Next Article in Journal
Prediction of Nodal Metastasis in Lung Cancer Using Deep Learning of Endobronchial Ultrasound Images
Previous Article in Journal
Identification of Endoplasmic Reticulum Stress-Related Subtypes, Infiltration Analysis of Tumor Microenvironment, and Construction of a Prognostic Model in Colorectal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of Resolution of Inflammation on Tumor Microenvironment: Exploring New Ways to Control Cancer Progression

1
Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
2
Institute of Experimental Endocrinology and Oncology (IEOS), CNR, 80131 Naples, Italy
3
Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
*
Authors to whom correspondence should be addressed.
Cancers 2022, 14(14), 3333; https://doi.org/10.3390/cancers14143333
Submission received: 10 June 2022 / Revised: 6 July 2022 / Accepted: 7 July 2022 / Published: 8 July 2022
(This article belongs to the Section Tumor Microenvironment)

Abstract

:

Simple Summary

The evolution of cancer is strongly influenced by the context in which tumor cells develop and grow, known as the tumor microenvironment (TME). The TME is constituted of a set of cells with different natures, which can produce various factors or interact with cancer cells, thus favoring or inhibiting cancer growth. Specific factors with the ability to shape the TME, in order to create an unfavorable context for tumor cells, are the Specialized Pro-resolving Mediators (SPMs). SPMs are small lipid molecules derived from ω-3 and ω-6 fatty acids, exerting the physiologic role of dampening the inflammatory responses and helping tissues to regain their homeostasis after insults. Here, we present the knowledge relative to the action of SPMs on each component of the TME and its effects on tumor growth and progression. These summarized findings highlight novel potential strategies to manage cancer progression.

Abstract

Non-resolving inflammation is an enabling feature of cancer. A novel super-family of lipid mediators termed Specialized Pro-resolving Mediators (SPMs) have a role as bioactive molecules mediating the resolution of inflammation in cancer biology. SPMs are derived from ω-3 and ω-6 polyunsaturated fatty acids through the activity of lipoxygenases. SPMs have been described to directly modulate cancer progression by interfering with the epithelial to mesenchymal transition and invasion of cancer cells. SPMs have also been demonstrated to act on several components of the tumor microenvironment (TME). Consistently with their natural immunomodulatory and anti-inflammatory properties, SPMs are able to reprogram macrophages to favor phagocytosis of cell debris, which are an important source of pro-inflammatory and pro-angiogenic signals; sustain a direct cytotoxic immune response against cancer cells; stimulate neutrophils anti-tumor activities; and inhibit the development of regulatory T and B cells, thus indirectly leading to enhanced anti-tumor immunity. Furthermore, the resolution pathways exert crucial anti-angiogenic functions in lung, liver, and gastrointestinal cancers, and inhibit cancer-associated fibroblast differentiation and functions in hepatocellular carcinoma and pancreatic cancer. The present review will be focused on the potential protective effects of resolution pathways against cancer, exerted by modulating different components of the TME.

1. Resolution of Inflammation

Chronic inflammation, together with genome instability, are now considered enabling features of cancer, fostering the other established hallmark functions (i.e., sustained proliferation and resistance to cell death, increased angiogenesis, and invasion capability, etc.) [1]. Consistently, several studies have demonstrated that inflammation intervenes in tumor initiation, growth, and progression in different cancer models [2].
In recent years, various mechanisms underlying the onset of a chronic inflammatory process have been defined. In particular, it is now clear that inflammation intensity and duration are the result of the balance of two active processes: the first is the activation of inflammatory response with production of pro-inflammatory mediators (e.g., eicosanoids and cytokines) and recruitment of innate immune cells; the second is its resolution process mediated by different actively produced mediators, whose goal is to dampen the inflammatory response and allow the return to tissue homeostasis [3].
While inflammation is characterized by the production of inflammatory mediators facilitating leukocyte recruitment and activation, the resolution phase stands out for pro-inflammatory mediator degradation and/or reduced production. When the concentrations of pro-inflammatory factors reach a plateau, neutrophils and the other leukocytes infiltrating the inflamed site initiate a switch in the mediators produced, from pro-inflammatory to pro-resolutive, in order to counteract further recruitment of inflammatory neutrophils and to favor non-inflammatory monocytes. An example of this fine adjustment is the change in the lipidic mediators present in the exudates: in the first phase of inflammation, prostaglandins and leukotrienes are abundant, while during the resolution process different kinds of lipidic autacoids can be detected (e.g., lipoxins and resolvins). Pro-resolving mediators not only limit the inflammatory response, but also contribute to tissue homeostasis, causing the clearance of apoptotic neutrophils (aka, efferocytosis), sustaining several steps of tissue repair, and limiting fibrosis and scar formation [4].
Different kinds of mediators can actively sustain the resolution response [4]. Lipidic mediators intervene in inflammation resolution: these autacoids are known as Specialized Pro-resolving Mediators [lipoxins (LX), resolvins (Rv), protectins (PD), and maresins (MaR)] [5]. Furthermore, annexin A1 (AnxA1), together with other proteins (e.g., adrenocorticotropic hormone, chemerin peptides, and galectin-1) can sustain the resolution program [4,6]. Interestingly, gaseous mediators (nitric oxide, hydrogen sulfide, and carbon monoxide), adenosine, and neuromodulators such as acetylcholine could also exert anti-inflammatory and pro-resolving functions [7,8].
An insufficient resolution response can lead to chronic inflammation, exactly as in the persistent activation of a pro-inflammatory program [9]. However, while the study of the resolution of inflammation in acute inflammatory response has been widely dissected [10], the investigation of its role in chronic inflammatory processes is more recent and therefore less detailed [11].

2. Specialized Pro-Resolving Mediators and Cancer

Specialized Pro-resolving Mediators (SPMs) are bioactive lipid molecules produced from ω-3 (docosahexaenoic (DHA) and eicosapentaenoic (EPA) acids) and ω-6 (arachidonic acid (AA)) polyunsaturated fatty acids (PUFA) thanks to the activity of specific enzymes (lipoxygenases (ALOX5, ALOX15)). Lipoxins (LXA4, LXB4) are SPMs derived from the ω-6 AA; E-series Resolvins (RvEs) from the ω-3 EPA; D-series Resolvins (RvDs), Protectins (PD), and Maresins (MaR) from the ω-3 DHA [12]. These molecules exert their action by interacting with G protein coupled receptors (e.g., GPR18, GPR32, GPR37, ChemR23, FPR2, BLT1) [13,14].
FPR2, aka ALX/FPR2, contributes to the resolution of inflammation by binding LXA4 [15,16,17], RvD1, and RvD3 [18,19]. RvD1 and RvD3 have also been described as binding GPR32 [16,17], together with RvD5 [14]. ChemR23 was identified as the high affinity receptor for RvE1 [20]. RvE1, in addition to ChemR23, has also been described as binding with low affinity the BLT1 receptor [21]. ChemR23 also recognizes RvE2 [22]. Merlin and coll. recently published that they were not able to observe ChemR23 activation upon RvE1 stimulation of HEK293 cells [23], thus suggesting that more in depth studies are needed to define the pairs of receptor–ligands or to describe context-dependent effects of these receptor(s). GPR18, an already known receptor for cannabinoids [24], also recognizes RvD2 [25]. GPR37 is a candidate receptor for NPD1/PD1 [26], together with BLT1 [23]. More recently, other studies have shown that MaR1 is able to bind and activate the LGR6 receptor [27] and that N-3 docosapentaenoic acid-derived resolvin D5 (RvD5n-3 DPA) can activate resolution responses through the engagement of GPR101 receptor [28].
Due to their crucial role in the regulation of inflammatory responses, SPM deficiency has been linked to several pathologic conditions in which an inflammatory microenvironment is established and is the cause of disease, including cancer [29].
The strongest evidence of the crucial role of SPMs in the modulation of cancer initiation and progression comes from the studies of Serhan et al., who demonstrated that the protective role of aspirin against some cancer types could be ascribed to its ability to mediate COX2 acetylation. This COX2 modification is responsible for the production of the SPM epimers (aka, aspirin-triggered (AT) lipoxins/epi-lipoxins and aspirin-triggered (AT) resolvins/epi-resolvins) [12]. These effects are typical of aspirin and not common to other nonsteroidal anti-inflammatory drugs (NSAIDs); consistently, aspirin triggers beneficial effects on tumorigenesis that have not been observed with NSAIDs [30,31].
Although the study of SPM functions in cancer is new and the data produced are still limited, some evidence in humans supports a tumor suppressor function of SPMs [29,32,33,34].
The cancer types in which a protective role of SPMs has been described are oral, gastric, colon, pancreatic, liver, lung cancers, melanoma, papilloma, and non-solid tumors such as leukemia [35].
In recent years, attention has been paid to the mechanisms by which SPMs could affect cancer progression. Obviously, several of these activities have been linked to the anti-inflammatory potential of SPMs and their impact on different components of the tumor microenvironment (TME), but the direct effect of SPMs on cancer cells survival, epithelial to mesenchymal transition (EMT), and invasion also deserves to be mentioned [9,34,36,37,38,39,40,41,42] (Table 1).
The present review will be focused on the lipid Specialized Pro-resolving Mediators and their role in cancer progression, pointing a magnifying glass at their effects on tumor stroma. In the following paragraphs, we will describe the most important reports present in the literature related to the effects of SPMs on the three component of the TME: the immune cell infiltrate, the vascular bed, and the mesenchymal component of tumor stroma.

3. Effects of SPMs on the Immune Cell Compartment Infiltrating Tumor Microenvironment

Tumor-infiltrating immune cells play a key role in the evolution of cancer. Typically, immune cells should recognize and eliminate cancer cells [56]. Despite this, cancer cells continue to grow and expand, due to several escape mechanisms [57]. Furthermore, several immune cell classes can orchestrate cancer growth, by producing mediators directly sustaining cancer progression [58].
The most abundant immune cell population infiltrating tumors are the macrophages, dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), T cells, mast cells, and natural killer (NK) cells. Some of them have been clearly associated to anti-tumor immune responses: lymphoid cells, including natural killer (NK) cells, CD8+ T lymphocytes, CD4+ helper T (Th) cells, pro-inflammatory macrophages (M1), and DCs. Moreover, MDSCs and regulatory T (Treg) cells have been shown to exert a pro-tumorigenic action [59,60].
In the context of anti-tumor immune response, a primary regulatory role has to be ascribed to immune checkpoint (IC) molecules (e.g., Programmed Cell Death-1 (PD-1), Programmed death ligand 1 (PD-L1), Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4), etc.). Due to its ability to dampen anti-tumor immune response, immune checkpoint blockade (ICB) is today considered, among the various types of immunotherapy, the pivotal approach [61]. It is now clear that a continuous influence of IC on TME, and vice versa, exists. Thus, it would also be simplistic not to consider, in the context of immune response to cancer, the possibility that resolution of inflammation could affect immune-checkpoint expression and functions.
In this scenario, an RNA-seq analysis of patients with head and neck squamous cell carcinoma showed that a higher score for RvD metabolism-associated markers directly correlates with a better clinical outcome. A high RvD score also associates to a signature suggestive of enhanced anti-tumor immunity, as demonstrated by its association to genes involved in the cytotoxic activity of immune cells (e.g., granzymes and perforin). Finally, a high RvD score is presumably linked to increased responsivity to immune-checkpoint inhibitors (ICI), as demonstrated by its significant correlation with the expression of four immune-checkpoint inhibitor targets (CD274, CTLA4, LAG3, and PDCD1) [62].
Consistently, preliminary data from Gartung et al. reported that, in a murine model of head and neck cancer, resolvins and immune checkpoint inhibitors (e.g., anti-PD-1) act synergistically, by suppressing tumor growth [63]. Furthermore, in a model of colorectal carcinoma (CRC), administration of AT-SPM regulated macrophage trafficking by stimulating the clearance of apoptotic cells and reduced the expression of the immunosuppressive receptor PD-1 in both macrophages and CD8+ T cells [64]. Similarly, SPM LXA4 was reported to reduce the expression of PD-L1 in a Kaposi’s sarcoma-associated herpesvirus (KSHV)-induced model [65].

