Skip to main content
Cancer Immunology, Immunotherapy : CII logoLink to Cancer Immunology, Immunotherapy : CII
. 2018 Jul 12;68(4):645–660. doi: 10.1007/s00262-018-2200-6

Interactions among myeloid regulatory cells in cancer

Viktor Umansky 1,2,, Gosse J Adema 3, Jaroslaw Baran 4, Sven Brandau 5, Jo A Van Ginderachter 6,7, Xiaoying Hu 1,2, Jadwiga Jablonska 5, Slavko Mojsilovic 8, Helen A Papadaki 9, Yago Pico de Coaña 10, Kim C M Santegoets 3, Juan F Santibanez 11,12, Karine Serre 13, Yu Si 5, Isabela Sieminska 4, Maria Velegraki 9, Zvi G Fridlender 14
PMCID: PMC11028297  PMID: 30003321

Abstract

Mounting evidence has accumulated on the critical role of the different myeloid cells in the regulation of the cancerous process, and in particular in the modulation of the immune reaction to cancer. Myeloid cells are a major component of host cells infiltrating tumors, interacting with each other, with tumor cells and other stromal cells, and demonstrating a prominent plasticity. We describe here various myeloid regulatory cells (MRCs) in mice and human as well as their relevant therapeutic targets. We first address the role of the monocytes and macrophages that can contribute to angiogenesis, immunosuppression and metastatic dissemination. Next, we discuss the differential role of neutrophil subsets in tumor development, enhancing the dual and sometimes contradicting role of these cells. A heterogeneous population of immature myeloid cells, MDSCs, was shown to be generated and accumulated during tumor progression as well as to be an important player in cancer-related immune suppression. Lastly, we discuss the role of myeloid DCs, which can either contribute to effective anti-tumor responses or play a more regulatory role. We believe that MRCs play a critical role in cancer-related immune regulation and suggest that future anti-cancer therapies will focus on these abundant cells.

Keywords: Myeloid regulatory cells, Mye-EUNITER, Macrophages, Neutrophils, Myeloid-derived suppressor cells, Dendritic cells

Introduction

Various investigations have presented strong data that different tumors were able to display immunogenic properties, stimulating robust anti-tumor immune responses, in which T cells were demonstrated to play a major role [1]. An increased infiltration of tumor lesions with T lymphocytes has been demonstrated in patients with a large spectrum of tumors to correlate markedly with better clinical outcome [2]. However, multiple studies in the last decade have indicated a critical role of myeloid cell subsets in the development of an immunosuppressive tumor microenvironment, resulting in cancer progression. On the other side, some myeloid cell subsets could be indispensable for the induction of efficient anti-tumor immune responses. Myeloid cells represent a major component of host cells infiltrating tumors and are characterized by an extraordinary plasticity [3]. They may not only interact with each other, with other host cells (especially T lymphocytes) and tumor cells but they can also undergo a conversion from one subset to another. Importantly, the recruitment and enrichment of myeloid cells with immunosuppressive/regulatory properties designated in this series of reviews as myeloid regulatory cells (MRCs), at the tumor site was found to be stimulated by chronic inflammatory conditions developing in the tumor microenvironment [4].

The phenotypic characterization of circulating and tumor-infiltrating MRCs is difficult due to the fact that MRC subsets could express overlapping set of markers. Last years were characterized by an ongoing discussion regarding the relationship between neutrophils and polymorphonuclear MDSCs (PMN-MDSCs) or tumor-associated macrophages (TAMs) and monocytic MDSCs (M-MDSCs). For example, classical neutrophils and PMN-MDSCs share markers [5]. Despite numerous efforts to determine the similarities and differences between these two cell populations, further studies are still needed to better identify their molecular identity and functional characteristics. A comprehensive analysis of the phenotypic markers, molecular signaling pathways and functional capacities of various MRC subsets is presented by Cassetta et al. and Bruger et al. [6] in companion reviews in this symposium-in-writing. In the current review, we will focus on the characterization of various MRC subpopulations, including monocytes/macrophages, neutrophils, MDSCs and DCs, their interactions and therapeutic targeting both in mouse tumor models and in cancer patients (Figs. 1, 2).

Fig. 1.

Fig. 1

Multiple effects and interactions among the different types of MRCs in cancer. Macrophages, M2 M2 macrophages, TAN tumor-associated neutrophils, N2 N2 TAN, Mo-MDSC monocytic myeloid-derived suppressor cells, PMN-MDSC polymorphonuclear MDSC, DC dendritic cells, Treg regulatory T cells

Fig. 2.

Fig. 2

Effects of tumor cells on MRC subsets and that of MRCs on T cell subpopulations in cancer. M2 M2 macrophages, N2 N2 tumor-associated neutrophils, Mo-MDSC monocytic myeloid-derived suppressor cells, PMN-MDSC polymorphonuclear MDSC, DC dendritic cells, Treg regulatory T cells

Monocytes and macrophages

Macrophages reside in every tissue and perform, under homeostatic conditions, crucial housekeeping functions. Tissue-resident macrophages seed the organs pre-birth from embryonal precursors and are maintained throughout life with a variable contribution of monocytes depending on the organ [7]. Although monocytes were traditionally considered to comprise the main source of tissue macrophages [8] it is now known that many of the tissue-resident macrophages are of embryonic origin, derived either directly from progenitors of the yolk-sac or the fetal liver [7, 9, 10]. Monocytes appear to adopt the phenotype of tissue-resident macrophages if the correct niche is available [11]. In tumors, macrophages can be abundant and contribute to angiogenesis, immunosuppression and metastasis; therefore, their enrichment is correlated with a worse clinical prognosis [12].

In multiple mouse tumor models, the majority of TAMs were shown to derive from classical Ly6Chi monocytes and is continuously replaced via monocyte recruitment [13]. Although the spleen may be a source of monocyte precursors, the largest fraction of TAM originates from the bone marrow [14]. However, in the spontaneous MMTV-PyMT and MMTV-Neu breast cancer models, the TAM pools are maintained through in situ proliferation of tissue-resident cells [15]. Moreover, tissue-resident microglia can contribute to the environment of brain tumors, and embryonic pancreatic macrophages are crucial tumor-promoting cells in pancreatic ductal adenocarcinoma, besides monocyte-derived cells [1618]. It has been demonstrated that monocytes entering the hypoxic tumor microenvironment turn into potent MDSCs followed by the differentiation into TAMs under the influence of hypoxia [19]. Importantly, hypoxia could determine the protumoral activity of TAMs [20], and preventing TAM from migrating into hypoxic areas resulted in a reduced tumor growth [21]. This monocyte/MDSC-macrophage differentiation in tumors and their acquisition of tumor-promoting functions was reported to be supported by macrophage colony stimulating factor (M-CSF) [22].

Many papers proposed to test the MHC class-II expression to define distinct TAM subsets (MHC-IIlo versus MHC-IIhi) [13, 23]. In this respect, MHC-IIlo TAM resemble M2 macrophages and are superior in suppressing T-cell activity and angiogenic activity, whereas MHC-IIhi TAM are more M1-like and more permissive to anti-tumor immunity [13, 18]. On the other side, monocytes were shown to differentiate in tumors into MHC-IIhiCD11chi (Tip)-DC that produced TNF and NO, captured antigens but suppress T cell functions [24]. Furthermore, TAM may hamper the stimulatory capacity of conventional DC and T cells through IL-10 production [23, 25].

Besides influencing primary tumor growth, macrophages are known to influence the dissemination of cancer cells to distant organs. Perivascular Tie2hi macrophages cause transient vascular permeability and cancer cell intravasation through the VEGF secretion [26]. Macrophages were shown in preclinical tumor models to contribute to metastasis establishment. For example, lung metastasis of breast cancer cells critically depends on the recruitment of Ly6Chi monocytes via the chemokine (C–C motif) ligand (CCL) 2; upon CCL2 triggering, monocytes produced CCL3, entrapping them at the metastatic site through interaction with C–C chemokine receptor (CCR) 1 [27]. Once retained in the lung, these metastasis-associated macrophages self-maintain via the autocrine production of M-CSF under the influence of Flt1 (VEGFR1) signaling [28]. Importantly, macrophages contribute to the preparation of the metastatic niche even before cancer cells reach the target organ. Thus, macrophage-secreted granulin was reported to induce liver fibrosis, supporting pancreatic cancer metastasis to this organ [29]. In addition, liver-tropic metastatic cells could secrete exosomes that induce the acquisition of pro-metastatic properties by liver macrophages [30].

Numerous clinical studies indicated that the expansion of circulating monocytes and increased macrophage numbers in the tumor microenvironment was adversely correlated with the clinical outcomes in many human cancers [3135]. It was shown that the survival of cancer stem cells could be supported by TAMs [36]. Furthermore, TAMs were shown to be involved in the reduced responsiveness of different cancers to standard chemotherapy [37, 38]. Moreover, different soluble factors secreted by TAMs in the tumor microenvironment such as epidermal growth factor (EGF), fibroblast growth factor (FGF), and transforming growth factor (TGF)-β, could affect the proliferation of cancer cells [39]. TAMs were also described to promote tumor growth by supporting neoangiogenesis due to their ability to secrete a number of angiogenic factors such as VEGF, platelet-derived growth factor (PDGF), placenta growth factor (PLGF), C–X–C motif chemokine ligand (CXCL) 8, FGF, and matrix metalloproteinases (MMPs) [40]. The subpopulation of TAMs contributing to angiogenesis was reported to be derived from Tie-2 expressing monocytes that could exert tumor-supporting effects in various human cancers [41, 42].

On the other side, monocytes and TAMs could promote metastasis independently of the induction of neovascularization. It has been shown that TAMs enhance the motility of breast cancer [43] and hepatocellular carcinoma cells [44]. The release of EGF as well as MMPs, which degrade extracellular matrix and augment the invasiveness of tumor cells, were reported among the underlying mechanisms of the pro-metastatic effect of TAMs [27, 45]. Moreover, TAMs have been reported to be involved in the formation of the pre-metastatic niches at the site of metastases [46].

Immunosuppressive properties of TAMs and their contribution to the immune escape of malignant cells are well documented. These cells have been reported to inhibit T cell activity both directly via PD-1/PD-L1 signaling [47] and indirectly by the upregulation of Treg functions [48]. Furthermore, TAMs infiltrating Hodgkin’s lymphoma promoted the induction of Th17 responses or immunosuppression via the expression of TGF-β and IL-10 [49]. In addition, in EBV-associated nasopharyngeal carcinoma, TAMs were described to express the enzyme IDO, resulting in T cell inhibition via tryptophan depletion [50]. In addition, TAMs could attract other myeloid cell populations with immunosuppressive properties such as MDSCs that led to further inhibition of anti-tumor immune responses [51, 52].

Since TAMs were considered to support tumor progression, strategies aimed to target macrophage recruitment and monocyte–macrophage differentiation as well as to reprogram TAM into anti-tumoral M1-like cells were developed to normalize the tumor vasculature, to increase an anti-tumor reactivity of T cells and to exert direct anti-tumor cytotoxic effects [53] (Table 1).

Table 1.

