms, including cytokine secretion, to inhibit the immune response. | |
- Their presence correlates with reduced cytolytic activity of immune cells. | |
Role of MDSCs in Immune Evasion |
- MDSCs play a key role in melanoma progression and immunosuppression. |
- They can differentiate into TAMs and contribute to the tumor microenvironment. | |
- MDSC levels correlate with disease progression and decreased T-cell activity. |
In conclusion, melanoma is a complex and immunologically evasive cancer that exploits various mechanisms to evade the host immune system, including the use of immune checkpoints, the release of exosomes, and the recruitment of immunosuppressive cells.
Understanding these mechanisms is essential for developing effective treatments and immunotherapies for melanoma.
Additionally, minimizing UVR exposure and promoting early diagnosis remain crucial in preventing melanoma development and improving patient outcomes.
Conflict of interests
None to declare.
Acknowledgments
Radu-Florin Fruntealată and Matei Marius contributed equally to this manuscript and thus share first authorship.
References
- 1.de Laat, van der, de Gruijl. Carcinogenesis induced by UVA (365-nm) radiation: the dose-time dependence of tumor formation in hairless mice. Carcinogenesis. 1997;18(5):1013–1020. doi: 10.1093/carcin/18.5.1013. [DOI] [PubMed] [Google Scholar]
- 2.Westerdahl J, Olsson H, Måsbäck A, Ingvar C, Jonsson N, Brandt L, Jönsson P-E, Möller T. Use of Sunbeds or Sunlamps and Malignant Melanoma in Southern Sweden. American Journal of Epidemiology. 1994;140(8):691–699. doi: 10.1093/oxfordjournals.aje.a117317. [DOI] [PubMed] [Google Scholar]
- 3.Fujimura T, Kambayashi Y, Fujisawa Y, Hidaka T, Aiba S. Tumor-Associated Macrophages: Therapeutic Targets for Skin Cancer. Frontiers in Oncology. 2018;8:3–3. doi: 10.3389/fonc.2018.00003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Wondrak GT, Roberts MJ, Cervantes-Laurean D, Jacobson MK, Jacobson EL. Proteins of the extracellular matrix are sensitizers of photo-oxidative stress in human skin cells. J Invest Dermatol. 2003;121(3):578–586. doi: 10.1046/j.1523-1747.2003.12414.x. [DOI] [PubMed] [Google Scholar]
- 5.Wondrak GT, Roberts MJ, Jacobson MK, Jacobson EL. Photosensitized growth inhibition of cultured human skin cells: mechanism and suppression of oxidative stress from solar irradiation of glycated proteins. J Invest Dermatol. 2002;119(2):489–498. doi: 10.1046/j.1523-1747.2002.01788.x. [DOI] [PubMed] [Google Scholar]
- 6.Davis LE, Shalin SC, Tackett AJ. Current state of melanoma diagnosis and treatment. Cancer biology & therapy. 2019;20(11):1366–1379. doi: 10.1080/15384047.2019.1640032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Vandyck HH, Hillen LM, Bosisio FM, van den, Zur Hausen, Winnepenninckx V. Rethinking the biology of metastatic melanoma: a holistic approach. Cancer Metastasis Rev. 2021;40(2):603–624. doi: 10.1007/s10555-021-09960-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Gutschner T, Diederichs S. The hallmarks of cancer: a long non-coding RNA point of view. RNA Biol. 2012;9(6):703–719. doi: 10.4161/rna.20481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sample A, He YY. Mechanisms and prevention of UV-induced melanoma. Photodermatol Photoimmunol Photomed. 2018;34(1):13–24. doi: 10.1111/phpp.12329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zaidi MR, Davis S, Noonan FP, Graff-Cherry C, Hawley TS, Walker RL, Feigenbaum L, Fuchs E, Lyakh L, Young HA, Hornyak TJ, Arnheiter H, Trinchieri G, Meltzer PS, De Fabo, Merlino G. Interferon-γ links ultraviolet radiation to melanomagenesis in mice. Nature. 2011;469(7331):548–553. doi: 10.1038/nature09666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Senft D, Sorolla A, Dewing A, Claps G, Lau E, Walker GJ, Ronai ZA. ATF2 alters melanocyte response and macrophage recruitment in UV-irradiated neonatal mouse skin. Pigment Cell Melanoma Res. 2015;28(4):481–484. doi: 10.1111/pcmr.12382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Tong S, Cinelli MA, El-Sayed NS, Huang H, Patel A, Silverman RB, Yang S. Inhibition of interferon-gamma-stimulated melanoma progression by targeting neuronal nitric oxide synthase (nNOS) Sci Rep. 2022;12(1):1701–1701. doi: 10.1038/s41598-022-05394-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Nasti TH, Cochran JB, Vachhani RV, McKay K, Tsuruta Y, Athar M, Timares L, Elmets CA. IL-23 Inhibits Melanoma Development by Augmenting DNA Repair and Modulating T Cell Subpopulations. The Journal of Immunology. 2017;198(2):950–961. doi: 10.4049/jimmunol.1601455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hoejberg L, Bastholt L, Schmidt H. Interleukin-6 and melanoma. Melanoma Res. 2012;22(5):327–333. doi: 10.1097/CMR.0b013e3283543d72. [DOI] [PubMed] [Google Scholar]
- 15.Sim GC, Radvanyi L. The IL-2 cytokine family in cancer immunotherapy. Cytokine & growth factor reviews. 2014;25(4):377–390. doi: 10.1016/j.cytogfr.2014.07.018. [DOI] [PubMed] [Google Scholar]
- 16.Payne R, Glenn L, Hoen H, Richards B, Smith JW, Lufkin R, Crocenzi TS, Urba WJ, Curti BD. Durable responses and reversible toxicity of high-dose interleukin-2 treatment of melanoma and renal cancer in a Community Hospital Biotherapy Program. Journal for immunotherapy of cancer. 2014;2(1):1–10. doi: 10.1186/2051-1426-2-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Butler MO, Friedlander P, Milstein MI, Mooney MM, Metzler G, Murray AP, Tanaka M, Berezovskaya A, Imataki O, Drury L. Establishment of antitumor memory in humans using in vitro-educated CD8+ T cells. Science translational medicine. 2011;3(80):80ra34–80ra34. doi: 10.1126/scitranslmed.3002207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cho D, Song H, Kim YM, Houh D, Hur DY, Park H, Yoon D, Pyun KH, Lee WJ, Kurimoto M. Endogenous interleukin-18 modulates immune escape of murine melanoma cells by regulating the expression of Fas ligand and reactive oxygen intermediates. Cancer research. 2000;60(10):2703–2709. [PubMed] [Google Scholar]
