Skip to main content
Oncoimmunology logoLink to Oncoimmunology
. 2012 Nov 1;1(8):1323–1343. doi: 10.4161/onci.22009

Trial watch

Prognostic and predictive value of the immune infiltrate in cancer

Laura Senovilla 1,2,3,, Erika Vacchelli 1,2,3,, Jerome Galon 4,5,6,7,8, Sandy Adjemian 1,2,3, Alexander Eggermont 2, Wolf Hervé Fridman 4,5,6,7, Catherine Sautès-Fridman 4,6,8, Yuting Ma 1,2,3, Eric Tartour 7,9, Laurence Zitvogel 2,10, Guido Kroemer 1,4,6,7,11,‡,*, Lorenzo Galluzzi 1,4,‡,*
PMCID: PMC3518505  PMID: 23243596

Abstract

Solid tumors are constituted of a variety of cellular components, including bona fide malignant cells as well as endothelial, structural and immune cells. On one hand, the tumor stroma exerts major pro-tumorigenic and immunosuppressive functions, reflecting the capacity of cancer cells to shape the microenvironment to satisfy their own metabolic and immunological needs. On the other hand, there is a component of tumor-infiltrating leucocytes (TILs) that has been specifically recruited in the attempt to control tumor growth. Along with the recognition of the critical role played by the immune system in oncogenesis, tumor progression and response to therapy, increasing attention has been attracted by the potential prognostic and/or predictive role of the immune infiltrate in this setting. Data from large clinical studies demonstrate indeed that a robust infiltration of neoplastic lesions by specific immune cell populations, including (but not limited to) CD8+ cytotoxic T lymphocytes, Th1 and Th17 CD4+ T cells, natural killer cells, dendritic cells, and M1 macrophages constitutes an independent prognostic indicator in several types of cancer. Conversely, high levels of intratumoral CD4+CD25+FOXP3+ regulatory T cells, Th2 CD4+ T cells, myeloid-derived suppressor cells, M2 macrophages and neutrophils have frequently been associated with dismal prognosis. So far, only a few studies have addressed the true predictive potential of TILs in cancer patients, generally comforting the notion that—at least in some clinical settings—the immune infiltrate can reliably predict if a specific patient will respond to therapy or not. In this Trial Watch, we will summarize the results of clinical trials that have evaluated/are evaluating the prognostic and predictive value of the immune infiltrate in the context of solid malignancies.

Keywords: biomarker, chemotherapy, cytokines, interferon γ, interleukin-10, plasmacytoid dendritic cells, transforming growth factor

Introduction

For a long time, the heterogeneity of solid tumors has been incredibly underestimated and neoplastic lesions have often been regarded as aggregates of relatively disorganized tumor cells supported by their own vasculature. During the last few decades, this oversimplified view has been challenged by a consistent volume of scientific literature generated from multiple laboratories worldwide. Nowadays, it is generally accepted that cancer cells within neoplastic lesions are highly heterogeneous, exhibiting rather distinct phenotypic, proliferative, differentiative and functional profiles.1 Two theories have been proposed to account for such a heterogeneity: the clonal evolution and the cancer stem cell model.2,3 According to the former, a mutant population of cancer cells would, at some stage, acquire a proliferative advantage and hence become the major driver behind tumorigenesis.2 The latter, which has been proposed much later and for which compelling evidence has accumulated only recently, advocates a hierarchical organization of cancer cells, with a prominent role for a subpopulation of stem-like cells that would sustain tumor growth.3 In addition, it has nowadays become evident that solid tumors are constituted of multiple cellular components, including bona fide malignant cells as well as endothelial, structural and immune cells.4,5 Often, such non-malignant cell populations largely outnumber tumor cells, a notion with important pathophysiological and therapeutic implications that has received appropriate attention only recently.6-8

Accumulating evidence indicates that malignant cells exert a major control on their non-malignant neighbors. Thus, most cancer cells not only promote angiogenesis to support tumor growth beyond the size limit that would be dictated by a poorly vascularized microenvironment,9,10 but also activate metabolic circuitries whereby stromal cells are de facto rewired to function as a feeder compartment, generating large amounts of energetic products such as lactate and ketone bodies.11,12 Cancer-associated fibroblasts (CAFs) are prominent sources of mitogenic and pro-angiogenic factors such as interleukin (IL)-6 and vascular endothelial growth factor (VEGF).13 Furthermore, cancer cells, either directly or through CAFs, produce a wide array of cytokines including transforming growth factor β (TGFβ) and IL-10 that exert potent immunosuppressive effects.14 Altogether, these observations demonstrate that during oncogenesis, malignant cells become capable of co-opting the local microenvironment in order to satisfy their own metabolic and immunological needs.

Although part of the immune infiltrate is constituted by immunosuppressive cells that are specifically recruited and/or instructed by the tumor to maintain an immunoprivileged microenvironment, some tumor-infiltrating leukocytes (TILs) reflect the attempt of the immune system to mount an antitumor response.7,15,16 Immunosuppressive TILs include (but are not limited to) CD4+CD25+FOXP3+ regulatory T cells (Tregs), T-helper 2 (Th2) CD4+ T cells, myeloid-derived suppressor cells (MDSCs), M2 macrophages and N2 neutrophils. Conversely, CD8+ cytotoxic T lymphocytes (CTLs), T-helper 1 (Th1) and T-helper 17 (Th17) CD4+ T cells, M1 macrophages, N1 neutrophils, natural killer (NK) cells and dendritic cells (DCs) most often promote antitumor responses.7,15

Of note, the major role that the immune system plays in oncogenesis, tumor progression and response to therapy has received appropriate credit only during the last decade. Previously, the immune system would be considered as a mere bystander of cancer, incapable of reacting as a result of conventional self-tolerance mechanisms.17 Now, it has become clear that inflammatory and immune reactions can exert both pro- and anti-tumor effects, depending on the specific context.17 In particular, the cancer immunoediting hypothesis postulates that the immune system initially can eradicate potentially tumorigenic cells as they develop, a capacity that is progressively lost when transformed cells acquire mutations that sustain immunosubversion and/or immunoevasion.17 Moreover, accumulating evidence indicates that the success of some anticancer therapies, including conventional cytotoxic compounds, radiotherapy as well as targeted agents, depends (at least partially) on the activation of anticancer immune responses.8 Along with these conceptual revolutions, increasing interest has been attracted by the possibility that the abundance of specific tumor-infiltrating cell populations might constitute a biomarker for risk stratification (i.e., a prognostic biomarker) or even predict the response of patients to specific therapies (i.e., would constitute a predictive biomarker).7,18

Along the lines of our Trial Watch series,19-25 we will discuss clinical trials that have investigated/are investigating the prognostic and/or predictive value of the immune infiltrate in the context of solid malignancies.

T Lymphocytes

T lymphocytes constitute a diversified group of immune cells sharing the expression of an antigen-specific T-cell receptor (TCR) and a few key signaling molecules, including CD3. T cells develop in the thymus and are locally selected for (1) not recognizing self antigens (negative selection) and (2) being capable to recognize non-self antigens when presented in the context of autologous MHC molecules (positive selection).26,27 T cells can be grossly subdivided based on the chains that compose their TCR into αβ and γδ cells, the latter constituting a small population that operate at the interface between innate and adaptive immunity.28 Alternatively, T lymphocytes can be classified based on the expression at the cell surface of either CD8, allowing CTLs to recognize antigenic epitopes presented on MHC class I molecules, or CD4, allowing helper T cells to recognize antigenic epitopes presented on MHC class II molecules. According to a perhaps oversimplified model, CD8+ CTLs can specifically recognize (via their TCR) and kill infected or malignant cells, whereas TCR-activated CD4+ T cells secrete stimulatory signals for CTLs as well as for other immune cell populations.29 In this setting, T-helper (Th) responses can be classified based on the cytokine cocktail that is secreted by activated CD4+ T cells. Thus, while Th1 responses (which promote antiviral and antitumor effects) mainly involve IL-2 and interferon (IFN) γ, their Th2 counterparts develop along with the secretion of IL-4, IL-5, IL-10 and TGFβ, de facto exerting pro-tumorigenic functions.30 The precise impact of Th17 responses (featuring the secretion of high levels of IL-17) on oncogenesis, tumor progression and response to therapy remains to be elucidated.31-33 Irrespective of this unresolved issue, a critical class of CD4+ cells is constituted by Tregs, CD4+CD25+FOXP3+ lymphocytes that exert potent and multifaceted immunosuppressive functions.34 Of note, activated CD4+ and CD8+ T lymphocytes can differentiate into central or effector memory T cells, both of which are characterized by a CD45RA-CD45RO+ phenotype (the former, but not the latter, also expressing CCR7).35,36 Memory T cells are important in that they drive secondary immune responses (i.e., immune responses against antigens that have already been in contact with the immune system), de facto underlying the efficacy of vaccination.35 All naïve (CD45RA+) T cells must be activated by antigen-presenting cells (APCs) such as macrophages or DCs, providing both a TCR-transduced signal (i.e., an antigenic epitope presented in association of self MHC molecules) and a co-stimulatory signal, such as that transduced by the binding of B7 molecules on APCs and CD28 on T cells.37,38

Whether tumor infiltration by T cells would influence disease outcome in cohorts of cancer patients was one of the first questions of tumor immunology, formulated even before the actual recognition of oncoimmunology as a self-standing discipline, and has been extensively addressed during the past 15 y.8 How this question has been approached provides interesting insights into the theoretical and technical advances that have been made in the field since then. Indeed, for a long period extending until today, intratumoral T cells have been quantified based on one or more of their major surface markers, namely CD3, CD4 and CD8. Although this approach may appear relatively gross in view of the extensive functional heterogeneity of T cells, it has generated a sizeable amount of literature demonstrating that high levels of intratumoral T cells generally constitute a positive prognostic factor. Thus, high levels of intratumoral CD3+ cells have been associated with improved disease outcomes in cohorts of carcinoma patients receiving neoadjuvant chemotherapy (n = 25);39 in surgically resected hepatocellular carcinoma (HCC) patients (n = not available), together with DC infiltration;40 in colorectal cancer patients (n = 160), highlighting a prominent role for the peritumoral infiltrate;41 in subjects affected by advanced ovarian carcinoma (n = 186); and in prostate cancer patients receiving IL-2-based immunotherapy (n = 24)42 or not (n = 59).43 Tumor infiltration by cytotoxic CD3+CD8+ CTLs has been attributed a positive prognostic value in cohorts of breast carcinoma patients (n = 1334), positively correlating with tumor grade and inversely correlating with age at diagnosis, estrogen receptor as well as progesterone receptor positivity;44 differentiated thyroid carcinoma patients (n = 398);45 subjects affected by oral squamous cell carcinoma (n = 132);46 glioblastoma patients vaccinated with DC-based immunotherapy (n = 23);47 subjects affected by esophageal carcinoma (n = 70);48 non-small cell lung carcinoma (NSCLC) patients (n = 199), highlighting a preponderant prognostic value for T cells infiltrating cancer nests and the tumor stroma;49 subjects affected by melanoma (n = 264 and n = 285), a setting in which T-cell infiltration appears to convey independent prognostic information;50,51 surgically resected HCC patients (n = 44);52 Merkel cell carcinoma patients (n = 146);53 subjects affected by colorectal carcinoma (CRC) (n = 447, n = 276, n = 41, n = 93, n = 152, n = 97; n = 160 and n = 470);54-61 ovarian carcinoma patients who underwent surgical resection (n = 70);62 prostatic adenocarcinoma patients (n = 325);63 and subjects affected by muscle-invasive urothelial carcinoma (n = 69).64 Similarly, increased intratumoral levels of both CD3+CD8+ CTLs and not better characterized CD3+CD4+ helper T cells have been associated with improved clinicopathological parameters in cohorts of head and neck carcinoma (HNC) patients treated with IRX-2-based immunotherapy (n = 15, n = 42 and n = 27);65-67 esophageal cancer patients who underwent tumor resection (n = 122 and n = 181), a setting in which NK-cell accumulation also conveyed prognostic information;68,69 surgically resected NSCLC patients (n = 335), highlighting a prominent prognostic value for CD4+ cells accumulating at stromal, as opposed to epithelial, sites;70 subjects affected by melanoma;71 surgically resected HCC patients (n = 163);72 pancreatic adenocarcinoma patients (n = 80), correlating with an intense infiltration by DCs;73 gallbladder cancer patients treated with curative surgery (n = 110), a setting in which also DC, but not NK-cell, infiltration, conveyed prognostic information;74 prostate carcinoma patients undergoing radical prostatectomy (n = 188), a setting in which B-cell infiltration also provided prognostic insights;75 vulval intraepithelial neoplasia patients receiving therapeutic human papillomavirus vaccination combined with the Toll-like receptor (TLR) 7 agonist imiquimod (n = 19),76 and patients affected by various solid tumors (including breast carcinoma, melanoma and renal cell carcinoma, RCC) undergoing IL-2-based immunotherapy.77 Of note, CD3+ CD4+ and CD8+ cells (together with CD20+ and CD57+ cells) have been shown to preferentially accumulate at the margins of breast carcinoma lesions ablated by high intensity focused ultrasound (n = 23 patients), as opposed to similar lesions removed by radical mastectomy (n = 25).78 Nevertheless, no direct assessments of clinical outcome were performed in this study.

More recently, the identification of intratumoral T cell subsets (and hence the evaluation of their prognostic/predictive value) has been significantly refined, thanks to the implementation of detection techniques that allow for the quantification of T-cell functional markers including proteins related to cytotoxicity (e.g., the granule components granzyme B and TIA-1), proliferative potential (e.g., the nuclear antigen Ki67), helper profile (e.g., intracellular IFNγ, IL-4, IL-10 and IL-17), activation status (e.g., CD69), memory status (e.g., CD45RO) and immunosuppressive capacity (e.g., FOXP3) Thus, tumor infiltration by CD8+granzyme B+ cells has been associated with longer survival in cohorts of CRC (n = 131) and ovarian carcinoma (n = not available) patients,79,80 but with worsened disease outcome in anal squamous cell carcinoma patients (n = 38).81 The levels of intratumoral CD8+TIA-1+ cells inversely correlated with tumor grade, while those of Tregs directly did so, in a cohort of pancreatic ductal adenocarcinoma patients (n = 198),82 and emerged as a positive prognostic factor among optimally debulked high-grade serous epithelial ovarian carcinoma patients (n = 199).83 Increased amounts of intratumoral CD8+Ki67+ cells have been linked to improved disease outcome in cohorts of RCC (n = 221) and CRC (n = 415 and n = 131) patients,79,84,85 but not of surgically resected NSCLC patients (n = 178), a setting in which the accumulation of CD8+Ki67+ cells in cancer cell nests actually constituted an independent unfavorable prognostic factor by multivariate analysis.86 High levels of intratumoral CD69+ cells have been associated with improved locoregional control of the tumor and longer survival in melanoma (n = 58) and HNC (n = 84) patients,87,88 although only in the latter setting additional lymphocytic markers were monitored (i.e., CD4). Robust tumor infiltration by CD8+CD45RO+ cells has been associated with improved disease outcome in large cohorts of CRC patients (n = 415, n = 602, n = 599, n = 87 and n = 768);85,89-93 NSCLC patients (n = 74), a setting in which T cells were found to stay in close proximity of mature DCs (mDCs) within a so-called “tumor-induced bronchus-associated lymphoid tissue”;94,95 and HCC patients who underwent surgical resection (n = 302).96

The development of Th1 responses, alone or linked to the suppression of Th2 responses, has been shown to constitute a positive prognostic marker in cohorts of breast carcinoma patients (n = 112 and n = 1123),97,98 pediatric medulloblastoma patients (n = 17);99 subjects awaiting surgery for esophageal cancer (n = 32), correlating with TNM stage;100 gastric cancer patients who underwent curative gastrectomy (n = 157);101 CRC patients (n = 103 and n = 125);102,103 and ovarian carcinoma patients (n = 99 and n = 40).104,105 The development of Th2 responses has been associated with no prognostic value in a cohort of CRC patients (n = 125), contrarily to Th1 and Th17 responses;103 with significantly improved disease- and event-free survival in a cohort of Hodgkin Lymphoma patients (n = 87), a setting that is dominated by Th2 immunity,106 and with worsened outcomes in pancreatic cancer patients (n = 69), correlating with the establishment of an immunosuppressive tumor microenvironment.107 On a similar note, while circulating CD4+ T cells isolated from healthy donors (n = 23) responded to an antigenic challenge by producing IFNγ and granulocyte macrophage colony-stimulating factor (GM-CSF), their counterparts obtained from prostate cancer patients (n = 44) did so by producing IL-5, thus manifesting a skew toward Th2 immunity.108 Th17 responses have been given no prognostic value in a cohort of HNC patients (n = 106),109 shown to constitute positive prognostic factors in cohorts of gastric (n = 192),110 esophageal (n = 181)69 and ovarian carcinoma (n = 201) patients,111 and linked to worsened disease outcome in cohorts of NSCLC (n = 52),112 HCC (n = 178),113 and CRC (n = 125 and n = 52) patients.103,114

Recently, great efforts have been dedicated to ascertain whether the levels of intratumoral CD4+CD25+FOXP3+ Tregs would have a prognostic or predictive value in cancer patients, with rather heterogeneous results. No significant association with disease outcome has been reported for Treg infiltration in cohorts of glioma (n = 135),115 glioblastoma (n = 29)116 and melanoma (n = 58 and n = 97) patients.87,117 Conversely, high levels of intratumoral Tregs have been associated with worsened disease outcomes in cohorts of breast carcinoma patients (n = 309 and n = 1445);118,119 subjects affected by gastric cancer (n = 80), highlighting a prominent prognostic valued for Treg localization, rather than absolute abundance;120 resected NSCLC patients (n = 100 and n = 87);121,122 individuals affected by melanoma (n = 66, n = 100 and n = 50);123-125 pancreatic ductal adenocarcinoma patients (n = 198);82 RCC patients receiving IL-2-based immunotherapy (n = 100);126 and ovarian carcinoma patients (n = 104).127 Conversely (and in part surprisingly), tumor infiltration by Tregs has been shown to constitute a positive prognostic factor in cohorts of HNC patients (n = 84 and n = 56), correlating with a better locoregional control of the tumor and prolonged overall survival;88,128 individuals affected by multiple types of lymphoma (n = 97 and n = 1019);129,130 CRC patients (n = 967, n = 57, n = 1420, n = 87, n = 768 and n = 76);92,93,131-134 cystectomized bladder carcinoma patients (n = 37);135 and optimally debulked high-grade serous epithelial ovarian carcinoma patients (n = 199).83 While in some settings, such as CRC, these findings may reflect the very peculiar nature of the oncogenic program, involving a prominent pro-inflammatory component,136 in others, such as bladder carcinoma, they are not easily reconciled with the current knowledge. One possibility is that studies specifically measuring Treg infiltration are intrinsically prone to imprecise determinations as the intratumoral amounts of Tregs often correlates with the levels of cytotoxic and/or helper T cells.87 Alternatively, such apparently odd results may reflect the existence of functionally heterogeneous FOXP3+ and FOXP3- cell populations (see below).

To circumvent (at least in part) this issue and obtain a more reliable prognostic/predictive indication, several studies have measured tumor infiltration by both FOXP3+ and CD4+FOXP3- or CD8+FOXP3- cells and integrated such assessments into a combined score (most often represented by the ratio between CD4+FOXP3- or CD8+FOXP3- and FOXP3+ cells). Invariably, tumors that are predominantly infiltrated by effector cells have a better outcome than tumors in which Tregs prevail, as demonstrated in cohorts of breast carcinoma (n = 56, n = 162 and n = 60),137-139 HNC (n = 106),109 lymphoma (n = 87),106 NSCLC (n = 64),140 HCC (n = 302),141 and ovarian carcinoma (n = 117, n = 306 and n = not available) patients.80,142,143

When this Trial Watch was being redacted (August 2012), official sources listed five clinical studies that would evaluate the predictive/prognostic value of intratumoral T cells in cohorts of cancer patients (Table 1). The first one (NCT00854282; current status: recruiting) aims at investigating the impact of intratumoral FOXP3+ cells on the overall survival of ovarian carcinoma patients undergoing surgical resection. The second one (NCT00896922; current status: unknown) intends to evaluate the prognostic significance of FOXP3+ cells (as well as that of CD68+ and CD163+ cells) in follicular lymphoma patients receiving immunochemotherapy. The third one (NCT01513408 current status: recruiting) will investigate if and how intratumoral CD8+ and FOXP3+ cells influence the survival and response to therapy of breast carcinoma patients treated with neoadjuvant therapy. Finally, two trials (NCT00673192; NCT00854269; current status: unknown) will evaluate the predictive/prognostic value of cellular (and humoral) immunity in cohorts of cervical cancer patients, yet the authors do not clearly state how they intend to quantify these parameters (source www.clinicaltrials.gov).

Table 1. Current clinical trials involving the prognostic/predictive evaluation of the intratumoral immune infiltrate.

Cell type Setting Phase Status Notes Ref.
B cells (?)
T cells (?)
Cervical carcinoma
Surgery
n.a.
Unknown
Not better specified approach and
clinicopathological parameters
NCT00673192
NCT00854269
FOXP3+ Tregs
Ovarian carcinoma
Surgery
n.a.
Recruiting
Not better specified approach
Primary outcome: OS
Secondary outcome: Severity and progression
NCT00854282
FOXP3+ Tregs
CD8+ T cells
Breast carcinoma
Neoadjuvant chemotherapy
n.a.
Recruiting
IHC detection of the CD8+/ FOXP3+ T-cell ratio
Primary outcome: OS
Secondary outcome: RFS, pCR and PathIm score
NCT01513408
FOXP3+ Tregs
TAMs
Follicular lymphoma
Immunochemotherapy
n.a.
Unknown
IHC detection of FOXP3+, CD68+ and CD163+ cells
Primary outcome: Prognostic significance
of FOXP3+ Tregs and TAMs
NCT00896922
FOXP3+ Tregs
T cells
Bladder cancer
Immunochemotherapy
n.a.
Ongoing,
Not recruiting
Not better specified approach
Primary outcome: OS
Secondary outcome: Response to therapy
NCT01198808
TAMs
Lung cancer
Not specified
n.a.
Unknown
Not better specified detection of M1 vs. M2 TAMs
Primary outcome: OS and response to therapy
Secondary outcome: Clinical presentation
NCT00690261
TAMs Wilms’ tumor
Not specified
n.a. Unknown Not better specified IHC approach
Primary outcome: Prognostic significance of TAMs
NCT01493817

Abbreviations: IHC, immunohistochemical; n.a., not available; OS, overall survival; PathIm, pathological-immunological; pCR, pathological complete response; TAM, tumor-associated macrophage; Treg, CD4+CD25+FOXP3+ regulatory T cell.

NK and NKT Cells

NK cells are a group of CD3-TCR- large granular lymphocytes that derive from common lymphoid progenitors that also generate B and T lymphocytes, the main anatomical sites for NK-cell differentiation being the bone marrow, spleen, thymus, lymph nodes and tonsils.144 NK cells play a fundamental role in innate immunity. Thus, owing to a diversified set of inhibitory and activating transmembrane receptors, NK cells are capable of specifically recognizing and killing virus-infected and transformed cells well before the initiation of an adaptive immune response.144,145 In addition, NK cells express the co-stimulatory receptor CD7, the adhesion molecule CD56, the glucuronosyltransferase CD57 (which is also expressed by T cells) and CD16 (FcγRIII), hence sharing with macrophages the ability to detect opsonized material.146 However, while macrophages respond to the engagement of CD16 by activating the phagocytic program (see below), NK cells mediate the lysis of opsonized cells by releasing perforin- and granzyme B-containing granules.144,145 Of note, a consistent proportion (~50%) of circulating NK cells expresses CD8, labeling an NK-cell subset that exhibits increased survival upon activation and enhanced effector functions.147 Along similar lines, NK cells, in particular those found in lymphoid tissues, express CD4, increasing their capacity to secrete cytokines upon activation and conferring them the ability to respond to the chemotactic factor IL-16.148 Beyond their innate cytotoxic activity against infected and malignant cells, NK cells are also important for adaptive immunity, in particular as they mediate the so-called “DC editing.”149,150 Thus, NK cells are capable of selectively killing DCs that fail to properly mature in response to activation stimuli (often as these cells express low levels of MHC class I and II molecules), de facto purging the DC repertoire from potentially tolerogenic cells.149,150 In line with their critical contributions to both the innate and adaptive arms of the immune response, NK cells have been recognized to mediate considerable antitumor effects more than one decade ago.151-154 In particular, NK cells appear to underlie the mechanism of immunosurveillance that (at least initially) prevents metastatic colonization.144,145 Recent data confirm that NK cells are required for the elicitation of potent tumor-specific CTL responses, presumably as they edit, hence optimizing, the DC repertoire.155

No statistically significant association between high intratumoral levels of NK cells, most often detected as CD56+ or CD57+ cells, and clinicopathological parameters has been detected in cohorts of esophageal carcinoma patients (n = 122), a setting in which T-cell infiltration was shown to strongly correlated with prognosis;68 stage I NSCLC patients (n = 40);156 and subjects affected by gallbladder cancer (n = 110), at odds with T-cell and DC infiltration.74 The findings by Cho et al. were disconfirmed by several subsequent studies, linking tumor infiltration by NK cells to improved disease outcomes in (primary resected) esophageal carcinoma patients,69,157,158 even in settings in which high levels of intratumoral DCs apparently did not correlate with prognosis.157 Similarly, robust tumor infiltration by NK cells has been associated with improved disease outcomes in cohorts of head and neck squamous cell carcinoma patients receiving intratumoral IL-12 (n = 10);159 subjects affected by gastric cancer (n = 146 and n = 169), irrespective of DC infiltration;160,161 HCC patients;162 and CRC patients who underwent curative colectomy (n = 157 and n = 93), correlating with the intratumoral abundance of T cells.57,163 Along similar lines, it has recently been demonstrated that parameters reflecting the functional status of circulating NK cells (in particular, IFNγ secreted in response to IL-2 or mDCs) can predict the long-term survival of gastrointestinal stromal tumor patients receiving imatinib mesylate therapy (n = 77).164,165

Natural killer T (NKT) cells constitute a lymphocyte subset sharing features of both T and NK cells. On one hand, similar to T lymphocytes, NKT cells are equipped with an αβ TCR and, optionally, with CD4 or CD8 co-receptors. On the other hand, NKT cells express CD16 and CD56, in thus far resembling NK cells.166 Upon activation, NKT cells produce large amounts of cytokines including IFNγ, IL-4 and GM-CSF, thus exerting potent antiviral and antitumor activity.166-168 Based on phenotypic markers and functional properties, NKT cells have been classified into three main groups.168 Type 1, “classical” or “invariant” NKT cells bear TCRs that are far more restricted in diversity than T-cell counterparts, obligatorily involving a Vα24 chain (Vα14 in mice).169 Type 2, “non-classical” or “diverse” NKT cells significantly resemble their invariant counterparts, with the notable exception that they can count on a non-limited TCR repertoire.166 Indeed, both these NKT-cell subsets fail to recognize antigenic peptides presented by MHC molecules, yet efficiently react to lipids and glycolipids presented by CD1d, thus playing a prominent role in innate immune responses to infectious agents such as Mycobacterium tuberculosis.170,171 Of note, whereas type 1 NKT cells are reactive against α-galactosylceramide, a component of the cell wall from Gram-negative lipopolysaccharide-negative bacteria, type 2 NKT cells are not.170,171 Finally, so-called “NKT-like” NKT cells are characterized by a normal TCR repertoire, by the capacity to recognize peptide-MHC complexes and by the expression of CD3 and CD56, de facto constituting the NKT-cell subset that most resembles T cells.168 Of note, NKT cells appear to mediate potent anticancer effects via multiple mechanisms, among which their capacity to selectively kill a peculiar subset of tumor-associated macrophages as well as MDSCs (see below).172,173 Moreover, cancer patients exhibit circulating and intratumoral NKT cells that are reduced in number and characterized by prominent functional alterations, perhaps owing to a defective crosstalk with DCs.174-177 Thus, NKT cells appear to be intimately involved in oncogenesis and tumor progression. In line with this notion, great efforts have recently been dedicated to the development of strategies that would harness the antitumor potential of NKT cells.178-180

This said, whether high intratumoral levels of CD3+CD57+ NKT cells are associated with improved clinical outcome remains to be elucidated, and perhaps may be influenced by disease setting and/or other clinicopathological variables. So far, only a few studies have addressed this issue. A high prevalence of intratumoral NKT cells has been associated with poor overall survival and reduced time-to-recurrence in a cohort of HCC patients (n = 42).181 Along similar lines, elevated levels of circulating CD3+CD57+ NKT cells have been shown to constitute (similar to tumor depth) an independent risk factor in a cohort of advanced (Stage III-IV) gastric carcinoma patients (n = 48).182 Conversely, robust tumor infiltration by invariant NKT (iNKT) cells has been indicated as an independent favorable prognostic factor in a cohort of CRC patients (n = 103), and has been suggested to predict the outcome of an NKT cell-targeted glycosphingolipid employed for the therapy of several distinct advanced solid tumors (n = 24) in the context of a Phase I clinical trial.178 Furthermore, the reconstitution of the iNKT-cell compartment appears to be required for the long-term remission of pediatric leukemia patients undergoing HLA-haploidentical stem cell transplantation, as assessed in a cohort of n = 34 individuals.183

When this Trial Watch was being redacted (August 2012), official sources listed no clinical studies that would investigate the predictive/prognostic value of NK- and NKT-cell infiltration in cohorts of cancer patients (source www.clinicaltrials.gov).

Dendritic Cells

DCs constitute a relatively small population of myeloid cells exhibiting a peculiar tree-like morphology, after which Ralph Steinman and colleagues originally named them (‘dendron’ is indeed the Greek term for tree) in 1973.184 DCs derive from myeloid bone marrow progenitors and can be found in practically all tissues, yet are highly enriched at sites where antigen exposure is most intense (e.g., lymphoid organs, the body surface, internal mucosae).185,186 In general, tissue-resident DCs are immature, i.e., they are characterized by a high capacity for taking up antigens but a limited potential for releasing cytokines, and they express (1) MHC class II molecules mostly in the late endosome-lysosomal compartment, (2) low levels of co-stimulatory molecules (e.g., OX40L, CD40, CD70, CD86) and (3) specific chemokine receptors.187 Following antigen uptake and the obligatory exposure to one among multiple maturation stimuli, including microbe-associated molecular patterns (MAMPs), endogenous damage-associated molecular patterns (DAMPs) and specific paracrine mediators, immature DCs (iDCs) acquire a novel surface phenotype and functional profile.188 As compared with iDCs, mDCs feature (1) a significantly compromised capacity to capture antigens, (2) increased levels of MHC class II molecules (including HLA-DR) and other components of the machinery for antigen presentation (e.g., CD1A, CD83, CD208/DC-LAMP, fascin) at the cell surface, (3) the expression of chemokine receptors that are required for their migration to lymphoid organs (e.g., CCR7) and (4) an improved capacity to secrete cytokines/chemokines.187 Notably, mDCs are highly efficient at eliciting adaptive immune responses, much more than other APCs including macrophages.189 Conversely, in the absence of maturation signals, iDCs efficiently present antigens to T cells in the context of inhibitory interactions, a response that is critical for the development of peripheral self tolerance.190

Multiple subsets of DCs have been described, regulating not only humoral vs. cellular immunity, but also more refined aspects of the latter.191-193 These include, but are not limited to: (1) human CD14+ dermal DCs, mainly stimulating naïve B cells to differentiate into antibody-producing plasma cells and memory B cells;194,195 (2) epidermal Langerhans cells, preferentially stimulating CD8+ T-cell responses;194,195 (3) circulating CD141+ DCs (the human homologs of murine CD8α+ DCs), perhaps constituting the DC subset most efficient at cross-presentation196-199 and (4) plasmacytoid DCs (pDCs).200,201 As opposed to their “conventional” counterparts (often indicated with the adjective “myeloid,” improperly, as all DCs have a myeloid origin), pDCs morphologically resemble to antibody-secreting plasma cells and express high levels of endosomal TLR7 and TLR9,202-204 providing them with a superior capacity to respond to MAMPs and DAMPs by secreting high levels of type I IFN (in both mice and humans) and IL-12 (only in mice).205-207 On histological sections, DCs are often identified as they express the Ca2+-binding protein S-100, with CD11c (one subunit of CR4, a heterodimeric receptor for the complement component iC3b) and CD123 (the α chain of the IL-3 receptor) being preferentially expressed to high levels by conventional DCs and pDCs, respectively.208,209 DCs occupy a central position in the immune system, operating as prominent APCs and orchestrating a wide repertoire of responses that span from the development of self tolerance to the induction of potent cellular and humoral immune responses.190,210 Accordingly, great efforts have been dedicated during the last couple of decades to the development of strategies that would harness the great immunostimulatory potential of DCs against cancer.19,187

Elevated intratumoral amounts of DCs have often, but not always, been associated with an improved clinical outcome, most likely owing to the fact that DCs exist in several functionally distinct subsets that cannot be appropriately discriminated by common immunohistochemical approaches. Thus, no statistically significant association between high levels of intratumoral DCs (most often identified as S100+ cells) and clinicopathological parameters including disease course has been detected in cohorts of esophageal carcinoma (n = 101) and lung cancer (n = 39) patients.157,211 Conversely, tumor infiltration by not-better characterized S100+ DCs has been associated with improved clinical outcomes in cohorts of nasopharyngeal carcinoma patients (n = 45), correlating with high levels of T cells and macrophages;212 subjects affected by oral squamous cell carcinoma (n = 132), a setting in which the high levels of intratumoral S100+ DCs correlated with the peritumoral accumulation of mDCs and T cells;46 esophageal cancer patients who underwent surgical resection (n = 88 and n = 203), in one setting correlating with the levels of expression of p53 by tumor cells;213,214 subjects affected by gastric carcinoma (n = 93; n = 165, n = 123, n = 169 and n = 92);161,215-218 high-risk melanoma patients receiving GM-CSF-based immunotherapy (n = 42);219 surgically resected HCC patients (n = 44 and n = not available);40,52 gallbladder cancer patients treated with curative surgery (n = 110);74 subjects affected by RCC (n = 69), negatively correlating with lymph node metastasis;220 and endometrial carcinoma patients (n = 115), upon both uni- and multivariate analyses.221 Tumor infiltration by mDCs (identified as fascin, CD1A, CD83 or CD208-expressing cells) has been linked to improved clinical outcomes in cohorts of diffuse large B-cell lymphoma patients (n = 48), with a particular propensity of DCs to accumulate within nodal, as opposed to extra-nodal, lesions;222 subjects affected by esophageal cancer (n = 67), correlating with the accumulation of T cells around tumor nests;223 breast carcinoma patients (n = 130), a setting in which CD83+ (but not CD1A+) DCs had an independent prognostic relevance;224 subjects affected by NSCLC (n = 74);94,95 melanoma patients (n = 82), again correlating with T-cell infiltration;225 and RCC patients receiving cytokine-based immunotherapy (n = 25).226 Along similar lines, tumor infiltration by Langerhans cells has been associated with prolonged overall survival in cohorts of nasopharyngeal carcinoma (n = 119) and NSCLC (n = 74) patients,95,227 apparently in contrast with the notion that these cells generally behave as iDCs and hence promote tolerance. Conversely, elevated intratumoral levels of CD208+ mDCs have been linked to worsened disease outcomes in cohorts of breast carcinoma patients (n = 152);228 subjects affected by gastric carcinoma (n = 128);229 and CRC patients (n = 104).230 Similar results have been obtained for CD123+ pDCs in patients affected by breast carcinoma (n = 152) and melanoma (n = 186).228,231 In the context of CRC, such a negative correlation between tumor-infiltrating mDCs and clinical outcome is rather expected, in line with the fact that high intratumoral levels of Tregs positively (rather than negatively, as in most other cancers)8 affect CRC prognosis.92,93,131-134 This said, the results obtained by Treilleux et al. and Ishigami et al. on cohorts of breast carcinoma and gastric cancer patients, respectively,228,229 are at odds with several previous studies,161,215-218,224 raising doubts on the actual prognostic/predictive value of DC infiltration in these settings.

When this Trial Watch was being redacted (August 2012), official sources listed no clinical studies that would evaluate as an endpoint the predictive/prognostic value of DC infiltration in cohorts of cancer patients (source www.clinicaltrials.gov).

Macrophages

Macrophages (literally “big eaters,” as from the Greek terms makros “large” and phagein “eat”) are tissue-resident myeloid cells generated by the differentiation of circulating monocytes following extravasation.232 Besides exhibiting a considerable size (Ø = 20–80 μm) and peculiar amoeboid movements, macrophages are characterized by the expression of cell surface markers including, but not limited to: CD14 (the co-receptor for the lipopolysaccharide sensor TLR4), CD16 (FcγRIII), CD31 (also known as PECAM1, a member of the immunoglobulin superfamily involved in phagocytosis), CD40 (a co-stimulatory molecule found on multiple APCs), CD68 (a glycoprotein that binds to low density lipoproteins), CD163 (a member of the scavenger cysteine-rich receptor superfamily involved in the clearance of hemoglobin/haptoglobin complexes), EMR1 (an adhesion molecule, the human ortholog to murine F4/80), lysozime M (a glycoside hydrolase that can damage the bacterial cell wall), MAC-1/CR3 (a CD11b/CD18 heterodimer operating both as a pattern recognition receptor and as a receptor for the complement component iC3b), MAC-3 (a receptor for galectin-3) and the mannose receptor C type 1 (MRC1, also known as CD206).232-234 Thanks to these and other plasma membrane receptors, macrophages exert prominent phagocytic functions, taking up free as well as opsonized particulate material within intracellular vesicles (endosomes) and ensuring its degradation. Being linked to the activation of the so-called “inflammasome,” and hence to the release of pyrogenic IL-1β and IL-18,235 such a phagocytic activity is particularly relevant as a first line of defense against invading pathogens.233 Moreover, it constitutes a major mechanism whereby the organism disposes of apoptotic corpses and necrotic debris.233 Macrophages also operate as bona fide APCs, hence processing internalized antigens and presenting them in the context of MHC class II molecules to elicit adaptive immune responses. Alongside, macrophages secrete cytokines that de facto dictate the type of immune response. Thus, whereas “classically” activated (M1) macrophages secreting IL-12 promote protective Th1 responses, “alternatively” activated (M2) macrophages favor the establishment of tolerance, owing to the secretion of multiple cytokines including IL-10.233,236 Of note, M2 macrophages can be phenotypically discriminated from their M1 counterparts as the former express high levels of CD163 and CD206, while the latter bear on their surface consistent amounts of HLA-DR.237

The first studies attempting to correlate intratumoral macrophages with disease outcome generated heterogeneous results, most likely owing to the fact that infiltration was assessed with anti-CD68 antibodies, which do not allow for the discrimination between M1 and M2 macrophage subsets. Thus, no statistically significant association between high levels of CD68+ cells within neoplastic lesions and clinicopathological parameters including disease course has been detected in cohorts of nasopharyngeal carcinoma (n = 119) and endometrial cancer (n = 109) patients.227,238 Tumor infiltration by CD68+ macrophages has been linked to worsened disease course in cohorts of breast carcinoma patients (n = 249), in relationship with increased microvessel density and VEGF expression;239 HNC patients who underwent surgery for the removal of laryngeal lesions (n = 98);240 patients subjected to potentially curative resection of esophageal cancers (n = 56 and n = 121);241,242 resected gastric cancer patients (n = 97),243 subjects with resected HCC (n = 137);244 lung adenocarcinoma patients (n = 113), again in association with increased local angiogenesis;245 pleural mesothelioma patients who underwent cytoreductive surgery (n = 52), a setting in which also circulating monocytes correlated with poor survival;246 melanoma patients (n = 58, n = 202 and n = 190), high macrophage counts being associated with markers of aggressive disease including Breslow thickness, ulceration and mitotic rate;87,247,248 subjects affected by ovarian carcinoma (n = 67);249 bladder cancer patients receiving intravesical instillations of the bacillus Calmette-Guérin (BCG) (n = 41);250 prostate cancer patients treated with hormonal therapy (n = 71), a setting in which macrophage infiltration was shown to predict resistance to treatment;251 and patients affected by renal pelvis and ureteral transitional cell carcinomas (n = 75).252 Conversely, tumor infiltration by macrophages (most often detected as CD68+ cells) has been correlated with improved disease outcome in cohorts of HNC patients bearing nasopharyngeal lesions (n = 45)212 or treated with IRX-2-based immunotherapy (n = 27);67 patients bearing differentiated thyroid carcinoma (n = 398);45 NSCLC patients who underwent surgical resection (n = 175)253 or receiving chemotherapy for stage IV lesions (n = 199);49 gastric cancer patients (n = 84), highlighting a correlation between intratumoral macrophages, intratumoral CD8+ T cells and tumor cell apoptosis;254 HCC patients who underwent surgical resection (n = 302);96 melanoma patients treated with oral BCG (n = 25);255 CRC patients (n = 97, n = 70, n = 446 and n = 160), overall suggesting a critical role of macrophages at the tumor invasive margin;60,256-258 and patients affected by various solid tumors (including breast carcinoma, melanoma and RCC) subjected to IL-2-based immunotherapy.77 Along similar lines, intense tumor infiltration by CD14+CD40+ macrophages has been shown to constitute an independent prognostic factor in a cohort of CRC patients (n = 31).259

Recently, clinical studies based on M2-specific phenotypic markers, most often CD163, have provided more consistent results. Thus, tumor infiltration by elevated amounts of CD163+ cells have been associated with poor clinical outcome in cohorts of breast carcinoma patients (n = 144), highlighting a critical role for macrophages located within the tumor stroma, but not tumor nests;260 subjects affected by lung adenocarcinoma (n = 65), correlating with lymphatic microvessel density;261 melanoma patients (n = 190), further highlighting the revelance of CD163+ cells within the tumor stroma;248 RCC patients (n = 66), although in this setting the association was statistically significant upon univariate, but not multivariate, analyses;262 primary leiomyosarcoma patients (n = 149);263 as well as in subjects affected by Hodgkin's lymphoma (n = 288), among which both CD68 and CD163 positivity were associated not only with poor overall survival but also with the presence of the Epstein-Barr virus within neoplastic cells.264

When this Trial Watch was being redacted (August 2012), official sources listed three clinical studies that would evaluate the predictive/prognostic value of intratumoral macrophages in cohorts of cancer patients (Table 1). The first one (NCT00690261; current status: recruiting) specifically aims at investigating impact of M1/M2 macrophage polarization on cancer progression and prognosis prediction among lung cancer patients with malignant pleural effusions. The second one (NCT00896922; current status: unknown) intends to evaluate the prognostic significance of CD68+ and CD163+ cells (as well as that of FOXP3+ cells) in follicular lymphoma patients receiving immunochemotherapy. The third one (NCT01493817; current status: active, not recruiting) aims at studying the relationship between tumor-associated macrophages and clinicopathological factors within a large assessment of prognostic/predictive biomarkers in patients affected by Wilms' tumor. (source www.clinicaltrials.gov).

Myeloid-Derived Suppressor Cells

The term MDSCs refers to a heterogeneous population of cells that are defined by their common myeloid origin, relatively immature state and capacity to potently suppress both the innate and the adaptive arms of cellular immunity (i.e., NK-cell, NKT-cell and T-cell responses).265,266 MDSCs are released from the bone marrow in response to a wide array of signals (including cytokines produced by malignant cells and tumor-associated stromal cells as well as mediators secreted by multiple cells following infection or trauma) and de facto derive from monocytic and granulocytic cell progenitors that would normally differentiate into DCs, macrophages, neutrophils, eosinophils or basophils.266-270 Accordingly, at least two distinct MDSC subsets can be identified based on morphological features and cell surface markers. Thus, whereas all human MDSCs express CD11b, CD33 (a myeloid lineage-specific lectin also known as SIGLEC-3) and CD124 (the α chain of the IL-4 receptor), granulocytic MDSCs can be discriminated from their myeloid counterparts as the former, but not the latter, express the VEGF receptor 1 (VEGFR1) and (low levels of) CD16. Conversely, monocytic MDSCs express CD14, low levels of HLA-DR and S100-A9 (a Ca2+-binding protein also known as MRP-14 and calgranulin B) while their granulocytic counterparts fail to do so.266,271-273 MDSCs can suppress the effector functions of NK, NKT and T cells by a variety of mechanisms including a strong conditioning of the local microenvironment that involves the production of reactive oxygen species, reactive nitrogen species and cytokines as well as the less direct cellular circuitries involving Tregs, NK cells and macrophages.274-277 Recently, the role of MDSCs in oncogenesis, tumor progression and response to therapy has begun to be unveiled. In particular, it has been shown that intratumoral MDSCs (which, optionally, can differentiate into tumor-associated macrophages) promote tumor growth not only as they exert immunosuppressive functions but also since they promote angiogenesis/lymphangiogenesis, both at the primary tumor site and at distant pre-metastatic niches.278,279 Of note, the levels of circulating MDSCs are altered in transplanted patients280 as well as in subjects affected by a large spectrum of tumors, often correlating with disease progression.266,273,275,281,282 This suggests that MDSCs play a prominent role in the pathogenesis of cancer.

Low levels of circulating MDSCs have been reported to constitute a positive prognostic/predictive factor in cohorts of untreated diffuse large B-cell lymphoma patients (n = 91), indicating a prominent role for the monocytic, rather than the granulocytic MDSC subset;283 stage II-IIIc breast carcinoma patients receiving neoadjuvant disodium glutathione disulfide-containing chemotherapy (n = 40);284 treatment-naïve, advanced NSCLC patients (n = 87);285 subjects affected by gastrointestinal neoplasms (n = 40), correlating with the circulating levels of IL-6 and IL-10;272 advanced melanoma patients treated with an oncolytic virus encoding GM-CSF (n = 50);125 RCC patients receiving the broad spectrum tyrosine kinase inhibitor sunitinib (n = 23);286 bladder carcinoma patients (n = 64), correlating with pathological grade and clinical stage;287 patients bearing esophageal, gastric or pancreatic neoplasms (n = 131);288 and patients affected by terminal tumors (n = not available).289 Conversely, low levels of MDSCs have been reported in high-risk (n = 3) neuroblastoma patients, as compared with their low-risk counterparts (n = 1), correlating with reduced levels of circulating IL-10.290 However, whether neuroblastoma patients truly benefit from high levels of circulating MDSCs and IL-10 will have to be confirmed in large patient cohorts. Intriguingly, it has been reported that the amount of psychological stress experienced by surgically resected breast carcinoma patients (n = 16) alters the levels of circulating MDSCs. This said, the actual implications of these findings for the therapeutic and psychological management of cancer patients remain to be elucidated.

When this Trial Watch was being redacted (August 2012), official sources listed no clinical studies that would evaluate the predictive/prognostic value of circulating or tumor-infiltrating MDSCs in cohorts of cancer patients (source www.clinicaltrials.gov).

B Cells

B cells are small circulating lymphocytes expressing (at least in the vast majority of cases) a monospecific B-cell receptor (BCR), i.e., a fully rearranged immunoglobulin (most often of the M or D type) inserted in the plasma membrane and associated with a signal transduction machinery involving CD79A and CD79B.291 In all mammals but rabbits, B cells are generated in the bone marrow, where they progressively mature along with the rearrangement of the immunoglobulin-coding genes.292 Bone marrow-resident BCR+ immature B cells express high levels of the IL-7 receptor, a phenotypic marker that they lose as they migrate to secondary lymphoid tissues (e.g., spleen, lymph nodes, Peyer's patches).293 Further surface markers that are routinely used for identifying B cells include CD19, CD20 and CD78, all of which start to be expressed along with the rearrangement of the immunoglobulin heavy chain-coding genes.294 CD19 (which is also expressed by follicular DCs) has been shown to cooperate with CD21 to form a multimeric receptor for several components of the complement system.295 Conversely, the functions of CD20 (which is currently employed as a target for monoclonal antibody-based therapies against B cell neoplasms)22 and CD78 are less characterized, although CD20 appears to required for optimal B-cell responses, in particular against T cell-independent antigens.296

B cells that recognize an antigen via their BCR can internalize, process and present it, complexed with MHC class II molecules, to CD4+ T cells. If the latter react to the same antigen by producing IL-4, B cells expand and mature either into antibody-secreting plasma B cells or into memory B cells. Optionally, prior to reach either of these terminal differentiation stages, B cells can undergo the so-called “class switching,” i.e., a further genetic rearrangement that shifts the production of antibodies from one class to another while maintaining specificity.297 Plasma B cells do not express CD19 and CD20, yet can be identified owing to the presence of CD78, CD38 (a cyclic ADP ribose hydrolase), the receptor for IL-6 and high levels of CD27, a member of the tumor necrosis factor α receptor (TNFR) superfamily.298 Memory B cells are characterized by the expression the BCR, CD19 and CD27, and — similar to their T-cell counterparts — play a critical role in secondary immune responses.299,300

B cells generally resemble T cells also in that they are subjected to multiple control mechanisms for the avoidance of autoimmune responses. Indeed, B cells bearing BCRs that recognize self antigens with high affinity are either driven into apoptotic cell death (clonal deletion), either allowed to rearrange their BCR to generate a new binding specificity (receptor editing), or permanently maintained in an unresponsive status (anergy).301,302 Finally, it should be noted that not all B cells stimulate immune responses. In particular, so-called regulatory B cells (Bregs) respond to antigens by secreting high amounts of IL-10 and TNFα (which inhibit CD8+ CTLs and favor the differentiation of Tregs) and/or by expressing cell death-inducing molecules such as FASL (to which activated B and T cells are particularly sensitive) on their surface, globally exerting potent immunosuppressive effects.303-307

Increased amounts of intratumoral or peritumoral CD20+ B cells (alone or together with other immune cells) have been associated with improved clinical outcome in cohorts of surgically resected HCC patients (n = 163);72 melanoma as well as superficial bladder carcinoma patients receiving BCG-based immunotherapy (n = 30 and n = 20);255,308 prostate carcinoma patients undergoing radical prostatectomy (n = 188), a setting in which T-cell infiltration also provided prognostic insights;75 and optimally debulked high-grade serous epithelial ovarian carcinoma patients (n = 199).83 The accumulation of CD20+ B cells at the invasive margin, at tumor cell nests or within the tumor stroma has been shown to constitute a good prognostic or predictive biomarker in surgically resected NSCLC patients (n = 335);70 in subjects affected by melanoma (n = 106)309 as well as in HNC patients receiving IRX-2-based immunotherapy (n = 27).67 Increased amounts of circulating CD19+ B cells that share clonotypic rearrangements with malignant plasma cells are a relatively common finding among multiple myeloma patients, and have been significantly associated (in a cohort of n = 521 patients) with low (rather than high) stage and longer (rather than shorter) overall survival.310

When this Trial Watch was being redacted (August 2012), official sources listed two clinical studies that would evaluate the predictive/prognostic value of humoral (and cellular) immunity in cohorts of cervical cancer patients (NCT00673192; NCT00854269) (Table 1). This said, the authors of these trials (current status: unknown) do not clearly state whether they intend to quantify intratumoral B cells or whether other, more direct, measures of humoral antitumor immunity will be employed (source www.clinicaltrials.gov).

Other Immune Cells

Neutrophil granulocytes (also known as polymorphonuclear neutrophils, owing to their peculiar multilobulated nuclear morphology) are the most abundant type of white blood cells in mammals.311 The term “neutrophil” derives from the fact that on hematoxylin and eosin (H&E) cytological preparations, these cells appear of a neutral pink (as opposed to other granulocytes, see below).312 In response to chemotactic signals, circulating neutrophils are the first to accumulate at sites of inflammation, where they provide a multipronged contribution to innate immune responses by: (1) secreting cytokines that recruit other immune cells; (2) operating as professional phagocytes, though only toward opsonized material (at odds with DCs and macrophages); (3) releasing pre-formed granules containing a wide spectrum of pro-inflammatory or antimicrobial proteins including myeloperoxidase, bactericidal/permeability-increasing protein (BPI), defensins, lactoferrin, cathelicidin, gelatinase and multiple proteases (e.g., elastase and cathepsins); and (4) liberating so-called neutrophil extracellular traps (NETs), i.e., networks of DNA- and protease-containing fibers that are particularly efficient at capturing and killing extracellular bacteria.311-313

Similar to macrophages, neutrophils intensely infiltrate neoplastic lesions.314,315 However, while the depletion of tumor-associated neutrophils (TANs) normally exacerbates CD8+ CTL-mediated antitumor effects, the contrary holds true in condition in which TGFβ is blocked.315 These observations point to the existence of two functionally distinct subsets of TANs: one with antitumor functions, which is generally repressed by TGFβ (N1 TAN), and one that exerts pro-tumorigenic effects (N2 TANs), which predominates in normal conditions.315 The surface markers that are routinely employed to identify neutrophils include CD11b, the adhesion molecule CD66 (which is also expressed by epithelial and endothelial cells, but not by other immune cells) and Ly6G, a glycosylphosphatidylinisotol-anchored protein of unknown function that can also be found on a subset of eosinophils, differentiating pre-monocytes, and pDCs.316,317 So far, no surface markers that would be useful to differentiate between N1 and N2 neutrophils have been reported. Neutrophilia (i.e., an increase in circulating neutrophils) is a normal finding in response to bacterial infection or during acute inflammation (for instance following extensive burns or heart attacks). In addition, neutrophilia is the most prominent hematological manifestation of multiple hematopoietic cancers including chronic myelogenous leukemia (CML), some cases of which are driven by the hyperproliferation of neutrophil progenitors.318

High levels of intra- or peritumoral neutrophils have been associated with worsened disease outcomes in cohorts of HCC (n = 238)319 and melanoma (n = 186)231 patients, as well as among subjects affected by renal pelvis and ureteral transitional cell carcinoma (n = 75).252 Along similar lines, tumor infiltration by neutrophils has been shown to predict dismal prognosis in a first cohort of surgically resected advanced gastric carcinoma patients (n = 212),320 but to constitute a positive prognostic factor (for women only) in a second clinical setting (at least apparently) of the same type (n = 273).321 This (perhaps only apparent) discrepancy will be clarified by the results of future clinical trials.

Eosinophil granulocytes (also known as polymorphonuclear eosinophils or acidophil granulocytes) account for about 1–6% white blood cells in mammals. The term “eosinophil” (literally “loving eosin,” from Greek) stems from the fact that on H&E cytological preparations, these cells acquire the intense brick red staining.322,323 Eosinophils, which are generated by myeloid precursors in the presence of IL-3, IL-5 and GM-CSF, not only play a prominent role against helminthic and viral infections, but also are involved in the pathogenesis of asthma and allergic responses, as they express (in an inducible fashion) high-affinity receptors for the Fc domain of type E immunoglobulins (FcεRI).322,323 Activated eosinophils can indeed secrete a wide array of cytokines (e.g., IL-2, IL-4, IL-5, IL-8 and IL-13), growth factors (e.g., TGFβ, VEGF), enzymes (e.g., elastase), lipid mediators (e.g., leukotrienes, prostaglandins) and ROS.322,323 In addition, eosinophils resemble neutrophils in that they can respond to stimulation by releasing pre-formed granules that contain potentially harmful proteins such as major basic protein (MBP), eosinophil cationic protein (ECP), eosinophil peroxidase (EPO) and eosinophil-derived neurotoxin (EDN).323,324 Activated eosinophils often express on their surface Ly6G, CD9 and CD44 (two cell-surface glycoproteins involved in cell-to-cell interactions, cell adhesion and migration), as well as CD69 (a lectin also expressed by activated T and NK cells).325,326 In addition, eosinophils can be detected (upon permeabilization) with antibodies targeting MBP and other granule proteins.327 Although eosinophils have been proposed to sustain tumor angiogenesis328 and eosinophilia (i.e., an increase in circulating eosinophils) is a relatively common finding in some types of cancer, the precise role that these cells play in oncogenesis, tumor progression and response to therapy has not yet been fully elucidated.322,323,328

Tumor infiltration by eosinophils has been reported to convey no prognostic/predictive information in two distinct cohorts of HNC patients (n = 248 and n = 76),329,330 a finding that was not confirmed by other studies. Indeed, high levels of intratumoral eosinophils have been associated with poor prognosis in two additional cohorts of HNC patients (n = 31 and n = 87),331,332 but with improved disease outcome in a third HNC patient group (n = 25), yet only among subjects bearing epidermal growth factor receptor (EGFR)+ lesions.333 Along similar lines, tumor infiltration by eosinophils has been reported to constitute a positive prognostic indicator in cohorts of gastric carcinoma patients (n = 647 and n = 324),334,335 glioma patients receiving a combinatorial immunotherapeutic regimen (n = 28),336 subjects affected by lung adenocarcinoma (n = not available),337 CRC patients (n = 126),338 and subjects affected by cervical carcinoma treated with radiotherapy (n = 14).339

Basophil granulocytes constitute the least common type of granulocytes, representing less than 1% circulating white blood cells. The term “basophil” (literally “loving basicity,” from Greek) reflects the fact that these cells are particularly prone to take up basic dyes, acquiring a dark purple/blue staining on H&E cytological preparations.340,341 Similar to eosinophils, basophils play a prominent role in the innate immune response against parasites, such as ticks, and contribute to the development of allergic reactions, owing to the fact that they constitutively express high levels of FcεRI.340,342 On activation, basophils release pre-formed granules that contain large amounts of heparin (an anticoagulant), histamine (a vasodilator) and lytic enzymes (e.g., elastase and lysophospholipase), as well as newly synthesized lipids (e.g., leukotrienes) and cytokines (e.g., IL-4). Altogether, these mediators increase the local blood flow, favor the recruitment of additional immune cells and preferentially drive the production of type E immunoglobulins, de facto contributing to the development of hypersensitivity.340,342-344 Besides FcεRI, basophils express CD69, CD123, CD49 (the α subunit of the heterodimeric integrin α2β1) and TLR4, but neither CD19 nor other relatively common markers of myeloid cells.340,341 Basophilia (i.e., an increase in circulating basophils) is relatively rare finding that can be observed in some cases of leukemia and lymphoma.345 This said, whether basophils modulate oncogenesis, tumor progression and response to therapy remains an open conundrum. Indeed, several studies demonstrate that basophilia is a negative prognostic factor for CML patients.346-348 Still, in this specific setting, the accumulation of circulating basophils is a manifestation of the disease rather than of an antitumor immune reaction.

Even though they derive from different myeloid progenitors, mast cells (also known as mastocytes) are often considered the tissue-resident counterparts of basophils, for multiple reasons.349 In particular, mast cells resemble basophils in that they express high levels of FcεRI and respond to activation stimuli by releasing IL-4, leukotrienes as well as pre-formed granules containing heparin and histamine.349-351 Similar to basophils, mastocytes (in particular those localized at mucosal surfaces) exert potent antiparasitic functions352 and have been involved in the pathogenesis of allergic reactions and autoimmune diseases.351 In addition, mast cells are known to promote wound healing and angiogenesis as well as to secrete a wide array of mediators that can profoundly influence immune responses including IL-10, TNFα and TGFβ.328,353,354 In line with this rather heterogeneous functional profile, mast cells have been proposed to exert either pro- or anti-tumor functions, depending on a variety of parameters including tumor type, presence of additional mediators secreted by other cells and (perhaps with a major influence) intratumoral vs. peritumoral localization.355,356 The surface of mastocytes presents high amounts of the IL-3 receptor as well as of the tyrosine kinase receptor KIT, which is also expressed by variety of non-hematopoietic cell types, including breast epithelial cells, germ cells and melanocytes.357,358 Moreover, on tissue sections subjected to immunohistochemistry, mast cells can be detected with antibodies that recognize members of the chymase family of serine proteases.359 As a note, murine mast cells also express high levels of CD34, a cell-surface sialomucin expressed by hematopoietic stem cells and vascular endothelial cells that regulate homotypic and heterotypic cell-to-cell interactions.357,360

Robust tumor infiltration by mast cells has been linked to worsened disease outcomes in cohorts of gastric carcinoma (n = 102),361 lung adenocarcinoma (n = 180),362 pancreatic cancer (n = 53 and n = 67),363,364 and RCC (n = 71) patients,365 as well as in subjects bearing renal pelvis and ureteral transitional cell carcinomas (n = 75),252 near-to-invariably correlating with increased tumor angiogenesis and metastatic invasion. Conversely, high intratumoral levels of mastocytes have been reported to constitute a positive prognostic factor in cohorts of breast carcinoma (n = 4444),366 HNC (n = 50),367 NSCLC (n = 175),253 CRC (n = 160)41 and ovarian carcinoma (n = 44)368 patients. Whether this apparent discrepancy reflects the differential impact of angiogenesis (which mast cells potently induce) on distinct types of cancer remains an unexplored possibility.

When this Trial Watch was being redacted (August 2012), official sources listed no clinical studies that would evaluate as an endpoint the predictive/prognostic value of tumor-infiltrating and/or circulating neutrophil, eosinophils, basophils and mast cells in cohorts of cancer patients (source www.clinicaltrials.gov).

Concluding Remarks

The results of the clinical studies discussed in this Trial Watch strongly support the contention that the abundance of specific TILs can be translated into reliable prognostic information. Although the number of trials that have specifically addressed this question is relatively low, encouraging results have been obtained also in support of the use of TILs as predictive biomarkers. Of note, at least in some instances, scores based on the type, density and localization of specific TIL populations have been shown to convey a prognostic value that is independent from and superior to that of conventional classifications such as the Duke’s stage and the UICC-TNM system.85,89,90 These observations, which for the most part stem from the pioneer work of Jerome Galon and collaborators,85,89-91,102,103 strongly suggest that immune scores should be integrated into the clinical practice to ameliorate risk stratification and hence aid in therapy-related decision making.18,369 As the immunophenotyping of TILs may provide novel prognostic/predictive information that is likely to influence the therapeutic management of cancer patients, a worldwide task force (including 22 international cancer centers) has recently been instituted for validating a quantitative standardized immune score in routine clinical settings.369

One emerging concept in this respect relates to the prognostic and/or predictive value of the exact localization of TILs within and around neoplastic lesions. Infiltration is indeed rather heterogeneous and distinct TIL types accumulate with differential kinetics at the tumor center, at its invasive margin and within tertiary lymphoid structures (TLSs) that are located in the proximity of the lesion.16,18,94 For instance, granulocytes, macrophages, mast cells and MDSCs often penetrate both tumor cores as well as invasive margins. Conversely, NK cells mainly localize within the tumor stroma, not engaging in direct physical interactions with malignant cells. CD8+ T cells preferentially accumulate at invasive margins, while their naïve and memory counterparts are found within TLSs, which also contain a large number of B cells and mDCs. On the contrary, iDCs are found within the tumor core, both in close contact with neoplastic cells and scattered within stromal components.7,370 A few reports have already provided evidence supporting to the notion that the accumulation of a specific TIL population (e.g., CD83+ DCs) at a specific site (e.g., the invasive margin) conveys prognostic information while the recruitment of the same cells to another location does not.371 Thus, the clinical implementation of an immune score will have to take into account not only the differential predictive and/or prognostic value of specific TILs, but also that of their intratumoral and/or peritumoral localization. This is further complicated by the fact that the infiltration of immune cells varies quite significantly across cancer types, indicating that the immune system reacts in a relatively specific fashion to distinct neoplasms.7,372

Another issue that must be taken into attentive consideration for the clinical implementation of immune infiltrate-based prognostic and/or predictive biomarkers reflects two aspects: (1) the methods whereby TILs are quantified, and (2) our hitherto insufficient knowledge on the phenotypic and functional biology of these cells. Both points are well exemplified by the fact that pioneer studies attempting to evaluate the relevance of intratumoral T cells for disease outcome relied on the immunohistochemical detection of CD3, an invariable component of the TCR signaling complex.373 Obviously, beyond the intrinsic limitations of immunohistochemistry (which until recently involved an obligate step of visual inspections that, by definition, is prone to operator-dependent bias), such an approach was entirely unreliable in that it quantified a plethora of functionally distinct CD3+ cells, encompassing tumor-reactive CD8+, CD4+ Th1 and Th17 T cells as well as immunosuppressive CD4+FOXP3+ and CD4+ Th2 cells.373 In addition to such relatively gross issues, which are increasingly being resolved along with the discovery of ever more specific surface markers, recent observations indicate that the phenotypic markers that are routinely employed to determine the functional profile of TILs may also be (at least in part) unreliable. Thus, it has recently been shown that a fraction of FOXP3+ cells is not committed to immunosuppressive functions but retain developmental plasticity, being able to differentiate into effector Th cells, in vitro.374 Along similar lines, some bona fide immunosuppressive FOXP3+ Tregs have been demonstrated to lose FOXP3 expression while retaining their functional profile.375 Thus, further investigation is warranted to gain deeper insights into the phenotypic and functional profiles of TILs, in turn allowing for the development of detection methods that better reflect the actual role of the immune infiltrate in antitumor responses.

Finally, a mention should be given to immune parameters other than the immune infiltrate that have been shown to influence disease outcome and/or response to therapy in cancer patients. This very large and heterogeneous group of biomarkers includes (but is not limited to) the levels of tumor associated antigen-specific antibodies,376,377 single-nucleotide polymorphisms in genes coding for factors involved in innate or adaptive immunity such as TLR4 and the purinergic receptor P2RX7,378-382 the abundance of specific immune cell populations (e.g., MDSCs) in the bloodstream,282,383,384 the ability of specific immune cell populations (e.g., NK cells) to maintain an elevated production of antitumor cytokines (e.g., IFNγ) in the course of therapy,165 as well as the expression profile of specific receptors (e.g., NKp30) on the surface of immune cell subsets (e.g., NK cells).164 The detailed discussion of these biomarkers largely exceeds the scope of this Trial Watch and can be found in refs. 8 and 15 This said, it can be speculated that the prognostic/predictive information conveyed by the abundance, type and localization of TILs (which mainly reflect local immune responses) might be further ameliorated, at least in some instances, by the development of an immune score that would also take into account systemic biomarkers. This possibility will have to be addressed in appropriate clinical settings.

Acknowledgments

Authors are supported by the Ligue contre le Cancer (équipes labelisées), AXA Chair for Longevity Research, Cancéropôle Ile-de-France, Institut National du Cancer (INCa), Fondation Bettencourt-Schueller, Fondation de France, Fondation pour la Recherche Médicale, Agence National de la Recherche, the European Commission (Apo-Sys, ArtForce, ChemoRes. Death-Train) and the LabEx Immuno-Oncology.

Glossary

Abbreviations:

APC

antigen-presenting cell

BCG

bacillus Calmette-Guérin

BCR

B-cell receptor

BPI

bactericidal/permeability-increasing protein

Breg

regulatory B cell

CAF

cancer-associated fibroblasts

CML

chronic myelogenous leukemia

CRC

colorectal carcinoma

CTL

cytotoxic T lymphocyte

DAMP

damage-associated molecular pattern

DC

dendritic cell

ECP

eosinophil cationic protein

EDN

eosinophil-derived neurotoxin

EGFR

epidermal growth factor receptor

EPO

eosinophil peroxidase

FcεRI

high-affinity type E immunoglobulin receptor

GM-CSF

granulocyte macrophage colony-stimulating factor

HCC

hepatocellular carcinoma

HNC

head and neck carcinoma

iDC

immature DC

IFN

interferon

IL

interleukin

iNKT

invariant NKT

mDC

mature DC

MAMP

microbe-associated molecular pattern

MBP

major basic protein

MDSC

myeloid-derived suppressor cell

MRC1

mannose receptor C type 1

NET

neutrophil extracellular trap

NK

natural killer

NKT

natural killer T

NSCLC

non-small cell lung carcinoma

pDC

plasmacytoid DC

RCC

renal cell carcinoma

TAN

tumor-associated neutrophil

TCR

T-cell receptor

TGFβ

transforming growth factor β

Th

T-helper

TIL

tumor-infiltrating leukocyte

Treg

CD4+CD25+FOXP3+ regulatory T cell

TLR

Toll-like receptor

TLS

tertiary lymphoid structure

TNM

tumor, node, metastasis

UICC

Union for International Cancer Control

VEGF

vascular endothelial growth factor

VEGFR1

VEGF receptor 1

Footnotes

References

  • 1.Galluzzi L, Vitale I, Kroemer G. Past, present, and future of molecular and cellular oncology. Front Oncol. 2011;1:1. doi: 10.3389/fonc.2011.00001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Nowell PC. The clonal evolution of tumor cell populations. Science. 1976;194:23–8. doi: 10.1126/science.959840. [DOI] [PubMed] [Google Scholar]
  • 3.Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11. doi: 10.1038/35102167. [DOI] [PubMed] [Google Scholar]
  • 4.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 5.Albini A, Sporn MB. The tumour microenvironment as a target for chemoprevention. Nat Rev Cancer. 2007;7:139–47. doi: 10.1038/nrc2067. [DOI] [PubMed] [Google Scholar]
  • 6.Flight MH. Drug screening: shedding light on tumour-stroma interactions. Nat Rev Drug Discov. 2010;9:360–1. doi: 10.1038/nrd3163. [DOI] [PubMed] [Google Scholar]
  • 7.Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer. 2012;12:298–306. doi: 10.1038/nrc3245. [DOI] [PubMed] [Google Scholar]
  • 8.Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov. 2012;11:215–33. doi: 10.1038/nrd3626. [DOI] [PubMed] [Google Scholar]
  • 9.Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011;473:298–307. doi: 10.1038/nature10144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Potente M, Gerhardt H, Carmeliet P. Basic and therapeutic aspects of angiogenesis. Cell. 2011;146:873–87. doi: 10.1016/j.cell.2011.08.039. [DOI] [PubMed] [Google Scholar]
  • 11.Galluzzi L, Kepp O, Kroemer G. Reverse Warburg: straight to cancer. Cell Cycle. 2012;11:1059. doi: 10.4161/cc.11.6.19746. [DOI] [PubMed] [Google Scholar]
  • 12.Witkiewicz AK, Whitaker-Menezes D, Dasgupta A, Philp NJ, Lin Z, Gandara R, et al. Using the “reverse Warburg effect” to identify high-risk breast cancer patients: stromal MCT4 predicts poor clinical outcome in triple-negative breast cancers. Cell Cycle. 2012;11:1108–17. doi: 10.4161/cc.11.6.19530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Räsänen K, Vaheri A. Activation of fibroblasts in cancer stroma. Exp Cell Res. 2010;316:2713–22. doi: 10.1016/j.yexcr.2010.04.032. [DOI] [PubMed] [Google Scholar]
  • 14.Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol. 2007;25:267–96. doi: 10.1146/annurev.immunol.25.022106.141609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zitvogel L, Kepp O, Kroemer G. Immune parameters affecting the efficacy of chemotherapeutic regimens. Nat Rev Clin Oncol. 2011;8:151–60. doi: 10.1038/nrclinonc.2010.223. [DOI] [PubMed] [Google Scholar]
  • 16.Fridman WH, Galon J, Pagès F, Tartour E, Sautès-Fridman C, Kroemer G. Prognostic and predictive impact of intra- and peritumoral immune infiltrates. Cancer Res. 2011;71:5601–5. doi: 10.1158/0008-5472.CAN-11-1316. [DOI] [PubMed] [Google Scholar]
  • 17.Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331:1565–70. doi: 10.1126/science.1203486. [DOI] [PubMed] [Google Scholar]
  • 18.Pagès F, Galon J, Dieu-Nosjean MC, Tartour E, Sautès-Fridman C, Fridman WH. Immune infiltration in human tumors: a prognostic factor that should not be ignored. Oncogene. 2010;29:1093–102. doi: 10.1038/onc.2009.416. [DOI] [PubMed] [Google Scholar]
  • 19.Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, et al. Trial Watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology. 2012;1 doi: 10.4161/onci.21494. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: Adoptive cell transfer immunotherapy. Oncoimmunology. 2012;1:306–15. doi: 10.4161/onci.19549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2012;1:699–716. doi: 10.4161/onci.20696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunology. 2012;1:28–37. doi: 10.4161/onci.1.1.17938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: FDA-approved Toll-like receptor agonists for cancer therapy. OncoImmunology. 2012;1 doi: 10.4161/onci.20931. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Vacchelli E, Galluzzi L, Eggermont A, Galon J, Tartour E, Zitvogel L, et al. Trial Watch: Immunostimulatory cytokines. Oncoimmunology. 2012;1:493–506. doi: 10.4161/onci.20459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Vacchelli E, Galluzzi L, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology. 2012;1:179–88. doi: 10.4161/onci.1.2.19026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Palmer E. Negative selection--clearing out the bad apples from the T-cell repertoire. Nat Rev Immunol. 2003;3:383–91. doi: 10.1038/nri1085. [DOI] [PubMed] [Google Scholar]
  • 27.Anderton SM, Wraith DC. Selection and fine-tuning of the autoimmune T-cell repertoire. Nat Rev Immunol. 2002;2:487–98. doi: 10.1038/nri842. [DOI] [PubMed] [Google Scholar]
  • 28.Bonneville M, O’Brien RL, Born WK. Gammadelta T cell effector functions: a blend of innate programming and acquired plasticity. Nat Rev Immunol. 2010;10:467–78. doi: 10.1038/nri2781. [DOI] [PubMed] [Google Scholar]
  • 29.Singer A, Adoro S, Park JH. Lineage fate and intense debate: myths, models and mechanisms of CD4- versus CD8-lineage choice. Nat Rev Immunol. 2008;8:788–801. doi: 10.1038/nri2416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kalinski P, Moser M. Consensual immunity: success-driven development of T-helper-1 and T-helper-2 responses. Nat Rev Immunol. 2005;5:251–60. doi: 10.1038/nri1569. [DOI] [PubMed] [Google Scholar]
  • 31.Zou W, Restifo NPT. T(H)17 cells in tumour immunity and immunotherapy. Nat Rev Immunol. 2010;10:248–56. doi: 10.1038/nri2742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ma Y, Aymeric L, Locher C, Mattarollo SR, Delahaye NF, Pereira P, et al. Contribution of IL-17-producing gamma delta T cells to the efficacy of anticancer chemotherapy. J Exp Med. 2011;208:491–503. doi: 10.1084/jem.20100269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science. 2011;334:1573–7. doi: 10.1126/science.1208347. [DOI] [PubMed] [Google Scholar]
  • 34.Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol. 2010;10:490–500. doi: 10.1038/nri2785. [DOI] [PubMed] [Google Scholar]
  • 35.Lefrançois L. Development, trafficking, and function of memory T-cell subsets. Immunol Rev. 2006;211:93–103. doi: 10.1111/j.0105-2896.2006.00393.x. [DOI] [PubMed] [Google Scholar]
  • 36.Lefrançois L. Dual personality of memory T cells. Trends Immunol. 2002;23:226–8. doi: 10.1016/S1471-4906(02)02190-7. [DOI] [PubMed] [Google Scholar]
  • 37.Acuto O, Michel F. CD28-mediated co-stimulation: a quantitative support for TCR signalling. Nat Rev Immunol. 2003;3:939–51. doi: 10.1038/nri1248. [DOI] [PubMed] [Google Scholar]
  • 38.Lenschow DJ, Walunas TL, Bluestone JA. CD28/B7 system of T cell costimulation. Annu Rev Immunol. 1996;14:233–58. doi: 10.1146/annurev.immunol.14.1.233. [DOI] [PubMed] [Google Scholar]
  • 39.Demaria S, Volm MD, Shapiro RL, Yee HT, Oratz R, Formenti SC, et al. Development of tumor-infiltrating lymphocytes in breast cancer after neoadjuvant paclitaxel chemotherapy. Clin Cancer Res. 2001;7:3025–30. [PubMed] [Google Scholar]
  • 40.Yin XY, Lu MD, Lai YR, Liang LJ, Huang JF. Prognostic significances of tumor-infiltrating S-100 positive dendritic cells and lymphocytes in patients with hepatocellular carcinoma. Hepatogastroenterology. 2003;50:1281–4. [PubMed] [Google Scholar]
  • 41.Nagtegaal ID, Marijnen CA, Kranenbarg EK, Mulder-Stapel A, Hermans J, van de Velde CJ, et al. Local and distant recurrences in rectal cancer patients are predicted by the nonspecific immune response; specific immune response has only a systemic effect--a histopathological and immunohistochemical study. BMC Cancer. 2001;1:7. doi: 10.1186/1471-2407-1-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Belldegrun A, Tso CL, Zisman A, Naitoh J, Said J, Pantuck AJ, et al. Interleukin 2 gene therapy for prostate cancer: phase I clinical trial and basic biology. Hum Gene Ther. 2001;12:883–92. doi: 10.1089/104303401750195854. [DOI] [PubMed] [Google Scholar]
  • 43.Richardsen E, Uglehus RD, Due J, Busch C, Busund LT. The prognostic impact of M-CSF, CSF-1 receptor, CD68 and CD3 in prostatic carcinoma. Histopathology. 2008;53:30–8. doi: 10.1111/j.1365-2559.2008.03058.x. [DOI] [PubMed] [Google Scholar]
  • 44.Mahmoud SM, Paish EC, Powe DG, Macmillan RD, Grainge MJ, Lee AH, et al. Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Clin Oncol. 2011;29:1949–55. doi: 10.1200/JCO.2010.30.5037. [DOI] [PubMed] [Google Scholar]
  • 45.Cunha LL, Morari EC, Guihen AC, Razolli D, Gerhard R, Nonogaki S, et al. Infiltration of a mixture of immune cells may be related to good prognosis in patients with differentiated thyroid carcinoma. Clin Endocrinol (Oxf) 2012 doi: 10.1111/j.1365-2265.2012.04482.x. In press. [DOI] [PubMed] [Google Scholar]
  • 46.Reichert TE, Scheuer C, Day R, Wagner W, Whiteside TL. The number of intratumoral dendritic cells and zeta-chain expression in T cells as prognostic and survival biomarkers in patients with oral carcinoma. Cancer. 2001;91:2136–47. doi: 10.1002/1097-0142(20010601)91:11<2136::AID-CNCR1242>3.0.CO;2-Q. [DOI] [PubMed] [Google Scholar]
  • 47.Prins RM, Soto H, Konkankit V, Odesa SK, Eskin A, Yong WH, et al. Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clin Cancer Res. 2011;17:1603–15. doi: 10.1158/1078-0432.CCR-10-2563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Schumacher K, Haensch W, Röefzaad C, Schlag PM. Prognostic significance of activated CD8(+) T cell infiltrations within esophageal carcinomas. Cancer Res. 2001;61:3932–6. [PubMed] [Google Scholar]
  • 49.Kawai O, Ishii G, Kubota K, Murata Y, Naito Y, Mizuno T, et al. Predominant infiltration of macrophages and CD8(+) T Cells in cancer nests is a significant predictor of survival in stage IV nonsmall cell lung cancer. Cancer. 2008;113:1387–95. doi: 10.1002/cncr.23712. [DOI] [PubMed] [Google Scholar]
  • 50.Clark WH, Jr., Elder DE, Guerry D, 4th, Braitman LE, Trock BJ, Schultz D, et al. Model predicting survival in stage I melanoma based on tumor progression. J Natl Cancer Inst. 1989;81:1893–904. doi: 10.1093/jnci/81.24.1893. [DOI] [PubMed] [Google Scholar]
  • 51.Clemente CG, Mihm MC, Jr., Bufalino R, Zurrida S, Collini P, Cascinelli N. Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer. 1996;77:1303–10. doi: 10.1002/(SICI)1097-0142(19960401)77:7<1303::AID-CNCR12>3.0.CO;2-5. [DOI] [PubMed] [Google Scholar]
  • 52.Cai XY, Gao Q, Qiu SJ, Ye SL, Wu ZQ, Fan J, et al. Dendritic cell infiltration and prognosis of human hepatocellular carcinoma. J Cancer Res Clin Oncol. 2006;132:293–301. doi: 10.1007/s00432-006-0075-y. [DOI] [PubMed] [Google Scholar]
  • 53.Paulson KG, Iyer JG, Tegeder AR, Thibodeau R, Schelter J, Koba S, et al. Transcriptome-wide studies of merkel cell carcinoma and validation of intratumoral CD8+ lymphocyte invasion as an independent predictor of survival. J Clin Oncol. 2011;29:1539–46. doi: 10.1200/JCO.2010.30.6308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Diederichsen AC, Hjelmborg JB, Christensen PB, Zeuthen J, Fenger C. Prognostic value of the CD4+/CD8+ ratio of tumour infiltrating lymphocytes in colorectal cancer and HLA-DR expression on tumour cells. Cancer Immunol Immunother. 2003;52:423–8. doi: 10.1007/s00262-003-0388-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Jass JR. Lymphocytic infiltration and survival in rectal cancer. J Clin Pathol. 1986;39:585–9. doi: 10.1136/jcp.39.6.585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lugli A, Karamitopoulou E, Panayiotides I, Karakitsos P, Rallis G, Peros G, et al. CD8+ lymphocytes/ tumour-budding index: an independent prognostic factor representing a ‘pro-/anti-tumour’ approach to tumour host interaction in colorectal cancer. Br J Cancer. 2009;101:1382–92. doi: 10.1038/sj.bjc.6605318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Menon AG, Janssen-van Rhijn CM, Morreau H, Putter H, Tollenaar RA, van de Velde CJ, et al. Immune system and prognosis in colorectal cancer: a detailed immunohistochemical analysis. Lab Invest. 2004;84:493–501. doi: 10.1038/labinvest.3700055. [DOI] [PubMed] [Google Scholar]
  • 58.Prall F, Dührkop T, Weirich V, Ostwald C, Lenz P, Nizze H, et al. Prognostic role of CD8+ tumor-infiltrating lymphocytes in stage III colorectal cancer with and without microsatellite instability. Hum Pathol. 2004;35:808–16. doi: 10.1016/j.humpath.2004.01.022. [DOI] [PubMed] [Google Scholar]
  • 59.Ropponen KM, Eskelinen MJ, Lipponen PK, Alhava E, Kosma VM. Prognostic value of tumour-infiltrating lymphocytes (TILs) in colorectal cancer. J Pathol. 1997;182:318–24. doi: 10.1002/(SICI)1096-9896(199707)182:3<318::AID-PATH862>3.0.CO;2-6. [DOI] [PubMed] [Google Scholar]
  • 60.Funada Y, Noguchi T, Kikuchi R, Takeno S, Uchida Y, Gabbert HE. Prognostic significance of CD8+ T cell and macrophage peritumoral infiltration in colorectal cancer. Oncol Rep. 2003;10:309–13. [PubMed] [Google Scholar]
  • 61.Sinicrope FA, Rego RL, Ansell SM, Knutson KL, Foster NR, Sargent DJ. Intraepithelial effector (CD3+)/regulatory (FoxP3+) T-cell ratio predicts a clinical outcome of human colon carcinoma. Gastroenterology. 2009;137:1270–9. doi: 10.1053/j.gastro.2009.06.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, et al. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci U S A. 2007;104:3360–5. doi: 10.1073/pnas.0611533104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Vesalainen S, Lipponen P, Talja M, Syrjänen K. Histological grade, perineural infiltration, tumour-infiltrating lymphocytes and apoptosis as determinants of long-term prognosis in prostatic adenocarcinoma. Eur J Cancer. 1994;30A:1797–803. doi: 10.1016/0959-8049(94)E0159-2. [DOI] [PubMed] [Google Scholar]
  • 64.Sharma P, Shen Y, Wen S, Yamada S, Jungbluth AA, Gnjatic S, et al. CD8 tumor-infiltrating lymphocytes are predictive of survival in muscle-invasive urothelial carcinoma. Proc Natl Acad Sci U S A. 2007;104:3967–72. doi: 10.1073/pnas.0611618104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Barrera JL, Verastegui E, Meneses A, Zinser J, de la Garza J, Hadden JW. Combination immunotherapy of squamous cell carcinoma of the head and neck: a phase 2 trial. Arch Otolaryngol Head Neck Surg. 2000;126:345–51. doi: 10.1001/archotol.126.3.345. [DOI] [PubMed] [Google Scholar]
  • 66.Hadden J, Verastegui E, Barrera JL, Kurman M, Meneses A, Zinser JW, et al. A trial of IRX-2 in patients with squamous cell carcinomas of the head and neck. Int Immunopharmacol. 2003;3:1073–81. doi: 10.1016/S1567-5769(03)00029-8. [DOI] [PubMed] [Google Scholar]
  • 67.Berinstein NL, Wolf GT, Naylor PH, Baltzer L, Egan JE, Brandwein HJ, et al. Increased lymphocyte infiltration in patients with head and neck cancer treated with the IRX-2 immunotherapy regimen. Cancer Immunol Immunother. 2012;61:771–82. doi: 10.1007/s00262-011-1134-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Cho Y, Miyamoto M, Kato K, Fukunaga A, Shichinohe T, Kawarada Y, et al. CD4+ and CD8+ T cells cooperate to improve prognosis of patients with esophageal squamous cell carcinoma. Cancer Res. 2003;63:1555–9. [PubMed] [Google Scholar]
  • 69.Lv L, Pan K, Li XD, She KL, Zhao JJ, Wang W, et al. The accumulation and prognosis value of tumor infiltrating IL-17 producing cells in esophageal squamous cell carcinoma. PLoS One. 2011;6:e18219. doi: 10.1371/journal.pone.0018219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Al-Shibli KI, Donnem T, Al-Saad S, Persson M, Bremnes RM, Busund LT. Prognostic effect of epithelial and stromal lymphocyte infiltration in non-small cell lung cancer. Clin Cancer Res. 2008;14:5220–7. doi: 10.1158/1078-0432.CCR-08-0133. [DOI] [PubMed] [Google Scholar]
  • 71.Tefany FJ, Barnetson RS, Halliday GM, McCarthy SW, McCarthy WH. Immunocytochemical analysis of the cellular infiltrate in primary regressing and non-regressing malignant melanoma. J Invest Dermatol. 1991;97:197–202. doi: 10.1111/1523-1747.ep12479662. [DOI] [PubMed] [Google Scholar]
  • 72.Wada Y, Nakashima O, Kutami R, Yamamoto O, Kojiro M. Clinicopathological study on hepatocellular carcinoma with lymphocytic infiltration. Hepatology. 1998;27:407–14. doi: 10.1002/hep.510270214. [DOI] [PubMed] [Google Scholar]
  • 73.Fukunaga A, Miyamoto M, Cho Y, Murakami S, Kawarada Y, Oshikiri T, et al. CD8+ tumor-infiltrating lymphocytes together with CD4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma. Pancreas. 2004;28:e26–31. doi: 10.1097/00006676-200401000-00023. [DOI] [PubMed] [Google Scholar]
  • 74.Nakakubo Y, Miyamoto M, Cho Y, Hida Y, Oshikiri T, Suzuoki M, et al. Clinical significance of immune cell infiltration within gallbladder cancer. Br J Cancer. 2003;89:1736–42. doi: 10.1038/sj.bjc.6601331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Kärjä V, Aaltomaa S, Lipponen P, Isotalo T, Talja M, Mokka R. Tumour-infiltrating lymphocytes: A prognostic factor of PSA-free survival in patients with local prostate carcinoma treated by radical prostatectomy. Anticancer Res. 2005;25(6C):4435–8. [PubMed] [Google Scholar]
  • 76.Daayana S, Elkord E, Winters U, Pawlita M, Roden R, Stern PL, et al. Phase II trial of imiquimod and HPV therapeutic vaccination in patients with vulval intraepithelial neoplasia. Br J Cancer. 2010;102:1129–36. doi: 10.1038/sj.bjc.6605611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Rubin JT, Elwood LJ, Rosenberg SA, Lotze MT. Immunohistochemical correlates of response to recombinant interleukin-2-based immunotherapy in humans. Cancer Res. 1989;49:7086–92. [PubMed] [Google Scholar]
  • 78.Lu P, Zhu XQ, Xu ZL, Zhou Q, Zhang J, Wu F. Increased infiltration of activated tumor-infiltrating lymphocytes after high intensity focused ultrasound ablation of human breast cancer. Surgery. 2009;145:286–93. doi: 10.1016/j.surg.2008.10.010. [DOI] [PubMed] [Google Scholar]
  • 79.Naito Y, Saito K, Shiiba K, Ohuchi A, Saigenji K, Nagura H, et al. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res. 1998;58:3491–4. [PubMed] [Google Scholar]
  • 80.Pölcher M, Braun M, Friedrichs N, Rudlowski C, Bercht E, Fimmers R, et al. Foxp3(+) cell infiltration and granzyme B(+)/Foxp3(+) cell ratio are associated with outcome in neoadjuvant chemotherapy-treated ovarian carcinoma. Cancer Immunol Immunother. 2010;59:909–19. doi: 10.1007/s00262-010-0817-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Grabenbauer GG, Lahmer G, Distel L, Niedobitek G. Tumor-infiltrating cytotoxic T cells but not regulatory T cells predict outcome in anal squamous cell carcinoma. Clin Cancer Res. 2006;12:3355–60. doi: 10.1158/1078-0432.CCR-05-2434. [DOI] [PubMed] [Google Scholar]
  • 82.Hiraoka N, Onozato K, Kosuge T, Hirohashi S. Prevalence of FOXP3+ regulatory T cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions. Clin Cancer Res. 2006;12:5423–34. doi: 10.1158/1078-0432.CCR-06-0369. [DOI] [PubMed] [Google Scholar]
  • 83.Milne K, Köbel M, Kalloger SE, Barnes RO, Gao D, Gilks CB, et al. Systematic analysis of immune infiltrates in high-grade serous ovarian cancer reveals CD20, FoxP3 and TIA-1 as positive prognostic factors. PLoS One. 2009;4:e6412. doi: 10.1371/journal.pone.0006412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Nakano O, Sato M, Naito Y, Suzuki K, Orikasa S, Aizawa M, et al. Proliferative activity of intratumoral CD8(+) T-lymphocytes as a prognostic factor in human renal cell carcinoma: clinicopathologic demonstration of antitumor immunity. Cancer Res. 2001;61:5132–6. [PubMed] [Google Scholar]
  • 85.Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pagès C, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313:1960–4. doi: 10.1126/science.1129139. [DOI] [PubMed] [Google Scholar]
  • 86.Wakabayashi O, Yamazaki K, Oizumi S, Hommura F, Kinoshita I, Ogura S, et al. CD4+ T cells in cancer stroma, not CD8+ T cells in cancer cell nests, are associated with favorable prognosis in human non-small cell lung cancers. Cancer Sci. 2003;94:1003–9. doi: 10.1111/j.1349-7006.2003.tb01392.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hillen F, Baeten CI, van de Winkel A, Creytens D, van der Schaft DW, Winnepenninckx V, et al. Leukocyte infiltration and tumor cell plasticity are parameters of aggressiveness in primary cutaneous melanoma. Cancer Immunol Immunother. 2008;57:97–106. doi: 10.1007/s00262-007-0353-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Badoual C, Hans S, Rodriguez J, Peyrard S, Klein C, Agueznay NelH, et al. Prognostic value of tumor-infiltrating CD4+ T-cell subpopulations in head and neck cancers. Clin Cancer Res. 2006;12:465–72. doi: 10.1158/1078-0432.CCR-05-1886. [DOI] [PubMed] [Google Scholar]
  • 89.Mlecnik B, Tosolini M, Kirilovsky A, Berger A, Bindea G, Meatchi T, et al. Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction. J Clin Oncol. 2011;29:610–8. doi: 10.1200/JCO.2010.30.5425. [DOI] [PubMed] [Google Scholar]
  • 90.Pagès F, Berger A, Camus M, Sanchez-Cabo F, Costes A, Molidor R, et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med. 2005;353:2654–66. doi: 10.1056/NEJMoa051424. [DOI] [PubMed] [Google Scholar]
  • 91.Pagès F, Kirilovsky A, Mlecnik B, Asslaber M, Tosolini M, Bindea G, et al. In situ cytotoxic and memory T cells predict outcome in patients with early-stage colorectal cancer. J Clin Oncol. 2009;27:5944–51. doi: 10.1200/JCO.2008.19.6147. [DOI] [PubMed] [Google Scholar]
  • 92.Lee WS, Park S, Lee WY, Yun SH, Chun HK. Clinical impact of tumor-infiltrating lymphocytes for survival in stage II colon cancer. Cancer. 2010;116:5188–99. doi: 10.1002/cncr.25293. [DOI] [PubMed] [Google Scholar]
  • 93.Nosho K, Baba Y, Tanaka N, Shima K, Hayashi M, Meyerhardt JA, et al. Tumour-infiltrating T-cell subsets, molecular changes in colorectal cancer, and prognosis: cohort study and literature review. J Pathol. 2010;222:350–66. doi: 10.1002/path.2774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Dieu-Nosjean MC, Antoine M, Danel C, Heudes D, Wislez M, Poulot V, et al. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol. 2008;26:4410–7. doi: 10.1200/JCO.2007.15.0284. [DOI] [PubMed] [Google Scholar]
  • 95.Sautès-Fridman C, Cherfils-Vicini J, Damotte D, Fisson S, Fridman WH, Cremer I, et al. Tumor microenvironment is multifaceted. Cancer Metastasis Rev. 2011;30:13–25. doi: 10.1007/s10555-011-9279-y. [DOI] [PubMed] [Google Scholar]
  • 96.Li YW, Qiu SJ, Fan J, Gao Q, Zhou J, Xiao YS, et al. Tumor-infiltrating macrophages can predict favorable prognosis in hepatocellular carcinoma after resection. J Cancer Res Clin Oncol. 2009;135:439–49. doi: 10.1007/s00432-008-0469-0. [DOI] [PubMed] [Google Scholar]
  • 97.Oldford SA, Robb JD, Codner D, Gadag V, Watson PH, Drover S. Tumor cell expression of HLA-DM associates with a Th1 profile and predicts improved survival in breast carcinoma patients. Int Immunol. 2006;18:1591–602. doi: 10.1093/intimm/dxl092. [DOI] [PubMed] [Google Scholar]
  • 98.Teschendorff AE, Gomez S, Arenas A, El-Ashry D, Schmidt M, Gehrmann M, et al. Improved prognostic classification of breast cancer defined by antagonistic activation patterns of immune response pathway modules. BMC Cancer. 2010;10:604. doi: 10.1186/1471-2407-10-604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Wiegering V, Eyrich M, Rutkowski S, Wölfl M, Schlegel PG, Winkler B. TH1 predominance is associated with improved survival in pediatric medulloblastoma patients. Cancer Immunol Immunother. 2011;60:693–703. doi: 10.1007/s00262-011-0981-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.van Sandick JW, Boermeester MA, Gisbertz SS, ten Berge IJ, Out TA, van der Pouw Kraan TC, et al. Lymphocyte subsets and T(h)1/T(h)2 immune responses in patients with adenocarcinoma of the oesophagus or oesophagogastric junction: relation to pTNM stage and clinical outcome. Cancer Immunol Immunother. 2003;52:617–24. doi: 10.1007/s00262-003-0406-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Ubukata H, Motohashi G, Tabuchi T, Nagata H, Konishi S, Tabuchi T. Evaluations of interferon-γ/interleukin-4 ratio and neutrophil/lymphocyte ratio as prognostic indicators in gastric cancer patients. J Surg Oncol. 2010;102:742–7. doi: 10.1002/jso.21725. [DOI] [PubMed] [Google Scholar]
  • 102.Camus M, Tosolini M, Mlecnik B, Pagès F, Kirilovsky A, Berger A, et al. Coordination of intratumoral immune reaction and human colorectal cancer recurrence. Cancer Res. 2009;69:2685–93. doi: 10.1158/0008-5472.CAN-08-2654. [DOI] [PubMed] [Google Scholar]
  • 103.Tosolini M, Kirilovsky A, Mlecnik B, Fredriksen T, Mauger S, Bindea G, et al. Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, th2, treg, th17) in patients with colorectal cancer. Cancer Res. 2011;71:1263–71. doi: 10.1158/0008-5472.CAN-10-2907. [DOI] [PubMed] [Google Scholar]
  • 104.Kusuda T, Shigemasa K, Arihiro K, Fujii T, Nagai N, Ohama K. Relative expression levels of Th1 and Th2 cytokine mRNA are independent prognostic factors in patients with ovarian cancer. Oncol Rep. 2005;13:1153–8. [PubMed] [Google Scholar]
  • 105.Marth C, Fiegl H, Zeimet AG, Müller-Holzner E, Deibl M, Doppler W, et al. Interferon-gamma expression is an independent prognostic factor in ovarian cancer. Am J Obstet Gynecol. 2004;191:1598–605. doi: 10.1016/j.ajog.2004.05.007. [DOI] [PubMed] [Google Scholar]
  • 106.Schreck S, Friebel D, Buettner M, Distel L, Grabenbauer G, Young LS, et al. Prognostic impact of tumour-infiltrating Th2 and regulatory T cells in classical Hodgkin lymphoma. Hematol Oncol. 2009;27:31–9. doi: 10.1002/hon.878. [DOI] [PubMed] [Google Scholar]
  • 107.De Monte L, Reni M, Tassi E, Clavenna D, Papa I, Recalde H, et al. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J Exp Med. 2011;208:469–78. doi: 10.1084/jem.20101876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Tassi E, Gavazzi F, Albarello L, Senyukov V, Longhi R, Dellabona P, et al. Carcinoembryonic antigen-specific but not antiviral CD4+ T cell immunity is impaired in pancreatic carcinoma patients. J Immunol. 2008;181:6595–603. doi: 10.4049/jimmunol.181.9.6595. [DOI] [PubMed] [Google Scholar]
  • 109.Zhang YL, Li J, Mo HY, Qiu F, Zheng LM, Qian CN, et al. Different subsets of tumor infiltrating lymphocytes correlate with NPC progression in different ways. Mol Cancer. 2010;9:4. doi: 10.1186/1476-4598-9-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Chen JG, Xia JC, Liang XT, Pan K, Wang W, Lv L, et al. Intratumoral expression of IL-17 and its prognostic role in gastric adenocarcinoma patients. Int J Biol Sci. 2011;7:53–60. doi: 10.7150/ijbs.7.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Kryczek I, Banerjee M, Cheng P, Vatan L, Szeliga W, Wei S, et al. Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments. Blood. 2009;114:1141–9. doi: 10.1182/blood-2009-03-208249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Chen X, Wan J, Liu J, Xie W, Diao X, Xu J, et al. Increased IL-17-producing cells correlate with poor survival and lymphangiogenesis in NSCLC patients. Lung Cancer. 2010;69:348–54. doi: 10.1016/j.lungcan.2009.11.013. [DOI] [PubMed] [Google Scholar]
  • 113.Zhang JP, Yan J, Xu J, Pang XH, Chen MS, Li L, et al. Increased intratumoral IL-17-producing cells correlate with poor survival in hepatocellular carcinoma patients. J Hepatol. 2009;50:980–9. doi: 10.1016/j.jhep.2008.12.033. [DOI] [PubMed] [Google Scholar]
  • 114.Liu J, Duan Y, Cheng X, Chen X, Xie W, Long H, et al. IL-17 is associated with poor prognosis and promotes angiogenesis via stimulating VEGF production of cancer cells in colorectal carcinoma. Biochem Biophys Res Commun. 2011;407:348–54. doi: 10.1016/j.bbrc.2011.03.021. [DOI] [PubMed] [Google Scholar]
  • 115.Heimberger AB, Abou-Ghazal M, Reina-Ortiz C, Yang DS, Sun W, Qiao W, et al. Incidence and prognostic impact of FoxP3+ regulatory T cells in human gliomas. Clin Cancer Res. 2008;14:5166–72. doi: 10.1158/1078-0432.CCR-08-0320. [DOI] [PubMed] [Google Scholar]
  • 116.Jacobs JF, Idema AJ, Bol KF, Grotenhuis JA, de Vries IJ, Wesseling P, et al. Prognostic significance and mechanism of Treg infiltration in human brain tumors. J Neuroimmunol. 2010;225:195–9. doi: 10.1016/j.jneuroim.2010.05.020. [DOI] [PubMed] [Google Scholar]
  • 117.Ladányi A, Mohos A, Somlai B, Liszkay G, Gilde K, Fejos Z, et al. FOXP3+ cell density in primary tumor has no prognostic impact in patients with cutaneous malignant melanoma. Pathol Oncol Res. 2010;16:303–9. doi: 10.1007/s12253-010-9254-x. [DOI] [PubMed] [Google Scholar]
  • 118.Bates GJ, Fox SB, Han C, Leek RD, Garcia JF, Harris AL, et al. Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. J Clin Oncol. 2006;24:5373–80. doi: 10.1200/JCO.2006.05.9584. [DOI] [PubMed] [Google Scholar]
  • 119.Mahmoud SM, Paish EC, Powe DG, Macmillan RD, Lee AH, Ellis IO, et al. An evaluation of the clinical significance of FOXP3+ infiltrating cells in human breast cancer. Breast Cancer Res Treat. 2011;127:99–108. doi: 10.1007/s10549-010-0987-8. [DOI] [PubMed] [Google Scholar]
  • 120.Mizukami Y, Kono K, Kawaguchi Y, Akaike H, Kamimura K, Sugai H, et al. Localisation pattern of Foxp3+ regulatory T cells is associated with clinical behaviour in gastric cancer. Br J Cancer. 2008;98:148–53. doi: 10.1038/sj.bjc.6604149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Shimizu K, Nakata M, Hirami Y, Yukawa T, Maeda A, Tanemoto K. 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. 2010;5:585–90. doi: 10.1097/JTO.0b013e3181d60fd7. [DOI] [PubMed] [Google Scholar]
  • 122.Tao H, Mimura Y, Aoe K, Kobayashi S, Yamamoto H, Matsuda E, et al. Prognostic potential of FOXP3 expression in non-small cell lung cancer cells combined with tumor-infiltrating regulatory T cells. Lung Cancer. 2012;75:95–101. doi: 10.1016/j.lungcan.2011.06.002. [DOI] [PubMed] [Google Scholar]
  • 123.Miracco C, Mourmouras V, Biagioli M, Rubegni P, Mannucci S, Monciatti I, et al. Utility of tumour-infiltrating CD25+FOXP3+ regulatory T cell evaluation in predicting local recurrence in vertical growth phase cutaneous melanoma. Oncol Rep. 2007;18:1115–22. [PubMed] [Google Scholar]
  • 124.Mougiakakos D, Johansson CC, Trocme E, All-Ericsson C, Economou MA, Larsson O, et al. Intratumoral forkhead box P3-positive regulatory T cells predict poor survival in cyclooxygenase-2-positive uveal melanoma. Cancer. 2010;116:2224–33. doi: 10.1002/cncr.24999. [DOI] [PubMed] [Google Scholar]
  • 125.Kaufman HL, Kim DW, DeRaffele G, Mitcham J, Coffin RS, Kim-Schulze S. Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Ann Surg Oncol. 2010;17:718–30. doi: 10.1245/s10434-009-0809-6. [DOI] [PubMed] [Google Scholar]
  • 126.Jensen HK, Donskov F, Nordsmark M, Marcussen N, von der Maase H. Increased intratumoral FOXP3-positive regulatory immune cells during interleukin-2 treatment in metastatic renal cell carcinoma. Clin Cancer Res. 2009;15:1052–8. doi: 10.1158/1078-0432.CCR-08-1296. [DOI] [PubMed] [Google Scholar]
  • 127.Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10:942–9. doi: 10.1038/nm1093. [DOI] [PubMed] [Google Scholar]
  • 128.Wansom D, Light E, Thomas D, Worden F, Prince M, Urba S, et al. UM Head Neck SPORE Program Infiltrating lymphocytes and human papillomavirus-16--associated oropharyngeal cancer. Laryngoscope. 2012;122:121–7. doi: 10.1002/lary.22133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Carreras J, Lopez-Guillermo A, Fox BC, Colomo L, Martinez A, Roncador G, et al. High numbers of tumor-infiltrating FOXP3-positive regulatory T cells are associated with improved overall survival in follicular lymphoma. Blood. 2006;108:2957–64. doi: 10.1182/blood-2006-04-018218. [DOI] [PubMed] [Google Scholar]
  • 130.Tzankov A, Meier C, Hirschmann P, Went P, Pileri SA, Dirnhofer S. Correlation of high numbers of intratumoral FOXP3+ regulatory T cells with improved survival in germinal center-like diffuse large B-cell lymphoma, follicular lymphoma and classical Hodgkin’s lymphoma. Haematologica. 2008;93:193–200. doi: 10.3324/haematol.11702. [DOI] [PubMed] [Google Scholar]
  • 131.Salama P, Phillips M, Grieu F, Morris M, Zeps N, Joseph D, et al. Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J Clin Oncol. 2009;27:186–92. doi: 10.1200/JCO.2008.18.7229. [DOI] [PubMed] [Google Scholar]
  • 132.Correale P, Rotundo MS, Del Vecchio MT, Remondo C, Migali C, Ginanneschi C, et al. Regulatory (FoxP3+) T-cell tumor infiltration is a favorable prognostic factor in advanced colon cancer patients undergoing chemo or chemoimmunotherapy. J Immunother. 2010;33:435–41. doi: 10.1097/CJI.0b013e3181d32f01. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Frey DM, Droeser RA, Viehl CT, Zlobec I, Lugli A, Zingg U, et al. High frequency of tumor-infiltrating FOXP3(+) regulatory T cells predicts improved survival in mismatch repair-proficient colorectal cancer patients. Int J Cancer. 2010;126:2635–43. doi: 10.1002/ijc.24989. [DOI] [PubMed] [Google Scholar]
  • 134.Correale P, Rotundo MS, Botta C, Del Vecchio MT, Ginanneschi C, Licchetta A, et al. Tumor infiltration by T lymphocytes expressing chemokine receptor 7 (CCR7) is predictive of favorable outcome in patients with advanced colorectal carcinoma. Clin Cancer Res. 2012;18:850–7. doi: 10.1158/1078-0432.CCR-10-3186. [DOI] [PubMed] [Google Scholar]
  • 135.Winerdal ME, Marits P, Winerdal M, Hasan M, Rosenblatt R, Tolf A, et al. FOXP3 and survival in urinary bladder cancer. BJU Int. 2011;108:1672–8. doi: 10.1111/j.1464-410X.2010.10020.x. [DOI] [PubMed] [Google Scholar]
  • 136.Ladoire S, Martin F, Ghiringhelli F. Prognostic role of FOXP3+ regulatory T cells infiltrating human carcinomas: the paradox of colorectal cancer. Cancer Immunol Immunother. 2011;60:909–18. doi: 10.1007/s00262-011-1046-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Ladoire S, Arnould L, Apetoh L, Coudert B, Martin F, Chauffert B, et al. Pathologic complete response to neoadjuvant chemotherapy of breast carcinoma is associated with the disappearance of tumor-infiltrating foxp3+ regulatory T cells. Clin Cancer Res. 2008;14:2413–20. doi: 10.1158/1078-0432.CCR-07-4491. [DOI] [PubMed] [Google Scholar]
  • 138.Ladoire S, Mignot G, Dabakuyo S, Arnould L, Apetoh L, Rébé C, et al. In situ immune response after neoadjuvant chemotherapy for breast cancer predicts survival. J Pathol. 2011;224:389–400. doi: 10.1002/path.2866. [DOI] [PubMed] [Google Scholar]
  • 139.Senovilla L, Vitale I, Martins I, Tailler M, Pailleret C, Michaud M, et al. An immunosurveillance mechanism controls cancer cell ploidy. Science. 2012 doi: 10.1126/science.1224922. In press. [DOI] [PubMed] [Google Scholar]
  • 140.Petersen RP, Campa MJ, Sperlazza J, Conlon D, Joshi MB, Harpole DH, Jr., et al. Tumor infiltrating Foxp3+ regulatory T-cells are associated with recurrence in pathologic stage I NSCLC patients. Cancer. 2006;107:2866–72. doi: 10.1002/cncr.22282. [DOI] [PubMed] [Google Scholar]
  • 141.Gao Q, Qiu SJ, Fan J, Zhou J, Wang XY, Xiao YS, et al. Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. J Clin Oncol. 2007;25:2586–93. doi: 10.1200/JCO.2006.09.4565. [DOI] [PubMed] [Google Scholar]
  • 142.Leffers N, Gooden MJ, de Jong RA, Hoogeboom BN, ten Hoor KA, Hollema H, et al. Prognostic significance of tumor-infiltrating T-lymphocytes in primary and metastatic lesions of advanced stage ovarian cancer. Cancer Immunol Immunother. 2009;58:449–59. doi: 10.1007/s00262-008-0583-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A. 2005;102:18538–43. doi: 10.1073/pnas.0509182102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Sun JC, Lanier LL. NK cell development, homeostasis and function: parallels with CD8⁺ T cells. Nat Rev Immunol. 2011;11:645–57. doi: 10.1038/nri3044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, et al. Innate or adaptive immunity? The example of natural killer cells. Science. 2011;331:44–9. doi: 10.1126/science.1198687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Milush JM, Long BR, Snyder-Cappione JE, Cappione AJ, 3rd, York VA, Ndhlovu LC, et al. Functionally distinct subsets of human NK cells and monocyte/DC-like cells identified by coexpression of CD56, CD7, and CD4. Blood. 2009;114:4823–31. doi: 10.1182/blood-2009-04-216374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Addison EG, North J, Bakhsh I, Marden C, Haq S, Al-Sarraj S, et al. Ligation of CD8alpha on human natural killer cells prevents activation-induced apoptosis and enhances cytolytic activity. Immunology. 2005;116:354–61. doi: 10.1111/j.1365-2567.2005.02235.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Bernstein HB, Plasterer MC, Schiff SE, Kitchen CM, Kitchen S, Zack JA. CD4 expression on activated NK cells: ligation of CD4 induces cytokine expression and cell migration. J Immunol. 2006;177:3669–76. doi: 10.4049/jimmunol.177.6.3669. [DOI] [PubMed] [Google Scholar]
  • 149.Moretta A. Natural killer cells and dendritic cells: rendezvous in abused tissues. Nat Rev Immunol. 2002;2:957–64. doi: 10.1038/nri956. [DOI] [PubMed] [Google Scholar]
  • 150.Moretta L, Ferlazzo G, Bottino C, Vitale M, Pende D, Mingari MC, et al. Effector and regulatory events during natural killer-dendritic cell interactions. Immunol Rev. 2006;214:219–28. doi: 10.1111/j.1600-065X.2006.00450.x. [DOI] [PubMed] [Google Scholar]
  • 151.Takeda K, Hayakawa Y, Smyth MJ, Kayagaki N, Yamaguchi N, Kakuta S, et al. Involvement of tumor necrosis factor-related apoptosis-inducing ligand in surveillance of tumor metastasis by liver natural killer cells. Nat Med. 2001;7:94–100. doi: 10.1038/83416. [DOI] [PubMed] [Google Scholar]
  • 152.Smyth MJ, Thia KY, Street SE, Cretney E, Trapani JA, Taniguchi M, et al. Differential tumor surveillance by natural killer (NK) and NKT cells. J Exp Med. 2000;191:661–8. doi: 10.1084/jem.191.4.661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Smyth MJ, Swann J, Cretney E, Zerafa N, Yokoyama WM, Hayakawa Y. NKG2D function protects the host from tumor initiation. J Exp Med. 2005;202:583–8. doi: 10.1084/jem.20050994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Smyth MJ, Swann J, Kelly JM, Cretney E, Yokoyama WM, Diefenbach A, et al. NKG2D recognition and perforin effector function mediate effective cytokine immunotherapy of cancer. J Exp Med. 2004;200:1325–35. doi: 10.1084/jem.20041522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Morandi B, Mortara L, Chiossone L, Accolla RS, Mingari MC, Moretta L, et al. Dendritic cell editing by activated natural killer cells results in a more protective cancer-specific immune response. PLoS One. 2012;7:e39170. doi: 10.1371/journal.pone.0039170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Karadayi S, Kayi Cangir A, Ozturk S, Dizbay Sak S, Akal M, Akay H. [The prognostic effect of natural killer cell infiltration to tumoral tissues in stage I non-small cell lung cancer] Tuberk Toraks. 2008;56:251–6. [PubMed] [Google Scholar]
  • 157.Cao FM, Zhang XH, Yan X, Wang JL, Wang XL, Zhang ZG, et al. [Prognostic significances of natural killer cells and dendritic cells infiltrations in esophageal squamous cell carcinoma] Ai Zheng. 2005;24:232–6. [PubMed] [Google Scholar]
  • 158.Hsia JY, Chen JT, Chen CY, Hsu CP, Miaw J, Huang YS, et al. Prognostic significance of intratumoral natural killer cells in primary resected esophageal squamous cell carcinoma. Chang Gung Med J. 2005;28:335–40. [PubMed] [Google Scholar]
  • 159.van Herpen CM, van der Laak JA, de Vries IJ, van Krieken JH, de Wilde PC, Balvers MG, et al. Intratumoral recombinant human interleukin-12 administration in head and neck squamous cell carcinoma patients modifies locoregional lymph node architecture and induces natural killer cell infiltration in the primary tumor. Clin Cancer Res. 2005;11:1899–909. doi: 10.1158/1078-0432.CCR-04-1524. [DOI] [PubMed] [Google Scholar]
  • 160.Ishigami S, Natsugoe S, Tokuda K, Nakajo A, Che X, Iwashige H, et al. Prognostic value of intratumoral natural killer cells in gastric carcinoma. Cancer. 2000;88:577–83. doi: 10.1002/(SICI)1097-0142(20000201)88:3<577::AID-CNCR13>3.0.CO;2-V. [DOI] [PubMed] [Google Scholar]
  • 161.Ishigami S, Natsugoe S, Tokuda K, Nakajo A, Xiangming C, Iwashige H, et al. Clinical impact of intratumoral natural killer cell and dendritic cell infiltration in gastric cancer. Cancer Lett. 2000;159:103–8. doi: 10.1016/S0304-3835(00)00542-5. [DOI] [PubMed] [Google Scholar]
  • 162.Zhu LY, Zhou J, Liu YZ, Pan WD. [Prognostic significance of natural killer cell infiltration in hepatocellular carcinoma] Ai Zheng. 2009;28:1198–202. doi: 10.5732/cjc.009.10284. [DOI] [PubMed] [Google Scholar]
  • 163.Coca S, Perez-Piqueras J, Martinez D, Colmenarejo A, Saez MA, Vallejo C, et al. The prognostic significance of intratumoral natural killer cells in patients with colorectal carcinoma. Cancer. 1997;79:2320–8. doi: 10.1002/(SICI)1097-0142(19970615)79:12<2320::AID-CNCR5>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
  • 164.Delahaye NF, Rusakiewicz S, Martins I, Ménard C, Roux S, Lyonnet L, et al. Alternatively spliced NKp30 isoforms affect the prognosis of gastrointestinal stromal tumors. Nat Med. 2011;17:700–7. doi: 10.1038/nm.2366. [DOI] [PubMed] [Google Scholar]
  • 165.Ménard C, Blay JY, Borg C, Michiels S, Ghiringhelli F, Robert C, et al. Natural killer cell IFN-gamma levels predict long-term survival with imatinib mesylate therapy in gastrointestinal stromal tumor-bearing patients. Cancer Res. 2009;69:3563–9. doi: 10.1158/0008-5472.CAN-08-3807. [DOI] [PubMed] [Google Scholar]
  • 166.Bendelac A, Savage PB, Teyton L. The biology of NKT cells. Annu Rev Immunol. 2007;25:297–336. doi: 10.1146/annurev.immunol.25.022106.141711. [DOI] [PubMed] [Google Scholar]
  • 167.Cui J, Shin T, Kawano T, Sato H, Kondo E, Toura I, et al. Requirement for Valpha14 NKT cells in IL-12-mediated rejection of tumors. Science. 1997;278:1623–6. doi: 10.1126/science.278.5343.1623. [DOI] [PubMed] [Google Scholar]
  • 168.Godfrey DI, MacDonald HR, Kronenberg M, Smyth MJ, Van Kaer L. NKT cells: what’s in a name? Nat Rev Immunol. 2004;4:231–7. doi: 10.1038/nri1309. [DOI] [PubMed] [Google Scholar]
  • 169.Taniguchi M, Harada M, Kojo S, Nakayama T, Wakao H. The regulatory role of Valpha14 NKT cells in innate and acquired immune response. Annu Rev Immunol. 2003;21:483–513. doi: 10.1146/annurev.immunol.21.120601.141057. [DOI] [PubMed] [Google Scholar]
  • 170.Kawano T, Cui J, Koezuka Y, Toura I, Kaneko Y, Motoki K, et al. CD1d-restricted and TCR-mediated activation of valpha14 NKT cells by glycosylceramides. Science. 1997;278:1626–9. doi: 10.1126/science.278.5343.1626. [DOI] [PubMed] [Google Scholar]
  • 171.Schofield L, McConville MJ, Hansen D, Campbell AS, Fraser-Reid B, Grusby MJ, et al. CD1d-restricted immunoglobulin G formation to GPI-anchored antigens mediated by NKT cells. Science. 1999;283:225–9. doi: 10.1126/science.283.5399.225. [DOI] [PubMed] [Google Scholar]
  • 172.Song L, Asgharzadeh S, Salo J, Engell K, Wu HW, Sposto R, et al. Valpha24-invariant NKT cells mediate antitumor activity via killing of tumor-associated macrophages. J Clin Invest. 2009;119:1524–36. doi: 10.1172/JCI37869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Mussai F, De Santo C, Cerundolo V. Interaction between invariant NKT cells and myeloid-derived suppressor cells in cancer patients: evidence and therapeutic opportunities. J Immunother. 2012;35:449–59. doi: 10.1097/CJI.0b013e31825be926. [DOI] [PubMed] [Google Scholar]
  • 174.van der Vliet HJ, Wang R, Yue SC, Koon HB, Balk SP, Exley MA. Circulating myeloid dendritic cells of advanced cancer patients result in reduced activation and a biased cytokine profile in invariant NKT cells. J Immunol. 2008;180:7287–93. doi: 10.4049/jimmunol.180.11.7287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Yoneda K, Morii T, Nieda M, Tsukaguchi N, Amano I, Tanaka H, et al. The peripheral blood Valpha24+ NKT cell numbers decrease in patients with haematopoietic malignancy. Leuk Res. 2005;29:147–52. doi: 10.1016/j.leukres.2004.06.005. [DOI] [PubMed] [Google Scholar]
  • 176.Gulubova M, Manolova I, Kyurkchiev D, Julianov A, Altunkova I. Decrease in intrahepatic CD56+ lymphocytes in gastric and colorectal cancer patients with liver metastases. APMIS. 2009;117:870–9. doi: 10.1111/j.1600-0463.2009.02547.x. [DOI] [PubMed] [Google Scholar]
  • 177.Jadidi-Niaragh F, Jeddi-Tehrani M, Ansaripour B, Razavi SM, Sharifian RA, Shokri F. Reduced frequency of NKT-like cells in patients with progressive chronic lymphocytic leukemia. Med Oncol. 2012 doi: 10.1007/s12032-012-0262-4. In press. [DOI] [PubMed] [Google Scholar]
  • 178.Giaccone G, Punt CJ, Ando Y, Ruijter R, Nishi N, Peters M, et al. A phase I study of the natural killer T-cell ligand alpha-galactosylceramide (KRN7000) in patients with solid tumors. Clin Cancer Res. 2002;8:3702–9. [PubMed] [Google Scholar]
  • 179.Motohashi S, Okamoto Y, Yoshino I, Nakayama T. Anti-tumor immune responses induced by iNKT cell-based immunotherapy for lung cancer and head and neck cancer. Clin Immunol. 2011;140:167–76. doi: 10.1016/j.clim.2011.01.009. [DOI] [PubMed] [Google Scholar]
  • 180.Yamasaki K, Horiguchi S, Kurosaki M, Kunii N, Nagato K, Hanaoka H, et al. Induction of NKT cell-specific immune responses in cancer tissues after NKT cell-targeted adoptive immunotherapy. Clin Immunol. 2011;138:255–65. doi: 10.1016/j.clim.2010.11.014. [DOI] [PubMed] [Google Scholar]
  • 181.Cariani E, Pilli M, Zerbini A, Rota C, Olivani A, Pelosi G, et al. Immunological and molecular correlates of disease recurrence after liver resection for hepatocellular carcinoma. PLoS One. 2012;7:e32493. doi: 10.1371/journal.pone.0032493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Akagi J, Baba H. Prognostic value of CD57(+) T lymphocytes in the peripheral blood of patients with advanced gastric cancer. Int J Clin Oncol. 2008;13:528–35. doi: 10.1007/s10147-008-0789-8. [DOI] [PubMed] [Google Scholar]
  • 183.de Lalla C, Rinaldi A, Montagna D, Azzimonti L, Bernardo ME, Sangalli LM, et al. Invariant NKT cell reconstitution in pediatric leukemia patients given HLA-haploidentical stem cell transplantation defines distinct CD4+ and CD4- subset dynamics and correlates with remission state. J Immunol. 2011;186:4490–9. doi: 10.4049/jimmunol.1003748. [DOI] [PubMed] [Google Scholar]
  • 184.Steinman RM, Cohn ZA. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J Exp Med. 1973;137:1142–62. doi: 10.1084/jem.137.5.1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–52. doi: 10.1038/32588. [DOI] [PubMed] [Google Scholar]
  • 186.Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007;449:419–26. doi: 10.1038/nature06175. [DOI] [PubMed] [Google Scholar]
  • 187.Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 2012;12:265–77. doi: 10.1038/nrc3258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Reis e Sousa C. Dendritic cells in a mature age. Nat Rev Immunol. 2006;6:476–83. doi: 10.1038/nri1845. [DOI] [PubMed] [Google Scholar]
  • 189.Steinman RM. Decisions about dendritic cells: past, present, and future. Annu Rev Immunol. 2012;30:1–22. doi: 10.1146/annurev-immunol-100311-102839. [DOI] [PubMed] [Google Scholar]
  • 190.Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol. 2003;21:685–711. doi: 10.1146/annurev.immunol.21.120601.141040. [DOI] [PubMed] [Google Scholar]
  • 191.Maldonado-López R, De Smedt T, Michel P, Godfroid J, Pajak B, Heirman C, et al. CD8alpha+ and CD8alpha- subclasses of dendritic cells direct the development of distinct T helper cells in vivo. J Exp Med. 1999;189:587–92. doi: 10.1084/jem.189.3.587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Pulendran B, Smith JL, Caspary G, Brasel K, Pettit D, Maraskovsky E, et al. Distinct dendritic cell subsets differentially regulate the class of immune response in vivo. Proc Natl Acad Sci U S A. 1999;96:1036–41. doi: 10.1073/pnas.96.3.1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Dudziak D, Kamphorst AO, Heidkamp GF, Buchholz VR, Trumpfheller C, Yamazaki S, et al. Differential antigen processing by dendritic cell subsets in vivo. Science. 2007;315:107–11. doi: 10.1126/science.1136080. [DOI] [PubMed] [Google Scholar]
  • 194.Klechevsky E, Morita R, Liu M, Cao Y, Coquery S, Thompson-Snipes L, et al. Functional specializations of human epidermal Langerhans cells and CD14+ dermal dendritic cells. Immunity. 2008;29:497–510. doi: 10.1016/j.immuni.2008.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Ueno H, Klechevsky E, Morita R, Aspord C, Cao T, Matsui T, et al. Dendritic cell subsets in health and disease. Immunol Rev. 2007;219:118–42. doi: 10.1111/j.1600-065X.2007.00551.x. [DOI] [PubMed] [Google Scholar]
  • 196.Bachem A, Güttler S, Hartung E, Ebstein F, Schaefer M, Tannert A, et al. Superior antigen cross-presentation and XCR1 expression define human CD11c+CD141+ cells as homologues of mouse CD8+ dendritic cells. J Exp Med. 2010;207:1273–81. doi: 10.1084/jem.20100348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Crozat K, Guiton R, Contreras V, Feuillet V, Dutertre CA, Ventre E, et al. The XC chemokine receptor 1 is a conserved selective marker of mammalian cells homologous to mouse CD8alpha+ dendritic cells. J Exp Med. 2010;207:1283–92. doi: 10.1084/jem.20100223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Jongbloed SL, Kassianos AJ, McDonald KJ, Clark GJ, Ju X, Angel CE, et al. Human CD141+ (BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens. J Exp Med. 2010;207:1247–60. doi: 10.1084/jem.20092140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Poulin LF, Salio M, Griessinger E, Anjos-Afonso F, Craciun L, Chen JL, et al. Characterization of human DNGR-1+ BDCA3+ leukocytes as putative equivalents of mouse CD8alpha+ dendritic cells. J Exp Med. 2010;207:1261–71. doi: 10.1084/jem.20092618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Swiecki M, Colonna M. Unraveling the functions of plasmacytoid dendritic cells during viral infections, autoimmunity, and tolerance. Immunol Rev. 2010;234:142–62. doi: 10.1111/j.0105-2896.2009.00881.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Reizis B, Colonna M, Trinchieri G, Barrat F, Gilliet M. Plasmacytoid dendritic cells: one-trick ponies or workhorses of the immune system? Nat Rev Immunol. 2011;11:558–65. doi: 10.1038/nri3027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Kadowaki N, Ho S, Antonenko S, Malefyt RW, Kastelein RA, Bazan F, et al. Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens. J Exp Med. 2001;194:863–9. doi: 10.1084/jem.194.6.863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Gibson SJ, Lindh JM, Riter TR, Gleason RM, Rogers LM, Fuller AE, et al. Plasmacytoid dendritic cells produce cytokines and mature in response to the TLR7 agonists, imiquimod and resiquimod. Cell Immunol. 2002;218:74–86. doi: 10.1016/S0008-8749(02)00517-8. [DOI] [PubMed] [Google Scholar]
  • 204.Kadowaki N, Liu YJ. Natural type I interferon-producing cells as a link between innate and adaptive immunity. Hum Immunol. 2002;63:1126–32. doi: 10.1016/S0198-8859(02)00751-6. [DOI] [PubMed] [Google Scholar]
  • 205.Siegal FP, Kadowaki N, Shodell M, Fitzgerald-Bocarsly PA, Shah K, Ho S, et al. The nature of the principal type 1 interferon-producing cells in human blood. Science. 1999;284:1835–7. doi: 10.1126/science.284.5421.1835. [DOI] [PubMed] [Google Scholar]
  • 206.Cella M, Facchetti F, Lanzavecchia A, Colonna M. Plasmacytoid dendritic cells activated by influenza virus and CD40L drive a potent TH1 polarization. Nat Immunol. 2000;1:305–10. doi: 10.1038/79747. [DOI] [PubMed] [Google Scholar]
  • 207.Gilliet M, Boonstra A, Paturel C, Antonenko S, Xu XL, Trinchieri G, et al. The development of murine plasmacytoid dendritic cell precursors is differentially regulated by FLT3-ligand and granulocyte/macrophage colony-stimulating factor. J Exp Med. 2002;195:953–8. doi: 10.1084/jem.20020045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Van Vré EA, Bosmans JM, Van Brussel I, Maris M, De Meyer GR, Van Schil PE, et al. Immunohistochemical characterisation of dendritic cells in human atherosclerotic lesions: possible pitfalls. Pathology. 2011;43:239–47. doi: 10.1097/PAT.0b013e328344e266. [DOI] [PubMed] [Google Scholar]
  • 209.Orii T, Takeda H, Kawata S, Maeda K, Yamakawa M. Differential immunophenotypic analysis of dendritic cell tumours. J Clin Pathol. 2010;63:497–503. doi: 10.1136/jcp.2009.067819. [DOI] [PubMed] [Google Scholar]
  • 210.Steinman RM, Idoyaga J. Features of the dendritic cell lineage. Immunol Rev. 2010;234:5–17. doi: 10.1111/j.0105-2896.2009.00888.x. [DOI] [PubMed] [Google Scholar]
  • 211.Zhao R, Feng H, Wang X, Hu D, Zhu T. [Study on the relationship between the dendritic cell infiltration in cancer tissues and prognosis in patients with lung cancer] Zhongguo Fei Ai Za Zhi. 2002;5:112–4. doi: 10.3779/j.issn.1009-3419.2002.02.10. [DOI] [PubMed] [Google Scholar]
  • 212.Gallo O, Bianchi S, Giannini A, Gallina E, Libonati GA, Fini-Storchi O. Correlations between histopathological and biological findings in nasopharyngeal carcinoma and its prognostic significance. Laryngoscope. 1991;101:487–93. doi: 10.1288/00005537-199105000-00008. [DOI] [PubMed] [Google Scholar]
  • 213.Ikeguchi M, Ikeda M, Tatebe S, Maeta M, Kaibara N. Clinical significance of dendritic cell infiltration in esophageal squamous cell carcinoma. Oncol Rep. 1998;5:1185–9. doi: 10.3892/or.5.5.1185. [DOI] [PubMed] [Google Scholar]
  • 214.Ishigami S, Natsugoe S, Matsumoto M, Okumura H, Sakita H, Nakashima S, et al. Clinical implications of intratumoral dendritic cell infiltration in esophageal squamous cell carcinoma. Oncol Rep. 2003;10:1237–40. [PubMed] [Google Scholar]
  • 215.Kakeji Y, Maehara Y, Korenaga D, Tsujitani S, Haraguchi M, Watanabe A, et al. Prognostic significance of tumor-host interaction in clinical gastric cancer: relationship between DNA ploidy and dendritic cell infiltration. J Surg Oncol. 1993;52:207–12. doi: 10.1002/jso.2930520402. [DOI] [PubMed] [Google Scholar]
  • 216.Ishigami S, Aikou T, Natsugoe S, Hokita S, Iwashige H, Tokushige M, et al. Prognostic value of HLA-DR expression and dendritic cell infiltration in gastric cancer. Oncology. 1998;55:65–9. doi: 10.1159/000011837. [DOI] [PubMed] [Google Scholar]
  • 217.Okuyama T, Maehara Y, Kakeji Y, Tsuijitani S, Korenaga D, Sugimachi K. Interrelation between tumor-associated cell surface glycoprotein and host immune response in gastric carcinoma patients. Cancer. 1998;82:1468–75. doi: 10.1002/(SICI)1097-0142(19980415)82:8<1468::AID-CNCR6>3.0.CO;2-5. [DOI] [PubMed] [Google Scholar]
  • 218.Saito H, Osaki T, Murakami D, Sakamoto T, Kanaji S, Ohro S, et al. Prediction of sites of recurrence in gastric carcinoma using immunohistochemical parameters. J Surg Oncol. 2007;95:123–8. doi: 10.1002/jso.20612. [DOI] [PubMed] [Google Scholar]
  • 219.Daud AI, Mirza N, Lenox B, Andrews S, Urbas P, Gao GX, et al. Phenotypic and functional analysis of dendritic cells and clinical outcome in patients with high-risk melanoma treated with adjuvant granulocyte macrophage colony-stimulating factor. J Clin Oncol. 2008;26:3235–41. doi: 10.1200/JCO.2007.13.9048. [DOI] [PubMed] [Google Scholar]
  • 220.Chen T, Zheng SB, Tan WL, Li ZG. J First Mil Med Univ. 2002;22:833–4. [Association of dendritic cell infiltration with prognosis and lymph node metastasis of renal cell carcinoma] [PubMed] [Google Scholar]
  • 221.Honig A, Schaller N, Dietl J, Backe J, Kammerer U. S100 as an immunohistochemically-detected marker with prognostic significance in endometrial carcinoma. Anticancer Res. 2005;25(3A):1747–53. [PubMed] [Google Scholar]
  • 222.Chang KC, Huang GC, Jones D, Lin YH. Distribution patterns of dendritic cells and T cells in diffuse large B-cell lymphomas correlate with prognoses. Clin Cancer Res. 2007;13:6666–72. doi: 10.1158/1078-0432.CCR-07-0504. [DOI] [PubMed] [Google Scholar]
  • 223.Furihata M, Ohtsuki Y, Sonobe H, Araki K, Ogata T, Toki T, et al. Prognostic significance of simultaneous infiltration of HLA-DR-positive dendritic cells and tumor infiltrating lymphocytes into human esophageal carcinoma. Tohoku J Exp Med. 1993;169:187–95. doi: 10.1620/tjem.169.187. [DOI] [PubMed] [Google Scholar]
  • 224.Iwamoto M, Shinohara H, Miyamoto A, Okuzawa M, Mabuchi H, Nohara T, et al. Prognostic value of tumor-infiltrating dendritic cells expressing CD83 in human breast carcinomas. Int J Cancer. 2003;104:92–7. doi: 10.1002/ijc.10915. [DOI] [PubMed] [Google Scholar]
  • 225.Ladányi A, Kiss J, Somlai B, Gilde K, Fejos Z, Mohos A, et al. Density of DC-LAMP(+) mature dendritic cells in combination with activated T lymphocytes infiltrating primary cutaneous melanoma is a strong independent prognostic factor. Cancer Immunol Immunother. 2007;56:1459–69. doi: 10.1007/s00262-007-0286-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Kobayashi M, Suzuki K, Yashi M, Yuzawa M, Takayashiki N, Morita T. Tumor infiltrating dendritic cells predict treatment response to immmunotherapy in patients with metastatic renal cell carcinoma. Anticancer Res. 2007;27:1137–41. [PubMed] [Google Scholar]
  • 227.Ma CX, Jia TC, Li XR, Zhand ZF, Yiao CB. Langerhans cells in nasopharyngeal carcinoma in relation to prognosis. In Vivo. 1995;9:225–9. [PubMed] [Google Scholar]
  • 228.Treilleux I, Blay JY, Bendriss-Vermare N, Ray-Coquard I, Bachelot T, Guastalla JP, et al. Dendritic cell infiltration and prognosis of early stage breast cancer. Clin Cancer Res. 2004;10:7466–74. doi: 10.1158/1078-0432.CCR-04-0684. [DOI] [PubMed] [Google Scholar]
  • 229.Ishigami S, Ueno S, Matsumoto M, Okumura H, Arigami T, Uchikado Y, et al. Prognostic value of CD208-positive cell infiltration in gastric cancer. Cancer Immunol Immunother. 2010;59:389–95. doi: 10.1007/s00262-009-0758-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Sandel MH, Dadabayev AR, Menon AG, Morreau H, Melief CJ, Offringa R, et al. Prognostic value of tumor-infiltrating dendritic cells in colorectal cancer: role of maturation status and intratumoral localization. Clin Cancer Res. 2005;11:2576–82. doi: 10.1158/1078-0432.CCR-04-1448. [DOI] [PubMed] [Google Scholar]
  • 231.Jensen TO, Schmidt H, Møller HJ, Donskov F, Høyer M, Sjoegren P, et al. Intratumoral neutrophils and plasmacytoid dendritic cells indicate poor prognosis and are associated with pSTAT3 expression in AJCC stage I/II melanoma. Cancer. 2012;118:2476–85. doi: 10.1002/cncr.26511. [DOI] [PubMed] [Google Scholar]
  • 232.Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5:953–64. doi: 10.1038/nri1733. [DOI] [PubMed] [Google Scholar]
  • 233.Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958–69. doi: 10.1038/nri2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Taylor PR, Martinez-Pomares L, Stacey M, Lin HH, Brown GD, Gordon S. Macrophage receptors and immune recognition. Annu Rev Immunol. 2005;23:901–44. doi: 10.1146/annurev.immunol.23.021704.115816. [DOI] [PubMed] [Google Scholar]
  • 235.Zitvogel L, Kepp O, Galluzzi L, Kroemer G. Inflammasomes in carcinogenesis and anticancer immune responses. Nat Immunol. 2012;13:343–51. doi: 10.1038/ni.2224. [DOI] [PubMed] [Google Scholar]
  • 236.Martinez FO, Helming L, Gordon S. Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol. 2009;27:451–83. doi: 10.1146/annurev.immunol.021908.132532. [DOI] [PubMed] [Google Scholar]
  • 237.Porcheray F, Viaud S, Rimaniol AC, Léone C, Samah B, Dereuddre-Bosquet N, et al. Macrophage activation switching: an asset for the resolution of inflammation. Clin Exp Immunol. 2005;142:481–9. doi: 10.1111/j.1365-2249.2005.02934.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Hashimoto I, Kodama J, Seki N, Hongo A, Miyagi Y, Yoshinouchi M, et al. Macrophage infiltration and angiogenesis in endometrial cancer. Anticancer Res. 2000;20(6C):4853–6. [PubMed] [Google Scholar]
  • 239.Tsutsui S, Yasuda K, Suzuki K, Tahara K, Higashi H, Era S. Macrophage infiltration and its prognostic implications in breast cancer: the relationship with VEGF expression and microvessel density. Oncol Rep. 2005;14:425–31. [PubMed] [Google Scholar]
  • 240.Morra B, Ferrero V, Bussi M, Pacchioni D, Cerrato M, Bussolati G. Peri and intratumoral macrophage infiltration in laryngeal carcinoma. An immunohistochemical study. Acta Otolaryngol. 1991;111:444–8. doi: 10.3109/00016489109137417. [DOI] [PubMed] [Google Scholar]
  • 241.Dutta S, Going JJ, Crumley AB, Mohammed Z, Orange C, Edwards J, et al. The relationship between tumour necrosis, tumour proliferation, local and systemic inflammation, microvessel density and survival in patients undergoing potentially curative resection of oesophageal adenocarcinoma. Br J Cancer. 2012;106:702–10. doi: 10.1038/bjc.2011.610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Koide N, Nishio A, Sato T, Sugiyama A, Miyagawa S. Significance of macrophage chemoattractant protein-1 expression and macrophage infiltration in squamous cell carcinoma of the esophagus. Am J Gastroenterol. 2004;99:1667–74. doi: 10.1111/j.1572-0241.2004.30733.x. [DOI] [PubMed] [Google Scholar]
  • 243.Ishigami S, Natsugoe S, Tokuda K, Nakajo A, Okumura H, Matsumoto M, et al. Tumor-associated macrophage (TAM) infiltration in gastric cancer. Anticancer Res. 2003;23(5A):4079–83. [PubMed] [Google Scholar]
  • 244.Ding T, Xu J, Wang F, Shi M, Zhang Y, Li SP, et al. High tumor-infiltrating macrophage density predicts poor prognosis in patients with primary hepatocellular carcinoma after resection. Hum Pathol. 2009;40:381–9. doi: 10.1016/j.humpath.2008.08.011. [DOI] [PubMed] [Google Scholar]
  • 245.Takanami I, Takeuchi K, Kodaira S. Tumor-associated macrophage infiltration in pulmonary adenocarcinoma: association with angiogenesis and poor prognosis. Oncology. 1999;57:138–42. doi: 10.1159/000012021. [DOI] [PubMed] [Google Scholar]
  • 246.Burt BM, Rodig SJ, Tilleman TR, Elbardissi AW, Bueno R, Sugarbaker DJ. Circulating and tumor-infiltrating myeloid cells predict survival in human pleural mesothelioma. Cancer. 2011;117:5234–44. doi: 10.1002/cncr.26143. [DOI] [PubMed] [Google Scholar]
  • 247.Storr SJ, Safuan S, Mitra A, Elliott F, Walker C, Vasko MJ, et al. Objective assessment of blood and lymphatic vessel invasion and association with macrophage infiltration in cutaneous melanoma. Mod Pathol. 2012;25:493–504. doi: 10.1038/modpathol.2011.182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Jensen TO, Schmidt H, Møller HJ, Høyer M, Maniecki MB, Sjoegren P, et al. Macrophage markers in serum and tumor have prognostic impact in American Joint Committee on Cancer stage I/II melanoma. J Clin Oncol. 2009;27:3330–7. doi: 10.1200/JCO.2008.19.9919. [DOI] [PubMed] [Google Scholar]
  • 249.Wan T, Liu JH, Zheng LM, Cai MY, Ding T. [Prognostic significance of tumor-associated macrophage infiltration in advanced epithelial ovarian carcinoma] Ai Zheng. 2009;28:323–7. [PubMed] [Google Scholar]
  • 250.Takayama H, Nishimura K, Tsujimura A, Nakai Y, Nakayama M, Aozasa K, et al. Increased infiltration of tumor associated macrophages is associated with poor prognosis of bladder carcinoma in situ after intravesical bacillus Calmette-Guerin instillation. J Urol. 2009;181:1894–900. doi: 10.1016/j.juro.2008.11.090. [DOI] [PubMed] [Google Scholar]
  • 251.Nonomura N, Takayama H, Nakayama M, Nakai Y, Kawashima A, Mukai M, et al. Infiltration of tumour-associated macrophages in prostate biopsy specimens is predictive of disease progression after hormonal therapy for prostate cancer. BJU Int. 2011;107:1918–22. doi: 10.1111/j.1464-410X.2010.09804.x. [DOI] [PubMed] [Google Scholar]
  • 252.Al’-Shukri CKh, Rybakova MG, Makovskaia AI, Tiurin AG, Kozlov VV, Ageev MN, et al. [Prognostic implication of morphometric stromal parameters of renal pelvis and ureteral transitional cell carcinomas] Urologiia. 2004:10–4. [PubMed] [Google Scholar]
  • 253.Welsh TJ, Green RH, Richardson D, Waller DA, O’Byrne KJ, Bradding P. Macrophage and mast-cell invasion of tumor cell islets confers a marked survival advantage in non-small-cell lung cancer. J Clin Oncol. 2005;23:8959–67. doi: 10.1200/JCO.2005.01.4910. [DOI] [PubMed] [Google Scholar]
  • 254.Ohno S, Inagawa H, Dhar DK, Fujii T, Ueda S, Tachibana M, et al. The degree of macrophage infiltration into the cancer cell nest is a significant predictor of survival in gastric cancer patients. Anticancer Res. 2003;23(6D):5015–22. [PubMed] [Google Scholar]
  • 255.Varella AD, Bandiera DC, de Amorim AR, Sr., Calvis LA, Santos IO, Escaleira N, et al. Treatment of disseminated malignant melanoma with high-dose oral BCG. Cancer. 1981;48:1353–62. doi: 10.1002/1097-0142(19810915)48:6<1353::AID-CNCR2820480617>3.0.CO;2-N. [DOI] [PubMed] [Google Scholar]
  • 256.Forssell J, Oberg A, Henriksson ML, Stenling R, Jung A, Palmqvist R. High macrophage infiltration along the tumor front correlates with improved survival in colon cancer. Clin Cancer Res. 2007;13:1472–9. doi: 10.1158/1078-0432.CCR-06-2073. [DOI] [PubMed] [Google Scholar]
  • 257.Lackner C, Jukic Z, Tsybrovskyy O, Jatzko G, Wette V, Hoefler G, et al. Prognostic relevance of tumour-associated macrophages and von Willebrand factor-positive microvessels in colorectal cancer. Virchows Arch. 2004;445:160–7. doi: 10.1007/s00428-004-1051-z. [DOI] [PubMed] [Google Scholar]
  • 258.Zhou Q, Peng RQ, Wu XJ, Xia Q, Hou JH, Ding Y, et al. The density of macrophages in the invasive front is inversely correlated to liver metastasis in colon cancer. J Transl Med. 2010;8:13. doi: 10.1186/1479-5876-8-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Kinouchi M, Miura K, Mizoi T, Ishida K, Fujibuchi W, Sasaki H, et al. Infiltration of CD40-positive tumor-associated macrophages indicates a favorable prognosis in colorectal cancer patients. Hepatogastroenterology. 2012;60 doi: 10.5754/hge12372. In press. [DOI] [PubMed] [Google Scholar]
  • 260.Medrek C, Pontén F, Jirström K, Leandersson K. The presence of tumor associated macrophages in tumor stroma as a prognostic marker for breast cancer patients. BMC Cancer. 2012;12:306. doi: 10.1186/1471-2407-12-306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Zhang BC, Gao J, Wang J, Rao ZG, Wang BC, Gao JF. Tumor-associated macrophages infiltration is associated with peritumoral lymphangiogenesis and poor prognosis in lung adenocarcinoma. Med Oncol. 2011;28:1447–52. doi: 10.1007/s12032-010-9638-5. [DOI] [PubMed] [Google Scholar]
  • 262.Komohara Y, Hasita H, Ohnishi K, Fujiwara Y, Suzu S, Eto M, et al. Macrophage infiltration and its prognostic relevance in clear cell renal cell carcinoma. Cancer Sci. 2011;102:1424–31. doi: 10.1111/j.1349-7006.2011.01945.x. [DOI] [PubMed] [Google Scholar]
  • 263.Lee CH, Espinosa I, Vrijaldenhoven S, Subramanian S, Montgomery KD, Zhu S, et al. Prognostic significance of macrophage infiltration in leiomyosarcomas. Clin Cancer Res. 2008;14:1423–30. doi: 10.1158/1078-0432.CCR-07-1712. [DOI] [PubMed] [Google Scholar]
  • 264.Kamper P, Bendix K, Hamilton-Dutoit S, Honoré B, Nyengaard JR, d’Amore F. Tumor-infiltrating macrophages correlate with adverse prognosis and Epstein-Barr virus status in classical Hodgkin’s lymphoma. Haematologica. 2011;96:269–76. doi: 10.3324/haematol.2010.031542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Marigo I, Dolcetti L, Serafini P, Zanovello P, Bronte V. Tumor-induced tolerance and immune suppression by myeloid derived suppressor cells. Immunol Rev. 2008;222:162–79. doi: 10.1111/j.1600-065X.2008.00602.x. [DOI] [PubMed] [Google Scholar]
  • 266.Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9:162–74. doi: 10.1038/nri2506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Bunt SK, Sinha P, Clements VK, Leips J, Ostrand-Rosenberg S. Inflammation induces myeloid-derived suppressor cells that facilitate tumor progression. J Immunol. 2006;176:284–90. doi: 10.4049/jimmunol.176.1.284. [DOI] [PubMed] [Google Scholar]
  • 268.Brandau S, Trellakis S, Bruderek K, Schmaltz D, Steller G, Elian M, et al. Myeloid-derived suppressor cells in the peripheral blood of cancer patients contain a subset of immature neutrophils with impaired migratory properties. J Leukoc Biol. 2011;89:311–7. doi: 10.1189/jlb.0310162. [DOI] [PubMed] [Google Scholar]
  • 269.Solito S, Falisi E, Diaz-Montero CM, Doni A, Pinton L, Rosato A, et al. A human promyelocytic-like population is responsible for the immune suppression mediated by myeloid-derived suppressor cells. Blood. 2011;118:2254–65. doi: 10.1182/blood-2010-12-325753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Rodriguez PC, Ernstoff MS, Hernandez C, Atkins M, Zabaleta J, Sierra R, et al. Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res. 2009;69:1553–60. doi: 10.1158/0008-5472.CAN-08-1921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Kotsakis A, Harasymczuk M, Schilling B, Georgoulias V, Argiris A, Whiteside TL. Myeloid-derived suppressor cell measurements in fresh and cryopreserved blood samples. J Immunol Methods. 2012;381:14–22. doi: 10.1016/j.jim.2012.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Mundy-Bosse BL, Young GS, Bauer T, Binkley E, Bloomston M, Bill MA, et al. Distinct myeloid suppressor cell subsets correlate with plasma IL-6 and IL-10 and reduced interferon-alpha signaling in CD4⁺ T cells from patients with GI malignancy. Cancer Immunol Immunother. 2011;60:1269–79. doi: 10.1007/s00262-011-1029-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Zhao F, Hoechst B, Duffy A, Gamrekelashvili J, Fioravanti S, Manns MP, et al. S100A9 a new marker for monocytic human myeloid-derived suppressor cells. Immunology. 2012;136:176–83. doi: 10.1111/j.1365-2567.2012.03566.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Talmadge JE. Pathways mediating the expansion and immunosuppressive activity of myeloid-derived suppressor cells and their relevance to cancer therapy. Clin Cancer Res. 2007;13:5243–8. doi: 10.1158/1078-0432.CCR-07-0182. [DOI] [PubMed] [Google Scholar]
  • 275.Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Krüger C, Manns MP, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells. Gastroenterology. 2008;135:234–43. doi: 10.1053/j.gastro.2008.03.020. [DOI] [PubMed] [Google Scholar]
  • 276.Sinha P, Clements VK, Bunt SK, Albelda SM, Ostrand-Rosenberg S. Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response. J Immunol. 2007;179:977–83. doi: 10.4049/jimmunol.179.2.977. [DOI] [PubMed] [Google Scholar]
  • 277.Hoechst B, Voigtlaender T, Ormandy L, Gamrekelashvili J, Zhao F, Wedemeyer H, et al. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology. 2009;50:799–807. doi: 10.1002/hep.23054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Kujawski M, Kortylewski M, Lee H, Herrmann A, Kay H, Yu H. Stat3 mediates myeloid cell-dependent tumor angiogenesis in mice. J Clin Invest. 2008;118:3367–77. doi: 10.1172/JCI35213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Mauti LA, Le Bitoux MA, Baumer K, Stehle JC, Golshayan D, Provero P, et al. Myeloid-derived suppressor cells are implicated in regulating permissiveness for tumor metastasis during mouse gestation. J Clin Invest. 2011;121:2794–807. doi: 10.1172/JCI41936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Hock BD, Mackenzie KA, Cross NB, Taylor KG, Currie MJ, Robinson BA, et al. Renal transplant recipients have elevated frequencies of circulating myeloid-derived suppressor cells. Nephrol Dial Transplant. 2012;27:402–10. doi: 10.1093/ndt/gfr264. [DOI] [PubMed] [Google Scholar]
  • 281.Porembka MR, Mitchem JB, Belt BA, Hsieh CS, Lee HM, Herndon J, et al. Pancreatic adenocarcinoma induces bone marrow mobilization of myeloid-derived suppressor cells which promote primary tumor growth. Cancer Immunol Immunother. 2012;61:1373–85. doi: 10.1007/s00262-011-1178-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Diaz-Montero CM, Salem ML, Nishimura MI, Garrett-Mayer E, Cole DJ, Montero AJ. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol Immunother. 2009;58:49–59. doi: 10.1007/s00262-008-0523-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Tadmor T, Fell R, Polliack A, Attias D. Absolute monocytosis at diagnosis correlates with survival in diffuse large B-cell lymphoma-possible link with monocytic myeloid-derived suppressor cells. Hematol Oncol. 2012 doi: 10.1002/hon.2019. In press. [DOI] [PubMed] [Google Scholar]
  • 284.Montero AJ, Diaz-Montero CM, Deutsch YE, Hurley J, Koniaris LG, Rumboldt T, et al. Phase 2 study of neoadjuvant treatment with NOV-002 in combination with doxorubicin and cyclophosphamide followed by docetaxel in patients with HER-2 negative clinical stage II-IIIc breast cancer. Breast Cancer Res Treat. 2012;132:215–23. doi: 10.1007/s10549-011-1889-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Liu CY, Wang YM, Wang CL, Feng PH, Ko HW, Liu YH, et al. 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. 2010;136:35–45. doi: 10.1007/s00432-009-0634-0. [DOI] [PubMed] [Google Scholar]
  • 286.Ko JS, Zea AH, Rini BI, Ireland JL, Elson P, Cohen P, et al. Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients. Clin Cancer Res. 2009;15:2148–57. doi: 10.1158/1078-0432.CCR-08-1332. [DOI] [PubMed] [Google Scholar]
  • 287.Yuan XK, Zhao XK, Xia YC, Zhu X, Xiao P. Increased circulating immunosuppressive CD14(+)HLA-DR(-/low) cells correlate with clinical cancer stage and pathological grade in patients with bladder carcinoma. J Int Med Res. 2011;39:1381–91. doi: 10.1177/147323001103900424. [DOI] [PubMed] [Google Scholar]
  • 288.Gabitass RF, Annels NE, Stocken DD, Pandha HA, Middleton GW. Elevated myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin-13. Cancer Immunol Immunother. 2011;60:1419–30. doi: 10.1007/s00262-011-1028-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Choi J, Suh B, Ahn YO, Kim TM, Lee JO, Lee SH, et al. CD15+/CD16low human granulocytes from terminal cancer patients: granulocytic myeloid-derived suppressor cells that have suppressive function. Tumour Biol. 2012;33:121–9. doi: 10.1007/s13277-011-0254-6. [DOI] [PubMed] [Google Scholar]
  • 290.Gowda M, Godder K, Kmieciak M, Worschech A, Ascierto ML, Wang E, et al. Distinct signatures of the immune responses in low risk versus high risk neuroblastoma. J Transl Med. 2011;9:170. doi: 10.1186/1479-5876-9-170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Chu PG, Arber DA. CD79: a review. Appl Immunohistochem Mol Morphol. 2001;9:97–106. doi: 10.1097/00022744-200106000-00001. [DOI] [PubMed] [Google Scholar]
  • 292.Anderson SM, Tomayko MM, Shlomchik MJ. Intrinsic properties of human and murine memory B cells. Immunol Rev. 2006;211:280–94. doi: 10.1111/j.0105-2896.2006.00398.x. [DOI] [PubMed] [Google Scholar]
  • 293.Mazzucchelli R, Durum SK. Interleukin-7 receptor expression: intelligent design. Nat Rev Immunol. 2007;7:144–54. doi: 10.1038/nri2023. [DOI] [PubMed] [Google Scholar]
  • 294.Jourdan M, Caraux A, Caron G, Robert N, Fiol G, Rème T, et al. Characterization of a transitional preplasmablast population in the process of human B cell to plasma cell differentiation. J Immunol. 2011;187:3931–41. doi: 10.4049/jimmunol.1101230. [DOI] [PubMed] [Google Scholar]
  • 295.Carter RH, Barrington RA. Signaling by the CD19/CD21 complex on B cells. Curr Dir Autoimmun. 2004;7:4–32. doi: 10.1159/000075685. [DOI] [PubMed] [Google Scholar]
  • 296.Kuijpers TW, Bende RJ, Baars PA, Grummels A, Derks IA, Dolman KM, et al. CD20 deficiency in humans results in impaired T cell-independent antibody responses. J Clin Invest. 2010;120:214–22. doi: 10.1172/JCI40231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Esser C, Radbruch A. Immunoglobulin class switching: molecular and cellular analysis. Annu Rev Immunol. 1990;8:717–35. doi: 10.1146/annurev.iy.08.040190.003441. [DOI] [PubMed] [Google Scholar]
  • 298.Calame KL, Lin KI, Tunyaplin C. Regulatory mechanisms that determine the development and function of plasma cells. Annu Rev Immunol. 2003;21:205–30. doi: 10.1146/annurev.immunol.21.120601.141138. [DOI] [PubMed] [Google Scholar]
  • 299.Kurosaki T, Aiba Y, Kometani K, Moriyama S, Takahashi Y. Unique properties of memory B cells of different isotypes. Immunol Rev. 2010;237:104–16. doi: 10.1111/j.1600-065X.2010.00939.x. [DOI] [PubMed] [Google Scholar]
  • 300.Tangye SG, Good KL. Human IgM+CD27+ B cells: memory B cells or “memory” B cells? J Immunol. 2007;179:13–9. doi: 10.4049/jimmunol.179.1.13. [DOI] [PubMed] [Google Scholar]
  • 301.Basten A, Silveira PA. B-cell tolerance: mechanisms and implications. Curr Opin Immunol. 2010;22:566–74. doi: 10.1016/j.coi.2010.08.001. [DOI] [PubMed] [Google Scholar]
  • 302.Luning Prak ET, Monestier M, Eisenberg RA. B cell receptor editing in tolerance and autoimmunity. Ann N Y Acad Sci. 2011;1217:96–121. doi: 10.1111/j.1749-6632.2010.05877.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Mauri C, Blair PA. Regulatory B cells in autoimmunity: developments and controversies. Nat Rev Rheumatol. 2010;6:636–43. doi: 10.1038/nrrheum.2010.140. [DOI] [PubMed] [Google Scholar]
  • 304.Mauri C, Bosma A. Immune regulatory function of B cells. Annu Rev Immunol. 2012;30:221–41. doi: 10.1146/annurev-immunol-020711-074934. [DOI] [PubMed] [Google Scholar]
  • 305.Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, et al. Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ. 2012;19:107–20. doi: 10.1038/cdd.2011.96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Kroemer G, Galluzzi L, Brenner C. Mitochondrial membrane permeabilization in cell death. Physiol Rev. 2007;87:99–163. doi: 10.1152/physrev.00013.2006. [DOI] [PubMed] [Google Scholar]
  • 307.Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, et al. Nomenclature Committee on Cell Death 2009 Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 2009;16:3–11. doi: 10.1038/cdd.2008.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Chang SG, Lee SJ, Huh JS, Lee JH. Changes in mucosal immune cells of bladder tumor patient after BCG intravesical immunotherapy. Oncol Rep. 2001;8:257–61. [PubMed] [Google Scholar]
  • 309.Ladányi A, Kiss J, Mohos A, Somlai B, Liszkay G, Gilde K, et al. Prognostic impact of B-cell density in cutaneous melanoma. Cancer Immunol Immunother. 2011;60:1729–38. doi: 10.1007/s00262-011-1071-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Kay NE, Leong T, Kyle RA, Greipp P, Billadeau D, Van Ness B, et al. Circulating blood B cells in multiple myeloma: analysis and relationship to circulating clonal cells and clinical parameters in a cohort of patients entered on the Eastern Cooperative Oncology Group phase III E9486 clinical trial. Blood. 1997;90:340–5. [PubMed] [Google Scholar]
  • 311.Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol. 2006;6:173–82. doi: 10.1038/nri1785. [DOI] [PubMed] [Google Scholar]
  • 312.Witko-Sarsat V, Rieu P, Descamps-Latscha B, Lesavre P, Halbwachs-Mecarelli L. Neutrophils: molecules, functions and pathophysiological aspects. Lab Invest. 2000;80:617–53. doi: 10.1038/labinvest.3780067. [DOI] [PubMed] [Google Scholar]
  • 313.Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–5. doi: 10.1126/science.1092385. [DOI] [PubMed] [Google Scholar]
  • 314.Fridlender ZG, Albelda SM. Tumor-associated neutrophils: friend or foe? Carcinogenesis. 2012;33:949–55. doi: 10.1093/carcin/bgs123. [DOI] [PubMed] [Google Scholar]
  • 315.Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, et al. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell. 2009;16:183–94. doi: 10.1016/j.ccr.2009.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316.Bao Y, Cao X. Revisiting the protective and pathogenic roles of neutrophils: Ly-6G is key! Eur J Immunol. 2011;41:2535–8. doi: 10.1002/eji.201141979. [DOI] [PubMed] [Google Scholar]
  • 317.Stocks SC, Ruchaud-Sparagano MH, Kerr MA, Grunert F, Haslett C, Dransfield I. CD66: role in the regulation of neutrophil effector function. Eur J Immunol. 1996;26:2924–32. doi: 10.1002/eji.1830261218. [DOI] [PubMed] [Google Scholar]
  • 318.Elliott MA. Chronic neutrophilic leukemia: a contemporary review. Curr Hematol Rep. 2004;3:210–7. [PubMed] [Google Scholar]
  • 319.Kuang DM, Zhao Q, Wu Y, Peng C, Wang J, Xu Z, et al. Peritumoral neutrophils link inflammatory response to disease progression by fostering angiogenesis in hepatocellular carcinoma. J Hepatol. 2011;54:948–55. doi: 10.1016/j.jhep.2010.08.041. [DOI] [PubMed] [Google Scholar]
  • 320.Zhao JJ, Pan K, Wang W, Chen JG, Wu YH, Lv L, et al. The prognostic value of tumor-infiltrating neutrophils in gastric adenocarcinoma after resection. PLoS One. 2012;7:e33655. doi: 10.1371/journal.pone.0033655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321.Caruso RA, Bellocco R, Pagano M, Bertoli G, Rigoli L, Inferrera C. Prognostic value of intratumoral neutrophils in advanced gastric carcinoma in a high-risk area in northern Italy. Mod Pathol. 2002;15:831–7. doi: 10.1097/01.MP.0000020391.98998.6B. [DOI] [PubMed] [Google Scholar]
  • 322.Kita H. Eosinophils: multifaceted biological properties and roles in health and disease. Immunol Rev. 2011;242:161–77. doi: 10.1111/j.1600-065X.2011.01026.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Rothenberg ME, Hogan SP. The eosinophil. Annu Rev Immunol. 2006;24:147–74. doi: 10.1146/annurev.immunol.24.021605.090720. [DOI] [PubMed] [Google Scholar]
  • 324.Bochner BS, Schleimer RP. Mast cells, basophils, and eosinophils: distinct but overlapping pathways for recruitment. Immunol Rev. 2001;179:5–15. doi: 10.1034/j.1600-065X.2001.790101.x. [DOI] [PubMed] [Google Scholar]
  • 325.Fernvik E, Halldén G, Hed J, Lundahl J. Intracellular and surface distribution of CD9 in human eosinophils. APMIS. 1995;103:699–706. doi: 10.1111/j.1699-0463.1995.tb01426.x. [DOI] [PubMed] [Google Scholar]
  • 326.Matsumoto K, Appiah-Pippim J, Schleimer RP, Bickel CA, Beck LA, Bochner BS. CD44 and CD69 represent different types of cell-surface activation markers for human eosinophils. Am J Respir Cell Mol Biol. 1998;18:860–6. doi: 10.1165/ajrcmb.18.6.3159. [DOI] [PubMed] [Google Scholar]
  • 327.Moqbel R, Barkans J, Bradley BL, Durham SR, Kay AB. Application of monoclonal antibodies against major basic protein (BMK-13) and eosinophil cationic protein (EG1 and EG2) for quantifying eosinophils in bronchial biopsies from atopic asthma. Clin Exp Allergy. 1992;22:265–73. doi: 10.1111/j.1365-2222.1992.tb03082.x. [DOI] [PubMed] [Google Scholar]
  • 328.Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008;8:618–31. doi: 10.1038/nrc2444. [DOI] [PubMed] [Google Scholar]
  • 329.Sassler AM, McClatchey KD, Wolf GT, Fisher SG, Veterans Administration Laryngeal Cooperative Study Group Eosinophilic infiltration in advanced laryngeal squamous cell carcinoma. Laryngoscope. 1995;105:413–6. doi: 10.1288/00005537-199504000-00014. [DOI] [PubMed] [Google Scholar]
  • 330.Tadbir AA, Ashraf MJ, Sardari Y. Prognostic significance of stromal eosinophilic infiltration in oral squamous cell carcinoma. J Craniofac Surg. 2009;20:287–9. doi: 10.1097/SCS.0b013e318199219b. [DOI] [PubMed] [Google Scholar]
  • 331.Alrawi SJ, Tan D, Stoler DL, Dayton M, Anderson GR, Mojica P, et al. Tissue eosinophilic infiltration: a useful marker for assessing stromal invasion, survival and locoregional recurrence in head and neck squamous neoplasia. Cancer J. 2005;11:217–25. doi: 10.1097/00130404-200505000-00008. [DOI] [PubMed] [Google Scholar]
  • 332.Horiuchi K, Mishima K, Ohsawa M, Sugimura M, Aozasa K. Prognostic factors for well-differentiated squamous cell carcinoma in the oral cavity with emphasis on immunohistochemical evaluation. J Surg Oncol. 1993;53:92–6. doi: 10.1002/jso.2930530209. [DOI] [PubMed] [Google Scholar]
  • 333.Fujii M, Yamashita T, Ishiguro R, Tashiro M, Kameyama K. Significance of epidermal growth factor receptor and tumor associated tissue eosinophilia in the prognosis of patients with nasopharyngeal carcinoma. Auris Nasus Larynx. 2002;29:175–81. doi: 10.1016/S0385-8146(01)00135-3. [DOI] [PubMed] [Google Scholar]
  • 334.Cuschieri A, Talbot IC, Weeden S, MRC Upper GI Cancer Working Party Influence of pathological tumour variables on long-term survival in resectable gastric cancer. Br J Cancer. 2002;86:674–9. doi: 10.1038/sj.bjc.6600161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335.Iwasaki K, Torisu M, Fujimura T. Malignant tumor and eosinophils. I. Prognostic significance in gastric cancer. Cancer. 1986;58:1321–7. doi: 10.1002/1097-0142(19860915)58:6<1321::AID-CNCR2820580623>3.0.CO;2-O. [DOI] [PubMed] [Google Scholar]
  • 336.Hayes RL, Arbit E, Odaimi M, Pannullo S, Scheff R, Kravchinskiy D, et al. Adoptive cellular immunotherapy for the treatment of malignant gliomas. Crit Rev Oncol Hematol. 2001;39:31–42. doi: 10.1016/S1040-8428(01)00122-6. [DOI] [PubMed] [Google Scholar]
  • 337.Takanami I, Takeuchi K, Gika M. Immunohistochemical detection of eosinophilic infiltration in pulmonary adenocarcinoma. Anticancer Res. 2002;22:2391–6. [PubMed] [Google Scholar]
  • 338.Fernández-Aceñero MJ, Galindo-Gallego M, Sanz J, Aljama A. Prognostic influence of tumor-associated eosinophilic infiltrate in colorectal carcinoma. Cancer. 2000;88:1544–8. doi: 10.1002/(SICI)1097-0142(20000401)88:7<1544::AID-CNCR7>3.0.CO;2-S. [DOI] [PubMed] [Google Scholar]
  • 339.Kapp DS, LiVolsi VA. Intense eosinophilic stromal infiltration in carcinoma of the uterine cervix: a clinicopathologic study of 14 cases. Gynecol Oncol. 1983;16:19–30. doi: 10.1016/0090-8258(83)90004-5. [DOI] [PubMed] [Google Scholar]
  • 340.Schroeder JT. Basophils beyond effector cells of allergic inflammation. Adv Immunol. 2009;101:123–61. doi: 10.1016/S0065-2776(08)01004-3. [DOI] [PubMed] [Google Scholar]
  • 341.Heneberg P. Mast cells and basophils: trojan horses of conventional lin- stem/progenitor cell isolates. Curr Pharm Des. 2011;17:3753–71. doi: 10.2174/138161211798357881. [DOI] [PubMed] [Google Scholar]
  • 342.Karasuyama H, Mukai K, Obata K, Tsujimura Y, Wada T. Nonredundant roles of basophils in immunity. Annu Rev Immunol. 2011;29:45–69. doi: 10.1146/annurev-immunol-031210-101257. [DOI] [PubMed] [Google Scholar]
  • 343.Karasuyama H, Mukai K, Tsujimura Y, Obata K. Newly discovered roles for basophils: a neglected minority gains new respect. Nat Rev Immunol. 2009;9:9–13. doi: 10.1038/nri2458. [DOI] [PubMed] [Google Scholar]
  • 344.Gauchat JF, Henchoz S, Mazzei G, Aubry JP, Brunner T, Blasey H, et al. Induction of human IgE synthesis in B cells by mast cells and basophils. Nature. 1993;365:340–3. doi: 10.1038/365340a0. [DOI] [PubMed] [Google Scholar]
  • 345.Yasuda H, Aritaka N, Ando J, Hirama M, Komatsu N, Hirano T. Chronic myelogenous leukemia with mild basophilia as the predominant manifestation at presentation. Intern Med. 2011;50:501–2. doi: 10.2169/internalmedicine.50.4695. [DOI] [PubMed] [Google Scholar]
  • 346.Rodriguez J, Cortes J, Smith T, O’Brien S, Rios MB, Talpaz M, et al. Determinants of prognosis in late chronic-phase chronic myelogenous leukemia. J Clin Oncol. 1998;16:3782–7. doi: 10.1200/JCO.1998.16.12.3782. [DOI] [PubMed] [Google Scholar]
  • 347.Hasford J, Baccarani M, Hoffmann V, Guilhot J, Saussele S, Rosti G, et al. Predicting complete cytogenetic response and subsequent progression-free survival in 2060 patients with CML on imatinib treatment: the EUTOS score. Blood. 2011;118:686–92. doi: 10.1182/blood-2010-12-319038. [DOI] [PubMed] [Google Scholar]
  • 348.Matsushima T, Handa H, Yokohama A, Nagasaki J, Koiso H, Kin Y, et al. Prevalence and clinical characteristics of myelodysplastic syndrome with bone marrow eosinophilia or basophilia. Blood. 2003;101:3386–90. doi: 10.1182/blood-2002-03-0947. [DOI] [PubMed] [Google Scholar]
  • 349.Prussin C, Metcalfe DD. 4. IgE, mast cells, basophils, and eosinophils. J Allergy Clin Immunol. 2003;111(Suppl):S486–94. doi: 10.1067/mai.2003.120. [DOI] [PubMed] [Google Scholar]
  • 350.Galli SJ, Grimbaldeston M, Tsai M. Immunomodulatory mast cells: negative, as well as positive, regulators of immunity. Nat Rev Immunol. 2008;8:478–86. doi: 10.1038/nri2327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351.Bischoff SC. Role of mast cells in allergic and non-allergic immune responses: comparison of human and murine data. Nat Rev Immunol. 2007;7:93–104. doi: 10.1038/nri2018. [DOI] [PubMed] [Google Scholar]
  • 352.Hopkin J. Immune and genetic aspects of asthma, allergy and parasitic worm infections: evolutionary links. Parasite Immunol. 2009;31:267–73. doi: 10.1111/j.1365-3024.2009.01104.x. [DOI] [PubMed] [Google Scholar]
  • 353.Yang Z, Zhang B, Li D, Lv M, Huang C, Shen GX, et al. Mast cells mobilize myeloid-derived suppressor cells and Treg cells in tumor microenvironment via IL-17 pathway in murine hepatocarcinoma model. PLoS One. 2010;5:e8922. doi: 10.1371/journal.pone.0008922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354.Ribatti D, Crivellato E. Mast cells, angiogenesis, and tumour growth. Biochim Biophys Acta. 2012;1822:2–8. doi: 10.1016/j.bbadis.2010.11.010. [DOI] [PubMed] [Google Scholar]
  • 355.Dyduch G, Kaczmarczyk K, Okoń K. Mast cells and cancer: enemies or allies? Pol J Pathol. 2012;63:1–7. [PubMed] [Google Scholar]
  • 356.Khazaie K, Blatner NR, Khan MW, Gounari F, Gounaris E, Dennis K, et al. The significant role of mast cells in cancer. Cancer Metastasis Rev. 2011;30:45–60. doi: 10.1007/s10555-011-9286-z. [DOI] [PubMed] [Google Scholar]
  • 357.Drew E, Merkens H, Chelliah S, Doyonnas R, McNagny KM. CD34 is a specific marker of mature murine mast cells. Exp Hematol. 2002;30:1211–8. doi: 10.1016/S0301-472X(02)00890-1. [DOI] [PubMed] [Google Scholar]
  • 358.Arber DA, Tamayo R, Weiss LM. Paraffin section detection of the c-kit gene product (CD117) in human tissues: value in the diagnosis of mast cell disorders. Hum Pathol. 1998;29:498–504. doi: 10.1016/S0046-8177(98)90066-1. [DOI] [PubMed] [Google Scholar]
  • 359.Sahashi K, Ibi T, Zhang G. [Immunohistochemical localization of chymase; a mast cell marker and clinical significance in diseased human skeletal muscle] Rinsho Shinkeigaku. 1997;37:615–20. [PubMed] [Google Scholar]
  • 360.Drew E, Merzaban JS, Seo W, Ziltener HJ, McNagny KM. CD34 and CD43 inhibit mast cell adhesion and are required for optimal mast cell reconstitution. Immunity. 2005;22:43–57. doi: 10.1016/j.immuni.2004.11.014. [DOI] [PubMed] [Google Scholar]
  • 361.Yano H, Kinuta M, Tateishi H, Nakano Y, Matsui S, Monden T, et al. Mast cell infiltration around gastric cancer cells correlates with tumor angiogenesis and metastasis. Gastric Cancer. 1999;2:26–32. doi: 10.1007/s101200050017. [DOI] [PubMed] [Google Scholar]
  • 362.Takanami I, Takeuchi K, Naruke M. Mast cell density is associated with angiogenesis and poor prognosis in pulmonary adenocarcinoma. Cancer. 2000;88:2686–92. doi: 10.1002/1097-0142(20000615)88:12<2686::AID-CNCR6>3.0.CO;2-6. [DOI] [PubMed] [Google Scholar]
  • 363.Chang DZ, Ma Y, Ji B, Wang H, Deng D, Liu Y, et al. Mast cells in tumor microenvironment promotes the in vivo growth of pancreatic ductal adenocarcinoma. Clin Cancer Res. 2011;17:7015–23. doi: 10.1158/1078-0432.CCR-11-0607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 364.Strouch MJ, Cheon EC, Salabat MR, Krantz SB, Gounaris E, Melstrom LG, et al. Crosstalk between mast cells and pancreatic cancer cells contributes to pancreatic tumor progression. Clin Cancer Res. 2010;16:2257–65. doi: 10.1158/1078-0432.CCR-09-1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365.Tuna B, Yorukoglu K, Unlu M, Mungan MU, Kirkali Z. Association of mast cells with microvessel density in renal cell carcinomas. Eur Urol. 2006;50:530–4. doi: 10.1016/j.eururo.2005.12.040. [DOI] [PubMed] [Google Scholar]
  • 366.Rajput AB, Turbin DA, Cheang MC, Voduc DK, Leung S, Gelmon KA, et al. Stromal mast cells in invasive breast cancer are a marker of favourable prognosis: a study of 4,444 cases. Breast Cancer Res Treat. 2008;107:249–57. doi: 10.1007/s10549-007-9546-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 367.Ranieri G, Achille G, Florio G, Labriola A, Marzullo F, Paradiso A, et al. [Biological-clinical significance of angiogenesis and mast cell infiltration in squamous cell carcinoma of the oral cavity] Acta Otorhinolaryngol Ital. 2001;21:171–8. [PubMed] [Google Scholar]
  • 368.Chan JK, Magistris A, Loizzi V, Lin F, Rutgers J, Osann K, et al. Mast cell density, angiogenesis, blood clotting, and prognosis in women with advanced ovarian cancer. Gynecol Oncol. 2005;99:20–5. doi: 10.1016/j.ygyno.2005.05.042. [DOI] [PubMed] [Google Scholar]
  • 369.Galon J, Pagès F, Marincola FM, Thurin M, Trinchieri G, Fox BA, et al. The immune score as a new possible approach for the classification of cancer. J Transl Med. 2012;10:1. doi: 10.1186/1479-5876-10-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 370.Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–44. doi: 10.1038/nature07205. [DOI] [PubMed] [Google Scholar]
  • 371.Gulubova MV, Ananiev JR, Vlaykova TI, Yovchev Y, Tsoneva V, Manolova IM. Role of dendritic cells in progression and clinical outcome of colon cancer. Int J Colorectal Dis. 2012;27:159–69. doi: 10.1007/s00384-011-1334-1. [DOI] [PubMed] [Google Scholar]
  • 372.Halama N, Michel S, Kloor M, Zoernig I, Benner A, Spille A, et al. Localization and density of immune cells in the invasive margin of human colorectal cancer liver metastases are prognostic for response to chemotherapy. Cancer Res. 2011;71:5670–7. doi: 10.1158/0008-5472.CAN-11-0268. [DOI] [PubMed] [Google Scholar]
  • 373.Guy CS, Vignali DA. Organization of proximal signal initiation at the TCR:CD3 complex. Immunol Rev. 2009;232:7–21. doi: 10.1111/j.1600-065X.2009.00843.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374.Komatsu N, Mariotti-Ferrandiz ME, Wang Y, Malissen B, Waldmann H, Hori S. Heterogeneity of natural Foxp3+ T cells: a committed regulatory T-cell lineage and an uncommitted minor population retaining plasticity. Proc Natl Acad Sci U S A. 2009;106:1903–8. doi: 10.1073/pnas.0811556106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 375.Hoffmann P, Boeld TJ, Eder R, Huehn J, Floess S, Wieczorek G, et al. Loss of FOXP3 expression in natural human CD4+CD25+ regulatory T cells upon repetitive in vitro stimulation. Eur J Immunol. 2009;39:1088–97. doi: 10.1002/eji.200838904. [DOI] [PubMed] [Google Scholar]
  • 376.Wang X, Yu J, Sreekumar A, Varambally S, Shen R, Giacherio D, et al. Autoantibody signatures in prostate cancer. N Engl J Med. 2005;353:1224–35. doi: 10.1056/NEJMoa051931. [DOI] [PubMed] [Google Scholar]
  • 377.Touzé A, Le Bidre E, Laude H, Fleury MJ, Cazal R, Arnold F, et al. High levels of antibodies against merkel cell polyomavirus identify a subset of patients with merkel cell carcinoma with better clinical outcome. J Clin Oncol. 2011;29:1612–9. doi: 10.1200/JCO.2010.31.1704. [DOI] [PubMed] [Google Scholar]
  • 378.Apetoh L, Ghiringhelli F, Tesniere A, Criollo A, Ortiz C, Lidereau R, et al. The interaction between HMGB1 and TLR4 dictates the outcome of anticancer chemotherapy and radiotherapy. Immunol Rev. 2007;220:47–59. doi: 10.1111/j.1600-065X.2007.00573.x. [DOI] [PubMed] [Google Scholar]
  • 379.Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat Med. 2009;15:1170–8. doi: 10.1038/nm.2028. [DOI] [PubMed] [Google Scholar]
  • 380.Thunberg U, Tobin G, Johnson A, Söderberg O, Padyukov L, Hultdin M, et al. Polymorphism in the P2X7 receptor gene and survival in chronic lymphocytic leukaemia. Lancet. 2002;360:1935–9. doi: 10.1016/S0140-6736(02)11917-9. [DOI] [PubMed] [Google Scholar]
  • 381.Vacchelli E, Galluzzi L, Rousseau V, Rigoni A, Tesniere A, Delahaye N, et al. Loss-of-function alleles of P2RX7 and TLR4 fail to affect the response to chemotherapy in non-small cell lung cancer. Oncoimmunology. 2012;1:271–8. doi: 10.4161/onci.18684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 382.Wiley JS, Dao-Ung LP, Gu BJ, Sluyter R, Shemon AN, Li C, et al. A loss-of-function polymorphic mutation in the cytolytic P2X7 receptor gene and chronic lymphocytic leukaemia: a molecular study. Lancet. 2002;359:1114–9. doi: 10.1016/S0140-6736(02)08156-4. [DOI] [PubMed] [Google Scholar]
  • 383.Eruslanov E, Neuberger M, Daurkin I, Perrin GQ, Algood C, Dahm P, et al. Circulating and tumor-infiltrating myeloid cell subsets in patients with bladder cancer. Int J Cancer. 2012;130:1109–19. doi: 10.1002/ijc.26123. [DOI] [PubMed] [Google Scholar]
  • 384.Ohki S, Shibata M, Gonda K, Machida T, Shimura T, Nakamura I, et al. Circulating myeloid-derived suppressor cells are increased and correlate to immune suppression, inflammation and hypoproteinemia in patients with cancer. Oncol Rep. 2012;28:453–8. doi: 10.3892/or.2012.1812. [DOI] [PubMed] [Google Scholar]

Articles from Oncoimmunology are provided here courtesy of Taylor & Francis

RESOURCES