Next Article in Journal
T-Regulatory Cells and Vaccination “Pay Attention and Do Not Neglect Them”: Lessons from HIV and Cancer Vaccine Trials
Next Article in Special Issue
Neutrophils and Granulocytic MDSC: The Janus God of Cancer Immunotherapy
Previous Article in Journal / Special Issue
Regulatory T Cells in the Tumor Microenvironment and Cancer Progression: Role and Therapeutic Targeting
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Monitoring of the Immune Dysfunction in Cancer Patients

by
Saskia J. A. M. Santegoets
,
Marij J. P. Welters
and
Sjoerd H. Van der Burg
*
Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
*
Author to whom correspondence should be addressed.
Vaccines 2016, 4(3), 29; https://doi.org/10.3390/vaccines4030029
Submission received: 7 July 2016 / Revised: 25 August 2016 / Accepted: 27 August 2016 / Published: 2 September 2016
(This article belongs to the Special Issue Mechanisms of Tumor Escape from Host Immunity)

Abstract

:
Immunotherapy shows promising clinical results in patients with different types of cancer, but its full potential is not reached due to immune dysfunction as a result of several suppressive mechanisms that play a role in cancer development and progression. Monitoring of immune dysfunction is a prerequisite for the development of strategies aiming to alleviate cancer-induced immune suppression. At this point, the level at which immune dysfunction occurs has to be established, the underlying mechanism(s) need to be known, as well as the techniques to assess this. While it is relatively easy to measure general signs of immune suppression, it turns out that accurate monitoring of the frequency and function of immune-suppressive cells is still difficult. A lack of truly specific markers, the phenotypic complexity among suppressive cells of the same lineage, but potentially with different functions and functional assays that may not cover every mechanistic aspect of immune suppression are among the reasons complicating proper assessments. Technical innovations in flow and mass cytometry will allow for more complete sets of markers to precisely determine phenotype and associated function. There is, however, a clear need for functional assays that recapitulate more of the mechanisms employed to suppress the immune system.

1. Introduction

The common treatment modalities for curing cancer or decreasing tumor load are surgery, chemotherapy and radiotherapy. Recent clinical success has also been reported by a fourth therapy option, called immunotherapy. In cancer immunotherapy, the immune system is reinforced to fight cancer either by: (i) increasing the number of cytotoxic lymphocytes (for example, by vaccination, by adoptive transfer of autologous tumor-specific T cells, receptor engineered T cells or ex vivo-activated NK cells); (ii) activating the endogenous tumor-specific T cells by agonistic antibodies against co-stimulatory molecules (e.g., CD28, CD137, CD154, tumor necrosis factor receptor superfamily member 4 (OX40) and inducible T cell costimulator (ICOS)); (iii) blocking the checkpoint pathways that act on the anti-tumor T cells through administration of antibodies against co-inhibitory molecules (e.g., cytotoxic T lymphocyte antigen 4 (CTLA-4), programmed cell death protein 1 (PD-1), T cell immunoglobulin and mucin protein 3 (TIM3), V-domain Immunoglobulin suppressor of T cell activation (VISTA)); (iv) alleviation of the suppression induced by regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), tumor-promoting macrophages (so-called type 2 macrophages (M2) or TAMs) or type 2 tumor-associated neutrophils (N2 or TANs); and/or (v) by making use of monoclonal antibodies (mAbs) directly targeting tumor cells (e.g., CD20, HER-2, CD52 and epidermal growth factor receptor (EGFR)). Notably, these tumor-directed mAbs will not be discussed in this review. While many clinical successes are reported with these treatments, mitigation of immune cell suppression has been found to be tough.
Examples of clinically-successful immunotherapeutic approaches are especially found among treatments for melanoma. Major clinical improvement has been obtained by using checkpoint blocking antibodies against CTLA-4, PD-1 and PD-1 ligand (PDL-1), as well as their combination in advanced melanoma patients [1,2,3,4]. Similar success has been observed in other types of immunogenic cancers [5,6,7]. Furthermore, the adoptive cell transfer (ACT) of tumor antigen-specific T cells obtained and expanded from autologous tumor infiltrating lymphocytes [8,9] or peripheral blood mononuclear cells (PBMC) [10,11] has resulted in durable clinical responses and complete responses in late-stage melanoma patients. Another successful adoptive cell therapy is by transfer of genetically-engineered chimeric antigen receptors (CAR)-expressing cytotoxic T cells. CAR T cells express an extracellular tumor antigen recognition domain that is composed of a single chain variable fragment from an antibody and intracellular signaling and/or co-stimulatory domains allowing for T cell activation [12,13]. As an example, adoptive transfer of CD19-CAR T cells has resulted in remarkable clinical responses in patients with B cell malignancies [13,14]. Furthermore, reinfusion of ex vivo cytokine-activated haplo-identical natural killer (NK) cells has also led to promising anti-tumor responses in patients with hematological malignancies [15,16].
Beside passive infusion of relative high numbers of tumor-specific T cells, one can also actively stimulate the autologous T cell repertoire of patients, for instance through provision of agonistic antibodies directed to co-stimulatory molecules on T cells [17,18,19,20,21] or by injecting vaccines. Therapeutic cancer vaccines come in many flavors. Vaccine platforms include DNA, RNA, proteins or synthetic long peptides (SLP) encoding for tumor-specific or tumor-associated antigens, as well as vector-based vaccines (e.g., dendritic cell (DC) vaccines, bacterial and viral vectors, nanoparticles) [22,23,24]. Especially in the pre-malignant setting, we and others have shown that multiple vaccine platforms induced complete and partial regressions [25,26,27,28,29]. In the cancer setting, progress is slower. Examples of therapeutic cancer vaccines with clinical benefit include Sipuleucel-T immunotherapy for castration-resistant prostate cancer [30,31], the oncolytic herpes virus Talimogene Laherparepvec (T-VEC) for advanced melanoma patients [32,33], the granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transfected tumor cell vaccine (GVAX) used to prime combined with Listeria monocytogenes-expressing Mesothelin booster vaccine for metastatic pancreas cancer [34,35] and multiple tumor antigen-loaded autologous DC vaccines for advanced melanoma, hepatocellular carcinoma, malignant glioma and renal cell carcinoma patients [36,37,38].
These studies demonstrate that tumor-specific T cells can successfully combat cancer. However, tumors may escape from the immune system by strategies aimed at avoiding T cell recognition, including HLA downregulation [39] and/or the deletion, modification or decreased expression of tumor antigens [40]. Moreover, in many cases, cancer-associated immune suppressive networks may kick in to impair the induction, activation, expansion or the execution of the effector function of tumor antigen-specific T cells. Here, we briefly review the current knowledge on the different levels of this immune dysfunction present in cancer patients and discuss how to monitor this (Figure 1), as this will be important when new immunotherapeutic strategies are designed in which the alleviation of immune suppression is one of the goals.

2. General Signs of Immune Suppression

Tumor cells are capable of releasing various cytokines (reviewed in [41,42,43]), including interleukin-1 (IL-1), IL-6, IL-10, prostaglandin E2 (PGE2), transforming growth factor beta (TGF-β), vascular endothelial growth factor (VEGF), macrophage colony-stimulating factor (M-CSF), macrophage migration inhibitory factor (MIF) and granulocyte-macrophage colony stimulating factor (GM-CSF) [44,45,46,47]. In addition, tumor cells can also shed alarm proteins, such as the major histocompatibility complex (MHC) class I polypeptide-related sequence A (MICA) or B7-H6, thereby dampening NKG2D-mediated activation of T cells and NK cells or NKp30-mediated activation of NK cells [48,49]. Furthermore, tumor cells can release soluble factors, like chemokines, matrix metalloproteinases (MMPs) [50] and gangliosides [51], exosomes (small endosomal membrane vesicles that are enriched for tumor biomarkers, such as DNA, RNA and proteins [52]), as well as lactate [53], via which they can suppress immune responses. Interestingly, all of these tumor-derived factors can operate locally within the tumor microenvironment (TME), but also spread systemically via the blood compartment to mediate distant effects (e.g., on bone marrow, lymph nodes and spleen) and thus mediate systemic immune suppression. Many of these factors can be measured in blood plasma and/or serum and are, therefore, studied for their use as biomarkers for tumor progression or treatment efficacy, but they may also be used to obtain indications for immune suppression.
Cytokine, chemokine and MMP levels are measured by ELISA, multiplex immunobead assays (i.e., Luminex or cytometric bead array) [54,55,56,57] or other assays, such as solid-phase proximity ligation assay or a combined immunoprecipitation and mass spectrometry assay [58,59]. Gangliosides can be detected in blood plasma by a chromatographic mass spectrometry-based assay [60], while antibodies against these gangliosides can be detected by ELISA. Tumor-derived exosomes can be isolated and characterized by novel state-of-the-art microfluidic lab-on-a-chip technology [61,62,63]. The secreted tumor-derived factors have been shown to impact lymphoid and myeloid subsets by skewing them towards immunosuppressive cells. They contribute to the inhibition of effector T cells and the induction of Tregs [64], as well as to the differentiation of myeloid cells into tolerogenic DCs [65,66,67], TAMs [68], MDSCs [69,70] or the attraction and/or induction of TANs [71,72] that share features with MDSCs [72]. Significant negative correlations between serum levels of IL-6 [73,74], TGF-β [75,76] and IL-10 [77] and disease-free and/or overall survival were observed in multiple cancer types, suggesting that the measurement of these soluble factors can be used to assess whether immune suppression can be expected.
Another parameter that can be used to assess general immune dysfunction in cancer patients is the abnormal frequencies of certain types of immune cells in blood. Abnormal high counts of leukocytes (leukocytosis) are one of these. Leukocytosis is observed in patients with colorectal cancer [78], lung cancer [79], cervical cancer [80] and ovarian cancer [81] and served as an indicator of poor prognosis in these patients. In the blood of advanced-stage cervical cancer patients, the number of monocytes were found to be much higher compared to those in healthy donors, and this was associated with immune suppression [82]. Similar observations were made in patients suffering from different types of cancer [83,84,85,86]. Alternatively, leukopenia, which is decreased numbers of leukocytes (or subsets), has also been observed in some types of cancer (e.g., head and neck, colon, lung cancer and Hodgkin lymphoma) [87,88]. Differential cell counts in whole blood, which are routinely done in every hospital, can reveal whether the total leukocyte, as well as the subset cell counts are within the normal range or are altered. Moreover, the lymphocyte to monocyte ratio, the platelet to lymphocyte ratio or others have been shown to be associated with the response to therapy and/or clinical outcome [89]. Most recently, it was found that an increased absolute number of lymphocytes [90] or a low neutrophil to lymphocyte ratio in the blood [91] were associated with less suppression and a better response to checkpoint therapy.
There are also functional approaches to assess systemic immune suppression through the analysis of general antigen-presenting cell (APC), T cell and NK cell function. The capacity of circulating APCs to stimulate T cells can be easily tested using a mixed lymphocyte reaction (MLR). In this assay, the capacity of APC to stimulate proliferation and cytokine production of alloreactive HLA-mismatched T cells is assessed [82,92]. In pancreatic cancer patients, the circulating DCs were not only found to be decreased in number, but also their stimulatory function was impaired when measured by MLR [93]. Furthermore, in other types of cancer, the functional capacity of DCs was found to be impaired [94,95,96,97]. To test the functional capability of circulating T cells, PBMC can also be tested for their responsiveness to activating signals. General T cell function can be assessed by analyzing the proliferation and/or cytokine production of PBMC by mitogenic (chemical) compounds like phytohemagglutinin (PHA) [98] and staphylococcal enterotoxin B (SEB) [99] in a non-HLA restricted and APC-independent manner. A more subtle approach to test T cell functionality is by analyzing T cell proliferation and/or cytokine production in response to common bacterial and viral antigens, the so-called recall antigens. Examples of such recall antigens are proteins derived from influenza virus, cytomegalovirus (CMV), Epstein-Barr virus (EBV), tetanus toxoid, Mycobacterium tuberculosis and Candida albicans. Alleviation of suppression, as measured by improved T cell function (either by increased proliferation or cytokine production) against recall antigens, could be observed in cancer patients upon anti-tumor therapy [82,100,101]. Of note, these analyses can give valuable information on the level of T cell suppression, as the absence of T cell responsiveness following the strong mitogenic PHA stimulation may reveal T cell intrinsic problems, and the absence of recall antigen-specific responses may be indicative of a state of more general tumor-induced immune suppression. To test the functional activity of circulating NK cells, which is often reduced in patients with cancer [102,103], PBMC can be tested for their cytotoxic activity against NK cell targets (i.e., MHC-devoid targets, such as K562 cells) by the standard 51chromium release assay or CD107a (lysosome-associated membrane protein 1 (LAMP-1)) flow cytometric degranulation assay [104].