3.1. SPM Effects on Innate Immune Cell Compartment

Due to their natural role in inhibiting the inflammatory response, most of the studies present in the literature, including those related to cancer, have been focused on the effects of SPMs in modulating the functions of innate immune cells (Table 1).
The most abundant innate immune cells infiltrating tumors are neutrophils and macrophages, which sometimes act against cancer cells through cytotoxic and phagocytic activities, while in other contexts they sustain tumor growth through the production of mediators hijacked by cancer cells to favor their spread [66].
Here, we will summarize the knowledge regarding SPM effects on neutrophils and macrophages. Furthermore, a paragraph will be dedicated to DCs, the innate immune cell population essential for the induction of an adaptive immune response.

3.1.1. Neutrophils

While several research programs have highlighted the role of macrophages in cancer progression [67], less information is available concerning the function of neutrophils in the context of the tumor microenvironment [46]. To date, most reports have sustained the pro-tumorigenic role of neutrophils, and more generally of polymorphonuclear (PMN) cells [46,48], while others have demonstrated that PMN are able to inhibit cancer progression [68,69,70,71]. The literature suggests that the different functions of neutrophils in supporting or inhibiting cancer depends on the tumor stage and the cancer tissue [71].
Nevertheless, neutrophils are targets of resolution molecules, as demonstrated by the evidence that BLT1 receptor expressed on neutrophil surface is crucial in the regulation of their mitochondrial functions. In more detail, the RvE1-BLT1 signal mediates the activation of apoptotic responses through the induction of reactive oxygen species (ROS) and subsequent activation of caspases, and through the inhibition of ERK and Akt anti-apoptotic signals [72]. Furthermore, GPR18 knockout in mice displayed reduced neutrophil infiltration at the infection site and reduced tissue recovery following injuries [25].
The few studies regarding the effects of pro-resolving mediators on cancer-associated neutrophils were directed to the definition of the effects of SPMs on neutrophil phenotype and functions and on the impact of these on cancer progression.
Mattoscio and coll. demonstrated that RvD1 reduces the growth and proliferation of tumors in vivo using a human papilloma virus (HPV) tumorigenesis model. This effect is mediated by the ability of RvD1 to change the phenotype of neutrophils in a “pro-resolving anti-cancer phenotype”, characterized by increased anti-tumor properties. In addition, RvD1 stimulated PMN to secrete increased levels of chemoattractants for monocytes able to inhibit tumor growth in vivo [46].
Vannitamby et al. focused their studies on the effects of AT-RvD1 on neutrophils in a mouse model of lung cancer. AT-RvD1 is produced by aspirin-acetylated COX2 and 5-lipoxygenase (ALOX5) enzymes [48]. The authors found that increased neutrophil infiltration in the TME of lung adenocarcinoma patients correlated with a low level of expression of ALOX5 enzyme. Based upon this observation, they investigated the possibility that AT-RvD1 could exert a therapeutic activity in a mice model of lung adenocarcinoma, demonstrating that AT-RvD1 treatment significantly reduced lung adenocarcinoma growth in vivo by reducing the infiltration of neutrophil, which is responsible for lymphocyte activity suppression [48].

3.1.2. Macrophages

Macrophages represent probably the most important target cell of SPMs. In the physiology of inflammation resolution, SPMs sustain the recruitment of the macrophages responsible for the non-inflammatory efferocytosis of apoptotic neutrophils [73,74].
The classic view of macrophage biology classifies them as M1, with pro-inflammatory, microbicidal, and anti-tumor functions; or as M2, displaying anti-inflammatory, immunosuppressive, and pro-tumorigenic properties [75]. However, this polarization model shows some limitations: it reflects well the characteristics of macrophages induced to polarize in vitro by specific cytokines and mediators, but it does not describe well the characteristics of macrophages present in tissues [76]. In the cancer context, tumor associated macrophages (TAM) display phenotypes that are a continuum between the two phenotypes, being continuously conditioned by the TME [76]. For example, the most abundant phenotype of macrophages in TME is M2, but these cells can exert a more efficient phagocytosis of cancer cells than M1 cells, thus potentially favoring an anti-tumor response [29].
A characterization of the lipid mediators produced by the two different classes of macrophages demonstrated that M1 produce pro-inflammatory lipid mediators, such as prostaglandins and leukotrienes, while M2 macrophages produce SPMs. This suggests that the different classes of lipid autacoids could be useful in re-shaping the macrophage phenotype to activate a protective anti-tumor response [29]. A critical role of resolution molecules in macrophage function regulation was also revealed in studies on SPM receptors knock-out: (i) macrophages deleted for GPR32 become unresponsive to RvD1 and lost the ability to polarize toward a pro-resolutive phenotype [19]; (ii) GPR18 knock-out in macrophages reduced their ability to phagocyte debris and dead cells [25]; (iii) ChemR23 blocking antibody reduced the phagocytic activity of RvE1-stimulated macrophages in acute and chronic inflammatory models [77,78]; and (iv) mice knocked-out for GPR37 displayed a reduced macrophage phagocytic activity in a model of inflammatory pain. [26].
Physiologically, SPMs increase macrophage survival [79], sustain the phagocytic activity of macrophages against microbes and apoptotic neutrophils [16,80], reduce the secretion of pro-inflammatory cytokines via inhibition of NF-kB [29,80], and, in the meantime, increase the production of anti-inflammatory cytokines [22,81].
Although it would be expected that the SPM-sustained changes of macrophage phenotype towards a M2 type would favor neoplastic progression, the available literature points to a protective anti-tumor effect of SPM-conditioned TAM [29,54]. The analysis of the effects of SPMs on TAM supports this evidence and demonstrates that the scenario is more complex than expected. Indeed, SPMs sustain changes in TAM that place them in between the M1 and M2 phenotypes [82,83,84].
Several research groups have investigated the role of SPMs in conditioning macrophage phenotype in the cancer context [29]. ATL-1, a synthetic analogue of LXA4, alters TAM profile by decreasing M2 surface markers, triggering ROS production, and increasing the cytotoxic and decreasing the anti-apoptotic properties of TAM against melanoma cancer cells. Consistently, ATL-1 inhibited cancer progression in a murine model by affecting macrophage phenotype [29]. Similarly, RvD1 and RvD2 inhibited cancer cell proliferation by affecting macrophage polarization in a prostate cancer model [47].
A very interesting research line has focused on the effects of SPMs on macrophage efferocytosis of chemotherapies-induced tumor cell debris. Debris of tumor cells have been demonstrated to be produced following several types of cancer therapies, including radiation and chemotherapy, and to sustain tumor engraftment, growth, and metastasis by activating a pro-inflammatory response [85,86]. Furthermore, tumor cell debris affect M1 TAM, inducing an immunosuppressive response, and thus limiting anti-cancer immunity [87,88].
The involvement of lipidic autacoids in the regulation of inflammatory response associated to cancer cell debris is now clear, since it has been recognized that they stimulate tumor growth by sustaining the production of the pro-inflammatory autacoid prostaglandin E2 (PGE2) in the TME [89]. On the other hand, in several tumor types, SPMs RvD1, RvD2, and RvE1 block the ability of tumor cell debris to sustain cancer progression via the activation of macrophage phagocytosis and clearance of cell debris [44]. SPMs sustained polarization of macrophages in these models is also characterized by a reduced production of pro-inflammatory cytokines [44]. Furthermore, the anti-tumor effect of aspirin was linked to its ability to induce the production of SPMs, enhance the macrophage phagocytosis of tumor cell debris, and reduce macrophage production of inflammatory mediators [45].

3.1.3. Dendritic Cells

This cell type represents a crucial link between innate immunity and the activation of the adaptive response, due to its ability to professionally present antigens to lymphocytes [90]. DCs express receptors to SPMs, thus representing a target of these molecules [91]. To date, evidence has been produced demonstrating that SPMs could affect DC maturation and functions, with the aim of reducing inflammation and allowing tissue restitution. SPMs reduce the DC migration and production of pro-inflammatory cytokines [92,93]. Furthermore, RvD1 reduces the levels of Major Histocompatibility Complex II (MHC II) and costimulatory molecules on DCs [94]. As an example, RvE1 treatment blocks DCs in an immature state [95] and prevents their migration to draining lymph-nodes, thus resulting in a reduced T cell response [96,97]. All these evidence has been produced in disease models in which SPMs work in order to facilitate tissue homeostasis (e.g., infection, autoimmune diseases, etc.), but currently no data have been published on SPM effects on DCs in a cancer context, where the separation between tissue homeostasis and tumor growth is less clear.

3.2. Effects of SPMs on the Adaptive Immune Cell Compartment of the TME

Very few studies are available on the effects of SPMs on the adaptive immune branch of the immune system, and even less in the specific context of cancer, although the potential immunomodulatory effects of SPMs on adaptive immune response could be of great interest [98].