- 19.Schwarz T. Mechanisms of UV-induced immunosuppression. Keio J Med. 2005;54(4):165–171. doi: 10.2302/kjm.54.165. [DOI] [PubMed] [Google Scholar]
- 20.Kripke ML, Fisher MS. Immunologic Parameters of Ultraviolet Carcinogenesis2. JNCI: Journal of the National Cancer Institute. 1976;57(1):211–215. doi: 10.1093/jnci/57.1.211. [DOI] [PubMed] [Google Scholar]
- 21.Kripke M, Fisher M. Immunologic aspects of tumor induction by ultraviolet radiation. National Cancer Institute Monograph. 1978;(50):179–183. [PubMed] [Google Scholar]
- 22.Hart PH, Norval M. Ultraviolet radiation-induced immunosuppression and its relevance for skin carcinogenesis. Photochemical & photobiological sciences. 2018;17(12):1872–1884. doi: 10.1039/c7pp00312a. [DOI] [PubMed] [Google Scholar]
- 23.Fortner GW, Kripke ML. In vitro reactivity of splenic lymphocytes from normal and UV-irradiated mice against syngeneic UV-induced tumors. Journal of Immunology. 1977;118(4):1483–1487. [PubMed] [Google Scholar]
- 24.Sun X, Zhang N, Yin C, Zhu B, Li X. Ultraviolet radiation and melanomagenesis: from mechanism to immunotherapy. Frontiers in Oncology. 2020;10:951–951. doi: 10.3389/fonc.2020.00951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Fantin A, Vieira JM, Gestri G, Denti L, Schwarz Q, Prykhozhij S, Peri F, Wilson SW, Ruhrberg C. Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF-mediated endothelial tip cell induction. Blood. 2010;116(5):829–840. doi: 10.1182/blood-2009-12-257832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Shreedhar VK, Pride MW, Sun Y, Kripke ML, Strickland FM. Origin and characteristics of ultraviolet-B radiation-induced suppressor T lymphocytes. The Journal of Immunology. 1998;161(3):1327–1335. [PubMed] [Google Scholar]
- 27.Rivas JM, Ullrich SE. The role of IL-4, IL-10, and TNF-α in the immune suppression induced by ultraviolet radiation. Journal of Leukocyte Biology. 1994;56(6):769–775. doi: 10.1002/jlb.56.6.769. [DOI] [PubMed] [Google Scholar]
- 28.Granstein RD, Matsui MS. UV radiation-induced immunosuppression and skin cancer. Cutis. 2004;74(5 Suppl):4–9. [PubMed] [Google Scholar]
- 29.Ullrich SE. Does exposure to UV radiation induce a shift to a Th‐2‐like immune reaction. Photochemistry and photobiology. 1996;64(2):254–258. doi: 10.1111/j.1751-1097.1996.tb02454.x. [DOI] [PubMed] [Google Scholar]
- 30.Schmitt D, Walterscheid J, Ullrich S. Reversal of ultraviolet radiation‐induced immune suppression by recombinant interleukin‐12: suppression of cytokine production. Immunology. 2000;101(1):90–96. doi: 10.1046/j.1365-2567.2000.00084.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, Matsumori A, Sasayama S, Mizoguchi A, Hiai H, Minato N. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science. 2001;291(5502):319–322. doi: 10.1126/science.291.5502.319. [DOI] [PubMed] [Google Scholar]
- 32.Nishimura H, Honjo T. PD-1: an inhibitory immunoreceptor involved in peripheral tolerance. Trends in immunology. 2001;22(5):265–268. doi: 10.1016/s1471-4906(01)01888-9. [DOI] [PubMed] [Google Scholar]
- 33.Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11(2):141–151. doi: 10.1016/s1074-7613(00)80089-8. [DOI] [PubMed] [Google Scholar]
- 34.Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. The Journal of experimental medicine. 2000;192(7):1027–1034. doi: 10.1084/jem.192.7.1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, Ahmed R. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439(7077):682–687. doi: 10.1038/nature04444. [DOI] [PubMed] [Google Scholar]
- 36.Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. The Journal of Immunology. 2004;173(2):945–954. doi: 10.4049/jimmunol.173.2.945. [DOI] [PubMed] [Google Scholar]
- 37.Baumeister SH, Freeman GJ, Dranoff G, Sharpe AH. Coinhibitory Pathways in Immunotherapy for Cancer. Annual Review of Immunology. 2016;34(1):539–573. doi: 10.1146/annurev-immunol-032414-112049. [DOI] [PubMed] [Google Scholar]
- 38.Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359(6382):1350–1355. doi: 10.1126/science.aar4060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, Iwai Y, Long AJ, Brown JA, Nunes R. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nature immunology. 2001;2(3):261–268. doi: 10.1038/85330. [DOI] [PubMed] [Google Scholar]
- 40.Ghoneim HE, Fan Y, Moustaki A, Abdelsamed HA, Dash P, Dogra P, Carter R, Awad W, Neale G, Thomas PG. De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell. 2017;170(1):142–157. doi: 10.1016/j.cell.2017.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Pauken KE, Sammons MA, Odorizzi PM, Manne S, Godec J, Khan O, Drake AM, Chen Z, Sen DR, Kurachi M. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science. 2016;354(6316):1160–1165. doi: 10.1126/science.aaf2807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Pereira RM, Hogan PG, Rao A, Martinez GJ. Transcriptional and epigenetic regulation of T cell hyporesponsiveness. Journal of leukocyte biology. 2017;102(3):601–615. doi: 10.1189/jlb.2RI0317-097R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Dong W, Wu X, Ma S, Wang Y, Nalin AP, Zhu Z, Zhang J, Benson DM, He K, Caligiuri MA, Yu J. The Mechanism of Anti-PD-L1 Antibody Efficacy against PD-L1-Negative Tumors Identifies NK Cells Expressing PD-L1 as a Cytolytic Effector. Cancer Discovery. 2019;9(10):1422–1437. doi: 10.1158/2159-8290.CD-18-1259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hartley G, Regan D, Guth A, Dow S. Regulation of PD-L1 expression on murine tumor-associated monocytes and macrophages by locally produced TNF-α. Cancer Immunology Immunotherapy. 2017;66:523–535. doi: 10.1007/s00262-017-1955-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Herbst RS, Soria J-C, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, McDermott DF, Powderly JD, Gettinger SN. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515(7528):563–567. doi: 10.1038/nature14011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Hartley GP, Chow L, Ammons DT, Wheat WH, Dow SW. Programmed cell death ligand 1 (PD-L1) signaling regulates macrophage proliferation and activation. Cancer immunology research. 2018;6(10):1260–1273. doi: 10.1158/2326-6066.CIR-17-0537. [DOI] [PubMed] [Google Scholar]