3. Immune Dysfunction through the Induction of Suppressor Cells

The role of lymphoid and myeloid suppressor cells in tumor development and progression has been studied extensively over the past decades [64,68,69,105,106]. By making use of cell-depleting agents or conditional cell ablation models based on the diphtheria toxin receptor, the role and contribution of specific immune cell subsets in the suppression of anti-tumor immune responses have been revealed in preclinical settings. Ablation of Tregs can result in dramatic tumor reduction and/or complete tumor clearance of large established tumors [107,108,109]. Similarly, the suppressive role of MDSC, TAM and TAN have also been demonstrated [110,111,112,113,114], emphasizing that several types of immune cells play an important role in suppressing an (initially) effective anti-tumor response.
Obviously, it is much harder to study the role of lymphoid and myeloid suppressor cells in human beings. Generally, the functional impact of such cells is determined by the association in that the frequency of certain phenotypic populations of immune cells is increased in the blood or tumor of patients with a higher stage of disease or in patients with a worse immunological response or clinical outcome. A major obstacle in this type of analysis is that the unambiguous enumeration of these immunosuppressive cell subsets is hampered by the absence of exclusive, highly specific markers for functionally-active cells. While in mice, specific markers for MDSC and Treg detection have been identified (Gr-1 and its isoforms Ly6C and Ly6G for MDSC and Foxp3 for Treg detection), in humans, the identification of these cells is more complex, as Gr-1 is not expressed on human leukocytes [115], and Foxp3 can also be expressed on activated non-regulatory T cells [116,117]. As a result, a multitude of human MDSC and Treg subsets with different phenotypes has been documented in several types of tumors in the last decades [118,119]. As an example, a recent in-depth phenotypic analysis of human Tregs revealed 22 distinct subpopulations [120], while the myeloid cell subpopulations exceeded one hundred [121]. This makes correct interpretation of data and comparison between studies difficult. To tackle the heterogeneity in current human MDSC and Treg phenotyping panels, proficiency panels and workshops aiming at harmonization of their detection through developing robust marker combinations and gating strategies are being performed [122,123].
So far, there were a number of studies showing that significantly higher levels of Tregs [124,125,126,127], MDSC [90,128,129,130,131], (tumor-associated) macrophages [85,132,133] and neutrophils [105,134,135] could be detected in the peripheral blood and TME of almost all types of cancer, merely in advanced stages of the disease, and these high levels usually negatively correlated with clinical outcome and/or survival.
Despite advances in the formulation of essential marker sets and gating strategies for such analyses, data on their functionality is still lacking and, as such, the link between phenotype and function. Since functional assessment of immune suppressor cells in the TME usually is not feasible due to limited tissue material, more in-depth analysis of (surrogate) markers for immune suppressor cell functionality would be an attractive approach to gain some more insight into their suppressive capacity. Examples of such markers include arginase 1 (Arg1), inducible nitric oxide synthase (iNOS), reactive oxygen species (ROS), TGF-β, indoleamine 2,3-dioxygenase (IDO) and IL-10, all of which can be expressed by myeloid suppressor cells [70,105,136]. The T cell-suppressive factors Arg1, ROS and IL-10 can also be produced by tumor-promoting TANs [137]. In addition, they can produce macrophage inflammatory protein 1-alpha (MIP-1α or CCL3), which recruits more myeloid cells into the tumor bed [70,72,105].
The activity of the enzyme Arg1 leads to increased L-arginine (L-arg) metabolism and converting it into ornithine and urea [138,139]. This way, it results in the deprivation of L-arg for the T cells, which is required for their proliferation and metabolism [140], and subsequently leads to impairment of T cell function [141,142]. Another enzyme involved in the metabolism of L-arg is iNOS, which converts L-arg into nitric oxide (NO) and citrulline. NO can mediate direct effects, but it can also be converted into peroxynitrite when ROS is available [143,144]. Peroxynitrites, which are secreted by MDSCs, can alter the TCR complex through inducing posttranslational modifications, dissociation of the TCR or selective degradation of the ζ-chain and, thereby, inhibit downstream signaling and T cell effector function [70,145]. This effect requires antigen presentation by MDSC, as in double transgenic mice, in which one T cell can recognize two antigens, it has been shown that the reactivity of T cells was only lost to the epitopes presented by the MDSC and not against the other antigen [145]. Notably, this shows that the MDSC can also suppress in an antigen-specific manner. ROS can also desensitize the TCR by inducing loss of TCR ζ-chain expression [146]. IDO exerts its suppressive effect by depletion of local L-tryptophan, thereby inhibiting T cell activation and proliferation, as well as inducing Tregs [147]. Arg1, iNOS and IDO expression can be detected by flow cytometry [148,149,150,151,152], Western blot [153], immunohistochemistry (IHC) [153] or quantitative PCR (qPCR) [154,155,156]. Arginase activity can be measured in cell lysates through measuring urea production [157,158]. ROS production can be measured by flow cytometry using an oxidation-sensitive dye named dichlorodihydrofluorescein diacetate (DCFDA) [148,155]. TGF-β and IL-10 are mostly detected by qPCR [156,159,160].
In the majority of laboratories, flow cytometry is generally limited to a maximum of 18 parameters (setting aside some experimental machines), and therefore, incorporation of these functional markers into existing flow panels is often not feasible. Currently, the novel multi-parameter single cell analysis technology named mass cytometry, or cytometry time-of-flight (CyTOF), would be an attractive alternative platform for incorporation of functional markers. CyTOF technology combines mass spectrometry with flow cytometry methodology. It utilizes target antibodies labeled to rare heavy metals for the simultaneous detection of up to 40 parameters [161,162,163] and, thus, permits a more in-depth single cell analysis. CyTOF’s ability to allow for in-depth profiling makes it a very attractive approach when complex and extensive immunophenotyping is desired.
A major drawback of using flow cytometry or CyTOF analysis is that it is a single cell analysis and, thus, does not provide any information about their location and functional orientation within the TME. A technique that holds great promise for the analysis of TME is multiplex IHC. Multiplex IHC permits the identification of up to 30 antigens on a single tissue slide or a group of different tissue samples (reviewed in [164]). Examples of such techniques include the multiplexed IHC combined with multispectral imaging system (Vectra platform [165]), sequential multiplex staining making use of repetitive staining and erase techniques [166,167] or image mass cytometry using a combination of mass cytometry, IHC and immunocytochemistry techniques [168]. These approaches are especially attractive because they allow the analysis of the TME on archival, paraffin-embedded tissue samples, and they retain the complete tumor (immune) contexture, i.e., this permits analysis of the location, density and functional orientation of the immune cell populations within the tumor and its surrounding stroma.
Taken together, the current and emerging monitoring strategies are well suited to study lymphoid and myeloid suppressor cells, especially at the level of quantification and immunophenotyping. Yet, one has to take into account that most of the evidence on the clinical relevance of these cells in suppressing anti-tumor immunity in humans was based on association and correlation and not based on formal proof, as can be obtained in murine studies. Therefore, we feel that it is of utmost importance that the functional capability of these suppressor cells be also analyzed.

4. Monitoring Functional Capacity of Lymphoid and Myeloid Suppressor Cells

4.1. Monitoring Suppressive Capacity of Tregs

Given their potential suppressive role in anti-tumor immunity, Tregs are often monitored in clinical (immunotherapy) trials. In the majority of studies, Treg assessment is restricted to phenotypic characterization and quantification. Yet, the absence of exclusive and highly specific Treg markers in humans and the presence of a high number of distinct subpopulations [120] complicates the analysis, leads to a multitude of reported phenotypic Treg definitions, as well as to a blurred picture regarding the association between Tregs and clinical outcome (reviewed by Whiteside et al. [117,119]). Especially since information on the suppressive capacity of the identified Treg population often is lacking in these studies, assessment of the suppressive capacity of each of the identified Treg populations, therefore, seems essential in the context of immunomonitoring.
The study of antigen-independent in vitro suppression of T cell reactivity was one of the first assays to functionally study the immune suppressive function of Tregs. It measures the inhibition of responder T cell proliferation, either through analyzing the dilution of cell tracking dyes (i.e., carboxyfluorescein succinimidyl ester (CSFE) and Cell Trace proliferation reagent) or by 3H-thymidine uptake by proliferating cells. In general, responder T cells are polyclonally stimulated (i.e., via soluble, bead- or plate-bound anti-CD3 ± anti-CD28 antibodies) and co-cultured with Tregs at different ratios [169,170,171,172]. Unfortunately, this approach requires large numbers of isolated Tregs, which are often not available in the context of clinical trials. A major advantage of measuring 3H-thymidine uptake over the dilution of tracking dyes is that it displays good sensitivity to detect suppression with low numbers of Treg cells [173]. In fact, a recent protocol by Tree and co-workers [172] permits Treg suppressive analysis with just 10,000 isolated Treg, illustrating that this assay could be used for immunomonitoring studies. Interestingly, this assay could be done in an APC-independent (with anti-CD3/anti-CD28-beads) and APC-dependent (autologous B cells + PHA) fashion. Studies in our lab confirmed the applicability of this assay for monitoring Treg suppressive function in cancer patients. Moreover, we were able to show that unlike the CFSE-based flow cytometric proliferation assay, this 3H-thymidine based assay is capable of detecting the suppressive function of healthy donor-derived Tregs, confirming that indeed the 3H-thymidine-based approach is much more sensitive.
Later, also antigen-dependent Treg suppression assays were developed as in the above-mentioned in vitro suppression assays, as quite a robust activation of responder T cells was provided by agonistic antibodies to CD3 and CD28, while this does not really reflect the cognate interactions between APC and T cells. Therefore, in an alternative setting, the suppressive effect of Tregs can be analyzed through evaluating their impact on tumor-associated antigen (TAA)-specific T cell reactivity in vitro. Using this setup, Tregs were found to suppress multiple tumor-specific responses in assays where Tregs were depleted prior to the assessment of tumor antigen-specific T cell reactivity in patients with urothelial cancer or colorectal cancer [174,175,176]. Similarly, the addition of Tregs to T cell cultures in which the effector cells specifically were stimulated with cognate antigen was also found to block antigen-specific effector function [177].
When it was realized that tumor-infiltrating Tregs are able to recognize tumor-derived antigens and can be expanded by cancer vaccines [178,179,180], assays that allowed the assessment of the suppressive capacity of these antigen-specific Tregs were developed. To test the suppressive effect of antigen-specific Treg cells, antigen-activated regulatory T cells can be co-cultured with either polyclonally-activated or antigen-specifically-activated responder cells, after which the inhibition of T cell proliferation, activation and/or cytokine production can be analyzed through different flow cytometric approaches, including activation marker expression, CFSE dilution for proliferation and cytokine production (IFN-γ, IL-2). Indeed, inhibition of responder cell proliferation could be shown upon stimulation with the cognate antigen employing this methodology [174,177,181].
Current approaches focus on direct suppression of effector T cells by Tregs as if it were one type of functional cell. Recent findings indicate that there is a high phenotypic and functional diversity in human Treg cells [120]. Moreover, different Treg subsets can be in different states of maturation, differentiation and activation (reviewed in [182,183,184]) and have distinct homing capabilities [185], suppressive mechanisms [186] and targets for suppression [187,188,189]. In addition, it has been shown that distinct subsets of Tregs can effectively regulate different types of Th cell responses in vivo [190]. This illustrates that it is very important to gain a full understanding of what Treg subset (phenotype) targets what type of immune responses, in which disease and by what mechanism, if one wants to perform the assay that is suitable for that situation. This requires the development of new and alternative methodologies. In the last couple of years, potential new ways to determine antigen specificity and activation status of Tregs have been developed. A candidate marker for the identification of such activated/antigen-specific Tregs is CD137 [191,192]. As described, combined CD137 and CD154 analysis following short-term (6 h) stimulation allows for the discrimination between CD137+CD154− Tregs (with stable Foxp3 expression) and CD137+CD154+ activated T cells (with instable Foxp3 expression) [191]. Alternatively, the transmembrane protein glycoprotein A repetitions predominant (GARP) has also been described to selectively identify activated human Foxp3+ Tregs following T cell receptor (TCR) stimulation [193,194]. Whether these markers will also be useful to identify antigen-specific Tregs ex vivo in human blood or tissue samples still needs to be elucidated.

4.2. Functional Analysis of Myeloid Cell Suppression

Myeloid cells with suppressive capacity are a heterogeneous population of immune regulatory cells originating from the common progenitor myeloid lineage that is arrested in differentiation to DCs, macrophages or granulocytes [195,196,197,198]. Their normal function is to control immunopathology, as they expand in the bone marrow and/or spleen upon infections, stress, trauma and other pathological conditions, such as cancer [199,200,201]. Analysis of the suppressive function of myeloid cells is still in its starting phase, since it is not known exactly what, when and how the suppression by myeloid on effector cells should be measured due to the heterogeneity of this cell population. It is not yet clear whether their suppressive capacity in vivo depends on cell-cell interaction or acts via the production of soluble factors that interfere with the function of anti-tumor effector cells (i.e., T cells, NK cells and B cells [106]).
Suppression factors produced by MDSC that have been identified are the aforementioned Arg1, iNOS, ROS and IDO, but also IL-10 and TGF-β [141,202]. Already almost 20 years ago, HLA-DRlow/-CD16+CD54+CD14+ IL-10-producing monocytes that were able to suppress autologous T cells were identified in ascites of ovarian cancer patients [203]. Moreover, TGF-β, and not Arg1 or iNOS, was shown to be involved in the suppressive capacity of CD14+HLA-DRlow/− cells obtained from melanoma patients [204].
Because of the lack of specific functional markers on myeloid-suppressive cells, it is more accepted to functionally access the capacity of these cells isolated from blood, bone marrow or TME in an in vitro cell-based assay. Whether close proximity or cell-to-cell contact is important for the suppressive efficacy of the myeloid cells is difficult to identify in in vitro assays, except for those earlier mentioned soluble mediators for which blocking antibodies exist. In a classic functional assay, isolated suppressive myeloid cells are co-cultured with activated responder T cells, and inhibition of the proliferation (often with 3H-thymidine incorporation or the flow-based technique) and cytokine production by these responder T cells is measured [143,205,206]. Using this assay, the responder T cells are often activated by anti-CD3/anti-CD28 beads or antibodies, which is not always mimicking the physiological situation in the tumor [207,208]. In melanoma patients, it was demonstrated that the proliferation and cytokine production (IFNγ) of autologous non-specific stimulated T cells were suppressed by 40% at the physiological ratios of CD14+HLA-DR-/low MDSC to T cells (1:4) [209]. Further analysis revealed that the suppressive molecules Arg1 and ROS were partly responsible for the suppressive effect. More importantly, cell-to-cell contact or close cell proximity was a prerequisite for this effect, as the suppressive capacity of the MDSCs was prevented in trans-well experiments [157,209].
l-arg deprivation and peroxynitrites can lead to loss or downregulation of CD3/ζ-chain (C247), which is part of the TCR complex in T cells and of the NKp46, NKp30 and CD16 killing receptor complexes on NK cells and, therefore, to the non-responsiveness of these two types of effector cells [210,211]. The expression of CD247 can be measured by quantitative PCR or flow cytometry. In advanced stage NSCLC patients, L-arginase- and iNOS-producing CD11b+CD14−CD15+CD33+ MDSCs were shown to downregulate CD247 on CD8+ T cells, thereby suppressing their function [212]. Downregulation of CD247 and high serum levels of nitric oxide (NO) and ROS are associated with high frequencies of circulating MDSCs and worse prognosis for cancer patients [106].
We have demonstrated that the depletion of CD14+ myeloid cells from the PBMC of patients with advanced cervical cancer or lung cancer resulted in the increase of T cell reactivity against recall antigens, but also unmasked tumor-specific T cell reactivity [82,213], albeit that the mechanism of action was not studied.
Most of the functional assays conducted with myeloid-suppressive cells focus on the suppression of effector T cells; however, in vivo, particular subsets of myeloid cells might (preferentially) suppress other immune cells, like DCs [214], macrophages [215,216], NK cells [217], Tregs [218] and B cells [106]. Moreover, often, circulating and bone-marrow-derived myeloid suppressor cells are used for studying their functional capabilities [128], whilst the tumor infiltrated or resident myeloid cells may have different suppressive potential. Indeed, in melanoma patients, CD11b+lineage− MDSCs isolated from the blood were able to suppress T cell proliferation, while the tumor infiltrating myeloid cells were not [219]. In contrast, CD11b+CD14− granulocytic MDSCs obtained from head and neck cancer (HNC) in patients were able to suppress non-specific stimulated T cells, while their blood-derived counterparts were not [158]. In addition, Corzo and co-workers also showed that MDSCs from peripheral blood (HNC patients) or lymph nodes (tumor-bearing mice) only suppressed tumor-specific T cell responses, but not the responsiveness of T cells to non-specific stimulation, while MDSCs isolated from tumors suppressed both types of T cell responses [158]. Altogether, this shows that the MDSC can suppress in an antigen-specific manner and that the type of suppression may depend on the compartment from where the MDSCs are obtained (i.e., periphery, lymph node, TME). Whereas harmonization of phenotypical analysis of MDSCs is going on [122], even more effort should be undertaken to also obtain consensus about the functional assays.

