GET THE APP

Polarization of Tumor Infiltrating Leukocytes from Innate Immunit
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2015) Volume 0, Issue 0

Polarization of Tumor Infiltrating Leukocytes from Innate Immunity and their role in the Pro-angiogenic Phenotype in NSCLC

Lorenzo Mortara1, Silvia Zanellato1,2, Barbara Bassani1,3, Andrea Imperatori2, Nicola Rotolo2, Lorenzo Dominioni2, Adriana Albini4, Douglas M Noonan1,3* and Antonino Bruno3
1Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
2Center for Thoracic Surgery, Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
3Science and Technology Center, IRCCS MultiMedica, Milano, Italy
4Department of Research and Statistics, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
*Corresponding Author: Douglas M Noonan, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy 21100, Italy, Tel: +390332217610, Fax: +390332217609 Email:

Abstract

Non-small cell lung cancer (NSCLC), the most frequent lung cancer (80%), can be phenotypically classified into two main subtypes: squamous cell carcinoma (SCC) and adenocarcinoma (ADC). While SCC has relatively rapid doubling times from the onset, ADC has longer doubling times initially that become reduced during tumor progression, suggesting a key role for the microenvironment. During lung tumor progression, a complex and dynamic interplay occurs between proliferating tumor cells and stromal, endothelial and immune tumor-conditioned host cells within the tumor microenvironment (TUMIC). Several factors within the TUMIC, such as hypoxia, cytokines and soluble factors, appear to blunt the anti-tumor immune response and polarize immune cells towards a pro-tumor phenotype. Phenotypically and functionally altered immune cells found in cancer patients include macrophages, neutrophils, myeloid, dendritic, and even NK cells. We studied tumor infiltrating (TINK) and tumor associated (TANK) NK cells in NSCLC. NSCLC TINKs and TANKs show similarities to decidual NK cells, being polarized toward tissue builders, rather than killers, and producing pro-angiogenic cytokines. The functionally polarized immune cells in NSCLC provide the stromal support and neovascularization required for NSCLC tumor expansion and progression in a feed-forward mechanism, leading to tumor progression. Further, systemic alterations of immune cells are also present in NSCLC patients. The precise knowledge of these immune cell alterations within the TUMIC has become crucial for diagnosis, targeted therapeutic intervention, as well as prevention, of NSCLC cancer.

Keywords: Innate immune cells, Polarization, Non-small cell lung cancer, Inflammation, tumor microenvironment, Angiogenesis

Introduction

Lung cancer, a predominant cause of cancer-related mortality in the developed world, is a heterogeneous disease with several histological subtypes. Approximately 80% of all lung cancers are non-small cell lung cancer (NSCLC). NSCLC can be phenotypically further divided into two main subtypes: squamous cell carcinoma (SCC) and adenocarcinoma (ADC) [1]. Standard chemotherapeutic treatments for this devastating disease are only partially effective, and show considerable toxicity, with only fewer than 20% of patients surviving after 5 years from diagnosis; the median survival is less than one year for metastatic disease [1,2]. Surgery represents a valid treatment for early disease, yet still most lung cancers are diagnosed when they become symptomatic and therefore at very late stages. Hence, alternative and innovative therapeutic strategies, including immunotherapies, are urgently needed.

Although in the past lung cancer was not considered an immunesensitive malignancy, currently there is increasing evidence that both principal NSCLC subtypes can evoke specific B and T cell immune responses. For this reason, in addition to standard intervention approaches, numerous research groups focus their efforts on studying the active specific stimulation of the host’s own immune system, termed “therapeutic vaccination”, such as genetically modified autologous tumor cells secreting immune-modulating cytokines, allogenic tumor cells, tumor antigen-pulsed dendritic cells (DC), or therapeutic immune-modulating agents reversing check-point blockades, in particular anti-CTLA-4 or anti-PD-1.

When isolated from host components, tumors cannot expand in mass, they remain a small, clinically indolent disease [3]. For tumor expansion and progression, tumor cells must interact with several host-derived cells consisting of stromal, inflammatory, immune and endothelial cells (ECs) that delineate a complex modified tissue compartment, termed the tumor microenvironment (TUMIC) [4]. Accumulating evidence shows the importance of TUMIC in shaping the tumor mass fate, regulating growth, progression, invasiveness, dissemination and clinical outcome. Among the host-dependent biological features of the tumor hallmarks defined by Hanahan and Weinberg [4] there are “evading immune destruction” and “tumorpromoting inflammation”, which together with the immune orchestration of angiogenesis, points out the key role of the immune system in neoplastic disease [5,6].

Tumor progression leads to tumor immune escape through an array of known and as yet unknown mechanisms. Thorough knowledge of these mechanisms and the resulting clinical situation will be fundamental to plan adequate active anti-tumor intervention in combination with conventional treatment modalities such as surgery and chemotherapy.

It is now clear that the TUMIC polarizes immune cells, in particular the innate compartment, (macrophages, myeloid-derived suppressor cells (MDSCs), regulatory dendritic cells (regDCs)), yet the TUMIC also has an influence on mast cells, cancer-associated fibroblasts (CAFs) and adaptive immunity. Recent data have shown that NK cells are also skewed to a pro-tumor pro-angiogenic phenotype in NSCLC [7-9]. The inflammatory response originating within the TUMIC is a crucial step of the disease, tightly linked to the tumor angiogenesis along with repression of adaptive immune system [6,10]. Tumorigenesis and progression are promoted by different molecules produced within the TUMIC, including pro-angiogenic factors and extracellular matrix-modifying enzymes that facilitate angiogenesis, invasion, and metastasis [11-14]. Moreover, inflammatory cells, by releasing reactive oxygen and nitrogen species, can accelerate genetic mutation events, thereby inducing a faster evolution toward malignancy [12,15].

Major NSCLC subtypes: adenocarcinoma and squamous cell carcinoma

In the past the two major clinical subtypes of NSCLC, adenocarcinoma and squamous cell carcinoma, were treated more or less the same. However, they are quite distinct biologically and clinically. More than two decades ago the studies of Arai et al. [16] showed that surgically resectable SCC features a volume doubling time about 25% shorter compared to resectable ADC. In the same case series, the faster doubling time was a negative prognostic factor, as the SCCs were, on average, more aggressive clinically [16]. At the Center for Thoracic Surgery of the University of Insubria we studied a series of 116 resectable lung cancers, and confirmed that the median tumor volume doubling time of SCC is significantly shorter than that of ADC, both in screen-detected and in incidentally detected NSCLC patients (Table 1). In addition, among the NSCLC cases diagnosed by CT imaging, a technology that identifies tumors at a very early stage, the ADCs with a minimally invasive phenotype (formerly known as bronchioloalveolar carcinomas-BAC) show very long doubling times [17,18]. However, ADCs at later stages appear to have more rapid doubling times, suggesting that while SCC starts out with a rapid doubling time, ADC shows a more complex evolution over time. The biological diversity of the two main NSCLC subtypes is further underlined by their different response to currently available chemotherapy regimens and targeted therapies.

Mode of lung cancer detection Adenocarcinoma (n=69) Squamous cell carcinoma (n=47)
Chest X-ray screening*      
n. of pts 25 16  
Median TVDT, days 109 80 0.0347
(IQR) (91-185) (56-108)  
Incidental**      
n. of pts 44 31  
Median TVDT, days 227 95 0.0108
(IQR) (108-349) (64-230)  
P° (screening vs. incidental) 0.0099 0.2128  

^TVDT calculated according to Schwartz formula [194] after comparison of two consecutive chest X-rays or CT-scans.
°Mann-Whitney test
*Annual repeat screen during population-based chest X-ray screening programme carried out in the Province of Varese, Italy in 1997-2011 [195].
**Asymptomatic subjects with incidental NSCLC detection, for whom previous imaging study was available for tumor size comparisons.
TVDT: Tumor Volume Doubling Time; IQR: Interquartile Range (25%-75%).

Table 1: Median tumor volume doubling time (TVDT)^ of 116 non-small cell lung cancers (NSCLC) by histology and by mode of detection (case series 1997-2011, Center for Thoracic Surgery, University of Insubria).

Currently, targeted therapies are used almost exclusively in lung ADC, and the distinction between subtypes is imperative not only in advanced but also in early stage NSCLC undergoing surgical resection, to optimize treatment in case of relapsing disease [19]. The epithelial growth factor receptor (EGFR) inhibitors erlotinib, gefitinib and afatinib are selectively indicated for treatment of NSCLCs with EGFR mutations, which predominantly occur in ADCs [20]. Similarly, the EML4-ALK rearrangement occurs predominantly in ADC cells [21], and it is associated with susceptibility to the targeted agent crizotinib, although acquired drug resistance almost inevitably develops [22]. Importantly, the angiogenesis inhibitor bevacizumab and the folate anti-metabolite pemetrexed are excluded from use in patients with SCC, as treatment with these molecules has been associated respectively with the occurrence of life-threatening hemorrhages or with lack of effectiveness [23].

Different “actors” and “scenarios” in the tumor microenvironment

Macrophages and tumor-associated macrophages:

Macrophages are innate immune cells characterized by high plasticity associated with nearly opposite functional programs induced by different microenvironment signals [24]. Crucial functions of macrophages range from phagocytosis, induction of inflammation, recognition and elimination of invading pathogens, antigen presentation and for resolution of inflammation and tissue repair, remodeling and maintenance of tissue homeostasis [25]. Two main subtypes of macrophages have been characterized: M1 (classically activated) macrophages, that possess high anti-microbial activities, pro-inflammatory function and display anti-tumor activity, mediated for example by production of reactive nitrogen and oxygen intermediates, and by synthesis of pro-inflammatory cytokines, and M2 (alternatively activated) macrophages that, conversely, are involved in tissue remodeling and repair activities, immunesuppressive effects, pro-angiogenic and pro-tumor functions. M1 macrophages are also characterized by a high capacity for IL-12 and IL-23 secretion, related to the induction or development of Th1 response, while M2 macrophages are closely related to the tumorassociated macrophage (TAM) profile and associated with an IL-10 secretory phenotype associated with Th2 and immune-suppression [24,25].

However, the M1/M2 macrophage paradigm represent extremes across a gradient of macrophage polarization, thus macrophages can display both M1 and M2 markers and M1 and M2 macrophages can be present in the same tissue [24]. In this context, it is conceivable that there is an evolving macrophage balance that integrates over time a changing phenotype in response to the microenvironment, depending on soluble tissue-derived signals and on cell-to-cell interactions, including the presence of polarized T lymphocytes, Treg and NK cells [26-28]. Experimental results have suggested that tumor-associated macrophages (TAMs) are an heterogeneous cell population with cells showing various degrees of polarization, also comprising Tie2- expressing monocytes/macrophages (TEMs) [29], and other immature myeloid cells, in particular myeloid-derived suppressive cells (MDSCs) [27]

In the natural history of cancer, TAMs become switched early on to an immature activation state, resembling the M2 macrophage polarization program. This includes low production of reactive oxygen species (ROS), poor release of inflammatory cytokines (IL-1β, IL-6, TNFα and IL-12), enhanced release of anti-inflammatory cytokines (IL-10), decreased efficiency in antigen presentation, an increase in pro-angiogenic features and immune-suppressive functions [30,31].

Several studies performed on different transgenic mouse models evaluating the potential role of TAMs in the tumor progression in vivo demonstrate that up-regulation of TAM number is associated with increased tumor growth [32], while their depletion or inhibition leads to a relevant inhibition of tumor angiogenesis and progression [33]. In multiple murine models of carcinomas, it has been well established that TAMs acquire a predominant role in the regulation of tumor angiogenesis, releasing crucial factors, such as vascular endothelial growth factor A (VEGF-A), matrix metalloproteinase 9 (MMP-9) and placental growth factor (PlGF) [26].

TAMs originate from blood monocytes recruited from the tumor vasculature as well as from adjacent tissues by different types of tumor microenvironment-derived chemokines (Figure 1), in particular MCSF (CSF-1), CCL2 (MCP-1), VEGF, Angiopoietin-2 (ANG-2), and CXCL12 (SDF-1) [26,27,34-36]. TAMs represent a relevant part of the inflammatory component infiltrating solid cancers, exerting a crucial role in the regulation of cancer-related inflammation and tumor progression [37]. Presence and quantity of TAMs have been correlated with a poor prognosis for patients with diverse types of tumors, including breast, prostate and bladder cancers [38,39].

clinical-cellular-immunology-tumor-insurgence

Figure 1: M2 pro-angiogenic macrophage polarization and tumorassociated macrophage in NSCLC. During tumor insurgence and progression a complex and diversified interplay occurs between tumor cells and stromal, endothelial and innate immune cells within the TUMIC and in particular M2 macrophages and TAMs. Several tumor-derived factors, such as hypoxia, chemokines e.g. CXCL1, CXCL2, IL-8, VEGF or from tumor conditioned myeloid cells, e.g. IL-6, CCL2, VEGF, MMPs, plasmin, and other molecules could play a relevant role in the triggering of the angiogenic switch, contributing to the tumor angiogenesis, lymphangiogenesis, the invasive process, and metastasis.

Accumulating evidence has shown that different types of cytokines, chemokines, growth factors and proteolytic enzymes can be produced by TAMs and these molecules have been suggested to act as key effectors in tumor angiogenesis and progression during the “angiogenic switch” [40,41]. However, in the case of NSCLC, current data are not conclusive concerning the prognostic role of the collective TAM tumor infiltrate in term of recurrence-free survival and overall survival. TAM density in NSCLC correlated with tumor IL-8 mRNA levels and high microvessel counts, suggesting a role of TAMs in the tumor angiogenesis [42,43], as well as with accelerated lymphangiogenesis [44] (Figure 1). However, this scenario still requires further investigation since there are still opposite and conflicting results on the role of TAMs in NSCLC [45-49].

In lung cancer, TAMs have been suggested to support tumor progression contributing to stroma formation and angiogenesis induction through the release of platelet-derived growth factor (PDGF), in association with TGFβ production by the cancer cells [26]. Through production of proteases, including MMP-1,-2,-3,-9, and-12, plasmin, urokinase-type plasminogen activator (uPA) and the uPA receptor, macrophages can also regulate the degradation of the extracellular matrix (ECM), weakening the connective tissue and favoring enhanced infiltration and metastasis.

Interestingly, a gene profile study on primary NSCLC tissues showed that TAMs are associated with higher gene expression of cathepsin K, MMP-9, uPA, VEGF, PDGF, HGF and COX-2 [50] when compared to monocytes from healthy donors. A high expression of cathepsin was observed in NSCLC patients with a poor prognosis [51]. Moreover, higher gene expression of MMP-9 and VEGF-A in TAMs were present in the later stage of disease as compared to early stages (Figures 1 and 2), including in patients with lymph node metastasis [50]. Functional studies using conditioned media derived from TAMs demonstrated that these cells release soluble factors able to improve cell lung cancer invasion and migration in vitro. TAM invasiveness has been associated with their ability to release of MMP-9 and uPA while targeting of these two molecules by mAbs significantly impaired TAM invasion.

clinical-cellular-immunology-innate-immune

Figure 2: Polarization of innate immune cells toward a pro angiogenic phenotype and function during NSCLC tumor growth and progression. The tumor mass fate, their invasive capacity, dissemination and clinical outcome are strictly dependent of the interactions with innate polarized pro-tumor and pro-angiogenic innate cells. Among these cells, M2/TAM, N2/TAN, regDC, MDSC, TINK and CAF are main actors playing key roles in shaping the TUMIC with capacities that lead to evasion from immune recognition and destruction, tumor-promoting inflammation and angiogenesis.

Hypoxia is a crucial condition for the development of several cancer types, inducing the necessary microenvironment for tumor establishment [52,53]. High TAM numbers accumulate in hypoxic areas of tumors and this process can triggers a pro-angiogenic program in these cells [54] as a function of IL-10 release, hypoxia and polarization [31]. Therefore, infiltrating TAMs represent an indirect pathway of promotion of angiogenesis, together with other tumorderived angiogenic molecules.

In a recent study on NSCLC adenocarcinomas [55], tumor infiltration by M2 macrophages was directly correlated with metastasis (Figure 1). These data highlight the role exerted by hypoxia in the development of polarized TAMs through the activation of ERK pathway. Further studies will be necessary to clarify which specific transcription factors are involved in the macrophage polarization process. Studies performed in the murine Lewis lung carcinoma (LLC) model also demonstrated that TAMs were associated with metastasis and angiogenesis both in vitro and in vivo. Tumor hypoxia selectively promoted the in vitro M2 polarization of murine macrophages in conjunction with stimulation of IL-6 [56]. IL-6 has been shown to be involved as a crucial activator for the oncogenic transcription factor STAT3 [56] that has been suggested to be involved in the M2 macrophage polarization [27] (Figure 1). Noteworthy, when hypoxia and IL-6-induced murine M2 macrophages were injected with LLC cells in vivo, the metastasis rate increased dramatically as well as the proportion of CD31-positive cells as compared to controls, suggesting a clear correlation between M2 macrophages and tumor angiogenesis [56].

Lactic acid released from hypoxic murine tumor (LLC or B16) cells supported the role of hypoxia in skewing functional M2 macrophage polarization [57]. A by-product of anaerobic glycolysis, lactic acid might play a crucial role in inducing expression of VEGF and arginase 1 (ARG1) in TAMs, a process that was mediated by hypoxia-inducible factor 1 alpha (HIF-1α). These studies directly suggest that the possibility to interfere with the M2-type polarization during tumor progression could represent an innovative therapeutic intervention strategy [58,59]. In vitro endothelial cell chemotaxis assays performed on human peripheral mononuclear cells exposed to conditioned media of the Calu 6 and A549 NSCLC cell lines showed a marked increased in monocyte angiogenic activity. This was correlated with enhanced levels of CXC chemokines: IL-8 (CXCL8), CXCL5, and CXCL1. Interestingly, macrophage angiogenic activity was enhanced by tumorderived macrophage inhibitory factor (MIF).

A xenograft model using the highly metastatic NCI-H460-LNM35 (LNM35) human lung carcinoma cell line, in comparison to the low metastatic cell line NCI-H460-N15 (N15), found that the highly metastatic cancer cells were associated with increased macrophage and neutrophil infiltration and lymph node metastasis in vivo. This phenomenon was correlated with human and murine expression of IL-1, and VEGF-A,-C and-D molecules [60]. Interestingly, the enhanced F4/80+ infiltrating macrophages, detected by immunohistochemistry, expressed specific markers for the M2-type, i.e. IL-10 and ARG1 mRNAs, as well as for both VEGF-A and VEGFC mRNAs (Figures 1 and 2). The high angiogenic and lymphangiogenic potential of LNM35 tumors appeared to be mediated by induction of CXC chemokines: IL-8 (CXCL8), CXCL5 and CXCL1 by cancer cells (Figure 1), in conjunction with a IL-1-driven inflammatory signaling that lead to the recruitment of M2-type macrophages.

Concerning the role of the TEM subset in tumor vascularization and progression, it has been reported that selective depletion, or inhibition of their migration capacities, results in substantial inhibition of both tumor angiogenesis and progression in various mouse tumor models [61,62]. TEMs represent a subset of already preprogrammed highly pro-angiogenic circulating human monocytes that can be detected in several tumor infiltrates, including lung carcinomas, near to the tumor blood vasculature as well as in the stroma [63]. In contrast to Tie2-monocytes, TEMs showed higher expression of MMP-9, VEGF-A, COX-2, and WNT5A [64]. Furthermore, exposure to ANG-2 induced enhanced tumor-promoting functions of TEMs, in particular increasing expression of two important pro-angiogenic enzymes: thymidine phosphorylase (TP) and cathepsin B (CTSB) [64]. Purified peripheral TEMs from healthy human donors, induced angiogenesis in xenotransplanted human tumors while Tie2- monocytes did not, suggesting a key role for TEMs in human cancer growth [63]. ANG-2 has also been implicated in lung metastasis and formation of the pre-metastatic niche by TEMs [65].

Neutrophils

Neutrophils are the most abundant circulating leukocytes and represent the first-line host defense against infectious microorganisms. In response to diverse inflammatory stimuli, neutrophils migrate from blood to infected tissues, where they efficiently contain pathogens through several strategies: phagocytosis which involves the microorganism engulfment and subsequent elimination in phagolysosomes and degranulation which results in potent antimicrobial lytic molecule release (including activation of myeloperossidase (MPO) and the respiratory burst) resulting in copious production of reactive oxygen species (ROS) (O−2, H2O2, HOCl) [66,67] and formation of neutrophil extracellular traps. In addition, neutrophils are also able to produce several cytokines (TNFα, IL-1β, IL-12) and chemokines (CXCL8, CXCL10, CCL3, and CCL4) (Figure 2) directly involved in the complex immune cell cross-talk in order to regulate the balance between humoral and cell-mediated immunity [68,69].

Neutrophils represent a significant inflammatory cell infiltrate component observed in a wide array of murine and human tumors, including head and neck cancer [70], renal cell carcinoma [71], melanoma [72], hepatocellular cancer [73], colon cancer [74], gastric cancer [75] and lung cancer [46,76], suggesting a potential involvement in tumor development.

Although the role of tumor-associated neutrophils (TANs) in cancer progression remains unclear and is currently debated, increased systemic neutrophil levels [77] and high intra-tumor neutrophil density were found to be a negative prognostic factor [76]. The first evidence of neutrophil infiltrated BAC was reviewed in 1998 when Bellocq et al. reported that an enhanced TAN number was significantly associated with a poor outcome in BAC patients [78]. Increased blood neutrophil numbers was associated with a negative prognosis and a poor overall survival (OS) in patients with advanced NSCLC [79] and with small cell lung carcinoma (SCLC) [80]. Intratumor CD66b+ neutrophils in resectable NSCLC were correlated with a significantly increased relapse incidence [76].

Like macrophages, neutrophils have also been reported to be able to display at least two different phenotypes associated with diverse biological behavior. Fridlender et al. demonstrated that in mouse models of lung cancer, TGFβ represents a crucial factor that induces a shift from an anti-tumor (N1) to a pro-tumor (N2) neutrophil phenotype (Figure 2). When TGFβ activity was blocked, N1 neutrophils were observed to be associated with direct tumor cell killing and CD8+ T cell activation, whereas N2 neutrophils were predominant in the control animals bearing tumors [81].

Early-stage NSCLC TANs displayed a CD62LlowCD54high phenotype with a characteristic pattern of chemokine receptors including CCR5, CCR7, CXCR3, and CXCR4 [82]. These TANs were able to release pro-inflammatory cytokines (CCL2, IL-8, CCL3, and IL-6) as well as the anti-inflammatory IL-1R antagonist (Figure 2). Both TANs and neutrophils isolated from distant non-malignant lung tissues were able to induce T cell proliferation and IFNγ production. TAN-T cell cross-talk induced an increase of CD54, CD86, OX40L, and 4-1BBL co-stimulatory molecules on the neutrophil surface, which fostered T cell proliferation in a positive-feedback loop [82].

Several recent studies have highlighted the importance of the neutrophil-to-lymphocyte ratio, a biomarker of the host systemic inflammatory response whose increase has been reported to be associated with a poor outcome in patients with SCLC [83] and NSCLC patients receiving standard chemotherapy [84]. In an in-vitro study, Hattar et al. demonstrated that the co-incubation of A549 with neutrophils induced proliferation of A549 cells by an elastasedependent mechanism. Moreover, a specific COX-2 inhibitor was able to decrease A549 proliferation in the presence of neutrophils [85]. Inhibition of COX-2 has been associated with reduction of lung cancer incidence [3]. Together these data suggested the potential relevance of the interactions between neutrophils and tumor cells that could lead to the release of inflammatory mediators that potentially are able to enhance tumor cell growth and inhibition of this process may help delay tumor onset.

Dendritic cells

 

Dendritic cells (DCs) are the most important antigen presenting cells (APCs) primarily involved in inducing the adaptive immune response, whereas immature DCs are able to prevent the activation of auto-reactive T cells and the onset of autoimmunity [86,87].

Following maturation, DCs give rise to a heterogeneous population of cells in which the two main actors are conventional DCs (cDCs) (also called myeloid (mDCs)) and plasmacytoid (pDCs) [88]. cDCs are able to secrete IL-12 and can be reproduced in vitro using CD34+ precursors or blood monocytes using appropriate stiumuli; when exposed to TGFβ they are able to differentiate into Langerhans cells (LCs). On the other hand, pDCs can release interferon-alpha (IFNα) and are derived from lymphoid precursors, express the surface marker CD123 (interleukin IL-3 receptor, IL-3R) and their growth is dependent on the presence of IL-3 [89-91].

During cancer promotion and progression, the maturation of DCs is strongly inhibited by different signals from the microenvironment and DCs remain in an immature state, thereby acquiring a tolerogenic and immunosuppressive properties [92,93].

Patients with NSCLC are characterized by reduced CD11c+ cDCs, in particular in lymph nodes of patients with metastasis or in lymph nodes located adjacent to the primary tumor in subjects without metastasis. Moreover, DCs within the tumor show low expression of HLA-DR antigens, suggesting that their ability for antigen presentation is severely impaired [94]. Among CD11c+ cDCs present in lung cancer, three major subpopulations have been identified in NSCLC: CD1a+(a marker that specifically identifies immature DCs [95]) Langerin+cells, CD1a+Langerin-cells, and CD1a-DC-SIGN +CD68-cells [96,97]. CD123+ pDCs have also been observed in lower numbers in tumor tissues as compared to healthy tissue [98].

CD83+ mature DCs are found in a very low numbers in tumor tissues, higher numbers are found in adjacent tissues [100]. NSCLC tumor tissues also contain three subsets of cDCs based on CD11c expression: high (CD11chigh cDC), intermediate (CD11cint cDC) and low (CD11c-pDC). Tumor-infiltrating CD11chigh cDCs show a lower state of maturation as compared with DCs from peripheral blood, and the stimulation of tumor-infiltrating cDCs by TLR4 or TLR8 ligands are only able to induce secretion of limited amounts of cytokines. CD11cint DCs represent a quarter of total DCs found in tumor and adjacent tissues, they express low levels of co-stimulatory molecules and high levels of immune-inhibitory molecules, such as B7-H1. The decreased numbers of CD11c+ pDCs found in tumors represent immature cells that are able to secret low amount of IFNα upon TLR9 stimulation [100].

High VEGF expression and DC infiltration have been reported to be inversely correlated in tumor specimens of NSCLC and are associated with a poor prognosis [101] suggesting that VEGF might inhibit or regulate DC recruitment and/or activation. Conversely, a high density of mature DCs can be considered a good predictor for clinical outcome in NSCLC, as it is correlated with prolonged survival and it may identify patients with early-stage tumors [102].

DCs with immunosuppressive functions (Figure 2) are termed regulatory DCs (regDCs) [103]. It was recently demonstrated that, in mice, development of LLC was associated with intra-tumor accumulation of regDCs [104].

NSCLC cells that are positive for programmed-death receptor ligand 1 (PD-L1) are associated with histological subtypes and overall survival. Patients with ADC or prognosis after surgery less than 3 years show higher expression rate of PD-L1, thus this molecule might be regarded as a poor prognostic factor. PD-L1 was detected in CD1a+ immature DCs in NSCLC, potentially maintaining DC in an immature state and contributing to the immune escape and disease progression [105]. The B7-H3 co-inhibitory molecule (a member of the PD-L family) is up-regulated in NSCLC-residing DC, this phenomenon correlates with lymph node metastasis [106]. High levels of circulating B7-H3+ DCs are associated with tumor stage and metastasis diffusion. NSCLC-derived DCs show immunosuppressive activities and are able to release large amounts of IL-10 (Figure 2), contributing to an immature phenotype and tolerogenic state [106]. DCs, like other immune cells, can be conditioned and polarized by tumor microenvironment into tolerogenic cells able to contribute to tumor progression, immunosuppression and angiogenesis by releasing TGFβ, polarizing T lymphocytes into pro-tumor Th2 cells (Figure 2) and activating Treg cells [104].

Myeloid-derived suppressor cells

Myeloid-derived suppressor cells (MDSCs) include a heterogeneous population of immature myeloid and myeloid progenitor cells endowed with immunosuppressive properties, pro-angiogenic potential, and able to support metastatic spread [107,108]. Human MDSCs are described as CD11b+, CD33+, CD16low, HLA-DRneg/low and can be classified into two major subsets based on CD14 marker expression: the CD14+ subtype, termed monocytic MDSCs (MMDSCs), and the CD14-CD15+ subtype that are polymorphonuclear MDSCs (PMN-MDSCs) [109]. The murine counterpart is characterized by the co-expression of Gr-1 and CD11b [108].

Tumor and inflammatory milieu-derived mediators such as IL-1β, IL-6, IL-10, VEGF and GM-CSF are crucial factors able to induce MDSC recruitment, expansion, and triggering functions [110]. Murine MDSCs exert a direct role in the promotion of tumor angiogenesis through release of soluble factors, such as MMP-9 and VEGF, and by their ability to trans-differentiate into ECs [111,112]. In contrast, the MMP inhibitor TIMP-2 targets NSCLC MDSCs and impairs their angiogenic and immunosupressive potential [113]. In addition, it has been proposed that murine MDSCs could act as regDCs, since once isolated from lungs of healthy mice and co-cultured with LCC conditioned medium, they acquire inhibitory functions. [104]. regDCs can also secrete TGFβ and IL-10 (Figure 2) resulting in the suppression of immune responses, mainly through inhibition of T helper, T cytotoxic lymphocytes and natural killer cells [114] as well as the induction of Tregs [115].

HLA-DR-/low M-MDSCs have been found to be increased in the peripheral blood of NSCLC patients as compared to healthy donors [116] and their numbers were associated with extrathoracic metastasis, as well as response to chemotherapy and tumor progression. MMDSCs appear to exert very low allo-stimulatory activity and show the ability to inhibit both autologous CD4+ and CD8+ T cell proliferation and IFNγ production in a cell-to-cell contact-dependent manner. MMDSCs have been shown to express the NADPH oxidase component gp91phox and are able to generate high level of reactive oxygen species (ROS) suggesting that their suppressive effect on T cells is mainly mediated by ROS production [116]. In advanced NSCLC patients, it has been shown that PMN-MDSC peripheral count was increased, and this subset, when co-cultured in vitro with CD8+ T cells, was able to reduce the expression of CD3ζ chain leading to the suppression of T cell proliferation and induction of apoptosis [117]. In peripheral blood the CD11b+CD14-MDSC subpopulation is decreased in advanced disease stage patients that are responsive to chemotherapy or in earlystage patients after tumor mass surgical resection [117].

Depletion of L-arginine by the enzyme ARG1 has been reported as an additional mechanism by which MDSCs are able to exert their immunosuppressive role. ARG1 is mainly produced by PMN-MDSC and the ARG1 levels in peripheral blood of NSCLC patients are correlated with PMN-MDSC count [118]. In different types of cancer, including NSCLC, an accumulation in the peripheral blood of immature CD66b+ PMN-MDSCs in the MDSC fraction with immunosuppressive activities has been found [119]. These cells display altered surface marker expression, longer survival and decreased ability to act as effector cells when compared to neutrophils derived from healthy subjects. Furthermore, PMN-MDSCs lack CXCR1 and CXCR2 receptors, suggesting an extravasation and tumor tissue infiltration defects in vivo [119]. In a murine model of lung cancer, treatment with mAb against the Gr1 or Ly6G markers depleted MDSCs, resulting in enhanced anti-tumor immune responses and inhibition of pro-angiogenic activities [120]. When MDSC depletion was induced using LLC-Ovalbumin expressing tumor cells, the treatment resulted in enhanced therapeutic vaccination responses with marked inhibition of tumor growth and strong reduction in tumor burden compared to controls.

Finally, MDSCs have been shown to be crucial in the modulation of immune responses, contributing to angiogenesis and promoting tumor progression and metastases [109]. Accumulating experimental data show an inverse correlation between MDSC number, cancer clinical stage, and prognosis, suggesting that MDSCs could be a potential marker correlating clinical outcome and response to therapy; larger prospective trials are needed to determine the role of MDSC as a biomarker [121].

Natural killer cells

Natural killer (NK) cells are innate immune cell effectors able to recognize and eliminate tumor and virus-infected cells. NKs constitute a heterogeneous population of large granular lymphocytes that comprise approximately 10-15% of peripheral blood mononuclear cells (PBMC) in humans. Several human NK cell subsets have been described on the basis of the expression of two main surface antigens, CD56 and CD16. CD56dimCD16+ NK cells constitutes about 90-95% of peripheral blood NK cells and are associated with target cell eradication through the secretion of perforin, granzyme and antibody dependent cellular cytotoxicity (ADCC) [122]. The second NK cell subset, CD56brightCD16-, represents about 5-10% of peripheral blood NK cells. These NK cells are poorly cytotoxic but able to release large amounts of cytokines, including IFNγ, TNFα, and GM-CSF. A peculiar third NK cell subset has been found in the decidua during implantation [123,124]. Decidual NK cells (dNKs) are CD56superbrightCD16-, have low cytotoxicity [125], are tolerogenic, participate in the protection of the developing embryo, and are involved in decidual angiogenesis producing copious quantities of angiogenic factors [125] as well as promoting decidual cellularity [126].

dNKs represent a clear example of the microenvironment involvement in shaping immune cell plasticity and response. A similar scenario has been described in the context of lung cancer. Carrega et al. showed that CD56brightCD16-subset represent the predominant NK subset infiltrating NSCLC [8]. Tumor infiltrating NK cells have been reported to exert poor cytotoxicity on target tumor cells [8,9,127-129]. We demonstrated that CD56brightCD16- NK cells infiltrating resectable NSCLC tumors, and even peripheral blood NK cells in patients with NSCLC, express pro-angiogenic cytokines (Figure 2) including VEGF, PlGF and IL-8 [7]. These factors are functionally active in vitro on endothelial cells. These data suggest that NK cells contribute to tumor angiogenesis in NSCLC. Like dNK cells and NK cells in some cases of tissue injury, the NSCLC tumor infiltrating NK cells (TINKs) and peripheral blood NK cells from oncology patients (tumor associated NK cells or TANKs) [130] have pro-angiogenic activities [7]. We also observed that the NK-associated pro-angiogenic activity was particularly pronounced in patients with SCC NSCLC, again suggesting that SCC subtype starts aggressively but remains constant over time, while ADC continues to gain increasing malignancy as time passes, in keeping with clinical observations. These data indicate that the NSCLC TUMIC exerts a potent polarizing effect on NK cells, resulting in phenotype and functional alterations of these cells both locally and systemically. Several TUMIC-derived/associated factors have been report to impair NK “normal function”, including acidity, hypoxia, immune suppressive cytokines (in particular TGFβ), exosomes.

TGFβ is one of the numerous TUMIC factors involved in the induction of immune cell polarization [131], and is expressed at high levels both in the TUMIC and in the decidua [130]. TGFβ has been found to polarize the CD56dimCD16-peripheral NK cells toward a decidual-like phenotype expressing CD56brightCD16-, KIR+ CD9+CD49a+ [130]. TGFβ has been shown to inhibit CD16-mediated human NK cell IFNγ-production and ADCC through SMAD3 [132]. TGFβ appears to contribute to the induction of the angiogenic switch of NK cells from healthy individuals [130].

An hypoxic microenvironment is another common theme between the decidua [133] and the TUMIC [134]. A combination of TGFβ, hypoxia, and a demethylating agent have been found to convert sorted peripheral blood CD56dimCD16+ NK cells into a dNK-like cell phenotype characterized by low cytotoxicity associated with high expression levels of VEGF, CD9 marker and KIRs [135].

Adenosine is a soluble immunomodulatory molecule acting through adenosine receptors (in particular A1, A2A, A2B, and A3) expressed on multiple immune subsets, including NK cells. Adenosine peaks during decidualization [136] and up to 20-fold increases in the extracellular fluid of solid carcinomas has been reported [137]. Once released in the extracellular environment, adenosine has been reported to impair NK cell normal function by decreasing TNFα secretion (following IL-2 stimulation), inhibiting cytotoxic granule exocytosis, repressing perforin and Fas ligand-mediated cytotoxic activity as well as cytokine production [138].

Finally, exosomes are abundantly released by tumor cells and present in the TUMIC, and represent another mechanism by which tumors regulate NK cell plasticity by impairing NK killing efficiency [139]. This impairment includes the down-regulating perforin/ granzyme production and/or NKG2D ligand expression [140]. The production of NKG2D ligand-bearing exosomes has also been proposed as a mechanism for tumor cell escape from immune recognition [141]. Moreover, the granzyme B-inhibitory serpin proteinase inhibitor-9 (PI-9) has also been identified inside exosomes [142] that could also play an important role in the resistance of tumor cells to NK cell lysis.

Mast Cells

Mast cells (MCs) represent another inflammatory cell type associated with high plasticity that are able to regulate different aspects of inflammatory responses, angiogenesis, allergic reactions, tissue repair, remodeling and tumor. MCs have been reported to accumulate in several types of tumors in response to diverse tumor-derived chemoattractant factors, including RANTES, CCL2, stem cell factor (SCF) [143]. MCs are able to release several pro-angiogenic factors, such as fibroblast growth factor 2 (FGF-2), VEGF-A,-B,-C,-D, IL-8, TNFα, TGFβ [144]. Moreover they store secretory granules preformed active serine proteases, including tryptase and chymase [145]. Tryptase is a strong stimulator of EC proliferation leading to promote vascular tube formation in vitro, and also a valuable activator of both MMPs and PA [146].

MC numbers within tumor tissues have been correlated with tumor angiogenesis, cancer progression and poor prognosis in lung adenocarcinomas [147-150]. Interestingly, the accumulation rate of MCs was significantly different between diverse types of NSCLC [151]. Proliferation of SCLC was found to be associated with the expression of H1, H2, H3 and H4 histamine-receptors [151]. H1-receptor inhibition resulted in an improved overall survival rate and decrease tumor proliferation by inhibiting mast cell recruitment and release of VEGF and HIF-1α [152]. Abundant MC accumulation in solid cancers, including lung carcinomas, has been showed to closely associate with the number of blood vessels surrounding solid tumors apparently sustaining tumor angiogenesis [153]. MC contribution to tumor angiogenesis is exerted by the release of growth factors, including VEGF, IL-8 and MMP-9 secretion, which in turn facilitates tumor invasiveness [144]. Studies using mast cell-deficient KitW/ KitWvor KitW-sh/KitW-sh mice showed that tumor-associated MCs significantly contribute to tumor angiogenesis, enhancing tumor growth and metastasis [154-156]. However, there are some controversial studies in NSCLC concerning the role of MCs as predictors of poor survival [147,148]. Although angiogenesis and MC density were found to be positively associated, only microvascular density, and not the MC count, was correlated with poor survival in SCC NSCLC patients [147]. A positive beneficial role for tumor MC accumulation together with CD68+ macrophage infiltration has been reported [47], adding some unresolved and conflicting features for defining the importance of MCs infiltrating NSCLC as a driving force in tumor angiogenesis and progression.

Cancer-associated fibroblasts

Cancer-associated fibroblasts (CAFs) are a major component of the TUMIC (Figure 2), regulating important tumor cell functions by secreting several cytokines, chemokines and growth factors, such as TGFβ, VEGF, CXCL12, HGF, FGF, ECM proteins and ECM degrading enzymes (MMPs) [157-159]. CAFs constitute a major portion of the reactive tumor cell stroma and play a crucial role in tumor progression. The main precursors of CAFs are normal fibroblasts, and the transdifferentiation of fibroblasts to CAFs is driven to a great extent by cancer-derived cytokines such as TGFβ [161]. The tumor-promoting effects of CAFs have been described as a hallmark of cancer, since they are directly associated with evasion of apoptosis, sustained angiogenesis promotion, and tissue invasion and metastasis [162]. CAFs are also able to secrete IL-6 (Figure 2) and cardiotrophinlike cytokine factor 1 (CLCF1), cytokines that play a role in the promotion of NSCLC growth [163]. Compared with normal fibroblasts, CAFs appear to increase the invasiveness of co-cultured NSCLC cells in vitro and also enhanced tumorigenicity of NSCLC cells lines in vivo [164]. Mechanisms involved appear to be enhancement of integrin α11, CTHRC1 , SULF1 , MFAP5 , CLU , and THBS2 expression [164]. These genes are regulated by the TGFβ1 signaling pathway [165,166], crucial for CAF differentiation and for the induction of epithelial to mesenchymal transition (EMT) [167].

The pro-metastatic role of CAFs in NSCLC was confirmed in coculture studies: CAFs induce an increase in motility of NSCLC cells through the expression of alpha-smooth muscle actin, and a decrease in proliferation through a SMAD3-dependent up-regulation of the growth inhibitory gene p21 (CDKN1A) [167]. CAFs secrete ECM degrading enzymes, in particular MMPs (Figure 2), facilitating tumor invasion and metastasis [159]. Through recruitment of endothelial precursor cells (EPCs) and monocytes [168-170], CAFs contribute to pro-angiogenic and tumor progression. In breast cancer, the proangiogenic properties exerted by CAFs is linked to their ability to recruit endothelial precursor cells (EPCs) via SDF-1/CXCL12 secretion [169] and monocytes via CCL2-CCR2A/2B signaling pathway [168]. The role of CAFs in the promotion of angiogenesis in the context NSCLC remains to be elucidated. Tissue fibrosis has been epidemiologically associated with increased risk for tumors, and recent studies have suggested a correlation between matrix rigidity and cancer insurgence [171,172]. However, using a genetic murine model, deletion of αSMAC+ fibroblasts actually stimulated progression of pancreatic cancer [173], indicating we still have a great deal to understand in the role of CAFs within the TUMIC.

T regulatory cells

Regulatory T cells (Tregs) are immunosuppressive lymphocytes able to impair auto-reactive T cell activity and maintain immunological self-tolerance. In NSCLC, Tregs (defined as CD4+CD25highFOXP3+ [12]) number is associated with advanced tumor growth and poor prognosis [175]. T cell infiltration in malignant and non-malignant lung tissues was found to be similar, and both tissue and tumorinfiltrating T cells show no functional impairment. However, CD4+CD25+CD127-Treg cells are present only in malignant tissues [177]. Additional data confirm that NSCLC patients display an increased percentage of Treg cells as compared to controls, and demonstrate that theTregcell count is increased in relation to tumor stage and higher in patients with metastasis as compared to nonmetastatic patients [176,177]. Furthermore, (Tregs) are found in all lung carcinoma subtypes, but with significant enrichment in adenocarcinoma respect to squamous carcinoma [178].

Increased accumulation of (Tregs) in NSCLC tissues is correlated with poor prognosis [179,180], in particular in smoking patients. Smoking patients characterized by a higher number of(Tregs) [181] have a significantly higher risk for recurrent disease [179]. Moreover, high Treg count in the regional lymph nodes is associated with an unfavorable prognostic factor even in lymph node negative patients [182]. The frequency of peripheral Tregs was found to be significantly higher in tumor of patients with pleural invasion, vessel invasion, lymphatic vessel invasion and recurrent disease [183].

Forkhead box protein 3 (FOXP3) and toll-like receptor 4 (TLR4) are relevant factors exerting a role in tumor escape and tumor growth. The expression of these two factors in NSCLC is higher than in normal lung tissue. FOXP3 expression correlates with lymph node metastasis and tumor staging and is aged-related, whereas TLR4 expression is related with tumor differentiation [184,185]. Moreover, T cell immunoglobulin-3 (TIM-3) is a negative regulatory molecule that plays a critical role in immune tolerance. Nearly 60% of FOXP3+ tumor-infiltrating lymphocytes are TIM-3+, and TIM-3 expression on CD4+ T cells correlated with poor clinical-pathological parameters of NSCLC patients, such as nodal metastasis and advanced cancer stages [186].

Induction of CD4+CD25+FOXP3+ Tregs occurs in vitro when CD4+ T cells are exposed to antigens or polyclonal activators in the presence of immunosuppressive cytokines, in particular IL-10 or TGFβ [187]. It is known that TGFβ is expressed in patients with high risk to develop NSCLC [188]. Both ADC and SSC patients exhibited higher levels of serum IL-10 and TGFβ than healthy controls [189]. TGFβ-mediated immunosuppression is in relationship with aberrant inflammation (COX-2/PGE2 pathway) and this is related to the induction of polarization to Tregs. PGE2 is able to stimulate the development of Tregs, both in vitro and in vivo, suggesting a crucial role of tumorderived COX-2 promotion of the Treg phenotype. COX-2/PGE2 and TGFβ are both implicated in tumorigenesis and are capable to generate peripherally induced Tregs [190,191].

TGFβ plasma concentration in NSCLC patients directly correlated with the frequency of circulating CD4+CD25+FOXP3+ Tregs. These cells display higher expression levels of FOXP3 if compared to Tregs of control subjects. It is of note that in resected lung tumor tissue specimens, a co-expression of TGFβ, COX-2, and FOXP3 is found [192]. Anti-CD25 IgG plasma level was significantly higher in patients with NSCLC than control subjects, in particular in patients at stage III of NSCLC, suggesting that antibody specificity could be used as a biomarker for prognosis of lung cancers in analogy with the enhancement of Treg count in peripheral blood [193]. Whilw it is clear that Tregs play a role in the promotion of tumor tolerance and immunosuppression their specific contribution to tumor angiogenesis in NSCLC still requires further investigation.

Concluding remarks

The NSCLC TUMIC is able to polarize most innate immune cells toward a pro-tumor phenotype. In some cases this can be extended systemically, with peripheral cells also being affected. Polarization within the TUMIC is then critical for permitting tumor angiogenesis, suppression and subversion of the adaptive immune system, leading to tumor progression. Thus the TUMIC and the immune cells within the TUMIC can be considered targets for prevention and therapy.

References

  1. Non-small cell lung cancer treatment (PDQ®): general information about non-small cell lung cancer (NSCLC).
  2. Imperatori A, Harrison RN, Leitch DN, Rovera F, Lepore G, et al. (2006) Lung cancer in Teesside (UK) and Varese (Italy): a comparison of management and survival.Thorax 61: 232-239.
  3. Albini A, Tosetti F, Li VW, Noonan DM, Li WW (2012) Cancer prevention by targeting angiogenesis.Nat Rev Clin Oncol 9: 498-509.
  4. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation.Cell 144: 646-674.
  5. Bruno A, Pagani A, Magnani E, Rossi T, Noonan DM, et al. (2014) Inflammatory angiogenesis and the tumor microenvironment as targets for cancer therapy and prevention.Cancer Treat Res 159: 401-426.
  6. Noonan DM, De Lerma Barbaro A, Vannini N, Mortara L, Albini A (2008) Inflammation, inflammatory cells and angiogenesis: decisions and indecisions.Cancer Metastasis Rev 27: 31-40.
  7. Bruno A, Focaccetti C, Pagani A, Imperatori AS, Spagnoletti M, et al. (2013) The proangiogenic phenotype of natural killer cells in patients with non-small cell lung cancer.Neoplasia 15: 133-142.
  8. Carrega P, Morandi B, Costa R, Frumento G, Forte G, et al. (2008) Natural killer cells infiltrating human nonsmall-cell lung cancer are enriched in CD56 bright CD16(-) cells and display an impaired capability to kill tumor cells.Cancer 112: 863-875.
  9. Platonova S, Cherfils-Vicini J, Damotte D, Crozet L, Vieillard V, et al. (2011) Profound coordinated alterations of intratumoral NK cell phenotype and function in lung carcinoma.Cancer Res 71: 5412-5422.
  10. de Visser KE, Eichten A, Coussens LM (2006) Paradoxical roles of the immune system during cancer development.Nat Rev Cancer 6: 24-37.
  11. DeNardo DG, Andreu P, Coussens LM (2010) Interactions between lymphocytes and myeloid cells regulate pro-versus anti-tumor immunity.Cancer Metastasis Rev 29: 309-316.
  12. Grivennikov SI, Greten FR, Karin M (2010) Immunity, inflammation, and cancer.Cell 140: 883-899.
  13. Qian BZ, Pollard JW (2010) Macrophage diversity enhances tumor progression and metastasis.Cell 141: 39-51.
  14. Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, et al. (2007) Mesenchymal stem cells within tumour stroma promote breast cancer metastasis.Nature 449: 557-563.
  15. Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A (2009) Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability.Carcinogenesis 30: 1073-1081.
  16. Arai T, Kuroishi T, Saito Y, Kurita Y, Naruke T, et al. (1994) Tumor doubling time and prognosis in lung cancer patients: evaluation from chest films and clinical follow-up study. Japanese Lung Cancer Screening Research Group.Jpn J Clin Oncol 24: 199-204.
  17. Aoki T, Nakata H, Watanabe H, Nakamura K, Kasai T, et al. (2000) Evolution of peripheral lung adenocarcinomas: CT findings correlated with histology and tumor doubling time.AJR Am J Roentgenol 174: 763-768.
  18. Hasegawa M, Sone S, Takashima S, Li F, Yang ZG, et al. (2000) Growth rate of small lung cancers detected on mass CT screening.Br J Radiol 73: 1252-1259.
  19. Langer CJ, Besse B, Gualberto A, Brambilla E, Soria JC (2010) The evolving role of histology in the management of advanced non-small-cell lung cancer.J Clin Oncol 28: 5311-5320.
  20. Roengvoraphoj M, Tsongalis GJ, Dragnev KH, Rigas JR (2013) Epidermal growth factor receptor tyrosine kinase inhibitors as initial therapy for non-small cell lung cancer: focus on epidermal growth factor receptor mutation testing and mutation-positive patients. Cancer treatment reviews 39: 839-50.
  21. Shaw AT, Yeap BY, Mino-Kenudson M, Digumarthy SR, Costa DB, et al. (2009) Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK.J Clin Oncol 27: 4247-4253.
  22. Doebele RC, Pilling AB, Aisner DL, Kutateladze TG, Le AT, et al. (2012) Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer.Clin Cancer Res 18: 1472-1482.
  23. Sandler AB, Schiller JH, Gray R, Dimery I, Brahmer J, et al. (2009) Retrospective evaluation of the clinical and radiographic risk factors associated with severe pulmonary hemorrhage in first-line advanced, unresectable non-small-cell lung cancer treated with Carboplatin and Paclitaxel plus bevacizumab. J Clin Oncol 27: 1405-12
  24. Sica A, Mantovani A (2012) Macrophage plasticity and polarization: in vivo veritas.J Clin Invest 122: 787-795.
  25. Sica A, Larghi P, Mancino A, Rubino L, Porta C, et al. (2008) Macrophage polarization in tumour progression.Semin Cancer Biol 18: 349-355.
  26. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A (2002) Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes.Trends Immunol 23: 549-555.
  27. Sica A, Bronte V (2007) Altered macrophage differentiation and immune dysfunction in tumor development.J Clin Invest 117: 1155-1166.
  28. Bellora F, Castriconi R, Dondero A, Reggiardo G, Moretta L, et al. (2010) The interaction of human natural killer cells with either unpolarized or polarized macrophages results in different functional outcomes.Proc Natl Acad Sci U S A 107: 21659-21664
  29. De Palma M, Venneri MA, Galli R, Sergi Sergi L, Politi LS, et al. (2005) Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell 8: 211-226
  30. Guo C, Buranych A, Sarkar D, Fisher PB, Wang XY (2013) The role of tumor-associated macrophages in tumor vascularization. Vascular cell 5: 20
  31. Wu WK, Llewellyn OP, Bates DO, Nicholson LB, Dick AD (2010) IL-10 regulation of macrophage VEGF production is dependent on macrophage polarisation and hypoxia.Immunobiology 215: 796-803.
  32. Bottazzi B, Walter S, Govoni D, Colotta F, Mantovani A (1992) Monocyte chemotactic cytokine gene transfer modulates macrophage infiltration, growth, and susceptibility to IL-2 therapy of a murine melanoma.J Immunol 148: 1280-1285.
  33. Lin EY, Nguyen AV, Russell RG, Pollard JW (2001) Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy.J Exp Med 193: 727-740.
  34. Saccani A, Schioppa T, Porta C, Biswas SK, Nebuloni M, et al. (2006) p50 nuclear factor-kappaB overexpression in tumor-associated macrophages inhibits M1 inflammatory responses and antitumor resistance. Cancer Res 66: 11432-40.
  35. Schioppa T, Uranchimeg B, Saccani A, Biswas SK, Doni A, et al. (2003) Regulation of the chemokine receptor CXCR4 by hypoxia.J Exp Med 198: 1391-1402.
  36. Murdoch C, Tazzyman S, Webster S, Lewis CE (2007) Expression of Tie-2 by human monocytes and their responses to angiopoietin-2.J Immunol 178: 7405-7411.
  37. Balkwill FR, Mantovani A (2012) Cancer-related inflammation: common themes and therapeutic opportunities.Semin Cancer Biol 22: 33-40.
  38. Chen P, Bonaldo P (2013) Role of macrophage polarization in tumor angiogenesis and vessel normalization: implications for new anticancer therapies.Int Rev Cell Mol Biol 301: 1-35.
  39. Lewis CE, Pollard JW (2006) Distinct role of macrophages in different tumor microenvironments.Cancer Res 66: 605-612.
  40. Bergers G, Benjamin LE (2003) Tumorigenesis and the angiogenic switch.Nat Rev Cancer 3: 401-410.
  41. Lin EY, Li JF, Gnatovskiy L, Deng Y, Zhu L, et al. (2006) Macrophages regulate the angiogenic switch in a mouse model of breast cancer.Cancer Res 66: 11238-11246.
  42. Yuan A, Yang PC, Yu CJ, Chen WJ, Lin FY, et al. (2000) Interleukin-8 messenger ribonucleic acid expression correlates with tumor progression, tumor angiogenesis, patient survival, and timing of relapse in non-small-cell lung cancer.Am J Respir Crit Care Med 162: 1957-1963.
  43. Chen JJ, Yao PL, Yuan A, Hong TM, Shun CT, et al. (2003) Up-regulation of tumor interleukin-8 expression by infiltrating macrophages: its correlation with tumor angiogenesis and patient survival in non-small cell lung cancer.Clin Cancer Res 9: 729-737.
  44. Zhang BC, Gao J, Wang J, Rao ZG, Wang BC, et al. (2011) Tumor-associated macrophages infiltration is associated with peritumoral lymphangiogenesis and poor prognosis in lung adenocarcinoma.Med Oncol 28: 1447-1452.
  45. Al-Shibli K, Al-Saad S, Donnem T, Persson M, Bremnes RM, et al. (2009) The prognostic value of intraepithelial and stromal innate immune system cells in non-small cell lung carcinoma.Histopathology 55: 301-312.
  46. Carus A, Ladekarl M, Hager H, Pilegaard H, Nielsen PS, et al. (2013) Tumor-associated neutrophils and macrophages in non-small cell lung cancer: no immediate impact on patient outcome.Lung Cancer 81: 130-137.
  47. Welsh TJ, Green RH, Richardson D, Waller DA, O'Byrne KJ, et al. (2005) Macrophage and mast-cell invasion of tumor cell islets confers a marked survival advantage in non-small-cell lung cancer.J Clin Oncol 23: 8959-8967.
  48. Dai F, Liu L, Che G, Yu N, Pu Q, et al. (2010) The number and microlocalization of tumor-associated immune cells are associated with patient's survival time in non-small cell lung cancer.BMC Cancer 10: 220.
  49. Ma J, Liu L, Che G, Yu N, Dai F, et al. (2010) The M1 form of tumor-associated macrophages in non-small cell lung cancer is positively associated with survival time.BMC Cancer 10: 112.
  50. Wang R, Zhang J, Chen S, Lu M, Luo X, et al. (2011) Tumor-associated macrophages provide a suitable microenvironment for non-small lung cancer invasion and progression.Lung Cancer 74: 188-196.
  51. Cordes C, Bartling B, Simm A, Afar D, Lautenschläger C, et al. (2009) Simultaneous expression of Cathepsins B and K in pulmonary adenocarcinomas and squamous cell carcinomas predicts poor recurrence-free and overall survival.Lung Cancer 64: 79-85.
  52. Vaupel P, Mayer A (2007) Hypoxia in cancer: significance and impact on clinical outcome.Cancer Metastasis Rev 26: 225-239.
  53. Swinson DE, Jones JL, Richardson D, Wykoff C, Turley H, et al. (2003) Carbonic anhydrase IX expression, a novel surrogate marker of tumor hypoxia, is associated with a poor prognosis in non-small-cell lung cancer.J Clin Oncol 21: 473-482.
  54. Murdoch C, Muthana M, Lewis CE (2005) Hypoxia regulates macrophage functions in inflammation.J Immunol 175: 6257-6263.
  55. Zhang J, Cao J, Ma S, Dong R, Meng W, et al. (2014) Tumor hypoxia enhances Non-Small Cell Lung Cancer metastasis by selectively promoting macrophage M2 polarization through the activation of ERK signaling.Oncotarget 5: 9664-9677.
  56. Zhang L, Alizadeh D, Van Handel M, Kortylewski M, Yu H, et al. (2009) Stat3 inhibition activates tumor macrophages and abrogates glioma growth in mice.Glia 57: 1458-1467.
  57. Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, et al. (2014) Functional polarization of tumour-associated macrophages by tumour-derived lactic acid.Nature 513: 559-563.
  58. Colombo MP, Mantovani A (2005) Targeting myelomonocytic cells to revert inflammation-dependent cancer promotion.Cancer Res 65: 9113-9116.
  59. Sica A, Schioppa T, Mantovani A, Allavena P (2006) Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti-cancer therapy.Eur J Cancer 42: 717-727.
  60. Watari K, Shibata T, Kawahara A, Sata K, Nabeshima H, et al. (2014) Tumor-derived interleukin-1 promotes lymphangiogenesis and lymph node metastasis through M2-type macrophages.PLoS One 9: e99568.
  61. Mazzieri R, Pucci F, Moi D, Zonari E, Ranghetti A, et al. (2011) Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells.Cancer Cell 19: 512-526.
  62. Bouvard C, Segaoula Z, De Arcangelis A, Galy-Fauroux I, Mauge L, et al. (2014) Tie2-dependent deletion of α6 integrin subunit in mice reduces tumor growth and angiogenesis.Int J Oncol 45: 2058-2064.
  63. Venneri MA, De Palma M, Ponzoni M, Pucci F, Scielzo C, et al. (2007) Identification of proangiogenic TIE2-expressing monocytes (TEMs) in human peripheral blood and cancer.Blood 109: 5276-5285.
  64. Coffelt SB, Tal AO, Scholz A, De Palma M, Patel S, et al. (2010) Angiopoietin-2 regulates gene expression in TIE2-expressing monocytes and augments their inherent proangiogenic functions.Cancer Res 70: 5270-5280.
  65. Albini A, Noonan DM (2012) Angiopoietin2 and tie2: tied to lymphangiogenesis and lung metastasis. New perspectives in antimetastatic antiangiogenic therapy.J Natl Cancer Inst 104: 429-431.
  66. Papayannopoulos V, Zychlinsky A (2009) NETs: a new strategy for using old weapons.Trends Immunol 30: 513-521.
  67. Borregaard N, Cowland JB (1997) Granules of the human neutrophilic polymorphonuclear leukocyte.Blood 89: 3503-3521.
  68. Scapini P, Lapinet-Vera JA, Gasperini S, Calzetti F, Bazzoni F, et al. (2000) The neutrophil as a cellular source of chemokines.Immunol Rev 177: 195-203.
  69. Di Carlo E, Forni G, Lollini P, Colombo MP, Modesti A, et al. (2001) The intriguing role of polymorphonuclear neutrophils in antitumor reactions.Blood 97: 339-345.
  70. Trellakis S, Bruderek K, Dumitru CA, Gholaman H, Gu X, et al. (2011) Polymorphonuclear granulocytes in human head and neck cancer: enhanced inflammatory activity, modulation by cancer cells and expansion in advanced disease.Int J Cancer 129: 2183-2193.
  71. Jensen HK, Donskov F, Marcussen N, Nordsmark M, Lundbeck F, et al. (2009) Presence of intratumoral neutrophils is an independent prognostic factor in localized renal cell carcinoma.J Clin Oncol 27: 4709-4717.
  72. Jensen TO, Schmidt H, Møller HJ, Donskov F, Høyer M, et al. (2012) Intratumoral neutrophils and plasmacytoid dendritic cells indicate poor prognosis and are associated with pSTAT3 expression in AJCC stage I/II melanoma.Cancer 118: 2476-2485.
  73. Li YW, Qiu SJ, Fan J, Zhou J, Gao Q, et al. (2011) Intratumoral neutrophils: a poor prognostic factor for hepatocellular carcinoma following resection.J Hepatol 54: 497-505.
  74. Rao HL, Chen JW, Li M, Xiao YB, Fu J, et al. (2012) Increased intratumoral neutrophil in colorectal carcinomas correlates closely with malignant phenotype and predicts patients' adverse prognosis.PLoS One 7: e30806.
  75. Caruso RA, Bellocco R, Pagano M, Bertoli G, Rigoli L, et al. (2002) Prognostic value of intratumoral neutrophils in advanced gastric carcinoma in a high-risk area in northern Italy.Mod Pathol 15: 831-837.
  76. Ilie M, Hofman V, Ortholan C, Bonnetaud C, Coëlle C, et al. (2012) Predictive clinical outcome of the intratumoral CD66b-positive neutrophil-to-CD8-positive T-cell ratio in patients with resectable nonsmall cell lung cancer.Cancer 118: 1726-1737.
  77. Teramukai S, Kitano T, Kishida Y, Kawahara M, Kubota K, et al. (2009) Pretreatment neutrophil count as an independent prognostic factor in advanced non-small-cell lung cancer: an analysis of Japan Multinational Trial Organisation LC00-03.Eur J Cancer 45: 1950-1958.
  78. Bellocq A, Antoine M, Flahault A, Philippe C, Crestani B, et al. (1998) Neutrophil alveolitis in bronchioloalveolar carcinoma: induction by tumor-derived interleukin-8 and relation to clinical outcome.Am J Pathol 152: 83-92.
  79. Paesmans M, Sculier JP, Libert P, Bureau G, Dabouis G, et al. (1995) Prognostic factors for survival in advanced non-small-cell lung cancer: univariate and multivariate analyses including recursive partitioning and amalgamation algorithms in 1,052 patients. The European Lung Cancer Working Party. J Clin Oncol 13: 1221-30
  80. Paesmans M, Sculier JP, Lecomte J, Thiriaux J, Libert P, et al. (2000) Prognostic factors for patients with small cell lung carcinoma: analysis of a series of 763 patients included in 4 consecutive prospective trials with a minimum follow-up of 5 years.Cancer 89: 523-533.
  81. Fridlender ZG, Albelda SM (2012) Tumor-associated neutrophils: friend or foe? Carcinogenesis 33:949-955.
  82. Eruslanov EB, Bhojnagarwala PS, Quatromoni JG, Stephen TL, Ranganathan A, et al. (2014) Tumor-associated neutrophils stimulate T cell responses in early-stage human lung cancer.J Clin Invest 124: 5466-5480.
  83. Kang MH, Go SI, Song HN, Lee A, Kim SH, et al. (2014) The prognostic impact of the neutrophil-to-lymphocyte ratio in patients with small-cell lung cancer.Br J Cancer 111: 452-460.
  84. Pinato DJ, Shiner RJ, Seckl MJ, Stebbing J, Sharma R, et al. (2014) Prognostic performance of inflammation-based prognostic indices in primary operable non-small cell lung cancer.Br J Cancer 110: 1930-1935.
  85. Hattar K, Franz K, Ludwig M, Sibelius U, Wilhelm J, et al. (2014) Interactions between neutrophils and non-small cell lung cancer cells: enhancement of tumor proliferation and inflammatory mediator synthesis. Cancer Immunol Immunother 63: 1297-306
  86. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, et al. (2000) Immunobiology of dendritic cells.Annu Rev Immunol 18: 767-811.
  87. Mellman I, Steinman RM (2001) Dendritic cells: specialized and regulated antigen processing machines.Cell 106: 255-258.
  88. Liu K, Nussenzweig MC (2010) Origin and development of dendritic cells.Immunol Rev 234: 45-54.
  89. Ardavín C, Martínez del Hoyo G, Martín P, Anjuère F, Arias CF, et al. (2001) Origin and differentiation of dendritic cells.Trends Immunol 22: 691-700.
  90. Trinchieri G1 (2003) Interleukin-12 and the regulation of innate resistance and adaptive immunity.Nat Rev Immunol 3: 133-146.
  91. Asselin-Paturel C, Trinchieri G (2005) Production of type I interferons: plasmacytoid dendritic cells and beyond.J Exp Med 202: 461-465.
  92. Lenahan C, Avigan D (2006) Dendritic cell defects in patients with cancer: mechanisms and significance.Breast Cancer Res 8: 101.
  93. Pinzon-Charry A, Maxwell T, López JA (2005) Dendritic cell dysfunction in cancer: a mechanism for immunosuppression.Immunol Cell Biol 83: 451-461.
  94. Zielinski P, Dyszkiewicz W, Piwkowski CT, Dworacki G, Gasiorowski L (2009) Can the condition of the cell microenvironment of mediastinal lymph nodes help predict the risk of metastases in non-small cell lung cancer? Cancer Epidemiol 33: 387-390.
  95. Coventry BJ, Morton J (2003) CD1a-positive infiltrating-dendritic cell density and 5-year survival from human breast cancer.Br J Cancer 89: 533-538.
  96. Thomachot MC, Bendriss-Vermare N, Massacrier C, Biota C, Treilleux I, et al. (2004) Breast carcinoma cells promote the differentiation of CD34+ progenitors towards 2 different subpopulations of dendritic cells with CD1a(high)CD86(-)Langerin-and CD1a(+)CD86(+)Langerin+ phenotypes. Int J Cancer 110: 710-20
  97. Soilleux EJ (2003) DC-SIGN (dendritic cell-specific ICAM-grabbing non-integrin) and DC-SIGN-related (DC-SIGNR): friend or foe? Clin Sci (Lond) 104: 437-446.
  98. Bergeron A, El-Hage F, Kambouchner M, Lecossier D, Tazi A (2006) Characterisation of dendritic cell subsets in lung cancer micro-environments.Eur Respir J 28: 1170-1177.
  99. van Cruijsen H, Ruiz MG, van der Valk P, de Gruijl TD, Giaccone G (2009) Tissue micro array analysis of ganglioside N-glycolyl GM3 expression and signal transducer and activator of transcription (STAT)-3 activation in relation to dendritic cell infiltration and microvessel density in non-small cell lung cancer.BMC Cancer 9: 180.
  100. Perrot I, Blanchard D, Freymond N, Isaac S, Guibert B, et al. (2007) Dendritic cells infiltrating human non-small cell lung cancer are blocked at immature stage.J Immunol 178: 2763-2769.
  101. Inoshima N, Nakanishi Y, Minami T, Izumi M, Takayama K, et al. (2002) The influence of dendritic cell infiltration and vascular endothelial growth factor expression on the prognosis of non-small cell lung cancer.Clin Cancer Res 8: 3480-3486.
  102. Dieu-Nosjean MC, Antoine M, Danel C, Heudes D, Wislez M, et al. (2008) Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures.J Clin Oncol 26: 4410-4417.
  103. Shurin GV, Ma Y, Shurin MR (2013) Immunosuppressive mechanisms of regulatory dendritic cells in cancer. Cancer Microenviron 6: 159-167.
  104. Zhong H, Gutkin DW, Han B, Ma Y, Keskinov AA, et al. (2014) Origin and pharmacological modulation of tumor-associated regulatory dendritic cells.Int J Cancer 134: 2633-2645.
  105.  Mu CY, Huang JA, Chen Y, Chen C, Zhang XG (2011) High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor infiltrating dendritic cells maturation. Med onco 28: 682-688.
  106. Schneider T, Hoffmann H, Dienemann H, Schnabel PA, Enk AH, et al. (2011) Non-small cell lung cancer induces an immunosuppressive phenotype of dendritic cells in tumor microenvironment by upregulating B7-H3.J Thorac Oncol 6: 1162-1168.
  107. Gabrilovich DI, Nagaraj S (2009) Myeloid-derived suppressor cells as regulators of the immune system.Nat Rev Immunol 9: 162-174.
  108. Peranzoni E, Zilio S, Marigo I, Dolcetti L, Zanovello P, et al. (2010) Myeloid-derived suppressor cell heterogeneity and subset definition.Curr Opin Immunol 22: 238-244.
  109. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V (2012) Coordinated regulation of myeloid cells by tumours.Nat Rev Immunol 12: 253-268.
  110. Bunt SK, Sinha P, Clements VK, Leips J, Ostrand-Rosenberg S (2006) Inflammation induces myeloid-derived suppressor cells that facilitate tumor progression.J Immunol 176: 284-290.
  111. Yang L, DeBusk LM, Fukuda K, Fingleton B, Green-Jarvis B, et al. (2004) Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis.Cancer Cell 6: 409-421.
  112. Murdoch C, Muthana M, Coffelt SB, Lewis CE (2008) The role of myeloid cells in the promotion of tumour angiogenesis.Nat Rev Cancer 8: 618-631.
  113. Guedez L, Jensen-Taubman S, Bourboulia D, Kwityn CJ, Wei B, et al. (2012) TIMP-2 targets tumor-associated myeloid suppressor cells with effects in cancer immune dysfunction and angiogenesis.J Immunother 35: 502-512.
  114. Hoechst B, Voigtlaender T, Ormandy L, Gamrekelashvili J, Zhao F, et al. (2009) Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor.Hepatology 50: 799-807.
  115. Filipazzi P, Huber V, Rivoltini L (2012) Phenotype, function and clinical implications of myeloid-derived suppressor cells in cancer patients.Cancer Immunol Immunother 61: 255-263.
  116. Huang A, Zhang B, Wang B, Zhang F, Fan KX, et al. (2013) Increased CD14(+)HLA-DR (-/low) myeloid-derived suppressor cells correlate with extrathoracic metastasis and poor response to chemotherapy in non-small cell lung cancer patients.Cancer Immunol Immunother 62: 1439-1451.
  117. Liu CY, Wang YM, Wang CL, Feng PH, Ko HW, et al. (2010) Population alterations of L-arginase-and inducible nitric oxide synthase-expressed CD11b+/CD14-/CD15+/CD33+ myeloid-derived suppressor cells and CD8+ T lymphocytes in patients with advanced-stage non-small cell lung cancer.J Cancer Res Clin Oncol 136: 35-45.
  118. Heuvers ME, Muskens F, Bezemer K, Lambers M, Dingemans AM, et al. (2013) Arginase-1 mRNA expression correlates with myeloid-derived suppressor cell levels in peripheral blood of NSCLC patients.Lung Cancer 81: 468-474.
  119. Brandau S, Trellakis S, Bruderek K, Schmaltz D, Steller G, et al. (2011) Myeloid-derived suppressor cells in the peripheral blood of cancer patients contain a subset of immature neutrophils with impaired migratory properties.J Leukoc Biol 89: 311-317.
  120. Srivastava MK, Zhu L, Harris-White M, Kar UK, Huang M, et al. (2012) Myeloid suppressor cell depletion augments antitumor activity in lung cancer.PLoS One 7: e40677.
  121. Montero AJ, Diaz-Montero CM, Kyriakopoulos CE, Bronte V, Mandruzzato S (2012) Myeloid-derived suppressor cells in cancer patients: a clinical perspective.J Immunother 35: 107-115.
  122. Cooper MA, Fehniger TA, Caligiuri MA (2001) The biology of human natural killer-cell subsets.Trends Immunol 22: 633-640.
  123. Hanna J, Goldman-Wohl D, Hamani Y, Avraham I, Greenfield C, et al. (2006) Decidual NK cells regulate key developmental processes at the human fetal-maternal interface.Nat Med 12: 1065-1074.
  124. Hanna J, Mandelboim O (2007) When killers become helpers.Trends Immunol 28: 201-206.
  125. Kopcow HD, Allan DS, Chen X, Rybalov B, Andzelm MM, et al. (2005) Human decidual NK cells form immature activating synapses and are not cytotoxic.Proc Natl Acad Sci U S A 102: 15563-15568.
  126. Ashkar AA, Di Santo JP, Croy BA (2000) Interferon gamma contributes to initiation of uterine vascular modification, decidual integrity, and uterine natural killer cell maturation during normal murine pregnancy. J Exp Med 192: 259-70
  127. Mamessier E, Sylvain A, Thibult ML, Houvenaeghel G, Jacquemier J, et al. (2011) Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity.J Clin Invest 121: 3609-3622.
  128. Rocca YS, Roberti MP, Arriaga JM, Amat M, Bruno L, et al. (2013) Altered phenotype in peripheral blood and tumor-associated NK cells from colorectal cancer patients.Innate Immun 19: 76-85.
  129. Carrega P, Bonaccorsi I, Di Carlo E, Morandi B, Paul P, et al. (2014) CD56(bright)perforin(low) noncytotoxic human NK cells are abundant in both healthy and neoplastic solid tissues and recirculate to secondary lymphoid organs via afferent lymph.J Immunol 192: 3805-3815.
  130. Bruno A, Ferlazzo G, Albini A, Noonan DM1 (2014) A think tank of TINK/TANKs: tumor-infiltrating/tumor-associated natural killer cells in tumor progression and angiogenesis.J Natl Cancer Inst 106: dju200.
  131. Flavell RA, Sanjabi S, Wrzesinski SH, Licona-Limón P (2010) The polarization of immune cells in the tumour environment by TGFbeta.Nat Rev Immunol 10: 554-567.
  132. Trotta R, Dal Col J, Yu J, Ciarlariello D, Thomas B, et al. (2008) TGF-beta utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and antibody-dependent cellular cytotoxicity in human NK cells.J Immunol 181: 3784-3792.
  133. Burton GJ, Jauniaux E, Charnock-Jones DS (2010) The influence of the intrauterine environment on human placental development.Int J Dev Biol 54: 303-312.
  134. Semenza GL (2010) Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics.Oncogene 29: 625-634.
  135. Cerdeira AS, Rajakumar A, Royle CM, Lo A, Husain Z, et al. (2013) Conversion of peripheral blood NK cells to a decidual NK-like phenotype by a cocktail of defined factors.J Immunol 190: 3939-3948.
  136.  Blackburn MR, Gao X, Airhart MJ, Skalko RG, Thompson LF, et al. (1992) Adenosine levels in the postimplantation mouse uterus: quantitation by HPLC-fluorometric detection and spatiotemporal regulation by 5'-nucleotidase and adenosine deaminase. Dev Dyn 194: 155-168.
  137. Blay J, White TD, Hoskin DW (1997) The extracellular fluid of solid carcinomas contains immunosuppressive concentrations of adenosine.Cancer Res 57: 2602-2605.
  138. Antonioli L, Blandizzi C, Pacher P, Haskó G (2013) Immunity, inflammation and cancer: a leading role for adenosine.Nat Rev Cancer 13: 842-857.
  139. Baginska J, Viry E, Paggetti J, Medves S, Berchem G, et al. (2013) The critical role of the tumor microenvironment in shaping natural killer cell-mediated anti-tumor immunity.Front Immunol 4: 490.
  140. Mincheva-Nilsson L, Baranov V (2014) Cancer exosomes and NKG2D receptor-ligand interactions: impairing NKG2D-mediated cytotoxicity and anti-tumour immune surveillance.Semin Cancer Biol 28: 24-30.
  141. Liu C, Yu S, Zinn K, Wang J, Zhang L, et al. (2006) Murine mammary carcinoma exosomes promote tumor growth by suppression of NK cell function.J Immunol 176: 1375-1385.
  142. Buschow SI, van Balkom BW, Aalberts M, Heck AJ, Wauben M, et al. (2010) MHC class II-associated proteins in B-cell exosomes and potential functional implications for exosome biogenesis.Immunol Cell Biol 88: 851-856.
  143. Conti P, Pang X, Boucher W, Letourneau R, Reale M, et al. (1997) Impact of Rantes and MCP-1 chemokines on in vivo basophilic cell recruitment in rat skin injection model and their role in modifying the protein and mRNA levels for histidine decarboxylase.Blood 89: 4120-4127.
  144. Almholt K, Johnsen M (2003) Stromal cell involvement in cancer.Recent Results Cancer Res 162: 31-42.
  145. Douaiher J, Succar J, Lancerotto L, Gurish MF, Orgill DP, et al. (2014) Development of mast cells and importance of their tryptase and chymase serine proteases in inflammation and wound healing.Adv Immunol 122: 211-252.
  146. Ribatti D, Ranieri G (2015) Tryptase, a novel angiogenic factor stored in mast cell granules.Exp Cell Res 332: 157-162.
  147. Ullah E, Nagi AH, Ashraf M (2013) Angiogenesis and mast cell density as predictors of patient survival in squamous cell carcinoma of lung.J Cancer Res Ther 9: 701-705.
  148. Ullah E, Nagi AH, Lail RA (2012) Angiogenesis and mast cell density in invasive pulmonary adenocarcinoma.J Cancer Res Ther 8: 537-541.
  149. Stoyanov E, Uddin M, Mankuta D, Dubinett SM, Levi-Schaffer F (2012) Mast cells and histamine enhance the proliferation of non-small cell lung cancer cells.Lung Cancer 75: 38-44.
  150. Xiao H, Lässer C, Shelke GV, Wang J, Rådinger M, et al. (2014) Mast cell exosomes promote lung adenocarcinoma cell proliferation-role of KIT-stem cell factor signaling.Cell Commun Signal 12: 64.
  151. Imada A, Shijubo N, Kojima H, Abe S (2000) Mast cells correlate with angiogenesis and poor outcome in stage I lung adenocarcinoma.Eur Respir J 15: 1087-1093.
  152. Jeong HJ, Oh HA, Nam SY, Han NR, Kim YS, et al. (2013) The critical role of mast cell-derived hypoxia-inducible factor-1α in human and mice melanoma growth.Int J Cancer 132: 2492-2501.
  153. Takanami I, Takeuchi K, Naruke M (2000) Mast cell density is associated with angiogenesis and poor prognosis in pulmonary adenocarcinoma.Cancer 88: 2686-2692.
  154. Starkey JR, Crowle PK, Taubenberger S (1988) Mast-cell-deficient W/Wv mice exhibit a decreased rate of tumor angiogenesis.Int J Cancer 42: 48-52.
  155. Coussens LM, Raymond WW, Bergers G, Laig-Webster M, Behrendtsen O, et al. (1999) Inflammatory mast cells up-regulate angiogenesis during squamous epithelial carcinogenesis.Genes Dev 13: 1382-1397.
  156. Soucek L, Lawlor ER, Soto D, Shchors K, Swigart LB, et al. (2007) Mast cells are required for angiogenesis and macroscopic expansion of Myc-induced pancreatic islet tumors.Nat Med 13: 1211-1218.
  157. Bremnes RM, Dønnem T, Al-Saad S, Al-Shibli K, Andersen S, et al. (2011) The role of tumor stroma in cancer progression and prognosis: emphasis on carcinoma-associated fibroblasts and non-small cell lung cancer.J Thorac Oncol 6: 209-217.
  158. Hughes CC (2008) Endothelial-stromal interactions in angiogenesis.Curr Opin Hematol 15: 204-209.
  159. Koukourakis MI, Giatromanolaki A, Harris AL, Sivridis E (2006) Comparison of metabolic pathways between cancer cells and stromal cells in colorectal carcinomas: a metabolic survival role for tumor-associated stroma.Cancer Res 66: 632-637.
  160. Siegel PM, Massagué J (2003) Cytostatic and apoptotic actions of TGF-beta in homeostasis and cancer.Nat Rev Cancer 3: 807-821.
  161. Pietras K, Ostman A (2010) Hallmarks of cancer: interactions with the tumor stroma.Exp Cell Res 316: 1324-1331.
  162. Vicent S, Sayles LC, Vaka D, Khatri P, Gevaert O, et al. (2012) Cross-species functional analysis of cancer-associated fibroblasts identifies a critical role for CLCF1 and IL-6 in non-small cell lung cancer in vivo.Cancer Res 72: 5744-5756.
  163. Navab R, Strumpf D, Bandarchi B, Zhu CQ, Pintilie M, et al. (2011) Prognostic gene-expression signature of carcinoma-associated fibroblasts in non-small cell lung cancer.Proc Natl Acad Sci U S A 108: 7160-7165.
  164. Honda E, Yoshida K, Munakata H (2010) Transforming growth factor-beta upregulates the expression of integrin and related proteins in MRC-5 human myofibroblasts.Tohoku J Exp Med 220: 319-327.
  165. Lu N, Carracedo S, Ranta J, Heuchel R, Soininen R, et al. (2010) The human alpha11 integrin promoter drives fibroblast-restricted expression in vivo and is regulated by TGF-beta1 in a Smad-and Sp1-dependent manner. Matrix biol 29: 166-176.
  166. Margadant C, Sonnenberg A (2010) Integrin-TGF-beta crosstalk in fibrosis, cancer and wound healing.EMBO Rep 11: 97-105.
  167. Kim SH, Choe C, Shin YS, Jeon MJ, Choi SJ, et al. (2013) Human lung cancer-associated fibroblasts enhance motility of non-small cell lung cancer cells in co-culture.Anticancer Res 33: 2001-2009.
  168. Ksiazkiewicz M, Gottfried E, Kreutz M, Mack M, Hofstaedter F, et al. (2010) Importance of CCL2-CCR2A/2B signaling for monocyte migration into spheroids of breast cancer-derived fibroblasts.Immunobiology 215: 737-747.
  169. Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, et al. (2005) Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion.Cell 121: 335-348.
  170. Hellevik T, Pettersen I, Berg V, Bruun J, Bartnes K, et al. (2013) Changes in the Secretory Profile of NSCLC-Associated Fibroblasts after Ablative Radiotherapy: Potential Impact on Angiogenesis and Tumor Growth. Translational oncology 6: 66-74
  171. Low BC, Pan CQ, Shivashankar GV, Bershadsky A, Sudol M, et al. (2014) YAP/TAZ as mechanosensors and mechanotransducers in regulating organ size and tumor growth.FEBS Lett 588: 2663-2670.
  172. O'Connor JW, Gomez EW (2014) Biomechanics of TGFβ-induced epithelial-mesenchymal transition: implications for fibrosis and cancer.Clin Transl Med 3: 23.
  173. Özdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, et al. (2014) Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival.Cancer Cell 25: 719-734.
  174. Tao H, Mimura Y, Aoe K, Kobayashi S, Yamamoto H, et al. (2012) Prognostic potential of FOXP3 expression in non-small cell lung cancer cells combined with tumor-infiltrating regulatory T cells.Lung Cancer 75: 95-101.
  175. Esendagli G, Bruderek K, Goldmann T, Busche A, Branscheid D, et al. (2008) Malignant and non-malignant lung tissue areas are differentially populated by natural killer cells and regulatory T cells in non-small cell lung cancer.Lung Cancer 59: 32-40.
  176. Erfani N, Mehrabadi SM, Ghayumi MA, Haghshenas MR, Mojtahedi Z, et al. (2012) Increase of regulatory T cells in metastatic stage and CTLA-4 over expression in lymphocytes of patients with non-small cell lung cancer (NSCLC).Lung Cancer 77: 306-311.
  177. Wang WJ, Tao Z, Gu W, Sun LH (2013) Variation of blood T lymphocyte subgroups in patients with non-small cell lung cancer.Asian Pac J Cancer Prev 14: 4671-4673.
  178. Black CC, Turk MJ, Dragnev K, Rigas JR (2013) Adenocarcinoma contains more immune tolerance regulatory t-cell lymphocytes (versus squamous carcinoma) in non-small-cell lung cancer.Lung 191: 265-270
  179. Petersen RP, Campa MJ, Sperlazza J, Conlon D, Joshi MB, et al. (2006) Tumor infiltrating Foxp3+ regulatory T-cells are associated with recurrence in pathologic stage I NSCLC patients.Cancer 107: 2866-2872
  180. Shimizu K, Nakata M, Hirami Y, Yukawa T, Maeda A, et al. (2010) Tumor-infiltrating Foxp3+ regulatory T cells are correlated with cyclooxygenase-2 expression and are associated with recurrence in resected non-small cell lung cancer.J Thorac Oncol 5: 585-590
  181. Roos-Engstrand E, Ekstrand-Hammarström B, Pourazar J, Behndig AF, Bucht A, et al. (2009) Influence of smoking cessation on airway T lymphocyte subsets in COPD.COPD 6: 112-120.
  182. Hanagiri T, Shigematsu Y, Shinohara S, Takenaka M, Oka S, et al. (2013) Clinical significance of the frequency of regulatory T cells in regional lymph node lymphocytes as a prognostic factor for non-small-cell lung cancer.Lung Cancer 81: 475-479.
  183. Hasegawa T, Suzuki H, Yamaura T, Muto S, Okabe N, et al. (2014) Prognostic value of peripheral and local forkhead box P3+ regulatory T cells in patients with non-small-cell lung cancer.Mol Clin Oncol 2: 685-694.
  184. Dimitrakopoulos FI, Papadaki H, Antonacopoulou AG, Kottorou A, Gotsis AD, et al. (2011) Association of FOXP3 expression with non-small cell lung cancer.Anticancer Res 31: 1677-1683.
  185. Fu HY, Li C, Yang W, Gai XD, Jia T, et al. (2013) FOXP3 and TLR4 protein expression are correlated in non-small cell lung cancer: implications for tumor progression and escape.Acta Histochem 115: 151-157.
  186. Gao X, Zhu Y, Li G, Huang H, Zhang G, et al. (2012) TIM-3 expression characterizes regulatory T cells in tumor tissues and is associated with lung cancer progression.PLoS One 7: e30676.
  187. Shevach EM (2009) Mechanisms of foxp3+ T regulatory cell-mediated suppression.Immunity 30: 636-645.
  188. Takizawa H, Tanaka M, Takami K, Ohtoshi T, Ito K, et al. (2001) Increased expression of transforming growth factor-beta1 in small airway epithelium from tobacco smokers and patients with chronic obstructive pulmonary disease (COPD).Am J Respir Crit Care Med 163: 1476-1483.
  189. Zhao L, Yang J, Wang HP, Liu RY (2013) Imbalance in the Th17/Treg and cytokine environment in peripheral blood of patients with adenocarcinoma and squamous cell carcinoma.Med Oncol 30: 461.
  190. Sharma S, Yang SC, Zhu L, Reckamp K, Gardner B, et al. (2005) Tumor cyclooxygenase-2/prostaglandin E2-dependent promotion of FOXP3 expression and CD4+ CD25+ T regulatory cell activities in lung cancer.Cancer Res 65: 5211-5220.
  191. Anumanthan G, Halder SK, Osada H, Takahashi T, Massion PP, et al. (2005) Restoration of TGF-beta signalling reduces tumorigenicity in human lung cancer cells.Br J Cancer 93: 1157-1167.
  192. Baratelli F, Lee JM, Hazra S, Lin Y, Walser TC, et al. (2010) PGE(2) contributes to TGF-beta induced T regulatory cell function in human non-small cell lung cancer.Am J Transl Res 2: 356-367.
  193. Ye L, Li X, Sun S, Guan S, Wang M, et al. (2013) A study of circulating anti-CD25 antibodies in non-small cell lung cancer.Clin Transl Oncol 15: 633-637.
  194. Schwartz M (1961) A biomathematical approach to clinical tumor growth.Cancer 14: 1272-1294.
  195. Dominioni L, Rotolo N, Mantovani W, Poli A, Pisani S, et al. (2012) A population-based cohort study of chest x-ray screening in smokers: lung cancer detection findings and follow-up.BMC Cancer 12: 18.
Citation: Mortara L, Zanellato S, Bassani B, Imperatori A, Rotolo N, et al. (2015) Polarization of Tumor Infiltrating Leukocytes from Innate Immunity and their role in the Pro-angiogenic Phenotype in NSCLC. J Clin Cell Immunol 6:312.

Copyright: © 2015 Mortara L, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top