- 47.Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, Rosenberg SA. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. The Journal of the American Society of Hematology. 2009;114(8):1537–1544. doi: 10.1182/blood-2008-12-195792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, Krzysiek R, Knutson KL, Daniel B, Zimmermann MC. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nature medicine. 2003;9(5):562–567. doi: 10.1038/nm863. [DOI] [PubMed] [Google Scholar]
- 49.Dammeijer F, van Gulijk, Mulder EE, Lukkes M, Klaase L, van den, van Nimwegen, Lau SP, Latupeirissa K, Schetters S. The PD-1/PD-L1-checkpoint restrains T cell immunity in tumor-draining lymph nodes. Cancer cell. 2020;38(5):685–700. doi: 10.1016/j.ccell.2020.09.001. [DOI] [PubMed] [Google Scholar]
- 50.Yearley JH, Gibson C, Yu N, Moon C, Murphy E, Juco J, Lunceford J, Cheng J, Chow LQ, Seiwert TY. PD-L2 expression in human tumors: relevance to anti-PD-1 therapy in cancer. Clinical cancer research. 2017;23(12):3158–3167. doi: 10.1158/1078-0432.CCR-16-1761. [DOI] [PubMed] [Google Scholar]
- 51.Obeid JM, Erdag G, Smolkin ME, Deacon DH, Patterson JW, Chen L, Bullock TN, Slingluff CL. PD-L1, PD-L2 and PD-1 expression in metastatic melanoma: Correlation with tumor-infiltrating immune cells and clinical outcome. Oncoimmunology. 2016;5(11):1235107–1235107. doi: 10.1080/2162402X.2016.1235107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Rodig N, Ryan T, Allen JA, Pang H, Grabie N, Chernova T, Greenfield EA, Liang SC, Sharpe AH, Lichtman AH, Freeman GJ. Endothelial expression of PD-L1 and PD-L2 down-regulates CD8+ T cell activation and cytolysis. European Journal of Immunology. 2003;33(11):3117–3126. doi: 10.1002/eji.200324270. [DOI] [PubMed] [Google Scholar]
- 53.Kaunitz GJ, Cottrell TR, Lilo M, Muthappan V, Esandrio J, Berry S, Xu H, Ogurtsova A, Anders RA, Fischer AH, Kraft S, Gerstenblith MR, Thompson CL, Honda K, Cuda JD, Eberhart CG, Handa JT, Lipson EJ, Taube JM. Melanoma subtypes demonstrate distinct PD-L1 expression profiles. Laboratory Investigation. 2017;97(9):1063–1071. doi: 10.1038/labinvest.2017.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Morales-Betanzos CA, Lee H, Gonzalez Ericsson, Balko JM, Johnson DB, Zimmerman LJ, Liebler DC. Quantitative Mass Spectrometry Analysis of PD-L1 Protein Expression, N-glycosylation and Expression Stoichiometry with PD-1 and PD-L2 in Human Melanoma. Mol Cell Proteomics. 2017;16(10):1705–1717. doi: 10.1074/mcp.RA117.000037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Li CW, Lim SO, Xia W, Lee HH, Chan LC, Kuo CW, Khoo KH, Chang SS, Cha JH, Kim T, Hsu JL, Wu Y, Hsu JM, Yamaguchi H, Ding Q, Wang Y, Yao J, Lee CC, Wu HJ, Sahin AA, Allison JP, Yu D, Hortobagyi GN, Hung MC. Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity. Nat Commun. 2016;7:12632–12632. doi: 10.1038/ncomms12632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Maher CM, Thomas JD, Haas DA, Longen CG, Oyer HM, Tong JY, Kim FJ. Small-Molecule Sigma1 Modulator Induces Autophagic Degradation of PD-L1. Mol Cancer Res. 2018;16(2):243–255. doi: 10.1158/1541-7786.MCR-17-0166. [DOI] [PubMed] [Google Scholar]
- 57.Hsu J-M, Li C-W, Lai Y-J, Hung M-C. Posttranslational Modifications of PD-L1 and Their Applications in Cancer Therapy. Cancer Research. 2018;78(22):6349–6353. doi: 10.1158/0008-5472.CAN-18-1892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zerdes I, Matikas A, Bergh J, Rassidakis GZ, Foukakis T. Genetic, transcriptional and post-translational regulation of the programmed death protein ligand 1 in cancer: biology and clinical correlations. Oncogene. 2018;37(34):4639–4661. doi: 10.1038/s41388-018-0303-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, Chen S, Klein AP, Pardoll DM, Topalian SL, Chen L. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med. 2012;4(127):127–137. doi: 10.1126/scitranslmed.3003689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, Lennon VA, Celis E, Chen L. Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nature Medicine. 2002;8(8):793–800. doi: 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
- 61.Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, Zaretsky JM, Sun L, Hugo W, Wang X, Parisi G, Saus CP, Torrejon DY, Graeber TG, Comin-Anduix B, Hu-Lieskovan S, Damoiseaux R, Lo RS, Ribas A. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep. 2017;19(6):1189–1201. doi: 10.1016/j.celrep.2017.04.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Thiem A, Hesbacher S, Kneitz H, di Primio, Heppt MV, Hermanns HM, Goebeler M, Meierjohann S, Houben R, Schrama D. IFN-gamma-induced PD-L1 expression in melanoma depends on p53 expression. Journal of Experimental & Clinical Cancer Research. 2019;38(1):397–397. doi: 10.1186/s13046-019-1403-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Casey SC, Tong L, Li Y, Do R, Walz S, Fitzgerald KN, Gouw AM, Baylot V, Gütgemann I, Eilers M, Felsher DW. MYC regulates the antitumor immune response through CD47 and PD-L1. Science. 2016;352(6282):227–231. doi: 10.1126/science.aac9935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Atefi M, Avramis E, Lassen A, Wong DJ, Robert L, Foulad D, Cerniglia M, Titz B, Chodon T, Graeber TG, Comin-Anduix B, Ribas A. Effects of MAPK and PI3K pathways on PD-L1 expression in melanoma. Clin Cancer Res. 2014;20(13):3446–3457. doi: 10.1158/1078-0432.CCR-13-2797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Jiang X, Zhou J, Giobbie-Hurder A, Wargo J, Hodi FS. The activation of MAPK in melanoma cells resistant to BRAF inhibition promotes PD-L1 expression that is reversible by MEK and PI3K inhibition. Clin Cancer Res. 2013;19(3):598–609. doi: 10.1158/1078-0432.CCR-12-2731. [DOI] [PubMed] [Google Scholar]