3.2.1. Effector T and B lymphocytes

Since the activation of T or B lymphocytes is the goal of the adaptive immune response, an active area of study focuses on SPM activity with T and B cells. The effector functions of the different T and B cell subsets are strictly linked to the characteristics of the produced cytokines [99].
The most plastic subset of human cells is probably represented by the CD4+ T lymphocytes: the effector functions against different classes of pathogens are assigned to T helper (Th)1, Th2, and Th17 subsets; follicular helper T (Thf) cells assist B lymphocytes in their maturation, and Treg cells, instead, control self-tolerance [100]. Chiurchiù and coll. investigated the effects of SPMs on T CD4+ lymphocytes, demonstrating that RvD1, RvD2, and MaR1 are able to modulate T cell differentiation programs suppressing Th1 and Th17 T cell subtypes, as well as their ability to produce pro-inflammatory cytokines and to increase, in the meantime, the production of Treg cells [101]. Similarly, the inhibition of cytokine production mediated by SPMs on CD4+ Th1 and Th17 T cells has also been described for PD1, RvD3, LXA4, and LXB4 [102,103]. This inhibitory effect of SPMs on cytokine production has a great impact on pro-inflammatory T cell subsets (i.e., Th1 and Th17) but a less important effect on the Th2 phenotype. An exception was reported for RvE1, which has been described to dampen the Th2 T cell response in a model of asthma [104,105].
Both in vitro and in vivo data support the hypothesis that SPMs are instead able to augment Treg generation, consistently with their ability to produce immunomodulatory factors and to exert pro-resolving functions [101,106,107]. Since more data are available on the effects of SPMs on regulatory subsets of lymphocytes, a specific paragraph will be dedicated to these.
Unfortunately, to date, no direct and structured data have been published on the effects of SPMs on the cytotoxic response of CD8+ T lymphocytes, although they represent the key to directly killing cancer cells. However, in support of a role of SPMs in sustaining an anti-tumor cytotoxic response, it has been observed that RvD1 and its precursor (DHA) affect natural killer (NK) cells, the innate immune counterpart of CD8+ T cells, by preventing their death and sustaining their tumoricidal activity [38]. Consistently, it has been demonstrated that LXA4 is able to improve the anti-tumor activities of CD8+ T cells indirectly, by reducing regulatory subsets of T and B cells [53]. Furthermore, it has been described that GPR18 is expressed in CD8+ T lymphocytes and regulates their development and homing in epithelia [108], suggesting the crucial importance of resolution responses in tumor immune response and immunotherapy [108]. Interestingly, it has been demonstrated that BLT1 regulates anti-tumor immune responses by favoring CD8+ T cell migration in melanoma [109] and the cervical cancer murine microenvironment [110].
Some authors have also addressed the role of inflammation resolution on B cell compartment. SPMs have been described as potent modulators of B cell functions [111]: RvD1 increases IgM and IgG and decreases IL-6 production from B cells, as well as favoring the differentiation of B cells in antibody secreting cells [112]. Several other reports have pointed to a role of SPMs in sustaining antibody production from B cells [113,114]. LXA4 has been described to be able to accelerate B cell migration to the spleen, increasing the response to antigens [115].
These data perfectly fit into the scenario of SPMs as negative modulators of inflammatory response when acting on the adaptive immune compartment, although no structured data are available in cancer context nor a clear definition of potential pro- or anti-tumor effects.

3.2.2. Regulatory Lymphocytes

The immune response against cancer cells is the result of the balance between immune effector cells such as CD8+ T cytotoxic cells or NK cells and immunosuppressive cells such as Treg or MDSC [116,117,118].
The resolution of inflammation affects Treg regulation and function(s). It has been demonstrated that SPMs (RvD1, RvD2, and Mar1) exert a regulatory role on the balance between pathogenic Th1 and tolerogenic Treg cells [101,119,120,121] during autoimmune responses. Furthermore, the reduced expression of ALOX15 enzyme in Treg is associated with a lower expression of Foxp3, the master regulator of Treg lineage, and alteration of several cellular metabolic pathways [122]. Consistently, GPR32 plays a crucial role in regulating the adaptive immune responses, as demonstrated by the evidence that its expression is crucial for the generation and function of Treg cells [101].
This evidence suggests that the resolution of inflammation could also impact on the role and function of Treg cells in cancer, although several questions remain open. The first point is that the Treg role in cancer initiation/progression is complex: the most accredited theory is that Treg cells exert a pro-tumor effect by inhibiting anti-cancer immune response; however, in some cancer contexts, an anti-tumor effect of Treg was described and associated with their ability to prevent tumor inflammation [123]. Indeed, it has been demonstrated that Treg depletion favors cancer development in a model of inflammation induced colon cancer [43]. Zhang and coll. postulated that Treg could promote early tumor growth, while inhibiting tumor progression later on [123]. Interestingly, they demonstrated that inhibition of Treg in large tumors favors growth, by inducing the generation of MDSCs in the TME, and that this phenomenon is associated with increased levels of LXA4 [123].
A specific subpopulation of B cells, namely B regulatory (Breg) cells, have been recently described as able to modulate the immune response in the context of TME [124]. Breg cells are able to produce large amounts of immunosuppressive cytokines (IL-10 and TGF-β) and to express immunoregulatory molecules on their membrane (e.g., PD-L1). Breg cells have been demonstrated to suppress T and NK cells responses in several models of tumor growth [125]. Furthermore, Breg cells facilitate the conversion of CD4+ T lymphocyte in TME in CD4+CD25+FoxP3+ Treg cells [125].
Wang and coll. demonstrated that LXA4 is able to suppress the generation of Breg cells in tumor-bearing mice, thus dampening tumor growth. Consistently, the depletion of Breg cells in mice abolishes the anti-tumor activity of LXA4 [53]. Interestingly, LXA4 not only affects Breg cell induction, but is also able to reduce the number of Treg cells, both in the draining lymph nodes and in the TME, and simultaneously to enhance the cytotoxic activities of T lymphocytes [53].
These data suggest that other studies are necessary to clarify the role of SPMs in the modulation of regulatory lymphocytes in the TME, in order to define the effects of SPMs on these immune populations and their impact on cancer progression.

4. Effects of SPMs on Cancer-Associated Fibroblasts

Cancer-associated fibroblasts (CAFs) are the most abundant cells in the TME, where they represent a key source of extracellular matrix components, thus contributing to the formation of a desmoplastic stroma and playing crucial roles during malignant cancer progression and metastasis [126].
SPMs promote the regeneration of damaged tissue by finely regulating the fibrotic response, both in the physiology of wound healing and in several pathologic scenarios in which an excessive fibrotic response is established [127]. This is the case of idiopathic pulmonary fibrosis (IPF), in which LXA4 has been reported to inhibit the pro-fibrotic pulmonary response through different mechanisms [128].
The effects of SPMs on CAFs have been studied and described in a few model systems. RvD1 has been demonstrated to inhibit the production of pro-tumorigenic mediators from CAFs in a model of hepatocellular carcinoma (HCC), thus suppressing EMT and the stemness of cancer cells [49]. Furthermore, LXA4 attenuates pancreatic tumor growth by inhibiting the function of human pancreatic stellate cells (hPSCs), which are the precursors of pancreatic CAFs; in more detail, LXA4 attenuates hPSC-induced desmoplasia, reverts hPSC activation into CAFs, and modulates several other hPSC tumor-promoting effects [55].
Since one of the problems of the fibrotic process associated with tumors, in addition to the growth-promoting effect, is the formation of a barrier that is difficult to penetrate for drugs, these papers suggest a further potential use of SPMs (i.e., RvD1 and LXA4) as useful agents, in conjunction with conventional therapy, to improve their efficacy.

5. Effects of SPMs on the Tumor Vascular Bed

Other than in immune cells, SPM receptors have also been described in endothelial cells (ECs) and vascular smooth muscle cells (VSMCs), and to be modulators of their phenotype and function(s) [13]. In particular, it has been described that ECs express ALX/FPR2, GPR32, GPR18, BLT1, and ChemR23 [16,129,130,131,132]. Among the other types, the anti-angiogenic properties of SPMs have also been described in several contexts [9], for example, mediated by the SPM (LXA4) ability to inhibit EC proliferation and migration, to interfere with vascular endothelial growth factor (VEGF) signaling, and to dampen VEGF receptor expression [133,134]. LXA4 has also been reported to modulate EC functions by increasing NO production [135]. Furthermore, most SPMs affect leukocyte–EC interactions by reducing the expression of adhesion molecules and the production of inflammatory cytokines [13]. Finally, the genetic ablation of GPR32 in ECs demonstrated that the receptor is crucial for endothelial cell integrity and barrier function [136].
However, to date, only a few studies have investigated the role of pro-resolving pathways in the modulation of cancer angiogenesis [9].
In a model of Kaposi Sarcoma (KS), LXA4 inhibited the secretion of angiogenic factors [137]. Similarly, LXA4 decreased the production of the angiogenic mediators from HCC cells [138]. Interestingly, SPMs (RvD4 or RvD5), in combination with anti-angiogenic therapy (i.e., the thrombospondin (TSP)-1 peptide 3 TSR or anti-VEGF via DC101), induced synergistic anti-tumor activity in xenograft models of ovarian cancer [138].
Related to cancers of the gastrointestinal (GI) tract, the authors recently described a novel function of SPMs in gastric cancer (GC), demonstrating that two SPMs (i.e., RvD1 and LXB4) suppress angiogenesis, thus inhibiting tumor growth [50]. A key modulator of SPM production was the Formyl Peptide Receptor 1 (FPR1), which functions in the GI tract as a tumor suppressor [139]. FPR1 is a member of an innate immune receptor family of formyl peptide receptors (FPR1, 2, and 3), which are pattern recognition receptors (PRRs) belonging to the G-protein coupled receptor family and able to recognize conserved microbe- or damage-associated molecular structures (PAMPs/DAMPs) and initiate both inflammation and its resolution, depending on the environmental context and on the specific ligand [140,141]. We demonstrated that GC cells silenced for FPR1, but not for FPR2 or FPR3, displayed an in vivo growth advantage, due to their increased ability to form new vessels. In culture, these cells displayed an increase in the production of pro-inflammatory and pro-angiogenic mediators and a drop in the levels of various components of the pro-resolving pathways (ALOX5/15, SPMs (RvD1 and LXB4), and SPM receptors (BLT1, ChemR23 and GPR32)) [50,139]. The same effects were observed when FPR1 was pharmacologically antagonized by cyclosporin H. Consistently, FPR1 activation or increased expression mediates the opposite effects [50]. FPR1-mediated anti-angiogenic potential depends on pro-resolving pathways, as witnessed by the finding that blockade of ALOXs or of SPM receptors (i.e., GPR32) blunted this FPR1 activity in GC cells. Moreover, the increased angiogenic potential of FPR1-depleted GC cells was reverted by exogenous administration of SPMs (RvD1 or LXB4) [50]. The blockade of the pro-resolving receptor GPR32, or of ALOX15, enhanced the angiogenesis and tumorigenic activity of GC cells, mimicking FPR1-depletion. Thus, GC cells are endowed with an intrinsic angiogenic potential, negatively controlled by SPMs. These, in turn, are positively controlled by FPR1. Consistently, a diet enriched in the precursors of SPMs, ω-3 or ω-6 PUFA, [142] inhibited the growth of FPR1-silenced GC cells in mice, by specifically impairing angiogenesis [50].
Several reports have also pointed to a crucial protective role of pro-resolving pathways in colorectal cancer (CRC) [143,144,145]. Unpublished results from the authors’ group also indicate a crucial anti-angiogenic function of SPMs in CRC: the activation of FPR1 mediated by the probiotic Lactobacillus Rhamnosus (LGG) or specific bacterial products in CRC cells caused an increase of pro-resolving mediators (RvD1 and LXB4) and the consequent inhibition of angiogenesis [51]. LGG is a commensal bacterium used as a probiotic and described as able to dampen the chronic inflammation associated with CRC development [146]. LGG sustains colonic wound healing in mice [147,148,149] and activates pro-apoptotic and anti-metastatic responses [150,151] through the interaction with FPR1 [147]. Our data highlight new pro-resolving and anti-angiogenic functions of this bacterium mediated by FPR1 [51].
By asking whether similar mechanisms could also intervene in cancers derived from other epithelia, we demonstrated that pro-resolving pathways could also exert anti-angiogenic activity in a lung cancer model; however, in this context, the innate immune receptor that controls SPM production is the Toll-like receptor 7 (TLR7) [52]. In this model system, TLR7 activation, similarly to FPR1 in the GI tract, sustains the increased expression of the enzymes responsible for SPMs biosynthesis, the increased secretion of SPMs from cancer cells, the augmented expression of SPM receptors, and finally the reduction of the angiogenic potential of lung cancer cells [52].