5. Conclusions

The measurement of immune dysfunction is important to improve current immunotherapeutic approaches and for the design of new strategies to alleviate immune suppression. This still requires a huge effort with respect to setting-up reliable phenotypic and functional monitoring tools of the suppressive cells, which are correlated with the clinical outcome of the cancer patients. When successful, this might lead to a higher percentage of patients who benefit from the treatment, as well as increase the duration of the clinical response.

Acknowledgments

This work was supported by a grant from the Dutch Cancer Society (KWF; Grant Number 2009-4400).

Author Contributions

All three authors contributed to writing and revising the review.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ACTadoptive cell transfer
APCantigen-presenting cell
Arg1arginase 1
CARchimeric antigen receptor
CFSEcarboxyfluorescein succinimidyl ester
CMVcytomegalovirus
CRCcolorectal cancer
CTCcirculating tumor cell
CTLA-4cytotoxic T lymphocyte antigen 4
CyTOFcytometry time-of-flight
DCdendritic cell
EBVEpstein-Barr virus
GM-CSFgranulocyte-macrophage colony stimulating factor
HPVhuman papillomavirus
ICOSinducible T cell co-stimulator
IDOindoleamine-2,3-deoxigenase
IHCimmunohistochemistry
ILinterleukin
iNOSinducible nitric oxide synthase
l-argl-arginine
M-CSFmacrophage colony stimulating factor
MDSCmyeloid-derived suppressor cells
MHCmajor histocompatibility complex
MICAMHC class I polypeptide-related sequence A
MIFmacrophage inhibitory factor
MLRmixed lymphocyte reaction
MMPmatrix metalloproteins
NKnatural killer
NOnitric oxide
OX40tumor necrosis factor receptor superfamily member 4
qPCRquantitative polymerase chain reaction
PBMCperipheral blood mononuclear cells
PD-1programmed cell death protein 1
PDL-1PD-1 ligand
PGE2prostaglandin E2
PHAphytohemagglutinin
ROSreactive oxygen species
SEBstaphylococcal enterotoxin B
SLPsynthetic long peptide
TGF-βtransforming growth factor receptor-β
Tregregulatory T cells
TAMtumor-associated macrophages
TANtumor-associated neutrophils
TCRT cell receptor
TIM3T cell immunoglobulin and mucin protein 3
TMEtumor micro-environment
T-VECTalimogene Laherparepvec
VEGFvascular endothelial growth factor
VISTAV-domain immunoglobulin suppressor of T cell activation