- 66.Alcázar I, Marqués M, Kumar A, Hirsch E, Wymann M, Carrera AC, Barber DF. Phosphoinositide 3-kinase gamma participates in T cell receptor-induced T cell activation. J Exp Med. 2007;204(12):2977–2987. doi: 10.1084/jem.20070366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.D'Souza WN, Chang C-F, Fischer AM, Li M, Hedrick SM. The Erk2 MAPK Regulates CD8 T Cell Proliferation and Survival1. The Journal of Immunology. 2008;181(11):7617–7629. doi: 10.4049/jimmunol.181.11.7617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Adachi K, Davis MM. T-cell receptor ligation induces distinct signaling pathways in naïve vs. antigen-experienced T cells. Proceedings of the National Academy of Sciences. 2011;108(4):1549–1554. doi: 10.1073/pnas.1017340108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Boussiotis VA, Chatterjee P, Li L. Biochemical Signaling of PD-1 on T Cells and Its Functional Implications. The Cancer Journal. 2014;20(4):265–271. doi: 10.1097/PPO.0000000000000059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Arasanz H, Gato-Cañas M, Zuazo M, Ibañez-Vea M, Breckpot K, Kochan G, Escors D. PD1 signal transduction pathways in T cells. Oncotarget. 2017;8(31):51936–51945. doi: 10.18632/oncotarget.17232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Parry RV, Chemnitz JM, Frauwirth KA, Lanfranco AR, Braunstein I, Kobayashi SV, Linsley PS, Thompson CB, Riley JL. CTLA-4 and PD-1 Receptors Inhibit T-Cell Activation by Distinct Mechanisms. Molecular and cellular biology. 2005;25(21):9543–9553. doi: 10.1128/MCB.25.21.9543-9553.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Patsoukis N, Duke-Cohan JS, Chaudhri A, Aksoylar H-I, Wang Q, Council A, Berg A, Freeman GJ, Boussiotis VA. Interaction of SHP-2 SH2 domains with PD-1 ITSM induces PD-1 dimerization and SHP-2 activation. Communications Biology. 2020;3(1):128–128. doi: 10.1038/s42003-020-0845-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Quigley M, Pereyra F, Nilsson B, Porichis F, Fonseca C, Eichbaum Q, Julg B, Jesneck JL, Brosnahan K, Imam S, Russell K, Toth I, Piechocka-Trocha A, Dolfi D, Angelosanto J, Crawford A, Shin H, Kwon DS, Zupkosky J, Francisco L, Freeman GJ, Wherry EJ, Kaufmann DE, Walker BD, Ebert B, Haining WN. Transcriptional analysis of HIV-specific CD8+ T cells shows that PD-1 inhibits T cell function by upregulating BATF. Nature Medicine. 2010;16(10):1147–1151. doi: 10.1038/nm.2232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Gibbons RM, Liu X, Pulko V, Harrington SM, Krco CJ, Kwon ED, Dong H. B7-H1 limits the entry of effector CD8(+) T cells to the memory pool by upregulating Bim. Oncoimmunology. 2012;1(7):1061–1073. doi: 10.4161/onci.20850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Fife BT, Pauken KE, Eagar TN, Obu T, Wu J, Tang Q, Azuma M, Krummel MF, Bluestone JA. Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal. Nature immunology. 2009;10(11):1185–1192. doi: 10.1038/ni.1790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Patsoukis N, Bardhan K, Chatterjee P, Sari D, Liu B, Bell LN, Karoly ED, Freeman GJ, Petkova V, Seth P, Li L, Boussiotis VA. PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat Commun. 2015;6:6692–6692. doi: 10.1038/ncomms7692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, Hashimoto-Tane A, Azuma M, Saito T. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med. 2012;209(6):1201–1217. doi: 10.1084/jem.20112741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Okazaki T, Maeda A, Nishimura H, Kurosaki T, Honjo T. PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc Natl Acad Sci U S A. 2001;98(24):13866–13871. doi: 10.1073/pnas.231486598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Patsoukis N, Li L, Sari D, Petkova V, Boussiotis VA. PD-1 Increases PTEN Phosphatase Activity While Decreasing PTEN Protein Stability by Inhibiting Casein Kinase 2. Molecular and cellular biology. 2013;33(16):3091–3098. doi: 10.1128/MCB.00319-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Qin W, Hu L, Zhang X, Jiang S, Li J, Zhang Z, Wang X. The Diverse Function of PD-1/PD-L Pathway Beyond Cancer. Frontiers in immunology. 2019;10:2298–2298. doi: 10.3389/fimmu.2019.02298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Leclerc M, Voilin E, Gros G, Corgnac S, de Montpréville, Validire P, Bismuth G, Mami-Chouaib F. Regulation of antitumour CD8 T-cell immunity and checkpoint blockade immunotherapy by Neuropilin-1. Nature Communications. 2019;10(1):3345–3345. doi: 10.1038/s41467-019-11280-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Tivol EA, Borriello F, Schweitzer AN, Lynch WP, Bluestone JA, Sharpe AH. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity. 1995;3(5):541–547. doi: 10.1016/1074-7613(95)90125-6. [DOI] [PubMed] [Google Scholar]
- 83.Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, Lee KP, Thompson CB, Griesser H, Mak TW. Lymphoproliferative Disorders with Early Lethality in Mice Deficient in Ctla-4. Science. 1995;270(5238):985–988. doi: 10.1126/science.270.5238.985. [DOI] [PubMed] [Google Scholar]
- 84.Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271(5256):1734–1736. doi: 10.1126/science.271.5256.1734. [DOI] [PubMed] [Google Scholar]
- 85.Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med. 2010;207(10):2187–2194. doi: 10.1084/jem.20100643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Fourcade J, Sun Z, Benallaoua M, Guillaume P, Luescher IF, Sander C, Kirkwood JM, Kuchroo V, Zarour HM. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med. 2010;207(10):2175–2186. doi: 10.1084/jem.20100637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Grosso JF, Kelleher CC, Harris TJ, Maris CH, Hipkiss EL, De Marzo, Anders R, Netto G, Getnet D, Bruno TC, Goldberg MV, Pardoll DM, Drake CG. LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. The Journal of Clinical Investigation. 2007;117(11):3383–3392. doi: 10.1172/JCI31184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, Hipkiss EL, Ravi S, Kowalski J, Levitsky HI, Powell JD, Pardoll DM, Drake CG, Vignali DA. Role of LAG-3 in regulatory T cells. Immunity. 2004;21(4):503–513. doi: 10.1016/j.immuni.2004.08.010. [DOI] [PubMed] [Google Scholar]
- 89.Lines JL, Pantazi E, Mak J, Sempere LF, Wang L, O'Connell S, Ceeraz S, Suriawinata AA, Yan S, Ernstoff MS, Noelle R. VISTA Is an Immune Checkpoint Molecule for Human T Cells. Cancer Research. 2014;74(7):1924–1932. doi: 10.1158/0008-5472.CAN-13-1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Sansom DM. CD28, CTLA-4 and their ligands: who does what and to whom. Immunology. 2000;101(2):169–177. doi: 10.1046/j.1365-2567.2000.00121.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Azuma M, Ito D, Yagita H, Okumura K, Phillips JH, Lanier LL, Somoza C. B70 antigen is a second ligand for CTLA-4 and CD28. Nature. 1993;366(6450):76–79. doi: 10.1038/366076a0. [DOI] [PubMed] [Google Scholar]
- 92.Greene JL, Leytze GM, Emswiler J, Peach R, Bajorath J, Cosand W, Linsley PS. Covalent dimerization of CD28/CTLA-4 and oligomerization of CD80/CD86 regulate T cell costimulatory interactions. J Biol Chem. 1996;271(43):26762–26771. doi: 10.1074/jbc.271.43.26762. [DOI] [PubMed] [Google Scholar]
- 93.Linsley PS, Brady W, Urnes M, Grosmaire LS, Damle NK, Ledbetter JA. CTLA-4 is a second receptor for the B cell activation antigen B7. J Exp Med. 1991;174(3):561–569. doi: 10.1084/jem.174.3.561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.van der, Bodian DL, Daenke S, Linsley P, Davis SJ. CD80 (B7-1) binds both CD28 and CTLA-4 with a low affinity and very fast kinetics. J Exp Med. 1997;185(3):393–403. doi: 10.1084/jem.185.3.393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kleffel S, Posch C, Barthel SR, Mueller H, Schlapbach C, Guenova E, Elco CP, Lee N, Juneja VR, Zhan Q, Lian CG, Thomi R, Hoetzenecker W, Cozzio A, Dummer R, Mihm MC, Jr undefined. Flaherty KT, Frank MH, Murphy GF, Sharpe AH, Kupper TS, Schatton T. Melanoma Cell-Intrinsic PD-1 Receptor Functions Promote Tumor Growth. Cell. 2015;162(6):1242–1256. doi: 10.1016/j.cell.2015.08.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Clark CA, Gupta HB, Sareddy G, Pandeswara S, Lao S, Yuan B, Drerup JM, Padron A, Conejo-Garcia J, Murthy K, Liu Y, Turk MJ, Thedieck K, Hurez V, Li R, Vadlamudi R, Curiel TJ. Tumor-Intrinsic PD-L1 Signals Regulate Cell Growth, Pathogenesis, and Autophagy in Ovarian Cancer and Melanoma. Cancer Res. 2016;76(23):6964–6974. doi: 10.1158/0008-5472.CAN-16-0258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Mo X, Zhang H, Preston S, Martin K, Zhou B, Vadalia N, Gamero AM, Soboloff J, Tempera I, Zaidi MR. Interferon-γ Signaling in Melanocytes and Melanoma Cells Regulates Expression of CTLA-4. Cancer Res. 2018;78(2):436–450. doi: 10.1158/0008-5472.CAN-17-1615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Contardi E, Palmisano GL, Tazzari PL, Martelli AM, Falà F, Fabbi M, Kato T, Lucarelli E, Donati D, Polito L, Bolognesi A, Ricci F, Salvi S, Gargaglione V, Mantero S, Alberghini M, Ferrara GB, Pistillo MP. CTLA-4 is constitutively expressed on tumor cells and can trigger apoptosis upon ligand interaction. International Journal of Cancer. 2005;117(4):538–550. doi: 10.1002/ijc.21155. [DOI] [PubMed] [Google Scholar]
- 99.Seliger B, Maio M, Cuaia O, Calabro L. Expression and function of CTLA4 in melanoma. Journal of Clinical Oncology. 2013;31(15):20040–20040. [Google Scholar]
- 100.Pistillo MP, Carosio R, Grillo F, Fontana V, Mastracci L, Morabito A, Banelli B, Tanda E, Cecchi F, Dozin B, Gualco M, Salvi S, Spagnolo F, Poggi A, Queirolo P. Phenotypic characterization of tumor CTLA-4 expression in melanoma tissues and its possible role in clinical response to Ipilimumab. Clin Immunol. 2020;215:108428–108428. doi: 10.1016/j.clim.2020.108428. [DOI] [PubMed] [Google Scholar]
- 101.Isola AL, Eddy K, Chen S. Biology, Therapy and Implications of Tumor Exosomes in the Progression of Melanoma. Cancers. 2016;8(12):110–110. doi: 10.3390/cancers8120110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hood JL, San RS, Wickline SA. Exosomes Released by Melanoma Cells Prepare Sentinel Lymph Nodes for Tumor Metastasis. Cancer Research. 2011;71(11):3792–3801. doi: 10.1158/0008-5472.CAN-10-4455. [DOI] [PubMed] [Google Scholar]
- 103.Hu L, Wickline SA, Hood JL. Magnetic resonance imaging of melanoma exosomes in lymph nodes. Magnetic Resonance in Medicine. 2015;74(1):266–271. doi: 10.1002/mrm.25376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Düchler M, Czernek L, Peczek L, Cypryk W, Sztiller-Sikorska M, Czyz M. Melanoma-Derived Extracellular Vesicles Bear the Potential for the Induction of Antigen-Specific Tolerance. Cells. 2019;8(7):665–665. doi: 10.3390/cells8070665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Taylor DD, Gerçel-Taylor C. Tumour-derived exosomes and their role in cancer-associated T-cell signalling defects. British Journal of Cancer. 2005;92(2):305–311. doi: 10.1038/sj.bjc.6602316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Muller L, Mitsuhashi M, Simms P, Gooding WE, Whiteside TL. Tumor-derived exosomes regulate expression of immune function-related genes in human T cell subsets. Scientific Reports. 2016;6(1):20254–20254. doi: 10.1038/srep20254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Sharma P, Diergaarde B, Ferrone S, Kirkwood JM, Whiteside TL. Melanoma cell-derived exosomes in plasma of melanoma patients suppress functions of immune effector cells. Scientific Reports. 2020;10(1):92–92. doi: 10.1038/s41598-019-56542-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Liu C, Yu S, Zinn K, Wang J, Zhang L, Jia Y, Kappes JC, Barnes S, Kimberly RP, Grizzle WE, Zhang HG. Murine mammary carcinoma exosomes promote tumor growth by suppression of NK cell function. J Immunol. 2006;176(3):1375–1385. doi: 10.4049/jimmunol.176.3.1375. [DOI] [PubMed] [Google Scholar]
- 109.Xiang X, Poliakov A, Liu C, Liu Y, Deng ZB, Wang J, Cheng Z, Shah SV, Wang GJ, Zhang L, Grizzle WE, Mobley J, Zhang HG. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer. 2009;124(11):2621–2633. doi: 10.1002/ijc.24249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Fujimura T, Kambayashi Y, Fujisawa Y, Hidaka T, Aiba S. Tumor-Associated Macrophages: Therapeutic Targets for Skin Cancer. Front Oncol. 2018;8:3–3. doi: 10.3389/fonc.2018.00003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Movahedi K, Laoui D, Gysemans C, Baeten M, Stangé G, Van den, Mack M, Pipeleers D, In't Veld, De Baetselier, Van Ginderachter. Different Tumor Microenvironments Contain Functionally Distinct Subsets of Macrophages Derived from Ly6C(high) Monocytes. Cancer Research. 2010;70(14):5728–5739. doi: 10.1158/0008-5472.CAN-09-4672. [DOI] [PubMed] [Google Scholar]
- 112.Palazón A, Aragonés J, Morales-Kastresana A, de Landázuri, Melero I. Molecular pathways: hypoxia response in immune cells fighting or promoting cancer. Clin Cancer Res. 2012;18(5):1207–1213. doi: 10.1158/1078-0432.CCR-11-1591. [DOI] [PubMed] [Google Scholar]
- 113.Bardi GT, Smith MA, Hood JL. Melanoma exosomes promote mixed M1 and M2 macrophage polarization. Cytokine. 2018;105:63–72. doi: 10.1016/j.cyto.2018.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Chen G, Huang AC, Zhang W, Zhang G, Wu M, Xu W, Yu Z, Yang J, Wang B, Sun H, Xia H, Man Q, Zhong W, Antelo LF, Wu B, Xiong X, Liu X, Guan L, Li T, Liu S, Yang R, Lu Y, Dong L, McGettigan S, Somasundaram R, Radhakrishnan R, Mills G, Lu Y, Kim J, Chen YH, Dong H, Zhao Y, Karakousis GC, Mitchell TC, Schuchter LM, Herlyn M, Wherry EJ, Xu X, Guo W. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 2018;560(7718):382–386. doi: 10.1038/s41586-018-0392-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Poggio M, Hu T, Pai CC, Chu B, Belair CD, Chang A, Montabana E, Lang UE, Fu Q, Fong L, Blelloch R. Suppression of Exosomal PD-L1 Induces Systemic Anti-tumor Immunity and Memory. Cell. 2019;177(2):414–427. doi: 10.1016/j.cell.2019.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Alb M, Sie C, Adam C, Chen S, Becker JC, Schrama D. Cellular and cytokine-dependent immunosuppressive mechanisms of grm1-transgenic murine melanoma. Cancer Immunology Immunotherapy. 2012;61(12):2239–2249. doi: 10.1007/s00262-012-1290-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Mairhofer DG, Ortner D, Tripp CH, Schaffenrath S, Fleming V, Heger L, Komenda K, Reider D, Dudziak D, Chen S, Becker JC, Flacher V, Stoitzner P. Impaired gp100-Specific CD8(+) T-Cell Responses in the Presence of Myeloid-Derived Suppressor Cells in a Spontaneous Mouse Melanoma Model. J Invest Dermatol. 2015;135(11):2785–2793. doi: 10.1038/jid.2015.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Prokopi A, Tripp CH, Tummers B, Komenda K, Hutter K, Cappellano G, Bellmann L, Efremova M, Trajanoski Z, Chen S, Clausen BE, Green DR, Stoitzner P. Abstract A102: Rescue of lost skin dendritic cells in melanoma is key for the resuscitation of antitumor T-cell responses. Cancer Immunology Research. 2019;7(2):102–102. [Google Scholar]
- 119.Tucci M, Passarelli A, Mannavola F, Felici C, Stucci LS, Cives M, Silvestris F. Immune System Evasion as Hallmark of Melanoma Progression: The Role of Dendritic Cells. Front Oncol. 2019;9:1148–1148. doi: 10.3389/fonc.2019.01148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Escors D. Tumour immunogenicity, antigen presentation and immunological barriers in cancer immunotherapy. New J Sci. 2014 doi: 10.1155/2014/734515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Chattopadhyay G, Shevach EM. Antigen-Specific Induced T Regulatory Cells Impair Dendritic Cell Function via an IL-10/MARCH1-Dependent Mechanism. The Journal of Immunology. 2013;191(12):5875–5884. doi: 10.4049/jimmunol.1301693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Mittal SK, Roche PA. Suppression of antigen presentation by IL-10. Curr Opin Immunol. 2015;34:22–27. doi: 10.1016/j.coi.2014.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Groux H, Bigler M, de Vries, Roncarolo MG. Inhibitory and stimulatory effects of IL-10 on human CD8+ T cells. J Immunol. 1998;160(7):3188–3193. [PubMed] [Google Scholar]
- 124.Laurent S, Carrega P, Saverino D, Piccioli P, Camoriano M, Morabito A, Dozin B, Fontana V, Simone R, Mortara L, Mingari MC, Ferlazzo G, Pistillo MP. CTLA-4 is expressed by human monocyte-derived dendritic cells and regulates their functions. Hum Immunol. 2010;71(10):934–941. doi: 10.1016/j.humimm.2010.07.007. [DOI] [PubMed] [Google Scholar]
- 125.Yao S, Wang S, Zhu Y, Luo L, Zhu G, Flies S, Xu H, Ruff W, Broadwater M, Choi IH, Tamada K, Chen L. PD-1 on dendritic cells impedes innate immunity against bacterial infection. Blood. 2009;113(23):5811–5818. doi: 10.1182/blood-2009-02-203141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Hobo W, Maas F, Adisty N, de Witte, Schaap N, van der, Dolstra H. siRNA silencing of PD-L1 and PD-L2 on dendritic cells augments expansion and function of minor histocompatibility antigen-specific CD8+ T cells. Blood. 2010;116(22):4501–4511. doi: 10.1182/blood-2010-04-278739. [DOI] [PubMed] [Google Scholar]
- 127.Oh SA, Wu D-C, Cheung J, Navarro A, Xiong H, Cubas R, Totpal K, Chiu H, Wu Y, Comps-Agrar L, Leader AM, Merad M, Roose-Germa M, Warming S, Yan M, Kim JM, Rutz S, Mellman I. PD-L1 expression by dendritic cells is a key regulator of T-cell immunity in cancer. Nature Cancer. 2020;1(7):681–691. doi: 10.1038/s43018-020-0075-x. [DOI] [PubMed] [Google Scholar]
- 128.Wang XB, Fan ZZ, Anton D, Vollenhoven AV, Ni ZH, Chen XF, Lefvert AK. CTLA4 is expressed on mature dendritic cells derived from human monocytes and influences their maturation and antigen presentation. BMC Immunology. 2011;12(1):21–21. doi: 10.1186/1471-2172-12-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Zhang W, Song Z, Xiao J, Liu X, Luo Y, Yang Z, Luo R, Li A. Blocking the PD-1/PD-L1 axis in dendritic cell-stimulated Cytokine-Induced Killer Cells with pembrolizumab enhances their therapeutic effects against hepatocellular carcinoma. Journal of Cancer. 2019;10(11):2578–2587. doi: 10.7150/jca.26961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Donia M, Andersen R, Kjeldsen JW, Fagone P, Munir S, Nicoletti F, Andersen MH, thor Straten, Svane IM. Aberrant Expression of MHC Class II in Melanoma Attracts Inflammatory Tumor-Specific CD4+ T- Cells, Which Dampen CD8+ T-cell Antitumor Reactivity. Cancer Research. 2015;75(18):3747–3759. doi: 10.1158/0008-5472.CAN-14-2956. [DOI] [PubMed] [Google Scholar]
- 131.Jayasingam SD, Citartan M, Thang TH, Mat Zin, Ang KC, Ch'ng ES. Evaluating the Polarization of Tumor-Associated Macrophages Into M1 and M2 Phenotypes in Human Cancer Tissue: Technicalities and Challenges in Routine Clinical Practice. Front Oncol. 2019;9:1512–1512. doi: 10.3389/fonc.2019.01512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Tcyganov E, Mastio J, Chen E, Gabrilovich DI. Plasticity of myeloid-derived suppressor cells in cancer. Curr Opin Immunol. 2018;51:76–82. doi: 10.1016/j.coi.2018.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Laoui D, Van Overmeire, Di Conza, Aldeni C, Keirsse J, Morias Y, Movahedi K, Houbracken I, Schouppe E, Elkrim Y, Karroum O, Jordan B, Carmeliet P, Gysemans C, De Baetselier, Mazzone M, Van Ginderachter. Tumor Hypoxia Does Not Drive Differentiation of Tumor-Associated Macrophages but Rather Fine-Tunes the M2-like Macrophage Population. Cancer Research. 2014;74(1):24–30. doi: 10.1158/0008-5472.CAN-13-1196. [DOI] [PubMed] [Google Scholar]
- 134.Salmi S, Siiskonen H, Sironen R, Tyynelä-Korhonen K, Hirschovits-Gerz B, Valkonen M, Auvinen P, Pasonen-Seppänen S. The number and localization of CD68+ and CD163+ macrophages in different stages of cutaneous melanoma. Melanoma Res. 2019;29(3):237–247. doi: 10.1097/CMR.0000000000000522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Falleni M, Savi F, Tosi D, Agape E, Cerri A, Moneghini L, Bulfamante GP. M1 and M2 macrophages' clinicopathological significance in cutaneous melanoma. Melanoma Res. 2017;27(3):200–210. doi: 10.1097/CMR.0000000000000352. [DOI] [PubMed] [Google Scholar]
- 136.Herwig MC, Bergstrom C, Wells JR, Höller T, Grossniklaus HE. M2/M1 ratio of tumor associated macrophages and PPAR-gamma expression in uveal melanomas with class 1 and class 2 molecular profiles. Exp Eye Res. 2013;107:52–58. doi: 10.1016/j.exer.2012.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Mäkitie T, Summanen P, Tarkkanen A, Kivelä T. Tumor-infiltrating macrophages (CD68(+) cells) and prognosis in malignant uveal melanoma. Invest Ophthalmol Vis Sci. 2001;42(7):1414–1421. [PubMed] [Google Scholar]
- 138.Ley K. M1 Means Kill; M2 Means Heal. The Journal of Immunology. 2017;199(7):2191–2193. doi: 10.4049/jimmunol.1701135. [DOI] [PubMed] [Google Scholar]
- 139.Buscher K, Ehinger E, Gupta P, Pramod AB, Wolf D, Tweet G, Pan C, Mills CD, Lusis AJ, Ley K. Natural variation of macrophage activation as disease-relevant phenotype predictive of inflammation and cancer survival. Nature Communications. 2017;8(1):16041–16041. doi: 10.1038/ncomms16041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Van Overmeire, Stijlemans B, Heymann F, Keirsse J, Morias Y, Elkrim Y, Brys L, Abels C, Lahmar Q, Ergen C, Vereecke L, Tacke F, De Baetselier, Van Ginderachter, Laoui D. M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization in the Tumor Microenvironment. Cancer Research. 2016;76(1):35–42. doi: 10.1158/0008-5472.CAN-15-0869. [DOI] [PubMed] [Google Scholar]
- 141.Georgoudaki AM, Prokopec KE, Boura VF, Hellqvist E, Sohn S, Östling J, Dahan R, Harris RA, Rantalainen M, Klevebring D, Sund M, Brage SE, Fuxe J, Rolny C, Li F, Ravetch JV, Karlsson MC. Reprogramming Tumor-Associated Macrophages by Antibody Targeting Inhibits Cancer Progression and Metastasis. Cell Rep. 2016;15(9):2000–2011. doi: 10.1016/j.celrep.2016.04.084. [DOI] [PubMed] [Google Scholar]
- 142.Kaneda MM, Messer KS, Ralainirina N, Li H, Leem CJ, Gorjestani S, Woo G, Nguyen AV, Figueiredo CC, Foubert P, Schmid MC, Pink M, Winkler DG, Rausch M, Palombella VJ, Kutok J, McGovern K, Frazer KA, Wu X, Karin M, Sasik R, Cohen EE, Varner JA. PI3Kγ is a molecular switch that controls immune suppression. Nature. 2016;539(7629):437–442. doi: 10.1038/nature19834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Vergadi E, Ieronymaki E, Lyroni K, Vaporidi K, Tsatsanis C. Akt Signaling Pathway in Macrophage Activation and M1/M2 Polarization. J Immunol. 2017;198(3):1006–1014. doi: 10.4049/jimmunol.1601515. [DOI] [PubMed] [Google Scholar]
- 144.Mouchemore KA, Sampaio NG, Murrey MW, Stanley ER, Lannutti BJ, Pixley FJ. Specific inhibition of PI3K p110δ inhibits CSF-1-induced macrophage spreading and invasive capacity. The FEBS Journal. 2013;280(21):5228–5236. doi: 10.1111/febs.12316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Huang SC, Smith AM, Everts B, Colonna M, Pearce EL, Schilling JD, Pearce EJ. Metabolic Reprogramming Mediated by the mTORC2-IRF4 Signaling Axis Is Essential for Macrophage Alternative Activation. Immunity. 2016;45(4):817–830. doi: 10.1016/j.immuni.2016.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Arredouani MS. Is the scavenger receptor MARCO a new immune checkpoint? OncoImmunology. 2014;3(10):e955709–e955709. doi: 10.4161/21624011.2014.955709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Mills CD, Kincaid K, Alt JM, Heilman MJ, Hill AM. M-1/M-2 Macrophages and the Th1/Th2 Paradigm1. The Journal of Immunology. 2000;164(12):6166–6173. doi: 10.4049/jimmunol.164.12.6166. [DOI] [PubMed] [Google Scholar]
- 148.Nevala WK, Vachon CM, Leontovich AA, Scott CG, Thompson MA, Markovic SN, Center ftMSGotMCC. Evidence of Systemic Th2-Driven Chronic Inflammation in Patients with Metastatic Melanoma. Clinical Cancer Research. 2009;15(6):1931–1939. doi: 10.1158/1078-0432.CCR-08-1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Lauerova L, Dusek L, Simickova M, Kocák I, Vagundová M, Zaloudík J, Kovarík J. Malignant melanoma associates with Th1/Th2 imbalance that coincides with disease progression and immunotherapy response. Neoplasma. 2002;49(3):159–166. [PubMed] [Google Scholar]
- 150.Enninga EA, Nevala WK, Holtan SG, Leontovich AA, Markovic SN. Galectin-9 modulates immunity by promoting Th2/M2 differentiation and impacts survival in patients with metastatic melanoma. Melanoma Res. 2016;26(5):429–441. doi: 10.1097/CMR.0000000000000281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nature immunology. 2010;11(10):889–896. doi: 10.1038/ni.1937. [DOI] [PubMed] [Google Scholar]
- 152.Massi D, Marconi C, Franchi A, Bianchini F, Paglierani M, Ketabchi S, Miracco C, Santucci M, Calorini L. Arginine metabolism in tumor-associated macrophages in cutaneous malignant melanoma: evidence from human and experimental tumors. Hum Pathol. 2007;38(10):1516–1525. doi: 10.1016/j.humpath.2007.02.018. [DOI] [PubMed] [Google Scholar]
- 153.Wang H, Yang L, Wang D, Zhang Q, Zhang L. Pro-tumor activities of macrophages in the progression of melanoma. Human Vaccines & Immunotherapeutics. 2017;13(7):1556–1562. doi: 10.1080/21645515.2017.1312043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Chen P, Huang Y, Bong R, Ding Y, Song N, Wang X, Song X, Luo Y. Tumor-Associated Macrophages Promote Angiogenesis and Melanoma Growth via Adrenomedullin in a Paracrine and Autocrine Manner. Clinical Cancer Research. 2011;17(23):7230–7239. doi: 10.1158/1078-0432.CCR-11-1354. [DOI] [PubMed] [Google Scholar]
- 155.Vignali DAA, Collison LW, Workman CJ. How regulatory T cells work. Nature Reviews Immunology. 2008;8(7):523–532. doi: 10.1038/nri2343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Viguier M, Lemaître F, Verola O, Cho M-S, Gorochov G, Dubertret L, Bachelez H, Kourilsky P, Ferradini L. Foxp3 Expressing CD4+CD25high Regulatory T Cells Are Overrepresented in Human Metastatic Melanoma Lymph Nodes and Inhibit the Function of Infiltrating T Cells1. The Journal of Immunology. 2004;173(2):1444–1453. doi: 10.4049/jimmunol.173.2.1444. [DOI] [PubMed] [Google Scholar]
- 157.Gray CP, Arosio P, Hersey P. Association of increased levels of heavy-chain ferritin with increased CD4+ CD25+ regulatory T-cell levels in patients with melanoma. Clin Cancer Res. 2003;9(7):2551–2559. [PubMed] [Google Scholar]
- 158.Fujii H, Josse J, Tanioka M, Miyachi Y, Husson F, Ono M. Regulatory T Cells in Melanoma Revisited by a Computational Clustering of FOXP3+ T Cell Subpopulations. J Immunol. 2016;196(6):2885–2892. doi: 10.4049/jimmunol.1402695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Magnuson AM, Kiner E, Ergun A, Park JS, Asinovski N, Ortiz-Lopez A, Kilcoyne A, Paoluzzi-Tomada E, Weissleder R, Mathis D, Benoist C. Identification and validation of a tumor-infiltrating Treg transcriptional signature conserved across species and tumor types. Proc Natl Acad Sci U S A. 2018;115(45):10672–10681. doi: 10.1073/pnas.1810580115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Ladányi A, Mohos A, Somlai B, Liszkay G, Gilde K, Fejos Z, Gaudi I, Tímár J. FOXP3+ cell density in primary tumor has no prognostic impact in patients with cutaneous malignant melanoma. Pathol Oncol Res. 2010;16(3):303–309. doi: 10.1007/s12253-010-9254-x. [DOI] [PubMed] [Google Scholar]
- 161.Leslie C, Bowyer SE, White A, Grieu-Iacopetta F, Trevenen M, Iacopetta B, Amanuel B, Millward M. FOXP3+ T regulatory lymphocytes in primary melanoma are associated with BRAF mutation but not with response to BRAF inhibitor. Pathology. 2015;47(6):557–563. doi: 10.1097/PAT.0000000000000314. [DOI] [PubMed] [Google Scholar]
- 162.Klages K, Mayer CT, Lahl K, Loddenkemper C, Teng MWL, Ngiow SF, Smyth MJ, Hamann A, Huehn J, Sparwasser T. Selective Depletion of Foxp3+ Regulatory T Cells Improves Effective Therapeutic Vaccination against Established Melanoma. Cancer Research. 2010;70(20):7788–7799. doi: 10.1158/0008-5472.CAN-10-1736. [DOI] [PubMed] [Google Scholar]
- 163.Rasku MA, Clem AL, Telang S, Taft B, Gettings K, Gragg H, Cramer D, Lear SC, McMasters KM, Miller DM, Chesney J. Transient T cell depletion causes regression of melanoma metastases. Journal of Translational Medicine. 2008;6(1):12–12. doi: 10.1186/1479-5876-6-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Gray CP, Arosio P, Hersey P. Heavy chain ferritin activates regulatory T cells by induction of changes in dendritic cells. Blood. 2002;99(9):3326–3334. doi: 10.1182/blood.v99.9.3326. [DOI] [PubMed] [Google Scholar]
- 165.Deng G. Tumor-infiltrating regulatory T cells: origins and features. Am J Clin Exp Immunol. 2018;7(5):81–87. [PMC free article] [PubMed] [Google Scholar]
- 166.Baumgartner J, Wilson C, Palmer B, Richter D, Banerjee A, McCarter M. Melanoma induces immunosuppression by up-regulating FOXP3(+) regulatory T cells. J Surg Res. 2007;141(1):72–77. doi: 10.1016/j.jss.2007.03.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Kawamoto H, Minato N. Myeloid cells. Int J Biochem Cell Biol. 2004;36(8):1374–1379. doi: 10.1016/j.biocel.2004.01.020. [DOI] [PubMed] [Google Scholar]
- 168.Condamine T, Ramachandran I, Youn JI, Gabrilovich DI. Regulation of tumor metastasis by myeloid-derived suppressor cells. Annu Rev Med. 2015;66:97–110. doi: 10.1146/annurev-med-051013-052304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Gabrilovich DI. Myeloid-Derived Suppressor Cells. Cancer Immunology Research. 2017;5(1):3–8. doi: 10.1158/2326-6066.CIR-16-0297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Umansky V, Sevko A, Gebhardt C, Utikal J. Myeloid-derived suppressor cells in malignant melanoma. J Dtsch Dermatol Ges. 2014;12(11):1021–1027. doi: 10.1111/ddg.12411. [DOI] [PubMed] [Google Scholar]
- 171.Schlecker E, Stojanovic A, Eisen C, Quack C, Falk CS, Umansky V, Cerwenka A. Tumor-infiltrating monocytic myeloid-derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth. J Immunol. 2012;189(12):5602–5611. doi: 10.4049/jimmunol.1201018. [DOI] [PubMed] [Google Scholar]
- 172.Jordan KR, Amaria RN, Ramirez O, Callihan EB, Gao D, Borakove M, Manthey E, Borges VF, McCarter MD. Myeloid-derived suppressor cells are associated with disease progression and decreased overall survival in advanced-stage melanoma patients. Cancer Immunology Immunotherapy. 2013;62(11):1711–1722. doi: 10.1007/s00262-013-1475-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Meyer C, Sevko A, Ramacher M, Bazhin AV, Falk CS, Osen W, Borrello I, Kato M, Schadendorf D, Baniyash M, Umansky V. Chronic inflammation promotes myeloid-derived suppressor cell activation blocking antitumor immunity in transgenic mouse melanoma model. Proceedings of the National Academy of Sciences. 2011;108(41):17111–17116. doi: 10.1073/pnas.1108121108. [DOI] [PMC free article] [PubMed] [Google Scholar]