Skip to main content
Cancer Immunology, Immunotherapy : CII logoLink to Cancer Immunology, Immunotherapy : CII
. 2007 Jun 15;56(11):1687–1700. doi: 10.1007/s00262-007-0343-y

Dynamic cross-talk between tumor and immune cells in orchestrating the immunosuppressive network at the tumor microenvironment

Diego O Croci 1, Mariano F Zacarías Fluck 2, María J Rico 2, Pablo Matar 2, Gabriel A Rabinovich 1,3,, O Graciela Scharovsky 2,
PMCID: PMC11030157  PMID: 17571260

Abstract

Accumulating evidence indicates that a dynamic cross-talk between tumors and the immune system can regulate tumor growth and metastasis. Increased understanding of the biochemical nature of tumor antigens and the molecular mechanisms responsible for innate and adaptive immune cell activation has revolutionized the fields of tumor immunology and immunotherapy. Both the protective effects of the immune system against tumor cells (immunosurveillance) and the evasion of tumor cells from immune attack (tumor-immune escape) have led to the concept of cancer immunoediting, a proposal which infers that a bidirectional interaction between tumor and inflammatory/regulatory cells is ultimately responsible for orchestrating the immunosuppressive network at the tumor site. In this context, a major challenge is the potentiation or redirection of tumor antigen-specific immune responses. The success in reaching this goal is highly dependent on an improved understanding of the interactions and mechanisms operating during the different phases of the cancer immunoediting process. In this review, we discuss the multiple defense and counterattack strategies that tumors have devised in order to evade immune attack and to thwart the effectiveness of several immunotherapeutic approaches.

Keywords: Inflammation, Immunosuppression, Tumor-immune escape, Tumor immunoediting

Cancer Immunoediting: dynamic cross-talk between the tumor and immune cells

The “cancer immunoediting” hypothesis, recently put forward by Schreiber et al. [31], has integrated mechanisms of tumor-immune escape with classical immunosurveillance theory. The renaissance of tumor immunosurveillance as part of the new concept of “immunoediting” emerges from the idea that the host’s immune system not only protects against tumor development, but can also inadvertently promote tumor growth by selecting tumor escape variants with reduced immunogenicity. It is proposed that the process of cancer immunoediting comprises three phases. The first phase, referred to as “elimination,” encompasses our traditional understanding of cancer immunosurveillance, in which cells of the innate and adaptive immune systems recognize and thereby destroy developing tumors, thus protecting the host against cancer. The second phase is “equilibrium,” a protracted period in which ongoing tumor growth and immune surveillance enter into a dynamic balance with one another. The third phase is “escape”, where tumor variants that can avoid immune-mediated destruction emerge and develop into clinically apparent neoplasms [31].

Certainly, a better understanding of the interactions between tumors and the immune system will lead to novel and more effective cancer immunotherapy strategies. As such, we summarize here the recent findings on different mechanisms leading to tumor-immune escape, including strategies employed by tumor cells that permit them to resist immune-mediated attack, particularly those that specifically thwart or counteract effector T cell responses.

Natural history of the relationship between tumors and the immune system

The first notion of a defense system recognizing tumor cells as foreign was postulated by Paul Ehrlich in the beginning of the last century [32]. In response to his pioneering foresight, the field of tumor antigens has evolved and several new concepts have emerged as central to our current understanding. Among them, we now understand that tumor tissue is not a plain box or static entity containing a fixed pool of different antigens, but it is instead a complex and evolving tissue modulated and sculpted by the cellular microenvironment. Likewise, it is important to recognize that a solid tumor is composed of different cellular compartments, including neoplastic tissue and stromal cells [49], both of which may develop strong interactions with the immune system.

Among the earliest supporters of Ehrlich’s hypothesis were Burnet and Thomas who postulated the theory of tumor-immune surveillance [11, 131]. Burnet and Thomas proposed that tumors appeared more frequently but were eliminated efficiently by the immune system, even before they were clinically detectable. However, in spite of the presence of an effective immune system, tumors arise and develop from normal tissues and invade surrounding and distant sites, suggesting that malignant cells have the capacity to devise multiple strategies to evade or to counterattack the immune response.

Mechanisms of tumor-immune resistance

Mechanisms used to elude recognition include tumor-induced impairment of antigen presentation, activation of negative costimulatory signals, and elaboration of immunosuppressive factors (Fig. 1). In addition, cancer cells may promote the expansion and/or recruitment of regulatory cell populations that can contribute to the immunosuppressive network; these populations include regulatory T cells (Tregs), myeloid suppressor cells, and distinct subsets of immature and mature regulatory dendritic cells (DCs).

Fig. 1.

Fig. 1

Immunosuppressive strategies used by tumors to evade immune responses. Tumors employ a plethora of immunosuppressive mechanisms, which may act in concert to counteract effective immune responses. These include tumor-induced impairment of the antigen presentation machinery, activation of negative costimulatory signals in the tumor microenvironment (CTLA-4/B7, PD-1/PD-L1, Fas/ FasL), elaboration of immunosuppressive factors (IL-10, TGF-β, galectin-1, gangliosides, PGE 2), and overexpression of indoleamine 2,3 dioxygenase (IDO). In addition, different regulatory cell populations contribute to this immunosuppressive network including CD4 + CD25 + regulatory T-cells (Tregs) and inducible T regulatory (Tr1) cells which negatively impact on the fate of effector T cells. Abbreviations: Gal-1 galectin-1, PGE 2 prostaglandin-E2, TGF-β transforming growth factor-β, MHC-I major histocompatibility complex, TAP transporter-associated protein

Alterations in the antigen presentation machinery

During the past decades, many laboratory groups participated in the identification and characterization of the structure and the molecular nature of different tumor antigens recognized by T cells [20, 30, 89]. However, to elude immune recognition, tumor cells employ different mechanisms to modify, down-regulate or even lose these antigens completely.

Mechanisms that include down-regulation of antigen expression can vary from decreased expression levels to a complete loss of one or more tumor antigens [107, 118, 124]. For example, the Melan-A/MART-1 antigen characteristic of melanoma can be silenced via its promoter, thus permitting the neoplastic tissue to avoid recognition by specific T cells [62, 113].

Tumors, like viruses, can also undergo “antigenic drift” in which the accumulation of point mutations result in cell surface antigens that are no longer recognized by the specific cytotoxic T cells [4].

The endogenous (class-I) antigen presentation machinery (APM) plays a crucial role in the generation of peptides from endogenously synthesized proteins, like tumor antigens, and in their presentation to CTL. Several defects in APM components, including the transporter associated with antigen processing (TAP) and components of the immunoproteosome (LMP-2, LMP-7), have been identified in head and neck, bladder and astrocytic tumors, among others [13, 29, 70, 111]. These findings support the notion that alterations in the APM might account, at least in part, for the resistance of tumor cells to immune recognition.

Also, in order to thwart CTL responses, tumor cells lose MHC molecules or alter their expression, thus rendering the tumor antigens invisible and, therefore resistant to CTLs. In this context, more than 50% of all the tumors show a complete loss of MHC class-I molecules or loss of at least a single allele. Several laboratories demonstrated that different types of tumors, including leukemia and melanoma, decrease the expression of HLA-A and HLA-B alleles [26, 38], and that such down-regulation has important prognostic value [12]. Moreover, complete loss of expression of HLA class I alleles is a phenotype found in many human tumors [53]. The HLA loss could be caused by different mutations involving β2-microglobulin genes [16]. In addition to the aforementioned HLA abnormalities, the HLA phenotype may be altered by the hemizygous loss of HLA-A, -B and -C alleles, generally caused by the presence of large deletions in chromosome 6 [51] or loss of single HLA alleles [110].

It is clearly established that cells that express lower levels of MHC class I molecules are more susceptible to lysis by NK cells [139]. In addition, expression of non-classical HLA molecules such as HLA-G and HLA-E, which belong to the group MHC class Ib, also modulate the immune response by inhibiting NK-cell-mediated cytotoxicity [108, 114, 141]. The expression of HLA-G in melanoma cells has been shown to contribute to cell evasion of immune response by hampering NK cell recognition [92], while the expression of HLA-E seems to play a role in the inhibition of NK cell-mediated lysis [1]. HLA-E binds to the leader peptide derived from the polymorphic classical major histocompatibility molecules. This peptide binding is highly specific and stabilizes the HLA-E protein, allowing it to migrate to the cell surface where it can interact with CD94/NKG2A receptors on NK cells. This interaction inhibits NK cell-mediated lysis of cells displaying HLA-E. If the leader peptide is not present in the endoplasmic reticulum, HLA-E is unstable and is degraded before it reaches the cell surface. In damaged cells, such as virally infected or tumor cells, down-regulation of classical HLA molecules (HLA-A, HLA-B and HLA-C) prevents stabilization of HLA-E by the leader peptide. Under these circumstances, HLA-E does not reach the cell surface and the cell is then vulnerable to lysis by NK cells. In addition, even when tumor cells express normal levels of MHC-I molecules and tumor-specific antigens (TSA), the lack of costimulation (B7.1/CD80 and B7.2/CD86) would provide another mechanism by which tumor cells can circumvent the immune system and remain undetectable [34].

Since a number of excellent reviews have been devoted to defects in antigen processing and presentation [29, 70], we will not discuss these mechanisms in detail and instead will focus on active immunosuppressive strategies developed by tumor cells.

Tumor cell resistance to apoptosis

Tumor cells possess a wide variety of mechanisms that permit them to resist CTL-induced apoptosis. Although, a direct role for these mechanisms in tumor-immune escape has only been shown in a few instances, it is likely that resistance to apoptosis is relevant not only for tumorigenesis, but also has influence on mechanisms of immunosurveillance and immunotherapy.

A critical strategy used by tumors to acquire resistance to apoptosis is the over-expression of anti-apoptotic molecules. The anti-apoptotic proteins FLIPL,S interfere with the induction of apoptosis at the level of death receptors (e.g. Fas/CD95) by inhibiting recruitment of caspase-8, but they do not prevent apoptosis induced by perforin/granzyme [47, 60]. Similarly, enhanced Bcl-2 expression correlates with the grade of malignancy of human tumors and protects different tumor types from apoptosis [137], and the anti-apoptotic Bcl-2 family members, Bcl-xL and Mcl-1 are up-regulated in several types of tumors and can confer resistance to multiple apoptotic stimuli [33]. Furthermore, the IAP family member, survivin, is expressed in a highly tumor-specific manner and can confer resistance to CTL-induced apoptosis [104]. Interestingly, in addition to anti-apoptotic effects associated with the inhibition of the apoptosome formation, survivin also has a role in cell cycle regulation [104].

In addition to blockade of the death receptor pathway, tumor cells can also resist killing by CTLs through direct interference with the perforin/granzyme cytotoxic pathway. In this regard, the serine protease inhibitor PI-9/SPI-6 that inhibits granzyme B is expressed in a variety of human and murine tumors and its overexpression results in resistance of tumor cells to CTLs [6, 76]. Finally, the expression of soluble receptors that act as decoys for death ligands may also interfere with apoptosis induction via death receptors [46]. In this regard, soluble Fas/CD95 and decoy receptor 3 (DcR3) interrupt Fas/CD95-mediated signaling and inhibit FasL-induced cell death [18].

Thus, resistance of tumor cells to the effector mechanisms of CTLs can be generated by inhibition of death receptor or granzyme/perforin pro-apoptotic pathways, leading not only to escape of the tumors from immunosurveillance, but also having a profound influence on the efficacy of immunotherapy strategies.

The immunosuppressive network

IDO (Indoleamine 2,3 dioxygenase)

A connection between elevated urinary tryptophan catabolites and bladder cancer was first reported in 1956 [9]. IDO is a heme-containing enzyme that catalyzes the oxidative breakdown of the essential amino acid tryptophan via the kynurenine pathway. Elevated levels of IDO-generated catabolites have been associated with a number of malignancies, including melanoma and colon and endometrial carcinoma. Although, this phenomenon was initially thought to be a consequence of the antitumor activity of IFN-γ, which stimulates expression of IDO in tumor cells [91], an independent mechanism action has been proposed for IDO, suggesting its participation in tolerance and immunosuppression in tumor-bearing hosts [81]. Specifically, Uyttenhove et al. demonstrated that expression of IDO by immunogenic mouse tumor cells prevents their rejection by pre-immunized mice. This effect was accompanied by diminished accumulation of antigen-specific T cells at the tumor site that was partially reversed by systemic administration of 1-methyl-tryptophan (1-MT), a competitive inhibitor of IDO [132]. Beside these effects of IDO activity within the tumor itself, other findings suggested that IDO might regulate the afferent arm of the antitumor immune response, at tumor-draining lymph nodes (TDLN) [82]. IDO appears to be able to create potent local, and even systemic, immunosuppression, by a mechanism of action that still remains to be clarified. However, there is currently sufficient evidence showing that deprivation of tryptophan favors the induction of apoptosis and cell cycle arrest in effector T cells [82]. On the basis of these findings, it is possible that IDO might render the TDLN a tolerizing microenvironment, and thus contribute to tumor-immune escape. Recently, it has been shown that IDO is under the control of the tumor suppression gene Bin1 [80].

A number of pharmacological compounds that could potentially function as specific IDO inhibitors are currently available [80, 132]. By potentiating antitumor immune responses in combination with chemotherapy or other immunotherapy strategies, IDO inhibitors may offer a novel possibility of overcoming tumor-immune resistance and of promoting tumor regression. In this regard, Muller et al. have shown that small molecule inhibitors of IDO cooperate with chemotherapeutic agents to elicit regression of established tumors [80].

Expansion of T regulatory cells (Tregs) and secretion of immunosuppressive cytokines

CD4+ CD25+ FoxP3+ Tregs cells were identified by Sakaguchi et al. [117] as a natural occurring CD4+ T cell subset (comprising 5–10% of all peripheral T cells) constitutively expressing CD25 that suppresses T cell responses in vivo [117]. Tregs contribute to the prevention of autoimmune disorders by controlling the activity of autoreactive T lymphocytes and by suppressing the proliferation of antigen-specific effector and bystander T cells [117]. Recent studies have demonstrated that immunosuppression mediated by Tregs is one of the most critical mechanisms of tumor-immune escape and a major hurdle for successful tumor immunotherapy [146]. By modulating the tumor microenvironment through the secretion of selected chemokines, cancer cells can actively prevent the induction of antitumor immunity through the differentiation, expansion and/or recruitment of Tregs. Indeed, recent work has identified a distinct set of chemokines that drive the recruitment of Tregs, namely thymus and activation-regulated chemokine (TARC or CCL-17) and macrophage-derived chemoattractant (MDC or CCL-22), both chemokines with affinity for the receptor, CCR4, expressed on Tregs [147].

Natural Tregs are found at a higher frequency in peripheral blood of cancer patients compared to healthy donors and have been shown to induce tolerance at the tumor microenvironment, facilitating metastatic spread of cancer cells [56, 68]. When Tregs were depleted in mice, transplantable tumors were efficiently rejected by the host immune system [120]. Interestingly, Dannull et al. recently demonstrated that vaccine-mediated antitumor immunity could be significantly enhanced following depletion of Tregs [23]. In addition, administration of anti-CD25 monoclonal antibody and/or anti-CTLA-4 antibody for a limited period of time also provoked effective tumor-specific immunity against syngeneic tumor cells [90]. Recently, Curiel et al. demonstrated that Tregs confer immune privilege to ovary tumor cells. The authors found that large numbers of CD4+ CD25+ Foxp3+ Tregs in tumors and malignant ascites inversely correlated with the survival of patients with malignant ovarian carcinoma. Interestingly, in this clinical setting, Tregs were recruited to the tumor site under the influence of the chemokine CCL22 produced by tumor cells and macrophages in the tumor microenvironment [14].

In addition to CD4+ CD25+ FoxP3+ Tregs’ recent studies highlighted the importance of inducible T regulatory (Tr1) cells and TH3 cells in suppressing T-cell responses [145]. The immunoregulatory functions of Tr1 and TH3 cells have been attributed to their capacity to secrete immunosuppressive cytokines such as IL-10 and transforming growth factor-β (TGF-β) [112]. In contrast, naturally occurring Tregs can suppress the effector immune response by cell-cell contact or by release of immunosuppressive factors [147].

In conjunction with the major roles of Tregs and Tr1 cells in suppressing effector T cell responses, it has been postulated that an imbalance in TH1/TH2 cytokine production may also be responsible for tumor cell evasion, with a shift toward a TH2 response and induction of immunosuppressive cytokines including IL-10 and IL-4 [54]. As an example of this phenomenon, low doses of the alkylating agent cyclophosphamide (Cy), induce a TH2 shift in the cytokine profile of syngeneic lymphoma-bearing rats, which may be in part responsible for its anti-metastatic effect. Such a treatment reduced the splenic production of IL-10, TGF-β and nitric oxide, and restored the lymphoproliferative capacity [7173]. In broad agreement with early studies of North et al., who showed that Cy favors the elimination of tumor-induced suppressor T cells [88], preliminary evidence in a rat lymphoma model supports the concept that Cy may selectively eliminate CD4+CD25+ Tregs (Scharovsky et al., unpublished data).

In conclusion, the idea that removal and/or inhibition of CD4+CD25+ Tregs or inducible Tr1 cells can abrogate immunological unresponsiveness to syngeneic tumors, has established novel strategies of evoking tumor immunity that would boost other cancer immunotherapy strategies.

Tolerogenic dendritic cells (DCs) and myeloid suppressor cells (MSCs) in cancer

Dendritic cells (DCs) are critically important for the generation and maintenance of an anti-tumor immune response [125]. Data from many laboratories obtained during the past few years indicate that defects in DCs are among the main factors responsible for tumor escape. DC abnormalities manifest in several major ways in the tumor microenvironment including: (a) decreased presence of functionally competent DCs, (b) accumulation of immature DCs, and (c) expansion of tolerogenic/regulatory DCs.

Since there are several excellent reviews that have been devoted to the study of DCs in cancer [125], we will only focus here on the role of MSCs in tumor-immune escape. MSCs represent a heterogeneous population of myeloid cells, including immature macrophages, granulocytes, DCs and other myeloid cells at earlier stages of differentiation, which can be identified in mice by expression of CD11b and Gr-1 [63] These cells are capable of inactivating both CD4+ and CD8+ T cells and are responsible for tumor-related immune dysfunction. The number of MSCs in tumor-bearing hosts increases steadily during tumor progression, since the tumor-derived factors that alter myeloid differentiation accumulate in proportion to tumor burden [87, 130]. The functional activity of MSCs involves the inhibition of IFN-γ production by CD8+ T cells in response to peptide epitopes presented by MHC-I molecules on the surface of MSCs [37]. This effect depends on MHC-I expression by MSCs, requires direct cell–cell contact, and is dependent on reactive oxygen species such as hydrogen peroxide [37]. Interestingly, although nitric oxide is required for MSCs-induced T cell inhibition, the enzyme arginase-I mediates the inhibition of allogeneic T cell responses by MSCs [10]. These cells can inhibit cytokine production by T cells and this effect is abrogated by the addition of the hydrogen peroxide scavenger catalase, implicating hydrogen peroxide as a critical effector molecule similarly to the mouse MSCs [10].

Alterations of signal transduction molecules: a mechanism driven by the tumor microenvironment with consequences for tumor-immune escape

Patients in advanced stages of cancer and mice with large transplantable tumors have compromised systemic immune responses with highly decreased delayed-type hypersensitivity (DTH) responses. In this regard, T and NK cells often exhibit alterations in their proliferative and cytotoxic capacities as well as in cytokine secretion [5]. Several observations showing alterations in signal transduction molecules in T and NK cells from both tumor-bearing mice and cancer patients provide a molecular basis to understand this immune dysfunction more completely [78, 138].

Of particular interest is the correlation observed between CD3-ζ expression and the disease stage in cancer patients [75]. Interestingly, evidence has been provided for tumor-induced degradation of the CD3-ζ chain. Tumor cells can induce the activation of intracellular peptidases in T lymphocytes that is responsible for decreased or absent expression of signal transduction molecules, including the CD3-ζ chain in activated T cells [140]. Finally, other mechanisms, including generation of free oxygen radicals and increased arginase activity within the tumor microenvironment, have been proposed to account for decreased CD3-ζ expression in cancer [57, 109]. In this regard, it has been shown that human T cells stimulated and cultured in the absence of L-arginine lose the expression of the CD3-ζ chain and demonstrate impaired proliferation and decreased cytokine production [109]. Therefore, the regulation of L-arginine concentration in the microenvironment could represent an important mechanism via which the expression of CD3-ζ chain is modulated, with critical consequences in TCR-mediated signaling and T cell function.

Recently, aberrant activation of STAT3 (signal transducer and activator of transcription 3) has been proposed as a novel mechanism of tumor-immune escape [136]. It was shown that STAT3 signaling in tumor cells suppresses both, innate and adaptive anti-tumor immune responses, further enhancing tumor progression [59]. Also, inhibition of STAT3 signaling up-regulates the expression of a subset of immunoregulatory genes, including a number of chemokines that promote recruitment of effector T cells [136]. Altogether, these observations suggest that the activation of STAT3 signaling contributes to the establishment of an immunosuppressive tumor microenvironment. Proof-of-concept studies in cell culture and animal models have validated STAT3 protein as a promising molecular target for novel cancer therapies, including small molecule inhibitors of STAT3 signaling [59, 136].

Other observations of tumor-induced alterations of T-cell signal transduction molecules have been provided from metastatic colorectal carcinoma patients who failed to respond to the tumor antigen epithelial mucin-1 (MUC-1) vaccine. These non-responding patients had T cells that lacked NF-κB p65; in contrast to those patients responding to treatment, who showed increased levels of NF-κB and augmented T-cell effector functions [55]. Interestingly, alterations in NF-κB family proteins, specifically the failure of p65 translocation to the nucleus, occur earlier and more frequently than the decrease in CD3-ζ chain and these defects are paralleled by an impaired ability to produce TH1 cytokines. These initial changes are then followed by a marked decrease in CTL functions [56].

Other immunosuppressive factors in the tumor microenvironment

Other tumor-derived or tumor-induced suppressive factors result in impaired T-cell function [102]. The production of prostaglandin E2 (PGE2) by macrophages is enhanced by tumor-derived factors, which induce an immunosuppressive state in glioma-bearing hosts [85]. In addition, RCAS1, a receptor binding cancer antigen that induces cell cycle arrest and apoptosis of effector T cells, has been identified as a tumor evasion mechanism in different tumor types, mainly those of gynecological origin [86].

The Fas (CD95)/FasL (CD95L) counterattack controversy

The Fas-dependent cell death pathway is regulated by cognate interactions between the Fas receptor (CD95) and its ligand FasL (CD95L). Expression of FasL has been reported in different solid tumors, including melanoma and colon carcinoma, and its expression has been shown to confer immune privilege to tumors by delivering death signals to Fas-positive effector T cells [116].

Although, considerable heterogeneity in cell surface expression of FasL has been detected even within a particular tumor cell line [40], and non-specific staining of specific FasL antibodies has been demonstrated [127], it has been shown that FasL-positive tumor cells can kill Fas-positive T cells in vitro, demonstrating functional significance of FasL expression [45].

Several lines of evidence support the involvement of the Fas system in tumor counterattack. Fais et al. demonstrated that purified microvesicle preparations from melanoma cell supernatants are able to induce FasL-mediated apoptosis in Fas-sensitive human T cells, providing evidence of a novel potential mechanism of tumor-immune escape [3]. However, certain experimental evidence seems to contradict the FasL counterattack hypothesis, as a pro-inflammatory function for FasL has been also demonstrated [17, 77]. Also, some results on tumor counterattack are controversial given that many factors may influence FasL activity in vivo, including the different levels and the kinetics of FasL expression on tumor cells, the release of different types of cytokines, such as TGF-β and IL-10, the levels of hypoxia at the tumor site, the extent of vascularization and the accessibility to immune cell infiltration [119]. A further complicating aspect in tumor-immune escape mediated by FasL is the role of soluble FasL (sFasL) [129], which is derived from cleavage of FasL by MMPs (matrix metalloproteinases), and can also counterattack CTLs, but to a lesser extent than FasL [42]. Thus, the altered expression of FasL and the shedding of sFasL may contribute to immune evasion by allowing tumor cells to escape from CTL-mediated cytotoxicity. Finally, other death factors, such as TNF-α and TRAIL, have been shown to eliminate T cells and might also contribute to suppression of T cell responses [84].

To further elucidate the pathophysiologic relevance of the tumor counterattack hypothesis, it will be essential to use defined experimental systems, in which all the above-mentioned elements are carefully controlled and modulated. These experiments will be critical to determine whether targeting tumor counterattack mechanisms might be beneficial or detrimental for cancer therapy.

Negative regulatory pathways: CTLA-4 and the PD-1/PD-L1 system

Compelling evidence indicates that costimulatory molecules with negative regulatory functions are expressed on the cell surface of tumor cells, effector T cells and Tregs. Undoubtedly, one of the best-studied regulatory signals is mediated by CTLA-4, a ligand for B7 expressed on activated T cells and naturally occurring Tregs [66, 94, 135]. Allison et al. were pioneers in demonstrating that blockade of CTLA-4 signaling might enhance anti-tumor responses [66]. Further studies supported this concept showing that antibody-mediated blockade of CTLA-4 enhances anti-tumor immunity elicited by a GM-CSF-transduced melanoma vaccine [44, 98].

In addition, the interaction between programmed death- 1 (PD-1) and programmed death receptor ligand-1 (PD-L1) represents a clear example of how negative costimulatory signals may contribute to create an immunosuppressive microenvironment at the tumor site [144]. PD-L1, engages the inducible inhibitory receptor on activated T cells called PD-1 and induces phosphorylation of an immunoreceptor tyrosine-based inhibitory motif (ITIM) [144]. Dong et al. demonstrated the presence of PD-L1 (also called B7-H1) on the cell surface of a wide range of tumors [28]. The expression of PD-L1 on tumor cells of diverse histological origins suggested that this molecule might contribute to tumor-immune escape. In fact, it has been demonstrated that cancer cell-associated PD-L1 promotes apoptosis of antigen-specific human T cell clones in vitro and in vivo [8]. In addition, blockade of PD-L1 enhances DC-mediated T-cell activation and limited tumor growth, suggesting another potential mechanism by which PD-L1 restrains T cell–mediated anti-tumor immunity [15].

Furthermore, Kryczek et al. recently reported that B7-H4, a B7 family molecule, identifies a novel suppressive macrophage population in human ovarian carcinoma, and that depletion of B7-H4+ tumor macrophages may represent a useful strategy to enhance T cell-mediated immunity in cancer [61].

Thus, blockade of the inhibitory pathways mediated by CTLA-4, PD-L1 or B7-H4 may be complementary approaches to augment tumor-specific T cell-mediated immunity for cancer immunotherapy.

The “sweet escape”: galectins and gangliosides

Galectins are animal lectins defined by their conserved amino acid sequences and affinity for poly-N-acetyllactosamine-containing glycoconjugates [99]. Galectin-1, a proto-type member of this family, has the potential to induce T-cell apoptosis and regulate cytokine secretion, thus inhibiting or skewing T-cell effector functions [99]. Accumulating evidence indicates the expression of galectin-1 in many different tumor types including astrocytoma, melanoma and prostate, breast and colon carcinomas [21]. Interestingly, in most cases a positive correlation exists between the expression of galectin-1 in tumor and stromal cells and the aggressiveness of these tumors [67].

Given the expression of galectin-1 in most malignant tumor types and the ability of this glycan-binding protein to down-modulate T-cell responses, we hypothesized that tumors may contribute to the immunosuppressive and anti-inflammatory microenvironment through the expression of galectin-1. By a combination of in vitro and in vivo experiments using knockdown transfectants, we established a link between galectin-1-mediated immunoregulation and its contribution to tumor-immune escape [115]. Interestingly, we observed a marked reduction of tumor mass (an effect which required intact CD4+ and CD8+ T cell responses) and the generation of a tumor-specific T-cell response following blockade of the inhibitory effects of galectin-1. Our observations suggest that galectin-1 contributes to immune privilege of tumors by modulating survival and effector functions of CD4+ and CD8+ T cells. Recently, Le et al. showed a link between tumor hypoxia and galectin-1 expression in head and neck squamous carcinoma cells [65]. Consistent with our findings, the authors found that galectin-1 expression negatively correlates with the presence of CD3+ T cells in tumor sections [65].

A very recent study conducted by Park et al. in human breast carcinoma demonstrates a tight correlation between galectin-1 expression levels in cancer-associated stromal tissue and various clinicopathologic parameters, including tumor invasiveness and lymph node metastases [52]. Moreover, we found high expression levels of galectin-1, sensitive to the immunosuppressive cytokine TGF-β, in two mammary adenocarcinoma cell lines and in a lung carcinoma cell line [24]. Accordingly, we proposed that TGF-β1 might trigger a Smad-dependent pathway to control galectin-1 expression, suggesting a possible cross-talk between different immunosuppressive factors in the regulation of tumor-immune escape. Furthermore, results from our laboratory demonstrate that low doses of Cy can modulate the kinetics of galectin-1 expression in tumor cells and its proapoptotic function in a B-cell lymphoma model [103, 143].

Accumulating evidence indicates that galectin-1 critically modulates other T cell functions including sensitization of T cells to FasL-induced cell death [74], inhibition of proximal TCR signaling [19] and suppression of TH1 and proinflammatory cytokines [101]. Moreover, galectin-1 has been shown to enhance the secretion of IL-10 by activated T cells [134]. In addition, a very recent study demonstrates that Tregs express high levels of galectin-1 and that this protein is critical for the immunosuppressive activity of these cells [39].

The information obtained so far suggests that potent and selective small inhibitors of galectin-1 could be designed that could be used to prevent the immunosuppressive activity of galectin-1. In fact, molecules with such properties have already been developed for galectin-1 and other galectins [2, 100, 123]. However, the possibility to target galectin-1 expression in cancer has been hindered by the limited understanding of the multifunctional modes of action of this protein and its particular biochemical properties. In addition, these limitations are further complicated by the potential compensatory functions of other members of the galectin family.

In this regard, galectin-2, -3 and -9 also regulate T-cell apoptosis and modulate cytokine production in vitro, suggesting potential roles in tumor-immune escape [67, 128]. Interestingly, galectin-3 has been shown to act in a dual manner, either protecting cells from apoptosis or promoting apoptosis depending on whether the protein acts intracellularly [142] or is added exogenously [126]. Yang et al. demonstrated that T-cell transfectants overexpressing galectin-3 display higher growth rates than control transfectants and are protected from apoptosis induced by a variety of agents including Fas ligation and staurosporine [35, 142]. Interestingly, recent findings showed that recombinant galectin-3 can signal apoptosis of human T cells through binding to cell surface glycoconjugates resulting in activation of the mitochondrial pathway, cytochrome c release and caspase-3 activation [35]. Furthermore, Hahn et al. suggested a functional cross-talk between intracellular and extracellular galectins in the regulation of T-cell death; the authors demonstrated that galectin-1-induced cell death is inhibited by intracellular expression of galectin-3 [41].

Interestingly, Demetriou et al. reported that galectin-3 may play a role in restricting TCR complex-initiated signal transduction by forming multivalent complexes with N-glycans on the TCR [27]. Thus, galectin-3 may influence T-cell interactions with APC and control T-cell activation by negatively regulating the immunological synapse.

Gangliosides are structurally diverse acidic glycosphingolipids that exist as clusters on the cell surface and contribute to the structure of lipid rafts [122]. They function as signaling intermediates in the regulation of multiple cellular responses by modulating the activities of various receptors [48]. Enhanced production of gangliosides has been observed in different tumor types [106]. Gangliosides can inhibit multiple steps of cellular immune responses, including antigen processing and presentation, T-cell proliferation, and production of cytokines such as IFN-γ [43, 93].

Biswas et al. examined the effect of human renal cell carcinoma (RCC)-derived gangliosides on T-cell apoptosis. Induction of T cell death by tumor-derived gangliosides was partially blocked by anti-GM2, suggesting that GM2 has a role in the T-cell dysfunction observed in RCC patients [7]. In addition, the same group found that gangliosides isolated from RCC induce mitochondrial permeability transition and inhibit the activation of NF-κB, suggesting the possible contribution of these glycosphingolipids to the suppression of the inflammatory response [133]. Interestingly, it has been observed that gangliosides isolated from RCC supernatants from some patients suppressed both TH1 (IFN-γ) and TH2 (IL-5) cytokine responses, whereas gangliosides from other tumor types suppressed only TH1-mediated immune responses [7]. Thus, the cytokine profile of the anti-tumor immune response in cancer patients seems to be, at least partially, influenced by the activity of different gangliosides expressed by tumors.

Immunostimulation: is cancer-associated inflammation a friend or a foe?

The phenomenon of immunostimulation was originally noticed by Richmond Prehn who established in 1971 that a mild immune response against certain tumor types might often stimulate rather than inhibit their development [96]. The findings on human breast cancer published by Stewart et al. confirmed Prehn’s postulation [125], as they found that the progression of certain types of human mammary carcinomas at the first stages of tumor evolution is stimulated by a mild inflammatory response.

What is the rationale supporting these findings? Similar to the cross-talk between different immune cell types, cytokines, chemokines and growth factors produced by inflammatory cells and the stroma can act as paracrine growth factors stimulating tumor growth and progression. In fact, tumors that appeared in cancer-prone mice immunized against a mutant oncoprotein grew faster than in non-immunized animals [121]. Interestingly, it has also been shown that CD4+ T cells can enhance skin cancer progression under certain circumstances [22]. Besides, recent findings support the notion that B lymphocytes are required for establishing chronic inflammatory states that promote de novo carcinogenesis [25]. Related to soluble factors released by immune cells, several publications proposed their association with immunostimulation. IL-10 and its receptor (IL-10R) can stimulate the growth of metastatic cells of a B-cell lymphoma, establishing an autocrine loop [105]. In addition, it has been demonstrated that liver metastases are enhanced in human colorectal carcinomas by increased production of IL-10, induced by carcinoembryonic antigen (CEA) [50]. Also, immune cells can release other type of soluble factors, like those involved in angiogenesis, a critical process for tumor progression [79]. In fact, serum levels of VEGF and/or other angiogenic molecules are positively associated with cancer progression [95]. In contrast, other studies report a critical role for VEGF in the suppression of DC differentiation and maturation [36].

Remarkably, Karin et al. demonstrated that inflammation-induced tumor growth depends on TNF-α production by host hematopoietic cells and NF-κB activation in tumor cells [69]. Inhibition of NF-κB in tumor cells can convert tumor growth into tumor regression [69]. Finally, in very elegant studies, Langowski et al. recently showed that IL-12 promotes infiltration of CTLs and subsequent immunosurveillance, while IL-23, which shares the p40 subunit with IL-12, promotes tumor-associated inflammatory responses mediated by the matrix metalloproteinase MMP-9 and angiogenic factors, but reduces CTL infiltration [64]. Thus, closely related cytokines may positively or negatively influence tumor progression by modulating the magnitude and quality of tumor-associated inflammatory responses.

Concluding remarks and future perspectives

There is currently no doubt concerning the wide variety of mechanisms that tumors employ in order to evade immune attack. These immune escape strategies range from a passive failure to express MHC or resistance to CTL-mediated cytotoxicity to more active strategies involving expression of inhibitory factors and immunosuppressive cytokines (e.g. TGF-β, IL-10, galectin-1, IDO), activation of negative regulatory pathways (e.g CTLA-4, PD-1/PD-L1, B7-H4), acquisition of counterattack mechanisms (FasL, TRAIL) and recruitment of regulatory immune cells (CD4+CD25+FoxP3+ Tregs, Tr1 cells, tolerogenic DCs and MSCs). Investigation during the past few years has provided novel insights into the cellular and molecular mechanisms involved in the bidirectional cross-talk between tumor cells and the immune system. Understanding this functional dialogue and the hierarchical status of different tumor-immune escape mechanisms at different stages of tumor progression will guide the design of novel therapeutic strategies aiming to destroy the “tumor fortress”. In this regard, it will be of particular interest to investigate the kinetics of the interactions between different inhibitory molecules and endogenous factors that influence the expansion and trafficking of Tregs, Tr1 and tolerogenic DCs within tumor-draining lymph nodes and the tumor surroundings. In addition it will be of critical importance to determine the effectiveness of combined strategies involving blockade of different inhibitory signals (PD-1/PD-L1, IDO, TGF-β, IL-10, VEGF, galectin-1) together with conventional chemotherapy, vaccination or adoptive transfer of effector CTLs. In fact, progress has been made in this direction by evaluating the effects of combined strategies, such as GM-CSF-secreting vaccines plus CTLA-4 blockade or OX-40 costimulation, and chemotherapy plus IDO blockade [58, 83, 97, 135]. The current wealth of available data promises a future scenario in which inhibition of tumor escape strategies and removal of inhibitory signals in the tumor microenvironment will be successful in combination with other therapeutic strategies to overcome immunological tolerance and promote tumor rejection.

Acknowledgments

We would like to give special thanks to Dr. Helene F. Rosenberg (NIAID, NIH, USA) for critical reading of the manuscript. We are also grateful to the members of our laboratories, in particular Marta Toscano, Juan Ilarregui, Germán Bianco and Mariana Salatino for their invaluable help. We apologize that we could not cite many excellent studies because of space limitations. We would like to give special thanks to Fundación Sales (Argentina) for continuous support. Work in the GAR`s laboratories is supported by grants from Cancer Research Institute (Elaine Shepard Investigator Award USA), John Simon Guggenheim Memorial Foundation (USA), Agencia de Promoción Científica y Tecnológica (PICT 2003-05-13787) and University of Buenos Aires (M091, Argentina). Work in O.G.S. laboratory is supported by grants from National University of Rosario (Argentina) and Fundación Sales (Argentina). G.A.R. and P.M are members of the scientific career of the National Research Council (CONICET, Argentina), O.G.S. is a member of the scientific career of the Research Council of the National University of Rosario (CIUNR, Argentina) and D.O.C. is a fellow of the University of Buenos Aires. M.J.R. thanks CONICET and M.F.Z.F. thanks ANPCyT and Fundación SALES for the fellowships granted.

Footnotes

Diego O. Croci, Mariano F. Zacarías Fluck contributed equally to this work.

Gabriel A. Rabinovich, O. Graciela Scharovsky contributed equally to this work and should be considered as senior authors.

Contributor Information

Gabriel A. Rabinovich, Email: gabyrabi@ciudad.com.ar

O. Graciela Scharovsky, Email: ogs@citynet.net.ar.

References

  • 1.Algarra I, Garcia-Lora A, Cabrera T, Ruiz-Cabello F, Garrido F. The selection of tumor variants with altered expression of classical and nonclassical MHC class I molecules: implications for tumor immune escape. Cancer Immunol Immunother. 2004;53:904–910. doi: 10.1007/s00262-004-0517-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Andre S, Pieters RJ, Vrasidas I, Kaltner H, Kuwabara I, Liu FT, Liskamp RM, Gabius HJ. Wedgelike glycodendrimers as inhibitors of binding of mammalian galectins to glycoproteins, lactose maxiclusters, and cell surface glycoconjugates. Chembiochem. 2001;2:822–830. doi: 10.1002/1439-7633(20011105)2:11<822::AID-CBIC822>3.0.CO;2-W. [DOI] [PubMed] [Google Scholar]
  • 3.Andreola G, Rivoltini L, Castelli C, Huber V, Perego P, Deho P, Squarcina P, Accornero P, Lozupone F, Lugini L, Stringaro A, Molinari A, Arancia G, Gentile M, Parmiani G, Fais S. Induction of lymphocyte apoptosis by tumor cell secretion of FasL-bearing microvesicles. J Exp Med. 2002;195:1303–1316. doi: 10.1084/jem.20011624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bai XF, Liu JQ, Joshi PS, Wang L, Yin L, Labanowska J, Heerema N, Zheng P, Liu Y. Different lineages of P1A-expressing cancer cells use divergent modes of immune evasion for T-cell adoptive therapy. Cancer Res. 2006;66:8241–8249. doi: 10.1158/0008-5472.CAN-06-0279. [DOI] [PubMed] [Google Scholar]
  • 5.Baxevanis CN, Papamichail M. Characterization of the anti-tumor immune response in human cancers and strategies for immunotherapy. Crit Rev Oncol Hematol. 1994;16:157–179. doi: 10.1016/1040-8428(94)90069-8. [DOI] [PubMed] [Google Scholar]
  • 6.Bird CH, Sutton VR, Sun J, Hirst CE, Novak A, Kumar S, Trapani JA, Bird PI. Selective regulation of apoptosis: the cytotoxic lymphocyte serpin proteinase inhibitor 9 protects against granzyme B-mediated apoptosis without perturbing the Fas cell death pathway. Mol Cell Biol. 1998;18:6387–6398. doi: 10.1128/mcb.18.11.6387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Biswas K, Richmond A, Rayman P, Biswas S, Thornton M, Sa G, Das T, Zhang R, Chahlavi A, Tannenbaum CS, Novick A, Bukowski R, Finke JH. GM2 expression in renal cell carcinoma: potential role in tumor-induced T-cell dysfunction. Cancer Res. 2006;66:6816–6825. doi: 10.1158/0008-5472.CAN-06-0250. [DOI] [PubMed] [Google Scholar]
  • 8.Blank C, Kuball J, Voelkl S, Wiendl H, Becker B, Walter B, Majdic O, Gajewski TF, Theobald M, Andreesen R, Mackensen A. Blockade of PD-L1 (B7-H1) augments human tumor-specific T cell responses in vitro. Int J Cancer. 2006;119:317–327. doi: 10.1002/ijc.21775. [DOI] [PubMed] [Google Scholar]
  • 9.Boyland E, Williams DC. The metabolism of tryptophan. 2. The metabolism of tryptophan in patients suffering from cancer of the bladder. Biochem J. 1956;64:578–582. doi: 10.1042/bj0640578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bronte V, Serafini P, De Santo C, Marigo I, Tosello V, Mazzoni A, Segal DM, Staib C, Lowel M, Sutter G, Colombo MP, Zanovello P. IL-4-induced arginase 1 suppresses alloreactive T cells in tumor-bearing mice. J Immunol. 2003;170:270–278. doi: 10.4049/jimmunol.170.1.270. [DOI] [PubMed] [Google Scholar]
  • 11.Burnet FM. The concept of immunological surveillance. Prog Exp Tumor Res. 1970;13:1–27. doi: 10.1159/000386035. [DOI] [PubMed] [Google Scholar]
  • 12.Cabrera T, Lara E, Romero JM, Maleno I, Real LM, Ruiz-Cabello F, Valero P, Camacho FM, Garrido F. HLA class I expression in metastatic melanoma correlates with tumor development during autologous vaccination. Cancer Immunol Immunother. 2007;56:709–717. doi: 10.1007/s00262-006-0226-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Cresswell AC, Sisley K, Laws D, Parsons MA, Rennie IG, Murray AK. Reduced expression of TAP-1 and TAP-2 in posterior uveal melanoma is associated with progression to metastatic disease. Melanoma Res. 2001;11:275–281. doi: 10.1097/00008390-200106000-00009. [DOI] [PubMed] [Google Scholar]
  • 14.Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, Evdemon-Hogan M, Conejo-Garcia JR, Zhang L, Burow M, Zhu Y, Wei S, Kryczek I, Daniel B, Gordon A, Myers L, Lackner A, Disis ML, Knutson KL, Chen L, Zou W. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10:942–949. doi: 10.1038/nm1093. [DOI] [PubMed] [Google Scholar]
  • 15.Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, Krzysiek R, Knutson KL, Daniel B, Zimmermann MC, David O, Burow M, Gordon A, Dhurandhar N, Myers L, Berggren R, Hemminki A, Alvarez RD, Emilie D, Curiel DT, Chen L, Zou W. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med. 2003;9:562–567. doi: 10.1038/nm863. [DOI] [PubMed] [Google Scholar]
  • 16.Chang CC, Ogino T, Mullins DW, Oliver JL, Yamshchikov GV, Bandoh N, Slingluff CL, Jr, Ferrone S. Defective human leukocyte antigen class I-associated antigen presentation caused by a novel beta2-microglobulin loss-of-function in melanoma cells. J Biol Chem. 2006;281:18763–18773. doi: 10.1074/jbc.M511525200. [DOI] [PubMed] [Google Scholar]
  • 17.Chen JJ, Sun Y, Nabel GJ. Regulation of the proinflammatory effects of Fas ligand (CD95L) Science. 1998;282:1714–1717. doi: 10.1126/science.282.5394.1714. [DOI] [PubMed] [Google Scholar]
  • 18.Cheng J, Zhou T, Liu C, Shapiro JP, Brauer MJ, Kiefer MC, Barr PJ, Mountz JD. Protection from Fas-mediated apoptosis by a soluble form of the Fas molecule. Science. 1994;263:1759–1762. doi: 10.1126/science.7510905. [DOI] [PubMed] [Google Scholar]
  • 19.Chung CD, Patel VP, Moran M, Lewis LA, Miceli MC. Galectin-1 induces partial TCR zeta-chain phosphorylation and antagonizes processive TCR signal transduction. J Immunol. 2000;165:3722–3729. doi: 10.4049/jimmunol.165.7.3722. [DOI] [PubMed] [Google Scholar]
  • 20.Dalgleish A, Pandha H. Tumor antigens as surrogate markers and targets for therapy and vaccines. Adv Cancer Res. 2007;96:175–190. doi: 10.1016/S0065-230X(06)96009-6. [DOI] [PubMed] [Google Scholar]
  • 21.Danguy A, Camby I, Kiss R. Galectins and cancer. Biochim Biophys Acta. 2002;1572:285–293. doi: 10.1016/s0304-4165(02)00315-x. [DOI] [PubMed] [Google Scholar]
  • 22.Daniel D, Meyer-Morse N, Bergsland EK, Dehne K, Coussens LM, Hanahan D. Immune enhancement of skin carcinogenesis by CD4+ T cells. J Exp Med. 2003;197:1017–1028. doi: 10.1084/jem.20021047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Dannull J, Su Z, Rizzieri D, Yang BK, Coleman D, Yancey D, Zhang A, Dahm P, Chao N, Gilboa E, Vieweg J. Enhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. J Clin Invest. 2005;115:3623–3633. doi: 10.1172/JCI25947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Daroqui CM, Ilarregui JM, Rubinstein N, Salatino M, Toscano MA, Vazquez P, Bakin A, Puricelli L, de Bal Kier Joffe E, Rabinovich GA. Regulation of galectin-1 expression by transforming growth factor beta1 in metastatic mammary adenocarcinoma cells: implications for tumor-immune escape. Cancer Immunol Immunother. 2007;56:491–499. doi: 10.1007/s00262-006-0208-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.de Visser KE, Korets LV, Coussens LM. De novo carcinogenesis promoted by chronic inflammation is B lymphocyte dependent. Cancer Cell. 2005;7:411–423. doi: 10.1016/j.ccr.2005.04.014. [DOI] [PubMed] [Google Scholar]
  • 26.Demanet C, Mulder A, Deneys V, Worsham MJ, Maes P, Claas FH, Ferrone S. Down-regulation of HLA-A and HLA-Bw6, but not HLA-Bw4, allospecificities in leukemic cells: an escape mechanism from CTL and NK attack? Blood. 2004;103:3122–3130. doi: 10.1182/blood-2003-07-2500. [DOI] [PubMed] [Google Scholar]
  • 27.Demetriou M, Granovsky M, Quaggin S, Dennis JW. Negative regulation of T-cell activation and autoimmunity by Mgat5 N-glycosylation. Nature. 2001;409:733–739. doi: 10.1038/35055582. [DOI] [PubMed] [Google Scholar]
  • 28.Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, Lennon VA, Celis E, Chen L. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800. doi: 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
  • 29.Drake CG, Jaffee E, Pardoll DM. Mechanisms of immune evasion by tumors. Adv Immunol. 2006;90:51–81. doi: 10.1016/S0065-2776(06)90002-9. [DOI] [PubMed] [Google Scholar]
  • 30.Dudley ME, Rosenberg SA. Adoptive-cell-transfer therapy for the treatment of patients with cancer. Nat Rev Cancer. 2003;3:666–675. doi: 10.1038/nrc1167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004;21:137–148. doi: 10.1016/j.immuni.2004.07.017. [DOI] [PubMed] [Google Scholar]
  • 32.Erlich P. Ueber den jetzigen. Stand der Karzinomforschung Ned Tijdschr Geneeskd. 1909;5:273–290. [Google Scholar]
  • 33.Findley HW, Gu L, Yeager AM, Zhou M. Expression and regulation of Bcl-2, Bcl-xl, and Bax correlate with p53 status and sensitivity to apoptosis in childhood acute lymphoblastic leukemia. Blood. 1997;89:2986–2993. [PubMed] [Google Scholar]
  • 34.Frydecka I, Kosmaczewska A, Bocko D, Ciszak L, Wolowiec D, Kuliczkowski K, Kochanowska I. Alterations of the expression of T-cell-related costimulatory CD28 and downregulatory CD152 (CTLA-4) molecules in patients with B-cell chronic lymphocytic leukaemia. Br J Cancer. 2004;90:2042–2048. doi: 10.1038/sj.bjc.6601833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Fukumori T, Takenaka Y, Yoshii T, Kim HR, Hogan V, Inohara H, Kagawa S, Raz A. CD29 and CD7 mediate galectin-3-induced type II T-cell apoptosis. Cancer Res. 2003;63:8302–8311. [PubMed] [Google Scholar]
  • 36.Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, Kavanaugh D, Carbone DP. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells [published erratum appears in Nat Med 1996 Nov;2(11):1267] Nat Med. 1996;2:1096–1103. doi: 10.1038/nm1096-1096. [DOI] [PubMed] [Google Scholar]
  • 37.Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J Immunol. 2001;166:5398–5406. doi: 10.4049/jimmunol.166.9.5398. [DOI] [PubMed] [Google Scholar]
  • 38.Garcia-Lora A, Algarra I, Garrido F. MHC class I antigens, immune surveillance, and tumor immune escape. J Cell Physiol. 2003;195:346–355. doi: 10.1002/jcp.10290. [DOI] [PubMed] [Google Scholar]
  • 39.Garin MI, Chu CC, Golshayan D, Cernuda-Morollon E, Wait R, Lechler RI. Galectin-1: a key effector of regulation mediated by CD4+CD25+ T cells. Blood. 2007;109:2058–2065. doi: 10.1182/blood-2006-04-016451. [DOI] [PubMed] [Google Scholar]
  • 40.Gastman BR, Johnson DE, Whiteside TL, Rabinowich H. Tumor-induced apoptosis of T lymphocytes: elucidation of intracellular apoptotic events. Blood. 2000;95:2015–2023. [PubMed] [Google Scholar]
  • 41.Hahn HP, Pang M, He J, Hernandez JD, Yang RY, Li LY, Wang X, Liu FT, Baum LG. Galectin-1 induces nuclear translocation of endonuclease G in caspase- and cytochrome c-independent T cell death. Cell Death Differ. 2004;11:1277–1286. doi: 10.1038/sj.cdd.4401485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hallermalm K, De Geer A, Kiessling R, Levitsky V, Levitskaya J. Autocrine secretion of Fas ligand shields tumor cells from Fas-mediated killing by cytotoxic lymphocytes. Cancer Res. 2004;64:6775–6782. doi: 10.1158/0008-5472.CAN-04-0508. [DOI] [PubMed] [Google Scholar]
  • 43.Heitger A, Ladisch S. Gangliosides block antigen presentation by human monocytes. Biochim Biophys Acta. 1996;1303:161–168. doi: 10.1016/0005-2760(96)00091-4. [DOI] [PubMed] [Google Scholar]
  • 44.Hodi FS, Mihm MC, Soiffer RJ, Haluska FG, Butler M, Seiden MV, Davis T, Henry-Spires R, MacRae S, Willman A, Padera R, Jaklitsch MT, Shankar S, Chen TC, Korman A, Allison JP, Dranoff G. Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci USA. 2003;100:4712–4717. doi: 10.1073/pnas.0830997100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Igney FH, Behrens CK, Krammer PH. CD95L mediates tumor counterattack in vitro but induces neutrophil-independent tumor rejection in vivo. Int J Cancer. 2005;113:78–87. doi: 10.1002/ijc.20538. [DOI] [PubMed] [Google Scholar]
  • 46.Igney FH, Krammer PH. Immune escape of tumors: apoptosis resistance and tumor counterattack. J Leukoc Biol. 2002;71:907–920. [PubMed] [Google Scholar]
  • 47.Irmler M, Thome M, Hahne M, Schneider P, Hofmann K, Steiner V, Bodmer JL, Schroter M, Burns K, Mattmann C, Rimoldi D, French LE, Tschopp J. Inhibition of death receptor signals by cellular FLIP. Nature. 1997;388:190–195. doi: 10.1038/40657. [DOI] [PubMed] [Google Scholar]
  • 48.Iwabuchi K, Yamamura S, Prinetti A, Handa K, Hakomori S. GM3-enriched microdomain involved in cell adhesion and signal transduction through carbohydrate-carbohydrate interaction in mouse melanoma B16 cells. J Biol Chem. 1998;273:9130–9138. doi: 10.1074/jbc.273.15.9130. [DOI] [PubMed] [Google Scholar]
  • 49.Jain R. Physiological resistance to the treatment of solid tumors. In: Teidier BA, editor. Drug resistance in oncology. New York: Dekker; 1993. pp. 87–105. [Google Scholar]
  • 50.Jessup JM, Samara R, Battle P, Laguinge LM. Carcinoembryonic antigen promotes tumor cell survival in liver through an IL-10-dependent pathway. Clin Exp Metastasis. 2004;21:709–717. doi: 10.1007/s10585-004-7705-z. [DOI] [PubMed] [Google Scholar]
  • 51.Jimenez P, Canton J, Collado A, Cabrera T, Serrano A, Real LM, Garcia A, Ruiz-Cabello F, Garrido F. Chromosome loss is the most frequent mechanism contributing to HLA haplotype loss in human tumors. Int J Cancer. 1999;83:91–97. doi: 10.1002/(sici)1097-0215(19990924)83:1<91::aid-ijc17>3.0.co;2-4. [DOI] [PubMed] [Google Scholar]
  • 52.Jung EJ, Moon HG, Cho BI, Jeong CY, Joo YT, Lee YJ, Hong SC, Choi SK, Ha WS, Kim JW, Lee CW, Lee JS, Park ST. Galectin-1 expression in cancer-associated stromal cells correlates tumor invasiveness and tumor progression in breast cancer. Int J Cancer. 2007;120:2331–2338. doi: 10.1002/ijc.22434. [DOI] [PubMed] [Google Scholar]
  • 53.Kageshita T, Ishihara T, Campoli M, Ferrone S. Selective monomorphic and polymorphic HLA class I antigenic determinant loss in surgically removed melanoma lesions. Tissue Antigens. 2005;65:419–428. doi: 10.1111/j.1399-0039.2005.00381.x. [DOI] [PubMed] [Google Scholar]
  • 54.Kidd P. Th1/Th2 balance: the hypothesis, its limitations, and implications for health and disease. Altern Med Rev. 2003;8:223–246. [PubMed] [Google Scholar]
  • 55.Kiessling R, Wasserman K, Horiguchi S, Kono K, Sjoberg J, Pisa P, Petersson M. Tumor-induced immune dysfunction. Cancer Immunol Immunother. 1999;48:353–362. doi: 10.1007/s002620050586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Knutson KL, Disis ML, Salazar LG. CD4 regulatory T cells in human cancer pathogenesis. Cancer Immunol Immunother. 2007;56:271–285. doi: 10.1007/s00262-006-0194-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kono K, Salazar-Onfray F, Petersson M, Hansson J, Masucci G, Wasserman K, Nakazawa T, Anderson P, Kiessling R. Hydrogen peroxide secreted by tumor-derived macrophages down-modulates signal-transducing zeta molecules and inhibits tumor-specific T cell-and natural killer cell-mediated cytotoxicity. Eur J Immunol. 1996;26:1308–1313. doi: 10.1002/eji.1830260620. [DOI] [PubMed] [Google Scholar]
  • 58.Korman AJ, Peggs KS, Allison JP. Checkpoint blockade in cancer immunotherapy. Adv Immunol. 2006;90:297–339. doi: 10.1016/S0065-2776(06)90008-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Kortylewski M, Kujawski M, Wang T, Wei S, Zhang S, Pilon-Thomas S, Niu G, Kay H, Mule J, Kerr WG, Jove R, Pardoll D, Yu H. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nat Med. 2005;11:1314–1321. doi: 10.1038/nm1325. [DOI] [PubMed] [Google Scholar]
  • 60.Krueger A, Baumann S, Krammer PH, Kirchhoff S. FLICE-inhibitory proteins: regulators of death receptor-mediated apoptosis. Mol Cell Biol. 2001;21:8247–8254. doi: 10.1128/MCB.21.24.8247-8254.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kryczek I, Zou L, Rodriguez P, Zhu G, Wei S, Mottram P, Brumlik M, Cheng P, Curiel T, Myers L, Lackner A, Alvarez X, Ochoa A, Chen L, Zou W. B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma. J Exp Med. 2006;203:871–881. doi: 10.1084/jem.20050930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kurnick JT, Ramirez-Montagut T, Boyle LA, Andrews DM, Pandolfi F, Durda PJ, Butera D, Dunn IS, Benson EM, Gobin SJ, van den Elsen PJ. A novel autocrine pathway of tumor escape from immune recognition: melanoma cell lines produce a soluble protein that diminishes expression of the gene encoding the melanocyte lineage melan-A/MART-1 antigen through down-modulation of its promoter. J Immunol. 2001;167:1204–1211. doi: 10.4049/jimmunol.167.3.1204. [DOI] [PubMed] [Google Scholar]
  • 63.Kusmartsev S, Gabrilovich DI. Role of immature myeloid cells in mechanisms of immune evasion in cancer. Cancer Immunol Immunother. 2006;55:237–45. doi: 10.1007/s00262-005-0048-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Langowski JL, Zhang X, Wu L, Mattson JD, Chen T, Smith K, Basham B, McClanahan T, Kastelein RA, Oft M. IL-23 promotes tumour incidence and growth. Nature. 2006;442:461–465. doi: 10.1038/nature04808. [DOI] [PubMed] [Google Scholar]
  • 65.Le QT, Shi G, Cao H, Nelson DW, Wang Y, Chen EY, Zhao S, Kong C, Richardson D, O’Byrne KJ, Giaccia AJ, Koong AC. Galectin-1: a link between tumor hypoxia and tumor immune privilege. J Clin Oncol. 2005;23:8932–8941. doi: 10.1200/JCO.2005.02.0206. [DOI] [PubMed] [Google Scholar]
  • 66.Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271:1734–1736. doi: 10.1126/science.271.5256.1734. [DOI] [PubMed] [Google Scholar]
  • 67.Liu F, Rabinovich G. Galectins as modulators of tumourprogression. Nat Rev Cancer. 2005;5(1):29–41. doi: 10.1038/nrc1527. [DOI] [PubMed] [Google Scholar]
  • 68.Liyanage UK, Moore TT, Joo HG, Tanaka Y, Herrmann V, Doherty G, Drebin JA, Strasberg SM, Eberlein TJ, Goedegebuure PS, Linehan DC. Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma. J Immunol. 2002;169:2756–2761. doi: 10.4049/jimmunol.169.5.2756. [DOI] [PubMed] [Google Scholar]
  • 69.Luo JL, Maeda S, Hsu LC, Yagita H, Karin M. Inhibition of NF-kappaB in cancer cells converts inflammation- induced tumor growth mediated by TNFalpha to TRAIL-mediated tumor regression. Cancer Cell. 2004;6:297–305. doi: 10.1016/j.ccr.2004.08.012. [DOI] [PubMed] [Google Scholar]
  • 70.Marincola FM, Jaffee EM, Hicklin DJ, Ferrone S. Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance. Adv Immunol. 2000;74:181–273. doi: 10.1016/s0065-2776(08)60911-6. [DOI] [PubMed] [Google Scholar]
  • 71.Matar P, Rozados VR, Gervasoni SI, Scharovsky GO. Th2/Th1 switch induced by a single low dose of cyclophosphamide in a rat metastatic lymphoma model. Cancer Immunol Immunother. 2002;50:588–596. doi: 10.1007/s00262-001-0237-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Matar P, Rozados VR, Gervasoni SI, Scharovsky OG. Down regulation of T-cell-derived IL-10 production by low-dose cyclophosphamide treatment in tumor-bearing rats restores in vitro normal lymphoproliferative response. Int Immunopharmacol. 2001;1:307–319. doi: 10.1016/s1567-5769(00)00028-x. [DOI] [PubMed] [Google Scholar]
  • 73.Matar P, Rozados VR, Gonzalez AD, Dlugovitzky DG, Bonfil RD, Scharovsky OG. Mechanism of antimetastatic immunopotentiation by low-dose cyclophosphamide. Eur J Cancer. 2000;36:1060–1066. doi: 10.1016/s0959-8049(00)00044-7. [DOI] [PubMed] [Google Scholar]
  • 74.Matarrese P, Tinari A, Mormone E, Bianco GA, Toscano MA, Ascione B, Rabinovich GA, Malorni W. Galectin-1 sensitizes resting human T lymphocytes to Fas (CD95)-mediated cell death via mitochondrial hyperpolarization, budding and fission. J Biol Chem. 2004;280(8):6969–6985. doi: 10.1074/jbc.M409752200. [DOI] [PubMed] [Google Scholar]
  • 75.Matsuda M, Petersson M, Lenkei R, Taupin JL, Magnusson I, Mellstedt H, Anderson P, Kiessling R. Alterations in the signal-transducing molecules of T cells and NK cells in colorectal tumor-infiltrating, gut mucosal and peripheral lymphocytes: correlation with the stage of the disease. Int J Cancer. 1995;61:765–772. doi: 10.1002/ijc.2910610605. [DOI] [PubMed] [Google Scholar]
  • 76.Medema JP, de Jong J, Peltenburg LT, Verdegaal EM, Gorter A, Bres SA, Franken KL, Hahne M, Albar JP, Melief CJ, Offringa R. Blockade of the granzyme B/perforin pathway through overexpression of the serine protease inhibitor PI-9/SPI-6 constitutes a mechanism for immune escape by tumors. Proc Natl Acad Sci USA. 2001;98:11515–11520. doi: 10.1073/pnas.201398198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Miwa K, Asano M, Horai R, Iwakura Y, Nagata S, Suda T. Caspase 1-independent IL-1beta release and inflammation induced by the apoptosis inducer Fas ligand. Nat Med. 1998;4:1287–1292. doi: 10.1038/3276. [DOI] [PubMed] [Google Scholar]
  • 78.Mizoguchi H, O’Shea JJ, Longo DL, Loeffler CM, McVicar DW, Ochoa AC. Alterations in signal transduction molecules in T lymphocytes from tumor-bearing mice. Science. 1992;258:1795–1798. doi: 10.1126/science.1465616. [DOI] [PubMed] [Google Scholar]
  • 79.Mor F, Quintana FJ, Cohen IR. Angiogenesis-inflammation cross-talk: vascular endothelial growth factor is secreted by activated T cells and induces Th1 polarization. J Immunol. 2004;172:4618–4623. doi: 10.4049/jimmunol.172.7.4618. [DOI] [PubMed] [Google Scholar]
  • 80.Muller AJ, DuHadaway JB, Donover PS, Sutanto-Ward E, Prendergast GC. Inhibition of indoleamine 2,3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy. Nat Med. 2005;11:312–329. doi: 10.1038/nm1196. [DOI] [PubMed] [Google Scholar]
  • 81.Munn DH. Indoleamine 2,3-dioxygenase, tumor-induced tolerance and counter-regulation. Curr Opin Immunol. 2006;18:220–225. doi: 10.1016/j.coi.2006.01.002. [DOI] [PubMed] [Google Scholar]
  • 82.Munn DH, Sharma MD, Hou D, Baban B, Lee JR, Antonia SJ, Messina JL, Chandler P, Koni PA, Mellor AL. Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Invest. 2004;114:280–290. doi: 10.1172/JCI21583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Murata S, Ladle BH, Kim PS, Lutz ER, Wolpoe ME, Ivie SE, Smith HM, Armstrong TD, Emens LA, Jaffee EM, Reilly RT. OX40 costimulation synergizes with GM-CSF whole-cell vaccination to overcome established CD8+ T cell tolerance to an endogenous tumor antigen. J Immunol. 2006;176:974–983. doi: 10.4049/jimmunol.176.2.974. [DOI] [PubMed] [Google Scholar]
  • 84.Nagata S. Apoptosis by death factor. Cell. 1997;88:355–365. doi: 10.1016/s0092-8674(00)81874-7. [DOI] [PubMed] [Google Scholar]
  • 85.Nakano Y, Kuroda E, Kito T, Yokota A, Yamashita U. Induction of macrophagic prostaglandin E2 synthesis by glioma cells. J Neurosurg. 2006;104:574–582. doi: 10.3171/jns.2006.104.4.574. [DOI] [PubMed] [Google Scholar]
  • 86.Nakashima M, Sonoda K, Watanabe T. Inhibition of cell growth and induction of apoptotic cell death by the human tumor-associated antigen RCAS1. Nat Med. 1999;5:938–942. doi: 10.1038/11383. [DOI] [PubMed] [Google Scholar]
  • 87.Nefedova Y, Huang M, Kusmartsev S, Bhattacharya R, Cheng P, Salup R, Jove R, Gabrilovich D. Hyperactivation of STAT3 is involved in abnormal differentiation of dendritic cells in cancer. J Immunol. 2004;172:464–474. doi: 10.4049/jimmunol.172.1.464. [DOI] [PubMed] [Google Scholar]
  • 88.North RJ. Cyclophosphamide-facilitated adoptive immunotherapy of an established tumor depends on elimination of tumor-induced suppressor T cells. J Exp Med. 1982;155:1063–1074. doi: 10.1084/jem.155.4.1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Novellino L, Castelli C, Parmiani G. A listing of human tumor antigens recognized by T cells: March 2004 update. Cancer Immunol Immunother. 2005;54:187–207. doi: 10.1007/s00262-004-0560-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Onizuka S, Tawara I, Shimizu J, Sakaguchi S, Fujita T, Nakayama E. Tumor rejection by in vivo administration of anti-CD25 (interleukin-2 receptor alpha) monoclonal antibody. Cancer Res. 1999;59:3128–3133. [PubMed] [Google Scholar]
  • 91.Ozaki Y, Edelstein MP, Duch DS. Induction of indoleamine 2,3-dioxygenase: a mechanism of the antitumor activity of interferon gamma. Proc Natl Acad Sci USA. 1988;85:1242–1246. doi: 10.1073/pnas.85.4.1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Paul P, Rouas-Freiss N, Khalil-Daher I, Moreau P, Riteau B, Le Gal FA, Avril MF, Dausset J, Guillet JG, Carosella ED. HLA-G expression in melanoma: a way for tumor cells to escape from immunosurveillance. Proc Natl Acad Sci USA. 1998;95:4510–4515. doi: 10.1073/pnas.95.8.4510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Peguet-Navarro J, Sportouch M, Popa I, Berthier O, Schmitt D, Portoukalian J. Gangliosides from human melanoma tumors impair dendritic cell differentiation from monocytes and induce their apoptosis. J Immunol. 2003;170:3488–3494. doi: 10.4049/jimmunol.170.7.3488. [DOI] [PubMed] [Google Scholar]
  • 94.Phan GQ, Yang JC, Sherry RM, Hwu P, Topalian SL, Schwartzentruber DJ, Restifo NP, Haworth LR, Seipp CA, Freezer LJ, Morton KE, Mavroukakis SA, Duray PH, Steinberg SM, Allison JP, Davis TA, Rosenberg SA. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci USA. 2003;100:8372–8377. doi: 10.1073/pnas.1533209100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Poon RT, Fan ST, Wong J. Clinical implications of circulating angiogenic factors in cancer patients. J Clin Oncol. 2001;19:1207–1225. doi: 10.1200/JCO.2001.19.4.1207. [DOI] [PubMed] [Google Scholar]
  • 96.Prehn RT, Lappe MA. An immunostimulation theory of tumor development. Transplant Rev. 1971;7:26–54. doi: 10.1111/j.1600-065x.1971.tb00462.x. [DOI] [PubMed] [Google Scholar]
  • 97.Pure E, Allison JP, Schreiber RD. Breaking down the barriers to cancer immunotherapy. Nat Immunol. 2005;6:1207–1210. doi: 10.1038/ni1205-1207. [DOI] [PubMed] [Google Scholar]
  • 98.Quezada SA, Peggs KS, Curran MA, Allison JP. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J Clin Invest. 2006;116:1935–1945. doi: 10.1172/JCI27745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Rabinovich GA, Baum LG, Tinari N, Paganelli R, Natoli C, Liu FT, Iacobelli S. Galectins and their ligands: amplifiers, silencers or tuners of the inflammatory response? Trends Immunol. 2002;23:313–320. doi: 10.1016/s1471-4906(02)02232-9. [DOI] [PubMed] [Google Scholar]
  • 100.Rabinovich GA, Cumashi A, Bianco GA, Ciavardelli D, Iurisci I, D’Egidio M, Piccolo E, Tinari N, Nifantiev N, Iacobelli S. Synthetic lactulose amines: novel class of anticancer agents that induce tumor-cell apoptosis and inhibit galectin-mediated homotypic cell aggregation and endothelial cell morphogenesis. Glycobiology. 2006;16:210–220. doi: 10.1093/glycob/cwj056. [DOI] [PubMed] [Google Scholar]
  • 101.Rabinovich GA, Daly G, Dreja H, Tailor H, Riera CM, Hirabayashi J, Chernajovsky Y. Recombinant galectin-1 and its genetic delivery suppress collagen-induced arthritis via T cell apoptosis. J Exp Med. 1999;190:385–398. doi: 10.1084/jem.190.3.385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol. 2007;25:267–296. doi: 10.1146/annurev.immunol.25.022106.141609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Rabinovich GA, Rubinstein N, Matar P, Rozados V, Gervasoni S, Scharovsky GO. The antimetastatic effect of a single low dose of cyclophosphamide involves modulation of galectin-1 and Bcl-2 expression. Cancer Immunol Immunother. 2002;50:597–603. doi: 10.1007/s00262-001-0238-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Reed JC. The Survivin saga goes in vivo. J Clin Invest. 2001;108:965–969. doi: 10.1172/JCI14123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Rico MJ, Matar P, Gervasoni SI, Bonfil RD, Calcaterra N, Scharovsky OG. The transition to the metastatic phenotype of rat lymphoma cells involves up-regulation of IL-10 receptor expression and IL-10 secretion. Clin Exp Metastasis. 2005;22:127–135. doi: 10.1007/s10585-005-5140-4. [DOI] [PubMed] [Google Scholar]
  • 106.Ritter G, Livingston PO. Ganglioside antigens expressed by human cancer cells. Semin Cancer Biol. 1991;2:401–409. [PubMed] [Google Scholar]
  • 107.Rivoltini L, Carrabba M, Huber V, Castelli C, Novellino L, Dalerba P, Mortarini R, Arancia G, Anichini A, Fais S, Parmiani G. Immunity to cancer: attack and escape in T lymphocyte-tumor cell interaction. Immunol Rev. 2002;188:97–113. doi: 10.1034/j.1600-065x.2002.18809.x. [DOI] [PubMed] [Google Scholar]
  • 108.Rodgers JR, Cook RG. MHC class Ib molecules bridge innate and acquired immunity. Nat Rev Immunol. 2005;5:459–471. doi: 10.1038/nri1635. [DOI] [PubMed] [Google Scholar]
  • 109.Rodriguez PC, Ochoa AC. T cell dysfunction in cancer: role of myeloid cells and tumor cells regulating amino acid availability and oxidative stress. Semin Cancer Biol. 2006;16:66–72. doi: 10.1016/j.semcancer.2005.10.001. [DOI] [PubMed] [Google Scholar]
  • 110.Rodriguez T, Mendez R, Roberts CH, Ruiz-Cabello F, Dodi IA, Lopez Nevot MA, Paco L, Maleno I, Marsh SG, Pawelec G, Garrido F. High frequency of homozygosity of the HLA region in melanoma cell lines reveals a pattern compatible with extensive loss of heterozygosity. Cancer Immunol Immunother. 2005;54:141–148. doi: 10.1007/s00262-004-0561-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Romero JM, Jimenez P, Cabrera T, Cozar JM, Pedrinaci S, Tallada M, Garrido F, Ruiz-Cabello F. Coordinated downregulation of the antigen presentation machinery and HLA class I/beta2-microglobulin complex is responsible for HLA-ABC loss in bladder cancer. Int J Cancer. 2005;113:605–610. doi: 10.1002/ijc.20499. [DOI] [PubMed] [Google Scholar]
  • 112.Roncarolo MG, Bacchetta R, Bordignon C, Narula S, Levings MK. Type 1 T regulatory cells. Immunol Rev. 2001;182:68–79. doi: 10.1034/j.1600-065x.2001.1820105.x. [DOI] [PubMed] [Google Scholar]
  • 113.Rosenberg SA. Progress in human tumour immunology and immunotherapy. Nature. 2001;411:380–384. doi: 10.1038/35077246. [DOI] [PubMed] [Google Scholar]
  • 114.Rouas-Freiss N, Bruel S, Menier C, Marcou C, Moreau P, Carosella ED. Switch of HLA-G alternative splicing in a melanoma cell line causes loss of HLA-G1 expression and sensitivity to NK lysis. Int J Cancer. 2005;117:114–122. doi: 10.1002/ijc.21151. [DOI] [PubMed] [Google Scholar]
  • 115.Rubinstein N, Alvarez M, Zwirner NW, Toscano MA, Ilarregui JM, Bravo A, Mordoh J, Fainboim L, Podhajcer OL, Rabinovich GA. Targeted inhibition of galectin-1 gene expression in tumor cells results in heightened T cell-mediated rejection; a potential mechanism of tumor-immune privilege. Cancer Cell. 2004;5:241–251. doi: 10.1016/s1535-6108(04)00024-8. [DOI] [PubMed] [Google Scholar]
  • 116.Ryan AE, Shanahan F, O’Connell J, Houston AM. Addressing the “Fas counterattack” controversy: blocking fas ligand expression suppresses tumor immune evasion of colon cancer in vivo. Cancer Res. 2005;65:9817–9823. doi: 10.1158/0008-5472.CAN-05-1462. [DOI] [PubMed] [Google Scholar]
  • 117.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155:1151–1164. [PubMed] [Google Scholar]
  • 118.Sanchez-Perez L, Kottke T, Diaz RM, Ahmed A, Thompson J, Chong H, Melcher A, Holmen S, Daniels G, Vile RG. Potent selection of antigen loss variants of B16 melanoma following inflammatory killing of melanocytes in vivo. Cancer Res. 2005;65:2009–2017. doi: 10.1158/0008-5472.CAN-04-3216. [DOI] [PubMed] [Google Scholar]
  • 119.Scaffidi C, Kirchhoff S, Krammer PH, Peter ME. Apoptosis signaling in lymphocytes. Curr Opin Immunol. 1999;11:277–285. doi: 10.1016/s0952-7915(99)80045-4. [DOI] [PubMed] [Google Scholar]
  • 120.Shimizu J, Yamazaki S, Sakaguchi S. Induction of tumor immunity by removing CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity. J Immunol. 1999;163:5211–5218. [PubMed] [Google Scholar]
  • 121.Siegel CT, Schreiber K, Meredith SC, Beck-Engeser GB, Lancki DW, Lazarski CA, Fu YX, Rowley DA, Schreiber H. Enhanced growth of primary tumors in cancer-prone mice after immunization against the mutant region of an inherited oncoprotein. J Exp Med. 2000;191:1945–1956. doi: 10.1084/jem.191.11.1945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Sorice M, Parolini I, Sansolini T, Garofalo T, Dolo V, Sargiacomo M, Tai T, Peschle C, Torrisi MR, Pavan A. Evidence for the existence of ganglioside-enriched plasma membrane domains in human peripheral lymphocytes. J Lipid Res. 1997;38:969–980. [PubMed] [Google Scholar]
  • 123.Sorme P, Qian Y, Nyholm PG, Leffler H, Nilsson UJ. Low micromolar inhibitors of galectin-3 based on 3′-derivatization of N-acetyllactosamine. Chembiochem. 2002;3:183–189. doi: 10.1002/1439-7633(20020301)3:2/3<183::aid-cbic183>3.0.co;2-#. [DOI] [PubMed] [Google Scholar]
  • 124.Spiotto MT, Rowley DA, Schreiber H. Bystander elimination of antigen loss variants in established tumors. Nat Med. 2004;10:294–8. doi: 10.1038/nm999. [DOI] [PubMed] [Google Scholar]
  • 125.Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol. 2003;21:685–711. doi: 10.1146/annurev.immunol.21.120601.141040. [DOI] [PubMed] [Google Scholar]
  • 126.Stillman BN, Hsu DK, Pang M, Brewer CF, Johnson P, Liu FT, Baum LG. Galectin-3 and galectin-1 bind distinct cell surface glycoprotein receptors to induce T cell death. J Immunol. 2006;176:778–789. doi: 10.4049/jimmunol.176.2.778. [DOI] [PubMed] [Google Scholar]
  • 127.Strater J, Walczak H, Hasel C, Melzner I, Leithauser F, Moller P. CD95 ligand (CD95L) immunohistochemistry: a critical study on 12 antibodies. Cell Death Differ. 2001;8:273–278. doi: 10.1038/sj.cdd.4400813. [DOI] [PubMed] [Google Scholar]
  • 128.Sturm A, Lensch M, Andre S, Kaltner H, Wiedenmann B, Rosewicz S, Dignass AU, Gabius HJ. Human galectin-2: novel inducer of T cell apoptosis with distinct profile of caspase activation. J Immunol. 2004;173:3825–3837. doi: 10.4049/jimmunol.173.6.3825. [DOI] [PubMed] [Google Scholar]
  • 129.Tanaka M, Suda T, Takahashi T, Nagata S. Expression of the functional soluble form of human fas ligand in activated lymphocytes. Embo J. 1995;14:1129–1135. doi: 10.1002/j.1460-2075.1995.tb07096.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Terabe M, Matsui S, Park JM, Mamura M, Noben-Trauth N, Donaldson DD, Chen W, Wahl SM, Ledbetter S, Pratt B, Letterio JJ, Paul WE, Berzofsky JA. Transforming growth factor-beta production and myeloid cells are an effector mechanism through which CD1d-restricted T cells block cytotoxic T lymphocyte-mediated tumor immunosurveillance: abrogation prevents tumor recurrence. J Exp Med. 2003;198:1741–1752. doi: 10.1084/jem.20022227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Thomas L. On immunosurveillance in human cancer. Yale J Biol Med. 1982;55:329–333. [PMC free article] [PubMed] [Google Scholar]
  • 132.Uyttenhove C, Pilotte L, Theate I, Stroobant V, Colau D, Parmentier N, Boon T, Van den Eynde BJ. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med. 2003;9:1269–1274. doi: 10.1038/nm934. [DOI] [PubMed] [Google Scholar]
  • 133.Uzzo RG, Rayman P, Kolenko V, Clark PE, Cathcart MK, Bloom T, Novick AC, Bukowski RM, Hamilton T, Finke JH. Renal cell carcinoma-derived gangliosides suppress nuclear factor-kappaB activation in T cells. J Clin Invest. 1999;104:769–776. doi: 10.1172/JCI6775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.van der Leij J, van den Berg A, Harms G, Eschbach H, Vos H, Zwiers P, van Weeghel R, Groen H, Poppema S, Visser L. Strongly enhanced IL-10 production using stable galectin-1 homodimers. Mol Immunol. 2007;44:506–513. doi: 10.1016/j.molimm.2006.02.011. [DOI] [PubMed] [Google Scholar]
  • 135.van Elsas A, Hurwitz AA, Allison JP. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J Exp Med. 1999;190:355–366. doi: 10.1084/jem.190.3.355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Wang T, Niu G, Kortylewski M, Burdelya L, Shain K, Zhang S, Bhattacharya R, Gabrilovich D, Heller R, Coppola D, Dalton W, Jove R, Pardoll D, Yu H. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat Med. 2004;10:48–54. doi: 10.1038/nm976. [DOI] [PubMed] [Google Scholar]
  • 137.Weller M, Malipiero U, Aguzzi A, Reed JC, Fontana A. Protooncogene bcl-2 gene transfer abrogates Fas/APO-1 antibody-mediated apoptosis of human malignant glioma cells and confers resistance to chemotherapeutic drugs and therapeutic irradiation. J Clin Invest. 1995;95:2633–2643. doi: 10.1172/JCI117965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Whiteside TL. Signaling defects in T lymphocytes of patients with malignancy. Cancer Immunol Immunother. 1999;48:346–352. doi: 10.1007/s002620050585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Whiteside TL, Herberman RB. The role of natural killer cells in immune surveillance of cancer. Curr Opin Immunol. 1995;7:704–710. doi: 10.1016/0952-7915(95)80080-8. [DOI] [PubMed] [Google Scholar]
  • 140.Wieckowski E, Wang GQ, Gastman BR, Goldstein LA, Rabinowich H. Granzyme B-mediated degradation of T-cell receptor zeta chain. Cancer Res. 2002;62:4884–4889. [PubMed] [Google Scholar]
  • 141.Wischhusen J, Friese MA, Mittelbronn M, Meyermann R, Weller M. HLA-E protects glioma cells from NKG2D-mediated immune responses in vitro: implications for immune escape in vivo. J Neuropathol Exp Neurol. 2005;64:523–528. doi: 10.1093/jnen/64.6.523. [DOI] [PubMed] [Google Scholar]
  • 142.Yang RY, Hsu DK, Liu FT. Expression of galectin-3 modulates T-cell growth and apoptosis. Proc Natl Acad Sci USA. 1996;93:6737–6742. doi: 10.1073/pnas.93.13.6737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Zacarias-Fluck MF, Rico MJ, Gervasoni SI, Ilarregui JM, Toscano MA, Rabinovich GA, Scharovsky GO. Low dose cyclophosphamide modulates galectin-1 expression and function in an experimental rat lymphoma model. Cancer Immunol Immunother. 2007;56:237–248. doi: 10.1007/s00262-006-0176-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Zha Y, Blank C, Gajewski TF. Negative regulation of T-cell function by PD-1. Crit Rev Immunol. 2004;24:229–237. doi: 10.1615/critrevimmunol.v24.i4.10. [DOI] [PubMed] [Google Scholar]
  • 145.Zhang X, Huang H, Yuan J, Sun D, Hou WS, Gordon J, Xiang J. CD4–8- dendritic cells prime CD4+ T regulatory 1 cells to suppress antitumor immunity. J Immunol. 2005;175:2931–2937. doi: 10.4049/jimmunol.175.5.2931. [DOI] [PubMed] [Google Scholar]
  • 146.Zou W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer. 2005;5:263–274. doi: 10.1038/nrc1586. [DOI] [PubMed] [Google Scholar]
  • 147.Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol. 2006;6:295–307. doi: 10.1038/nri1806. [DOI] [PubMed] [Google Scholar]

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

RESOURCES