Abstract
Dendritic cells (DC) play unique and diverse roles in the tumor occurrence, development, progression and response to therapy. First of all, DC can actively uptake tumor-associated antigens, process them and present antigenic peptides to T cells inducing and maintaining tumor-specific T cell responses. DC interaction with different immune effector cells may also support innate antitumor immunity, as well as humoral responses also known to inhibit tumor development in certain cases. On the other hand, DC are recruited to the tumor site by specific tumor-derived and stroma-derived factors, which may also impair DC maturation, differentiation and function, thus resulting in the deficient formation of antitumor immune response or development of DC-mediated tolerance and immune suppression. Identification of DC-stimulating and DC-suppressing/polarizing factors in the tumor environment and the mechanism of DC modulation are important for designing effective DC-based vaccines and for recovery of immunodeficient resident DC responsible for maintenance of clinically relevant antitumor immunity in patients with cancer. DC-targeting tumor-derived factors and their effects on resident and administered DC in the tumor milieu are described and discussed in this review.
Keywords: Immunosuppression, Dendritic cells, Tolerance, Tumor immunoenvironment, Regulatory dendritic cells, Cytokines
Introduction
The development and progression of a tumor is a complex process that includes multidirectorial interactions between malignant and non-malignant cells occurring systemically in lymphoid and non-lymphoid tissues and in the local tumor milieu. Active and passive communications between different cell types direct formation of the tumor microenvironment and may result in either elimination of cancerous cells or their growth and spreading. Immune effector cells are able to recognize tumor cells, initiate the antitumor immune reactions and erase tumors, while immune regulatory cells support tumor escape from immune attack by different mechanisms and pathways. Tumor- and tumor stroma-derived factors include both “danger signals” activating immune responses and various molecules suppressing and polarizing immune cells, thus leading to tumor survival and progression. Although many of these factors are identified and characterized, there are no effective tools to control their levels or expression in the tumor environment in order to shift their balance from the protumorigenic to proimmunogenic outcome in a clinical setting.
Dendritic cells (DC) are well-characterized antigen-presenting cells known to play a key role in initiating and maintaining the antitumor immunity, bridging innate and adaptive immune responses, and sustaining immune tolerance. The proven role of DC in the tumor immunosurveillance supported designing and testing of DC-based vaccines for different types of cancer, which, however, demonstrated different levels of efficacy and feasibility [1]. Further analyses of DC functioning and longevity in the tumor environment revealed that DC may be inhibited or functionally polarized and thus unable to support the development of antitumor immunity [2]. Furthermore, new data demonstrated that DC polarized in the tumor milieu were immunosuppressive and tolerogenic, and supported tumor growth and spreading [3]. It is well established now that there are several phenotypic functional subsets of tumor-associated DC: fully functional immunostimulatory DC, abnormal functionally deficient DC, dying DC and polarized immunosuppressive regulatory DC (regDC) [4, 5]. Thus, identification of tumor-derived factors targeting DC, understanding the mechanisms of DC modulation in cancer and revealing the means of changing the balance between DC-altering molecules in the tumor environment will not only improve the efficacy of DC vaccine and allow development of novel DC-based and DC-targeting therapies, but also provide novel perspectives for revealing complex cellular interactions during cancer development and therapy [6].
Tumor-derived DC-activating factors
DC are proven to play a central role in the initiation and regulation of antitumor immunity—proactive anticancer immunosurveillance mechanism—which suggests that in many instances, DC uptake malignant cells or their bodies and respond to local stimulation leading to DC maturation, emigration from the tumor mass and homing in the regional lymph nodes to present tumor antigens to antigen-specific T cells. Immunogenic signals released by dying tumor cells are able to prompt antigen uptake, antigen processing and antigen presentation by DC. Cancerous cells may undergo cell death due to hypoxia, nutrient deprivation and innate immune responses resulting in the release of host-derived damage-associated molecular pattern (DAMP), sometimes termed alarmins or danger signals, as an indicator of dying, stressed or dead cells. Such type of apoptotic and necroptotic cell death is characterized by the induction of endoplasmic reticulum stress and autophagy, exposure of calreticulin on the cell surface, the secretion of adenosine triphophosphate and the release of the chromatin-binding protein high-mobility group box 1 (HMGB1) [7]. DAMP-associated proteins include endogenous molecules, such as HMGB1, heat-shock proteins (HSP), histones, the S100 family of proteins and serum amyloid A, whereas nonprotein DAMP includes heparin sulfate, uric acid, ATP, DNA (genomic and mitochondrial DNA) and RNA [8].
HMGB1 is one of the HMGB family members consisting of three domains—HMGB A box, HMG B box and the C-terminal acidic tail [9]. HMGB1 functions to protect cells from injury in normal conditions, but may serve as DAMP in inflammation, cancer, sepsis, trauma and autoimmunity. HMGB1 receptors include toll-like receptors (TLR) TLR-2, TLR-4 and TLR-9, receptor for advanced glycation end product (RAGE), CD24, α-synuclein filaments, proteoglycans, the T cell immunoglobulin domain and mucin domain-3 (TIM-3), N-methyl-d-aspartate receptor (NMDAR) and the triggering receptor expressed on myeloid cell-1 (TREM1) [9, 10]. HMGB1, via its B box domain, has been reported to induce phenotypic maturation of DC demonstrated by increased expression of CD83, CD54, CD80, CD40, CD58 and MHC class II molecules and decreased CD206 expression [11, 12]. The B box also caused increased secretion of the proinflammatory cytokines IL-12, IL-6, IL-1α, IL-8, TNF-α and RANTES. Saenz et al. [13] showed that peptide Hp91, whose sequence corresponds to an area within the B box domain of HMGB1, activates DC and acts as an adjuvant in vivo. Hp91-induced DC activation is mediated by a MyD88- and TLR4-dependent pathway involving p38 MAPK and NF-κB. Thus, HMGB1 released by dying cells may be a signal of tissue or cellular injury that, when sensed by DC, induces or enhances an immune reaction. Interestingly, new results revealed that HMGB1 secretion during cervical carcinogenesis could support the achievement of a tolerogenic activity by plasmacytoid DC [14].
There is rising evidence that S100 proteins, which are structurally similar to calmodulin, may also act as DAMP and might play a role similar to HMGB1 in regulating tissue injury and inflammation operating via the same receptors [15]. This is a family of 24 related calcium-binding proteins that display differential tissue and cell-type expression profiles in vertebrates. The functions of intracellular S100 proteins have been extensively studied, and most members participate in the regulation of a diverse set of intracellular processes such as cell cycle progression, cell proliferation, differentiation and migration, protein degradation, cytoskeletal organization, protein phosphorylation and transcriptional factor activity [16]. Recently, it has been shown that S100 proteins released from different cell types may serve as useful markers of disease activity including such diseases as asthma, chronic obstructive pulmonary disease, colitis, rheumatoid arthritis, Alzheimer’s disease and cancer [17]. Furthermore, S100A8 and S100A9, with well-proven activity in inflammation, have been progressively recognized not only as markers, but also as new regulators with essential roles in modulating tumor growth and progression [18].
The S100 genes of the A series are sited in a region of chromosome 1 which is prone to rearrangements, linking these S100 proteins with cancer [19]. Marked changes in the expression of S100B, S100A2, S100A4, S100A6, S100A8/A9 and S100P have been reported for different types of cancer [20, 21]. For instance, serum S100B protein has been suggested as a biomarker of malignant melanoma [22], and serum S100A2 and S100A6 may have the potential for being used as a NSCLC biomarker [23]. Although it is known that S100A8 and S100A9 proteins can regulate the differentiation and function of different myeloid cells, including DC [24], new data allow speculating that other S100 proteins released in the tumor microenvironment may also affect DC activity. For instance, DC have been shown to require S100A4 for activating T cells [25].
Another well-established DAMP in cancer are heat-shock proteins, which function as intracellular chaperones and have been implicated in the activation and bridging of innate and adaptive immune systems. Studies based on molecular weight and phylogenetics have separated five major HSP families; however, only HSP96, HSP90, HSP70, HSP110 and HSP170 have proven immunogenic interactions as membrane-bound and extracellular components [26]. Stimulation of cross-presentation is achieved through binding of HSP to distinct cell surface receptors followed by antigen internalization, processing and presentation. For instance, HSP-CD91 binding on immune cells can assist in DC maturation, secretion of cytokines and T helper cell priming [27]. HSP70 binds to immature DC and induces their maturation as evidenced by an increase in CD40, CD86 and CD83 expression and enhanced ability to present model antigen to specific CTL [28]. Tumor-derived antigenic peptide or protein complexes with HSP can also be involved in immune activation via cross-presenting the chaperoned proteins to DC when they are released from necrotic tumor cells or secreted in response to cellular stress [29].
Tumor-derived DC-suppressive factors
Dendritic cells, together with other immune cells, such cytotoxic T cells, macrophages, NK cells, γδ T cells and B cells, play an important role in cancer immunosurveillance—the ability to recognize and destroy newly ascending malignant cells. As cancer is linked to certain defects in immunosurveillance, newly appearing cancerous cells progressively gain a variety of mechanisms to escape immune recognition and elimination that favor further tumor survival and progression. Immune escape is the consequence of a direct or indirect cross talk between malignant cells and the immune system occurring in the local tumor microenvironment as well as at a systemic level. Soluble mediators produced by both tumor cells and stromal cells represent crucial performers in this cross talk (Table 1).
Table 1.
Factors | Effect on DC | Known signaling pathways | References |
---|---|---|---|
VEGF | Inhibition of differentiation, maturation, migration; induction of apoptosis | NF-κB | [32] |
TGF-β | Suppression of maturation and function | SMAD, STAT3 | [40, 45] |
GDF-15 | Inhibition of costimulatory molecule expression, IL-12 production, T cell stimulatory activity; increasing of TGF-β expression | [46] | |
RANKL | Tolerogenic repolarization | [65] | |
IL-10 | Inhibition of maturation, induction of tolerogenic phenotype, redirection of differentiation | p38 MAPK, STAT3 | [52, 54, 55] |
IL-8 | Impaired migration | PI3K, AKT, PKC, MAPK | [115] |
IL-6 | Effect on growth, longevity, differentiation, migration | MAPK, STAT3, NF-κB | [56, 60] |
CSF-1 | Suppression of differentiation | PI3K | [64] |
CCL2 | Regulation of migration and maturation | PI3K | [67] |
MIP3a | NF-κB, MAPK, JAK/STAT | [68] | |
SDF-1 | [69] | ||
MUC1 | Regulation of chemotaxis, induction of semimaturation | [74] | |
PSA | Inhibition of maturation, longevity and function | MAPK, STAT3, NF-κB | [71] |
HCG | Induction of tolerogenic phenotype | [76] | |
Gangliosides | Inhibition of dendropoiesis, longevity and function | IRAK-M | [79, 80] |
Prostaglandins | Regulation of differentiation | RAS-MAPK, PI3K/AKT, PKA | [116] |
Lactic acid | Inhibition of differentiation | NF-κB | [89, 90] |
Adenosine | Alteration of differentiation and function and attraction to Treg cells | Epac1–Rap1-dependent pathways | [91] [94] [93] |
Neuropeptides | Regulation of generation, function and longevity | Multiple pathways | [3, 109] |
HLA-G | Induce tolerance | [117] | |
Microvesicles | Impair differentiation | Multiple pathways | [112, 113] |
Tumor-derived DC-suppressive factors: growth factors, cytokines and chemokines
Vascular endothelial growth factor (VEGF) is a secreted heparin-binding protein produced by the majority of tumors and responsible for the formation of tumor neovasculature and tumor development [30]. The increased serum level of VEGF correlates with poor prognosis in patients with different types of cancer [31]. VEGF seems to be the first identified tumor-derived factor affecting DC: It inhibits differentiation and maturation of DC via binding and activation of two tyrosine kinase receptors, VEGFR-1 and VEGFR-2 [32]. VEGF inhibits the development and maturation of DC in vitro and in vivo by blocking NF-κB activation in hematopoietic progenitor cells [33, 34]. Not only exposure of cultured DC to VEGF affects DC differentiation by induction of apoptosis, alteration of DC phenotypic profile and increasing CXCR4 expression, DC numbers in the peripheral blood of patients with cancer inversely correlate with VEGF serum levels [35]. VEGF molecules are also known to regulate DC migration and homing by recruiting immature myeloid cells and immature DC from the bone marrow to the tumor site [36].
As inhibition of the VEGF pathways has become an appreciated approach in the treatment of cancers, it is important to mention that inhibition of VEGF signaling by using VEGF-Trap, which significantly increased the proportion of mature DC in patients with refractory solid tumors, enrolled into a phase I clinical trials [37]. Direct in vitro studies showed that the inhibition of VEGF expression in breast cancer cells by small interfering RNA effectively recovered differentiation and maturation of DC inhibited by tumor cells and increased DC potential to induced tumor-specific cytotoxic T cells [38].
Different members of the transforming growth factor beta (TGF-β) superfamily have been shown to regulate activity and differentiation of DC [39]. Indeed, DC express both type-1 TGF receptors, including activin receptor 1 (ACVR1), ACVR1B, bone morphogenetic protein receptor (BMPR) 1A, BMPR1B and TGF-β receptor (TGFBR)1, and type-2 TGF receptors, including TGFBR2, BMPR2, ACVR2A and ACVR2B to respond to different TGF-β family ligands. DC also express all components of the principal signaling cascades, such as the R-SMAD, receptor-regulated SMAD—SMAD1/5/8 and SMAD2/3, and the Co-SMAD—SMAD4 [40]. However, with the exception of TGF-β, the role of different members of the TGF-β superfamily in DC regulation in the tumor microenvironment has not been yet established. TGF-β overexpression in the tumor milieu may be associated with suppressed DC maturation and function resulting in the defects of the tumor-specific immune responses [41]. Tumor-derived TGF-β has been shown to be responsible for downregulating the expression of DC maturation markers CD83, CD80, CD86 and MHC II molecules [42] and inhibiting the expression of proinflammatory cytokines inducing DC maturation, such as TNF-α, IL-1, IL-12 and IFN-α, while promoting the release of regulatory cytokines, including TGF-β itself [43]. TGF-β family ligands also affect DC motility and migration through the regulation of the expression of chemokines and chemokine receptors [39]. TGF-β might also induce apoptosis in DC [44].
These and other data showing decreased maturation of DC under the influence of TGF-β suggest that tumor-derived TGF-β can significantly suppress DC function and their ability to initiate antitumor immune responses. Additional results revealed that TGF-β can not only inhibit DC function, but may also polarize DC into immunosuppressive tolerogenic phenotype. These regulatory DC (regDC) display a strong ability to suppress proliferation of effector T cells and induce differentiation of T cells into regulatory (Treg) T cells [3, 5]. For instance, TGF-β has been shown to increase the expression of programmed death ligand 1 (PD-L1) and signal transducers and activators of transcription 3 (STAT3) in DC in both a time- and dose-dependent manner [45].
The DC-targeting effects of other members of the TGF-β superfamily produced in the tumor microenvironment are not well investigated. We have evaluated whether the ligands from the activin subfamily are expressed by tumor cell lines and can alter DC phenotype and function. We found that Nodal that is produced by different tumor cells and shares the SMAD2/3 signaling pathway with TGF-β1 can polarize DC into the regulatory phenotype by upregulating COX2 expression (Agassandian et al. in preparation).
Growth differentiation factor-15 [GDF-15, macrophage inhibitory cytokine-1 (MIC-1)], a divergent member of the TGF-β family of proteins, was detected in tissues and serum samples in patients with glioblastoma, and ovarian, prostate, gastric and colorectal cancers. GDF-15 was shown to inhibit the expression of CD83, CD86 and HLA-DR on DC, downregulate IL-12 and upregulate TGF-β1 production and activate phagocytic but inhibit T cell stimulatory activity of DC in vitro. Furthermore, GDF-15 suppressed DC potential to stimulate tumor-specific immune responses in vivo [46].
Interleukin-10 (IL-10), also known as human cytokine synthesis inhibitory factor (CSIF), is an anti-inflammatory cytokine primarily produced by monocytes and lymphocytes. However, IL-10 release has been reported from some tumors including melanoma, multiple myeloma and lung cancer [47, 48]. Tumor-derived IL-10 has been shown to have an inhibitory effect on DC maturation and the T cell stimulatory ability of DC [49]. In addition, increased serum levels of IL-10 also correlate with profound numerical deficiency and immature phenotype of circulating DC subsets in patients with hepatocellular carcinoma, indicating a dominant correlation between tumor-derived IL-10 and defects in DC differentiation [50].
The inhibitory effect of IL-10 on the expression of costimulatory and MHC molecules on DC has been well documented [51]. For instance, tumor-derived IL-10 not only inhibits DC maturation and longevity and induces DC tolerance, but also inhibits CD40 expression, suppresses CD40-dependent IL-12 production, decreases chemokine receptor expression, blocks antigen presentation and induces upregulation of B7-H1 expression on DC [52, 53]. It is likely that p38 MAPK and STAT3 pathways are involved in the inhibition of DC by tumor-derived IL-10 [54]. Recent microarray studies showed that IL-10 not only inhibited DC function but also redirected differentiation of DC into cells with a different phenotype leading to a decreased pool of DC precursors [55].
IL-6, originally identified as a B cell differentiation factor, is known to display a plethora of effects on cell growth, differentiation, longevity and migration during immune responses, hematopoiesis and inflammation [56]. Overproduction of IL-6 is proved to be associated with a functional defect in DC from cancer patients [57]. For instance, increased plasma levels of IL-8 and IL-6 were detected in epithelial ovarian cancer patients, and production of both cytokines by cultured epithelial ovarian cancer cell lines was also reported [58]. Interestingly, immunosuppression of human DC by culture supernatant of ovarian tumor cells was reversed when the production of IL-6 and IL-8 was blocked. Furthermore, tumor-derived IL-6 affects the differentiation of hematopoietic progenitor cells and monocytes, including the macrophage and DC in vitro [59], and may be responsible for tolerogenic phenotype of DC [60]. It has been reported that IL-6-associated DC suppression could be normalized by JAK2/STAT3 inhibitor AG490 [54]. As STAT3 has been shown to be involved in the effect of IL-6 on the differentiation and maturation of DC, the tumor-induced phosphorylated STAT3 in DC could be regarded as a promising target for cancer immunotherapy [60].
Macrophage colony-stimulating factor (M-CSF), also known as colony-stimulating factor 1 (CSF-1), a major regulator of the mononuclear phagocytic lineage, is expressed in human breast and renal cell carcinomas, and its increased expression is a predictor of a poor prognosis [61]. CSF-1 not only modulates tumor progression and metastasis by recruiting and regulating macrophages, but also suppresses the differentiation of DC [62]. In addition, tumor-derived CSF-1 may inhibit the differentiation of hematopoietic progenitor CD34+ cells into DC and induce cord blood monocyte to differentiate into tolerogenic DC [63]. Modulation of DC differentiation by CSF-1 is mediated by the PI3K-dependent pathway and delayed caspase activation in monocytes [64].
New data revealed receptor activator of nuclear factor κ-B ligand (RANKL), a TNF family member, as a DC-targeting tumor-derived factor that downregulated IL-12 and upregulated IL-10 production by DC and polarized DC into regDC that induce FoxP3+ Treg cell differentiation [65].
It is well known that the recruitment of different subsets of myeloid cells varies according to the chemokine receptor expression profile and depends on the maturation stage of a particular cell population [53]. Several tumor-derived chemokines may target DC altering their migration and maturation statuses. Immature DC can be recruited into tumor inner location by tumor-derived chemotactic factors including CCL2, CCL20/MIP3a, CCL25, CCL5, CXCL12, CXCL1 and CXCL5, while mature DC reside in the surrounding areas of the tumor [66, 67]. These data suggest that the tumor-derived chemokines, produced by malignant or stromal cells, play an important role in the location and homing of tumor-infiltrating DC, as well as the maintenance of the immature status of DC. For instance, human ovarian epithelial tumor cells express high levels of CXCL12, also known as stromal cell-derived factor-1 (SDF-1), which binds to CXCR4 expressed on precursor of plasmacytoid DC and induces their chemotaxis, transmigration and adhesion [68]. The binding of CXCL12 to CXCR4 induces intracellular signaling through several different pathways initiating signaling cascades regulating chemotaxis, survival and proliferation [69]. Importantly, new data demonstrate that melanoma-derived factors can change maturation and activation of tissue-resident DC subsets and that the extent to which DC function is changed by these factors correlates with the in vivo tumorigenicity of melanomas [70].
Tumor-derived DC-suppressive factors: tumor “antigens”
Prostate-specific antigen (PSA), a serine protease overexpressed in most prostate cancers, was the first tumor-associated antigen shown to inhibit maturation, longevity and function of DC [71]. Addition of active PSA to DC cultures resulted in significant inhibition of DC generation (dendropoiesis) and maturation, shown by the levels of expression of CD83, CD80, CD86 and HLA-DR molecules. The ability of DC to induce T cell proliferation was also inhibited by PSA-treated DC. Other study also showed that the endogenous factors presented in the serum of patients with prostate cancer inhibited the generation of functionally active DC from CD14+ monocyte in vitro. This inhibition of DC maturation by serum from prostate cancer patients has a positive relationship with the levels of serum-free PSA [72], suggesting that PSA may be involved in impairment of resident DC in prostate cancer.
Cell surface-associated Mucin 1 (MUC1) is a glycoprotein overexpressed in many tumor cells. In normal cells, MUC1 forms a protective layer against the attack of microbes and toxic chemicals; however, oversecretion of MUC1 provides cancerous cells with an increased invasiveness, metastasis and resistance to effective immune response [73]. It has been shown that MUC1 could chemoattract immature DC to the tumor site and induce semimaturation of DC subverting DC function. When cultured with MUC1 glycoprotein, human monocyte-derived DC displayed upregulated expression of D83, CD80, CD86, CD40 and MHC class II; however, these DC also produced greater amounts of IL-6, TNF-α and IL-10, but fail to make IL-12. When these DC were cocultured with CD4 + T cells, they induced production of IL-13 and IL-5 and lower levels of IL-2, thus failing to induce a type 1 response [74]. Other studies also showed that tumor-derived mucin profoundly affected the cytokine expression in monocyte-derived DC and transferred them into IL-10highIL-12low regDC with a poor ability to induce protective Th1 responses [75]. These findings provide evidence that tumor-derived MUC1 may be responsible for the impaired DC maturation and function in certain types of cancer, identifying MUC1 as an additional mechanism of tumor escape from immune surveillance and disclosing the existence of tolerogenic DC in MAC1-positive cancers.
Finally, human chorionic gonadotropin (HCG), which serves as an important tumor marker for trophoblastic disease, choriocarcinoma and testicular cancer, has been shown to upregulate the expression of indoleamine-2, 3-dioxygenase (IDO) in DC [76]. IDO is the rate-limiting enzyme in the degradation pathway of tryptophan, an essential amino acid required for cell proliferation, was reported to be the mechanism of T cell suppression induced by IDO-expressing DC [77].
Tumor-derived DC-suppressive factors: other molecules
Gangliosides, found predominantly in the nervous system, are also expressed by the neuroectodermal cell-originated tumors as membrane-bound or shaded glycosphingolipids and can be detected in the tumor microenvironment and the bloodstream. Gangliosides act as cell surface receptors and markers and also participate in intercellular communication, cell signaling, cell cycling and cell motility [78]. It is well documented that neuroblastoma-derived gangliosides regulate the development of the tumor immunity by inhibiting dendropoiesis, longevity and function of DC [79]. Melanoma-produced gangliosides were also reported to impair DC differentiation and induce their apoptosis [80, 81].
Prostanoids, a subclass of eicosanoids consisting of the prostaglandins, prostacyclins and thromboxanes, are known mediators of inflammatory, anaphylactic reactions and vasoconstriction. Prostanoids have been found elevated in many tumors and implicated in tumor-associated subversion of the immune functions. For instance, prostaglandin PGE2 has been proposed as the principal prostanoid associated with colorectal tumors since PGE2 levels and the activity of cyclooxygenases (COX) are elevated in patients with colon cancer and correlate with tumor size and patient survival [82]. Furthermore, PGE2 was found to be responsible for the reduced differentiation of DC from CD34+ precursors [83]. Furthermore, PGE2 can serve as a mediator of DC tolerance since it upregulates the expression of IDO1 in DC resulting in the differentiation of Treg cells and in the inhibition of antigen-specific stimulatory potential of DC [84].
Polyamines (putrescine, spermidine and spermine) are naturally occurring aliphatic cations essential for cell growth and have been reported to be increased in prostate, colon and breast cancers [85, 86]. Spermine has been shown to induce altered maturation and impaired function in DC in vitro, while a significant inverse correlation between spermine level and the percentage of IL-12-expressing DC was found in patients with breast cancer [87].
Tumor-derived lactic acid, the end product of glycolysis, is known to affect cancerous cells, adjacent stroma and endothelial cells in the local tumor microenvironment reprogramming metabolism, promoting tumor inflammation and stimulating tumor angiogenesis [88]. On the other side, lactic acid is known as an important agent affecting DC by inhibiting IL-12 production and antigen presentation in the tumor environment, which may markedly support tumor escape from immune recognition [89]. Moreover, high lactate concentrations distress the differentiation of DC from monocytes decreasing the inflammatory phenotype of DC and inducing the production of IL-10 [90].
Recently, a metabolic stress- or hypoxia-associated adenosine accumulation has been suggested as one of the important drivers for an inhibition of antitumor immune response. Adenosine concentrations in tumor tissue are in the 50–100 μM range, while in normal tissues, they are found to be in the range of 10–100 nM [91]. After adenosine is released into the extracellular areas, it exerts numerous immunomodulatory effects via adenosine receptors expressed on different immune cells, including DC. Adenosine could alter differentiation of DC precursors into CD11c+Gr-1+ DC with a strong stimulatory effect on Th17 cells [92]. Adenosine-differentiated DC expressed a number of angiogenic, proinflammatory and immunosuppressive molecules, including TGF-β, IL-10, VEGF, IL-6, IL-8, COX-2 and IDO, and could support tumor growth if injected into tumors in mice [93]. Adenosine may also attract DC to Treg cells via Epac1–Rap1-dependent pathways rendering them less stimulatory [94].
New reports began to uncover an intricate role of lipids and lipid accumulation in DC functioning and how the triglycerides (triacylglycerol, TAG) in the context of tumors may alter DC activity and longevity. As DC develop and mature, they take on a “lacy” appearance composing an amplified presence of fat and glycogen-containing lipid body droplets [95], and lipid production and consumption play critical roles in DC biology [96]. It is therefore of interest that elevated levels of lipids, particularly TAG, were described in substantial proportion of DC in mice bearing lymphoma and breast and colon cancers and patients with cancer [97]. Lipid accumulation in DC was due to an increased uptake of extracellular lipids induced by the upregulation of scavenger receptor A, and lipid-laden DC displayed a reduced potential to process antigens. This receptor is primarily responsible for the uptake of modified lipids, and several molecular species of oxygenated lipids in tumor-bearing animals responsible for their uptake and accumulation in DC via scavenger receptor A-dependent pathway have been recently identified [98]. Similarly, it has been recently reported that mesothelioma promotes lipid acquisition by DC, which was accompanied by reduced antigen processing and elevated expression of the costimulatory molecules and production of IL-10 [99].
Elevated expression of the lipoprotein lipase (LPL) and the fatty acid-binding protein (FABP4), as well as increased serum levels of triacylglycerol in radiation-induced tumors, may explain the mechanisms of lipid accumulation in DC in tumor-bearing hosts [100]. Furthermore, tumor-associated DC have been shown to display an activation of the unfolded protein response (UPR) seen as the appearance of high levels of an endoplasmic reticulum (ER) stress response factor XBP1 (X-box-binding protein 1). XBP1 activation, attributed to reactive oxygen species (ROS) in tumor, which induces lipid peroxidation, induced a triglyceride biosynthetic program in tumor-associated DC causing abnormal lipid accumulation and consequent suppression of DC ability to maintain antitumor T cell responses [101]. Interestingly, XBP1 is not only an important component of UPR, but also an essential nuclear transcription factor. Many XBP1 target genes are fatty acid synthesis enzymes. Enriched production of fatty acids results in the formation of lipid droplets inside the cytoplasm and extension of the ER compartment due to efficient intracellular membrane formation [102].
Many neuropeptides are known to be synthesized and released by tumor cell lines and primary tumor cells and are detected in the tumor microenvironment [103]. Interestingly, some of these regulatory peptides may also affect function of DC and influence the development of antitumor immune responses. For instance, substance P induces tumor cell proliferation, migration, invasion, intratumor angiogenesis and metastasis development [104], and suppresses phagocytic activity of monocyte-derived and resident DC [105]. Neuropeptide Y may function as a growth and angiogenic peptide controlling the inflammatory and immunologic tumor responses [106]; its expression was found to correlate with the progression of cutaneous melanoma upregulating tumor invasiveness [107]. Neuropeptide Y was also shown to induce a Th2 polarizing profile of DC through the increased expression of IL-6 and IL-10 [108]. Tumor-derived bombesin, neuromedin B and gastrin-releasing peptide were reported to inhibit maturation of DC assessed as downregulation of CD40, CD80 and CD86 expression, decreased IL-12 production and attenuated potential to stimulate T cell proliferation [109].
Interestingly, new data revealed that death receptor 6 (DR6), a member of the TNF receptor superfamily known to be overexpressed on many tumor cells, may affect DC generation and function. It was shown that DR6, which is expressed on tumor cells and may be cleaved from the surface of tumor cells by the membrane-associated matrix metalloproteinase (MMP)-14, could induce apoptosis in >50 % of monocytes differentiating into DC and changed phenotype and cytokine expression in the resulting immature DC [110].
Tumor-derived microvesicles are the heterogeneous group of membrane-bound particles that are shed from the surface of tumor cells into the extracellular environment. Proteins, lipids, glycoproteins, glycolipids, peptides, RNA, microRNA and DNA are included in this complex cargo, which suggests that microvesicles can use multipronged mechanisms to regulate cell interactions in the tumor milieu facilitating tumor progression [111]. The growing body of evidence demonstrates that tumor-derived microvesicles or exosomes can alter myeloid cell function in the tumor microenvironment by impairing monocyte differentiation into DC and promoting the generation of a myeloid immunosuppressive cell subset [112]. For instance, exosomes purified from the lung cancer biopsies and containing the epidermal growth factor receptor (EGFR) have been shown to induce tolerogenic DC, which stimulated the differentiation of tumor antigen-specific Treg cells that could suppress the tumor-specific CD8 + T cells [113]. Furthermore, pancreatic cancer-derived exosomal miRNA has been recently reported to inhibit mRNA expression in DC resulting in decreased expression of MHC molecules and induction of immune tolerance [114].
Conclusions
Dendritic cells, the strongest functional antigen-presenting cells, essentially contribute to the induction and regulation of innate and adaptive immunity. By interacting with NK, NKT, T and B lymphocytes, macrophages, neutrophils, mast cells and various non-immune cells, DC play a central role in regulating immunologic and tolerogenic responses maintaining the stability of immune homeostasis. Furthermore, providing a critical link between the innate and adaptive immunity and regulating both the humoral and the cellular immune response, DC are primary involved in the developing and sustaining of cancer immunosurveillance and may profoundly impact tumor development and progression in patients. Therefore, it is not surprising that DC are also a key target and important player in the different pathways evolved by tumor cells to escape immune recognition and elimination. New insights gained over the last decades have revealed multiple mechanisms of immune regulation during tumor growth and progression, with DC likely to achieve a unique role in forming an immunosuppressive and tolerogenic milieu that supports cancer progression and blocks antitumor immunity. Although numerous studies identified and characterized various molecules produced in the tumor microenvironment that can suppress DC generation and function or polarize DC phenotype (Table 1), the most interesting and probably important question in this regard has not yet been answered—why do different tumors with quite diverse microenvironment utilize identical mechanisms to affect DC and why these pathways may be rather dissimilar in tumors of the same origin or the same type?
In relation to this question, there is an interesting fact that some of tumor-derived factors may display an opposite effect on DC depending on their concentration, the presence of other factors and cells or the stage of tumor development. Probably, TNF-α and prostaglandins are the most interesting examples from this category.
The balance between DC-stimulating and DC-suppressing factors in the tumor microenvironment at each particular time of tumor development and progression represents a unique parameter of maintaining immunologic activity in the tumor milieu (Fig. 1). Knowledge of intercellular and intracellular circuits that shape immunogenic and tolerogenic phenotype of DC in cancer will provide necessary insights for developing adjuvant treatments to alleviate immunosuppression in the tumor microenvironment and improving the clinical efficacy of cancer vaccines and alternative immunotherapies for cancer.
Acknowledgments
This work was supported in part by the National Institutes of Health (NIH), Grant RO1 CA154369 (to Shurin).
Abbreviations
- ACVR1
Activin receptor 1
- BMPR
Bone morphogenetic protein receptor
- COX
Cyclooxygenases
- CSF-1
Colony-stimulating factor 1
- CSIF
Cytokine synthesis inhibitory factor
- CTL
Cytotoxic T cell(s)
- DAMP
Damage-associated molecular pattern
- DC
Dendritic cell(s)
- DR6
Death receptor 6
- ER
Endoplasmic reticulum
- FABP4
Fatty acid-binding protein
- GDF-15
Growth differentiation factor-15
- HCG
Human chorionic gonadotropin
- HMGB1
Chromatin-binding protein high-mobility group box 1
- HSP
Heat-shock protein(s)
- IDO
Indoleamine-2, 3-dioxygenase
- IL-10
Interleukin-10
- LPL
Lipoprotein lipase
- M-CSF
Macrophage colony-stimulating factor
- MIC-1
Macrophage inhibitory cytokine-1
- MMP
Matrix metalloproteinase
- MUC1
Mucin 1
- NMDAR
N-methyl-d-aspartate receptor
- PD-L1
Programmed death ligand 1
- PSA
Prostate-specific antigen
- RAGE
Receptor for advanced glycation end product
- regDC
Regulatory dendritic cell(s)
- ROS
Reactive oxygen species
- SDF-1
Stromal cell-derived factor-1
- STAT3
Signal transducers and activators of transcription 3
- TGF-β
Transforming growth factor beta
- TGFBR
TGF-β receptor
- TIM-3
T cell immunoglobulin domain and mucin domain-3
- TLR
Toll-like receptor
- Treg
Regulatory T cell(s)
- TREM1
Triggering receptor expressed on myeloid cell-1
- UPR
Unfolded protein response
- VEGF
Vascular endothelial growth factor
- XBP1
X-box-binding protein 1
Compliance with ethical standards
Conflict of interest
The authors declare that they have no conflict of interest.
Footnotes
This paper is a Focussed Research Review based on a presentation given at the Fourth International Conference on Cancer Immunotherapy and Immunomonitoring (CITIM 2015), held in Ljubljana, Slovenia, 27th–30th April 2015. It is part of a series of Focussed Research Reviews and meeting report in Cancer Immunology, Immunotherapy.
References
- 1.Zhong H, Shurin MR, Han B. Optimizing dendritic cell-based immunotherapy for cancer. Expert Rev Vaccin. 2007;6(3):333–345. doi: 10.1586/14760584.6.3.333. [DOI] [PubMed] [Google Scholar]
- 2.Ma Y, Shurin GV, Gutkin DW, Shurin MR. Tumor associated regulatory dendritic cells. Semin Cancer Biol. 2012;22(4):298–306. doi: 10.1016/j.semcancer.2012.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Shurin GV, Ma Y, Shurin MR. Immunosuppressive mechanisms of regulatory dendritic cells in cancer. Cancer Microenviron. 2013;6(2):159–167. doi: 10.1007/s12307-013-0133-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Shurin GV, Ouellette CE, Shurin MR. Regulatory dendritic cells in the tumor immunoenvironment. Cancer Immunol Immunother. 2012;61(2):223–230. doi: 10.1007/s00262-011-1138-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ma Y, Shurin GV, Peiyuan Z, Shurin MR. Dendritic cells in the cancer microenvironment. J Cancer. 2013;4(1):36–44. doi: 10.7150/jca.5046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Shurin MR, Naiditch H, Zhong H, Shurin GV. Regulatory dendritic cells: new targets for cancer immunotherapy. Cancer Biol Ther. 2011;11(11):988–992. doi: 10.4161/cbt.11.11.15543. [DOI] [PubMed] [Google Scholar]
- 7.Garg AD, Dudek-Peric AM, Romano E, Agostinis P. Immunogenic cell death. Int J Dev Biol. 2015;59(1–3):131–140. doi: 10.1387/ijdb.150061pa. [DOI] [PubMed] [Google Scholar]
- 8.Bianchi ME. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol. 2007;81(1):1–5. doi: 10.1189/jlb.0306164. [DOI] [PubMed] [Google Scholar]
- 9.Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, Sun X, Wang H, Wang Q, Tsung A, Billiar TR, Zeh HJ, 3rd, Lotze MT, Tang D. HMGB1 in health and disease. Mol Aspects Med. 2014;40:1–116. doi: 10.1016/j.mam.2014.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol. 2010;28:367–388. doi: 10.1146/annurev.immunol.021908.132603. [DOI] [PubMed] [Google Scholar]
- 11.Saenz R, Souza Cda S, Huang CT, Larsson M, Esener S, Messmer D. HMGB1-derived peptide acts as adjuvant inducing immune responses to peptide and protein antigen. Vaccine. 2010;28(47):7556–7562. doi: 10.1016/j.vaccine.2010.08.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Saenz R, Futalan D, Leutenez L, Eekhout F, Fecteau JF, Sundelius S, Sundqvist S, Larsson M, Hayashi T, Minev B, Carson D, Esener S, Messmer B, Messmer D. TLR4-dependent activation of dendritic cells by an HMGB1-derived peptide adjuvant. J Transl Med. 2014;12:211. doi: 10.1186/1479-5876-12-211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Saenz R, Messmer B, Futalan D, Tor Y, Larsson M, Daniels G, Esener S, Messmer D. Activity of the HMGB1-derived immunostimulatory peptide Hp91 resides in the helical C-terminal portion and is enhanced by dimerization. Mol Immunol. 2014;57(2):191–199. doi: 10.1016/j.molimm.2013.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Demoulin S, Herfs M, Somja J, Roncarati P, Delvenne P, Hubert P. HMGB1 secretion during cervical carcinogenesis promotes the acquisition of a tolerogenic functionality by plasmacytoid dendritic cells. Int J Cancer. 2015;137(2):345–358. doi: 10.1002/ijc.29389. [DOI] [PubMed] [Google Scholar]
- 15.Chen B, Miller AL, Rebelatto M, Brewah Y, Rowe DC, Clarke L, Czapiga M, Rosenthal K, Imamichi T, Chen Y, Chang CS, Chowdhury PS, Naiman B, Wang Y, Yang D, Humbles AA, Herbst R, Sims GP. S100A9 induced inflammatory responses are mediated by distinct damage associated molecular patterns (DAMP) receptors in vitro and in vivo. PLoS ONE. 2015;10(2):e0115828. doi: 10.1371/journal.pone.0115828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Donato R, Cannon BR, Sorci G, Riuzzi F, Hsu K, Weber DJ, Geczy CL. Functions of S100 proteins. Curr Mol Med. 2013;13(1):24–57. doi: 10.2174/156652413804486214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Lee TH, Jang AS, Park JS, Kim TH, Choi YS, Shin HR, Park SW, Uh ST, Choi JS, Kim YH, Kim Y, Kim S, Chung IY, Jeong SH, Park CS. Elevation of S100 calcium binding protein A9 in sputum of neutrophilic inflammation in severe uncontrolled asthma. Ann Allergy Asthma Immunol. 2013;111(4):268–275. doi: 10.1016/j.anai.2013.06.028. [DOI] [PubMed] [Google Scholar]
- 18.Srikrishna G. S100A8 and S100A9: new insights into their roles in malignancy. J Innate Immun. 2012;4(1):31–40. doi: 10.1159/000330095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Engelkamp D, Schafer BW, Mattei MG, Erne P, Heizmann CW. Six S100 genes are clustered on human chromosome 1q21: identification of two genes coding for the two previously unreported calcium-binding proteins S100D and S100E. Proc Natl Acad Sci USA. 1993;90(14):6547–6551. doi: 10.1073/pnas.90.14.6547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Zhu L, Kohda F, Nakahara T, Chiba T, Tsuji G, Hachisuka J, Ito T, Tu Y, Moroi Y, Uchi H, Furue M. Aberrant expression of S100A6 and matrix metalloproteinase 9, but not S100A2, S100A4, and S100A7, is associated with epidermal carcinogenesis. J Dermatol Sci. 2013;72(3):311–319. doi: 10.1016/j.jdermsci.2013.07.005. [DOI] [PubMed] [Google Scholar]
- 21.Basso D, Fogar P, Plebani M. The S100A8/A9 complex reduces CTLA4 expression by immature myeloid cells: implications for pancreatic cancer-driven immunosuppression. Oncoimmunology. 2013;2(6):e24441. doi: 10.4161/onci.24441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Harpio R, Einarsson R. S100 proteins as cancer biomarkers with focus on S100B in malignant melanoma. Clin Biochem. 2004;37(7):512–518. doi: 10.1016/j.clinbiochem.2004.05.012. [DOI] [PubMed] [Google Scholar]
- 23.Wang T, Liang Y, Thakur A, Zhang S, Yang T, Chen T, Gao L, Chen M, Ren H. Diagnostic significance of S100A2 and S100A6 levels in sera of patients with non-small cell lung cancer. Tumour Biol. 2015 doi: 10.1007/s13277-015-4057-z. [DOI] [PubMed] [Google Scholar]
- 24.Averill MM, Barnhart S, Becker L, Li X, Heinecke JW, Leboeuf RC, Hamerman JA, Sorg C, Kerkhoff C, Bornfeldt KE. S100A9 differentially modifies phenotypic states of neutrophils, macrophages, and dendritic cells: implications for atherosclerosis and adipose tissue inflammation. Circulation. 2011;123(11):1216–1226. doi: 10.1161/CIRCULATIONAHA.110.985523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bruhn S, Fang Y, Barrenas F, Gustafsson M, Zhang H, Konstantinell A, Kronke A, Sonnichsen B, Bresnick A, Dulyaninova N, Wang H, Zhao Y, Klingelhofer J, Ambartsumian N, Beck MK, Nestor C, Bona E, Xiang Z, Benson M. A generally applicable translational strategy identifies S100A4 as a candidate gene in allergy. Sci Transl Med. 2014;6(218):218ra4. doi: 10.1126/scitranslmed.3007410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ampie L, Choy W, Lamano JB, Fakurnejad S, Bloch O, Parsa AT. Heat shock protein vaccines against glioblastoma: from bench to bedside. J Neurooncol. 2015;123(3):441–448. doi: 10.1007/s11060-015-1837-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Pawaria S, Binder RJ. CD91-dependent programming of T-helper cell responses following heat shock protein immunization. Nat Commun. 2011;2:521. doi: 10.1038/ncomms1524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kuppner MC, Gastpar R, Gelwer S, Nossner E, Ochmann O, Scharner A, Issels RD. The role of heat shock protein (hsp70) in dendritic cell maturation: hsp70 induces the maturation of immature dendritic cells but reduces DC differentiation from monocyte precursors. Eur J Immunol. 2001;31(5):1602–1609. doi: 10.1002/1521-4141(200105)31:5<1602::AID-IMMU1602>3.0.CO;2-W. [DOI] [PubMed] [Google Scholar]
- 29.Binder RJ, Srivastava PK. Peptides chaperoned by heat-shock proteins are a necessary and sufficient source of antigen in the cross-priming of CD8 + T cells. Nat Immunol. 2005;6(6):593–599. doi: 10.1038/ni1201. [DOI] [PubMed] [Google Scholar]
- 30.Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9(6):669–676. doi: 10.1038/nm0603-669. [DOI] [PubMed] [Google Scholar]
- 31.Toi M, Matsumoto T, Bando H. Vascular endothelial growth factor: its prognostic, predictive, and therapeutic implications. Lancet Oncol. 2001;2(11):667–673. doi: 10.1016/S1470-2045(01)00556-3. [DOI] [PubMed] [Google Scholar]
- 32.Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, Kavanaugh D, Carbone DP. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med. 1996;2(10):1096–1103. doi: 10.1038/nm1096-1096. [DOI] [PubMed] [Google Scholar]
- 33.Gabrilovich D, Ishida T, Oyama T, Ran S, Kravtsov V, Nadaf S, Carbone DP. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood. 1998;92(11):4150–4166. [PubMed] [Google Scholar]
- 34.Shi Y, Yu P, Zeng D, Qian F, Lei X, Zhao Y, Tang B, Hao Y, Luo H, Chen J, Tan Y. Suppression of vascular endothelial growth factor abrogates the immunosuppressive capability of murine gastric cancer cells and elicits antitumor immunity. FEBS J. 2014;281(17):3882–3893. doi: 10.1111/febs.12923. [DOI] [PubMed] [Google Scholar]
- 35.Della Porta M, Danova M, Rigolin GM, Brugnatelli S, Rovati B, Tronconi C, Fraulini C, Russo Rossi A, Riccardi A, Castoldi G. Dendritic cells and vascular endothelial growth factor in colorectal cancer: correlations with clinicobiological findings. Oncology. 2005;68(2–3):276–284. doi: 10.1159/000086784. [DOI] [PubMed] [Google Scholar]
- 36.Kim R, Emi M, Tanabe K, Arihiro K. Tumor-driven evolution of immunosuppressive networks during malignant progression. Cancer Res. 2006;66(11):5527–5536. doi: 10.1158/0008-5472.CAN-05-4128. [DOI] [PubMed] [Google Scholar]
- 37.Fricke I, Mirza N, Dupont J, Lockhart C, Jackson A, Lee JH, Sosman JA, Gabrilovich DI. Vascular endothelial growth factor-trap overcomes defects in dendritic cell differentiation but does not improve antigen-specific immune responses. Clin Cancer Res. 2007;13(16):4840–4848. doi: 10.1158/1078-0432.CCR-07-0409. [DOI] [PubMed] [Google Scholar]
- 38.Wang H, Zhang L, Zhang S, Li Y. Inhibition of vascular endothelial growth factor by small interfering RNA upregulates differentiation, maturation and function of dendritic cells. Exper Therap Med. 2015;9(1):120–124. doi: 10.3892/etm.2014.2059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Seeger P, Musso T, Sozzani S. The TGF-beta superfamily in dendritic cell biology. Cytokine Growth Fact Rev. 2015;26(6):647–657. doi: 10.1016/j.cytogfr.2015.06.002. [DOI] [PubMed] [Google Scholar]
- 40.Yasmin N, Konradi S, Eisenwort G, Schichl YM, Seyerl M, Bauer T, Stockl J, Strobl H. Beta-Catenin promotes the differentiation of epidermal Langerhans dendritic cells. J Invest Derm. 2013;133(5):1250–1259. doi: 10.1038/jid.2012.481. [DOI] [PubMed] [Google Scholar]
- 41.Brown RD, Pope B, Murray A, Esdale W, Sze DM, Gibson J, Ho PJ, Hart D, Joshua D. Dendritic cells from patients with myeloma are numerically normal but functionally defective as they fail to up-regulate CD80 (B7-1) expression after huCD40LT stimulation because of inhibition by transforming growth factor-beta1 and interleukin-10. Blood. 2001;98(10):2992–2998. doi: 10.1182/blood.V98.10.2992. [DOI] [PubMed] [Google Scholar]
- 42.Kel JM, Girard-Madoux MJ, Reizis B, Clausen BE. TGF-beta is required to maintain the pool of immature Langerhans cells in the epidermis. J Immunol. 2010;185(6):3248–3255. doi: 10.4049/jimmunol.1000981. [DOI] [PubMed] [Google Scholar]
- 43.Lievens D, Habets KL, Robertson AK, Laouar Y, Winkels H, Rademakers T, Beckers L, Wijnands E, Boon L, Mosaheb M, Ait-Oufella H, Mallat Z, Flavell RA, Rudling M, Binder CJ, Gerdes N, Biessen EA, Weber C, Daemen MJ, Kuiper J, Lutgens E. Abrogated transforming growth factor beta receptor II (TGFbetaRII) signalling in dendritic cells promotes immune reactivity of T cells resulting in enhanced atherosclerosis. Eur Heart J. 2013;34(48):3717–3727. doi: 10.1093/eurheartj/ehs106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ito M, Minamiya Y, Kawai H, Saito S, Saito H, Nakagawa T, Imai K, Hirokawa M, Ogawa J. Tumor-derived TGFbeta-1 induces dendritic cell apoptosis in the sentinel lymph node. J Immun. 2006;176(9):5637–5643. doi: 10.4049/jimmunol.176.9.5637. [DOI] [PubMed] [Google Scholar]
- 45.Song S, Yuan P, Wu H, Chen J, Fu J, Li P, Lu J, Wei W. Dendritic cells with an increased PD-L1 by TGF-beta induce T cell anergy for the cytotoxicity of hepatocellular carcinoma cells. Int Immunopharmacol. 2014;20(1):117–123. doi: 10.1016/j.intimp.2014.02.027. [DOI] [PubMed] [Google Scholar]
- 46.Zhou Z, Li W, Song Y, Wang L, Zhang K, Yang J, Zhang W, Su H, Zhang Y. Growth differentiation factor-15 suppresses maturation and function of dendritic cells and inhibits tumor-specific immune response. PLoS ONE. 2013;8(11):e78618. doi: 10.1371/journal.pone.0078618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Smith DR, Kunkel SL, Burdick MD, Wilke CA, Orringer MB, Whyte RI, Strieter RM. Production of interleukin-10 by human bronchogenic carcinoma. Am J Pathol. 1994;145(1):18–25. [PMC free article] [PubMed] [Google Scholar]
- 48.Gu ZJ, Costes V, Lu ZY, Zhang XG, Pitard V, Moreau JF, Bataille R, Wijdenes J, Rossi JF, Klein B. Interleukin-10 is a growth factor for human myeloma cells by induction of an oncostatin M autocrine loop. Blood. 1996;88(10):3972–3986. [PubMed] [Google Scholar]
- 49.Kim KD, Lim HY, Lee HG, Yoon DY, Choe YK, Choi I, Paik SG, Kim YS, Yang Y, Lim JS. Apolipoprotein A-I induces IL-10 and PGE2 production in human monocytes and inhibits dendritic cell differentiation and maturation. Biochem Biophys Res Commun. 2005;338(2):1126–1136. doi: 10.1016/j.bbrc.2005.10.065. [DOI] [PubMed] [Google Scholar]
- 50.Beckebaum S, Zhang X, Chen X, Yu Z, Frilling A, Dworacki G, Grosse-Wilde H, Broelsch CE, Gerken G, Cicinnati VR. Increased levels of interleukin-10 in serum from patients with hepatocellular carcinoma correlate with profound numerical deficiencies and immature phenotype of circulating dendritic cell subsets. Clin Cancer Res. 2004;10(21):7260–7269. doi: 10.1158/1078-0432.CCR-04-0872. [DOI] [PubMed] [Google Scholar]
- 51.Williams LM, Ricchetti G, Sarma U, Smallie T, Foxwell BM. Interleukin-10 suppression of myeloid cell activation: a continuing puzzle. Immunology. 2004;113(3):281–292. doi: 10.1111/j.1365-2567.2004.01988.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Shurin MR, Yurkovetsky ZR, Tourkova IL, Balkir L, Shurin GV. Inhibition of CD40 expression and CD40-mediated dendritic cell function by tumor-derived IL-10. Int J Cancer. 2002;101(1):61–68. doi: 10.1002/ijc.10576. [DOI] [PubMed] [Google Scholar]
- 53.Shurin MR, Shurin GV, Lokshin A, Yurkovetsky ZR, Gutkin DW, Chatta G, Zhong H, Han B, Ferris RL. Intratumoral cytokines/chemokines/growth factors and tumor infiltrating dendritic cells: friends or enemies? Cancer Metas Rev. 2006;25(3):333–356. doi: 10.1007/s10555-006-9010-6. [DOI] [PubMed] [Google Scholar]
- 54.Oosterhoff D, Lougheed S, van de Ven R, Lindenberg J, van Cruijsen H, Hiddingh L, Kroon J, van den Eertwegh AJ, Hangalapura B, Scheper RJ, de Gruijl TD. Tumor-mediated inhibition of human dendritic cell differentiation and function is consistently counteracted by combined p38 MAPK and STAT3 inhibition. Oncoimmunology. 2012;1(5):649–658. doi: 10.4161/onci.20365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Schwarz AM, Banning-Eichenseer U, Seidel K, Mauz-Korholz C, Korholz D, Staege MS. Impact of interleukin-10 on phenotype and gene expression during early monocyte differentiation into dendritic cells. Anticancer Res. 2013;33(11):4791–4798. [PubMed] [Google Scholar]
- 56.Hirano T. Interleukin 6 and its receptor: 10 years later. Int Rev Immunol. 1998;16(3–4):249–284. doi: 10.3109/08830189809042997. [DOI] [PubMed] [Google Scholar]
- 57.Ratta M, Fagnoni F, Curti A, Vescovini R, Sansoni P, Oliviero B, Fogli M, Ferri E, Della Cuna GR, Tura S, Baccarani M, Lemoli RM. Dendritic cells are functionally defective in multiple myeloma: the role of interleukin-6. Blood. 2002;100(1):230–237. doi: 10.1182/blood.V100.1.230. [DOI] [PubMed] [Google Scholar]
- 58.Nishio H, Yaguchi T, Sugiyama J, Sumimoto H, Umezawa K, Iwata T, Susumu N, Fujii T, Kawamura N, Kobayashi A, Park J, Aoki D, Kawakami Y. Immunosuppression through constitutively activated NF-kappaB signalling in human ovarian cancer and its reversal by an NF-kappaB inhibitor. Br J Cancer. 2014;110(12):2965–2974. doi: 10.1038/bjc.2014.251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Chomarat P, Banchereau J, Davoust J, Palucka AK. IL-6 switches the differentiation of monocytes from dendritic cells to macrophages. Nat Immunol. 2000;1(6):510–514. doi: 10.1038/82763. [DOI] [PubMed] [Google Scholar]
- 60.Alshamsan A. Induction of tolerogenic dendritic cells by IL-6-secreting CT26 colon carcinoma. Immunopharmacol Immunotoxicol. 2012;34(3):465–469. doi: 10.3109/08923973.2011.625034. [DOI] [PubMed] [Google Scholar]
- 61.Yang L, Wu Q, Xu L, Zhang W, Zhu Y, Liu H, Xu J, Gu J. Increased expression of colony stimulating factor-1 is a predictor of poor prognosis in patients with clear-cell renal cell carcinoma. BMC Cancer. 2015;15:67. doi: 10.1186/s12885-015-1076-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Lin EY, Gouon-Evans V, Nguyen AV, Pollard JW. The macrophage growth factor CSF-1 in mammary gland development and tumor progression. J Mammary Gland Biol Neoplasia. 2002;7(2):147–162. doi: 10.1023/A:1020399802795. [DOI] [PubMed] [Google Scholar]
- 63.Menetrier-Caux C, Montmain G, Dieu MC, Bain C, Favrot MC, Caux C, Blay JY. Inhibition of the differentiation of dendritic cells from CD34(+) progenitors by tumor cells: role of interleukin-6 and macrophage colony-stimulating factor. Blood. 1998;92(12):4778–4791. [PubMed] [Google Scholar]
- 64.Lo AS, Gorak-Stolinska P, Bachy V, Ibrahim MA, Kemeny DM, Maher J. Modulation of dendritic cell differentiation by colony-stimulating factor-1: role of phosphatidylinositol 3′-kinase and delayed caspase activation. J Leukoc Biol. 2007;82(6):1446–1454. doi: 10.1189/jlb.0307142. [DOI] [PubMed] [Google Scholar]
- 65.Demoulin SA, Somja J, Duray A, Guenin S, Roncarati P, Delvenne PO, Herfs MF, Hubert PM. Cervical (pre)neoplastic microenvironment promotes the emergence of tolerogenic dendritic cells via RANKL secretion. Oncoimmunology. 2015;4(6):e1008334. doi: 10.1080/2162402X.2015.1008334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Perrot I, Blanchard D, Freymond N, Isaac S, Guibert B, Pacheco Y, Lebecque S. Dendritic cells infiltrating human non-small cell lung cancer are blocked at immature stage. J Immunol. 2007;178(5):2763–2769. doi: 10.4049/jimmunol.178.5.2763. [DOI] [PubMed] [Google Scholar]
- 67.Stoitzner P, Green LK, Jung JY, Price KM, Atarea H, Kivell B, Ronchese F. Inefficient presentation of tumor-derived antigen by tumor-infiltrating dendritic cells. Cancer Immunol Immunother. 2008;57(11):1665–1673. doi: 10.1007/s00262-008-0487-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zou W, Machelon V, Coulomb-L’Hermin A, Borvak J, Nome F, Isaeva T, Wei S, Krzysiek R, Durand-Gasselin I, Gordon A, Pustilnik T, Curiel DT, Galanaud P, Capron F, Emilie D, Curiel TJ. Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacytoid precursor dendritic cells. Nat Med. 2001;7(12):1339–1346. doi: 10.1038/nm1201-1339. [DOI] [PubMed] [Google Scholar]
- 69.Teicher BA, Fricker SP. CXCL12 (SDF-1)/CXCR4 pathway in cancer. Clin Cancer Res. 2010;16(11):2927–2931. doi: 10.1158/1078-0432.CCR-09-2329. [DOI] [PubMed] [Google Scholar]
- 70.Hargadon KM, Bishop JD, Brandt JP, Hand ZC, Ararso YT, Forrest OA. Melanoma-derived factors alter the maturation and activation of differentiated tissue-resident dendritic cells. Immunol Cell Biol. 2016;94(1):24–38. doi: 10.1038/icb.2015.58. [DOI] [PubMed] [Google Scholar]
- 71.Aalamian M, Tourkova IL, Chatta GS, Lilja H, Huland E, Huland H, Shurin GV, Shurin MR. Inhibition of dendropoiesis by tumor derived and purified prostate specific antigen. J Urol. 2003;170(5):2026–2030. doi: 10.1097/01.ju.0000091264.46134.b7. [DOI] [PubMed] [Google Scholar]
- 72.Aalamian-Matheis M, Chatta GS, Shurin MR, Huland E, Huland H, Shurin GV. Inhibition of dendritic cell generation and function by serum from prostate cancer patients: correlation with serum-free PSA. Adv Exp Med Biol. 2007;601:173–182. doi: 10.1007/978-0-387-72005-0_18. [DOI] [PubMed] [Google Scholar]
- 73.Pillai K, Pourgholami MH, Chua TC, Morris DL. MUC1 as a potential target in anticancer therapies. Am J Clin Oncol. 2015;38(1):108–118. doi: 10.1097/COC.0b013e31828f5a07. [DOI] [PubMed] [Google Scholar]
- 74.Carlos CA, Dong HF, Howard OM, Oppenheim JJ, Hanisch FG, Finn OJ. Human tumor antigen MUC1 is chemotactic for immature dendritic cells and elicits maturation but does not promote Th1 type immunity. J Immunol. 2005;175(3):1628–1635. doi: 10.4049/jimmunol.175.3.1628. [DOI] [PubMed] [Google Scholar]
- 75.Rughetti A, Pellicciotta I, Biffoni M, Backstrom M, Link T, Bennet EP, Clausen H, Noll T, Hansson GC, Burchell JM, Frati L, Taylor-Papadimitriou J, Nuti M. Recombinant tumor-associated MUC1 glycoprotein impairs the differentiation and function of dendritic cells. J Immunol. 2005;174(12):7764–7772. doi: 10.4049/jimmunol.174.12.7764. [DOI] [PubMed] [Google Scholar]
- 76.Ueno A, Cho S, Cheng L, Wang J, Hou S, Nakano H, Santamaria P, Yang Y. Transient upregulation of indoleamine 2,3-dioxygenase in dendritic cells by human chorionic gonadotropin downregulates autoimmune diabetes. Diabetes. 2007;56(6):1686–1693. doi: 10.2337/db06-1727. [DOI] [PubMed] [Google Scholar]
- 77.Hwu P, Du MX, Lapointe R, Do M, Taylor MW, Young HA. Indoleamine 2,3-dioxygenase production by human dendritic cells results in the inhibition of T cell proliferation. J Immunol. 2000;164(7):3596–3599. doi: 10.4049/jimmunol.164.7.3596. [DOI] [PubMed] [Google Scholar]
- 78.Palmano K, Rowan A, Guillermo R, Guan J, McJarrow P. The role of gangliosides in neurodevelopment. Nutrients. 2015;7(5):3891–3913. doi: 10.3390/nu7053891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Shurin GV, Shurin MR, Bykovskaia S, Shogan J, Lotze MT, Barksdale EM., Jr Neuroblastoma-derived gangliosides inhibit dendritic cell generation and function. Cancer Res. 2001;61(1):363–369. [PubMed] [Google Scholar]
- 80.Peguet-Navarro J, Sportouch M, Popa I, Berthier O, Schmitt D, Portoukalian J. Gangliosides from human melanoma tumors impair dendritic cell differentiation from monocytes and induce their apoptosis. J Immunol. 2003;170(7):3488–3494. doi: 10.4049/jimmunol.170.7.3488. [DOI] [PubMed] [Google Scholar]
- 81.Bennaceur K, Popa I, Chapman JA, Migdal C, Peguet-Navarro J, Touraine JL, Portoukalian J. Different mechanisms are involved in apoptosis induced by melanoma gangliosides on human monocyte-derived dendritic cells. Glycobiology. 2009;19(6):576–582. doi: 10.1093/glycob/cwp015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Pugh S, Thomas GA. Patients with adenomatous polyps and carcinomas have increased colonic mucosal prostaglandin E2. Gut. 1994;35(5):675–678. doi: 10.1136/gut.35.5.675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Sombroek CC, Stam AG, Masterson AJ, Lougheed SM, Schakel MJ, Meijer CJ, Pinedo HM, van den Eertwegh AJ, Scheper RJ, de Gruijl TD. Prostanoids play a major role in the primary tumor-induced inhibition of dendritic cell differentiation. J Immunol. 2002;168(9):4333–4343. doi: 10.4049/jimmunol.168.9.4333. [DOI] [PubMed] [Google Scholar]
- 84.Trabanelli S, Lecciso M, Salvestrini V, Cavo M, Ocadlikova D, Lemoli RM, Curti A. PGE2-induced IDO1 inhibits the capacity of fully mature DCs to elicit an in vitro antileukemic immune response. J Immunol Res. 2015;2015:253191. doi: 10.1155/2015/253191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Schipper RG, Romijn JC, Cuijpers VM, Verhofstad AA. Polyamines and prostatic cancer. Biochem Soc Trans. 2003;31(2):375–380. doi: 10.1042/bst0310375. [DOI] [PubMed] [Google Scholar]
- 86.Erbas H, Bal O, Cakir E. Effect of rosuvastatin on arginase enzyme activity and polyamine production in experimental breast cancer. Balkan Med J. 2015;32(1):89–95. doi: 10.5152/balkanmedj.2015.15611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Della Bella S, Gennaro M, Vaccari M, Ferraris C, Nicola S, Riva A, Clerici M, Greco M, Villa ML. Altered maturation of peripheral blood dendritic cells in patients with breast cancer. Br J Cancer. 2003;89(8):1463–1472. doi: 10.1038/sj.bjc.6601243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Doherty JR, Cleveland JL. Targeting lactate metabolism for cancer therapeutics. J Clin Invest. 2013;123(9):3685–3692. doi: 10.1172/JCI69741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Gottfried E, Kunz-Schughart LA, Ebner S, Mueller-Klieser W, Hoves S, Andreesen R, Mackensen A, Kreutz M. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood. 2006;107(5):2013–2021. doi: 10.1182/blood-2005-05-1795. [DOI] [PubMed] [Google Scholar]
- 90.Nasi A, Fekete T, Krishnamurthy A, Snowden S, Rajnavolgyi E, Catrina AI, Wheelock CE, Vivar N, Rethi B. Dendritic cell reprogramming by endogenously produced lactic acid. J Immunol. 2013;191(6):3090–3099. doi: 10.4049/jimmunol.1300772. [DOI] [PubMed] [Google Scholar]
- 91.Vaupel P, Mayer A. Hypoxia-driven adenosine accumulation: a crucial microenvironmental factor promoting tumor progression. Adv Exp Med Biol. 2016;876:177–183. doi: 10.1007/978-1-4939-3023-4_22. [DOI] [PubMed] [Google Scholar]
- 92.Liang D, Zuo A, Shao H, Chen M, Kaplan HJ, Sun D. A2B adenosine receptor activation switches differentiation of bone marrow cells to a CD11c(+)Gr-1(+) dendritic cell subset that promotes the Th17 response. Immun Inflamm Dis. 2015;3(4):360–373. doi: 10.1002/iid3.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Novitskiy SV, Ryzhov S, Zaynagetdinov R, Goldstein AE, Huang Y, Tikhomirov OY, Blackburn MR, Biaggioni I, Carbone DP, Feoktistov I, Dikov MM. Adenosine receptors in regulation of dendritic cell differentiation and function. Blood. 2008;112(5):1822–1831. doi: 10.1182/blood-2008-02-136325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ring S, Pushkarevskaya A, Schild H, Probst HC, Jendrossek V, Wirsdorfer F, Ledent C, Robson SC, Enk AH, Mahnke K. Regulatory T cell-derived adenosine induces dendritic cell migration through the Epac-Rap1 pathway. J Immunol. 2015;194(8):3735–3744. doi: 10.4049/jimmunol.1401434. [DOI] [PubMed] [Google Scholar]
- 95.Maroof A, English NR, Bedford PA, Gabrilovich DI, Knight SC. Developing dendritic cells become ‘lacy’ cells packed with fat and glycogen. Immunology. 2005;115(4):473–483. doi: 10.1111/j.1365-2567.2005.02181.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Dong H, Bullock TN. Metabolic influences that regulate dendritic cell function in tumors. Front Immunol. 2014;5:24. doi: 10.3389/fimmu.2014.00024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Herber DL, Cao W, Nefedova Y, Novitskiy SV, Nagaraj S, Tyurin VA, Corzo A, Cho HI, Celis E, Lennox B, Knight SC, Padhya T, McCaffrey TV, McCaffrey JC, Antonia S, Fishman M, Ferris RL, Kagan VE, Gabrilovich DI. Lipid accumulation and dendritic cell dysfunction in cancer. Nat Med. 2010;16(8):880–886. doi: 10.1038/nm.2172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Tyurin VA, Cao W, Tyurina YY, Gabrilovich DI, Kagan VE. Mass-spectrometric characterization of peroxidized and hydrolyzed lipids in plasma and dendritic cells of tumor-bearing animals. Biochem Biophys Res Com. 2011;413(1):149–153. doi: 10.1016/j.bbrc.2011.08.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Gardner JK, Mamotte CD, Patel P, Yeoh TL, Jackaman C, Nelson DJ. Mesothelioma tumor cells modulate dendritic cell lipid content, phenotype and function. PLoS ONE. 2015;10(4):e0123563. doi: 10.1371/journal.pone.0123563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Gao F, Liu C, Guo J, Sun W, Xian L, Bai D, Liu H, Cheng Y, Li B, Cui J, Zhang C, Cai J. Radiation-driven lipid accumulation and dendritic cell dysfunction in cancer. Sci Rep. 2015;5:9613. doi: 10.1038/srep09613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Cubillos-Ruiz JR, Silberman PC, Rutkowski MR, Chopra S, Perales-Puchalt A, Song M, Zhang S, Bettigole SE, Gupta D, Holcomb K, Ellenson LH, Caputo T, Lee AH, Conejo-Garcia JR, Glimcher LH. ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell. 2015;161(7):1527–1538. doi: 10.1016/j.cell.2015.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Wu R, Zhang QH, Lu YJ, Ren K, Yi GH. Involvement of the IRE1alpha-XBP1 pathway and XBP1s-dependent transcriptional reprogramming in metabolic diseases. DNA Cell Biol. 2015;34(1):6–18. doi: 10.1089/dna.2014.2552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Scanlon CS, Banerjee R, Inglehart RC, Liu M, Russo N, Hariharan A, van Tubergen EA, Corson SL, Asangani IA, Mistretta CM, Chinnaiyan AM, D’Silva NJ. Galanin modulates the neural niche to favour perineural invasion in head and neck cancer. Nat Commun. 2015;6:6885. doi: 10.1038/ncomms7885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Covenas R, Munoz M. Cancer progression and substance P. Histol Histopathol. 2014;29(7):881–890. doi: 10.14670/HH-29.881. [DOI] [PubMed] [Google Scholar]
- 105.Voedisch S, Rochlitzer S, Veres TZ, Spies E, Braun A. Neuropeptides control the dynamic behavior of airway mucosal dendritic cells. PLoS ONE. 2012;7(9):e45951. doi: 10.1371/journal.pone.0045951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Toda M, Suzuki T, Hosono K, Hayashi I, Hashiba S, Onuma Y, Amano H, Kurihara Y, Kurihara H, Okamoto H, Hoka S, Majima M. Neuronal system-dependent facilitation of tumor angiogenesis and tumor growth by calcitonin gene-related peptide. Proc Natl Acad Sci USA. 2008;105(36):13550–13555. doi: 10.1073/pnas.0800767105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Gilaberte Y, Roca MJ, Garcia-Prats MD, Coscojuela C, Arbues MD, Vera-Alvarez JJ. Neuropeptide Y expression in cutaneous melanoma. J Am Acad Dermatol. 2012;66(6):e201–e208. doi: 10.1016/j.jaad.2011.02.015. [DOI] [PubMed] [Google Scholar]
- 108.Buttari B, Profumo E, Domenici G, Tagliani A, Ippoliti F, Bonini S, Businaro R, Elenkov I, Rigano R. Neuropeptide Y induces potent migration of human immature dendritic cells and promotes a Th2 polarization. FASEB J. 2014;28(7):3038–3049. doi: 10.1096/fj.13-243485. [DOI] [PubMed] [Google Scholar]
- 109.Makarenkova VP, Shurin GV, Tourkova IL, Balkir L, Pirtskhalaishvili G, Perez L, Gerein V, Siegfried JM, Shurin MR. Lung cancer-derived bombesin-like peptides down-regulate the generation and function of human dendritic cells. J Neuroimmunol. 2003;145(1–2):55–67. doi: 10.1016/j.jneuroim.2003.09.009. [DOI] [PubMed] [Google Scholar]
- 110.DeRosa DC, Ryan PJ, Okragly A, Witcher DR, Benschop RJ. Tumor-derived death receptor 6 modulates dendritic cell development. Cancer Immunol Immunother. 2008;57(6):777–787. doi: 10.1007/s00262-007-0413-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.D’Souza-Schorey C, Clancy JW. Tumor-derived microvesicles: shedding light on novel microenvironment modulators and prospective cancer biomarkers. Genes Dev. 2012;26(12):1287–1299. doi: 10.1101/gad.192351.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Valenti R, Huber V, Filipazzi P, Pilla L, Sovena G, Villa A, Corbelli A, Fais S, Parmiani G, Rivoltini L. Human tumor-released microvesicles promote the differentiation of myeloid cells with transforming growth factor-beta-mediated suppressive activity on T lymphocytes. Cancer Res. 2006;66(18):9290–9298. doi: 10.1158/0008-5472.CAN-06-1819. [DOI] [PubMed] [Google Scholar]
- 113.Huang SH, Li Y, Zhang J, Rong J, Ye S. Epidermal growth factor receptor-containing exosomes induce tumor-specific regulatory T cells. Cancer Invest. 2013;31(5):330–335. doi: 10.3109/07357907.2013.789905. [DOI] [PubMed] [Google Scholar]
- 114.Ding G, Zhou L, Qian Y, Fu M, Chen J, Chen J, Xiang J, Wu Z, Jiang G, Cao L. Pancreatic cancer-derived exosomes transfer miRNAs to dendritic cells and inhibit RFXAP expression via miR-212-3p. Oncotarget. 2015;6(30):29877–29888. doi: 10.18632/oncotarget.4924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Feijoo E, Alfaro C, Mazzolini G, Serra P, Penuelas I, Arina A, Huarte E, Tirapu I, Palencia B, Murillo O, Ruiz J, Sangro B, Richter JA, Prieto J, Melero I. Dendritic cells delivered inside human carcinomas are sequestered by interleukin-8. Int J Cancer. 2005;116(2):275–281. doi: 10.1002/ijc.21046. [DOI] [PubMed] [Google Scholar]
- 116.Yang L, Yamagata N, Yadav R, Brandon S, Courtney RL, Morrow JD, Shyr Y, Boothby M, Joyce S, Carbone DP, Breyer RM. Cancer-associated immunodeficiency and dendritic cell abnormalities mediated by the prostaglandin EP2 receptor. J Clin Invest. 2003;111(5):727–735. doi: 10.1172/JCI16492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Ristich V, Liang S, Zhang W, Wu J, Horuzsko A. Tolerization of dendritic cells by HLA-G. Eur J Immunol. 2005;35(4):1133–1142. doi: 10.1002/eji.200425741. [DOI] [PubMed] [Google Scholar]