6. Conclusions

Inflammation has long been considered a hallmark of cancer, both for its ability to intervene in cancer initiation and to sustain cancer progression. Recently, the discovery of the resolution of inflammation opened the way for new therapeutic possibilities that, rather than block the inflammatory response, could instead support the induction of resolution of the inflammation.
Among others, Specialized Pro-resolving Mediators (SPMs) are “bioactive lipids” already described as key players in regulating inflammatory response and tissue homeostasis in several physiologic and pathologic conditions, by acting on several cell types in the context of inflammatory reactions. The present review focused on the possibility that SPMs could shape the TME, to affect cancer cell growth and progression.
Strong evidence has been provided supporting the possibility that SPMs act on the innate immune cell compartment infiltrating tumors from an anti-cancer perspective (Figure 1).
Indeed, SPMs favor the reduction of the inflammatory response and the acquisition of a “pro-resolving” phenotype in these cells, which contributes to the anti-tumor response. Less data are available on the effects of SPMs on the adaptive immune compartment, and no clear definition of the function of SPMs in the context of an adaptive anti-cancer response has been defined (Figure 1).
Interestingly, SPMs have been described to act on various other cells constituting tumor stroma (Figure 1). Consistently with their natural anti-fibrotic and anti-angiogenic properties, SPMs have been described as able to counteract the CAF ability to sustain cancer progression and to dampen the angiogenic response vital for cancer dissemination.
Although further studies are needed to define the role of these bioactive lipids in cancer, they appear to be a potential and promising approach to fight cancer progression.

Author Contributions

F.L. and M.M.; writing-original draft preparation, N.P. and R.M.M.; writing-review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

PRIN 2017 no. 2017XJ38A4; Istituto Superiore di Oncologia grant (MIUR PON01_02782/12); POR Campania FESR 2014ߝ12020 “SATIN” grant; POR Campania FESR 2014-2020 “RARE.PLAT.NET” grant.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Fishbein, A.; Hammock, B.D.; Serhan, C.N.; Panigrahy, D. Carcinogenesis: Failure of resolution of inflammation? Pharmacol. Ther. 2021, 218, 107670. [Google Scholar] [CrossRef] [PubMed]
  3. Fredman, G. DELineating resolution of inflammation. Nat. Immunol. 2019, 20, 2–3. [Google Scholar] [CrossRef] [PubMed]
  4. Sugimoto, M.A.; Sousa, L.P.; Pinho, V.; Perretti, M.; Teixeira, M.M. Resolution of Inflammation: What Controls Its Onset? Front. Immunol. 2016, 7, 160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Serhan, C.N.; Savill, J. Resolution of inflammation: The beginning programs the end. Nat. Immunol. 2005, 6, 1191–1197. [Google Scholar] [CrossRef]
  6. Perretti, M.; D’Acquisto, F. Annexin A1 and glucocorticoids as effectors of the resolution of inflammation. Nat. Rev. Immunol. 2009, 9, 62–70. [Google Scholar] [CrossRef] [PubMed]
  7. Hasko, G.; Cronstein, B. Regulation of inflammation by adenosine. Front. Immunol. 2013, 4, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Mirakaj, V.; Dalli, J.; Granja, T.; Rosenberger, P.; Serhan, C.N. Vagus nerve controls resolution and pro-resolving mediators of inflammation. J. Exp. Med. 2014, 211, 1037–1048. [Google Scholar] [CrossRef] [Green Version]
  9. Prevete, N.; Liotti, F.; Amoresano, A.; Pucci, P.; de Paulis, A.; Melillo, R.M. New perspectives in cancer: Modulation of lipid metabolism and inflammation resolution. Pharmacol. Res. 2018, 128, 80–87. [Google Scholar] [CrossRef]
  10. Serhan, C.N.; Gupta, S.K.; Perretti, M.; Godson, C.; Brennan, E.; Li, Y.; Soehnlein, O.; Shimizu, T.; Werz, O.; Chiurchiu, V.; et al. The Atlas of Inflammation Resolution (AIR). Mol. Asp. Med. 2020, 74, 100894. [Google Scholar] [CrossRef]
  11. Schett, G.; Neurath, M.F. Resolution of chronic inflammatory disease: Universal and tissue–specific concepts. Nat. Commun. 2018, 9, 3261. [Google Scholar] [CrossRef] [PubMed]
  12. Serhan, C.N.; Chiang, N.; Dalli, J.; Levy, B.D. Lipid mediators in the resolution of inflammation. Cold Spring Harb. Perspect. Biol. 2014, 7, a016311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Diaz Del Campo, L.S.; Rodrigues-Diez, R.; Salaices, M.; Briones, A.M.; Garcia-Redondo, A.B. Specialized Pro-Resolving Lipid Mediators: New Therapeutic Approaches for Vascular Remodeling. Int. J. Mol. Sci. 2022, 23, 3592. [Google Scholar] [CrossRef] [PubMed]
  14. Luo, X.; Gu, Y.; Tao, X.; Serhan, C.N.; Ji, R.R. Resolvin D5 Inhibits Neuropathic and Inflammatory Pain in Male but Not Female Mice: Distinct Actions of D-Series Resolvins in Chemotherapy-Induced Peripheral Neuropathy. Front. Pharmacol. 2019, 10, 745. [Google Scholar] [CrossRef]
  15. Gronert, K.; Gewirtz, A.; Madara, J.L.; Serhan, C.N. Identification of a human enterocyte lipoxin A4 receptor that is regulated by interleukin (IL)-13 and interferon gamma and inhibits tumor necrosis factor alpha-induced IL-8 release. J. Exp. Med. 1998, 187, 1285–1294. [Google Scholar] [CrossRef]
  16. Krishnamoorthy, S.; Recchiuti, A.; Chiang, N.; Yacoubian, S.; Lee, C.H.; Yang, R.; Petasis, N.A.; Serhan, C.N. Resolvin D1 binds human phagocytes with evidence for proresolving receptors. Proc. Natl. Acad. Sci. USA 2010, 107, 1660–1665. [Google Scholar] [CrossRef] [Green Version]
  17. Dalli, J.; Consalvo, A.P.; Ray, V.; Di Filippo, C.; D’Amico, M.; Mehta, N.; Perretti, M. Proresolving and tissue-protective actions of annexin A1-based cleavage-resistant peptides are mediated by formyl peptide receptor 2/lipoxin A4 receptor. J. Immunol. 2013, 190, 6478–6487. [Google Scholar] [CrossRef] [Green Version]
  18. Krishnamoorthy, S.; Recchiuti, A.; Chiang, N.; Fredman, G.; Serhan, C.N. Resolvin D1 receptor stereoselectivity and regulation of inflammation and proresolving microRNAs. Am. J. Pathol. 2012, 180, 2018–2027. [Google Scholar] [CrossRef] [Green Version]
  19. Arnardottir, H.H.; Dalli, J.; Norling, L.V.; Colas, R.A.; Perretti, M.; Serhan, C.N. Resolvin D3 Is Dysregulated in Arthritis and Reduces Arthritic Inflammation. J. Immunol. 2016, 197, 2362–2368. [Google Scholar] [CrossRef] [Green Version]
  20. Back, M.; Powell, W.S.; Dahlen, S.E.; Drazen, J.M.; Evans, J.F.; Serhan, C.N.; Shimizu, T.; Yokomizo, T.; Rovati, G.E. Update on leukotriene, lipoxin and oxoeicosanoid receptors: IUPHAR Review 7. Br. J. Pharmacol. 2014, 171, 3551–3574. [Google Scholar] [CrossRef] [Green Version]
  21. Arita, M.; Ohira, T.; Sun, Y.P.; Elangovan, S.; Chiang, N.; Serhan, C.N. Resolvin E1 selectively interacts with leukotriene B4 receptor BLT1 and ChemR23 to regulate inflammation. J. Immunol. 2007, 178, 3912–3917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Oh, S.F.; Dona, M.; Fredman, G.; Krishnamoorthy, S.; Irimia, D.; Serhan, C.N. Resolvin E2 formation and impact in inflammation resolution. J. Immunol. 2012, 188, 4527–4534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Merlin, J.; Park, J.; Vandekolk, T.H.; Fabb, S.A.; Allinne, J.; Summers, R.J.; Langmead, C.J.; Riddy, D.M. Multipathway In Vitro Pharmacological Characterization of Specialized Proresolving G Protein-Coupled Receptors. Mol. Pharmacol. 2022, 101, 246–256. [Google Scholar] [CrossRef]
  24. Finlay, D.B.; Joseph, W.R.; Grimsey, N.L.; Glass, M. GPR18 undergoes a high degree of constitutive trafficking but is unresponsive to N-Arachidonoyl Glycine. PeerJ 2016, 4, e1835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Chiang, N.; Dalli, J.; Colas, R.A.; Serhan, C.N. Identification of resolvin D2 receptor mediating resolution of infections and organ protection. J. Exp. Med. 2015, 212, 1203–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Bang, S.; Xie, Y.K.; Zhang, Z.J.; Wang, Z.; Xu, Z.Z.; Ji, R.R. GPR37 regulates macrophage phagocytosis and resolution of inflammatory pain. J. Clin. Investig. 2018, 128, 3568–3582. [Google Scholar] [CrossRef] [Green Version]
  27. Chiang, N.; Libreros, S.; Norris, P.C.; de la Rosa, X.; Serhan, C.N. Maresin 1 activates LGR6 receptor promoting phagocyte immunoresolvent functions. J. Clin. Investig. 2019, 129, 5294–5311. [Google Scholar] [CrossRef] [Green Version]
  28. Flak, M.B.; Koenis, D.S.; Sobrino, A.; Smith, J.; Pistorius, K.; Palmas, F.; Dalli, J. GPR101 mediates the pro-resolving actions of RvD5n-3 DPA in arthritis and infections. J. Clin. Investig. 2020, 130, 359–373. [Google Scholar] [CrossRef] [Green Version]
  29. Lavy, M.; Gauttier, V.; Poirier, N.; Barille-Nion, S.; Blanquart, C. Specialized Pro-Resolving Mediators Mitigate Cancer-Related Inflammation: Role of Tumor-Associated Macrophages and Therapeutic Opportunities. Front. Immunol. 2021, 12, 702785. [Google Scholar] [CrossRef]
  30. Sleire, L.; Forde, H.E.; Netland, I.A.; Leiss, L.; Skeie, B.S.; Enger, P.O. Drug repurposing in cancer. Pharmacol. Res. 2017, 124, 74–91. [Google Scholar] [CrossRef]
  31. Ramon, S.; Woeller, C.F.; Phipps, R.P. The influence of Cox-2 and bioactive lipids on hematological cancers. Curr. Angiogenesis 2013, 2, 135–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Liu, H.; Zeng, J.; Huang, W.; Xu, Q.; Ye, D.; Sun, R.; Zhang, D. Colorectal Cancer Is Associated with a Deficiency of Lipoxin A4, an Endogenous Anti-inflammatory Mediator. J. Cancer 2019, 10, 4719–4730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Thun, M.J.; Jacobs, E.J.; Patrono, C. The role of aspirin in cancer prevention. Nat. Rev. Clin. Oncol. 2012, 9, 259–267. [Google Scholar] [CrossRef] [PubMed]
  34. Claria, J.; Lee, M.H.; Serhan, C.N. Aspirin-triggered lipoxins (15-epi-LX) are generated by the human lung adenocarcinoma cell line (A549)-neutrophil interactions and are potent inhibitors of cell proliferation. Mol. Med. 1996, 2, 583–596. [Google Scholar] [CrossRef] [Green Version]
  35. Sulciner, M.L.; Gartung, A.; Gilligan, M.M.; Serhan, C.N.; Panigrahy, D. Targeting lipid mediators in cancer biology. Cancer Metastasis Rev. 2018, 37, 557–572. [Google Scholar] [CrossRef] [PubMed]
  36. Tsai, W.H.; Shih, C.H.; Wu, H.Y.; Chien, H.Y.; Chiang, Y.C.; Lai, S.L.; Hsu, S.C.; Kou, Y.R.; Hsu, H.C. Role of lipoxin A4 in the cell-to-cell interaction between all-trans retinoic acid-treated acute promyelocytic leukemic cells and alveolar macrophages. J. Cell. Physiol. 2012, 227, 1123–1129. [Google Scholar] [CrossRef] [PubMed]
  37. Chen, Y.; Hao, H.; He, S.; Cai, L.; Li, Y.; Hu, S.; Ye, D.; Hoidal, J.; Wu, P.; Chen, X. Lipoxin A4 and its analogue suppress the tumor growth of transplanted H22 in mice: The role of antiangiogenesis. Mol. Cancer Ther. 2010, 9, 2164–2174. [Google Scholar] [CrossRef] [Green Version]
  38. Halder, R.C.; Almasi, A.; Sagong, B.; Leung, J.; Jewett, A.; Fiala, M. Curcuminoids and omega-3 fatty acids with anti-oxidants potentiate cytotoxicity of natural killer cells against pancreatic ductal adenocarcinoma cells and inhibit interferon gamma production. Front. Physiol. 2015, 6, 129. [Google Scholar] [CrossRef]
  39. Lee, H.J.; Park, M.K.; Lee, E.J.; Lee, C.H. Resolvin D1 inhibits TGF-beta1-induced epithelial mesenchymal transition of A549 lung cancer cells via lipoxin A4 receptor/formyl peptide receptor 2 and GPR32. Int. J. Biochem. Cell Biol. 2013, 45, 2801–2807. [Google Scholar] [CrossRef]
  40. Canny, G.O.; Lessey, B.A. The role of lipoxin A4 in endometrial biology and endometriosis. Mucosal Immunol. 2013, 6, 439–450. [Google Scholar] [CrossRef] [Green Version]
  41. Zhou, X.Y.; Li, Y.S.; Wu, P.; Wang, H.M.; Cai, Z.Y.; Xu, F.Y.; Ye, D.Y. Lipoxin A(4) inhibited hepatocyte growth factor-induced invasion of human hepatoma cells. Hepatol. Res. Off. J. Jpn. Soc. Hepatol. 2009, 39, 921–930. [Google Scholar] [CrossRef]
  42. Ye, Y.; Scheff, N.N.; Bernabe, D.; Salvo, E.; Ono, K.; Liu, C.; Veeramachaneni, R.; Viet, C.T.; Viet, D.T.; Dolan, J.C.; et al. Anti-cancer and analgesic effects of resolvin D2 in oral squamous cell carcinoma. Neuropharmacology 2018, 139, 182–193. [Google Scholar] [CrossRef] [PubMed]
  43. Erdman, S.E.; Sohn, J.J.; Rao, V.P.; Nambiar, P.R.; Ge, Z.; Fox, J.G.; Schauer, D.B. CD4+CD25+ regulatory lymphocytes induce regression of intestinal tumors in ApcMin/+ mice. Cancer Res. 2005, 65, 3998–4004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Sulciner, M.L.; Serhan, C.N.; Gilligan, M.M.; Mudge, D.K.; Chang, J.; Gartung, A.; Lehner, K.A.; Bielenberg, D.R.; Schmidt, B.; Dalli, J.; et al. Resolvins suppress tumor growth and enhance cancer therapy. J. Exp. Med. 2018, 215, 115–140. [Google Scholar] [CrossRef]
  45. Gilligan, M.M.; Gartung, A.; Sulciner, M.L.; Norris, P.C.; Sukhatme, V.P.; Bielenberg, D.R.; Huang, S.; Kieran, M.W.; Serhan, C.N.; Panigrahy, D. Aspirin-triggered proresolving mediators stimulate resolution in cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 6292–6297. [Google Scholar] [CrossRef] [Green Version]
  46. Mattoscio, D.; Isopi, E.; Lamolinara, A.; Patruno, S.; Medda, A.; De Cecco, F.; Chiocca, S.; Iezzi, M.; Romano, M.; Recchiuti, A. Resolvin D1 reduces cancer growth stimulating a protective neutrophil-dependent recruitment of anti-tumor monocytes. J. Exp. Clin. Cancer Res. CR 2021, 40, 129. [Google Scholar] [CrossRef]
  47. Shan, K.; Feng, N.; Cui, J.; Wang, S.; Qu, H.; Fu, G.; Li, J.; Chen, H.; Wang, X.; Wang, R.; et al. Resolvin D1 and D2 inhibit tumour growth and inflammation via modulating macrophage polarization. J. Cell. Mol. Med. 2020, 24, 8045–8056. [Google Scholar] [CrossRef]
  48. Vannitamby, A.; Saad, M.I.; Aloe, C.; Wang, H.; Kumar, B.; Vlahos, R.; Selemidis, S.; Irving, L.; Steinfort, D.; Jenkins, B.J.; et al. Aspirin-Triggered Resolvin D1 Reduces Proliferation and the Neutrophil to Lymphocyte Ratio in a Mutant KRAS-Driven Lung Adenocarcinoma Model. Cancers 2021, 13, 3224. [Google Scholar] [CrossRef]
  49. Sun, L.; Wang, Y.; Wang, L.; Yao, B.; Chen, T.; Li, Q.; Liu, Z.; Liu, R.; Niu, Y.; Song, T.; et al. Resolvin D1 prevents epithelial-mesenchymal transition and reduces the stemness features of hepatocellular carcinoma by inhibiting paracrine of cancer-associated fibroblast-derived COMP. J. Exp. Clin. Cancer Res. CR 2019, 38, 170. [Google Scholar] [CrossRef] [PubMed]
  50. Prevete, N.; Liotti, F.; Illiano, A.; Amoresano, A.; Pucci, P.; de Paulis, A.; Melillo, R.M. Formyl peptide receptor 1 suppresses gastric cancer angiogenesis and growth by exploiting inflammation resolution pathways. Oncoimmunology 2017, 6, e1293213. [Google Scholar] [CrossRef] [Green Version]
  51. Liotti, F.; Marotta, M.; Sorriento, D.; Pagliuca, C.; Caturano, C.; Mantova, G.; Scaglione, E.; Salvatore, P.; Melillo, R.M.; Prevete, N. The probiotic Lactobacillus rhamnosus LGG restrains the angiogenic potential of colorectal carcinoma cells by activating a pro-resolving program via Formyl Peptide Receptor 1. Mol. Oncol. 2022. [Google Scholar] [CrossRef]
  52. Liotti, F.; Marotta, M.; Sorriento, D.; Pone, E.; Morra, F.; Melillo, R.M.; Prevete, N. Toll-Like Receptor 7 Mediates Inflammation Resolution and Inhibition of Angiogenesis in Non-Small Cell Lung Cancer. Cancers 2021, 13, 740. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, Z.; Cheng, Q.; Tang, K.; Sun, Y.; Zhang, K.; Zhang, Y.; Luo, S.; Zhang, H.; Ye, D.; Huang, B. Lipid mediator lipoxin A4 inhibits tumor growth by targeting IL-10-producing regulatory B (Breg) cells. Cancer Lett. 2015, 364, 118–124. [Google Scholar] [CrossRef]
  54. Simoes, R.L.; De-Brito, N.M.; Cunha-Costa, H.; Morandi, V.; Fierro, I.M.; Roitt, I.M.; Barja-Fidalgo, C. Lipoxin A4 selectively programs the profile of M2 tumor-associated macrophages which favour control of tumor progression. Int. J. Cancer 2017, 140, 346–357. [Google Scholar] [CrossRef]
  55. Schnittert, J.; Heinrich, M.A.; Kuninty, P.R.; Storm, G.; Prakash, J. Reprogramming tumor stroma using an endogenous lipid lipoxin A4 to treat pancreatic cancer. Cancer Lett. 2018, 420, 247–258. [Google Scholar] [CrossRef] [PubMed]
  56. Labani-Motlagh, A.; Ashja-Mahdavi, M.; Loskog, A. The Tumor Microenvironment: A Milieu Hindering and Obstructing Antitumor Immune Responses. Front. Immunol. 2020, 11, 940. [Google Scholar] [CrossRef]
  57. Galli, F.; Aguilera, J.V.; Palermo, B.; Markovic, S.N.; Nistico, P.; Signore, A. Relevance of immune cell and tumor microenvironment imaging in the new era of immunotherapy. J. Exp. Clin. Cancer Res. CR 2020, 39, 89. [Google Scholar] [CrossRef]
  58. Wang, J.; Li, D.; Cang, H.; Guo, B. Crosstalk between cancer and immune cells: Role of tumor-associated macrophages in the tumor microenvironment. Cancer Med. 2019, 8, 4709–4721. [Google Scholar] [CrossRef]
  59. Pages, F.; Galon, J.; Dieu-Nosjean, M.C.; Tartour, E.; Sautes-Fridman, C.; Fridman, W.H. Immune infiltration in human tumors: A prognostic factor that should not be ignored. Oncogene 2010, 29, 1093–1102. [Google Scholar] [CrossRef] [Green Version]
  60. Bremnes, R.M.; Al-Shibli, K.; Donnem, T.; Sirera, R.; Al-Saad, S.; Andersen, S.; Stenvold, H.; Camps, C.; Busund, L.T. The role of tumor-infiltrating immune cells and chronic inflammation at the tumor site on cancer development, progression, and prognosis: Emphasis on non-small cell lung cancer. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2011, 6, 824–833. [Google Scholar] [CrossRef] [Green Version]
  61. Petitprez, F.; Meylan, M.; de Reynies, A.; Sautes-Fridman, C.; Fridman, W.H. The Tumor Microenvironment in the Response to Immune Checkpoint Blockade Therapies. Front. Immunol. 2020, 11, 784. [Google Scholar] [CrossRef] [PubMed]
  62. Mattoscio, D.; Ferri, G.; Miccolo, C.; Chiocca, S.; Romano, M.; Recchiuti, A. Gene Expression of the D-Series Resolvin Pathway Predicts Activation of Anti-Tumor Immunity and Clinical Outcomes in Head and Neck Cancer. Int. J. Mol. Sci. 2022, 23, 6473. [Google Scholar] [CrossRef] [PubMed]
  63. Gartung, A.; Gilligan, M.M.; Bielenberg, D.R.; Fishbein, A.; Wells, S.; Huang, S.; Kieran, M.W.; Serhan, C.N.; Panigrahy, D. Synergy between Resolvins and Immune Checkpoint Blockade in a Novel Transplantable FANCC(−/−) Murine Head and Neck Tumor Model. Faseb J. 2019, 33, 496.10. [Google Scholar] [CrossRef]
  64. De Matteis, R.; Flak, M.B.; Gonzalez-Nunez, M.; Austin-Williams, S.; Palmas, F.; Colas, R.A.; Dalli, J. Aspirin activates resolution pathways to reprogram T cell and macrophage responses in colitis-associated colorectal cancer. Sci. Adv. 2022, 8, eabl5420. [Google Scholar] [CrossRef]
  65. Asha, K.; Balfe, N.; Sharma-Walia, N. Concurrent Control of the Kaposi’s Sarcoma-Associated Herpesvirus Life Cycle through Chromatin Modulation and Host Hedgehog Signaling: A New Prospect for the Therapeutic Potential of Lipoxin A4. J. Virol. 2020, 94, e02177-19. [Google Scholar] [CrossRef]
  66. Lecoultre, M.; Dutoit, V.; Walker, P.R. Phagocytic function of tumor-associated macrophages as a key determinant of tumor progression control: A review. J. Immunother. Cancer 2020, 8, e001408. [Google Scholar] [CrossRef]
  67. Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416. [Google Scholar] [CrossRef]
  68. Coffelt, S.B.; Wellenstein, M.D.; de Visser, K.E. Neutrophils in cancer: Neutral no more. Nat. Rev. Cancer 2016, 16, 431–446. [Google Scholar] [CrossRef] [Green Version]
  69. Powell, D.R.; Huttenlocher, A. Neutrophils in the Tumor Microenvironment. Trends Immunol. 2016, 37, 41–52. [Google Scholar] [CrossRef] [Green Version]
  70. Gentles, A.J.; Newman, A.M.; Liu, C.L.; Bratman, S.V.; Feng, W.; Kim, D.; Nair, V.S.; Xu, Y.; Khuong, A.; Hoang, C.D.; et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 2015, 21, 938–945. [Google Scholar] [CrossRef]
  71. Shaul, M.E.; Fridlender, Z.G. Tumour-associated neutrophils in patients with cancer. Nat. Rev. Clin. Oncol. 2019, 16, 601–620. [Google Scholar] [CrossRef] [PubMed]
  72. El Kebir, D.; Gjorstrup, P.; Filep, J.G. Resolvin E1 promotes phagocytosis-induced neutrophil apoptosis and accelerates resolution of pulmonary inflammation. Proc. Natl. Acad. Sci. USA 2012, 109, 14983–14988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Serhan, C.N. Pro-resolving lipid mediators are leads for resolution physiology. Nature 2014, 510, 92–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Doran, A.C.; Yurdagul, A., Jr.; Tabas, I. Efferocytosis in health and disease. Nat. Rev. Immunol. 2020, 20, 254–267. [Google Scholar] [CrossRef]
  75. Mills, C.D.; Kincaid, K.; Alt, J.M.; Heilman, M.J.; Hill, A.M. M-1/M-2 macrophages and the Th1/Th2 paradigm. J. Immunol. 2000, 164, 6166–6173. [Google Scholar] [CrossRef] [Green Version]
  76. Sica, A.; Mantovani, A. Macrophage plasticity and polarization: In vivo veritas. J. Clin. Investig. 2012, 122, 787–795. [Google Scholar] [CrossRef]
  77. Ohira, T.; Arita, M.; Omori, K.; Recchiuti, A.; Van Dyke, T.E.; Serhan, C.N. Resolvin E1 receptor activation signals phosphorylation and phagocytosis. J. Biol. Chem. 2010, 285, 3451–3461. [Google Scholar] [CrossRef] [Green Version]
  78. Trilleaud, C.; Gauttier, V.; Biteau, K.; Girault, I.; Belarif, L.; Mary, C.; Pengam, S.; Teppaz, G.; Thepenier, V.; Danger, R.; et al. Agonist anti-ChemR23 mAb reduces tissue neutrophil accumulation and triggers chronic inflammation resolution. Sci. Adv. 2021, 7, eabd1453. [Google Scholar] [CrossRef]
  79. Prieto, P.; Cuenca, J.; Traves, P.G.; Fernandez-Velasco, M.; Martin-Sanz, P.; Bosca, L. Lipoxin A4 impairment of apoptotic signaling in macrophages: Implication of the PI3K/Akt and the ERK/Nrf-2 defense pathways. Cell Death Differ. 2010, 17, 1179–1188. [Google Scholar] [CrossRef] [Green Version]
  80. Prescott, D.; McKay, D.M. Aspirin-triggered lipoxin enhances macrophage phagocytosis of bacteria while inhibiting inflammatory cytokine production. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 301, G487–G497. [Google Scholar] [CrossRef]
  81. Croasdell, A.; Thatcher, T.H.; Kottmann, R.M.; Colas, R.A.; Dalli, J.; Serhan, C.N.; Sime, P.J.; Phipps, R.P. Resolvins attenuate inflammation and promote resolution in cigarette smoke-exposed human macrophages. Am. J. Physiol. Lung Cell. Mol. Physiol. 2015, 309, L888–L901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Dalli, J.; Zhu, M.; Vlasenko, N.A.; Deng, B.; Haeggstrom, J.Z.; Petasis, N.A.; Serhan, C.N. The novel 13S,14S-epoxy-maresin is converted by human macrophages to maresin 1 (MaR1), inhibits leukotriene A4 hydrolase (LTA4H), and shifts macrophage phenotype. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2013, 27, 2573–2583. [Google Scholar] [CrossRef] [Green Version]
  83. Schmid, M.; Gemperle, C.; Rimann, N.; Hersberger, M. Resolvin D1 Polarizes Primary Human Macrophages toward a Proresolution Phenotype through GPR32. J. Immunol. 2016, 196, 3429–3437. [Google Scholar] [CrossRef] [Green Version]
  84. Herova, M.; Schmid, M.; Gemperle, C.; Hersberger, M. ChemR23, the receptor for chemerin and resolvin E1, is expressed and functional on M1 but not on M2 macrophages. J. Immunol. 2015, 194, 2330–2337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Correa, M.; Machado, J., Jr.; Carneiro, C.R.; Pesquero, J.B.; Bader, M.; Travassos, L.R.; Chammas, R.; Jasiulionis, M.G. Transient inflammatory response induced by apoptotic cells is an important mediator of melanoma cell engraftment and growth. Int. J. Cancer 2005, 114, 356–363. [Google Scholar] [CrossRef]
  86. Chaurio, R.A.; Munoz, L.E.; Maueroder, C.; Janko, C.; Harrer, T.; Furnrohr, B.G.; Niederweis, M.; Bilyy, R.; Schett, G.; Herrmann, M.; et al. The progression of cell death affects the rejection of allogeneic tumors in immune-competent mice-implications for cancer therapy. Front. Immunol. 2014, 5, 560. [Google Scholar] [CrossRef] [Green Version]
  87. Kim, S.H.; Saeidi, S.; Zhong, X.; Gwak, S.Y.; Muna, I.A.; Park, S.A.; Kim, S.H.; Na, H.K.; Joe, Y.; Chung, H.T.; et al. Breast cancer cell debris diminishes therapeutic efficacy through heme oxygenase-1-mediated inactivation of M1-like tumor-associated macrophages. Neoplasia 2020, 22, 606–616. [Google Scholar] [CrossRef]
  88. Weigert, A.; Mora, J.; Sekar, D.; Syed, S.; Brune, B. Killing Is Not Enough: How Apoptosis Hijacks Tumor-Associated Macrophages to Promote Cancer Progression. Adv. Exp. Med. Biol. 2016, 930, 205–239. [Google Scholar] [CrossRef]
  89. Karpisheh, V.; Nikkhoo, A.; Hojjat-Farsangi, M.; Namdar, A.; Azizi, G.; Ghalamfarsa, G.; Sabz, G.; Yousefi, M.; Yousefi, B.; Jadidi-Niaragh, F. Prostaglandin E2 as a potent therapeutic target for treatment of colon cancer. Prostaglandins Other Lipid Mediat. 2019, 144, 106338. [Google Scholar] [CrossRef]
  90. Hilligan, K.L.; Ronchese, F. Antigen presentation by dendritic cells and their instruction of CD4+ T helper cell responses. Cell. Mol. Immunol. 2020, 17, 587–599. [Google Scholar] [CrossRef]
  91. Chen, K.; Le, Y.; Liu, Y.; Gong, W.; Ying, G.; Huang, J.; Yoshimura, T.; Tessarollo, L.; Wang, J.M. A critical role for the g protein-coupled receptor mFPR2 in airway inflammation and immune responses. J. Immunol. 2010, 184, 3331–3335. [Google Scholar] [CrossRef] [Green Version]
  92. Kojima, F.; Kapoor, M.; Kawai, S.; Crofford, L.J. New insights into eicosanoid biosynthetic pathways: Implications for arthritis. Expert Rev. Clin. Immunol. 2006, 2, 277–291. [Google Scholar] [CrossRef] [PubMed]
  93. Aliberti, J.; Hieny, S.; Reis e Sousa, C.; Serhan, C.N.; Sher, A. Lipoxin-mediated inhibition of IL-12 production by DCs: A mechanism for regulation of microbial immunity. Nat. Immunol. 2002, 3, 76–82. [Google Scholar] [CrossRef] [PubMed]
  94. Hua, J.; Jin, Y.; Chen, Y.; Inomata, T.; Lee, H.; Chauhan, S.K.; Petasis, N.A.; Serhan, C.N.; Dana, R. The resolvin D1 analogue controls maturation of dendritic cells and suppresses alloimmunity in corneal transplantation. Investig. Ophthalmol. Vis. Sci. 2014, 55, 5944–5951. [Google Scholar] [CrossRef] [PubMed]
  95. Arita, M.; Yoshida, M.; Hong, S.; Tjonahen, E.; Glickman, J.N.; Petasis, N.A.; Blumberg, R.S.; Serhan, C.N. Resolvin E1, an endogenous lipid mediator derived from omega-3 eicosapentaenoic acid, protects against 2,4,6-trinitrobenzene sulfonic acid-induced colitis. Proc. Natl. Acad. Sci. USA 2005, 102, 7671–7676. [Google Scholar] [CrossRef] [Green Version]
  96. Sawada, Y.; Honda, T.; Hanakawa, S.; Nakamizo, S.; Murata, T.; Ueharaguchi-Tanada, Y.; Ono, S.; Amano, W.; Nakajima, S.; Egawa, G.; et al. Resolvin E1 inhibits dendritic cell migration in the skin and attenuates contact hypersensitivity responses. J. Exp. Med. 2015, 212, 1921–1930. [Google Scholar] [CrossRef] [Green Version]
  97. Vassiliou, E.K.; Kesler, O.M.; Tadros, J.H.; Ganea, D. Bone marrow-derived dendritic cells generated in the presence of resolvin E1 induce apoptosis of activated CD4+ T cells. J. Immunol. 2008, 181, 4534–4544. [Google Scholar] [CrossRef] [Green Version]
  98. Duffney, P.F.; Falsetta, M.L.; Rackow, A.R.; Thatcher, T.H.; Phipps, R.P.; Sime, P.J. Key roles for lipid mediators in the adaptive immune response. J. Clin. Investig. 2018, 128, 2724–2731. [Google Scholar] [CrossRef]
  99. Golubovskaya, V.; Wu, L. Different Subsets of T Cells, Memory, Effector Functions, and CAR-T Immunotherapy. Cancers 2016, 8, 36. [Google Scholar] [CrossRef] [Green Version]
  100. Geginat, J.; Paroni, M.; Maglie, S.; Alfen, J.S.; Kastirr, I.; Gruarin, P.; De Simone, M.; Pagani, M.; Abrignani, S. Plasticity of human CD4 T cell subsets. Front. Immunol. 2014, 5, 630. [Google Scholar] [CrossRef]
  101. Chiurchiu, V.; Leuti, A.; Dalli, J.; Jacobsson, A.; Battistini, L.; Maccarrone, M.; Serhan, C.N. Proresolving lipid mediators resolvin D1, resolvin D2, and maresin 1 are critical in modulating T cell responses. Sci. Transl. Med. 2016, 8, 353ra111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Ariel, A.; Li, P.L.; Wang, W.; Tang, W.X.; Fredman, G.; Hong, S.; Gotlinger, K.H.; Serhan, C.N. The docosatriene protectin D1 is produced by TH2 skewing and promotes human T cell apoptosis via lipid raft clustering. J. Biol. Chem. 2005, 280, 43079–43086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Ariel, A.; Chiang, N.; Arita, M.; Petasis, N.A.; Serhan, C.N. Aspirin-triggered lipoxin A4 and B4 analogs block extracellular signal-regulated kinase-dependent TNF-alpha secretion from human T cells. J. Immunol. 2003, 170, 6266–6272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Aoki, H.; Hisada, T.; Ishizuka, T.; Utsugi, M.; Ono, A.; Koga, Y.; Sunaga, N.; Nakakura, T.; Okajima, F.; Dobashi, K.; et al. Protective effect of resolvin E1 on the development of asthmatic airway inflammation. Biochem. Biophys. Res. Commun. 2010, 400, 128–133. [Google Scholar] [CrossRef] [PubMed]
  105. Aoki, H.; Hisada, T.; Ishizuka, T.; Utsugi, M.; Kawata, T.; Shimizu, Y.; Okajima, F.; Dobashi, K.; Mori, M. Resolvin E1 dampens airway inflammation and hyperresponsiveness in a murine model of asthma. Biochem. Biophys. Res. Commun. 2008, 367, 509–515. [Google Scholar] [CrossRef]
  106. Raphael, I.; Nalawade, S.; Eagar, T.N.; Forsthuber, T.G. T cell subsets and their signature cytokines in autoimmune and inflammatory diseases. Cytokine 2015, 74, 5–17. [Google Scholar] [CrossRef] [Green Version]
  107. Poisson, L.M.; Suhail, H.; Singh, J.; Datta, I.; Denic, A.; Labuzek, K.; Hoda, M.N.; Shankar, A.; Kumar, A.; Cerghet, M.; et al. Untargeted Plasma Metabolomics Identifies Endogenous Metabolite with Drug-like Properties in Chronic Animal Model of Multiple Sclerosis. J. Biol. Chem. 2015, 290, 30697–30712. [Google Scholar] [CrossRef] [Green Version]
  108. Wang, X.; Sumida, H.; Cyster, J.G. GPR18 is required for a normal CD8alphaalpha intestinal intraepithelial lymphocyte compartment. J. Exp. Med. 2014, 211, 2351–2359. [Google Scholar] [CrossRef] [Green Version]
  109. Chheda, Z.S.; Sharma, R.K.; Jala, V.R.; Luster, A.D.; Haribabu, B. Chemoattractant Receptors BLT1 and CXCR3 Regulate Antitumor Immunity by Facilitating CD8+ T Cell Migration into Tumors. J. Immunol. 2016, 197, 2016–2026. [Google Scholar] [CrossRef] [Green Version]
  110. Sharma, R.K.; Chheda, Z.; Jala, V.R.; Haribabu, B. Expression of leukotriene B(4) receptor-1 on CD8(+) T cells is required for their migration into tumors to elicit effective antitumor immunity. J. Immunol. 2013, 191, 3462–3470. [Google Scholar] [CrossRef] [Green Version]
  111. Ramon, S.; Baker, S.F.; Sahler, J.M.; Kim, N.; Feldsott, E.A.; Serhan, C.N.; Martinez-Sobrido, L.; Topham, D.J.; Phipps, R.P. The specialized proresolving mediator 17-HDHA enhances the antibody-mediated immune response against influenza virus: A new class of adjuvant? J. Immunol. 2014, 193, 6031–6040. [Google Scholar] [CrossRef] [PubMed]
  112. Ramon, S.; Gao, F.; Serhan, C.N.; Phipps, R.P. Specialized proresolving mediators enhance human B cell differentiation to antibody-secreting cells. J. Immunol. 2012, 189, 1036–1042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Kosaraju, R.; Guesdon, W.; Crouch, M.J.; Teague, H.L.; Sullivan, E.M.; Karlsson, E.A.; Schultz-Cherry, S.; Gowdy, K.; Bridges, L.C.; Reese, L.R.; et al. B Cell Activity Is Impaired in Human and Mouse Obesity and Is Responsive to an Essential Fatty Acid upon Murine Influenza Infection. J. Immunol. 2017, 198, 4738–4752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Teague, H.; Fhaner, C.J.; Harris, M.; Duriancik, D.M.; Reid, G.E.; Shaikh, S.R. n-3 PUFAs enhance the frequency of murine B-cell subsets and restore the impairment of antibody production to a T-independent antigen in obesity. J. Lipid Res. 2013, 54, 3130–3138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Cheng, Q.; Wang, Z.; Ma, R.; Chen, Y.; Yan, Y.; Miao, S.; Jiao, J.; Cheng, X.; Kong, L.; Ye, D. Lipoxin A4 protects against lipopolysaccharide-induced sepsis by promoting innate response activator B cells generation. Int. Immunopharmacol. 2016, 39, 229–235. [Google Scholar] [CrossRef]
  116. Lesokhin, A.M.; Merghoub, T.; Wolchok, J.D. Myeloid-derived suppressor sells and the efficacy of CD8(+) T-cell immunotherapy. Oncoimmunology 2013, 2, e22764. [Google Scholar] [CrossRef] [Green Version]
  117. Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef] [Green Version]
  118. Orentas, R.J.; Kohler, M.E.; Johnson, B.D. Suppression of anti-cancer immunity by regulatory T cells: Back to the future. Semin. Cancer Biol. 2006, 16, 137–149. [Google Scholar] [CrossRef]
  119. Luo, B.; Han, F.; Xu, K.; Wang, J.; Liu, Z.; Shen, Z.; Li, J.; Liu, Y.; Jiang, M.; Zhang, Z.Y.; et al. Resolvin D1 Programs Inflammation Resolution by Increasing TGF-beta Expression Induced by Dying Cell Clearance in Experimental Autoimmune Neuritis. J. Neurosci. Off. J. Soc. Neurosci. 2016, 36, 9590–9603. [Google Scholar] [CrossRef] [Green Version]
  120. Cheng, T.; Ding, S.; Liu, S.; Li, X.; Tang, X.; Sun, L. Resolvin D1 Improves the Treg/Th17 Imbalance in Systemic Lupus Erythematosus Through miR-30e-5p. Front. Immunol. 2021, 12, 668760. [Google Scholar] [CrossRef]
  121. Krishnamoorthy, N.; Burkett, P.R.; Dalli, J.; Abdulnour, R.E.; Colas, R.; Ramon, S.; Phipps, R.P.; Petasis, N.A.; Kuchroo, V.K.; Serhan, C.N.; et al. Cutting edge: Maresin-1 engages regulatory T cells to limit type 2 innate lymphoid cell activation and promote resolution of lung inflammation. J. Immunol. 2015, 194, 863–867. [Google Scholar] [CrossRef] [PubMed]
  122. Marques, R.M.; Gonzalez-Nunez, M.; Walker, M.E.; Gomez, E.A.; Colas, R.A.; Montero-Melendez, T.; Perretti, M.; Dalli, J. Loss of 15-lipoxygenase disrupts Treg differentiation altering their pro-resolving functions. Cell Death Differ. 2021, 28, 3140–3160. [Google Scholar] [CrossRef] [PubMed]
  123. Zhang, B.; Jia, H.; Liu, J.; Yang, Z.; Jiang, T.; Tang, K.; Li, D.; Huang, C.; Ma, J.; Shen, G.X.; et al. Depletion of regulatory T cells facilitates growth of established tumors: A mechanism involving the regulation of myeloid-derived suppressor cells by lipoxin A4. J. Immunol. 2010, 185, 7199–7206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Catalan, D.; Mansilla, M.A.; Ferrier, A.; Soto, L.; Oleinika, K.; Aguillon, J.C.; Aravena, O. Immunosuppressive Mechanisms of Regulatory B Cells. Front. Immunol. 2021, 12, 611795. [Google Scholar] [CrossRef]
  125. Schwartz, M.; Zhang, Y.; Rosenblatt, J.D. B cell regulation of the anti-tumor response and role in carcinogenesis. J. Immunother. Cancer 2016, 4, 40. [Google Scholar] [CrossRef] [Green Version]
  126. Xing, F.; Saidou, J.; Watabe, K. Cancer associated fibroblasts (CAFs) in tumor microenvironment. Front. Biosci. 2010, 15, 166–179. [Google Scholar] [CrossRef] [Green Version]
  127. Recchiuti, A.; Serhan, C.N. Pro-Resolving Lipid Mediators (SPMs) and Their Actions in Regulating miRNA in Novel Resolution Circuits in Inflammation. Front. Immunol. 2012, 3, 298. [Google Scholar] [CrossRef] [Green Version]
  128. Roach, K.M.; Feghali-Bostwick, C.A.; Amrani, Y.; Bradding, P. Lipoxin A4 Attenuates Constitutive and TGF-beta1-Dependent Profibrotic Activity in Human Lung Myofibroblasts. J. Immunol. 2015, 195, 2852–2860. [Google Scholar] [CrossRef] [Green Version]
  129. Chiang, N.; Arita, M.; Serhan, C.N. Anti-inflammatory circuitry: Lipoxin, aspirin-triggered lipoxins and their receptor ALX. Prostaglandins Leukot. Essent. Fat. Acids 2005, 73, 163–177. [Google Scholar] [CrossRef]
  130. Offertaler, L.; Mo, F.M.; Batkai, S.; Liu, J.; Begg, M.; Razdan, R.K.; Martin, B.R.; Bukoski, R.D.; Kunos, G. Selective ligands and cellular effectors of a G protein-coupled endothelial cannabinoid receptor. Mol. Pharmacol. 2003, 63, 699–705. [Google Scholar] [CrossRef] [Green Version]
  131. Qiu, H.; Johansson, A.S.; Sjostrom, M.; Wan, M.; Schroder, O.; Palmblad, J.; Haeggstrom, J.Z. Differential induction of BLT receptor expression on human endothelial cells by lipopolysaccharide, cytokines, and leukotriene B4. Proc. Natl. Acad. Sci. USA 2006, 103, 6913–6918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Kaur, J.; Adya, R.; Tan, B.K.; Chen, J.; Randeva, H.S. Identification of chemerin receptor (ChemR23) in human endothelial cells: Chemerin-induced endothelial angiogenesis. Biochem. Biophys. Res. Commun. 2010, 391, 1762–1768. [Google Scholar] [CrossRef] [Green Version]
  133. Cezar-de-Mello, P.F.; Nascimento-Silva, V.; Villela, C.G.; Fierro, I.M. Aspirin-triggered Lipoxin A4 inhibition of VEGF-induced endothelial cell migration involves actin polymerization and focal adhesion assembly. Oncogene 2006, 25, 122–129. [Google Scholar] [CrossRef] [Green Version]
  134. Fierro, I.M.; Kutok, J.L.; Serhan, C.N. Novel lipid mediator regulators of endothelial cell proliferation and migration: Aspirin-triggered-15R-lipoxin A(4) and lipoxin A(4). J. Pharmacol. Exp. Ther. 2002, 300, 385–392. [Google Scholar] [CrossRef] [PubMed]
  135. Paul-Clark, M.J.; Van Cao, T.; Moradi-Bidhendi, N.; Cooper, D.; Gilroy, D.W. 15-epi-lipoxin A4-mediated induction of nitric oxide explains how aspirin inhibits acute inflammation. J. Exp. Med. 2004, 200, 69–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Chattopadhyay, R.; Raghavan, S.; Rao, G.N. Resolvin D1 via prevention of ROS-mediated SHP2 inactivation protects endothelial adherens junction integrity and barrier function. Redox Biol. 2017, 12, 438–455. [Google Scholar] [CrossRef] [PubMed]
  137. Marginean, A.; Sharma-Walia, N. Lipoxins exert antiangiogenic and anti-inflammatory effects on Kaposi’s sarcoma cells. Transl. Res. J. Lab. Clin. Med. 2015, 166, 111–133. [Google Scholar] [CrossRef]
  138. Jin, Y.; Arita, M.; Zhang, Q.; Saban, D.R.; Chauhan, S.K.; Chiang, N.; Serhan, C.N.; Dana, R. Anti-angiogenesis effect of the novel anti-inflammatory and pro-resolving lipid mediators. Investig. Ophthalmol. Vis. Sci. 2009, 50, 4743–4752. [Google Scholar] [CrossRef]
  139. Prevete, N.; Liotti, F.; Visciano, C.; Marone, G.; Melillo, R.M.; de Paulis, A. The formyl peptide receptor 1 exerts a tumor suppressor function in human gastric cancer by inhibiting angiogenesis. Oncogene 2015, 34, 3826–3838. [Google Scholar] [CrossRef]
  140. Takeuchi, O.; Akira, S. Pattern recognition receptors and inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef] [Green Version]
  141. Prevete, N.; Liotti, F.; Marone, G.; Melillo, R.M.; de Paulis, A. Formyl peptide receptors at the interface of inflammation, angiogenesis and tumor growth. Pharmacol. Res. 2015, 102, 184–191. [Google Scholar] [CrossRef] [PubMed]
  142. Gobbetti, T.; Dalli, J.; Colas, R.A.; Federici Canova, D.; Aursnes, M.; Bonnet, D.; Alric, L.; Vergnolle, N.; Deraison, C.; Hansen, T.V.; et al. Protectin D1n-3 DPA and resolvin D5n-3 DPA are effectors of intestinal protection. Proc. Natl. Acad. Sci. USA 2017, 114, 3963–3968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Ungaro, F.; D’Alessio, S.; Danese, S. The Role of Pro-Resolving Lipid Mediators in Colorectal Cancer-Associated Inflammation: Implications for Therapeutic Strategies. Cancers 2020, 12, 2060. [Google Scholar] [CrossRef] [PubMed]
  144. Gravina, A.G.; Prevete, N.; Tuccillo, C.; De Musis, C.; Romano, L.; Federico, A.; de Paulis, A.; D’Argenio, G.; Romano, M. Peptide Hp(2-20) accelerates healing of TNBS-induced colitis in the rat. United Eur. Gastroenterol. J. 2018, 6, 1428–1436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Prevete, N.; de Paulis, A.; Sgambato, D.; Melillo, R.M.; D’Argenio, G.; Romano, L.; Zagari, R.M.; Romano, M. Role of Formyl Peptide Receptors in Gastrointestinal Healing. Curr. Pharm. Des. 2018, 24, 1966–1971. [Google Scholar] [CrossRef]
  146. Banna, G.L.; Torino, F.; Marletta, F.; Santagati, M.; Salemi, R.; Cannarozzo, E.; Falzone, L.; Ferrau, F.; Libra, M. Lactobacillus rhamnosus GG: An Overview to Explore the Rationale of Its Use in Cancer. Front. Pharmacol. 2017, 8, 603. [Google Scholar] [CrossRef] [Green Version]
  147. Alam, A.; Leoni, G.; Wentworth, C.C.; Kwal, J.M.; Wu, H.; Ardita, C.S.; Swanson, P.A.; Lambeth, J.D.; Jones, R.M.; Nusrat, A.; et al. Redox signaling regulates commensal-mediated mucosal homeostasis and restitution and requires formyl peptide receptor 1. Mucosal Immunol. 2014, 7, 645–655. [Google Scholar] [CrossRef] [Green Version]
  148. Alam, A.; Leoni, G.; Quiros, M.; Wu, H.; Desai, C.; Nishio, H.; Jones, R.M.; Nusrat, A.; Neish, A.S. The microenvironment of injured murine gut elicits a local pro-restitutive microbiota. Nat. Microbiol. 2016, 1, 15021. [Google Scholar] [CrossRef] [Green Version]
  149. Gao, J.; Li, Y.; Wan, Y.; Hu, T.; Liu, L.; Yang, S.; Gong, Z.; Zeng, Q.; Wei, Y.; Yang, W.; et al. A Novel Postbiotic From Lactobacillus rhamnosus GG with a Beneficial Effect on Intestinal Barrier Function. Front. Microbiol. 2019, 10, 477. [Google Scholar] [CrossRef] [Green Version]
  150. Orlando, A.; Linsalata, M.; Russo, F. Antiproliferative effects on colon adenocarcinoma cells induced by co-administration of vitamin K1 and Lactobacillus rhamnosus GG. Int. J. Oncol. 2016, 48, 2629–2638. [Google Scholar] [CrossRef] [Green Version]
  151. Escamilla, J.; Lane, M.A.; Maitin, V. Cell-free supernatants from probiotic Lactobacillus casei and Lactobacillus rhamnosus GG decrease colon cancer cell invasion in vitro. Nutr. Cancer 2012, 64, 871–878. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Effect of SPMs on TME components.
Figure 1. Effect of SPMs on TME components.
Cancers 14 03333 g001
Table 1. Cancer models and the mechanisms by which specific SPMs inhibit cancer progression by directly acting on tumor cells or by targeting different components of the TME.
Table 1. Cancer models and the mechanisms by which specific SPMs inhibit cancer progression by directly acting on tumor cells or by targeting different components of the TME.
SPMMechanism(s)Cancer ModelReferences
Direct effects on cancer cellsLXA4suppression of cancer cell growth and invasionlung[34]
suppression of cancer cell growth and invasionhepatocellular carcinoma[41]
reduction of tumor cell migrationacute cell leukaemia[41]
anti-estrogenic activityendometrium[40]
RvD1inhibition of cancer cell proliferationoral[42]
inhibition of epithelial to mesenchymal transitionlung[39]
Effects on TMERvD1targeting of regulatory cellscolon[43]
clearance of cell debris by macrophagepancreas, lung[44,45]
modulation of neutrophil phenotype and recruitment of anti-cancer monocytespapilloma[46]
modulation of TAM phenotypeprostate[47]
increase of NK functionpancreas[38]
reduction neutrophil infiltrationlung[48]
inhibition of CAF pro-tumorigenic mediatorsHCC[49]
reduction of angiogenic responsestomach, colon, lung[50,51,52]
RvD2clearance of cell debris by macrophagepancreas, lung[44,45]
modulation of TAM phenotypeprostate[47]
RvE1clearance of cell debris by macrophagepancreas, lung[44,45]
LXA4targeting Breg cells and increasing cytotoxic T cell activityliver[53]
modulation of TAM phenotypemelanoma[54]
inhibition of CAF precursorspancreas[55]
LXB4reduction of angiogenic responsestomach, colon, lung[50,51,52]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liotti, F.; Marotta, M.; Melillo, R.M.; Prevete, N. The Impact of Resolution of Inflammation on Tumor Microenvironment: Exploring New Ways to Control Cancer Progression. Cancers 2022, 14, 3333. https://doi.org/10.3390/cancers14143333

AMA Style

Liotti F, Marotta M, Melillo RM, Prevete N. The Impact of Resolution of Inflammation on Tumor Microenvironment: Exploring New Ways to Control Cancer Progression. Cancers. 2022; 14(14):3333. https://doi.org/10.3390/cancers14143333

Chicago/Turabian Style

Liotti, Federica, Maria Marotta, Rosa Marina Melillo, and Nella Prevete. 2022. "The Impact of Resolution of Inflammation on Tumor Microenvironment: Exploring New Ways to Control Cancer Progression" Cancers 14, no. 14: 3333. https://doi.org/10.3390/cancers14143333

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop