Differential Macrophage Programming in the Tumor Microenvironment

8
Differential macrophage programming in the tumor microenvironment Brian Ruffell 1 , Nesrine I. Affara 1 and Lisa M. Coussens 1, 2* 1 Department of Pathology, University of California, San Francisco, 513 Parnassus Ave, HSW450C, San Francisco, CA 94143, USA 2 Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, 513 Parnassus Ave, HSW450C, San Francisco, CA 94143, USA Of the multiple unique stromal cell types common to solid tumors, tumor-associated macrophages (TAMs) are significant for fostering tumor progression. The pro- tumor properties of TAMs derive from regulation of angiogenic programming, production of soluble media- tors that support proliferation, survival and invasion of malignant cells, and direct and indirect suppression of cytotoxic T cell activity. These varied activities are de- pendent on the polarization state of TAMs that is regu- lated in part by local concentrations of cytokines and chemokines, as well as varied interactions of TAMs with normal and degraded components of the extracellular matrix. Targeting molecular pathways regulating TAM polarization holds great promise for anticancer therapy. Macrophages in solid malignancies Macrophages are important residents of all tissues, where they play crucial roles in regulating tissue homeostasis. In this context, tissue-resident macrophages assist with com- bating infection [1,2], resolving acute inflammation [3], and in regulating metabolic response to tissue stress [4,5]. This broad range of functions, and the accompanying plasticity required to permit such adaptive responses, also implicates macrophages in several chronic pathological conditions including diabetes and atherosclerosis [57]. Solid tumors represent an extreme example of a dysregu- lated tissue, and multiple characteristics of tumors, includ- ing hypoxia [8] and abundant cell death [9], help direct macrophage function towards attempting homeostatic res- toration. In the context of a tumor however, this represents a maladaptive response that instead helps drive tumor growth through promotion of angiogenic programs, tissue remodeling, ectopic survival of malignant cells, and devel- opment of immunosuppressive microenvironments that blunt cytotoxic T cell activities [10]. More recently, it has been demonstrated that polarization of macrophages towards tumor-promoting phenotypes is not exclusively the result of thwarted tissue homeostasis, but instead a more active process driven by what are probably reciprocal interactions with both malignant and stromal cells in the local microenvironment [10,11]. Thus, in addition to dis- cussing well-accepted functions for tumor-associated macrophages (TAMs; Box 1), this review also focuses on recently recognized molecular and cellular mechanisms underlying TAM polarization within tumor microenviron- ments, and the therapeutic implications of these findings. TAM function With the exception of non-small cell lung carcinoma [12,13], patient prognosis in solid tumors is generally described as correlating inversely with TAM density and expression signatures [10,14]. TAMs have also been relat- ed to particular functional roles in human tumors, with an established association between TAM presence and densi- ty of tumor vasculature in several carcinomas [1518] including breast [1924], as well as increased local inva- sion and/or metastasis in melanoma [25], breast [26,27], ovarian [15,28], colorectal [29,30], pancreatic neuroendo- crine [31] and bladder [32] cancer. Less established in humans is a role for TAMs in local immune suppression, although there have been several reports in ovarian cancer [3335]. A potential caveat of these studies is that most rely on immunohistochemical detection of CD68 for measure- ment of TAM density. However, CD68 is not a specific marker of macrophages and is expressed by other stromal populations (Box 1), including those that may have over- lapping function with TAMs (Box 2). Angiogenesis Experimental studies using murine carcinoma models have clearly demonstrated that TAMs regulate vascular pro- gramming of tumors. Important for this activity is TAM production of vascular endothelial growth factor A (VEGFA), because overexpression of VEGFA partially reverses the effects of macrophage depletion [36], and macrophage-spe- cific loss of VEGFA results in vascular normalization [37]. In some tumor models, production of matrix metalloproteinase (MMP)-9 by TAMs mediates VEGFA bioavailability, thus providing an alternative, but still VEGF-dependent route for promoting angiogenesis [38,39]. Similarly, the TAM produc- tion of placental growth factor (PIGF), a homolog of VEGFA that selectively binds VEGF receptor 1 (VEGFR1), also stimulates angiogenesis in tumors [40] and may in part explain resistance to VEGFA and VEGFR targeted thera- pies in the clinic [4143]. Several other factors produced by macrophages are also known to regulate vascular program- ming [4446], but their roles in tumorigenesis are not firmly established and therefore are not discussed herein. Recently a novel subset of macrophages expressing Tie2, a receptor for angiopoeitins (ANGs) was described Review Corresponding author: Coussens, L.M. ([email protected]) * Current address: Department of Cell and Developmental Biology, Knight Cancer Institute, Oregon Health and Sciences University, Rm 5508, Richard Jones Hall, 3181 SW Sam Jackson Rd, Portland OR 97239-3098, USA. 1471-4906/$ see front matter ß 2012 Elsevier Ltd. All rights reserved. doi:10.1016/j.it.2011.12.001 Trends in Immunology, March 2012, Vol. 33, No. 3 119

Transcript of Differential Macrophage Programming in the Tumor Microenvironment

  • heM

    co,of

    Reviewa maladaptive response that instead helps drive tumorgrowth through promotion of angiogenic programs, tissueremodeling, ectopic survival of malignant cells, and devel-opment of immunosuppressive microenvironments thatblunt cytotoxic T cell activities [10]. More recently, ithas been demonstrated that polarization of macrophagestowards tumor-promoting phenotypes is not exclusivelythe result of thwarted tissue homeostasis, but instead amore active process driven by what are probably reciprocalinteractions with both malignant and stromal cells in thelocal microenvironment [10,11]. Thus, in addition to dis-cussing well-accepted functions for tumor-associatedmacrophages (TAMs; Box 1), this review also focuses on

    productionofvascular endothelialgrowth factor A (VEGFA),because overexpression of VEGFA partially reverses theeffects of macrophage depletion [36], and macrophage-spe-cific loss of VEGFA results in vascular normalization [37]. Insome tumor models, production of matrix metalloproteinase(MMP)-9 by TAMs mediates VEGFA bioavailability, thusprovidingan alternative, but stillVEGF-dependentroute forpromoting angiogenesis [38,39]. Similarly, the TAM produc-tion of placental growth factor (PIGF), a homolog of VEGFAthat selectively binds VEGF receptor 1 (VEGFR1), alsostimulates angiogenesis in tumors [40] and may in partexplain resistance to VEGFA and VEGFR targeted thera-pies in the clinic [4143]. Several other factors produced bymacrophages are also known to regulate vascular program-ming [4446], but their roles in tumorigenesis are not firmlyestablished and therefore are not discussed herein.

    Recently a novel subset of macrophages expressingTie2, a receptor for angiopoeitins (ANGs) was described

    Corresponding author: Coussens, L.M. ([email protected])* Current address: Department of Cell and Developmental Biology, Knight Cancer

    Institute, Oregon Health and Sciences University, Rm 5508, Richard Jones Hall, 3181SW Sam Jackson Rd, Portland OR 97239-3098, USA.Differential macropin the tumor microBrian Ruffell1, Nesrine I. Affara1 and Lisa 1Department of Pathology, University of California, San Francis2Helen Diller Family Comprehensive Cancer Center, University San Francisco, CA 94143, USA

    Of the multiple unique stromal cell types common tosolid tumors, tumor-associated macrophages (TAMs)are significant for fostering tumor progression. The pro-tumor properties of TAMs derive from regulation ofangiogenic programming, production of soluble media-tors that support proliferation, survival and invasion ofmalignant cells, and direct and indirect suppression ofcytotoxic T cell activity. These varied activities are de-pendent on the polarization state of TAMs that is regu-lated in part by local concentrations of cytokines andchemokines, as well as varied interactions of TAMs withnormal and degraded components of the extracellularmatrix. Targeting molecular pathways regulating TAMpolarization holds great promise for anticancer therapy.

    Macrophages in solid malignanciesMacrophages are important residents of all tissues, wherethey play crucial roles in regulating tissue homeostasis. Inthis context, tissue-resident macrophages assist with com-bating infection [1,2], resolving acute inflammation [3],and in regulating metabolic response to tissue stress[4,5]. This broad range of functions, and the accompanyingplasticity required to permit such adaptive responses, alsoimplicates macrophages in several chronic pathologicalconditions including diabetes and atherosclerosis [57].Solid tumors represent an extreme example of a dysregu-lated tissue, and multiple characteristics of tumors, includ-ing hypoxia [8] and abundant cell death [9], help directmacrophage function towards attempting homeostatic res-toration. In the context of a tumor however, this represents1471-4906/$ see front matter 2012 Elsevier Ltd. All rights reserved. doi:10.1016/j.it.2011.12age programmingnvironment. Coussens1,2*

    513 Parnassus Ave, HSW450C, San Francisco, CA 94143, USACalifornia, San Francisco, 513 Parnassus Ave, HSW450C,

    recently recognized molecular and cellular mechanismsunderlying TAM polarization within tumor microenviron-ments, and the therapeutic implications of these findings.

    TAM functionWith the exception of non-small cell lung carcinoma[12,13], patient prognosis in solid tumors is generallydescribed as correlating inversely with TAM density andexpression signatures [10,14]. TAMs have also been relat-ed to particular functional roles in human tumors, with anestablished association between TAM presence and densi-ty of tumor vasculature in several carcinomas [1518]including breast [1924], as well as increased local inva-sion and/or metastasis in melanoma [25], breast [26,27],ovarian [15,28], colorectal [29,30], pancreatic neuroendo-crine [31] and bladder [32] cancer. Less established inhumans is a role for TAMs in local immune suppression,although there have been several reports in ovarian cancer[3335]. A potential caveat of these studies is that most relyon immunohistochemical detection of CD68 for measure-ment of TAM density. However, CD68 is not a specificmarker of macrophages and is expressed by other stromalpopulations (Box 1), including those that may have over-lapping function with TAMs (Box 2).

    AngiogenesisExperimental studies using murine carcinoma models haveclearly demonstrated that TAMs regulate vascular pro-gramming of tumors. Important for this activity is TAM.001 Trends in Immunology, March 2012, Vol. 33, No. 3 119

  • Box 1. Identity crisis

    As their name implies, TAMs are found within or proximal toprimary tumors, and represent a mature population of terminallydifferentiated myeloid-lineage cells [55]. This location distinguishesthem from metastasis-associated macrophages [128], and they arephenotypically distinct from the heterogeneous population ofimmature myeloid cells that predominantly accumulate in theperiphery of tumor-bearing individuals, and are associated withimmune suppression [80]. Identifying TAMs can be difficult how-ever, as there are no lineage-defining markers for macrophages[128], and marker expression can vary by activation status andtissue localization [129]. In general, both human and mouse TAMscan be identified via flow cytometry through high surface expres-sion of CD11b, CD14 and MHCII/HLA-DR, in addition to the commonleukocyte antigen CD45. High expression of MHCII differentiates

    ReviewTAMs from immature myeloid cells, as does low expression of Ly6Cin mice and CD34 in humans [128]. Murine TAMs are also commonlyidentified by expression of F4/80, an EGF-transmembrane 7 familymolecule of unknown function. However, not all macrophagepopulations express F4/80, and it has been observed on Langerhanscells in the skin and on eosinophils in adipose tissue. Dendritic cellsalso express MHCII, and subsets express CD11b and CD14, whereasthe most commonly used marker for dendritic cells, CD11c, isexpressed constitutively by certain tissue macrophages and inducedby inflammatory conditions such as those found in the tumormicroenvironment. The problem of accurately identifying TAMs ismore acute in humans because studies rely almost exclusively onsingle maker detection of CD68 via immunohistochemistry. Inaddition to other leukocyte populations, CD68 is expressed byfibroblasts, and at least for breast cancer is not a specific marker forTAMs [99]. Thus, although human studies are referenced here, thefunctions ascribed to TAMs based on correlations between TAMdensity and clinical parameters require validation in some tissues.[47]. Tie2+ TAMs tend to be closely associated withtumor vasculature, and have been found to be crucialfor angiogenesis in orthotopic [47] and transgenic tumormodels [48]. This activity depends in part on endothelialcell-produced ANG2 and the Tie2 receptor that directlocalization of Tie-2-expressing cells along the vasculature[48]. Notably, antagonists that block ANG2 restrict tumorgrowth in mouse models of de novo mammary carcinogen-esis, that is, MMTV-PyMT mice [49], a property thathas thus far not been observed in tumors following block-ade of TAM infiltration [50], or by myeloid-specificVegfa ablation [37], thus hinting at a unique mode ofaction either downstream or parallel to VEGFA bioactivi-ty [48].

    Box 2. Tumor commune

    Although TAMs may constitute the majority of immune cells insome tumors, additional myeloid and mesenchyme-derived cellspervade tumors and are known to overlap functionally with TAMs.This includes granulocytic mast cells, neutrophils and immaturemyeloid cells that can be angiogenic, immunosuppressive, andpromote metastasis through pathways akin to those employed byTAMs [128]. Cancer-associated fibroblasts have also been shown topromote vascularization through SDF-1 [130] and IL-1b [131],whereas mesenchymal stem cells promote metastasis throughCCL5 [132] and are potentially immunosuppressive [133]. Mono-cyte-derived cells with a fibroblast morphology, or fibrocytes, areincreased during chronic inflammation, and based upon their dualmacrophage/fibroblast phenotype [70] are probably an emergingcell population involved in tumorigenesis.

    120Invasion and metastasisThe most comprehensively described mechanism throughwhich TAMs promote solid tumor development is by pro-viding factors that enhance invasion of malignant cells intoectopic tissue. This activity centers around a paracrineinteraction loop involving macrophage-expressed epider-mal growth factor (EGF) and epithelial cell-expressedcolony-stimulating factor (CSF)1, also known as macro-phage CSF [51]. Increased expression of CSF1 is a signifi-cant mechanism underlying macrophage recruitment intotissues [50,5254] after CSF1 binding to its high affinityreceptor (CSF1R) expressed on resident macrophages andsome macrophage precursors. This interaction promotesmacrophage proliferation, survival and tissue recruitmentduring development (e.g. branching morphogenesis in themammary gland) [55], homeostasis [56], and pathologicaltissue remodeling processes such as those associated withacute tissue injury [53,5759] and during solid tumordevelopment [50]. Chemokines such as CC ligand(CCL)2 and stromal-derived factor-1 (SDF-1; CXCL12)are also important for TAM recruitment with demonstrat-ed roles in models of glioblastoma, melanoma, and cervicaland prostate cancer [38,39,6065]. Crucially, in addition tosupporting TAM recruitment, activation of intracellularsignaling pathways downstream of the CSF1CSF1R in-teraction significantly enhances EGF expression, which inturn regulates epithelial cell migration in some tissues[51,6668].

    Macrophages also regulate composition and structure ofextracellular matrix (ECM) through their deposition ofECM components, for example, various types of collagens,and breakdown of these same components via their releaseof MMPs, serine proteases and cathepsins [6971]. Migra-tion on and through ECM is a necessary aspect of cellmigration [72], and thus by extension, TAMs are thought toregulate tumor and stromal cell migration/invasionthrough ECM. This activity has been directly demonstrat-ed in vitro [73,74] and in vivo with mice genetically defi-cient for cathepsin B, cathepsin S and urokinase/plasminogen activator (uPA), all of which derive primarilyfrom TAMs in the tumor microenvironment [7577]. MMP-dependent cleavage of the ECM [69], collagen deposition[78], and alteration of collagen structure [79] are alsopotential sources of regulation mediated by TAMs, butformal demonstration of these is lacking.

    Immune suppressionSolid tumors are well recognized for repressing the activityof cytotoxic T cells, and are thus characterized grossly asimmunosuppressive. Although regulatory T (Treg) cellshave garnered much attention owing to this capability,recent literature also recognizes macrophages as majordeterminants of immune suppression in solid tumors. Al-though the mechanisms underlying these activities are lesswell characterized as compared to myeloid-derived suppres-sor cells, for example [80], TAMs do typically express severalgenes with immunosuppressive potential [68,81,82], andare capable of directly limiting T cell proliferation during

    Trends in Immunology March 2012, Vol. 33, No. 3classic in vitro suppressive assays [8,34,50,83]. TAMs alsoact through intermediates to regulate immune suppression,as has been reported for recruitment of Treg cells by CCL22

  • [33], and may be important mediators of T cell recruitmentbased upon the inverse correlation between the presence ofCD68+ TAMs and CD8+ T cells in human breast cancer, andthe enhanced CD8+ T cell infiltration observed during che-motherapy in the MMTV-PyMT model following blockade ofthe CSF1CSF1R pathway [50].

    In murine tumor models, suppression of CD8+ T cellproliferation by TAMs is at least partly dependent on me-tabolism of L-arginine via arginase-1 or inducible nitricoxide synthase (iNOS) [8,83], and resulting production ofoxygen radicals or nitrogen species [84,85]. By contrast,suppression of CD8+ T cells by human TAMs can occurindependent of L-arginine metabolism [34], and insteadmay rely on the B7 family of molecules as has been describedfor B7-H1 (programmed cell death 1 ligand 1; PD-L1) inhepatocellular carcinoma [86], and B7-H4 in ovarian cancer[34]. This distinction may be a result of differences between

    human and murine macrophages, as expression of Arginase-1 by the former does not correlate with macrophage polari-zation. Alternatively, the tissue-specific microenvironmentcould dictate the mechanism of TAM suppression. As anexample of this, it has been described that ovarian TAMs areexposed to minimal levels of interleukin (IL)-4, which inother circumstances may downregulate B7-H4 expression[34]. TAMs from murine mammary and human breastcarcinomas however are exposed to significantly higheramounts of IL-4/13 [68,87], and thus one would anticipateminimal expression of B7-H4 in TAMs from these tissues.

    Mechanisms of TAM polarizationThe description of macrophage activation as either classi-cal [M1; interferon (IFN)g/lipopolysaccharide (LPS)-de-pendent] or alternative (M2; IL-4/IL-13/IL-10/FcgR-dependent) has provided a necessary framework for the

    MacrophageEndothelial cell

    HypoxiaNucleus

    oph

    Hypoxic Invasive

    eliapoxcleu

    F4/80LectinHypoxiaDAPI

    ayed

    Review Trends in Immunology March 2012, Vol. 33, No. 3Macr

    Perivascular

    EndothHyNu

    Figure 1. TAM localization within unique tumor microenvironments. Centrally displa late-stage tumor from the MMTV-PyMT mouse model of mammary carcinogenesis.

    vasculature as revealed by perfusion with tomato lectin (green) and DAPI nuclear stain (

    TAMs within a hypoxic region, at a locally invasive edge, in a normoxic area within thage

    Intratumoral

    l cellias

    TRENDS in Immunology

    is an immunofluorescent confocal micrograph of F4/80+ macrophages (red) withinAlso shown are areas of hypoxia detected with pimonidazole (yellow), functional

    blue). Clockwise from top left, insets display enlarged graphical representations of

    e tumor, and associated with the vasculature.

    121

  • understanding of TAM polarization [88]. However, eventhough the M1/M2 designations represent extreme ends ofa scale, the concept is an oversimplification of the diversityof TAM phenotypes evident simply based on their locali-zation within tumors (Figure 1). TAMs do not becomepolarized by virtue of their location per se, but insteadreceive signals from the particular microenvironment inwhich they reside (Figure 2). These heterotypic signalsmay overlap, for example, IL-4 promotes both Egf andArginase-1 expression by TAMs [68], whereas other signalsmay be unique to particular microenvironments, such ashypoxic zones within solid tumors that also induce Argi-nase-1 expression [8], but which are not found along vas-culature where invasive macrophages are localized. Fromthese two examples, all murine TAMs would be anticipatedto express higher levels of arginase-1 than normal tissuemacrophages, but the suppressive capabilities of intratu-moral TAMs in hypoxic regions would be even further

    enhanced. Thus, in addition to differentiation of TAMs,from either Tie2+ [89] or other monocyte precursors [83]that may preferentially home to specific localizations, onecan envision that integration of these distinct signalsresult in production of an array of TAM populations/phe-notypes with unique tumor-regulating properties [10]. It isimportant to note that the composition of the immunemicroenvironment [68,90,91] and the overall polarizationstate of TAMs becomes more favorable towards growthduring tumor progression [92]. The functional role ofmacrophages during tumor initiation [93,94] thereforediffers from that during tumor progression.

    Immune microenvironmentConsistent with the original description of alternativeactivation, the type 2 cytokine IL-4 [68,77], and immuno-globulin (Ig) signaling through activating types of Fcgreceptors [91], exert significant regulation on TAMs in

    (d)

    Treg

    Treg

    CTLTh2/NKT B cell

    IL-10

    CSF1

    IL4R

    CCL2

    CCL22

    ROS

    ECMdegradation

    Proteases

    B7-H

    1

    L-ar

    ginine

    EGFTAM

    FcRiNOSArg-1

    ie2

    EP2/4CCR/2CSF1R

    IL-10R

    C

    PD-1

    COX-2 PGE

    IL-4/IL-13IL-10

    Igs

    Imm

    un

    em

    icro

    enviro

    nm

    ent

    Imm

    un

    esuppression

    Invasion

    Tum

    or

    deriv

    ed

    (e)

    (a)

    (b)

    Neoplastic cells

    omo

    sulti

    at re

    ) Sig

    mun

    ct s

    Review Trends in Immunology March 2012, Vol. 33, No. 3ECMdegradation

    Hif1a

    TLRs THypoxia

    HMGB1

    2

    Hom

    eost

    atic

    imba

    lanc

    e

    (c)

    Neoplastic cells

    Figure 2. TAMs as central regulators of the tumor microenvironment. Factors that pr

    into those derived from the immune system, actively produced by tumor cells, or re

    factors such as immune complexes. (b) Neoplastic cells can produce chemokines th

    well as directly producing immunosuppressive molecules such as IL-10 and PGE2. (c

    cell death. These signals all direct the protumor functions of TAMs (df) including im

    suppression can occur through soluble or cell surface mediators, and may be indireectopic tissue can be promoted through directed release of cytokines such as EGF, or

    migration or increase chemoattractant bioavailability. (f) In addition to the interplay of

    factors, subsets of TAMs expressing the Tie2 receptor interact with mural cells/pericyte

    122PIGF

    VEGFA

    ANG2Muralcells

    Angiog

    enesis

    (f)

    TRENDS in Immunology

    te the polarization of TAMs towards a protumor phenotype (ac) can be subdivided

    ng from tissue stress. (a) From leukocytes, this includes cytokines and other soluble

    cruit macrophages, including CSF1 and CCL2, depending on the tissue involved, as

    ns of dysregulated tissues include leaky vasculature, hypoxia, ECM remodeling and

    e suppression, tumor cell dissemination, and promoting angiogenesis. (d) Immune

    uch as through the recruitment of regulatory T cells. (e) Neoplastic cell invasion of through protease-dependent ECM remodeling that may directly affect neoplastic

    TAMs with endothelial cells through production of VEGFA and other angiogenic

    s to regulate vascular structure.

  • their ability to direct TAM protumor phenotype. IL-13derived from malignant epithelial cells or TH2-polarizedCD4+ T cells [87], or CD1d-restricted natural killer T(NKT) cells [95], may have similar effects on TAM polariza-tion due to overlapping IL-13 and IL-4 signaling cascadesthat lead to signal transducer and activator of transcription(STAT)6 activation, but this has yet to be proven in vivo.Likewise, activation of STAT3 by IL-10 suppresses IL-12[96] and tumor necrosis factor-a (TNF-a) expression [97] byTAMs, but the source of IL-10 and the relevance of thispathway to tumor development is unclear. Intriguingly,although IL-4 from TH2-polarized CD4

    + T cells is necessaryfor TAM programming in the MMTV-PyMT mammarycarcinoma model for EGF expression and promotion ofmetastasis, the absence of Igs in B-cell-deficient mice doesnot affect tumorigenesis [68]. Conversely, autoantibodies inthe K14-HPV16 mouse model of squamous carcinogenesisdrive TAM-dependent angiogenesis by FcRg-dependentmechanisms [91], whereas animals deficient in CD4+ T cellshave only mildly reduced tumor incidence [98]. These para-doxical findings probably indicate tissue-specific dependen-cies in each respective tumor model, and argue for detailedanalysis of counterpart human tissue as a precursor toclinical translation because human tumors could vary con-siderably in leukocyte composition.

    Such human/mouse differences have been described inbreast/mammary cancer. Notably, leukocyte composition inmurine mammary carcinomas is dominated by TAMs,whereas lymphocytic infiltrate, predominately CD4+T cells,comprises the majority of immune cells in human breastcarcinomas [68,99]. CD4+T cells from human breast cancersexpress high levels of IFNg, resulting in protein concentra-tions >10-fold higher than IL-4 or IL-13 in human tumors[87]. This suggests divergent cytokine milieus betweenhuman and mouse tissue, because tumor CD4+ T cells fromMMTV-PyMT-derived mammary tumors exhibit high ex-pression of IL-4, IL-13 and IL-10, but produce minimal IFNg[68]. Given the strong polarizing effects of these cytokines,and the potential for synergistic effects on macrophagephenotype [100], transcriptional profiling of purified humanTAMs should prove informative in relating functional datain mouse models to correlative studies in patients.

    Tumor-derived mediatorsIn addition to its potential production by multiple leuko-cyte subsets, IL-10 is secreted in vitro by many humancarcinoma cell lines [101], which in some instances actuallyreflects its origin in vivo [102]. Well established as a broadimmunosuppressive molecule, in vitro administration ofIL-10 to macrophages inhibits production of proinflamma-tory cytokines and chemokines [103], and reduces surfaceexpression of major histocompatibility complex (MHC)II,and the co-stimulatory molecules CD80 and CD86 [104].IL-10 also synergizes with IL-4 to induce Arginase-1 ex-pression in macrophages, possibly through induction of IL-4 receptor a [103]. As mentioned however, the source of IL-10 in tumor microenvironments is unclear, and may evenderive from TAMs themselves [96,97,105]. Interestingly,

    Reviewthe ability of TAMs to produce IL-10 has been associatedwith another molecule produced by tumor cells, prosta-glandin E2 (PGE2), suggesting that this may also regulateTAM polarization [97,106] through prostaglandin (EP) 2and EP4 receptors [107]. The importance of PGE2 in canceris inferred from the preventative effects of cyclooxygenase-2 (COX-2) inhibitors including aspirin in colon [108,109]and respective transgenic mouse models [108], as well as aless prominent effect in several other cancers [110]. In theAPCMin/+ mouse model of intestinal tumorigenesis, COX-2inhibition reduces expression of Arginase-1 and increasesthat of CXCL1 [111]; both changes that are also associatedwith repolarization of TAMs. Whether this repolarizationis important for therapeutic efficacy of COX-2 inhibitors isunknown however, because PGE2 has pleiotropic effects onmultiple aspects of tumor development [108].

    Homeostatic imbalanceHypoxic conditions exist within solid tumors in areasdistal from functional vasculature (Figure 1). Althoughnutrient deprivation is the goal of antiangiogenic therapy,the resulting hypoxia actually seems to promote malig-nant conversion and metastasis [112]. This response tooxygen availability is mediated primarily through hypox-ia-inducible factor (HIF)-1a and HIF-2a; both of whichalso regulate macrophage function [113]. Using LysM-cremice to induce myeloid-specific loss of either HIF-1a orHIF-2a, two recent reports have established that theprotumor functions of TAMs are likewise dependent onHIFs [8,114]. Loss of HIF-1a limited Arginase-1 expres-sion and the suppressive capabilities of TAMs in theMMTV-PyMT model [8], whereas loss of HIF-2a reducedTAM recruitment through lower chemokine receptor ex-pression in models of inflammatory hepatocellular andcolon carcinoma [114]. Despite the divergent mechanismsimplicated in each study, both observed reduced tumorvolume and progression, suggesting either tissue-specificroles for TAMs and/or involvement of overlapping path-ways regulated by HIF-1a or HIF-2a. This includes thepossible induction of an angiogenic response that was notthoroughly evaluated in either study, but which has beenassociated with TAMs localized to hypoxic regions oftumors [83,115]

    The mislocalization of cellular and extracellular compo-nents is another prominent feature of tumors due to celldeath and dysregulated tissue architecture, respectively.The presence of extracellular ATP, high-mobility group box1 protein (HMGB1), and other normally intracellular mole-cules is detected by a class of receptors on the surface ofmacrophages called Toll-like receptors (TLRs) [6]. Al-though TLR signaling in dendritic cells is important forgenerating an adaptive immune response following cyto-toxic therapies through TLR4 recognition of HMGB1 [116],both TLR4 and TLR2 signaling promote growth of cell linesin the lung through induced TNF-a production by macro-phages [117,118]. For TLR2, this can be mediated bytumor-derived versican [118], but other ECM componentsincluding biglycan and hyaluronan also induce proinflam-matory cytokine expression by macrophages via TLR2 andTLR4 [119], and are potentially important in dictatingTAM polarization. Crucially, these ECM components do

    Trends in Immunology March 2012, Vol. 33, No. 3not bind TLRs in their native form in noninflamed tissue,but become TLR ligands following degradation by proteasecleavage or interaction with reactive oxygen or nitrogen

    123

  • species, thereby forming putative sensory pathways fordetection of inflammation and tissue disruption.

    Concluding remarks: targets for therapyThe pathways that engage and mediate the maladaptiveresponse of TAMs present attractive therapeutic targets;several of which have already shown promise in the pre-clinical arena, and improve therapeutic responses to che-motherapy [37,50,120]. Therapeutic strategies directed atTAMs can be grouped crudely into four prospective themes:blocking effector function, limiting recruitment, repro-gramming, or preventing protumor polarization. Monoclo-nal antibodies and small molecular inhibitors targeting theVEGF and EGF pathways are already approved for treat-ment of various carcinomas alone or in combination, al-though none were designed specifically to target TAMfunction, and their clinical efficacy has been mixed[121,122]. It has recently been established that blockingTAM recruitment and/or survival in solids tumors (inmurine models) improves efficacy of cytotoxic therapies[50,123], in a manner dependent upon CD8+ T cells [50].Although monoclonal antibodies against CD11b have beenunsuccessful as single agents for the treatment of inflam-matory disorders, antagonists targeting the CSF1CSF1Rpathway in breast cancer and the CCL2CCR2 axis inprostate cancer are now in early phase clinical trials.

    We anticipate emerging therapeutics to focus on repo-larization as a method to invoke the antitumor potential ofTAMs, as has been reported in pancreatic ductal adeno-carcinoma for agonist antibodies against the co-stimulato-ry molecule CD40 [120], and the use of Tie2+ monocytes todelivery IFNa to tumors [124]. Additional approachesinclude synthetic TLR ligands such as CpG [125] or imi-quimod [126], although reports that TLR signaling andnuclear factor (NF)-kB activation in TAMs promote tumorgrowth [117,118,127] suggest that TLR ligands must beused in a multitargeted approach [125]. Based upon ourfindings in mouse models of mammary and squamouscarcinogenesis, we are currently evaluating whether block-ing protumor polarization of TAMs, as opposed to directrepolarization, is similarly efficacious in preclinical modelsin combination with chemotherapy. Although this mayseem like a distinction without a difference, we hypothe-size that this approach may be less prone to refractoryresponses and adverse autoimmune side effects.

    AcknowledgmentsThe authors thank Dr Anna Wasiuk for helpful discussion. This work wassupported by a Department of Defense Breast Cancer Research Program(BCRP) Fellowship to B.R., and grants from the NIH/NCI (R01CA130980,R01CA132566, R01CA140943, R01 CA155331), the Department ofDefense BCRP Era of Hope Scholar Expansion Award (W81XWH-10-BCRP-EOHS-EXP) and W81XWH-08-PRMRP-IIRA to L.M.C.

    References1 Gordon, S. (2003) Alternative activation of macrophages. Nat. Rev.Immunol. 3, 2335

    2 Russell, D.G. et al. (2009) Foamy macrophages and the progression ofthe human tuberculosis granuloma. Nat. Immunol. 10, 943948

    3 Serhan, C.N. and Savill, J. (2005) Resolution of inflammation: thebeginning programs the end. Nat. Immunol. 6, 11911197

    Review4 Hotamisligil, G.S. and Erbay, E. (2008) Nutrient sensing andinflammation in metabolic diseases. Nat. Rev. Immunol. 8, 923934

    1245 Lumeng, C.N. and Saltiel, A.R. (2011) Inflammatory links betweenobesity and metabolic disease. J. Clin. Invest. 121, 21112117

    6 Medzhitov, R. (2008) Origin and physiological roles of inflammation.Nature 454, 428435

    7 Tabas, I. (2010) Macrophage death and defective inflammationresolution in atherosclerosis. Nat. Rev. Immunol. 10, 3646

    8 Doedens, A.L. et al. (2010) Macrophage expression of hypoxia-inducible factor-1{alpha} suppresses T-cell function and promotestumor progression. Cancer Res. 70, 74657475

    9 Lotze, M.T. and Tracey, K.J. (2005) High-mobility group box 1 protein(HMGB1): nuclear weapon in the immune arsenal. Nat. Rev.Immunol. 5, 331342

    10 Qian, B.Z. and Pollard, J.W. (2010) Macrophage diversity enhancestumor progression and metastasis. Cell 141, 3951

    11 Ruffell, B. et al. (2010) Lymphocytes in cancer development:polarization towards pro-tumor immunity. Cytokine Growth FactorRev. 21, 310

    12 Kerr, K.M. et al. (1998) Partial regression in primary carcinoma of thelung: does it occur? Histopathology 33, 5563

    13 Kim, D.W. et al. (2008) High tumour islet macrophage infiltrationcorrelates with improved patient survival but not with EGFRmutations, gene copy number or protein expression in resectednon-small cell lung cancer. Br. J. Cancer 98, 11181124

    14 Bingle, L. et al. (2002) The role of tumour-associated macrophages intumour progression: implications for new anticancer therapies. J.Pathol. 196, 254265

    15 Salvesen, H.B. and Akslen, L.A. (1999) Significance of tumour-associated macrophages, vascular endothelial growth factor andthrombospondin-1 expression for tumour angiogenesis andprognosis in endometrial carcinomas. Int. J. Cancer 84, 538543

    16 Nishie, A. et al. (1999) Macrophage infiltration and heme oxygenase-1expression correlate with angiogenesis in human gliomas. Clin.Cancer Res. 5, 11071113

    17 Lissbrant, I.F. et al. (2000) Tumor associated macrophages in humanprostate cancer: relation to clinicopathological variables and survival.Int. J. Oncol. 17, 445451

    18 Fujimoto, J. et al. (2000) Clinical implications of expression ofinterleukin 8 related to angiogenesis in uterine cervical cancers.Cancer Res. 60, 26322635

    19 Leek, R.D. et al. (1996) Association of macrophage infiltration withangiogenesis and prognosis in invasive breast carcinoma. Cancer Res.56, 46254629

    20 Leek, R.D. et al. (2000) Macrophage infiltration is associated withVEGF and EGFR expression in breast cancer. J. Pathol. 190, 430436

    21 Uzzan, B. et al. (2004) Microvessel density as a prognostic factor inwomen with breast cancer: a systematic review of the literature andmeta-analysis. Cancer Res. 64, 29412955

    22 Tsutsui, S. et al. (2005) Macrophage infiltration and its prognosticimplications in breast cancer: the relationship with VEGF expressionand microvessel density. Oncol. Rep. 14, 425431

    23 Chen, J.J. et al. (2005) Tumor-associated macrophages: the double-edged sword in cancer progression. J. Clin. Oncol. 23, 953964

    24 Bolat, F. et al. (2006) Microvessel density, VEGF expression, andtumor-associated macrophages in breast tumors: correlations withprognostic parameters. J. Exp. Clin. Cancer Res. 25, 365372

    25 Varney, M.L. et al. (2005) Tumour-associated macrophage infiltration,neovascularization and aggressiveness in malignant melanoma: roleof monocyte chemotactic protein-1 and vascular endothelial growthfactor-A. Melanoma Res. 15, 417425

    26 Robinson, B.D. et al. (2009) Tumor microenvironment of metastasis inhuman breast carcinoma: a potential prognostic marker linked tohematogenous dissemination. Clin. Cancer Res. 15, 24332441

    27 Beck, A.H. et al. (2009) The macrophage colony-stimulating factor 1response signature in breast carcinoma. Clin. Cancer Res. 15, 778787

    28 Kawamura, K. et al. (2009) Detection of M2 macrophages and colony-stimulating factor 1 expression in serous and mucinous ovarianepithelial tumors. Pathol. Int. 59, 300305

    29 Bailey, C. et al. (2007) Chemokine expression is associated with theaccumulation of tumour associated macrophages (TAMs) andprogression in human colorectal cancer. Clin. Exp. Metastasis 24,121130

    Trends in Immunology March 2012, Vol. 33, No. 330 Kang, J.C. et al. (2010) Intratumoral macrophage counts correlate withtumor progression in colorectal cancer. J. Surg. Oncol. 102, 242248

  • 31 Pyonteck, S.M. et al. (2011) Deficiency of the macrophage growthfactor CSF-1 disrupts pancreatic neuroendocrine tumor development.Oncogene DOI: 10.1038/onc.2011.337

    32 Hanada, T. et al. (2000) Prognostic value of tumor-associatedmacrophage count in human bladder cancer. Int. J. Urol. 7, 263269

    33 Curiel, T.J. et al. (2004) Specific recruitment of regulatory T cells inovarian carcinoma fosters immune privilege and predicts reducedsurvival. Nat. Med. 10, 942949

    34 Kryczek, I. et al. (2006) B7-H4 expression identifies a novelsuppressive macrophage population in human ovarian carcinoma.J. Exp. Med. 203, 871881

    35 Kryczek, I. et al. (2007) Relationship between B7-H4, regulatory Tcells, and patient outcome in human ovarian carcinoma. Cancer Res.67, 89008905

    36 Lin, E.Y. et al. (2007) VEGF restores delayed tumor progression intumors depleted of macrophages. Mol. Oncol. 1, 288302

    37 Stockmann, C. et al. (2008) Deletion of vascular endothelial growthfactor in myeloid cells accelerates tumorigenesis. Nature 456, 814818

    38 Giraudo, E. et al. (2004) An amino-bisphosphonate targets MMP-9-expressing macrophages and angiogenesis to impair cervicalcarcinogenesis. J. Clin. Invest. 114, 623633

    39 Du, R. et al. (2008) HIF1alpha induces the recruitment of bonemarrow-derived vascular modulatory cells to regulate tumorangiogenesis and invasion. Cancer Cell 13, 206220

    40 Rolny, C. et al. (2011) HRG inhibits tumor growth and metastasis byinducing macrophage polarization and vessel normalization throughdownregulation of PlGF. Cancer Cell 19, 3144

    41 Willett, C.G. et al. (2005) Surrogate markers for antiangiogenictherapy and dose-limiting toxicities for bevacizumab with radiationand chemotherapy: continued experience of a phase I trial in rectalcancer patients. J. Clin. Oncol. 23, 81368139

    42 Motzer, R.J. et al. (2006) Activity of SU11248, a multitargetedinhibitor of vascular endothelial growth factor receptor andplatelet-derived growth factor receptor, in patients with metastaticrenal cell carcinoma. J. Clin. Oncol. 24, 1624

    43 Fischer, C. et al. (2007) Anti-PlGF inhibits growth of VEGF(R)-inhibitor-resistant tumors without affecting healthy vessels. Cell131, 463475

    44 Dirkx, A.E. et al. (2006) Monocyte/macrophage infiltration in tumors:modulators of angiogenesis. J. Leukoc. Biol. 80, 11831196

    45 Murdoch, C. et al. (2008) The role of myeloid cells in the promotion oftumour angiogenesis. Nat. Rev. Cancer 8, 618631

    46 Motz, G.T. and Coukos, G. (2011) The parallel lives of angiogenesisand immunosuppression: cancer and other tales. Nat. Rev. Immunol.11, 702711

    47 De Palma, M. et al. (2005) Tie2 identifies a hematopoietic lineage ofproangiogenic monocytes required for tumor vessel formation and amesenchymal population of pericyte progenitors. Cancer Cell 8, 211226

    48 Mazzieri, R. et al. (2011) Targeting the ANG2/TIE2 axis inhibitstumor growth and metastasis by impairing angiogenesis anddisabling rebounds of proangiogenic myeloid cells. Cancer Cell 19,512526

    49 Guy, C.T. et al. (1992) Induction of mammary tumors by expression ofpolyomavirus middle T oncogene: a transgenic mouse model formetastatic disease. Mol. Cell. Biol. 12, 954961

    50 DeNardo, D.G. et al. (2011) Leukocyte complexity predicts breastcancer survival and functionally regulates response tochemotherapy. Cancer Discov. 1, 5467

    51 Condeelis, J. and Pollard, J.W. (2006) Macrophages: obligate partnersfor tumor cell migration, invasion, and metastasis. Cell 124, 263266

    52 Aharinejad, S. et al. (2004) Colony-stimulating factor-1 blockade byantisense oligonucleotides and small interfering RNAs suppressesgrowth of human mammary tumor xenografts in mice. Cancer Res. 64,53785384

    53 Kubota, Y. et al. (2009) M-CSF inhibition selectively targetspathological angiogenesis and lymphangiogenesis. J. Exp. Med.206, 10891102

    54 Abraham, D. et al. (2010) Stromal cell-derived CSF-1 blockadeprolongs xenograft survival of CSF-1-negative neuroblastoma. Int.J. Cancer 126, 13391352

    Review55 Pollard, J.W. (2009) Trophic macrophages in development anddisease. Nat. Rev. Immunol. 9, 25927056 Coussens, L.M. and Pollard, J.W. (2010) Leukocytes in mammarydevelopment and cancer. Cold Spring Harb. Perspect. Biol. 3, DOI:10.1101/cshperspect.a003285

    57 Isbel, N.M. et al. (2001) Tubules are the major site of M-CSFproduction in experimental kidney disease: correlation with localmacrophage proliferation. Kidney Int. 60, 614625

    58 Frangogiannis, N.G. et al. (2003) MCSF expression is induced inhealing myocardial infarcts and may regulate monocyte andendothelial cell phenotype. Am. J. Physiol. Heart Circ. Physiol.285, H483H492

    59 Joly, S. et al. (2009) Cooperative phagocytes: resident microglia andbone marrow immigrants remove dead photoreceptors in retinallesions. Am. J. Pathol. 174, 23102323

    60 Bottazzi, B. et al. (1992) Monocyte chemotactic cytokine gene transfermodulates macrophage infiltration, growth, and susceptibility to IL-2therapy of a murine melanoma. J. Immunol. 148, 12801285

    61 Gazzaniga, S. et al. (2007) Targeting tumor-associated macrophagesand inhibition of MCP-1 reduce angiogenesis and tumor growth in ahuman melanoma xenograft. J. Invest. Dermatol. 127, 20312041

    62 Pahler, J.C. et al. (2008) Plasticity in tumor-promoting inflammation:impairment of macrophage recruitment evokes a compensatoryneutrophil response. Neoplasia 10, 329340

    63 Fujimoto, H. et al. (2009) Stromal MCP-1 in mammary tumors inducestumor-associated macrophage infiltration and contributes to tumorprogression. Int. J. Cancer 125, 12761284

    64 Qian, B.Z. et al. (2011) CCL2 recruits inflammatory monocytes tofacilitate breast-tumour metastasis. Nature 475, 222225

    65 Mizutani, K. et al. (2009) The chemokine CCL2 increases prostatetumor growth and bone metastasis through macrophage andosteoclast recruitment. Neoplasia 11, 12351242

    66 Wyckoff, J. et al. (2004) A paracrine loop between tumor cells andmacrophages is required for tumor cell migration in mammarytumors. Cancer Res. 64, 70227029

    67 Wyckoff, J.B. et al. (2007) Direct visualization of macrophage-assistedtumor cell intravasation in mammary tumors. Cancer Res. 67, 26492656

    68 DeNardo, D.G. et al. (2009) CD4(+) T cells regulate pulmonarymetastasis of mammary carcinomas by enhancing protumorproperties of macrophages. Cancer Cell 16, 91102

    69 Kessenbrock, K. et al. (2010) Matrix metalloproteinases: regulators ofthe tumor microenvironment. Cell 141, 5267

    70 Reilkoff, R.A. et al. (2011) Fibrocytes: emerging effector cells inchronic inflammation. Nat. Rev. Immunol. 11, 427435

    71 Mason, S.D. and Joyce, J.A. (2011) Proteolytic networks in cancer.Trends Cell Biol. 21, 228237

    72 Roussos, E.T. et al. (2011) Chemotaxis in cancer. Nat. Rev. Cancer 11,573587

    73 Mohamed, M.M. and Sloane, B.F. (2006) Cysteine cathepsins:multifunctional enzymes in cancer. Nat. Rev. Cancer 6, 764775

    74 Sameni, M. et al. (2009) Imaging and quantifying the dynamics oftumor-associated proteolysis. Clin. Exp. Metastasis 26, 299309

    75 Almholt, K. et al. (2005) Reduced metastasis of transgenic mammarycancer in urokinase-deficient mice. Int. J. Cancer 113, 525532

    76 Vasiljeva, O. et al. (2006) Tumor cell-derived and macrophage-derivedcathepsin B promotes progression and lung metastasis of mammarycancer. Cancer Res. 66, 52425250

    77 Gocheva, V. et al. (2010) IL-4 induces cathepsin protease activity intumor-associated macrophages to promote cancer growth andinvasion. Genes Dev. 24, 241255

    78 Ingman, W.V. et al. (2006) Macrophages promote collagenfibrillogenesis around terminal end buds of the developingmammary gland. Dev. Dyn. 235, 32223229

    79 Levental, K.R. et al. (2009) Matrix crosslinking forces tumorprogression by enhancing integrin signaling. Cell 139, 891906

    80 Gabrilovich, D.I. and Nagaraj, S. (2009) Myeloid-derived suppressorcells as regulators of the immune system. Nat. Rev. Immunol. 9,162174

    81 Biswas, S.K. et al. (2006) A distinct and unique transcriptional programexpressed by tumor-associated macrophages (defective NF-kappaB andenhanced IRF-3/STAT1 activation). Blood 107, 21122122

    82 Ojalvo, L.S. et al. (2009) High-density gene expression analysis of

    Trends in Immunology March 2012, Vol. 33, No. 3tumor-associated macrophages from mouse mammary tumors. Am. J.Pathol. 174, 10481064

    125

  • Trends in Immunology March 2012, Vol. 33, No. 383 Movahedi, K. et al. (2010) Different tumor microenvironments containfunctionally distinct subsets of macrophages derived from Ly6C(high)monocytes. Cancer Res. 70, 57285739

    84 Molon, B. et al. (2011) Chemokine nitration prevents intratumoralinfiltration of antigen-specific T cells. J. Exp. Med. 208, 19491962

    85 Lu, T. et al. (2011) Tumor-infiltrating myeloid cells induce tumor cellresistance to cytotoxic T cells in mice. J. Clin. Invest. 121, 40154029

    86 Kuang, D.M. et al. (2009) Activated monocytes in peritumoral stromaof hepatocellular carcinoma foster immune privilege and diseaseprogression through PD-L1. J. Exp. Med. 206, 13271337

    87 Pedroza-Gonzalez, A. et al. (2011) Thymic stromal lymphopoietinfosters human breast tumor growth by promoting type 2inflammation. J. Exp. Med. 208, 479490

    88 Biswas, S.K. and Mantovani, A. (2010) Macrophage plasticity andinteraction with lymphocyte subsets: cancer as a paradigm. Nat.Immunol. 11, 889896

    89 Pucci, F. et al. (2009) A distinguishing gene signature shared bytumor-infiltrating Tie2-expressing monocytes (TEMs), bloodresident monocytes and embryonic macrophages suggestscommon functions and developmental relationships. Blood 114,901914

    90 Clark, C.E. et al. (2007) Dynamics of the immune reaction topancreatic cancer from inception to invasion. Cancer Res. 67,95189527

    91 Andreu, P. et al. (2010) FcRgamma activation regulatesinflammation-associated squamous carcinogenesis. Cancer Cell 17,121134

    92 Zaynagetdinov, R. et al. (2011) A critical role for macrophages inpromotion of urethane-induced lung carcinogenesis. J. Immunol. 187,57035711

    93 Pikarsky, E. et al. (2004) NF-kappaB functions as a tumour promoterin inflammation-associated cancer. Nature 431, 461466

    94 Kang, T.W. et al. (2011) Senescence surveillance of pre-malignanthepatocytes limits liver cancer development. Nature 479, 547551

    95 Terabe, M. et al. (2003) Transforming growth factor-beta productionand myeloid cells are an effector mechanism through which CD1d-restricted T cells block cytotoxic T lymphocyte-mediated tumorimmunosurveillance: abrogation prevents tumor recurrence. J. Exp.Med. 198, 17411752

    96 Sica, A. et al. (2000) Autocrine production of IL-10 mediates defectiveIL-12 production and NF-kappa B activation in tumor-associatedmacrophages. J. Immunol. 164, 762767

    97 Kambayashi, T. et al. (1995) Potential involvement of IL-10 insuppressing tumor-associated macrophages. Colon-26-derivedprostaglandin E2 inhibits TNF-alpha release via a mechanisminvolving IL-10. J. Immunol. 154, 33833390

    98 Daniel, D. et al. (2003) Immune enhancement of skin carcinogenesisby CD4+ T cells. J. Exp. Med. 197, 10171028

    99 Ruffell, B. et al. (2011) Leukocyte composition of human breast cancer.Proc. Natl. Acad. Sci. U.S.A. DOI: 10.1073/pnas.1104303108

    100 Ruffell, B. et al. (2011) Differential use of chondroitin sulfate toregulate hyaluronan binding by receptor CD44 in inflammatoryand interleukin 4-activated macrophages. J. Biol. Chem. 286,1917919190

    101 Gastl, G.A. et al. (1993) Interleukin-10 production by humancarcinoma cell lines and its relationship to interleukin-6expression. Int. J. Cancer 55, 96101

    102 Kim, J. et al. (1995) IL-10 production in cutaneous basal andsquamous cell carcinomas. A mechanism for evading the local Tcell immune response. J. Immunol. 155, 22402247

    103 Lang, R. et al. (2002) Shaping gene expression in activated and restingprimary macrophages by IL-10. J. Immunol. 169, 22532263

    104 Moore, K.W. et al. (2001) Interleukin-10 and the interleukin-10receptor. Annu. Rev. Immunol. 19, 683765

    105 Allavena, P. et al. (2010) Engagement of the mannose receptor bytumoral mucins activates an immune suppressive phenotype inhuman tumor-associated macrophages. Clin. Dev. Immunol. 2010,547179

    106 Heusinkveld, M. et al. (2011) M2 macrophages induced byprostaglandin E2 and IL-6 from cervical carcinoma are switched toactivated M1 macrophages by CD4+ Th1 cells. J. Immunol. 187,

    Review11571165

    126107 Akaogi, J. et al. (2004) Prostaglandin E2 receptors EP2 and EP4 areup-regulated in peritoneal macrophages and joints of pristane-treatedmice and modulate TNF-alpha and IL-6 production. J. Leukoc. Biol.76, 227236

    108 Greenhough, A. et al. (2009) The COX-2/PGE2 pathway: key roles inthe hallmarks of cancer and adaptation to the tumourmicroenvironment. Carcinogenesis 30, 377386

    109 Rothwell, P.M. et al. (2010) Long-term effect of aspirin on colorectalcancer incidence and mortality: 20-year follow-up of five randomisedtrials. Lancet 376, 17411750

    110 Rothwell, P.M. et al. (2011) Effect of daily aspirin on long-term risk ofdeath due to cancer: analysis of individual patient data fromrandomised trials. Lancet 377, 3141

    111 Nakanishi, Y. et al. (2011) COX-2 inhibition alters the phenotype oftumor-associated macrophages from M2 to M1 in ApcMin/+ mousepolyps. Carcinogenesis 32, 13331339

    112 Paez-Ribes, M. et al. (2009) Antiangiogenic therapy elicits malignantprogression of tumors to increased local invasion and distantmetastasis. Cancer Cell 15, 220231

    113 Imtiyaz, H.Z. and Simon, M.C. (2010) Hypoxia-inducible factors asessential regulators of inflammation. Curr. Top. Microbiol. Immunol.345, 105120

    114 Imtiyaz, H.Z. et al. (2010) Hypoxia-inducible factor 2alpha regulatesmacrophage function in mouse models of acute and tumorinflammation. J. Clin. Invest. 120, 26992714

    115 Lewis, J.S. et al. (2000) Expression of vascular endothelial growthfactor by macrophages is up-regulated in poorly vascularized areas ofbreast carcinomas. J. Pathol. 192, 150158

    116 Apetoh, L. et al. (2007) Toll-like receptor 4-dependent contribution ofthe immune system to anticancer chemotherapy and radiotherapy.Nat. Med. 13, 10501059

    117 Luo, J.L. et al. (2004) Inhibition of NF-kappaB in cancer cells convertsinflammation- induced tumor growth mediated by TNFalpha toTRAIL-mediated tumor regression. Cancer Cell 6, 297305

    118 Kim, S. et al. (2009) Carcinoma-produced factors activate myeloidcells through TLR2 to stimulate metastasis. Nature 457, 102106

    119 Sorokin, L. (2010) The impact of the extracellular matrix oninflammation. Nat. Rev. Immunol. 10, 712723

    120 Beatty, G.L. et al. (2011) CD40 agonists alter tumor stroma and showefficacy against pancreatic carcinoma in mice and humans. Science331, 16121616

    121 Ciardiello, F. and Tortora, G. (2008) EGFR antagonists in cancertreatment. N. Engl. J. Med. 358, 11601174

    122 Kerbel, R.S. (2008) Tumor angiogenesis. N. Engl. J. Med. 358, 20392049

    123 Ahn, G.O. et al. (2010) Inhibition of Mac-1 (CD11b/CD18) enhancestumor response to radiation by reducing myeloid cell recruitment.Proc. Natl. Acad. Sci. U.S.A. 107, 83638368

    124 De Palma, M. et al. (2008) Tumor-targeted interferon-alpha deliveryby Tie2-expressing monocytes inhibits tumor growth and metastasis.Cancer Cell 14, 299311

    125 Guiducci, C. et al. (2005) Redirecting in vivo elicited tumor infiltratingmacrophages and dendritic cells towards tumor rejection. Cancer Res.65, 34373446

    126 Schon, M.P. and Schon, M. (2008) TLR7 and TLR8 as targets in cancertherapy. Oncogene 27, 190199

    127 Hagemann, T. et al. (2008) Re-educating tumor-associatedmacrophages by targeting NF-kappaB. J. Exp. Med. 205, 12611268

    128 Joyce, J.A. and Pollard, J.W. (2009) Microenvironmental regulation ofmetastasis. Nat. Rev. Cancer 9, 239252

    129 Hashimoto, D. et al. (2011) Dendritic cell and macrophageheterogeneity in vivo. Immunity 35, 323335

    130 Orimo, A. et al. (2005) Stromal fibroblasts present in invasive humanbreast carcinomas promote tumor growth and angiogenesis throughelevated SDF-1/CXCL12 secretion. Cell 121, 335348

    131 Erez, N. et al. (2010) Cancer-associated fibroblasts are activated inincipient neoplasia to orchestrate tumor-promoting inflammation inan NF-kappaB-dependent manner. Cancer Cell 17, 135147

    132 Karnoub, A.E. et al. (2007) Mesenchymal stem cells within tumourstroma promote breast cancer metastasis. Nature 449, 557563

    133 Uccelli, A. et al. (2008) Mesenchymal stem cells in health and disease.

    Nat. Rev. Immunol. 8, 726736

    Differential macrophage programming in the tumor microenvironmentMacrophages in solid malignanciesTAM functionAngiogenesisInvasion and metastasisImmune suppression

    Mechanisms of TAM polarizationImmune microenvironmentTumor-derived mediatorsHomeostatic imbalance

    Concluding remarks: targets for therapyAcknowledgmentsReferences