Targeting of monocytes/macrophages

Target Drug Mechanism of action Therapeutic indication
Anti-CCL2 antibody Carlumab Inhibition of recruitment and differentiation of macrophages

Metastatic castration-resistant prostate cancer (CNTO 888)

Advanced solid tumor (NCT01204996)

CCR2 antagonist PF-04136309 Inhibition of recruitment of macrophages Pancreatic ductal adenocarcinoma (NCT01413022)
CCR5 antagonist Maraviroc Inhibition of recruitment of macrophages Advanced stage CRC
Anti-CSF-1R antibody Emactuzumab (RG7155) Inhibition of differentiation and recruitment macrophages Diffuse-type tenosynovial giant-cell tumor
CSF-1R inhibitor BLZ945 Inhibition of macrophage differentiation and recruitment Glioblastoma multiforme
Anti-CSF-1R antibody AFS98 Inhibition of macrophage differentiation and recruitment Breast carcinoma
CSF-1R inhibitor GW2580 Inhibition of macrophage differentiation and recruitment Ovarian cancer
Macrophages Clodronate in liposomes Induction of apoptosis in macrophages Breast and prostate cancer with bone metastasis
Anti-CD47 antibody Hu5F9-G4 Induction of apoptosis in macrophages Solid tumors
Macrophages Trabectedin Inhibition of survival and killing of macrophages Ovarian cancer
TAM IFN-γ Reprogramming of TAM towards an anti-tumor phenotype Ovarian cancer
Anti-CD40 agonist antibody CP-870,893 Reprogramming of TAM towards an anti-tumor phenotype Advanced stage pancreatic cancer

Neutrophils

Neutrophils, the most abundant myeloid blood cells, play a crucial role in inflammation-driven tumorigenesis. Only recently, it has been recognized that neutrophils are not a homogeneous cell population and that their differentiation and phenotype could be modulated by the tumor milieu, resulting in diverse phenotypic and functional states [5456]. Neutrophils in tumor-bearing hosts have been shown to exert contradictory functional activities, leading to potentiation or inhibition of cancer progression. Anti-tumor N1 neutrophils have a hypersegmented nucleus typical for mature neutrophils, while pro-tumor N2 population consist of cells with immature-banded nuclei [57]. It is not clear yet if diverse tumor-associated neutrophils (TANs) are different neutrophil subpopulations or rather flexible and dynamic cells that change their activity due to environmental cues. Furthermore, the differentiation between N2 TANs and PMN-MDSCs in the tumor is difficult due to overlapping phenotypic and functional properties of these closely related granulocytic subpopulations.

In a tumor situation, the number of circulating neutrophils continuously increases with tumor progression, possibly due to elevated emergency myelopoiesis [58]. It has been recently proposed that upon cancer pressure, extramedullary granulopoiesis occurred in the spleen that became a site of production and a reservoir of TANs [59]. Neutrophil migration into tissues and tumors is induced by specific chemokines (e.g., CXCL1, CXCL2 or CXCL4), cytokines (e.g., TNF-α and IFN-γ) and cell adhesion molecules [e.g., lymphocyte function associated antigen (LFA)-1 and macrophage (Mac)-1 antigen] [6062]. The primary tumor milieu influences migratory capacities of neutrophils that accumulate in certain organs and form a pre-metastatic niche [6365]. Neutrophils can accumulate in large numbers in pre-metastatic organs and release factors attracting tumor cells and facilitating their proliferation such as BV8, S100A8 and S100A9 [64]. Moreover, neutrophils support the seeding of tumor cells in secondary target organs by the release of neutrophil extracellular traps [66] or by the establishment of an immunosuppressive environment in target organs [67]. Conversely, neutrophils may acquire a cytotoxic phenotype-limiting metastatic seeding [64, 68].

Neutrophils represent a significant portion of tumor-infiltrating myeloid cells. TANs differ significantly from their blood counterparts as their activation and tissue association seem to be strongly dependent on the tumor milieu. TANs were reported to exert pro- or anti-tumor effects, depending on cytokines available in the tumor microenvironment (e.g., TGF-β or IFNs) [64, 65, 69, 70]. TANs were shown to produce pro-angiogenic and proteolytic factors that support tumor angiogenesis such as VEGF or MMP9 [61]. They were also implicated in promoting tumor growth via matrix degradation, and by the stimulation of tumor cell proliferation, survival and metastasis [64, 65]. Furthermore, neutrophils were shown to recruit other cells to the tumor, suppressing adaptive immune responses [71, 72].

On the other hand, neutrophils were described to have anti-tumor properties, including the capacity to kill tumor cells via direct or antibody-dependent cell cytotoxicity [73, 74] or by boosting T cell-mediated anti-tumor responses [57, 75]. Furthermore, neutrophils could enhance the efficiency of some immunotherapies. They favored photodynamic therapy-induced CD8 T cell activation [76], promoted T cell recruitment and improved the efficacy of BCG immunotherapy of bladder cancer [77], and exerted cytotoxicity under immunotherapy with tumor-specific monoclonal antibodies in melanoma and breast cancer [78].

Data regarding neutrophils in cancer patients are very scarce, and their nature and function in the tumor microenvironment are largely unknown. The lack of sizeable biopsy samples together with the poor characterization of appropriate functional and phenotypic markers for the myeloid subsets in patients are major issues to be resolved. Gene signatures from over 10,000 cancer biopsies demonstrated the presence of neutrophils in over 30 solid malignancies [79]. Strikingly, neutrophil signatures emerged as the most significant adverse cancer-wide prognostic population [79]. This is consistent with the growing number of studies that link high levels of intra-tumoral neutrophils to poor clinical outcome [80].

The main mechanisms of pro-tumoral functions of human circulating neutrophils are associated with their immunosuppressive and angiogenic properties [81]. The immunosuppressive mechanisms include production of arginase-1 (Arg-1), reactive oxygen and nitrogen species to impair T cell activation and proliferation [82, 83]. Neutrophils have also been shown to be important sources of VEGF and MMP9 [84].

As in mouse models, the protective effect of neutrophils was shown in patients undergoing therapy. For example, higher TAN density in colorectal cancer was associated with better response to 5-FU-based chemotherapy [85]. Moreover, the administration of IFN-α in chronic myeloid leukemia and melanoma patients stimulated the release of TRAIL by neutrophils, inducing apoptosis of TRAIL-sensitive cancer cells [86].

Circulating blood neutrophils may be separated using a density gradient into normal neutrophils (called high density, HDNs or normal density, NDNs) and low-density LDNs, co-separated in the mononuclear fraction. Of note, there is no clear immunotype that fits LDNs or HDNs since they often share the same surface markers and functions. Nevertheless, LDNs are enriched for immature cells and activated neutrophils compared with HDN [87, 88]. Neutrophils were found to be accumulated in the peripheral blood of patients with various types of cancer, especially in advanced stages [89, 90]. Furthermore, neutrophilia was reported to be associated with poor prognosis in many tumors such as bronchoalveolar carcinoma [91] and metastatic melanoma [89]. A high neutrophil to lymphocyte ratio (NLR) caused by tumor-induced neutrophilia and lymphocyte apoptosis is a robust marker of poor clinical outcome in cancer patients. The NLR has been validated as an independent prognostic factor in a variety of tumor types [92].

There is still little information on tumor-infiltrating neutrophils and their clinical relevance is only beginning to emerge. Nevertheless, TANs were demonstrated to be associated with poor clinical outcome in patients with renal cancer [93], non-small-cell lung carcinoma [94] and melanoma [95]. It has been reported that the enrichment of TANs was associated with metastases in various tumor entities and they were suggested to be involved in metastatic process [96]. Neutrophils infiltrating bronchoalveolar and cholangiocellular carcinoma have been shown to produce hepatocyte growth factor (HGF), enhancing the invasive capacity of cancer cells [97]. The recruitment of neutrophils within the tumor microenvironment relies on several chemokines, including but not limited to, CXCL8/IL-8, CXCL5 and macrophage migration inhibitory factor [5456, 88]. Therefore, most of therapeutic strategies to target neutrophils in tumor-bearing hosts are dealing with the blocking of their migration into the tumor site (Table 2).

Table 2.

Targeting of neutrophils

Target Drug Mechanism of action Therapeutic indication
CXCR1 and CXCR2 inhibitor Reparixin Inhibition of neutrophil migration into the tumor

Triple-negative breast cancer (NCT02370238)

HER2-negative metastatic breast cancer (NCT02001974)

G-CSF Anti-G-CSF antibodies Inhibition of neutrophil mobilization and angiogenesis Pancreatic adenocarcinoma
TGF-β receptor inhibitor Galunisertib (LY2157299 monohydrate) N2–N1 shift Glioma

Myeloid-derived suppressor cells

One of the important consequences of chronic inflammatory conditions typical for the tumor microenvironment is the generation and accumulation of immunosuppressive myeloid cells designated as MDSCs [98103]. These cells were shown to exert a strong capacity to inhibit anti-tumor functions of T and NK cells [99, 102107]. Numerous studies indicated that a variety of inflammatory factors produced by tumor and host cells, including IFN-γ, IL-1β, prostaglandin E2, IL-6, IL-10, IL-13, COX-2, TGF-β, complement component C5a, VEGF, G-CSF, M-CSF, GM-CSF, MMP-9, CCL2, CCL3, CCL4, CCL5, S100A8, S100A9, etc. were involved in MDSC enrichment and activation [99102, 108]. Most of these factors use STAT3 and janus kinase signaling pathways that trigger signals for cell survival, proliferation, differentiation and apoptosis [99, 103, 108].

MDSCs represent a heterogeneous population of myeloid cells with a strong immunosuppressive capacity [99, 102, 105, 106]. In humans, PMN-MDSCs can be recognized as CD11b+ or CD33+, CD15+ or CD66b+, and CD14 while M-MDSCs are CD11b+ or CD33+, CD14+, and HLA-DRlow cells; Lin (including CD3, CD14, CD15, CD19, and CD56) HLA-DRCD33+ cells are considered as early stage (e)MDSC [103]. Mouse MDSCs are characterized by Gr1 and CD11b expression and contain three subsets: PMN-MDSC characterized as CD11b+Ly6G+Ly6Clow, M-MDSCs as CD11b+Ly6GLy6Chigh, as well as non-PMN-MDSCs and non-M-MDSCs defined as CD11b+Ly6GmedLy6Cmed cells [103].

MDSC subpopulations effectively inhibit T lymphocyte activity through various mechanisms, including the upregulation of Arg-1 expression that is responsible for arginine depletion; stimulation of inducible NO synthase (iNOS), leading to the production of NO; secretion of reactive nitrogen and oxygen species; activation of IDO, resulting in tryptophan depletion; upregulation of PD-L1 expression. Furthermore, MDSCs produce high levels of immunosuppressive cytokines such as IL-10 and TGF-β [99, 102, 103, 108, 109]. Interestingly, M-MDSCs mediate immunosuppression mainly via NO, whereas PMN-MDSCs produce large amounts of ROS and express high levels of Arg-1. However, both subsets could use common immunosuppressive molecules such as PD-L1, IL-10 and TGF-β [87, 102, 103, 108, 109]. In addition, murine non-PMN- and non-M-MDSCs could also possess immunosuppressive functions mainly via IL-10 [110]. A recent study directly compared the immunosuppressive capacity and clinical relevance of the three human circulating MDSC subsets and identified mature PMN-MDSCs as dominant inhibitors of T cell functions mediated by Arg-1 [111].

For a detailed critical review of MDSC-T cell functional interactions, we refer the reader to the companion review by Bruger et al. [6] in this symposium-in-writing series.

In addition to the direct T cell inhibition, MDSCs are able to induce and recruit Treg via TGF-β and IL-10 production and CD40-CD40L signaling [112, 113]. Moreover, MDSCs may induce Th17 cell polarization from naive CD4+ T cells through the production of IL-1β, IL-6, IL-23 and NO [114]. On the other hand, IL-17 produced by Th17 cells was shown to upregulate the expression of Arg-1, IDO and COX-2 in a mouse breast cancer model, boosting thereby the immunosuppressive activity of MDSCs [115].

In tumors, MDSC suppress effector T cells not only directly but also by the generation of M2 TAMs and N2 TANs. As a result of this cross talk, IL-10 produced by MDSC not only inhibits IL-12 and TNF-α but also stimulates IL-10 production in macrophages that in turn enhances the release of IL-10 by MDSCs [51, 52]. Tumor-infiltrating MDSCs could also directly differentiate into potent immunosuppressive TAMs [116]. Furthermore, TGF-β secreted by tumor stroma cells, including MDSCs [99, 102], was reported to convert neutrophils into N2 TANs producing CCL17, a well-known chemoattractant for Treg [57]. Similar to M2 TAMs, MDSCs were shown to inhibit IL-12 and induce IL-10 production by DC [117]. Moreover, MDSC-derived VEGF and IL-10 could downregulate the expression of MHC class II and co-stimulatory molecules on DC via activation of STAT3 [118]. In addition, the antigen uptake by DC was found to be diminished in the presence of MDSCs [119]. This results in the inhibition of the DC capacity to stimulate T cell-mediated anti-tumor immune responses.

Numerous studies reported on the accumulation of highly immunosuppressive MDSCs in patients with various tumors, including hepatocellular carcinoma, melanoma, prostate cancer, bladder cancer, non-small cell lung cancer, head and neck squamous cell carcinoma as well as breast, gastric and colorectal cancer, which indicates the clinical significance of these cells [105, 118, 120125]. Interestingly, HLA-DRCD33+CD11b+CD14+ M-MDSCs were also detected in patients with EBV-associated lymphoid tumor, the extranodal natural killer NK/T cell lymphoma, which can develop after chronic active Epstein–Barr virus (EBV) infection [126]. In another EBV-associated tumor, nasopharyngeal carcinoma, an expansion of CD33+ MDSCs was found to be due to a latent membrane protein-1 in tumor cells that could induce the production of IL-1β, IL-6 and GM-CSF critical for MDSC generation [127]. An increased MDSC frequency in the peripheral blood was found to correlate with tumor progression and worse clinical outcome in patients with different tumors [105, 118, 120, 122, 123, 128, 129]. Moreover, several publications described that the decreased frequency and immunosuppressive function of both M- and PMN-MDSCs correlated with beneficial therapeutic effects in cancer patients treated with the negative immune checkpoint inhibitors [122, 130, 131].

Besides immunosuppressive functions, MDSCs may contribute to the remodeling of the tumor microenvironment by producing VEGF, FGF and MMPs. These factors stimulate tumor neoangiogenesis as well as cancer cell motility and invasion [106, 107]. Interestingly, MDSCs were reported to transdifferentiate towards endothelial cells contributing to tumor angiogenesis [132]. Moreover, TGF-β, HGF, and EGF produced by tumor-infiltrating MDSCs were found to contribute to cancer-associated epithelial to mesenchymal transition [133].

Given a critical role of MDSC in tumor progression, several strategies to neutralize these cells were developed, including (1) prevention of MDSC generation; (2) MDSC depletion or blocking their expansion and activation; (3) inhibition of MDSC recruitment; and (4) blocking MDSC immunosuppressive function [102, 106, 109, 134136] (Table 3).

Table 3.

Targeting of MDSCs

Target Drug Mechanism of action Therapeutic indication
iNOS inhibitors Phosphodiesterase-5 (PDE-5) inhibitors: tadalafil, sildenafil, nitro-aspirin Inhibition of MDSC function

Multiple myeloma (NCT01374217)

Head and neck cancer (NCT00843635)

Non-small cell lung carcinoma (NCT00752115)

Pancreatic cancer (NCT01342224)

Colorectal cancer (NCT00331786)

Arginase inhibitors Celecoxib, N-hydroxy-l-Arginine (NOHA), N(G)-Nitro-l-Arginine, Methyl Ester (l-NAME) Inhibition of MDSC function Colon cancer
ROS inhibitors Bardoxolone methyl (CDDO-Me) Inhibition of MDSC function Pancreatic cancer (RTA 402-C-0702)
Anti-glycan antibodies Receptor for Advanced glycation end products (RAGE) Inhibition of MDSC migration Colon cancer
CSF-1R inhibitor GW2580 Inhibition of MDSC migration Prostate cancer
Retinoid-activated transcriptional regulators All-trans retinoic acid Promotion of MDSC maturation

Renal cell carcinoma

Lung adenocarcinoma

Small cell lung cancer

Triterpenoids RTA 408 Promotion of MDSC maturation Melanoma
Vitamins 25-hydroxy-vitamin D Promotion of MDSC maturation Head and neck cancer
MMP9 Biphosphonates Inhibition of MDSC generation Pancreatic cancer
STAT3

Cucurbitacin B (CuB)

STAT3 DECOY

AZD9150

Sunitinib

Inhibition of MDSC generation

Advanced lung cancer

Head and neck cancer

Advanced hepatocellular carcinoma

Unknown

Cisplatin

5-Fluorouracil

Paclitaxel

MDSC depletion

Bronchoalveolar carcinoma

Lewis lung carcinoma

IL-6R Gemcitabine MDSC depletion Lung cancer
CCR5 Soluble fusion protein mCCR5-Ig MDSC depletion Melanoma
BRAF inhibitors Vemurafenib MDSC depletion Melanoma
Fas ligand IL-2 and anti-CD40 agonistic antibody MDSC depletion Renal adenocarcinoma
CTLA-4 Ipilimumab MDSC depletion Melanoma

Dendritic cells

DCs can be divided into myeloid and plasmacytoid DCs. In mice, myeloid DCs could be further subdivided in classical type I DCs (cDC1) (CD11c+/CD8α+) and type 2 DCs (cDC2) (CD11c+/CD11b+) cells [137]. The cDC1, which in tissues express CD103, can efficiently cross-present antigens to CD8+ T cells, while CD11b+ DCs mainly present antigens on MHC class II to CD4+ T cells [138]. Besides these classical DC subsets, monocytes and M-MDSCs can also contribute to the pool of tumor-infiltrating DCs by differentiating into inflammatory DCs [139]. In mice, these cells can be identified as MHC-II+/CD11b+/CD11c+/F4/80+/Ly6C+ and also express CD64 and FcεRI, which can be used to distinguish inflammatory DCs from classical DCs and macrophages [139]. Studying the composition of myeloid DCs in different tumor models, Laoui et al. [24] demonstrated that CD103+ DCs were generally the smallest subset, whereas CD11b+ cDC2 were always well represented. Interestingly, the number of monocyte-derived/inflammatory DCs varies widely in different tumor models [24].

Depending on the factors in the tumor microenvironment, myeloid DC subsets can either contribute to effective anti-tumor responses or show a more immature and/or regulatory phenotype. The presence of IL-10, IL-6 and VEGF in the tumor microenvironment induces prolonged STAT3 activation in DCs [140]. This limits DC maturation and IL-12 production and induces the production of the immunosuppressive cytokine IL-10. Furthermore, tumor-derived TLR2 ligands were found to stimulate an autocrine secretion of IL-10 and IL-6 by DCs and to enhance expression of the corresponding cytokine receptors, thus boosting STAT3 activation and DC dysfunction [141]. In addition to these cytokines, TGF-β and other immunoregulatory agents such as prostaglandins, lactic acid, adenosine, galectins and mucins were reported to play a role in the induction of immunosuppressive DCs [140, 142].

The accumulation of lipids in DCs could further contribute to their dysfunction in the tumor microenvironment [143, 144]. Lipid-loaded DCs did not differ in the expression of MHC or co-stimulatory molecules but had a reduced capacity to process antigens. Tumor-infiltrating DCs displayed the highest levels of lipids. In the spleen of tumor-bearing mice, increased lipid levels could be detected in both cDC1 and cDC2. The presence of oxidized lipids was also shown to inhibit cross-presentation [144].

Although immunosuppressive DCs displayed no unique markers, they often showed a reduced expression of maturation markers (CD80, CD86) and/or increased expression of inhibitory receptors such as PD-L1 and immunoglobulin-like transcripts (ILTs), limiting T cell activation. It has been reported that monocyte-derived DCs exhibited regulatory properties with increased iNOS expression and IL-10 to IL-12 ratio, resulting in the inhibition of CD4+ and CD8+ T cell proliferation in a lung carcinoma model [24].

Tumor-infiltrating DCs were reported to activate anti-tumor functions of other myeloid cells by producing IFN-β that induced anti-tumor polarization of TANs [145]. This type I IFN response was largely dependent on tumor-infiltrating myeloid DCs in multiple tumor models, and was induced via the STING pathway, through the recognition of tumor DNA [146, 147]. Moreover, IFN-β reduced the accumulation of proangiogenic TANs by influencing the expression of CXCR2 and its ligands CXCL1, CXCL2 and CXCL5 [60, 61]. In addition, Type I IFNs have been shown to induce MDSC maturation and reduce their immunosuppressive activity [148]. Importantly, IFNs were also demonstrated to inhibit the recruitment of TAMs [149].

IL-12 production by mature DCs can also broadly affect MRCs, reversing the suppressive function of MDSCs [150] and TAMs [151]. Furthermore, IL-12 is known as a potent inducer of IFN-γ production by T cells and NK cells. However, tumor-infiltrating DCs often show a regulatory/dysfunctional phenotype with low levels of IL-12 and increased IL-10 and TGFβ production, leading to an enhanced suppressive function of MRCs rather than enforcing their immune stimulatory activity. These immunosuppressive cytokines can support the development of pro-tumoral macrophages and neutrophils [57, 152, 153]. Tumor-infiltrating regulatory DCs also produced significantly higher levels of the chemokines CCL2, CCL4 and CXCL1, attracting monocytes and neutrophils as compared to other DC populations [24]. In this way, regulatory DCs could sustain the immunosuppressive tumor microenvironment created together with the other MRCs. Therefore, it is crucial to interrupt this vicious cycle of immunosuppression for effective anti-tumor immunotherapy.

Intratumoral DCs mainly affect the tumor progression or regression via an activation or inhibition of T cells and NK cells or induction of Treg. It has been recently demonstrated that tumor-infiltrating CD103+ cDC1 could not only induce an antigen cross-presentation to CD8+ T cells, but also support the T cell recruitment into the tumor [154]. However, when intratumoral DCs gain regulatory functions they are not able to efficiently activate T cells but can rather counteract T cell functions, protecting tumor cells from immune-mediated killing [155].

Similar to mouse-circulating myeloid DCs, their human counterparts could be divided into two main subsets: CD141+ (BDCA3+) cDC1 (so-called cross-presenting subset) and CD1c+ (BDCA1+) cDC2 [137, 138]. In addition to these classical DC subsets, monocyte-derived “inflammatory DCs” could also be found under inflammatory conditions in humans. They express HLA-DR, CD11c, BDCA1, CD1a, FcεRI, CD206, CD172a, CD14 and CD11b markers [138]. The presence of these DC subsets has been described in different human tumors, including lung and colorectal cancer [138]. As suggested from mouse studies, the accumulation of cDC1 was found to correlate with better clinical outcome in patients with various tumors [156, 157].

Most factors known to trigger regulatory DCs in mice have also been shown to induce a regulatory phenotype in human DCs in vitro. Indeed, tumor cell-conditioned medium could stimulate STAT3 activation, limit DC maturation, decrease IL-12 to IL-10 ratio and increase lipid levels in human monocyte-derived DCs [140, 143, 158, 159]. Therefore, one of the critical questions to address is how to prevent the detrimental effects of the tumor microenvironment on DC function in cancer patients. To this end, both small-molecule inhibitors and antibodies targeting DCs are currently under investigation to block their tolerogenic capacities and activate their immunostimulatory functions to increase the efficiency of tumor immunotherapy [160, 161] (Table 4). An additional novel strategy to target all abovementioned subsets of MRCs in cancer involves vaccination against immunosuppressive proteins expressed by these cells, e.g., IDO or Arg-1 [162].

Table 4.

Targeting of DCs

Target Drug Mechanism of action Therapeutic indication
Anti-PD-1 antibody Nivolumab, pembrolizumab Enhancement of T cell activation

Advanced melanoma

Non-small cell lung cancer castration-resistant prostate cancer

Renal cell carcinoma

Colorectal cancer

Anti-Tim-3 antibody Anti-Tim-3 antibodies Enhancement of T cell proliferation Hepatocellular Carcinoma

Conclusion

In the current review that is a part of a symposium-in-writing on MRCs generated following the MYE-EUNITER COST action, we combined the knowledge of many researchers in the field, and described the effects and importance of MRCs in the development and progression of cancer. The myeloid immune system, affected by powerful influences from the tumor and stroma, becomes a crucial player in determining the fate of cancer cells and metastases. As we have shown, all the different types of myeloid cells, i.e., monocytes and macrophages, neutrophils, MDSCs and DCs can have both supportive and detrimental roles in cancer, and their regulation is a potential key target in future anti-cancer therapeutics. The understanding of the regulatory effects of these cells has grown tremendously during the last decade, and it is now clear that these cells are important players that should be explored and targeted in the battle to conquer cancer.

Abbreviations

Arg

Arginase

CCL

C–C motif ligand

cDC

Classical dendritic cell

CXCL

C–X–C motif ligand

DCs

Dendritic cells

EBV

Epstein–Barr virus

EGF

Epidermal growth factor

FGF

Fibroblast growth factor

HDNs

High-density neutrophils

HGF

Hepatocyte growth factor

iNOS

Inducible NO synthase

LDNs

Low-density neutrophils

LFA

Lymphocyte function-associated antigen

Mac

Macrophage antigen

M-CSF

Macrophage colony-stimulating factor

M-MDSCs

Monocytic MDSCs

MMPs

Matrix metalloproteinases

MRCs

Myeloid regulatory cells

NLR

Neutrophil to lymphocyte ratio

PDGF

Platelet-derived growth factor

PLGF

Placenta growth factor

PMN-MDSCs

Polymorphonuclear MDSCs

TAMs

Tumor-associated macrophages

TANs

Tumor-associated neutrophils

TGF

Transforming growth factor

Author contributions

VU, ZGF: writing and revision of the manuscript, revision of the tables and figures. GA, JB, SB, JAVG, JJ, HAP, KCMS, JFS, KS, MV: writing and revision of the manuscript and preparation of the tables. YPC, IS: preparation of the figures. XH, SM, YS: revision of the manuscript and figures.

Funding

This work was supported by COST (European Cooperation in Science and Technology) and the COST Action BM1404 Mye-EUNITER (http://www.mye-euniter.eu). COST is part of the EU Framework Programme Horizon 2020. This work was also supported by Grants from the Cooperation between German Cancer Research Center (DKFZ) and Ministry of Science, Technology and Space of Israel (MOST) in Cancer Research (CA181 to V. Umansky and Z.G. Fridlender).

Compliance with ethical standards

Conflict of interest

The authors declare no conflict of interest.

Footnotes

This paper is part of a Symposium-in-Writing in Cancer Immunology, Immunotherapy by members of the European Network of Investigators Triggering Exploratory Research on Myeloid Regulatory Cells (Mye-EUNITER network).

References

  • 1.Giraldo NA, Becht E, Remark R, Damotte D, Sautès-Fridman C, Fridman WH. The immune contexture of primary and metastatic human tumours. Curr Opin Immunol. 2014;27:8–15. doi: 10.1016/j.coi.2014.01.001. [DOI] [PubMed] [Google Scholar]
  • 2.Coulie PG, Van den Eynde BJ, van der Bruggen P, Boon T. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer. 2014;14:135–146. doi: 10.1038/nrc3670. [DOI] [PubMed] [Google Scholar]
  • 3.Bonavita E, Galdiero MR, Jaillon S, Mantovani A. Phagocytes as corrupted policemen in cancer-related inflammation. Adv Cancer Res. 2015;128:141–171. doi: 10.1016/bs.acr.2015.04.013. [DOI] [PubMed] [Google Scholar]
  • 4.Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883–899. doi: 10.1016/j.cell.2010.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Brandau S, Moses K, Lang S. The kinship of neutrophils and granulocytic myeloid-derived suppressor cells in cancer: cousins, siblings or twins? Semin Cancer Biol. 2013;23:171–182. doi: 10.1016/j.semcancer.2013.02.007. [DOI] [PubMed] [Google Scholar]
  • 6.Bruger AM, Dorhoi A, Esendagli G, Barczyk-Kahlert K, van der Bruggen P, Lipoldova M, Perecko T, Santibanez J, Saraiva M, Van Ginderachter JA, Brandau S. How to measure the immunosuppressive activity of MDSC: assays, problems and potential solutions. Cancer Immunol Immunother. 2018 doi: 10.1007/s00262-018-2170-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ginhoux F, Jung S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat Rev Immunol. 2014;14:392–404. doi: 10.1038/nri3671. [DOI] [PubMed] [Google Scholar]
  • 8.Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958–969. doi: 10.1038/nri2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L, Garner H, Trouillet C, de Bruijn MF, Geissmann F, Rodewald HR. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature. 2015;518:547–551. doi: 10.1038/nature13989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Hoeffel G, Chen J, Lavin Y, Low D, Almeida FF, See P, Beaudin AE, Lum J, Low I, Forsberg EC, Poidinger M, Zolezzi F, Larbi A, Ng LG, Chan JK, Greter M, Becher B, Samokhvalov IM, Merad M, Ginhoux F. C-Myb(+) erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity. 2015;42:665–678. doi: 10.1016/j.immuni.2015.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ginhoux F, Guilliams M. Tissue-resident macrophage ontogeny and homeostasis. Immunity. 2016;44:439–449. doi: 10.1016/j.immuni.2016.02.024. [DOI] [PubMed] [Google Scholar]
  • 12.Movahedi K, Van Ginderachter JA. The ontogeny and microenvironmental regulation of tumor-associated macrophages. Antioxid Redox Signal. 2016;25:775–791. doi: 10.1089/ars.2016.6704. [DOI] [PubMed] [Google Scholar]
  • 13.Movahedi K, Laoui D, Gysemans C, Baeten M, Stange G, Van den Bossche J, Mack M, Pipeleers D, In’tVeld P, De Baetselier P, Van Ginderachter JA. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010;70:5728–5739. doi: 10.1158/0008-5472.CAN-09-4672. [DOI] [PubMed] [Google Scholar]
  • 14.Shand FH, Ueha S, Otsuji M, Koid SS, Shichino S, Tsukui T, Kosugi-Kanaya M, Abe J, Tomura M, Ziogas J, Matsushima K. Tracking of intertissue migration reveals the origins of tumor-infiltrating monocytes. Proc Natl Acad Sci USA. 2014;111:7771–7776. doi: 10.1073/pnas.1402914111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Tymoszuk P, Evens H, Marzola V, Wachowicz K, Wasmer MH, Datta S, Muller-Holzner E, Fiegl H, Bock G, van Rooijen N, Theurl I, Doppler W. In situ proliferation contributes to accumulation of tumor-associated macrophages in spontaneous mammary tumors. Eur J Immunol. 2014;44:2247–2262. doi: 10.1002/eji.201344304. [DOI] [PubMed] [Google Scholar]
  • 16.Bowman RL, Klemm F, Akkari L, Pyonteck SM, Sevenich L, Quail DF, Dhara S, Simpson K, Gardner EE, Iacobuzio-Donahue CA, Brennan CW, Tabar V, Gutin PH, Joyce JA. Macrophage ontogeny underlies differences in tumor-specific education in brain malignancies. Cell Rep. 2016;17:2445–2459. doi: 10.1016/j.celrep.2016.10.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhu Y, Herndon JM, Sojka DK, Kim KW, Knolhoff BL, Zuo C, Cullinan DR, Luo J, Bearden AR, Lavine KJ, Yokoyama WM, Hawkins WG, Fields RC, Randolph GJ, DeNardo DG. Tissue-resident macrophages in pancreatic ductal adenocarcinoma originate from embryonic hematopoiesis and promote tumor progression. Immunity. 2017;47:323–338. doi: 10.1016/j.immuni.2017.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kiss M, Van Gassen S, Movahedi K, Saeys Y, Laoui D. Myeloid cell heterogeneity in cancer: not a single cell alike. Cell Immunol. 2018 doi: 10.1016/j.cellimm.2018.02.008. [DOI] [PubMed] [Google Scholar]
  • 19.Corzo CA, Condamine T, Lu L, Cotter MJ, Youn JI, Cheng P, Cho HI, Celis E, Quiceno DG, Padhya T, McCaffrey TV, McCaffrey JC, Gabrilovich DI. HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med. 2010;207:2439–2453. doi: 10.1084/jem.20100587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Laoui D, Van Overmeire E, Di Conza G, Aldeni C, Keirsse J, Morias Y, Movahedi K, Houbracken I, Schouppe E, Elkrim Y, Karroum O, Jordan B, Carmeliet P, Gysemans C, De Baetselier P, Mazzone M, Van Ginderachter JA. Tumor hypoxia does not drive differentiation of tumor-associated macrophages but rather fine-tunes the M2-like macrophage population. Cancer Res. 2014;74:24–30. doi: 10.1158/0008-5472.CAN-13-1196. [DOI] [PubMed] [Google Scholar]
  • 21.Casazza A, Laoui D, Wenes M, Rizzolio S, Bassani N, Mambretti M, Deschoemaeker S, Van Ginderachter JA, Tamagnone L, Mazzone Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity. Cancer Cell. 2013;24:695–709. doi: 10.1016/j.ccr.2013.11.007. [DOI] [PubMed] [Google Scholar]
  • 22.Van Overmeire E, Stijlemans B, Heymann F, Keirsse J, Morias Y, Elkrim Y, Brys L, Abels C, Lahmar Q, Ergen C, Vereecke L, Tacke F, De Baetselier P, Van Ginderachter JA, Laoui D. M-CSF and GM-CSF receptor signaling differentially regulate monocyte maturation and macrophage polarization in the tumor microenvironment. Cancer Res. 2016;76:35–42. doi: 10.1158/0008-5472.CAN-15-0869. [DOI] [PubMed] [Google Scholar]
  • 23.Ruffell B, Chang-Strachan D, Chan V, Rosenbusch A, Ho CM, Pryer N, Daniel D, Hwang ES, Rugo HS, Coussens LM. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell. 2014;26:623–637. doi: 10.1016/j.ccell.2014.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Laoui D, Keirsse J, Morias Y, Van Overmeire E, Geeraerts X, Elkrim Y, Kiss M, Bolli E, Lahmar Q, Sichien D, Serneels J, Scott CL, Boon L, De Baetselier P, Mazzone M, Guilliams M, Van Ginderachter JA. The tumor microenvironment harbors ontogenically distinct dendritic cell populations with opposing effects on tumor immunity. Nat Commun. 2016;7:13720. doi: 10.1038/ncomms13720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Han Q, Shi H, Liu F. CD163(+) M2-type tumor-associated macrophage support the suppression of tumor-infiltrating T cells in osteosarcoma. Int Immunopharmacol. 2016;34:101–106. doi: 10.1016/j.intimp.2016.01.023. [DOI] [PubMed] [Google Scholar]
  • 26.Harney AS, Arwert EN, Entenberg D, Wang Y, Guo P, Qian BZ, Oktay MH, Pollard JW, Jones JG, Condeelis JS. Real-time imaging reveals local, transient vascular permeability, and tumor cell intravasation stimulated by TIE2hi macrophage-derived VEGFA. Cancer Discov. 2015;5:932–943. doi: 10.1158/2159-8290.CD-15-0012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kitamura T, Qian BZ, Soong D, Cassetta L, Noy R, Sugano G, Kato Y, Li J, Pollard JW. CCL2-induced chemokine cascade promotes breast cancer metastasis by enhancing retention of metastasis-associated macrophages. J Exp Med. 2015;212:1043–1059. doi: 10.1084/jem.20141836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Qian BZ, Zhang H, Li J, He T, Yeo EJ, Soong DY, Carragher NO, Munro A, Chang A, Bresnick AR, Lang RA, Pollard JW. FLT1 signaling in metastasis-associated macrophages activates an inflammatory signature that promotes breast cancer metastasis. J Exp Med. 2015;212:1433–1448. doi: 10.1084/jem.20141555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Nielsen SR, Quaranta V, Linford A, Emeagi P, Rainer C, Santos A, Ireland L, Sakai T, Sakai K, Kim YS, Engle D, Campbell F, Palmer D, Ko JH, Tuveson DA, Hirsch E, Mielgo A, Schmid MC. Macrophage-secreted granulin supports pancreatic cancer metastasis by inducing liver fibrosis. Nat Cell Biol. 2016;18:549–560. doi: 10.1038/ncb3340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, Singh S, Williams C, Soplop N, Uryu K, Pharmer L, King T, Bojmar L, Davies AE, Ararso Y, Zhang T, Zhang H, Hernandez J, Weiss JM, Dumont-Cole VD, Kramer K, Wexler LH, Narendran A, Schwartz GK, Healey JH, Sandstrom P, Labori KJ, Kure EH, Grandgenett PM, Hollingsworth MA, de Sousa M, Kaur S, Jain M, Mallya K, Batra SK, Jarnagin WR, Brady MS, Fodstad O, Muller V, Pantel K, Minn AJ, Bissell MJ, Garcia BA, Kang Y, Rajasekhar VK, Ghajar CM, Matei I, Peinado H, Bromberg J, Lyden D. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527:329–335. doi: 10.1038/nature15756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kumagai S, Marumo S, Shoji T, Sakuramoto M, Hirai T, Nishimura T, Arima N, Fukui M, Huang CL. Prognostic impact of preoperative monocyte counts in patients with resected lung adenocarcinoma. Lung Cancer. 2014;85:457–464. doi: 10.1016/j.lungcan.2014.06.015. [DOI] [PubMed] [Google Scholar]
  • 32.Porrata LF, Ristow K, Colgan JP, Habermann TM, Witzig TE, Inwards DJ, Ansell SM, Micallef IN, Johnston PB, Nowakowski GS, Thompson C, Markovic SN. Peripheral blood lymphocyte/monocyte ratio at diagnosis and survival in classical Hodgkin’s lymphoma. Haematologica. 2012;97:262–269. doi: 10.3324/haematol.2011.050138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Gwak JM, Jang MH, Kim DI, Seo AN, Park SY. Prognostic value of tumor-associated macrophages according to histologic locations and hormone receptor status in breast cancer. PLoS One. 2015;10:e0125728. doi: 10.1371/journal.pone.0125728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Lin CN, Wang CJ, Chao YJ, Lai MD, Shan YS. The significance of the co-existence of osteopontin and tumor-associated macrophages in gastric cancer progression. BMC Cancer. 2015;15:128. doi: 10.1186/s12885-015-1114-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Di Caro G, Cortese N, Castino GF, Grizzi F, Gavazzi F, Ridolfi C, Capretti G, Mineri R, Todoric J, Zerbi A, Allavena P, Mantovani A, Marchesi F. Dual prognostic significance of tumour-associated macrophages in human pancreatic adenocarcinoma treated or untreated with chemotherapy. Gut. 2016;65:1710–1720. doi: 10.1136/gutjnl-2015-309193. [DOI] [PubMed] [Google Scholar]
  • 36.Lu H, Clauser KR, Tam WL, Fröse J, Ye X, Eaton EN, Reinhardt F, Donnenberg VS, Bhargava R, Carr SA, Weinberg RA. A breast cancer stem cell niche supported by juxtacrine signalling from monocytes and macrophages. Nat Cell Biol. 2014;16:1105–1117. doi: 10.1038/ncb3041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, Belaygorod L, Carpenter D, Collins L, Piwnica-Worms D, Hewitt S, Udupi GM, Gallagher WM, Wegner C, West BL, Wang-Gillam A, Goedegebuure P, Linehan DC, DeNardo DG. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 2013;73:1128–1141. doi: 10.1158/0008-5472.CAN-12-2731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ruffell B, Coussens LM. Macrophages and therapeutic resistance in cancer. Cancer Cell. 2015;27:462–472. doi: 10.1016/j.ccell.2015.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kim J, Bae JS. Tumor-associated macrophages and neutrophils in tumor microenvironment. Mediators Inflamm. 2016;2016:6058147. doi: 10.1155/2016/6058147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008;8:618–631. doi: 10.1038/nrc2444. [DOI] [PubMed] [Google Scholar]
  • 41.De Palma M, Venneri MA, Galli R, Sergi L, Politi LS, Sampaolesi M, Naldini L. Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell. 2005;8:211–226. doi: 10.1016/j.ccr.2005.08.002. [DOI] [PubMed] [Google Scholar]
  • 42.Turrini R, Pabois A, Xenarios I, Coukos G, Delaloye JF, Doucey MA. TIE-2 expressing monocytes in human cancers. Oncoimmunology. 2017;6:e1303585. doi: 10.1080/2162402X.2017.1303585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wyckoff J, Wang W, Lin EY, Wang Y, Pixley F, Stanley ER, Graf T, Pollard JW, Segall J, Condeelis J. A paracrine loop between tumor cells and macrophages is required for tumor cell migration in mammary tumors. Cancer Res. 2004;64:7022–7029. doi: 10.1158/0008-5472.CAN-04-1449. [DOI] [PubMed] [Google Scholar]
  • 44.Wang H, Wang X, Li X, Fan Y, Li G, Guo C, Zhu F, Zhang L, Shi Y. CD68(+)HLA-DR(+) M1-like macrophages promote motility of HCC cells via NF-kappaB/FAK pathway. Cancer Lett. 2014;345:91–99. doi: 10.1016/j.canlet.2013.11.013. [DOI] [PubMed] [Google Scholar]
  • 45.Chen Q, Zhang XH, Massague J. Macrophage binding to receptor VCAM-1 transmits survival signals in breast cancer cells that invade the lungs. Cancer Cell. 2011;20:538–549. doi: 10.1016/j.ccr.2011.08.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Smith HA, Kang Y. The metastasis-promoting roles of tumor-associated immune cells. J Mol Med (Berl) 2013;91:411–429. doi: 10.1007/s00109-013-1021-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Kuang DM, Zhao Q, Peng C, Xu J, Zhang JP, Wu C, Zheng L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med. 2009;206:1327–1337. doi: 10.1084/jem.20082173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Savage ND, de Boer T, Walburg KV, Joosten SA, van Meijgaarden K, Geluk A, Ottenhoff TH. Human anti-inflammatory macrophages induce Foxp3+GITR+CD25+ regulatory T cells, which suppress via membrane-bound TGFbeta-1. J Immunol. 2008;181:2220–2226. doi: 10.4049/jimmunol.181.3.2220. [DOI] [PubMed] [Google Scholar]
  • 49.de la Cruz-Merino L, Lejeune M, Nogales Fernández E, Henao Carrasco F, Grueso López A, Illescas Vacas A, Pulla MP, Callau C, Álvaro T. Role of immune escape mechanisms in Hodgkin’s lymphoma development and progression: a whole new world with therapeutic implications. Clin Dev Immunol. 2012;2012:756353. doi: 10.1155/2012/756353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Liu WL, Lin YH, Xiao H, Xing S, Chen H, Chi PD, Zhang G. Epstein–Barr virus infection induces indoleamine 2,3-dioxygenase expression in human monocyte-derived macrophages through p38/mitogen-activated protein kinase and NF-kappaB pathways: impairment in T cell functions. J Virol. 2014;88:6660–6671. doi: 10.1128/JVI.03678-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Beury DW, Parker KH, Nyandjo M, Sinha P, Carter KA, Ostrand-Rosenberg S. Cross-talk among myeloid-derived suppressor cells, macrophages, and tumor cells impacts the inflammatory milieu of solid tumors. J Leukoc Biol. 2014;96:1109–1118. doi: 10.1189/jlb.3A0414-210R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Sinha P, Clements VK, Bunt SK, Albelda SM, Ostrand-Rosenberg S. Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response. J Immunol. 2007;179:977–983. doi: 10.4049/jimmunol.179.2.977. [DOI] [PubMed] [Google Scholar]
  • 53.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. doi: 10.1038/nrclinonc.2016.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Dumitru CA, Gholaman H, Trellakis S, Bruderek K, Dominas N, Gu X, Bankfalvi A, Whiteside TL, Lang S, Brandau S. Tumor-derived macrophage migration inhibitory factor modulates the biology of head and neck cancer cells via neutrophil activation. Int J Cancer. 2011;129:859–869. doi: 10.1002/ijc.25991. [DOI] [PubMed] [Google Scholar]
  • 55.Sionov RV, Fridlender ZG, Granot Z. The multifaceted roles neutrophils play in the tumor microenvironment. Cancer Microenviron. 2015;8:125–158. doi: 10.1007/s12307-014-0147-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Granot Z, Jablonska J. Distinct functions of neutrophil in cancer and its regulation. Mediators Inflamm. 2015;2015:701067. doi: 10.1155/2015/701067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, Worthen GS, Albelda SM. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell. 2009;16:183–194. doi: 10.1016/j.ccr.2009.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Strauss L, Sangaletti S, Consonni FM, Szebeni G, Morlacchi S, Totaro MG, Porta C, Anselmo A, Tartari S, Doni A, Zitelli F, Tripodo C, Colombo MP, Sica A. RORC1 regulates tumor-promoting “emergency” granulo-monocytopoiesis. Cancer Cell. 2015;28:253–269. doi: 10.1016/j.ccell.2015.07.006. [DOI] [PubMed] [Google Scholar]
  • 59.Cortez-Retamozo V, Etzrodt M, Newton A, Rauch PJ, Chudnovskiy A, Berger C, Ryan RJ, Iwamoto Y, Marinelli B, Gorbatov R, Forghani R, Novobrantseva TI, Koteliansky V, Figueiredo JL, Chen JW, Anderson DG, Nahrendorf M, Swirski FK, Weissleder R, Pittet MJ. Origins of tumor-associated macrophages and neutrophils. Proc Natl Acad Sci USA. 2012;109:2491–2496. doi: 10.1073/pnas.1113744109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Jablonska J, Wu CF, Andzinski L, Leschner S, Weiss S. CXCR2-mediated tumor-associated neutrophil recruitment is regulated by IFN-beta. Int J Cancer. 2014;134:1346–1358. doi: 10.1002/ijc.28551. [DOI] [PubMed] [Google Scholar]
  • 61.Jablonska J, Leschner S, Westphal K, Lienenklaus S, Weiss S. Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model. J Clin Invest. 2010;120:1151–1164. doi: 10.1172/JCI37223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kobayashi Y. The role of chemokines in neutrophil biology. Front Biosci. 2008;13:2400–2407. doi: 10.2741/2853. [DOI] [PubMed] [Google Scholar]
  • 63.Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, MacDonald DD, Jin DK, Shido K, Kerns SA, Zhu Z, Hicklin D, Wu Y, Port JL, Altorki N, Port ER, Ruggero D, Shmelkov SV, Jensen KK, Rafii S, Lyden D. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438:820–827. doi: 10.1038/nature04186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Granot Z, Henke E, Comen EA, King TA, Norton L, Benezra R. Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell. 2011;20:300–314. doi: 10.1016/j.ccr.2011.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Wu CF, Andzinski L, Kasnitz N, Kröger A, Klawonn F, Lienenklaus S, Weiss S, Jablonska J. The lack of type I interferon induces neutrophil-mediated pre-metastatic niche formation in the mouse lung. Int J Cancer. 2015;137:837–847. doi: 10.1002/ijc.29444. [DOI] [PubMed] [Google Scholar]
  • 66.Cools-Lartigue J, Spicer J, McDonald B, Gowing S, Chow S, Giannias B, Bourdeau F, Kubes P, Ferri L. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Invest. 2013;123:3446–3458. doi: 10.1172/JCI67484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Coffelt SB, Kersten K, Doornebal CW, Weiden J, Vrijland K, Hau CS, Verstegen NJM, Ciampricotti M, Hawinkels LJAC, Jonkers J, de Visser KE. IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature. 2015;522:345–348. doi: 10.1038/nature14282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.López-Lago MA, Posner S, Thodima VJ, Molina AM, Motzer RJ, Chaganti RS. Neutrophil chemokines secreted by tumor cells mount a lung antimetastatic response during renal cell carcinoma progression. Oncogene. 2013;32:1752–1760. doi: 10.1038/onc.2012.201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Brandau S, Dumitru CA, Lang S. Protumor and antitumor functions of neutrophil granulocytes. Semin Immunopathol. 2013;35:163–176. doi: 10.1007/s00281-012-0344-6. [DOI] [PubMed] [Google Scholar]
  • 70.Coffelt SB, Wellenstein MD, de Visser KE. Neutrophils in cancer: neutral no more. Nat Rev Cancer. 2016;16:431–446. doi: 10.1038/nrc.2016.52. [DOI] [PubMed] [Google Scholar]
  • 71.Leliefeld PH, Koenderman L, Pillay J. How neutrophils shape adaptive immune responses. Front Immunol. 2015;6:471. doi: 10.3389/fimmu.2015.00471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 2013;13:159–175. doi: 10.1038/nri3399. [DOI] [PubMed] [Google Scholar]
  • 73.Dallegri F, Ottonello L. Neutrophil-mediated cytotoxicity against tumour cells: state of art. Arch Immunol Ther Exp (Warsz) 1992;40:39–42. [PubMed] [Google Scholar]
  • 74.van Egmond M, Bakema JE. Neutrophils as effector cells for antibody-based immunotherapy of cancer. Semin Cancer Biol. 2013;23:190–199. doi: 10.1016/j.semcancer.2012.12.002. [DOI] [PubMed] [Google Scholar]
  • 75.Beauvillain C, Delneste Y, Scotet M, Peres A, Gascan H, Guermonprez P, Barnaba V, Jeannin P. Neutrophils efficiently cross-prime naive T cells in vivo. Blood. 2007;110:2965–2973. doi: 10.1182/blood-2006-12-063826. [DOI] [PubMed] [Google Scholar]
  • 76.Kousis PC, Henderson BW, Maier PG, Gollnick SO. Photodynamic therapy enhancement of antitumor immunity is regulated by neutrophils. Cancer Res. 2007;67:10501–10510. doi: 10.1158/0008-5472.CAN-07-1778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Suttmann H, Riemensberger J, Bentien G, Schmaltz D, Stöckle M, Jocham D, Böhle A, Brandau S. Neutrophil granulocytes are required for effective Bacillus Calmette–Guérin immunotherapy of bladder cancer and orchestrate local immune responses. Cancer Res. 2006;66:8250–8257. doi: 10.1158/0008-5472.CAN-06-1416. [DOI] [PubMed] [Google Scholar]
  • 78.Albanesi M, Mancardi DA, Jönsson F, Iannascoli B, Fiette L, Di Santo JP, Lowell CA, Bruhns P. Neutrophils mediate antibody-induced antitumor effects in mice. Blood. 2013;122:3160–3164. doi: 10.1182/blood-2013-04-497446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim D, Nair VS, Xu Y, Khuong A, Hoang CD, Diehn M, West RB, Plevritis SK, Alizadeh AA. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. 2015;21:938–945. doi: 10.1038/nm.3909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Shen M, Hu P, Donskov F, Wang G, Liu Q, Du J. Tumor-associated neutrophils as a new prognostic factor in cancer: A systematic review and meta-analysis. PLoS One. 2014;9:e98259. doi: 10.1371/journal.pone.0098259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Suh B, Ahn YO, Kim TM, Lee JO, Lee SH, Heo DS. CD15+/CD16low human granulocytes from terminal cancer patients: granulocytic myeloid-derived suppressor cells that have suppressive function. Tumour Biol. 2012;33:121–129. doi: 10.1007/s13277-011-0254-6. [DOI] [PubMed] [Google Scholar]
  • 82.Schmielau J, Finn OJ. Activated granulocytes and granulocyte-derived hydrogen peroxide are the underlying mechanism of suppression of T-cell function in advanced cancer patients. Cancer Res. 2001;61:4756–4760. [PubMed] [Google Scholar]
  • 83.Sippel TR, White J, Nag K, Tsvankin V, Klaassen M, Kleinschmidt-DeMasters BK, Waziri A. Neutrophil degranulation and immunosuppression in patients with GBM: restoration of cellular immune function by targeting arginase I. Clin Cancer Res. 2011;17:6992–7002. doi: 10.1158/1078-0432.CCR-11-1107. [DOI] [PubMed] [Google Scholar]
  • 84.Bausch D, Pausch T, Krauss T, Hopt UT, Fernandez-del-Castillo C, Warshaw AL, Thayer SP, Keck T. Neutrophil granulocyte derived MMP-9 is a VEGF independent functional component of the angiogenic switch in pancreatic ductal adenocarcinoma. Angiogenesis. 2011;14:235–243. doi: 10.1007/s10456-011-9207-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Galdiero MR, Bianchi P, Grizzi F, Di Caro G, Basso G, Ponzetta A, Bonavita E, Barbagallo M, Tartari S, Polentarutti N, Malesci A, Marone G, Roncalli M, Laghi L, Garlanda C, Mantovani A, Jaillon S. Occurrence and significance of tumor-associated neutrophils in patients with colorectal cancer. Int J Cancer. 2016;139:446–456. doi: 10.1002/ijc.30076. [DOI] [PubMed] [Google Scholar]
  • 86.Tecchio C, Huber V, Scapini P, Calzetti F, Margotto D, Todeschini G, Pilla L, Martinelli G, Pizzolo G, Rivoltini L, Cassatella MA. IFNalpha-stimulated neutrophils and monocytes release a soluble form of TNF-related apoptosis-inducing ligand (TRAIL/Apo-2 ligand) displaying apoptotic activity on leukemic cells. Blood. 2004;103:3837–3844. doi: 10.1182/blood-2003-08-2806. [DOI] [PubMed] [Google Scholar]
  • 87.Scapini P, Marini O, Tecchio C, Cassatella MA. Human neutrophils in the saga of cellular heterogeneity: insights and open questions. Immunol Rev. 2016;273:48–60. doi: 10.1111/imr.12448. [DOI] [PubMed] [Google Scholar]
  • 88.Moses K, Brandau Human neutrophils: Their role in cancer and relation to myeloid-derived suppressor cells. Semin Immunol. 2016;28:187–196. doi: 10.1016/j.smim.2016.03.018. [DOI] [PubMed] [Google Scholar]
  • 89.Schmidt H, Bastholt L, Geertsen P, Christensen IJ, Larsen S, Gehl J, von der Maase H. Elevated neutrophil and monocyte counts in peripheral blood are associated with poor survival in patients with metastatic melanoma: a prognostic model. Br J Cancer. 2005;93:273–278. doi: 10.1038/sj.bjc.6602702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Eruslanov EB, Bhojnagarwala PS, Quatromoni JG, Stephen TL, Ranganathan A, Deshpande C, Akimova T, Vachani A, Litzky L, Hancock WW, Conejo-Garcia JR, Feldman M, Albelda SM, Singhal S. Tumor-associated neutrophils stimulate T cell resposnses in early-stage human lung cancer. J Clin Invest. 2014;124:5466–5480. doi: 10.1172/JCI77053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Bellocq A, Antoine M, Flahault A, Philippe C, Crestani B, Bernaudin JF, Mayaud C, Milleron B, Baud L, Cadranel J. Neutrophil alveolitis in bronchioloalveolar carcinoma: induction by tumor-derived interleukin-8 and relation to clinical outcome. Am J Pathol. 1998;152:83–92. [PMC free article] [PubMed] [Google Scholar]
  • 92.Templeton AJ, McNamara MG, Šeruga B, Vera-Badillo FE, Aneja P, Ocaña A, Leibowitz-Amit R, Sonpavde G, Knox JJ, Tran B, Tannock IF, Amir E. Prognostic role of neutrophil-to-lymphocyte ratio in solid tumors: a systematic review and meta-analysis. J Natl Cancer Inst. 2014;106:dju124. doi: 10.1093/jnci/dju124. [DOI] [PubMed] [Google Scholar]
  • 93.Jensen HK, Donskov F, Marcussen N, Nordsmark M, Lundbeck F, von der Maase H. Presence of intratumoral neutrophils is an independent prognostic factor in localized renal cell carcinoma. J Clin Oncol. 2009;27:4709–4717. doi: 10.1200/JCO.2008.18.9498. [DOI] [PubMed] [Google Scholar]
  • 94.Ilie M, Hofman V, Ortholan C, Bonnetaud C, Coëlle C, Mouroux J, Hofman P. Predictive clinical outcome of the intratumoral CD66b-positive neutrophil-to-CD8-positive T-cell ratio in patients with resectable nonsmall cell lung cancer. Cancer. 2012;118:1726–1737. doi: 10.1002/cncr.26456. [DOI] [PubMed] [Google Scholar]
  • 95.Jensen TO, Schmidt H, Møller HJ, Donskov F, Høyer M, Sjoegren P, Christensen IJ, Steiniche T. Intratumoral neutrophils and plasmacytoid dendritic cells indicate poor prognosis and are associated with pSTAT3 expression in AJCC stage I/II melanoma. Cancer. 2012;118:2476–2485. doi: 10.1002/cncr.26511. [DOI] [PubMed] [Google Scholar]
  • 96.Tabariès S, Ouellet V, Hsu BE, Annis MG, Rose AA, Meunier L, Carmona E, Tam CE, Mes-Masson AM, Siegel PM. Granulocytic immune infiltrates are essential for the efficient formation of breast cancer liver metastases. Breast Cancer Res. 2015;17:45. doi: 10.1186/s13058-015-0558-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Wislez M, Rabbe N, Marchal J, Milleron B, Crestani B, Mayaud C, Antoine M, Soler P, Cadranel J. Hepatocyte growth factor production by neutrophils infiltrating bronchioloalveolar subtype pulmonary adenocarcinoma: role in tumor progression and death. Cancer Res. 2003;63:1405–1412. [PubMed] [Google Scholar]
  • 98.Ostrand-Rosenberg S, Sinha P. Myeloid-derived suppressor cells: linking inflammation and cancer. J Immunol. 2009;182:4499–4506. doi: 10.4049/jimmunol.0802740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 2012;12:253–268. doi: 10.1038/nri3175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Kanterman J, Sade-Feldman M, Baniyash M. New insights into chronic inflammation-induced immunosuppression. Semin Cancer Biol. 2012;22:307–318. doi: 10.1016/j.semcancer.2012.02.008. [DOI] [PubMed] [Google Scholar]
  • 101.Umansky V, Sevko A. Overcoming immunosuppression in the melanoma microenvironment induced by chronic inflammation. Cancer Immunol Immunother. 2012;61:275–282. doi: 10.1007/s00262-011-1164-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Parker KH, Beury DW, Ostrand-Rosenberg S. Myeloid-derived suppressor cells: critical cells driving immune suppression in the tumor microenvironment. Adv Cancer Res. 2015;128:95–139. doi: 10.1016/bs.acr.2015.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Greten TF, Mandruzzato S, Murray PJ, Ochoa A, Ostrand-Rosenberg S, Rodriguez PC, Sica A, Umansky V, Vonderheide RH, Gabrilovich DI. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun. 2016;7:12150. doi: 10.1038/ncomms12150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Hoechst B, Voigtlaender T, Ormandy L, Gamrekelashvili J, Zhao F, Wedemeyer H, Lehner F, Manns MP, Greten TF, Korangy F. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology. 2009;50:799–807. doi: 10.1002/hep.23054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Filipazzi P, Huber V, Rivoltini L. Phenotype, function and clinical implications of myeloid-derived suppressor cells in cancer patients. Cancer Immunol Immunother. 2012;61:255–263. doi: 10.1007/s00262-011-1161-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Poschke I, Kiessling R. On the armament and appearances of human myeloid-derived suppressor cells. Clin Immunol. 2012;144:250–268. doi: 10.1016/j.clim.2012.06.003. [DOI] [PubMed] [Google Scholar]
  • 107.Solito S, Marigo I, Pinton L, Damuzzo V, Mandruzzato S, Bronte V. Myeloid-derived suppressor cell heterogeneity in human cancers. Ann NY Acad Sci. 2014;1319:47–65. doi: 10.1111/nyas.12469. [DOI] [PubMed] [Google Scholar]
  • 108.Gabrilovich DI. Myeloid-derived suppressor cells. Cancer Immunol Res. 2017;5:3–8. doi: 10.1158/2326-6066.CIR-16-0297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. J Clin Invest. 2015;125:3356–3364. doi: 10.1172/JCI80005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Su Z, Ni P, Zhou C, Wang J. Myeloid-derived suppressor cells in cancers and inflammatory diseases: angel or demon? Scand J Immunol. 2016;84:255–261. doi: 10.1111/sji.12473. [DOI] [PubMed] [Google Scholar]
  • 111.Lang S, Bruderek K, Kaspar C, Höing O, Dominas N, Hussain T, Droege F, Eyth C, Hadaschik B, Brandau S. Clinical relevance and suppressive capacity of human MDSC subsets. Clin Cancer Res. 2018 doi: 10.1158/1078-0432.CCR-17-3726. [DOI] [PubMed] [Google Scholar]
  • 112.Pan PY, Ma G, Weber KJ, Ozao-Choy J, Wang G, Yin B, Divino CM, Chen SH. Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res. 2010;70:99–108. doi: 10.1158/0008-5472.CAN-09-1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Krüger C, Manns MP, Greten TF, Korangy F. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells. Gastroenterology. 2008;135:234–243. doi: 10.1053/j.gastro.2008.03.020. [DOI] [PubMed] [Google Scholar]
  • 114.Obermajer N, Wong JL, Edwards RP, Chen K, Scott M, Khader S, Kolls JK, Odunsi K, Billiar TR, Kalinski P. Induction and stability of human Th17 cells require endogenous NOS2 and cGMP-dependent NO signaling. J Exp Med. 2013;210:1433–1445. doi: 10.1084/jem.20121277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Novitskiy SV, Pickup MW, Gorska AE, Owens P, Chytil A, Aakre M, Wu H, Shyr Y, Moses HL. TGF-β receptor II loss promotes mammary carcinoma progression by Th17 dependent mechanisms. Cancer Discov. 2011;1:430–441. doi: 10.1158/2159-8290.CD-11-0100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Kumar V, Cheng P, Condamine T, Mony S, Languino LR, McCaffrey JC, Hockstein N, Guarino M, Masters G, Penman E, Denstman F, Xu X, Altieri DC, Du H, Yan C, Gabrilovich DI. CD45 phosphatase inhibits STAT3 transcription factor activity in myeloid cells and promotes tumor-associated macrophage differentiation. Immunity. 2016;44:303–315. doi: 10.1016/j.immuni.2016.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Hu CE, Gan J, Zhang RD, Cheng YR, Huang GJ. Up-regulated myeloid-derived suppressor cell contributes to hepatocellular carcinoma development by impairing dendritic cell function. Scand J Gastroenterol. 2011;46:156–164. doi: 10.3109/00365521.2010.516450. [DOI] [PubMed] [Google Scholar]
  • 118.Zhang B, Wang Z, Wu L, Zhang M, Li W, Ding J, Zhu J, Wei H, Zhao K. Circulating and tumor-infiltrating myeloid-derived suppressor cells in patients with colorectal carcinoma. PLoS One. 2013;8:e57114. doi: 10.1371/journal.pone.0057114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Poschke I, Mao Y, Adamson L, Salazar-Onfray F, Masucci G, Kiessling R. Myeloid-derived suppressor cells impair the quality of dendritic cell vaccines. Cancer Immunol Immunother. 2012;61:827–838. doi: 10.1007/s00262-011-1143-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Solito S, Falisi E, Diaz-Montero CM, Doni A, Pinton L, Rosato A, Francescato S, Basso G, Zanovello P, Onicescu G, Garrett-Mayer E, Montero AJ, Bronte V, Mandruzzato S. A human promyelocytic-like population is responsible for the immune suppression mediated by myeloid-derived suppressor cells. Blood. 2011;118:2254–2265. doi: 10.1182/blood-2010-12-325753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Jordan KR, Amaria RN, Ramirez O, Callihan EB, Gao D, Borakove M, Manthey E, Borges VF, McCarter MD. Myeloid-derived suppressor cells are associated with disease progression and decreased overall survival in advanced-stage melanoma patients. Cancer Immunol Immunother. 2013;62:1711–1722. doi: 10.1007/s00262-013-1475-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Pico de Coaña Y, Poschke I, Gentilcore G, Mao Y, Nyström M, Hansson J, Masucci GV, Kiessling R. Ipilimumab treatment results in an early decrease in the frequency of circulating granulocytic myeloid-derived suppressor cells as well as their Arginase1 production. Cancer Immunol Res. 2013;1:158–162. doi: 10.1158/2326-6066.CIR-13-0016. [DOI] [PubMed] [Google Scholar]
  • 123.Weide B, Martens A, Zelba H, Derhovanessian E, Bailur JK, Kyzirakos C, Pflugfelder A, Eigentler TK, Di Giacomo AM, Maio M, Aarntzen EH, de Vries J, Sucker A, Schadendorf D, Büttner P, Garbe C, Pawelec G. Myeloid-derived suppressor cells predict survival of advanced melanoma patients: comparison with regulatory T cells and NY-ESO-1- or Melan-A-specific T cells. Clin Cancer Res. 2014;20:1601–1609. doi: 10.1158/1078-0432.CCR-13-2508. [DOI] [PubMed] [Google Scholar]
  • 124.Condamine T, Dominguez GA, Youn JI, Kossenkov AV, Mony S, Alicea-Torres K, Tcyganov E, Hashimoto A, Nefedova Y, Lin C, Partlova S, Garfall A, Vogl DT, Xu X, Knight SC, Malietzis G, Lee GH, Eruslanov E, Albelda SM, Wang X, Mehta JL, Bewtra M, Rustgi A, Hockstein N, Witt R, Masters G, Nam B, Smirnov D, Sepulveda MA, Gabrilovich DI. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci Immunol. 2016 doi: 10.1126/sciimmunol.aaf8943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Elliot LA, Doherty GA, Sheahan K, Ryan EJ. Human tumor-infiltrating myeloid cells: phenotypic and functional diversity. Front Immunol. 2017;8:86. doi: 10.3389/fimmu.2017.00086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Zhang H, Li ZL, Ye SB, Ouyang LY, Chen YS, He J, Huang HQ, Zeng YX, Zhang XS, Li J. Myeloid-derived suppressor cells inhibit T cell proliferation in human extranodal NK/T cell lymphoma: a novel prognostic indicator. Cancer Immunol Immunother. 2015;64:1587–1599. doi: 10.1007/s00262-015-1765-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Cai TT, Ye SB, Liu YN, He J, Chen QY, Mai HQ, Zhang CX, Cui J, Zhang XS, Busson P, Zeng YX, Li J. LMP1-mediated glycolysis induces myeloid-derived suppressor cell expansion in nasopharyngeal carcinoma. PLoS Pathog. 2017;13:e1006503. doi: 10.1371/journal.ppat.1006503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Jiang H, Gebhardt C, Umansky L, Beckhove P, Schulze TJ, Utikal J, Umansky V. Elevated chronic inflammatory factors and myeloid-derived suppressor cells indicate poor prognosis in advanced melanoma patients. Int J Cancer. 2015;136:2352–2360. doi: 10.1002/ijc.29297. [DOI] [PubMed] [Google Scholar]
  • 129.Romano A, Parrinello NL, Vetro C, Forte S, Chiarenza A, Figuera A, Motta G, Palumbo GA, Ippolito M, Consoli U, Di Raimondo F. Circulating myeloid-derived suppressor cells correlate with clinical outcome in Hodgkin Lymphoma patients treated up-front with a risk-adapted strategy. Br J Haematol. 2015;168:689–700. doi: 10.1111/bjh.13198. [DOI] [PubMed] [Google Scholar]
  • 130.Sade-Feldman M, Kanterman J, Klieger Y, Ish-Shalom E, Olga M, Saragovi A, Shtainberg H, Lotem M, Baniyash M. Clinical significance of circulating CD33+ CD11b+ HLA-DR myeloid cells in patients with stage IV melanoma treated with ipilimumab. Clin Cancer Res. 2016;22:5661–5672. doi: 10.1158/1078-0432.CCR-15-3104. [DOI] [PubMed] [Google Scholar]
  • 131.Gebhardt C, Sevko A, Jiang H, Lichtenberger R, Reith M, Tarnanidis K, Holland-Letz T, Umansky L, Beckhove P, Sucker A, Schadendorf D, Utikal J, Umansky V. Myeloid cells and related chronic inflammatory factors as novel predictive markers in melanoma treatment with ipilimumab. Clin Cancer Res. 2015;21:5453–5459. doi: 10.1158/1078-0432.CCR-15-0676. [DOI] [PubMed] [Google Scholar]
  • 132.Yang L, DeBusk LM, Fukuda K, Fingleton B, Green-Jarvis B, Shyr Y, Matrisian LM, Carbone DP, Lin PC. Expansion of myeloid immune suppressor Gr+ CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell. 2004;6:409–421. doi: 10.1016/j.ccr.2004.08.031. [DOI] [PubMed] [Google Scholar]
  • 133.Toh B, Wang X, Keeble J, Sim WJ, Khoo K, Wong WC, Kato M, Prevost-Blondel A, Thiery JP, Abastado JP. Mesenchymal transition and dissemination of cancer cells is driven by myeloid-derived suppressor cells infiltrating the primary tumor. PLoS Biol. 2011;9:e1001162. doi: 10.1371/journal.pbio.1001162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Kao J, Ko EC, Eisenstein S, Sikora AG, Fu S, Chen SH. Targeting immune suppressing myeloid-derived suppressor cells in oncology. Crit Rev Oncol Hematol. 2011;77:12–19. doi: 10.1016/j.critrevonc.2010.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.De Sanctis F, Solito S, Ugel S, Molon B, Bronte V, Marigo I. MDSCs in cancer: conceiving new prognostic and therapeutic targets. Biochim Biophys Acta. 2016;1865:35–48. doi: 10.1016/j.bbcan.2015.08.001. [DOI] [PubMed] [Google Scholar]
  • 136.Umansky V, Blattner C, Fleming V, Hu X, Gebhardt C, Altevogt P, Utikal J. Myeloid-derived suppressor cells and tumor escape from immune surveillance. Semin Immunopathol. 2017;39:295–305. doi: 10.1007/s00281-016-0597-6. [DOI] [PubMed] [Google Scholar]
  • 137.Guilliams M, Ginhoux F, Jakubzick C, Naik SH, Onai N, Schraml BU, Segura E, Tussiwand R, Yona S. Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat Rev Immunol. 2014;14:571–578. doi: 10.1038/nri3712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Ueno H, Palucka AK, Banchereau J. The expanding family of dendritic cell subsets. Nat Biotechnol. 2010;28:813–815. doi: 10.1038/nbt0810-813. [DOI] [PubMed] [Google Scholar]
  • 139.Veglia F, Gabrilovich DI. Dendritic cells in cancer: the role revisited. Curr Opin Immunol. 2017;45:43–51. doi: 10.1016/j.coi.2017.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Gabrilovich D. Mechanisms and functional significance of tumour-induced dendritic-cell defects. Nat Rev Immunol. 2004;4:941–952. doi: 10.1038/nri1498. [DOI] [PubMed] [Google Scholar]
  • 141.Tang M, Diao J, Gu H, Khatri I, Zhao J, Cattral MS. Toll-like receptor 2 activation promotes tumor dendritic cell dysfunction by regulating IL-6 and IL-10 receptor signaling. Cell Rep. 2015;13:2851–2864. doi: 10.1016/j.celrep.2015.11.053. [DOI] [PubMed] [Google Scholar]
  • 142.Pinzon-Charry A, Maxwell T, Lopez JA. Dendritic cell dysfunction in cancer: a mechanism for immunosuppression. Immunol Cell Biol. 2005;83:451–461. doi: 10.1111/j.1440-1711.2005.01371.x. [DOI] [PubMed] [Google Scholar]
  • 143.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;16:880–886. doi: 10.1038/nm.2172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Ramakrishnan R, Tyurin VA, Veglia F, Condamine T, Amoscato A, Mohammadyani D, Johnson JJ, Zhang LM, Klein-Seetharaman J, Celis E, Kagan VE, Gabrilovich DI. Oxidized lipids block antigen cross-presentation by dendritic cells in cancer. J Immunol. 2014;192:2920–2931. doi: 10.4049/jimmunol.1302801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Andzinski L, Kasnitz N, Stahnke S, Wu CF, Gereke M, von Köckritz-Blickwede M, Schilling B, Brandau S, Weiss S, Jablonska J. Type I IFNs induce anti-tumor polarization of tumor associated neutrophils in mice and human. Int J Cancer. 2016;138:1982–1993. doi: 10.1002/ijc.29945. [DOI] [PubMed] [Google Scholar]
  • 146.Andzinski L, Spanier J, Kasnitz N, Kröger A, Jin L, Brinkmann MM, Kalinke U, Weiss S, Jablonska J, Lienenklaus S. Growing tumors induce a local STING dependent Type I IFN response in dendritic cells. Int J Cancer. 2016;139:1350–1357. doi: 10.1002/ijc.30159. [DOI] [PubMed] [Google Scholar]
  • 147.Woo SR, Fuertes MB, Corrales L, Spranger S, Furdyna MJ, Leung MY, Duggan R, Wang Y, Barber GN, Fitzgerald KA, Alegre ML, Gajewski TF. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity. 2014;41:830–842. doi: 10.1016/j.immuni.2014.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Zoglmeier C, Bauer H, Nörenberg D, Wedekind G, Bittner P, Sandholzer N, Rapp M, Anz D, Endres S, Bourquin C. CpG blocks immunosuppression by myeloid-derived suppressor cells in tumor-bearing mice. Clin Cancer Res. 2011;17:1765–1775. doi: 10.1158/1078-0432.CCR-10-2672. [DOI] [PubMed] [Google Scholar]
  • 149.U’Ren L, Guth A, Kamstock D, Dow S. Type I interferons inhibit the generation of tumor-associated macrophages. Cancer Immunol Immunother. 2010;59:587–598. doi: 10.1007/s00262-009-0776-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Steding CE, Wu ST, Zhang Y, Jeng MH, Elzey BD, Kao C. The role of interleukin-12 on modulating myeloid-derived suppressor cells, increasing overall survival and reducing metastasis. Immunology. 2011;133:221–238. doi: 10.1111/j.1365-2567.2011.03429.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Watkins SK, Egilmez NK, Suttles J, Stout RD. IL-12 rapidly alters the functional profile of tumor-associated and tumor-infiltrating macrophages in vitro and in vivo. J Immunol. 2007;178:1357–1362. doi: 10.4049/jimmunol.178.3.1357. [DOI] [PubMed] [Google Scholar]
  • 152.Mantovani A, Allavena P. The interaction of anticancer therapies with tumor-associated macrophages. J Exp Med. 2015;212:435–445. doi: 10.1084/jem.20150295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Bazzoni F, Tamassia N, Rossato M, Cassatella MA. Understanding the molecular mechanisms of the multifaceted IL-10-mediated anti-inflammatory response: lessons from neutrophils. Eur J Immunol. 2010;40:2360–2368. doi: 10.1002/eji.200940294. [DOI] [PubMed] [Google Scholar]
  • 154.Spranger S, Dai D, Horton B, Gajewski TF. Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell. 2017;31:711–723. doi: 10.1016/j.ccell.2017.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Ma Y, Shurin GV, Peiyuan Z, Shurin MR. Dendritic cells in the cancer microenvironment. J Cancer. 2013;4:36–44. doi: 10.7150/jca.5046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Broz ML, Binnewies M, Boldajipour B, Nelson AE, Pollack JL, Erle DJ, Barczak A, Rosenblum MD, Daud A, Barber DL, Amigorena S, Van’t Veer LJ, Sperling AI, Wolf DM, Krummel MF. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell. 2014;26:638–652. doi: 10.1016/j.ccell.2014.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Sluijter BJ, van den Hout MF, Koster BD, van Leeuwen PA, Schneiders FL, van de Ven R, Molenkamp BG, Vosslamber S, Verweij CL, van den Tol MP, van den Eertwegh AJ, Scheper RJ, de Gruijl TD. Arming the melanoma sentinel lymph node through local administration of CpG-B and GM-CSF: recruitment and activation of BDCA3/CD141(+) dendritic cells and enhanced cross-presentation. Cancer Immunol Res. 2015;3:495–505. doi: 10.1158/2326-6066.CIR-14-0165. [DOI] [PubMed] [Google Scholar]
  • 158.Bharadwaj U, Li M, Zhang R, Chen C, Yao Q. Elevated interleukin-6 and G-CSF in human pancreatic cancer cell conditioned medium suppress dendritic cell differentiation and activation. Cancer Res. 2007;67:5479–5488. doi: 10.1158/0008-5472.CAN-06-3963. [DOI] [PubMed] [Google Scholar]
  • 159.Michielsen AJ, O’Sullivan JN, Ryan EJ. Tumor conditioned media from colorectal cancer patients inhibits dendritic cell maturation. Oncoimmunology. 2012;1:751–753. doi: 10.4161/onci.19570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Macri C, Dumont C, Johnston AP, Mintern JD. Targeting dendritic cells: a promising strategy to improve vaccine effectiveness. Clin Transl Immunology. 2016;5:e66. doi: 10.1038/cti.2016.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Kitamura H, Ohno Y, Toyoshima Y, Ohtake J, Homma S, Kawamura H, Takahashi N, Taketomi A. Interleukin-6/STAT3 signaling as a promising target to improve the efficacy of cancer immunotherapy. Cancer Sci. 2017;108:1947–1952. doi: 10.1111/cas.13332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Andersen MH. The targeting of immunosuppressive mechanisms in hematological malignancies. Leukemia. 2014;28:1784–1792. doi: 10.1038/leu.2014.108. [DOI] [PubMed] [Google Scholar]

Articles from Cancer Immunology, Immunotherapy : CII are provided here courtesy of Springer

RESOURCES