References

  1. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef] [PubMed]
  2. Robert, C.; Thomas, L.; Bondarenko, I.; O’Day, S.; Weber, J.; Garbe, C.; Lebbe, C.; Baurain, J.F.; Testori, A.; Grob, J.J.; et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 2011, 364, 2517–2526. [Google Scholar] [CrossRef] [PubMed]
  3. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-pd-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [PubMed]
  4. Wolchok, J.D.; Kluger, H.; Callahan, M.K.; Postow, M.A.; Rizvi, N.A.; Lesokhin, A.M.; Segal, N.H.; Ariyan, C.E.; Gordon, R.A.; Reed, K.; et al. Nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med. 2013, 369, 122–133. [Google Scholar] [CrossRef] [PubMed]
  5. Garon, E.B.; Rizvi, N.A.; Hui, R.; Leighl, N.; Balmanoukian, A.S.; Eder, J.P.; Patnaik, A.; Aggarwal, C.; Gubens, M.; Horn, L.; et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 2015, 372, 2018–2028. [Google Scholar] [CrossRef] [PubMed]
  6. Brahmer, J.; Reckamp, K.L.; Baas, P.; Crino, L.; Eberhardt, W.E.; Poddubskaya, E.; Antonia, S.; Pluzanski, A.; Vokes, E.E.; Holgado, E.; et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N. Engl. J. Med. 2015, 373, 123–135. [Google Scholar] [CrossRef] [PubMed]
  7. Khoja, L.; Butler, M.O.; Kang, S.P.; Ebbinghaus, S.; Joshua, A.M. Pembrolizumab. J. Immunother. Cancer 2015. [Google Scholar] [CrossRef] [PubMed]
  8. Rosenberg, S.A.; Yang, J.C.; Sherry, R.M.; Kammula, U.S.; Hughes, M.S.; Phan, G.Q.; Citrin, D.E.; Restifo, N.P.; Robbins, P.F.; Wunderlich, J.R.; et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using t-cell transfer immunotherapy. Clin. Cancer Res. 2011, 17, 4550–4557. [Google Scholar] [CrossRef] [PubMed]
  9. Dudley, M.E.; Yang, J.C.; Sherry, R.; Hughes, M.S.; Royal, R.; Kammula, U.; Robbins, P.F.; Huang, J.; Citrin, D.E.; Leitman, S.F.; et al. Adoptive cell therapy for patients with metastatic melanoma: Evaluation of intensive myeloablative chemoradiation preparative regimens. J. Clin. Oncol. 2008, 26, 5233–5239. [Google Scholar] [CrossRef] [PubMed]
  10. Verdegaal, E.M.; Visser, M.; Ramwadhdoebe, T.H.; van der Minne, C.E.; van Steijn, J.A.; Kapiteijn, E.; Haanen, J.B.; van der Burg, S.H.; Nortier, J.W.; Osanto, S. Successful treatment of metastatic melanoma by adoptive transfer of blood-derived polyclonal tumor-specific CD4+ and CD8+ t cells in combination with low-dose interferon-alpha. Cancer Immunol. Immunother. 2011, 60, 953–963. [Google Scholar] [CrossRef] [PubMed]
  11. Vignard, V.; Lemercier, B.; Lim, A.; Pandolfino, M.C.; Guilloux, Y.; Khammari, A.; Rabu, C.; Echasserieau, K.; Lang, F.; Gougeon, M.L.; et al. Adoptive transfer of tumor-reactive melan-a-specific ctl clones in melanoma patients is followed by increased frequencies of additional melan-a-specific t cells. J. Immunol. 2005, 175, 4797–4805. [Google Scholar] [CrossRef] [PubMed]
  12. Maus, M.V.; June, C.H. Making better chimeric antigen receptors for adoptive T-cell therapy. Clin. Cancer Res. 2016, 22, 1875–1884. [Google Scholar] [CrossRef] [PubMed]
  13. Turtle, C.J.; Maloney, D.G. Clinical trials of cd19-targeted car-modified t cell therapy; a complex and varied landscape. Expert Rev. Hematol. 2016, 9, 719–721. [Google Scholar] [CrossRef] [PubMed]
  14. Davila, M.L.; Sadelain, M. Biology and clinical application of car T cells for B cell malignancies. Int. J. Hematol. 2016, 104, 6–17. [Google Scholar] [CrossRef] [PubMed]
  15. Rubnitz, J.E.; Inaba, H.; Ribeiro, R.C.; Pounds, S.; Rooney, B.; Bell, T.; Pui, C.H.; Leung, W. Nkaml: A pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J. Clin. Oncol. 2010, 28, 955–959. [Google Scholar] [CrossRef] [PubMed]
  16. Curti, A.; Ruggeri, L.; D’Addio, A.; Bontadini, A.; Dan, E.; Motta, M.R.; Trabanelli, S.; Giudice, V.; Urbani, E.; Martinelli, G.; et al. Successful transfer of alloreactive haploidentical kir ligand-mismatched natural killer cells after infusion in elderly high risk acute myeloid leukemia patients. Blood 2011, 118, 3273–3279. [Google Scholar] [CrossRef] [PubMed]
  17. Yonezawa, A.; Chester, C.; Rajasekaran, N.; Kohrt, H.E. Harnessing the innate immune system to treat cancer: Enhancement of antibody-dependent cellular cytotoxicity with anti-CD137 ab. Chin. Clin. Oncol. 2016. [Google Scholar] [CrossRef]
  18. Young, K.H.; Baird, J.R.; Savage, T.; Cottam, B.; Friedman, D.; Bambina, S.; Messenheimer, D.J.; Fox, B.; Newell, P.; Bahjat, K.S.; et al. Optimizing timing of immunotherapy improves control of tumors by hypofractionated radiation therapy. PLoS ONE 2016, 11, e0157164. [Google Scholar] [CrossRef] [PubMed]
  19. Sanmamed, M.F.; Pastor, F.; Rodriguez, A.; Perez-Gracia, J.L.; Rodriguez-Ruiz, M.E.; Jure-Kunkel, M.; Melero, I. Agonists of co-stimulation in cancer immunotherapy directed against CD137, OX40, GITR, CD27, CD28, and ICOS. Semin. Oncol. 2015, 42, 640–655. [Google Scholar] [CrossRef] [PubMed]
  20. Aspeslagh, S.; Postel-Vinay, S.; Rusakiewicz, S.; Soria, J.C.; Zitvogel, L.; Marabelle, A. Rationale for anti-OX40 cancer immunotherapy. Eur. J. Cancer 2016, 52, 50–66. [Google Scholar] [CrossRef] [PubMed]
  21. Ahrends, T.; Babala, N.; Xiao, Y.; Yagita, H.; van Eenennaam, H.; Borst, J. Cd27 agonism plus pd-1 blockade recapitulates CD4+ t-cell help in therapeutic anticancer vaccination. Cancer Res. 2016, 76, 2921–2931. [Google Scholar] [CrossRef] [PubMed]
  22. Melief, C.J.; van Hall, T.; Arens, R.; Ossendorp, F.; van der Burg, S.H. Therapeutic cancer vaccines. J. Clin. Investig. 2015, 125, 3401–3412. [Google Scholar] [CrossRef] [PubMed]
  23. Romero, P.; Banchereau, J.; Bhardwaj, N.; Cockett, M.; Disis, M.L.; Dranoff, G.; Gilboa, E.; Hammond, S.A.; Hershberg, R.; Korman, A.J.; et al. The human vaccines project: A roadmap for cancer vaccine development. Sci. Transl. Med. 2016. [Google Scholar] [CrossRef] [PubMed]
  24. Van der Burg, S.H.; Arens, R.; Ossendorp, F.; van Hall, T.; Melief, C.J.M. Vaccines for established cancer: Overcoming the challenges posed by immune evasion. Nat. Rev. Cancer 2016, 16, 219–233. [Google Scholar] [CrossRef] [PubMed]
  25. Kenter, G.G.; Welters, M.J.; Valentijn, A.R.; Lowik, M.J.; Berends-van der Meer, D.M.; Vloon, A.P.; Essahsah, F.; Fathers, L.M.; Offringa, R.; Drijfhout, J.W.; et al. Vaccination against hpv-16 oncoproteins for vulvar intraepithelial neoplasia. N. Engl. J. Med. 2009, 361, 1838–1847. [Google Scholar] [CrossRef] [PubMed]
  26. Van Poelgeest, M.I.; Welters, M.J.; Vermeij, R.; Stynenbosch, L.F.; Loof, N.M.; Berends-van der Meer, D.M.; Lowik, M.J.; Hamming, I.L.; van Esch, E.M.; Hellebrekers, B.W.; et al. Vaccination against oncoproteins of hpv16 for noninvasive vulvar/vaginal lesions: Lesion clearance is related to the strength of the t-cell response. Clin. Cancer Res. 2016, 22, 2342–2350. [Google Scholar] [CrossRef] [PubMed]
  27. Daayana, S.; Elkord, E.; Winters, U.; Pawlita, M.; Roden, R.; Stern, P.L.; Kitchener, H.C. Phase ii trial of imiquimod and hpv therapeutic vaccination in patients with vulval intraepithelial neoplasia. Br. J. Cancer 2010, 102, 1129–1136. [Google Scholar] [CrossRef] [PubMed]
  28. Fiander, A.N.; Tristram, A.J.; Davidson, E.J.; Tomlinson, A.E.; Man, S.; Baldwin, P.J.; Sterling, J.C.; Kitchener, H.C. Prime-boost vaccination strategy in women with high-grade, noncervical anogenital intraepithelial neoplasia: Clinical results from a multicenter phase ii trial. Int. J. Gynecol. Cancer 2006, 16, 1075–1081. [Google Scholar] [CrossRef] [PubMed]
  29. Trimble, C.L.; Morrow, M.P.; Kraynyak, K.A.; Shen, X.; Dallas, M.; Yan, J.; Edwards, L.; Parker, R.L.; Denny, L.; Giffear, M.; et al. Safety, efficacy, and immunogenicity of vgx-3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: A randomised, double-blind, placebo-controlled phase 2b trial. Lancet 2015, 386, 2078–2088. [Google Scholar] [CrossRef]
  30. Kantoff, P.W.; Higano, C.S.; Shore, N.D.; Berger, E.R.; Small, E.J.; Penson, D.F.; Redfern, C.H.; Ferrari, A.C.; Dreicer, R.; Sims, R.B.; et al. Sipuleucel-t immunotherapy for castration-resistant prostate cancer. N. Engl. J. Med. 2010, 363, 411–422. [Google Scholar] [CrossRef] [PubMed]
  31. Gulley, J.L.; Mulders, P.; Albers, P.; Banchereau, J.; Bolla, M.; Pantel, K.; Powles, T. Perspectives on sipuleucel-T: Its role in the prostate cancer treatment paradigm. Oncoimmunology 2016, 5, e1107698. [Google Scholar] [CrossRef] [PubMed]
  32. Andtbacka, R.H.; Kaufman, H.L.; Collichio, F.; Amatruda, T.; Senzer, N.; Chesney, J.; Delman, K.A.; Spitler, L.E.; Puzanov, I.; Agarwala, S.S.; et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J. Clin. Oncol. 2015, 33, 2780–2788. [Google Scholar] [CrossRef] [PubMed]
  33. Puzanov, I.; Milhem, M.M.; Minor, D.; Hamid, O.; Li, A.; Chen, L.; Chastain, M.; Gorski, K.S.; Anderson, A.; Chou, J.; et al. Talimogene laherparepvec in combination with ipilimumab in previously untreated, unresectable stage iiib-iv melanoma. J. Clin. Oncol. 2016. [Google Scholar] [CrossRef] [PubMed]
  34. Le, D.T.; Brockstedt, D.G.; Nir-Paz, R.; Hampl, J.; Mathur, S.; Nemunaitis, J.; Sterman, D.H.; Hassan, R.; Lutz, E.; Moyer, B.; et al. A live-attenuated listeria vaccine (anz-100) and a live-attenuated listeria vaccine expressing mesothelin (crs-207) for advanced cancers: Phase i studies of safety and immune induction. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2012, 18, 858–868. [Google Scholar] [CrossRef] [PubMed]
  35. Le, D.T.; Wang-Gillam, A.; Picozzi, V.; Greten, T.F.; Crocenzi, T.; Springett, G.; Morse, M.; Zeh, H.; Cohen, D.; Fine, R.L.; et al. Safety and survival with GVAX pancreas prime and listeria monocytogenes-expressing mesothelin (CRS-207) boost vaccines for metastatic pancreatic cancer. J. Clin. Oncol. 2015, 33, 1325–1333. [Google Scholar] [CrossRef] [PubMed]
  36. Wilgenhof, S.; Corthals, J.; Van Nuffel, A.M.; Benteyn, D.; Heirman, C.; Bonehill, A.; Thielemans, K.; Neyns, B. Long-term clinical outcome of melanoma patients treated with messenger rna-electroporated dendritic cell therapy following complete resection of metastases. Cancer Immunol. Immunother. 2015, 64, 381–388. [Google Scholar] [CrossRef] [PubMed]
  37. Lee, J.H.; Lee, Y.; Lee, M.; Heo, M.K.; Song, J.S.; Kim, K.H.; Lee, H.; Yi, N.J.; Lee, K.W.; Suh, K.S.; et al. A phase i/iia study of adjuvant immunotherapy with tumour antigen-pulsed dendritic cells in patients with hepatocellular carcinoma. Br. J. Cancer 2015, 113, 1666–1676. [Google Scholar] [CrossRef] [PubMed]
  38. Anguille, S.; Smits, E.L.; Lion, E.; van Tendeloo, V.F.; Berneman, Z.N. Clinical use of dendritic cells for cancer therapy. Lancet Oncol. 2014, 15, e257–e267. [Google Scholar] [CrossRef]
  39. Garrido, F.; Aptsiauri, N.; Doorduijn, E.M.; Garcia Lora, A.M.; van Hall, T. The urgent need to recover mhc class i in cancers for effective immunotherapy. Curr. Opin. Immunol. 2016, 39, 44–51. [Google Scholar] [CrossRef] [PubMed]
  40. Verdegaal, E.M.E.; de Miranda, N.F.C.C.; Visser, M.; Harryvan, T.; van Buuren, M.M.; Andersen, R.S.; Hadrup, S.R.; van der Minne, C.E.; Schotte, R.; Spits, H.; et al. Neoantigen landscape dynamics during human melanoma-T cell interactions. Nature 2016, 536, 91–95. [Google Scholar] [CrossRef] [PubMed]
  41. Lippitz, B.E. Cytokine patterns in patients with cancer: A systematic review. Lancet Oncol. 2013, 14, e218–e228. [Google Scholar] [CrossRef]
  42. Pahl, J.; Cerwenka, A. Tricking the balance: Nk cells in anti-cancer immunity. Immunobiology 2015. [Google Scholar] [CrossRef] [PubMed]
  43. Wu, A.A.; Drake, V.; Huang, H.S.; Chiu, S.; Zheng, L. Reprogramming the tumor microenvironment: Tumor-induced immunosuppressive factors paralyze t cells. Oncoimmunology 2015, 4, e1016700. [Google Scholar] [CrossRef] [PubMed]
  44. Dijkgraaf, E.M.; Welters, M.J.; Nortier, J.W.; van der Burg, S.H.; Kroep, J.R. Interleukin-6/interleukin-6 receptor pathway as a new therapy target in epithelial ovarian cancer. Curr. Pharm. Des. 2012, 18, 3816–3827. [Google Scholar] [CrossRef] [PubMed]
  45. Biswas, S.K.; Mantovani, A. Macrophage plasticity and interaction with lymphocyte subsets: Cancer as a paradigm. Nat. Immunol. 2010, 11, 889–896. [Google Scholar] [CrossRef] [PubMed]
  46. Heusinkveld, M.; de Vos van Steenwijk, P.J.; Goedemans, R.; Ramwadhdoebe, T.H.; Gorter, A.; Welters, M.J.; van Hall, T.; van der Burg, S.H. M2 macrophages induced by prostaglandin e2 and il-6 from cervical carcinoma are switched to activated m1 macrophages by CD4+ Th1 cells. J. Immunol. 2011, 187, 1157–1165. [Google Scholar] [CrossRef] [PubMed]
  47. O’Reilly, C.; Doroudian, M.; Mawhinney, L.; Donnelly, S.C. Targeting MIF in cancer: Therapeutic strategies, current developments, and future opportunities. Med. Res. Rev. 2016, 36, 440–460. [Google Scholar] [CrossRef] [PubMed]
  48. Schlecker, E.; Fiegler, N.; Arnold, A.; Altevogt, P.; Rose-John, S.; Moldenhauer, G.; Sucker, A.; Paschen, A.; von Strandmann, E.P.; Textor, S.; et al. Metalloprotease-mediated tumor cell shedding of B7-H6, the ligand of the natural killer cell-activating receptor NKp30. Cancer Res. 2014, 74, 3429–3440. [Google Scholar] [CrossRef] [PubMed]
  49. Motz, G.T.; Coukos, G. Deciphering and reversing tumor immune suppression. Immunity 2013, 39, 61–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Brown, G.T.; Murray, G.I. Current mechanistic insights into the roles of matrix metalloproteinases in tumour invasion and metastasis. J. Pathol. 2015, 237, 273–281. [Google Scholar] [CrossRef] [PubMed]
  51. Krengel, U.; Bousquet, P.A. Molecular recognition of gangliosides and their potential for cancer immunotherapies. Front. Immunol. 2014. [Google Scholar] [CrossRef] [PubMed]
  52. Yang, C.; Robbins, P.D. The roles of tumor-derived exosomes in cancer pathogenesis. Clin. Dev. Immunol. 2011. [Google Scholar] [CrossRef] [PubMed]
  53. Romero-Garcia, S.; Moreno-Altamirano, M.M.; Prado-Garcia, H.; Sanchez-Garcia, F.J. Lactate contribution to the tumor microenvironment: Mechanisms, effects on immune cells and therapeutic relevance. Front. Immunol. 2016. [Google Scholar] [CrossRef] [PubMed]
  54. Kaskas, N.M.; Moore-Medlin, T.; McClure, G.B.; Ekshyyan, O.; Vanchiere, J.A.; Nathan, C.A. Serum biomarkers in head and neck squamous cell cancer. JAMA Otolaryngol. Head Neck Surg. 2014, 140, 5–11. [Google Scholar] [CrossRef] [PubMed]
  55. Carvalho, S.; Troost, E.G.; Bons, J.; Menheere, P.; Lambin, P.; Oberije, C. Prognostic value of blood-biomarkers related to hypoxia, inflammation, immune response and tumour load in non-small cell lung cancer—A survival model with external validation. Radiother. Oncol. 2016, 119, 487–494. [Google Scholar] [CrossRef] [PubMed]
  56. Chang, W.J.; Du, Y.; Zhao, X.; Ma, L.Y.; Cao, G.W. Inflammation-related factors predicting prognosis of gastric cancer. World J. Gastroenterol. 2014, 20, 4586–4596. [Google Scholar] [CrossRef] [PubMed]
  57. Tran, H.T.; Liu, Y.; Zurita, A.J.; Lin, Y.; Baker-Neblett, K.L.; Martin, A.M.; Figlin, R.A.; Hutson, T.E.; Sternberg, C.N.; Amado, R.G.; et al. Prognostic or predictive plasma cytokines and angiogenic factors for patients treated with pazopanib for metastatic renal-cell cancer: A retrospective analysis of phase 2 and phase 3 trials. Lancet Oncol. 2012, 13, 827–837. [Google Scholar] [CrossRef]
  58. Ghanipour, L.; Darmanis, S.; Landegren, U.; Glimelius, B.; Pahlman, L.; Birgisson, H. Detection of biomarkers with solid-phase proximity ligation assay in patients with colorectal cancer. Transl. Oncol. 2016, 9, 251–255. [Google Scholar] [CrossRef] [PubMed]
  59. Lin, D.; Alborn, W.E.; Slebos, R.J.; Liebler, D.C. Comparison of protein immunoprecipitation-multiple reaction monitoring with elisa for assay of biomarker candidates in plasma. J. Proteome Res. 2013, 12, 5996–6003. [Google Scholar] [CrossRef] [PubMed]
  60. Garcia, A.D.; Chavez, J.L.; Mechref, Y. Rapid and sensitive lc-esi-ms of gangliosides. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2014, 947–948, 1–7. [Google Scholar] [CrossRef] [PubMed]
  61. Szajnik, M.; Czystowska-Kuzmicz, M.; Elishaev, E.; Whiteside, T.L. Biological markers of prognosis, response to therapy and outcome in ovarian carcinoma. Expert Rev. Mol. Diagn. 2016, 16, 811–826. [Google Scholar] [CrossRef] [PubMed]
  62. Wu, L.; Zhang, X.; Zhang, B.; Shi, H.; Yuan, X.; Sun, Y.; Pan, Z.; Qian, H.; Xu, W. Exosomes derived from gastric cancer cells activate nf-kappab pathway in macrophages to promote cancer progression. Tumour Biol. 2016. [Google Scholar] [CrossRef]
  63. He, M.; Zeng, Y. Microfluidic exosome analysis toward liquid biopsy for cancer. J. Lab. Autom. 2016. [Google Scholar] [CrossRef] [PubMed]
  64. Takeuchi, Y.; Nishikawa, H. Roles of regulatory t cells in cancer immunity. Int. Immunol. 2016, 25, 401–409. [Google Scholar] [CrossRef] [PubMed]
  65. Tesone, A.J.; Rutkowski, M.R.; Brencicova, E.; Svoronos, N.; Perales-Puchalt, A.; Stephen, T.L.; Allegrezza, M.J.; Payne, K.K.; Nguyen, J.M.; Wickramasinghe, J.; et al. Satb1 overexpression drives tumor-promoting activities in cancer-associated dendritic cells. Cell Rep. 2016, 14, 1774–1786. [Google Scholar] [CrossRef] [PubMed]
  66. Demoulin, S.A.; Somja, J.; Duray, A.; Guenin, S.; Roncarati, P.; Delvenne, P.O.; Herfs, M.F.; Hubert, P.M. Cervical (pre)neoplastic microenvironment promotes the emergence of tolerogenic dendritic cells via rankl secretion. Oncoimmunology 2015, 4, e1008334. [Google Scholar] [CrossRef] [PubMed]
  67. Seliger, B.; Massa, C. The dark side of dendritic cells: Development and exploitation of tolerogenic activity that favor tumor outgrowth and immune escape. Front. Immunol. 2013. [Google Scholar] [CrossRef] [PubMed]
  68. Mantovani, A.; Sica, A. Macrophages, innate immunity and cancer: Balance, tolerance, and diversity. Curr. Opin. Immunol. 2010, 22, 231–237. [Google Scholar] [CrossRef] [PubMed]
  69. Arina, A.; Corrales, L.; Bronte, V. Enhancing t cell therapy by overcoming the immunosuppressive tumor microenvironment. Semin Immunol. 2016, 28, 54–63. [Google Scholar] [CrossRef] [PubMed]
  70. Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef] [PubMed]
  71. Powell, D.R.; Huttenlocher, A. Neutrophils in the tumor microenvironment. Trends Immunol. 2016, 37, 41–52. [Google Scholar] [CrossRef] [PubMed]
  72. Moses, K.; Brandau, S. Human neutrophils: Their role in cancer and relation to myeloid-derived suppressor cells. Semin Immunol. 2016, 28, 187–196. [Google Scholar] [CrossRef] [PubMed]
  73. Dobrzycka, B.; Mackowiak-Matejczyk, B.; Terlikowska, K.M.; Kulesza-Bronczyk, B.; Kinalski, M.; Terlikowski, S.J. Serum levels of IL-6, IL-8 and crp as prognostic factors in epithelial ovarian cancer. Eur. Cytokine Netw. 2013, 24, 106–113. [Google Scholar] [PubMed]
  74. Wang, Z.; Wu, P.; Wu, D.; Zhang, Z.; Hu, G.; Zhao, S.; Lai, Y.; Huang, J. Prognostic and clinicopathological significance of serum interleukin-6 expression in colorectal cancer: A systematic review and meta-analysis. OncoTargets Ther. 2015, 8, 3793–3801. [Google Scholar] [CrossRef] [PubMed]
  75. Tas, F.; Yasasever, C.T.; Karabulut, S.; Tastekin, D.; Duranyildiz, D. Serum transforming growth factor-beta1 levels may have predictive and prognostic roles in patients with gastric cancer. Tumour Biol. J. Int. Soc. Oncodevelop. Biol. Med. 2015, 36, 2097–2103. [Google Scholar] [CrossRef] [PubMed]
  76. Jakubowska, K.; Naumnik, W.; Niklinska, W.; Chyczewska, E. Clinical significance of hmgb-1 and tgf-beta level in serum and balf of advanced non-small cell lung cancer. Adv. Exp. Med. Biol. 2015, 852, 49–58. [Google Scholar] [PubMed]
  77. Zhao, S.; Wu, D.; Wu, P.; Wang, Z.; Huang, J. Serum il-10 predicts worse outcome in cancer patients: A meta-analysis. PLoS ONE 2015, 10, e0139598. [Google Scholar] [CrossRef] [PubMed]
  78. Moghadamyeghaneh, Z.; Hanna, M.H.; Carmichael, J.C.; Mills, S.D.; Pigazzi, A.; Stamos, M.J. Preoperative leukocytosis in colorectal cancer patients. J. Am. Coll. Surg. 2015, 221, 207–214. [Google Scholar] [CrossRef] [PubMed]
  79. Holgersson, G.; Sandelin, M.; Hoye, E.; Bergstrom, S.; Henriksson, R.; Ekman, S.; Nyman, J.; Helsing, M.; Friesland, S.; Holgersson, M.; et al. Swedish lung cancer radiation study group: The prognostic value of anaemia, thrombocytosis and leukocytosis at time of diagnosis in patients with non-small cell lung cancer. Med. Oncol. 2012, 29, 3176–3182. [Google Scholar] [CrossRef] [PubMed]
  80. Mabuchi, S.; Matsumoto, Y.; Isohashi, F.; Yoshioka, Y.; Ohashi, H.; Morii, E.; Hamasaki, T.; Aozasa, K.; Mutch, D.G.; Kimura, T. Pretreatment leukocytosis is an indicator of poor prognosis in patients with cervical cancer. Gynecol. Oncol. 2011, 122, 25–32. [Google Scholar] [CrossRef] [PubMed]
  81. So, K.A.; Hong, J.H.; Jin, H.M.; Kim, J.W.; Song, J.Y.; Lee, J.K.; Lee, N.W. The prognostic significance of preoperative leukocytosis in epithelial ovarian carcinoma: A retrospective cohort study. Gynecol. Oncol. 2014, 132, 551–555. [Google Scholar] [CrossRef] [PubMed]
  82. Welters, M.J.; van der Sluis, T.C.; van Meir, H.; Loof, N.M.; van Ham, V.J.; van Duikeren, S.; Santegoets, S.J.; Arens, R.; de Kam, M.L.; Cohen, A.F.; et al. Vaccination during myeloid cell depletion by cancer chemotherapy fosters robust T cell responses. Sci. Transl. Med. 2016. [Google Scholar] [CrossRef] [PubMed]
  83. Bishara, S.; Griffin, M.; Cargill, A.; Bali, A.; Gore, M.E.; Kaye, S.B.; Shepherd, J.H.; Van Trappen, P.O. Pre-treatment white blood cell subtypes as prognostic indicators in ovarian cancer. Eur. J. Obstet. Gynecol. Reprod. Biol. 2008, 138, 71–75. [Google Scholar] [CrossRef] [PubMed]
  84. Matsuo, K.; Hom, M.S.; Moeini, A.; Machida, H.; Takeshima, N.; Roman, L.D.; Sood, A.K. Significance of monocyte counts on tumor characteristics and survival outcome of women with endometrial cancer. Gynecol. Oncol. 2015, 138, 332–338. [Google Scholar] [CrossRef] [PubMed]
  85. Schmidt, H.; Bastholt, L.; Geertsen, P.; Christensen, I.J.; Larsen, S.; Gehl, J.; von der Maase, H. Elevated neutrophil and monocyte counts in peripheral blood are associated with poor survival in patients with metastatic melanoma: A prognostic model. Br. J. Cancer 2005, 93, 273–278. [Google Scholar] [CrossRef] [PubMed]
  86. Yang, Y.Q.; Liang, J.H.; Wu, J.Z.; Wang, L.; Qu, X.Y.; Cao, L.; Zhao, X.L.; Huang, D.P.; Fan, L.; Li, J.Y.; et al. Elevated absolute monocyte count predicts unfavorable outcomes in patients with angioimmunoblastic T-cell lymphoma. Leuk Res. 2016, 42, 88–92. [Google Scholar] [CrossRef] [PubMed]
  87. Kou, F.; Lu, Z.; Li, J.; Zhang, X.; Lu, M.; Zhou, J.; Wang, X.; Gong, J.; Gao, J.; Li, J.; et al. Pretreatment lymphopenia is an easily detectable predictive and prognostic marker in patients with metastatic esophagus squamous cell carcinoma receiving first-line chemotherapy. Cancer Med. 2016, 5, 778–786. [Google Scholar] [CrossRef] [PubMed]
  88. Hancock, B.W.; Dunsmore, I.R.; Swan, H.T. Lymphopenia: A bad prognostic factor in hodgkin’s disease. Scand. J. Haematol. 1982, 29, 193–199. [Google Scholar] [CrossRef] [PubMed]
  89. Huerta, S.; Patel, P. Leukocytosis in colorectal cancer. J. Am. Coll. Surg. 2015, 221, 996–997. [Google Scholar] [CrossRef] [PubMed]
  90. Martens, A.; Wistuba-Hamprecht, K.; Yuan, J.; Postow, M.A.; Wong, P.; Capone, M.; Madonna, G.; Khammari, A.; Schilling, B.; Sucker, A.; et al. Increases in absolute lymphocytes and circulating CD4+ and CD8+ t cells are associated with positive clinical outcome of melanoma patients treated with ipilimumab. Am. Assoc. Cancer Res. 2016. [Google Scholar] [CrossRef] [PubMed]
  91. Ferrucci, P.F.; Ascierto, P.A.; Pigozzo, J.; Del Vecchio, M.; Maio, M.; Antonini Cappellini, G.C.; Guidoboni, M.; Queirolo, P.; Savoia, P.; Mandala, M.; et al. Baseline neutrophils and derived neutrophil-to-lymphocyte ratio: Prognostic relevance in metastatic melanoma patients receiving ipilimumab. Ann. Oncol. 2016, 27, 732–738. [Google Scholar] [CrossRef] [PubMed]
  92. Guidos, C.; Sinha, A.A.; Lee, K.C. Functional differences and complementation between dendritic cells and macrophages in T-cell activation. Immunology 1987, 61, 269–276. [Google Scholar] [PubMed]
  93. Yanagimoto, H.; Takai, S.; Satoi, S.; Toyokawa, H.; Takahashi, K.; Terakawa, N.; Kwon, A.H.; Kamiyama, Y. Impaired function of circulating dendritic cells in patients with pancreatic cancer. Clin. Immunol. 2005, 114, 52–60. [Google Scholar] [CrossRef] [PubMed]
  94. Rickmann, M.; Macke, L.; Sundarasetty, B.S.; Stamer, K.; Figueiredo, C.; Blasczyk, R.; Heuser, M.; Krauter, J.; Ganser, A.; Stripecke, R. Monitoring dendritic cell and cytokine biomarkers during remission prior to relapse in patients with FLT3-ITD acute myeloid leukemia. Ann. Hematol. 2013, 92, 1079–1090. [Google Scholar] [CrossRef] [PubMed]
  95. Ni, X.; Duvic, M. Dendritic cells and cutaneous t-cell lymphomas. G Ital. Dermatol. Venereol. 2011, 146, 103–113. [Google Scholar] [PubMed]
  96. Gigante, M.; Blasi, A.; Loverre, A.; Mancini, V.; Battaglia, M.; Selvaggi, F.P.; Maiorano, E.; Napoli, A.; Castellano, G.; Storkus, W.J.; et al. Dysfunctional dc subsets in rcc patients: Ex vivo correction to yield an effective anti-cancer vaccine. Mol. Immunol. 2009, 46, 893–901. [Google Scholar] [CrossRef] [PubMed]
  97. Mirza, N.; Fishman, M.; Fricke, I.; Dunn, M.; Neuger, A.M.; Frost, T.J.; Lush, R.M.; Antonia, S.; Gabrilovich, D.I. All-trans-retinoic acid improves differentiation of myeloid cells and immune response in cancer patients. Cancer Res. 2006, 66, 9299–9307. [Google Scholar] [CrossRef] [PubMed]
  98. Clerici, M.; Merola, M.; Ferrario, E.; Trabattoni, D.; Villa, M.L.; Stefanon, B.; Venzon, D.J.; Shearer, G.M.; De Palo, G.; Clerici, E. Cytokine production patterns in cervical intraepithelial neoplasia: Association with human papillomavirus infection. J. Natl. Cancer Inst. 1997, 89, 245–250. [Google Scholar] [CrossRef] [PubMed]
  99. McCloskey, T.W.; Haridas, V.; Pontrelli, L.; Pahwa, S. Response to superantigen stimulation in peripheral blood mononuclear cells from children perinatally infected with human immunodeficiency virus and receiving highly active antiretroviral therapy. Clin. Diagn. Lab. Immunol. 2004, 11, 957–962. [Google Scholar] [CrossRef] [PubMed]
  100. Waeckerle-Men, Y.; Uetz-von Allmen, E.; Fopp, M.; von Moos, R.; Bohme, C.; Schmid, H.P.; Ackermann, D.; Cerny, T.; Ludewig, B.; Groettrup, M.; et al. Dendritic cell-based multi-epitope immunotherapy of hormone-refractory prostate carcinoma. Cancer Immunol. Immunother. 2006, 55, 1524–1533. [Google Scholar] [CrossRef] [PubMed]
  101. Grenga, I.; Donahue, R.N.; Lepone, L.M.; Richards, J.; Schlom, J. A fully human IgG1 anti-PD-L1 mab in an in vitro assay enhances antigen-specific T-cell responses. Clin. Transl. Immunol. 2016, 5, e83. [Google Scholar] [CrossRef] [PubMed]
  102. Dahlberg, C.I.; Sarhan, D.; Chrobok, M.; Duru, A.D.; Alici, E. Natural killer cell-based therapies targeting cancer: Possible strategies to gain and sustain anti-tumor activity. Front. Immunol. 2015, 6, 605. [Google Scholar] [CrossRef] [PubMed]
  103. Guillerey, C.; Huntington, N.D. Targeting natural killer cells in cancer immunotherapy. Nat. Immunol. 2016, 17, 1025–1036. [Google Scholar] [CrossRef] [PubMed]
  104. Alter, G.; Malenfant, J.M.; Altfeld, M. Cd107a as a functional marker for the identification of natural killer cell activity. J. Immunol. Methods 2004, 294, 15–22. [Google Scholar] [CrossRef] [PubMed]
  105. Galdiero, M.R.; Bonavita, E.; Barajon, I.; Garlanda, C.; Mantovani, A.; Jaillon, S. Tumor associated macrophages and neutrophils in cancer. Immunobiology 2013, 218, 1402–1410. [Google Scholar] [CrossRef] [PubMed]
  106. Baniyash, M. Myeloid-derived suppressor cells as intruders and targets: Clinical implications in cancer therapy. Cancer Immunol. Immunother. 2016, 65, 857–867. [Google Scholar] [CrossRef] [PubMed]
  107. Teng, M.W.; Ngiow, S.F.; von Scheidt, B.; McLaughlin, N.; Sparwasser, T.; Smyth, M.J. Conditional regulatory T-cell depletion releases adaptive immunity preventing carcinogenesis and suppressing established tumor growth. Cancer Res. 2010, 70, 7800–7809. [Google Scholar] [CrossRef] [PubMed]
  108. Klages, K.; Mayer, C.T.; Lahl, K.; Loddenkemper, C.; Teng, M.W.; Ngiow, S.F.; Smyth, M.J.; Hamann, A.; Huehn, J.; Sparwasser, T. Selective depletion of foxp3+ regulatory t cells improves effective therapeutic vaccination against established melanoma. Cancer Res. 2010, 70, 7788–7799. [Google Scholar] [CrossRef] [PubMed]
  109. Li, X.; Kostareli, E.; Suffner, J.; Garbi, N.; Hammerling, G.J. Efficient treg depletion induces T-cell infiltration and rejection of large tumors. Eur. J. Immunol. 2010, 40, 3325–3335. [Google Scholar] [CrossRef] [PubMed]
  110. Liu, J.; Zhang, N.; Li, Q.; Zhang, W.; Ke, F.; Leng, Q.; Wang, H.; Chen, J.; Wang, H. Tumor-associated macrophages recruit CCR6+ regulatory t cells and promote the development of colorectal cancer via enhancing CCL20 production in mice. PLoS ONE 2011, 6, e19495. [Google Scholar] [CrossRef] [PubMed]
  111. Takahashi, T.; Ibata, M.; Yu, Z.; Shikama, Y.; Endo, Y.; Miyauchi, Y.; Nakamura, M.; Tashiro-Yamaji, J.; Miura-Takeda, S.; Shimizu, T.; et al. Rejection of intradermally injected syngeneic tumor cells from mice by specific elimination of tumor-associated macrophages with liposome-encapsulated dichloromethylene diphosphonate, followed by induction of cd11b+/ccr3−/GR-1− cells cytotoxic against the tumor cells. Cancer Immunol. Immunother. 2009, 58, 2011–2023. [Google Scholar] [PubMed]
  112. Srivastava, M.K.; Zhu, L.; Harris-White, M.; Kar, U.K.; Huang, M.; Johnson, M.F.; Lee, J.M.; Elashoff, D.; Strieter, R.; Dubinett, S.; et al. Myeloid suppressor cell depletion augments antitumor activity in lung cancer. PLoS ONE 2012, 7, e40677. [Google Scholar] [CrossRef]
  113. Zeisberger, S.M.; Odermatt, B.; Marty, C.; Zehnder-Fjallman, A.H.; Ballmer-Hofer, K.; Schwendener, R.A. Clodronate-liposome-mediated depletion of tumour-associated macrophages: A new and highly effective antiangiogenic therapy approach. Br. J. Cancer 2006, 95, 272–281. [Google Scholar] [CrossRef] [PubMed]
  114. Morales, J.K.; Kmieciak, M.; Graham, L.; Feldmesser, M.; Bear, H.D.; Manjili, M.H. Adoptive transfer of her2/neu-specific t cells expanded with alternating gamma chain cytokines mediate tumor regression when combined with the depletion of myeloid-derived suppressor cells. Cancer Immunol. Immunother. 2009, 58, 941–953. [Google Scholar] [CrossRef] [PubMed]
  115. Damuzzo, V.; Pinton, L.; Desantis, G.; Solito, S.; Marigo, I.; Bronte, V.; Mandruzzato, S. Complexity and challenges in defining myeloid-derived suppressor cells. Cytom. B Clin Cytom. 2015, 88, 77–91. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, J.; Ioan-Facsinay, A.; van der Voort, E.I.; Huizinga, T.W.; Toes, R.E. Transient expression of foxp3 in human activated nonregulatory cd4+ t cells. European J. Immunol. 2007, 37, 129–138. [Google Scholar] [CrossRef] [PubMed]
  117. Whiteside, T.L.; Schuler, P.; Schilling, B. Induced and natural regulatory t cells in human cancer. Expert Opin. Biol. Ther. 2012, 12, 1383–1397. [Google Scholar] [CrossRef] [PubMed]
  118. Solito, S.; Marigo, I.; Pinton, L.; Damuzzo, V.; Mandruzzato, S.; Bronte, V. Myeloid-derived suppressor cell heterogeneity in human cancers. Ann. N. Y. Acad. Sci. 2014, 1319, 47–65. [Google Scholar] [CrossRef] [PubMed]
  119. Whiteside, T.L. What are regulatory T cells (Treg) regulating in cancer and why? Semin Cancer Biol. 2012, 22, 327–334. [Google Scholar] [CrossRef] [PubMed]
  120. Mason, G.M.; Lowe, K.; Melchiotti, R.; Ellis, R.; de Rinaldis, E.; Peakman, M.; Heck, S.; Lombardi, G.; Tree, T.I. Phenotypic complexity of the human regulatory T cell compartment revealed by mass cytometry. J. Immunol. 2015, 195, 2030–2037. [Google Scholar] [CrossRef] [PubMed]
  121. Dijkgraaf, E.M.; Santegoets, S.J.; Reyners, A.K.; Goedemans, R.; Nijman, H.W.; van Poelgeest, M.I.; van Erkel, A.R.; Smit, V.T.; Daemen, T.A.; van der Hoeven, J.J.; et al. A phase 1/2 study combining gemcitabine, pegintron and p53 slp vaccine in patients with platinum-resistant ovarian cancer. Oncotarget 2015, 6, 32228–32243. [Google Scholar] [PubMed]
  122. Mandruzzato, S.; Brandau, S.; Britten, C.M.; Bronte, V.; Damuzzo, V.; Gouttefangeas, C.; Maurer, D.; Ottensmeier, C.; van der Burg, S.H.; Welters, M.J.; et al. Toward harmonized phenotyping of human myeloid-derived suppressor cells by flow cytometry: Results from an interim study. Cancer Immunol. Immunother. 2016, 65, 161–169. [Google Scholar] [CrossRef] [PubMed]
  123. Santegoets, S.J.A.M.; Dijkgraaf, E.M.; Battaglia, A.; Beckhove, P.; Britten, C.M.; Gallimore, A.; Godkin, A.; Gouttefangeas, C.; de Gruijl, T.D.; Koenen, H.J.P.M.; et al. Monitoring regulatory t cells in clinical samples: Consensus on an essential marker set and gating strategy for regulatory T cell analysis by flow cytometry. Cancer Immunol. Immunother. 2015, 64, 1271–1286. [Google Scholar] [CrossRef] [PubMed]
  124. Ikemoto, T.; Yamaguchi, T.; Morine, Y.; Imura, S.; Soejima, Y.; Fujii, M.; Maekawa, Y.; Yasutomo, K.; Shimada, M. Clinical roles of increased populations of Foxp3+CD4+ T cells in peripheral blood from advanced pancreatic cancer patients. Pancreas 2006, 33, 386–390. [Google Scholar] [CrossRef] [PubMed]
  125. Griffiths, R.W.; Elkord, E.; Gilham, D.E.; Ramani, V.; Clarke, N.; Stern, P.L.; Hawkins, R.E. Frequency of regulatory T cells in renal cell carcinoma patients and investigation of correlation with survival. Cancer Immunol. Immunother. 2007, 56, 1743–1753. [Google Scholar] [CrossRef] [PubMed]
  126. Hansen, G.L.; Gaudernack, G.; Brunsvig, P.F.; Cvancarova, M.; Kyte, J.A. Immunological factors influencing clinical outcome in lung cancer patients after telomerase peptide vaccination. Cancer Immunol. Immunother. 2015, 64, 1609–1621. [Google Scholar] [CrossRef] [PubMed]
  127. Nishikawa, H.; Sakaguchi, S. Regulatory T cells in cancer immunotherapy. Curr. Opin. Immunol. 2014, 27, 1–7. [Google Scholar] [CrossRef] [PubMed]
  128. Solito, S.; Falisi, E.; Diaz-Montero, C.M.; Doni, A.; Pinton, L.; Rosato, A.; Francescato, S.; Basso, G.; Zanovello, P.; Onicescu, G.; et al. A human promyelocytic-like population is responsible for the immune suppression mediated by myeloid-derived suppressor cells. Blood 2011, 118, 2254–2265. [Google Scholar] [CrossRef] [PubMed]
  129. Walter, S.; Weinschenk, T.; Stenzl, A.; Zdrojowy, R.; Pluzanska, A.; Szczylik, C.; Staehler, M.; Brugger, W.; Dietrich, P.-Y.; Mendrzyk, R.; et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat. Med. 2012, 18, 1254–1261. [Google Scholar] [CrossRef] [PubMed]
  130. Filipazzi, P.; Pilla, L.; Mariani, L.; Patuzzo, R.; Castelli, C.; Camisaschi, C.; Maurichi, A.; Cova, A.; Rigamonti, G.; Giardino, F.; et al. Limited induction of tumor cross-reactive T cells without a measurable clinical benefit in early melanoma patients vaccinated with human leukocyte antigen class I-modified peptides. Am. Assoc. Cancer Res. 2012, 18, 6485–6496. [Google Scholar] [CrossRef] [PubMed]
  131. Weide, B.; Martens, A.; Zelba, H.; Stutz, C.; Derhovanessian, E.; Di Giacomo, A.M.; Maio, M.; Sucker, A.; Schilling, B.; Schadendorf, D.; et al. Myeloid-derived suppressor cells predict survival of patients with advanced melanoma: Comparison with regulatory T cells and NY-ESO-1- or melan-A-specific T cells. Clin. Cancer Res. 2014, 20, 1601–1609. [Google Scholar] [CrossRef] [PubMed]
  132. Lin, G.N.; Peng, J.W.; Xiao, J.J.; Liu, D.Y.; Xia, Z.J. Prognostic impact of circulating monocytes and lymphocyte-to-monocyte ratio on previously untreated metastatic non-small cell lung cancer patients receiving platinum-based doublet. Med. Oncol. 2014. [Google Scholar] [CrossRef] [PubMed]
  133. Zhang, Q.W.; Liu, L.; Gong, C.Y.; Shi, H.S.; Zeng, Y.H.; Wang, X.Z.; Zhao, Y.W.; Wei, Y.Q. Prognostic significance of tumor-associated macrophages in solid tumor: A meta-analysis of the literature. PLoS ONE 2012, 7, e50946. [Google Scholar] [CrossRef] [PubMed]
  134. Trellakis, S.; Bruderek, K.; Dumitru, C.A.; Gholaman, H.; Gu, X.; Bankfalvi, A.; Scherag, A.; Hutte, J.; Dominas, N.; Lehnerdt, G.F.; et al. Polymorphonuclear granulocytes in human head and neck cancer: Enhanced inflammatory activity, modulation by cancer cells and expansion in advanced disease. Int. J. Cancer 2011, 129, 2183–2193. [Google Scholar] [CrossRef] [PubMed]
  135. Arigami, T.; Okumura, H.; Matsumoto, M.; Uchikado, Y.; Uenosono, Y.; Kita, Y.; Owaki, T.; Mori, S.; Kurahara, H.; Kijima, Y.; et al. Analysis of the fibrinogen and neutrophil-lymphocyte ratio in esophageal squamous cell carcinoma: A promising blood marker of tumor progression and prognosis. Medicine 2015, 94, e1702. [Google Scholar] [CrossRef] [PubMed]
  136. Sica, A.; Bronte, V. Altered macrophage differentiation and immune dysfunction in tumor development. J. Clin. Investig. 2007, 117, 1155–1166. [Google Scholar] [CrossRef] [PubMed]
  137. Jablonska, E.; Puzewska, W.; Grabowska, Z.; Jablonski, J.; Talarek, L. Vegf, il-18 and no production by neutrophils and their serum levels in patients with oral cavity cancer. Cytokine 2005, 30, 93–99. [Google Scholar] [CrossRef] [PubMed]
  138. Grabon, W.; Mielczarek-Puta, M.; Chrzanowska, A.; Baranczyk-Kuzma, A. L-arginine as a factor increasing arginase significance in diagnosis of primary and metastatic colorectal cancer. Clin. Biochem. 2009, 42, 353–357. [Google Scholar] [CrossRef] [PubMed]
  139. Polat, M.F.; Taysi, S.; Polat, S.; Boyuk, A.; Bakan, E. Elevated serum arginase activity levels in patients with breast cancer. Surg. Today 2003, 33, 655–661. [Google Scholar] [CrossRef] [PubMed]
  140. Wu, G.; Morris, S.M., Jr. Arginine metabolism: Nitric oxide and beyond. Biochem. J. 1998, 336, 1–17. [Google Scholar] [CrossRef] [PubMed]
  141. Bronte, V.; Zanovello, P. Regulation of immune responses by l-arginine metabolism. Nat. Rev. Immunol. 2005, 5, 641–654. [Google Scholar] [CrossRef] [PubMed]
  142. Bronte, V.; Serafini, P.; Mazzoni, A.; Segal, D.M.; Zanovello, P. L-arginine metabolism in myeloid cells controls t-lymphocyte functions. Trends Immunol. 2003, 24, 302–306. [Google Scholar] [CrossRef]
  143. Youn, J.I.; Nagaraj, S.; Collazo, M.; Gabrilovich, D.I. Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J. Immunol. 2008, 181, 5791–5802. [Google Scholar] [CrossRef] [PubMed]
  144. Kusmartsev, S.; Nefedova, Y.; Yoder, D.; Gabrilovich, D.I. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J. Immunol. 2004, 172, 989–999. [Google Scholar] [CrossRef] [PubMed]
  145. Nagaraj, S.; Schrum, A.G.; Cho, H.I.; Celis, E.; Gabrilovich, D.I. Mechanism of T cell tolerance induced by myeloid-derived suppressor cells. J. Immunol. 2010, 184, 3106–3116. [Google Scholar] [CrossRef] [PubMed]
  146. Nagaraj, S.; Gupta, K.; Pisarev, V.; Kinarsky, L.; Sherman, S.; Kang, L.; Herber, D.L.; Schneck, J.; Gabrilovich, D.I. Altered recognition of antigen is a mechanism of CD8+ t cell tolerance in cancer. Nat. Med. 2007, 13, 828–835. [Google Scholar] [CrossRef] [PubMed]
  147. Munn, D.H.; Mellor, A.L. IDO in the tumor microenvironment: Inflammation, counter-regulation, and tolerance. Trends Immunol. 2016, 37, 193–207. [Google Scholar] [CrossRef] [PubMed]
  148. Kotsakis, A.; Harasymczuk, M.; Schilling, B.; Georgoulias, V.; Argiris, A.; Whiteside, T.L. Myeloid-derived suppressor cell measurements in fresh and cryopreserved blood samples. J. Immunol. Methods 2012, 381, 14–22. [Google Scholar] [CrossRef] [PubMed]
  149. Zhang, H.; Li, Z.L.; Ye, S.B.; Ouyang, L.Y.; Chen, Y.S.; He, J.; Huang, H.Q.; Zeng, Y.X.; Zhang, X.S.; Li, J. Myeloid-derived suppressor cells inhibit t cell proliferation in human extranodal nk/t cell lymphoma: A novel prognostic indicator. Cancer Immunol. Immunother. 2015, 64, 1587–1599. [Google Scholar] [CrossRef] [PubMed]
  150. Idorn, M.; Kollgaard, T.; Kongsted, P.; Sengelov, L.; Thor Straten, P. Correlation between frequencies of blood monocytic myeloid-derived suppressor cells, regulatory T cells and negative prognostic markers in patients with castration-resistant metastatic prostate cancer. Cancer Immunol. Immunother. 2014, 63, 1177–1187. [Google Scholar] [CrossRef] [PubMed]
  151. Chevolet, I.; Speeckaert, R.; Schreuer, M.; Neyns, B.; Krysko, O.; Bachert, C.; Hennart, B.; Allorge, D.; van Geel, N.; Van Gele, M.; et al. Characterization of the in vivo immune network of IDO, tryptophan metabolism, PD-L1, and CTLA-4 in circulating immune cells in melanoma. Oncoimmunology 2015, 4, e982382. [Google Scholar] [CrossRef] [PubMed]
  152. Furuzawa-Carballeda, J.; Lima, G.; Uribe-Uribe, N.; Avila-Casado, C.; Mancilla, E.; Morales-Buenrostro, L.E.; Perez-Garrido, J.; Perez, M.; Cardenas, G.; Llorente, L.; et al. High levels of ido-expressing CD16+ peripheral cells, and tregs in graft biopsies from kidney transplant recipients under belatacept treatment. Transpl. Proc. 2010, 42, 3489–3496. [Google Scholar] [CrossRef] [PubMed]
  153. Raber, P.L.; Thevenot, P.; Sierra, R.; Wyczechowska, D.; Halle, D.; Ramirez, M.E.; Ochoa, A.C.; Fletcher, M.; Velasco, C.; Wilk, A.; et al. Subpopulations of myeloid-derived suppressor cells impair T cell responses through independent nitric oxide-related pathways. Int. J. Cancer 2014, 134, 2853–2864. [Google Scholar] [CrossRef] [PubMed]
  154. Gielen, P.R.; Schulte, B.M.; Kers-Rebel, E.D.; Verrijp, K.; Bossman, S.A.; Ter Laan, M.; Wesseling, P.; Adema, G.J. Elevated levels of polymorphonuclear myeloid-derived suppressor cells in patients with glioblastoma highly express S100A8/9 and arginase and suppress T cell function. Neuro Oncol. 2016, 18, 1253–1264. [Google Scholar] [CrossRef] [PubMed]
  155. Huang, A.; Zhang, B.; Wang, B.; Zhang, F.; Fan, K.X.; Guo, Y.J. Increased CD14(+)HLA-DR (-/low) myeloid-derived suppressor cells correlate with extrathoracic metastasis and poor response to chemotherapy in non-small cell lung cancer patients. Cancer Immunol. Immunother. 2013, 62, 1439–1451. [Google Scholar] [CrossRef] [PubMed]
  156. Mougiakakos, D.; Jitschin, R.; von Bahr, L.; Poschke, I.; Gary, R.; Sundberg, B.; Gerbitz, A.; Ljungman, P.; Le Blanc, K. Immunosuppressive CD14+HLA-DRlow/Neg IDO+ myeloid cells in patients following allogeneic hematopoietic stem cell transplantation. Leukemia 2013, 27, 377–388. [Google Scholar] [CrossRef] [PubMed]
  157. Vasquez-Dunddel, D.; Pan, F.; Zeng, Q.; Gorbounov, M.; Albesiano, E.; Fu, J.; Blosser, R.L.; Tam, A.J.; Bruno, T.; Zhang, H.; et al. STAT3 regulates arginase-I in myeloid-derived suppressor cells from cancer patients. J. Clin. Investig. 2013, 123, 1580–1589. [Google Scholar] [CrossRef] [PubMed]
  158. Corzo, C.A.; Condamine, T.; Lu, L.; Cotter, M.J.; Youn, J.I.; Cheng, P.; Cho, H.I.; Celis, E.; Quiceno, D.G.; Padhya, T.; et al. HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J. Exp. Med. 2010, 207, 2439–2453. [Google Scholar] [CrossRef] [PubMed]
  159. Tembhre, M.K.; Parihar, A.S.; Sharma, V.K.; Sharma, A.; Chattopadhyay, P.; Gupta, S. Alteration in regulatory T cells and programmed cell death 1-expressing regulatory T cells in active generalized vitiligo and their clinical correlation. Br. J. Dermatol. 2015, 172, 940–950. [Google Scholar] [CrossRef] [PubMed]
  160. Li, J.; Srivastava, R.M.; Ettyreddy, A.; Ferris, R.L. Cetuximab ameliorates suppressive phenotypes of myeloid antigen presenting cells in head and neck cancer patients. J. Immunother. Cancer 2015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Bandura, D.R.; Baranov, V.I.; Ornatsky, O.I.; Antonov, A.; Kinach, R.; Lou, X.; Pavlov, S.; Vorobiev, S.; Dick, J.E.; Tanner, S.D. Mass cytometry: Technique for real time single cell multitarget immunoassay based on inductively coupled plasma time-of-flight mass spectrometry. Anal. Chem. 2009, 81, 6813–6822. [Google Scholar] [CrossRef] [PubMed]
  162. Bendall, S.C.; Simonds, E.F.; Qiu, P.; Amir el, A.D.; Krutzik, P.O.; Finck, R.; Bruggner, R.V.; Melamed, R.; Trejo, A.; Ornatsky, O.I.; et al. Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science 2011, 332, 687–696. [Google Scholar] [CrossRef] [PubMed]
  163. Bendall, S.C.; Nolan, G.P.; Roederer, M.; Chattopadhyay, P.K. A deep profiler’s guide to cytometry. Trends Immunol. 2012, 33, 323–332. [Google Scholar] [CrossRef] [PubMed]
  164. Dixon, A.R.; Bathany, C.; Tsuei, M.; White, J.; Barald, K.F.; Takayama, S. Recent developments in multiplexing techniques for immunohistochemistry. Expert Rev. Mol. Diagn. 2015, 15, 1171–1186. [Google Scholar] [CrossRef] [PubMed]
  165. Huang, W.; Hennrick, K.; Drew, S. A colorful future of quantitative pathology: Validation of vectra technology using chromogenic multiplexed immunohistochemistry and prostate tissue microarrays. Hum. Pathol. 2013, 44, 29–38. [Google Scholar] [CrossRef] [PubMed]
  166. Glass, G.; Papin, J.A.; Mandell, J.W. Simple: A sequential immunoperoxidase labeling and erasing method. J. Histochem. Cytochem. 2009, 57, 899–905. [Google Scholar] [CrossRef] [PubMed]
  167. Remark, R.; Merghoub, T.; Damotte, D.; Wochok, J.D.; Merad, M.; Gnjatic, S. Abstract b109: In-depth tissue analysis using multiplexed immunohistochemical consecutive staining on single slide. Cancer Immunol. Res. 2016. [Google Scholar] [CrossRef]
  168. Giesen, C.; Wang, H.A.; Schapiro, D.; Zivanovic, N.; Jacobs, A.; Hattendorf, B.; Schuffler, P.J.; Grolimund, D.; Buhmann, J.M.; Brandt, S.; et al. Highly multiplexed imaging of tumor tissues with subcellular resolution by mass cytometry. Nat. Methods 2014, 11, 417–422. [Google Scholar] [CrossRef] [PubMed]
  169. Schuler, P.J.; Harasymczuk, M.; Schilling, B.; Lang, S.; Whiteside, T.L. Separation of human cd4+cd39+ t cells by magnetic beads reveals two phenotypically and functionally different subsets. J. Immunol. Methods 2011, 369, 59–68. [Google Scholar] [CrossRef] [PubMed]
  170. Strauss, L.; Bergmann, C.; Gooding, W.; Johnson, J.T.; Whiteside, T.L. The frequency and suppressor function of cd4+cd25highfoxp3+ t cells in the circulation of patients with squamous cell carcinoma of the head and neck. Clin. Cancer Res. 2007, 13, 6301–6311. [Google Scholar] [CrossRef] [PubMed]
  171. Lai, C.; August, S.; Albibas, A.; Behar, R.; Cho, S.Y.; Polak, M.E.; Theaker, J.; MacLeod, A.S.; French, R.R.; Glennie, M.J.; et al. Ox40+ regulatory t cells in cutaneous squamous cell carcinoma suppress effector t-cell responses and associate with metastatic potential. Clin. Cancer Res. 2016. [Google Scholar] [CrossRef] [PubMed]
  172. Yang, J.H.; Cutler, A.J.; Ferreira, R.C.; Reading, J.L.; Cooper, N.J.; Wallace, C.; Clarke, P.; Smyth, D.J.; Boyce, C.S.; Gao, G.J.; et al. Natural variation in interleukin-2 sensitivity influences regulatory t-cell frequency and function in individuals with long-standing type 1 diabetes. Diabetes 2015, 64, 3891–3902. [Google Scholar] [CrossRef] [PubMed]
  173. Brusko, T.M.; Hulme, M.A.; Myhr, C.B.; Haller, M.J.; Atkinson, M.A. Assessing the in vitro suppressive capacity of regulatory t cells. Immunol. Investig. 2007, 36, 607–628. [Google Scholar] [CrossRef] [PubMed]
  174. Bonertz, A.; Weitz, J.; Pietsch, D.H.; Rahbari, N.N.; Schlude, C.; Ge, Y.; Juenger, S.; Vlodavsky, I.; Khazaie, K.; Jaeger, D.; et al. Antigen-specific tregs control t cell responses against a limited repertoire of tumor antigens in patients with colorectal carcinoma. J. Clin. Investig. 2009, 119, 3311–3321. [Google Scholar] [CrossRef] [PubMed]
  175. Horn, T.; Grab, J.; Schusdziarra, J.; Schmid, S.; Maurer, T.; Nawroth, R.; Wolf, P.; Pritsch, M.; Gschwend, J.E.; Kubler, H.R.; et al. Antitumor t cell responses in bladder cancer are directed against a limited set of antigens and are modulated by regulatory t cells and routine treatment approaches. Int. J. Cancer 2013, 133, 2145–2156. [Google Scholar] [CrossRef] [PubMed]
  176. Scurr, M.; Bloom, A.; Pembroke, T.; Srinivasan, R.; Brown, C.; Smart, K.; Bridgeman, H.; Davies, M.; Hargest, R.; Phillips, S.; et al. Escalating regulation of 5t4-specific ifn-gamma+ cd4+ t cells distinguishes colorectal cancer patients from healthy controls and provides a target for in vivo therapy. Cancer Immunol. Res. 2013. [Google Scholar] [CrossRef] [PubMed]
  177. Piersma, S.J.; van der Hulst, J.M.; Kwappenberg, K.M.; Goedemans, R.; van der Minne, C.E.; van der Burg, S.H. Influenza matrix 1-specific human CD4+ Foxp3+ and Foxp3(−) regulatory t cells can be detected long after viral clearance. Eur. J. Immunol. 2010, 40, 3064–3074. [Google Scholar] [CrossRef] [PubMed]
  178. Zhou, G.; Levitsky, H.I. Natural regulatory t cells and de novo-induced regulatory t cells contribute independently to tumor-specific tolerance. J. Immunol. 2007, 178, 2155–2162. [Google Scholar] [CrossRef] [PubMed]
  179. Zhou, G.; Drake, C.G.; Levitsky, H.I. Amplification of tumor-specific regulatory T cells following therapeutic cancer vaccines. Blood 2006, 107, 628–636. [Google Scholar] [CrossRef] [PubMed]
  180. Vence, L.; Palucka, A.K.; Fay, J.W.; Ito, T.; Liu, Y.J.; Banchereau, J.; Ueno, H. Circulating tumor antigen-specific regulatory t cells in patients with metastatic melanoma. Proc. Natl. Acad. Sci. USA 2007, 104, 20884–20889. [Google Scholar] [CrossRef] [PubMed]
  181. Van der Burg, S.H.; Piersma, S.J.; de Jong, A.; van der Hulst, J.M.; Kwappenberg, K.M.; van den Hende, M.; Welters, M.J.; Van Rood, J.J.; Fleuren, G.J.; Melief, C.J.; et al. Association of cervical cancer with the presence of CD4+ regulatory T cells specific for human papillomavirus antigens. Proc. Natl. Acad. Sci. USA 2007, 104, 12087–12092. [Google Scholar] [CrossRef] [PubMed]
  182. Benoist, C.; Mathis, D. Treg cells, life history, and diversity. Cold Spring Harb. Perspect. Biol. 2012, 4, a007021. [Google Scholar] [CrossRef] [PubMed]
  183. Sakaguchi, S.; Miyara, M.; Costantino, C.M.; Hafler, D.A. Foxp3+ regulatory t cells in the human immune system. Nat. Rev. Immunol. 2010, 10, 490–500. [Google Scholar] [CrossRef] [PubMed]
  184. Miyara, M.; Sakaguchi, S. Human Foxp3(+)CD4(+) regulatory t cells: Their knowns and unknowns. Immunol. Cell Biol. 2011, 89, 346–351. [Google Scholar] [CrossRef] [PubMed]
  185. Davidson, T.S.; Shevach, E.M. Polyclonal treg cells modulate T effector cell trafficking. Eur. J. Immunol. 2011, 41, 2862–2870. [Google Scholar] [CrossRef] [PubMed]
  186. Chattopadhyay, G.; Shevach, E.M. Antigen-specific induced T regulatory cells impair dendritic cell function via an IL-10/MARCH1-dependent mechanism. J. Immunol. 2013, 191, 5875–5884. [Google Scholar] [CrossRef] [PubMed]
  187. Littwitz-Salomon, E.; Akhmetzyanova, I.; Vallet, C.; Francois, S.; Dittmer, U.; Gibbert, K. Activated regulatory T cells suppress effector NK cell responses by an IL-2-mediated mechanism during an acute retroviral infection. Retrovirology 2015. [Google Scholar] [CrossRef] [PubMed]
  188. Okamura, T.; Sumitomo, S.; Morita, K.; Iwasaki, Y.; Inoue, M.; Nakachi, S.; Komai, T.; Shoda, H.; Miyazaki, J.; Fujio, K.; et al. TGF-beta3-expressing CD4+CD25(−)lag3+ regulatory t cells control humoral immune responses. Nat. Commun. 2015. [Google Scholar] [CrossRef] [PubMed]
  189. Shevach, E.M. Biological functions of regulatory T cells. Adv. Immunol. 2011, 112, 137–176. [Google Scholar] [PubMed]
  190. Duhen, T.; Duhen, R.; Lanzavecchia, A.; Sallusto, F.; Campbell, D.J. Functionally distinct subsets of human Foxp3+ Treg cells that phenotypically mirror effector th cells. Blood 2012, 119, 4430–4440. [Google Scholar] [CrossRef] [PubMed]
  191. Schoenbrunn, A.; Frentsch, M.; Kohler, S.; Keye, J.; Dooms, H.; Moewes, B.; Dong, J.; Loddenkemper, C.; Sieper, J.; Wu, P.; et al. A converse 4–1bb and CD40 ligand expression pattern delineates activated regulatory t cells (treg) and conventional t cells enabling direct isolation of alloantigen-reactive natural foxp3+ treg. J. Immunol. 2012, 189, 5985–5994. [Google Scholar] [CrossRef] [PubMed]
  192. Bacher, P.; Kniemeyer, O.; Schonbrunn, A.; Sawitzki, B.; Assenmacher, M.; Rietschel, E.; Steinbach, A.; Cornely, O.A.; Brakhage, A.A.; Thiel, A.; et al. Antigen-specific expansion of human regulatory T cells as a major tolerance mechanism against mucosal fungi. Mucosal Immunol. 2014, 7, 916–928. [Google Scholar] [CrossRef] [PubMed]
  193. Wang, R.; Kozhaya, L.; Mercer, F.; Khaitan, A.; Fujii, H.; Unutmaz, D. Expression of garp selectively identifies activated human foxp3+ regulatory t cells. Proc. Natl. Acad. Sci. USA 2009, 106, 13439–13444. [Google Scholar] [CrossRef] [PubMed]
  194. Tran, D.Q.; Andersson, J.; Wang, R.; Ramsey, H.; Unutmaz, D.; Shevach, E.M. Garp (LRRC32) is essential for the surface expression of latent Tgf-beta on platelets and activated Foxp3+ regulatory T cells. Proc. Natl. Acad. Sci. USA 2009, 106, 13445–13450. [Google Scholar] [CrossRef] [PubMed]
  195. Keskinov, A.A.; Shurin, M.R. Myeloid regulatory cells in tumor spreading and metastasis. Immunobiology 2015, 220, 236–242. [Google Scholar] [CrossRef] [PubMed]
  196. Peranzoni, E.; Zilio, S.; Marigo, I.; Dolcetti, L.; Zanovello, P.; Mandruzzato, S.; Bronte, V. Myeloid-derived suppressor cell heterogeneity and subset definition. Curr. Opin. Immunol. 2010, 22, 238–244. [Google Scholar] [CrossRef] [PubMed]
  197. Youn, J.I.; Gabrilovich, D.I. The biology of myeloid-derived suppressor cells: The blessing and the curse of morphological and functional heterogeneity. Eur. J. Immunol. 2010, 40, 2969–2975. [Google Scholar] [CrossRef] [PubMed]
  198. Solito, S.; Pinton, L.; Damuzzo, V.; Mandruzzato, S. Highlights on molecular mechanisms of mdsc-mediated immune suppression: Paving the way for new working hypotheses. Immunol. Investig. 2012, 41, 722–737. [Google Scholar] [CrossRef] [PubMed]
  199. Cuenca, A.G.; Delano, M.J.; Kelly-Scumpia, K.M.; Moreno, C.; Scumpia, P.O.; Laface, D.M.; Heyworth, P.G.; Efron, P.A.; Moldawer, L.L. A paradoxical role for myeloid-derived suppressor cells in sepsis and trauma. Mol. Med. 2011, 17, 281–292. [Google Scholar] [CrossRef] [PubMed]
  200. Ostrand-Rosenberg, S.; Sinha, P. Myeloid-derived suppressor cells: Linking inflammation and cancer. J. Immunol. 2009, 182, 4499–4506. [Google Scholar] [CrossRef] [PubMed]
  201. Stromnes, I.M.; Brockenbrough, J.S.; Izeradjene, K.; Carlson, M.A.; Cuevas, C.; Simmons, R.M.; Greenberg, P.D.; Hingorani, S.R. Targeted depletion of an mdsc subset unmasks pancreatic ductal adenocarcinoma to adaptive immunity. Gut 2014, 63, 1769–1781. [Google Scholar] [CrossRef] [PubMed]
  202. Novitskiy, S.V.; Ryzhov, S.; Zaynagetdinov, R.; Goldstein, A.E.; Huang, Y.; Tikhomirov, O.Y.; Blackburn, M.R.; Biaggioni, I.; Carbone, D.P.; Feoktistov, I.; et al. Adenosine receptors in regulation of dendritic cell differentiation and function. Blood 2008, 112, 1822–1831. [Google Scholar] [CrossRef] [PubMed]
  203. Loercher, A.E.; Nash, M.A.; Kavanagh, J.J.; Platsoucas, C.D.; Freedman, R.S. Identification of an il-10-producing HLA-DR-negative monocyte subset in the malignant ascites of patients with ovarian carcinoma that inhibits cytokine protein expression and proliferation of autologous t cells. J. Immunol. 1999, 163, 6251–6260. [Google Scholar] [PubMed]
  204. Filipazzi, P.; Valenti, R.; Huber, V.; Pilla, L.; Canese, P.; Iero, M.; Castelli, C.; Mariani, L.; Parmiani, G.; Rivoltini, L. Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J. Clin. Oncol. 2007, 25, 2546–2553. [Google Scholar] [CrossRef] [PubMed]
  205. Mandruzzato, S.; Solito, S.; Falisi, E.; Francescato, S.; Chiarion-Sileni, V.; Mocellin, S.; Zanon, A.; Rossi, C.R.; Nitti, D.; Bronte, V.; et al. IL4ralpha+ myeloid-derived suppressor cell expansion in cancer patients. J. Immunol. 2009, 182, 6562–6568. [Google Scholar] [CrossRef] [PubMed]
  206. Duffy, A.; Zhao, F.; Haile, L.; Gamrekelashvili, J.; Fioravanti, S.; Ma, C.; Kapanadze, T.; Compton, K.; Figg, W.D.; Greten, T.F. Comparative analysis of monocytic and granulocytic myeloid-derived suppressor cell subsets in patients with gastrointestinal malignancies. Cancer Immunol. Immunother. 2013, 62, 299–307. [Google Scholar] [CrossRef] [PubMed]
  207. Solito, S.; Bronte, V.; Mandruzzato, S. Antigen specificity of immune suppression by myeloid-derived suppressor cells. J. Leukoc. Biol. 2011, 90, 31–36. [Google Scholar] [CrossRef] [PubMed]
  208. Hock, B.D.; McKenzie, J.L. Suppression of cd3/cd28 antibody stimulated responses by human granulocytic myeloid-derived suppressor cells: Fact or artefact? Immunol. Lett. 2013, 152, 151–152. [Google Scholar] [CrossRef] [PubMed]
  209. Poschke, I.; Mougiakakos, D.; Hansson, J.; Masucci, G.V.; Kiessling, R. Immature immunosuppressive CD14+HLA-DR-/low cells in melanoma patients are stat3hi and overexpress CD80, CD83, and DC-sign. Cancer Res. 2010, 70, 4335–4345. [Google Scholar] [CrossRef] [PubMed]
  210. Rodriguez, P.C.; Zea, A.H.; Culotta, K.S.; Zabaleta, J.; Ochoa, J.B.; Ochoa, A.C. Regulation of t cell receptor CD3zeta chain expression by l-arginine. J. Biol. Chem. 2002, 277, 21123–21129. [Google Scholar] [CrossRef] [PubMed]
  211. Srivastava, M.K.; Sinha, P.; Clements, V.K.; Rodriguez, P.; Ostrand-Rosenberg, S. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res. 2010, 70, 68–77. [Google Scholar] [CrossRef] [PubMed]
  212. Liu, C.Y.; Wang, Y.M.; Wang, C.L.; Feng, P.H.; Ko, H.W.; Liu, Y.H.; Wu, Y.C.; Chu, Y.; Chung, F.T.; Kuo, C.H.; 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. [Google Scholar] [CrossRef] [PubMed]
  213. Talebian Yazdi, M.; Loof, N.M.; Franken, K.L.; Taube, C.; Oostendorp, J.; Hiemstra, P.S.; Welters, M.J.; van der Burg, S.H. Local and systemic XAGE-1B-specific immunity in patients with lung adenocarcinoma. Cancer Immunol. Immunother. 2015, 64, 1109–1121. [Google Scholar] [CrossRef] [PubMed]
  214. Cheng, P.; Corzo, C.A.; Luetteke, N.; Yu, B.; Nagaraj, S.; Bui, M.M.; Ortiz, M.; Nacken, W.; Sorg, C.; Vogl, T.; et al. Inhibition of dendritic cell differentiation and accumulation of myeloid-derived suppressor cells in cancer is regulated by s100a9 protein. J. Exp. Med. 2008, 205, 2235–2249. [Google Scholar] [CrossRef] [PubMed]
  215. Beury, D.W.; Parker, K.H.; Nyandjo, M.; Sinha, P.; Carter, K.A.; Ostrand-Rosenberg, S. Cross-talk among myeloid-derived suppressor cells, macrophages, and tumor cells impacts the inflammatory milieu of solid tumors. J. Leukoc. Biol. 2014, 96, 1109–1118. [Google Scholar] [CrossRef] [PubMed]
  216. Sinha, P.; Clements, V.K.; Bunt, S.K.; Albelda, S.M.; Ostrand-Rosenberg, S. Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response. J. Immunol. 2007, 179, 977–983. [Google Scholar] [CrossRef] [PubMed]
  217. Hoechst, B.; Voigtlaender, T.; Ormandy, L.; Gamrekelashvili, J.; Zhao, F.; Wedemeyer, H.; Lehner, F.; Manns, M.P.; Greten, T.F.; Korangy, F. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology 2009, 50, 799–807. [Google Scholar] [CrossRef] [PubMed]
  218. Hoechst, B.; Gamrekelashvili, J.; Manns, M.P.; Greten, T.F.; Korangy, F. Plasticity of human th17 cells and itregs is orchestrated by different subsets of myeloid cells. Blood 2011, 117, 6532–6541. [Google Scholar] [CrossRef] [PubMed]
  219. Gros, A.; Turcotte, S.; Wunderlich, J.R.; Ahmadzadeh, M.; Dudley, M.E.; Rosenberg, S.A. Myeloid cells obtained from the blood but not from the tumor can suppress T-cell proliferation in patients with melanoma. Clin. Cancer Res. 2012, 18, 5212–5223. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview and measurement of the different levels of immune dysfunction present in cancer patients. Immune dysfunction can be analyzed through measuring general signs of immune suppression in the blood (top left) and through measuring the frequency and function of suppressor cells in the blood and tumor tissue (bottom left). The different parameters that can be measured studying these levels of immune dysfunction are depicted (middle), as well as the techniques that can be used (right). Abbreviations used: APC: antigen-presenting cell; Arg1: arginase 1; CBC: complete blood count; CMV: cytomegalovirus; DBC: differential blood count; EBV: Epstein-Barr virus; GARP: glycoprotein A repetitions predominant; ICS: intracellular cytokine staining; IDO: indoleamine-2,3-deoxigenase; IHC: immunohistochemistry; iNOS: inducible nitric oxide synthase; MDSC: myeloid-derived suppressor cells; MLR: mixed lymphocyte reaction; NK: natural killer; NO: nitric oxide; PBMC: peripheral blood mononuclear cells; PHA: phytohemagglutinin; qPCR: quantitative polymerase chain reaction; ROS: reactive oxygen species; SEB: staphylococcal enterotoxin B; TAA: tumor-associated antigen; TAM: tumor-associated macrophages; TAN: tumor-associated neutrophils; TGF-β: transforming growth factor receptor-β; Treg: regulatory T cells.
Figure 1. Overview and measurement of the different levels of immune dysfunction present in cancer patients. Immune dysfunction can be analyzed through measuring general signs of immune suppression in the blood (top left) and through measuring the frequency and function of suppressor cells in the blood and tumor tissue (bottom left). The different parameters that can be measured studying these levels of immune dysfunction are depicted (middle), as well as the techniques that can be used (right). Abbreviations used: APC: antigen-presenting cell; Arg1: arginase 1; CBC: complete blood count; CMV: cytomegalovirus; DBC: differential blood count; EBV: Epstein-Barr virus; GARP: glycoprotein A repetitions predominant; ICS: intracellular cytokine staining; IDO: indoleamine-2,3-deoxigenase; IHC: immunohistochemistry; iNOS: inducible nitric oxide synthase; MDSC: myeloid-derived suppressor cells; MLR: mixed lymphocyte reaction; NK: natural killer; NO: nitric oxide; PBMC: peripheral blood mononuclear cells; PHA: phytohemagglutinin; qPCR: quantitative polymerase chain reaction; ROS: reactive oxygen species; SEB: staphylococcal enterotoxin B; TAA: tumor-associated antigen; TAM: tumor-associated macrophages; TAN: tumor-associated neutrophils; TGF-β: transforming growth factor receptor-β; Treg: regulatory T cells.
Vaccines 04 00029 g001

Share and Cite

MDPI and ACS Style

Santegoets, S.J.A.M.; Welters, M.J.P.; Van der Burg, S.H. Monitoring of the Immune Dysfunction in Cancer Patients. Vaccines 2016, 4, 29. https://doi.org/10.3390/vaccines4030029

AMA Style

Santegoets SJAM, Welters MJP, Van der Burg SH. Monitoring of the Immune Dysfunction in Cancer Patients. Vaccines. 2016; 4(3):29. https://doi.org/10.3390/vaccines4030029

Chicago/Turabian Style

Santegoets, Saskia J. A. M., Marij J. P. Welters, and Sjoerd H. Van der Burg. 2016. "Monitoring of the Immune Dysfunction in Cancer Patients" Vaccines 4, no. 3: 29. https://doi.org/10.3390/vaccines4030029

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop