Tumors evolve mechanisms to escape immune control by a process called immune editing, which provides a selective pressure in the tumor microenvironment that could lead to malignant progression. A variety of tumor-derived factors contribute to the emergence of complex local and regional immunosuppressive networks, including vascular endothelial growth factor, interleukin-10, transforming growth factor-β, prostaglandin E2, and soluble phosphatidylserine, soluble Fas, soluble Fas ligand, and soluble MHC class I–related chain A proteins. Although deposited at the primary tumor site, these secreted factors could extend immunosuppressive effects into the local lymph nodes and the spleen, promoting invasion and metastasis. Vascular endothelial growth factors play a key role in recruiting immature myeloid cells from the bone marrow to enrich the microenvironment as tumor-associated immature dendritic cells and tumor-associated macrophages. The understanding of the immunosuppressive networks that evolve is incomplete, but several features are emerging. Accumulation of tumor-associated immature dendritic cells may cause roving dendritic cells and T cells to become suppressed by the activation of indoleamine 2,3-dioxygenase and arginase I by tumor-derived growth factors. Soluble phosphatidylserines support tumor-associated macrophages by stimulating the release of anti-inflammatory mediators that block antitumor immune responses. Soluble Fas, soluble FasL, and soluble MHC class I–related chain A proteins may help tumor cells escape cytolysis by cytotoxic T cells and natural killer cells, possibly by counterattacking immune cells and causing their death. In summary, tumor-derived factors drive the evolution of an immunosuppressive network which ultimately extends immune evasion from the primary tumor site to peripheral sites in patients with cancer. (Cancer Res 2006; 66(11): 5527-36)

The host immune system is regulated by innate and adaptive immune responses to protect it from the attack of foreign entities, such as bacteria and viruses. Cancer cells can grow by escaping from the attack of immune cells, thus, disrupting the host immune system, which is progressively suppressed as a result of tumor progression and metastasis (1, 2). The molecular mechanisms by which cancer cells evade the host immune system have been investigated in mouse models and clinical samples. Several important mechanisms have been documented, such as those involving loss of tumor antigen (3), alteration of HLA class I antigen (4), defective death receptor signaling (5), lack of costimulation (6), immunosuppressive cytokines (7), and immunosuppressive T cells (8). The immunosuppressive cytokines, such as interleukin (IL)-10 and transforming growth factor-β (TGF-β), are secreted from tumor cells and inhibit the maturation of dendritic cells (DC) and T cell function through regulatory T cells (Treg; ref. 9). In addition to these tumor-derived cytokines, the small molecule, prostaglandin E2 (PGE2) inhibits T cell function as well as IL-10 and TGF-β (10).

Another important tumor-derived soluble factor (TDSF) is the vascular endothelial growth factor (VEGF), which acts as a chemoattractant for the production of immature myeloid cells (iMC) from the bone marrow, which are then recruited to the primary tumor site through the peripheral circulation in a mouse model (11). The iMCs produce immature dendritic cells (iDC) and macrophages, which are modulated by TDSFs in the tumor microenvironment to produce tumor-associated iDCs (TiDC) and tumor-associated macrophages (TAM). The activities of indoleamine 2,3-dioxygenase and arginase I in TiDCs and TAMs are increased by TDSFs, such as IL-10 and TGF-β, to produce reactive oxygen species (ROS; refs. 12, 13); thereby, maturation of DCs and T cell proliferation are inhibited, which leads to apoptosis. The accumulation of TiDCs in the primary tumor site could be extended to the secondary lymphoid organs, such as regional lymph nodes and spleen—and the cells might possibly recirculate to the peripheral vessels—thereby providing cancer cells with an immunosuppressive network that extends the immune evasion and promotes tumor progression and metastasis (14).

Other TDSFs, such as soluble Fas ligand (sFasL) and soluble MHC class I–related chain A gene (sMICA) products, also play important roles in immune evasion, which inhibit Fas- and NKG2D-mediated killing of immune cells (15, 16). Another TDSF, soluble phosphatidylserine (sPS), acts as an inducer of an anti-inflammatory response to TAMs, resulting in the release of anti-inflammatory mediators, such as IL-10, TGF-β, and PGE2, that inhibit an immune response to DCs and T cells (17). The altered tumor surface antigen, such as FasL, also causes immune evasion by counterattacking immune cells, leading to cell death (18). In addition, the soluble forms of FasL and MICA, sFasL and sMICA, are able to inhibit Fas and the NKG2D-mediated death of immune cells (19, 20). Thus, it is likely that TDSFs play pivotal roles in constituting immunosuppressive networks that aid tumor progression and metastasis. Indeed, the immunosuppressive networks derived from these TDSFs can be a critical factor in causing the unsatisfactory clinical response that is usually seen in the immunotherapy of advanced cancer, and they remain an important obstacle to be overcome in the interaction between tumors and the immune system in the tumor microenvironment (1, 21). This article discusses the role of TDSFs in establishing the immunosuppressive networks that extend from the primary tumor site to the peripheral vessels, and how the immunosuppressive networks are critical obstacles to immunotherapy in patients with cancer. A better and more comprehensive understanding of the evolution of the immunosuppressive networks derived from tumor cells may provide new insights that may help break the immune suppression by cancer cells.

Since the connection between inflammation caused by leukocytes in cancer tissue and progression of cancer was noted by Virchow in 1863 (22), studies of the inflammatory microenvironment of cancer tissues have helped to reveal the links between cancer and inflammation (23). Leukocytes infiltrate cancer tissues, particularly in the hypoxic tumor lesion, and the monocyte-macrophage lineage is a major component of the leukocyte infiltrate of tumors. Some cancers, such as liver, gastrointestinal, and gynecologic malignancies, in which the inflammatory process is a cofactor in carcinogenesis, are attributable to infections in these organs (24). Inflammatory mediators, including reactive oxygen and nitrogen intermediates, prostaglandins, and inflammatory cytokines, contribute to tumor development by inducing protumor mutations, resistance to apoptosis, and environmental changes, such as stimulation of angiogenesis, that confer a survival advantage to a susceptible cell.

Despite the fact that the progression and metastasis of cancer cells are mediated by proinflammatory responses in the tumor microenvironment, the immune response is regulated in anti-inflammatory situations by TDSFs such as sPS. Phosphatidylserine, an ATP-dependent aminophospholipid, triggers the phagocytosis of apoptotic cells by phagocytes such as macrophages and DCs. The clearance of apoptotic cells is mainly mediated by macrophages influenced by the interaction between phosphatidylserine and phosphatidylserine receptor. Phagocytosis of apoptotic cells by macrophages occurs without the induction of inflammatory reactions (25, 26). The presence of apoptotic cells during macrophage activation has been shown to increase the secretion of anti-inflammatory mediators, such as IL-10, TGF-β, and PGE2, and to decrease the secretion of proinflammatory cytokines such as IL-12 (27). Although phagocytosis of both apoptotic and necrotic cells is mediated by the phosphatidylserine/phosphatidylserine receptor interaction, the reduced cytotoxicity of TAMs after ingestion of apoptotic cells may be explained by the anti-inflammatory response to cancer cells that was caused by several negative mediators. The sequential immune responses by TAMs and DCs can be abrogated by sPS derived from cancer cells. In the sequential interactions, the tumor-derived sPS reacts with phosphatidylserine receptor in TAMs and DCs to inhibit the innate immune response. The enforced anti-inflammatory response by sPS—not only by interaction with the phosphatidylserine receptor on TAMs, but also with the phosphatidylserine receptor on iDCs to inhibit their maturation—may cause immune escape and tumor proliferation. Furthermore, the tumor-derived sPS bind to their own phosphatidylserine receptor, resulting in the production of IL-10, TGF-β, and PGE2 to facilitate tumor proliferation.

During the anti-inflammatory response, the TGF-β derived from TAMs prevents the maturation of antigen-presenting DCs, thereby inhibiting the activation of natural killer (NK) cells and CTLs (28). In addition, TGF-β promotes the proliferation of stromal cells, such as fibroblasts and macrophages, and induces them to secrete angiogenic and cell survival factors that stimulate tumor proliferation, including cyclooxygenase-2 and VEGF through nuclear factor κB (29). These stromal cells also produce Th2 cytokines, such as IL-4 and IL-10, as well as PGE2 as a result of TGF-β secretion. The increased PGE2 concentration decreases the expression of MHC class I and II antigens in DCs (30). Under tumor-specific conditions, such as hypoxia and glucose starvation, macrophages produce hydroxide as well as TGF-β, and subsequently the oxidized macrophages, TAMs, also produce Th2 cytokines such as IL-4 and IL-10, which might be involved in immune escape (31). The increased levels of the cytokines, IL-4, IL-10, PGE2, and TGF-β enhance the expressions of c-FLIP, Bcl-xL, and Mcl-1, which are involved in the resistance to apoptosis (3234). Thus, the production of IL-10, TGF-β, and PGE2 from TAMs—mediated by sPS or hydroxide—plays a critical role in immune escape. Because immune cells are suppressed during the anti-inflammatory response, the defective removal of apoptotic cells promotes an inflammatory response to cancer cells. In conjunction with the secretion of the cytokines, IL-10 and TGF-β, in the tumor microenvironment, cancer cells promote their own growth by activating antiapoptotic pathways concomitant with tumor angiogenesis activated by matrix metalloproteinase 2 (MMP-2) and MMP-9 (35, 36).

VEGF is an important growth factor for endothelial cells in tumor angiogenesis, which is activated by hypoxia-inducible factor-1 in the tumor microenvironment (37). In addition, VEGF plays a critical role as a chemoattractant in the induction of iMC from bone marrow (38). The induced iMCs, such as iDC and macrophages, flow into peripheral vessels, and they are recruited to primary tumor sites. These cells are either committed to apoptosis or modulated by TDSFs to become resistant to apoptosis in the tumor site with decreased Fas and increased Bcl-2 expression, which are attributed to TiDCs and TAMs (39). These tumor-associated iMCs are able to distribute to secondary lymphoid organs such as regional lymph nodes (e.g., sentinel lymph nodes) and spleen, where they are recirculated to peripheral vessels. In fact, the expansion of iMCs with immunosuppressive cell characteristics has been reported in patients with head and neck cancer (40), gastric cancer (41), and colon cancer (42). In the mouse tumor model, the iMCs are characterized by the expression of Gr-1 and CD11b markers, including iDCs, macrophages, and granulocytes. These cells suppress antigen-specific CD8+ T cells via direct cell-cell contact, mediated by ROS (43). Accumulation of H2O2 in these cells—in which arginase (Arg) activity plays an important role—was a major contributor to this increased pool of ROS. The iMCs could be induced by several TDSFs, such as IL-3, SDF-1, and VEGF from bone marrow, and then recruited to tumor sites through the peripheral circulation (44, 45). In particular, the TiDCs are resistant to apoptosis and show increased arginase I activity. Arginase I is an enzyme that converts the amino acid l-arginine to ornithine and urea, which inhibits T cell proliferation and functions in coordination with TGF-β. Furthermore, l-arginine is metabolized to nitric oxide and hydroxide by inducible nitric oxide synthase; the metabolites inhibit Th1 cytokine production and T cell function, and decrease expression of the T cell receptor-ζ (TCR-ζ) that leads to T cell apoptosis (46). Thus, VEGF-induced iMCs play a critical role in inhibiting innate and adaptive immune responses in cancer cells. The induction of iMCs by tumor-derived chemoattractants is constitutive, whereas the induction of iMCs by bacterial infection and potent immunogens is transient and declined after several hours (47). In this regard, VEGF inhibited the differentiation of thymus precursors, resulting in thymus atrophy (48). It is likely that the constitutive induction of iMCs, their recruitment to the tumor site, and their functional modulation by TDSFs are critically involved in the evolution of the immunosuppressive network in patients with cancer.

NF-κB is a transcription factor that plays an important role in multiple cellular functions such as an immune response associated with an inflammatory response (49). Although NF-κB activation is required for the maturation of DCs and activation of T cells in an immune response, the activation of signal transducers and activator of transcription 3 in Gr-1 CD11b+ cells inhibits NF-κB activation as well as VEGF and PGE2, whereby the maturation of DCs is blocked (50). In addition, the TAMs, freshly isolated from spleens of tumor-bearing or naïve mice, were able to inhibit the T cell–mediated immune response in vitro via induction of T cell apoptosis, which was responsible for signal transducer and activator of transcription 1 activation. Arginase and nitric oxide were both responsible for the apoptotic mechanism, and were present in TAMs (13). Of interest, coincubation of CD8+ T cells with Gr-1 CD11b+ cells immediately induced apoptosis of the T cells, whereas CD4+ cells were less sensitive to Gr-1 CD11b+-mediated apoptosis (51). One reason for the increased sensitivity of CD8+ T cells to Gr-1 CD11b+-mediated apoptosis may be explained by the observation that iMCs express MHC class I but very little or no MHC class II antigens. The population of iMCs is a heterogeneous mixture of precursors of different myeloid cells and early progenitors, therefore, whether the inhibitory effect of iMCs is limited to CD8+ T cells or is extended to CD4+ T cells may depend on the expression of MHC antigens on the iMC surface. In fact, the increased sensitivity of CD8+ T cells to apoptosis—which is due to the elevated Bax/Bcl-2 ratio on the T cells, Fas/Fas-ligand interactions, and high levels of FasL expression on the tumor—have been observed in patients with head and neck cancer (52), breast cancer (53), and oral cancer (54). Furthermore, microvesicle-associated FasL-induced decreased expression of TCR-ζ, which is a substrate of caspase-3, leads to altered signal transduction in T cells and apoptosis (55). The altered T cell dysfunction was attributed to the induction of apoptosis due to ROS and hydroxide derived from l-arginine because T cells stimulated and cultured in the absence of l-arginine present a sustained down-regulation of TCR-ζ, exhibit a decreased rate of proliferation, and a significantly diminished production of IFN-γ (46). Although inhibition by iDCs of T cell function is mediated by cell-cell contact, iDCs produce anti-inflammatory cytokines, such as IL-10 and TGF-β, that lead to an increase in arginase I activity, resulting in the production of ROS, which is involved in the inhibition of T cell function (43). The functional roles of TDSFs and iMCs are summarized in Fig. 1.

Figure 1.

Overview of immunosuppressive networks supported by tumor-derived factors. The TDSFs, such as VEGF and SDF-1, act as chemoattractants that recruit iMCs from bone marrow into peripheral vessels, from where they are then recruited to the primary tumor site. Accumulated iMCs are functionally and biochemically modulated to become TiDCs and TAMs, resulting in the suppression of the maturation of DCs and T cells, thereby helping to promote the immune evasion. Immature DCs induce immune tolerance by either deleting T cells or inducing Tregs. The TiDCs and TAMs are distributed to secondary lymphoid organs such as sentinel lymph nodes and spleen, and possibly recirculated to the peripheral vessels, which leads to the formation of the immunosuppressive network for tumor progression and metastasis. TAMs, isolated from the spleens of tumor-bearing hosts, inhibit the T cell–mediated immune response via induction of T cell apoptosis. The detailed explanation of this process is provided in the text. Summary: TDSFs, such as VEGF and SDF-1, lead to the immunosuppressive network for tumor progression and metastasis by recruiting into the tumor site iMCs that are modulated for immune evasion.

Figure 1.

Overview of immunosuppressive networks supported by tumor-derived factors. The TDSFs, such as VEGF and SDF-1, act as chemoattractants that recruit iMCs from bone marrow into peripheral vessels, from where they are then recruited to the primary tumor site. Accumulated iMCs are functionally and biochemically modulated to become TiDCs and TAMs, resulting in the suppression of the maturation of DCs and T cells, thereby helping to promote the immune evasion. Immature DCs induce immune tolerance by either deleting T cells or inducing Tregs. The TiDCs and TAMs are distributed to secondary lymphoid organs such as sentinel lymph nodes and spleen, and possibly recirculated to the peripheral vessels, which leads to the formation of the immunosuppressive network for tumor progression and metastasis. TAMs, isolated from the spleens of tumor-bearing hosts, inhibit the T cell–mediated immune response via induction of T cell apoptosis. The detailed explanation of this process is provided in the text. Summary: TDSFs, such as VEGF and SDF-1, lead to the immunosuppressive network for tumor progression and metastasis by recruiting into the tumor site iMCs that are modulated for immune evasion.

Close modal

The accumulation of iDCs is found not only at the primary tumor site but could also be extended to the secondary lymphoid organs and peripheral vessels, which facilitates constitution of the immunosuppressive network that supports tumor growth in patients with cancer. In fact, increased numbers of iDCs have been shown in lymph nodes and peripheral blood in patients with head and neck, breast, and non–small cell lung cancers according to their tumor progression, and these iDCs are able to inhibit DC maturation and T cell function (56, 57). The TDSF-induced new iDCs from bone marrow are recruited to the primary site and are educated by biochemical modulation to show, for example, resistance to apoptosis and increased activity of arginase I. The TiDCs may be different from tumor-associated DCs because the latter are committed to apoptosis. However, some TiDCs are converted to mature tumor-associated DCs, and some of them may be able to activate T cells as antigen-presenting cells. In contrast, TiDCs are able to exert immunosuppressive activity that affects not only the primary tumor site but also secondary lymphoid organs, including regional lymph nodes and spleen, and extends to peripheral vessels. This immunosuppressive network is associated with tumor progression and affects the poor prognosis for patients with advanced cancer. The evolution of immunosuppressive activity in cancer patients is also revealed in the sentinel lymph nodes of patients with melanoma (58) and breast cancers (59, 60). A high degree of down-regulation of TCR-ζ is observed in the sentinel lymph nodes of patients with breast cancer (61). Furthermore, in our preliminary study we observed that expression of the costimulatory molecules, such as CD80 and CD86 in DCs, was down-regulated in the sentinel lymph nodes in breast cancer patients, and the expression of CD83 was also decreased, indicating that the increased iDCs in the sentinel lymph nodes might play a critical role in an evolving immune evasion. In addition, given that increased levels of IL-10 and decreased levels of IL-12 were observed in the sentinel lymph nodes, the Th2-dominant situation also contributes to immune evasion in the sentinel lymph nodes.4

4

Unpublished results.

The spleen is a secondary lymphoid organ that can be involved in an antitumor immune response. In the second half of the 1970s, the immune suppressor cells, including DCs and T cells, were first described in the mouse tumor model (62, 63). The number of these suppressor cells in the spleen increases together with tumor growth, and they are now identified as plasmacytoid DCs (pDC; ref. 64) and CD4+ CD25+ Tregs (65). Some TiDCs can be converted to pDCs, the pDCs are being recruited to tumor sites, mediated by CXCR4/CXCL12 (SDF-1) interaction between pDCs and tumor cells. Tumor-derived CXCL12 mediates the trafficking of pDCs that express CXCR4 to the tumor (45). pDCs originate in iMCs in the bone marrow; iMCs are induced by TDSFs, such as VEGF, that are recruited to tumor sites through interaction with chemokines and chemokine receptors, such as the CXCR4/CXCL12 interaction. CXCL12 protects tumor pDCs (TpDC) from apoptosis, these cells can be phenotypically and functionally modulated in the tumor microenvironment (45). Functional pDCs are found in the local tumor microenvironment of patients with ovarian cancer (45), melanoma (66), and head and neck squamous cell carcinoma (67). TpDCs induce significant production of IL-10 by T cells that suppress myeloid DC (mDC)-induced tumor antigen–specific T cell effector functions. Despite the fact that activated peripheral blood pDCs stimulate tumor-specific T cell and IFN-γ production, TpDCs show reduced expression of toll-like receptor 9 (TLR9) for inducing IFN-α. CXCR4-CXCL12 signals are implicated in the trafficking of pDCs into tumors, and they are further involved in tumor vascularization and metastasis by evoking immune evasion through functional modulation of the pDCs.

CD4+ CD25+ Tregs play a crucial role in regulating the immune response and in maintaining immune homeostasis (68). Naturally occurring Tregs represent a subset of Tregs that suppresses immune responses through a cell-cell contact mechanism without specific antigen stimulation (68). In comparison, antigen-induced Tregs mediate immune suppression either through cell-cell contact or through secretion of soluble factors such as IL-10 and TGF-β (69). The function of Tregs, which are involved in establishing immunologic tolerance in cancer immunosuppression, has been reported to be the inhibition of CD8+ and CD4+ T cells. Several studies have reported increased frequencies of CD4+ CD25+ Tregs in blood, malignant effusions, draining lymph nodes, and tumor tissues, thus, associating an impaired immune response to cancer with a high frequency and/or hyperactivity of Tregs (8, 70). The immunosuppressive cytokines, such as IL-10 and TGF-β, derived from tumor cells, induce Tregs (71), which activate the activity of indoleamine 2,3-dioxygenase for the inhibition of T cell function. Furthermore, immature DCs tend to induce immune tolerance rather than immunity by either deleting reactive T cells or inducing Tregs (72). The functional and molecular characterization of Tregs has been made possible by the recent association of cell markers with immunoregulatory activity—such as the tumor necrosis factor (TNF) family molecule GITR (73), CTL antigen-4 (74), and Foxp3 (71)—which affect both antitumor immunity and autoimmunity.

The cytolytic function of CTLs and NK cells in killing tumor cells is mediated by either the death receptor–dependent pathway or perforin-induced granzyme B, a serine protease, which leads to apoptosis. The Fas-dependent cell death pathway is regulated by the Fas receptor on tumor cells and its ligand FasL on CTLs. The soluble form of Fas, sFas, which is derived from the cleavage of Fas by MMPs in tumor cells, is increased according to tumor progression in patients with breast cancer (75) and other malignancies (76, 77). Tumor-derived sFas can interact with asL on CTLs, whereby Fas-mediated apoptosis is inhibited. In addition, the altered expression of FasL on tumor cells is able to counterattack CTLs, leading to their apoptosis (78). This mechanism may also activate FasL-mediated activation-induced cell death for CTLs (79). The soluble form of FasL, sFasL, which is derived from cleavage of FasL by MMPs, can also counterattack CTLs but to a lesser extent than FasL (80). In fact, tumor cells of various cellular origins were sensitized to Fas-mediated cytotoxicity by MMP inhibitors (81). sFasL is also able to inhibit the interaction of Fas/FasL between tumor cells and CTLs in Fas-mediated apoptosis. Furthermore, internalization of sFasL into Fas in tumor cells down-regulates Fas expression, which contributes to the development of resistance to Fas-mediated apoptosis. Thus, the altered expressions of FasL and the soluble factors, sFas and sFasL, contribute to immune evasion by allowing tumor cells to escape from immune attack by CTLs.

The other effectors, NK cells, play an important role in antitumor immune responses via NKG2D-mediated apoptosis (82). This pathway is regulated by the interaction between the NKG2D receptor on NK and CD8+ T cells, and its ligand, MICA on tumor cells. The soluble form of MICA (sMICA), derived from tumor cells, could inhibit NKG2D-mediated apoptosis by interfering with the NKG2D/MICA interaction between tumor and NK cells (83). Tumor-derived sMICA internalizes with the NKG2D receptor on NK cells, resulting in the down-regulation of the expression of NKG2D, thereby decreasing NK-mediated killing of MICA+ tumor cells (84). Thus, it is likely that not only the soluble death receptors, such as sFas derived from tumor cells, but also the altered expression of death receptor ligands, such as FasL and MICA and their soluble forms, play a role in immune evasion by tumor cells (Fig. 2).

Figure 2.

Possible roles of sFas, sFasL, and sMICA in promoting immune escape. Tumor-derived sFas interacts with FasL on CTL, resulting in the inhibition of Fas-mediated cell death. FasL and sFasL are able to counterattack CTLs, which leads to the apoptosis of CTLs, and sFasL competes with FasL of CTLs for binding to tumor cells. Tumor-derived sMICA internalizes with the NKG2D receptor on NK cells, which leads to its down-regulation and results in the inhibition of NKG2D-mediated cell death. sMICA competes with MICA for binding to NK cells. Summary: TDSFs, sFas, sFasL, and sMICA help tumor cells escape Fas and NKG2D-mediated cell death by competing with FasL of CTLs and MICA of tumor cells.

Figure 2.

Possible roles of sFas, sFasL, and sMICA in promoting immune escape. Tumor-derived sFas interacts with FasL on CTL, resulting in the inhibition of Fas-mediated cell death. FasL and sFasL are able to counterattack CTLs, which leads to the apoptosis of CTLs, and sFasL competes with FasL of CTLs for binding to tumor cells. Tumor-derived sMICA internalizes with the NKG2D receptor on NK cells, which leads to its down-regulation and results in the inhibition of NKG2D-mediated cell death. sMICA competes with MICA for binding to NK cells. Summary: TDSFs, sFas, sFasL, and sMICA help tumor cells escape Fas and NKG2D-mediated cell death by competing with FasL of CTLs and MICA of tumor cells.

Close modal

Given that the evolution of the immunosuppressive network from the primary tumor site plays a critical role in maintaining immunosuppression in cancer patients—mediated by TDSFs and altered tumor surface antigens according to tumor progression—the simplest and best way to eliminate the immunosuppressive network is by removal of the primary tumor and eradication of tumor cells from the cancer patient. However, despite complete surgical resection of a primary tumor with regional lymph node dissection, residual tumor cells may still exist, and these tumor cells would be able to grow and reconstitute the immunosuppressive network during tumor progression. It is conceivable, though, that residual tumor cells after surgical resection may be completely removed by adjuvant therapy such as chemotherapy and immunotherapy. Complete eradication of residual tumor cells by chemotherapy can increase the cure rate of patients with cancer, and activation of the antitumor immune response may provide an increase in cure rate either by complete eradication of tumor cells or prolongation of disease-free survival by controlling the number of residual tumor cells following surgical resection. Unfortunately, the potential contribution to survival benefit of effective immunotherapy as an adjuvant after surgical resection has not yet been established in solid tumors. Nevertheless, immune activation by massive tumor cell death after neoadjuvant chemotherapy may be effective in antagonizing the anti-inflammatory response in immune cells, which disrupts immune tolerance mediated by iMCs. Induction of massive cell death, including apoptotic and nonapoptotic cell death, activates the immune response, which is mediated by the engulfment of dead cells by DCs and increased release of tumor antigens or tumor-associated antigens to secondary lymphoid organs such as sentinel lymph nodes and spleen. Because the sentinel lymph nodes in breast cancer are involved in immune suppression, activation of T cells through cross-priming by mature DCs enables them to attack tumor cells in the primary and other tumor sites. Therefore, activation of the immune response in sentinel lymph nodes may result in not only a tumor response but also prolongation of survival after neoadjuvant chemotherapy. Similarly, the immunosuppressive myeloid cells accumulated in the spleen of patients with advanced gastric cancer may be modulated to provide immune activation after neoadjuvant chemotherapy. Thus, the potential immunostimulation after neoadjuvant chemotherapy for solid tumors, such as breast and gastrointestinal cancer, will be of great interest. The molecular mechanism of the immune activation and its clinical significance will also need to be clarified in future studies.

Over the past two decades, several modalities for immunotherapy, such as administration of cytokines, transfer of T cells and DCs, have been tried in various types of cancer patients, and a recent development of specific cancer vaccines as adoptive immunotherapy has emerged. Although a number of clinical studies have been conducted for various types of cancer, the clinical response rate was only ∼2.6%, hence, a cancer vaccine is not yet a reality (21). The reason for the low response rate may be that the clinical trials were conducted on advanced cases and that most of them had failed previous chemotherapy. In these situations, the immune evasion that originated at the primary tumor site and extended via its suppressive network to secondary lymphoid organs and peripheral vessels could not be disrupted by vaccine treatment alone, even though a novel tumor antigen was used for immune activation. Current clinical trials have shifted to combination therapy with vaccine together with anticancer drugs or radiation. The rationale for using combination treatment is that massive tumor cell death may be expected, followed by immune activation with loading of tumor antigens and tumor-associated antigens. Because a key factor in an immune response is the release and loading of tumor antigens and tumor-associated antigens derived from massive tumor cell death and subsequent activation of DCs, it is likely that massive tumor cell death is critical for generating an effective immune response against the anti-inflammatory response to immune cells. In general, tumor antigens and tumor-associated antigens are down-regulated in stromal cells in the tumor microenvironment because the stromal cells, such as the tumor-associated fibroblasts surrounding tumor cells, increase interstitial fluid pressure (IFP), which inhibits the release of tumor cells and antigens to secondary lymphoid organs. Furthermore, the extracellular matrix binds tumor antigen (85), and fibroblasts and endothelial cells compete with DCs for the antigen (86), whereby many tumor antigens are down-regulated, thus allowing tumor progression (87). A decrease in IFP following massive tumor cell death facilitates the release and loading of tumor antigens, thereby activating immune cells (Fig. 3). Although apoptotic cells are engulfed by DCs, and the tumor-associated antigens are processed for MHC class I antigen cross-presentation to CD8+ T cells, causing their activation (88), necrotic cells are proinflammatory and may be more suitable than apoptotic cells for the activation of T cells (89). In addition, nonapoptotic cell death, autophagy, is attributed to the presentation of MHC class II antigen for the regulation of CD4+ T cells (90). It is unclear which type of cell death is most applicable for the activation of T cells and the generation of an antitumor immune response in a clinical situation.

Figure 3.

Role of IFP in immune evasion. The bone marrow–derived myeloid cells, such as iDCs and TAMs, and non-bone marrow–derived cells, such as fibroblasts, endothelium, and extracellular matrix, increase the IFP. The increased IFP subsequently decreases the tumor antigen level because extracellular matrix binds to antigen, and endothelium/fibroblasts compete with DCs for antigen, which leads to immune ignorance. Massive tumor cell death induced by anticancer treatment decreases IFP, resulting in an increased release of tumor cells and tumor antigens to secondary lymphoid organs. Nevertheless, accumulation of iDCs in sentinel lymph nodes inhibits the maturation of DCs and a tumor-specific T cell response, which leads to immune tolerance. Summary: nontumorous cells increase IFP, which decreases tumor antigen level and causes immune ignorance. Massive tumor cell death decreases IFP and provokes a tumor-specific immune response.

Figure 3.

Role of IFP in immune evasion. The bone marrow–derived myeloid cells, such as iDCs and TAMs, and non-bone marrow–derived cells, such as fibroblasts, endothelium, and extracellular matrix, increase the IFP. The increased IFP subsequently decreases the tumor antigen level because extracellular matrix binds to antigen, and endothelium/fibroblasts compete with DCs for antigen, which leads to immune ignorance. Massive tumor cell death induced by anticancer treatment decreases IFP, resulting in an increased release of tumor cells and tumor antigens to secondary lymphoid organs. Nevertheless, accumulation of iDCs in sentinel lymph nodes inhibits the maturation of DCs and a tumor-specific T cell response, which leads to immune tolerance. Summary: nontumorous cells increase IFP, which decreases tumor antigen level and causes immune ignorance. Massive tumor cell death decreases IFP and provokes a tumor-specific immune response.

Close modal

Given that VEGF plays a dual role—by being involved in tumor angiogenesis and functioning as a chemoattractant inducing iMCs from bone marrow—the blockage of VEGF by anti-VEGF antibody could modulate the immunosuppressive network that is initiated from the primary tumor site (91). Continuous recombinant VEGF infusion resulted in the dramatic inhibition of dendritic cell development, associated with an increase in the production of B cells and immature Gr-1(+) myeloid cells (11). Infusion of VEGF was associated with inhibition of the activity of the transcription factor NF-κB in bone marrow progenitor cells. VEGF itself affected polypotent stem cells, resulting in the observed abnormal hematopoiesis. In contrast, treatment with anti-VEGF antibody decreased the accumulation of iMCs from bone marrow and improved DC function in tumor-bearing hosts (92). However, the inhibition of VEGF is a partial effect because soluble VEGF cannot completely be blocked by anti-VEGF antibody. Nevertheless, the inhibition of VEGF as an inducer of iMCs may be valuable for recovering the immune response, particularly in combination with anticancer drugs. In addition, it should be noted that treatment with anti-VEGF antibody decreases IFP, which may enhance the immunomodulatory action of anti-VEGF antibody in combination treatment (93). Recent reports of clinical trials for colon cancer (94), non–small cell lung cancer (95), and breast cancer (96) revealed the substantial survival benefits of bevacizumab, a humanized anti-VEGF monoclonal antibody, in combination with standard chemotherapy. The reproducible survival benefit of bevacizumab for various types of solid tumors may be explained by the decrease in accumulated TiDCs and TAMs and their functional inhibition, which could facilitate the activation of DCs and T cells following tumor cell death by anticancer drugs.

Because the immune cells in tumor-bearing hosts are regulated as part of the anti-inflammatory response, a proinflammatory response to DCs is necessary for the activation of a tumor-specific immune response. One possible approach is the use of cytosine-phosphate-guanosine (CpG) oligodeoxyribonucleotides (ODN), among which, CpG-DNA was the first one shown to possess an immunostimulatory effect in cancer patients treated with a bacterial lysate (97). This effect is induced by unmethylated CpG dinucleotide, which is a specific base pair sequence that conferred immunostimulatory activity to bacterial DNA and synthetic ODNs containing these same sequences (98). The recognition of the specific CpG motif is mediated by TLR9 as part of an effector mechanism (99). CpG activates macrophages, DCs, B cells, and NK cells, causing increases in various factors such as IL-1, IL-6, IL-12, TNF-α, IFN-α, costimulatory molecules, MHC class II, and IFN-γ for innate immune responses (Fig. 4). CpG 7909, a class B type CpG ODN, which is characterized by a phosphorothioate backbone without poly-G tails, is being tried as an adjuvant in a phase II trial for lung cancer. A recent report indicated that patients with melanoma who received vaccinations of CpG 7909 mixed with a melanoma antigen A analogue peptide, and incomplete Freund's adjuvant, exhibited rapid and strong antigen-specific T cell responses: the frequency of Melan-A-specific T cells increased to >3% of the circulating CD8+ T cells, which was one order of magnitude higher than the frequency seen in control patients treated similarly but without CpG (100). Furthermore, combination treatment with CpG ODNs and anticancer drugs or radiation enhanced the therapeutic effect in an orthotopic tumor model (101, 102). In addition, antisense Bcl-2 (AS Bcl-2, oblimersen sodium), containing two CpG motifs, induced an immunostimulatory response, as shown by splenomegaly and an increase in IL-12 in a mouse model, which may be shifted to a Th1 immune response (103). In fact, a recent study showed an increase in IgM level over time for responders among patients with relapsed multiple myeloma treated with AS Bcl-2 in combination with dexamethasone and thalidomide (104). Although potential survival benefits of AS Bcl-2 in combination with anticancer drugs have been suggested for metastatic melanoma and chronic lymphocytic leukemia, the clinical relevance still remains to be determined. Further clinical studies are being continued for solid tumors and other malignancies to clarify the clinical efficacy of AS Bcl-2 in cancer therapy.

Figure 4.

Toll receptor ligands such as CpG ODNs weaken immunosuppression to improve therapeutic response. Top, anti-inflammatory response to immune cells mediated by macrophages through phosphatidylserine and its receptor (phosphatidylserine/phosphatidylserine receptor) interaction. The dead tumor cells induced by anticancer treatment are removed by phagocytosis by macrophages through phosphatidylserine/phosphatidylserine receptor interaction. Hence, anti-inflammatory mediators, such as IL-10, TGF-β, and PGE2, are released, which causes the constitutive anti-inflammatory response to immune cells. Bottom, effect of CpG ODNs on tumor-specific immune response mediated by TLR9 in pDCs in combination with anticancer treatment. Cotreatment by CpG ODNs and anticancer drugs or radiation activates pDCs through TLR9, which could be shifted to a Th1 immune response. The Th1 response activates iDCs and macrophages for the production of TNF-α, IL-Iβ, and nitric oxide, which provokes a proinflammatory response that connects to a tumor-specific T cell response. Summary: the combination of CpG ODNs and anticancer treatment activates tumor-specific immune response, which is mediated by TLR9 on pDCs and subsequent T cell activation.

Figure 4.

Toll receptor ligands such as CpG ODNs weaken immunosuppression to improve therapeutic response. Top, anti-inflammatory response to immune cells mediated by macrophages through phosphatidylserine and its receptor (phosphatidylserine/phosphatidylserine receptor) interaction. The dead tumor cells induced by anticancer treatment are removed by phagocytosis by macrophages through phosphatidylserine/phosphatidylserine receptor interaction. Hence, anti-inflammatory mediators, such as IL-10, TGF-β, and PGE2, are released, which causes the constitutive anti-inflammatory response to immune cells. Bottom, effect of CpG ODNs on tumor-specific immune response mediated by TLR9 in pDCs in combination with anticancer treatment. Cotreatment by CpG ODNs and anticancer drugs or radiation activates pDCs through TLR9, which could be shifted to a Th1 immune response. The Th1 response activates iDCs and macrophages for the production of TNF-α, IL-Iβ, and nitric oxide, which provokes a proinflammatory response that connects to a tumor-specific T cell response. Summary: the combination of CpG ODNs and anticancer treatment activates tumor-specific immune response, which is mediated by TLR9 on pDCs and subsequent T cell activation.

Close modal

Cancer immune evasion initiated from the primary tumor site, which is mediated by TDSFs and altered tumor surface antigens, is able to extend to secondary lymphoid organs and peripheral vessels, resulting in the formation of an immunosuppressive network that inhibits an antitumor immune response. The increased number of iMCs recruited from the bone marrow, triggered by VEGF and other TDSFs, as well as their TiDCs and TAM, play a critical role in suppressing the immune response at not only the primary site but also at secondary lymphoid organs and peripheral vessels. Given that tumor cells are regulated by a proinflammatory mechanism that stimulates tumor growth, whereas immune cells are regulated by an anti-inflammatory mechanism that causes immune suppression, proinflammatory or anti-inflammatory treatments have been controversial and considered as a double-edged sword in cancer therapy. However, because a proinflammatory response is mediated by massive tumor cell death, and other immunostimulatory agents are crucial for provoking an antitumor immune response, a proinflammatory response does not necessarily promote tumor growth. In contrast, anti-inflammatory treatment with, for example, NF-κB inhibitor may affect the activation of immune cells. Selective targeted therapy for effects downstream of NF-κB may be more effective than nonspecific inhibition of NF-κB. Thus, primary cancer therapy needs to be focused on inducing a proinflammatory response, which can be connected to the activation of an immune response to tumor antigens and tumor-associated antigens in cancer cells.

Many previous reports indicated that tumors infiltrated with lymphocytes or mature DCs had a good prognosis. Furthermore, recent reports on the use of DNA microarrays for gene expression analysis also suggested that an increase in expression of immune response–related genes is associated with a good prognosis (105, 106). Despite the fact that treatment with anticancer drugs and molecular targeting agents induces infiltration of T cells into the tumor, the molecular mechanism by which anticancer treatment activates an immune response against the tumor-induced immunosuppressive network in individual cancer patients still remains to be elucidated. Further studies will be required for the analysis of individual immune responses to tumor cells following anticancer treatment to disrupt and modulate the immunosuppressive network. Such studies may provide new insights and help to overcome a critical obstacle in cancer therapy.

1
Zou W. Immunosuppressive networks in the tumour environment and their therapeutic relevance.
Nat Rev Cancer
2005
;
5
:
263
–74.
2
Whiteside TL. Immunobiology of head and neck cancer.
Cancer Metastasis Rev
2005
;
24
:
95
–105.
3
Jager D, Jager E, Knuth A. Immune responses to tumour antigens: implications for antigen specific immunotherapy of cancer.
J Clin Pathol
2001
;
54
:
669
–74.
4
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
–10.
5
French LE, Tschopp J. Defective death receptor signaling as a cause of tumor immune escape.
Semin Cancer Biol
2002
;
12
:
51
–5.
6
Byrne SN, Halliday GM. High levels of Fas ligand and MHC class II in the absence of CD80 or CD86 expression and a decreased CD4+ T cell Infiltration, enables murine skin tumours to progress.
Cancer Immunol Immunother
2003
;
52
:
396
–402.
7
Toutirais O, Chartier P, Dubois D, et al. Constitutive expression of TGF-β1, interleukin-6 and interleukin-8 by tumor cells as a major component of immune escape in human ovarian carcinoma.
Eur Cytokine Netw
2003
;
14
:
246
–55.
8
Somasundaram R, Jacob L, Swoboda R, et al. Inhibition of cytolytic T lymphocyte proliferation by autologous CD4+/CD25+ regulatory T cells in a colorectal carcinoma patient is mediated by transforming growth factor-β.
Cancer Res
2002
;
62
:
5267
–72.
9
Berger CL, Tigelaar R, Cohen J, et al. Cutaneous T-cell lymphoma: malignant proliferation of T-regulatory cells.
Blood
2005
;
105
:
1640
–7.
10
He X, Stuart JM. Prostaglandin E2 selectively inhibits human CD4+ T cells secreting low amounts of both IL-2 and IL-4.
J Immunol
1999
;
163
:
6173
–9.
11
Gabrilovich D, Ishida T, Oyama T, et al. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo.
Blood
1998
;
92
:
4150
–66.
12
Munn DH, Sharma MD, Lee JR, et al. Potential regulatory function of human dendritic cells expressing indoleamine 2,3-dioxygenase.
Science
2002
;
297
:
1867
–70.
13
Kusmartsev S, Gabrilovich DI. STAT1 signaling regulates tumor-associated macrophage-mediated T cell deletion.
J Immunol
2005
;
174
:
4880
–91.
14
Almand B, Clark JI, Nikitina E, et al. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer.
J Immunol
2001
;
166
:
678
–89.
15
Webb SD, Sherratt JA, Fish RG. Cells behaving badly: a theoretical model for the Fas/FasL system in tumour immunology.
Math Biosci
2002
;
179
:
113
–29.
16
Doubrovina ES, Doubrovin MM, Vider E, et al. Evasion from NK cell immunity by MHC class I chain-related molecules expressing colon adenocarcinoma.
J Immunol
2003
;
171
:
6891
–9.
17
Kim R, Emi M, Tanabe K. Cancer cell immune escape and tumor progression by exploitation of anti-inflammatory and pro-inflammatory responses.
Cancer Biol Ther
2005
;
4
:
924
–33.
18
Huber V, Fais S, Iero M, et al. Human colorectal cancer cells induce T-cell death through release of proapoptotic microvesicles: role in immune escape.
Gastroenterology
2005
;
128
:
1796
–804.
19
Song E, Chen J, Ouyang N, Su F, Wang M, Heemann U. Soluble Fas ligand released by colon adenocarcinoma cells induces host lymphocyte apoptosis: an active mode of immune evasion in colon cancer.
Br J Cancer
2001
;
85
:
1047
–54.
20
Groh V, Wu J, Yee C, Spies T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation.
Nature
2002
;
419
:
734
–8.
21
Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines.
Nat Med
2004
;
10
:
909
–15.
22
Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow?
Lancet
2001
;
357
:
539
–45.
23
Pikarsky E, Porat RM, Stein I, et al. NF-κB functions as a tumour promoter in inflammation-associated cancer.
Nature
2004
;
431
:
461
–6.
24
Coussens LM, Werb Z. Inflammation and cancer.
Nature
2002
;
420
:
860
–7.
25
Maderna P, Godson C. Phagocytosis of apoptotic cells and the resolution of inflammation.
Biochim Biophys Acta
2003
;
1639
:
141
–51.
26
Gregory CD, Devitt A. The macrophage and the apoptotic cell: an innate immune interaction viewed simplistically?
Immunology
2004
;
113
:
1
–14.
27
Kim S, Elkon KB, Ma X. Transcriptional suppression of interleukin-12 gene expression following phagocytosis of apoptotic cells.
Immunity
2004
;
21
:
643
–59.
28
Bondanza A, Zimmermann VS, Rovere-Querini P, et al. Inhibition of phosphatidylserine recognition heightens the immunogenicity of irradiated lymphoma cells in vivo.
J Exp Med
2004
;
200
:
1157
–65.
29
Kim R, Emi M, Tanabe K, Uchida Y, Toge T. The role of Fas ligand and transforming growth factor β in tumor progression: molecular mechanisms of immune privilege via Fas-mediated apoptosis and potential targets for cancer therapy.
Cancer
2004
;
100
:
2281
–91.
30
Harizi H, Juzan M, Grosset C, Rashedi M, Gualde N. Dendritic cells issued in vitro from bone marrow produce PGE2 that contributes to the immunomodulation induced by antigen-presenting cells.
Cell Immunol
2001
;
209
:
19
–28.
31
Murata Y, Shimamura T, Hamuro J. The polarization of Th1/Th2 balance is dependent on the intracellular thiol redox status of macrophages due to the distinctive cytokine production.
Int Immunol
2002
;
14
:
201
–12.
32
Espana L, Fernandez Y, Rubio N, Torregrosa A, Blanco J, Sierra A. Overexpression of Bcl-xL in human breast cancer cells enhances organ-selective lymph node metastasis.
Breast Cancer Res Treat
2004
;
87
:
33
–44.
33
Jung YJ, Kim JY, Park JH. TGF-β1 inhibits Fas-mediated apoptosis by regulating surface Fas and cFLIPL expression in human leukaemia/lymphoma cells.
Int J Mol Med
2004
;
13
:
99
–104.
34
Santiago B, Galindo M, Palao G, Pablos JL. Intracellular regulation of Fas-induced apoptosis in human fibroblasts by extracellular factors and cycloheximide.
J Immunol
2004
;
172
:
560
–6.
35
Vakkila J, Lotze MT. Inflammation and necrosis promote tumour growth.
Nat Rev Immunol
2004
;
4
:
641
–8.
36
Zhang D, Bar-Eli M, Meloche S, Brodt P. Dual regulation of MMP-2 expression by the type 1 insulin-like growth factor receptor: the phosphatidylinositol 3-kinase/Akt and Raf/ERK pathways transmit opposing signals.
J Biol Chem
2004
;
279
:
19683
–90.
37
Tang N, Wang L, Esko J, et al. Loss of HIF-1α in endothelial cells disrupts a hypoxia-driven VEGF autocrine loop necessary for tumorigenesis.
Cancer Cell
2004
;
6
:
485
–95.
38
Bellamy WT, Richter L, Sirjani D, et al. Vascular endothelial cell growth factor is an autocrine promoter of abnormal localized immature myeloid precursors and leukemia progenitor formation in myelodysplastic syndromes.
Blood
2001
;
97
:
1427
–34.
39
Hasebe H, Sato K, Yanagie H, et al. Bcl-2, Bcl-xL and c-FLIPL potentially regulate the susceptibility of human peripheral blood monocyte-derived dendritic cells to cell death at different developmental stages.
Biomed Pharmacother
2002
;
56
:
144
–51.
40
Young MR, Wright MA, Pandit R. Myeloid differentiation treatment to diminish the presence of immune-suppressive CD34+ cells within human head and neck squamous cell carcinomas.
J Immunol
1997
;
159
:
990
–6.
41
Kusmartsev SA, Kusmartseva IN, Afanasyev SG, Cherdyntseva NV. Immunosuppressive cells in bone marrow of patients with stomach cancer.
Adv Exp Med Biol
1998
;
451
:
189
–94.
42
Bronte V, Chappell DB, Apolloni E, et al. Unopposed production of granulocyte-macrophage colony-stimulating factor by tumors inhibits CD8+ T cell responses by dysregulating antigen-presenting cell maturation.
J Immunol
1999
;
162
:
5728
–37.
43
Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI. 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
–99.
44
Storozynsky E, Woodward JG, Frelinger JG, Lord EM. Interleukin-3 and granulocyte-macrophage colony-stimulating factor enhance the generation and function of dendritic cells.
Immunology
1999
;
97
:
138
–49.
45
Zou W, Machelon V, Coulomb-L'Hermin A, et al. Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacytoid precursor dendritic cells.
Nat Med
2001
;
7
:
1339
–46.
46
Zea AH, Rodriguez PC, Culotta KS, et al. l-Arginine modulates CD3ζ expression and T cell function in activated human T lymphocytes.
Cell Immunol
2004
;
232
:
21
–31.
47
Porgador A, Snyder D, Gilboa E. Induction of antitumor immunity using bone marrow-generated dendritic cells.
J Immunol
1996
;
156
:
2918
–26.
48
Ohm JE, Gabrilovich DI, Sempowski GD, et al. VEGF inhibits T-cell development and may contribute to tumor-induced immune suppression.
Blood
2003
;
101
:
4878
–86.
49
Ruland J, Mak TW. Transducing signals from antigen receptors to nuclear factor κB.
Immunol Rev
2003
;
193
:
93
–100.
50
Nefedova Y, Cheng P, Gilkes D, et al. Activation of dendritic cells via inhibition of Jak2/STAT3 signaling.
J Immunol
2005
;
175
:
4338
–46.
51
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
–406.
52
Kim JW, Tsukishiro T, Johnson JT, Whiteside TL. Expression of pro- and antiapoptotic proteins in circulating CD8+ T cells of patients with squamous cell carcinoma of the head and neck.
Clin Cancer Res
2004
;
10
:
5101
–10.
53
Bauernhofer T, Kuss I, Friebe-Hoffmann U, et al. Role of prolactin receptor and CD25 in protection of circulating T lymphocytes from apoptosis in patients with breast cancer.
Br J Cancer
2003
;
88
:
1301
–9.
54
Reichert TE, Strauss L, Wagner EM, Gooding W, Whiteside TL. Signaling abnormalities, apoptosis, and reduced proliferation of circulating and tumor-infiltrating lymphocytes in patients with oral carcinoma.
Clin Cancer Res
2002
;
8
:
3137
–45.
55
Kim JW, Wieckowski E, Taylor DD, Reichert TE, Watkins S, Whiteside TL. Fas ligand-positive membranous vesicles isolated from sera of patients with oral cancer induce apoptosis of activated T lymphocytes.
Clin Cancer Res
2005
;
11
:
1010
–20.
56
Almand B, Resser JR, Lindman B, et al. Clinical significance of defective dendritic cell differentiation in cancer.
Clin Cancer Res
2000
;
6
:
1755
–66.
57
Kusmartsev S, Gabrilovich DI. Immature myeloid cells and cancer-associated immune suppression.
Cancer Immunol Immunother
2002
;
51
:
293
–8.
58
Lee JH, Torisu-Itakara H, Cochran AJ, et al. Quantitative analysis of melanoma-induced cytokine-mediated immunosuppression in melanoma sentinel nodes.
Clin Cancer Res
2005
;
11
:
107
–12.
59
Schule JM, Bergkvist L, Hakansson L, Gustafsson B, Hakansson A. CD28 expression in sentinel node biopsies from breast cancer patients in comparison with CD3-ζ chain expression.
J Transl Med
2004
;
2
:
45
.
60
Poindexter NJ, Sahin A, Hunt KK, Grimm EA. Analysis of dendritic cells in tumor-free and tumor-containing sentinel lymph nodes from patients with breast cancer.
Breast Cancer Res
2004
;
6
:
R408
–15.
61
Schule J, Bergkvist L, Hakansson L, Gustafsson B, Hakansson A. Down-regulation of the CD3-ζ chain in sentinel node biopsies from breast cancer patients.
Breast Cancer Res Treat
2002
;
74
:
33
–40.
62
Glaser M, Kirchner H, Holden HT, Herberman RB. Inhibition of cell-mediated cytotoxicity against tumor-associated antigens by suppressor cells from tumor-bearing mice.
J Natl Cancer Inst
1976
;
56
:
865
–7.
63
Poupon MF, Kolb JP, Lespinats G. Evidence for splenic suppressor cells in C3H/He, T-cell-deprived C3H/He, and nude mice bearing a 3-methylcholanthrene-induced fibrosarcoma.
J Natl Cancer Inst
1976
;
57
:
1241
–7.
64
Wakkach A, Fournier N, Brun V, Breittmayer JP, Cottrez F, Groux H. Characterization of dendritic cells that induce tolerance and T regulatory 1 cell differentiation in vivo.
Immunity
2003
;
18
:
605
–17.
65
Nolte-'t Hoen EN, Wagenaar-Hilbers JP, Boot EP, et al. Identification of a CD4+CD25+ T cell subset committed in vivo to suppress antigen-specific T cell responses without additional stimulation.
Eur J Immunol
2004
;
34
:
3016
–27.
66
Vermi W, Bonecchi R, Facchetti F, et al. Recruitment of immature plasmacytoid dendritic cells (plasmacytoid monocytes) and myeloid dendritic cells in primary cutaneous melanomas.
J Pathol
2003
;
200
:
255
–68.
67
Hartmann E, Wollenberg B, Rothenfusser S, et al. Identification and functional analysis of tumor-infiltrating plasmacytoid dendritic cells in head and neck cancer.
Cancer Res
2003
;
63
:
6478
–87.
68
Sakaguchi S. Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self.
Nat Immunol
2005
;
6
:
345
–52.
69
Shevach EM. CD4+ CD25+ suppressor T cells: more questions than answers.
Nat Rev Immunol
2002
;
2
:
389
–400.
70
Liyanage UK, Moore TT, Joo HG, et al. 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
–61.
71
Chen W, Jin W, Hardegen N, et al. Conversion of peripheral CD4+CD25− naive T cells to CD4+CD25+ regulatory T cells by TGF-β induction of transcription factor Foxp3.
J Exp Med
2003
;
198
:
1875
–86.
72
Moser M. Dendritic cells in immunity and tolerance—do they display opposite functions?
Immunity
2003
;
19
:
5
–8.
73
Curotto de Lafaille MA, Lafaille JJ. CD4(+) regulatory T cells in autoimmunity and allergy.
Curr Opin Immunol
2002
;
14
:
771
–8.
74
Read S, Malmstrom V, Powrie F. Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation.
J Exp Med
2000
;
192
:
295
–302.
75
Sheen-Chen SM, Chen HS, Eng HL, Chen WJ. Circulating soluble Fas in patients with breast cancer.
World J Surg
2003
;
27
:
10
–3.
76
Kondera-Anasz Z, Mielczarek-Palacz A, Sikora J. Soluble Fas receptor and soluble Fas ligand in the serum of women with uterine tumors.
Apoptosis
2005
;
10
:
1143
–9.
77
Enjoji M, Yamaguchi K, Nakashima M, et al. Serum levels of soluble molecules associated with evasion of immune surveillance: a study in biliary disease.
Liver Int
2004
;
24
:
330
–4.
78
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.
79
Li JH, Rosen D, Sondel P, Berke G. Immune privilege and FasL: two ways to inactivate effector cytotoxic T lymphocytes by FasL-expressing cells.
Immunology
2002
;
105
:
267
–77.
80
Hoffmann TK, Dworacki G, Tsukihiro T, et al. Spontaneous apoptosis of circulating T lymphocytes in patients with head and neck cancer and its clinical importance.
Clin Cancer Res
2002
;
8
:
2553
–62.
81
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
–82.
82
Smyth MJ, Swann J, Cretney E, Zerafa N, Yokoyama WM, Hayakawa Y. NKG2D function protects the host from tumor initiation.
J Exp Med
2005
;
202
:
583
–8.
83
Holdenrieder S, Stieber P, Peterfi A, Nagel D, Steinle A, Salih HR. Soluble MICA in malignant diseases.
Int J Cancer
2006
;
118
:
684
–7.
84
Raffaghello L, Prigione I, Airoldi I, et al. Downregulation and/or release of NKG2D ligands as immune evasion strategy of human neuroblastoma.
Neoplasia
2004
;
6
:
558
–68.
85
Juprelle-Soret M, Wattiaux-De Coninck S, Wattiaux R. Subcellular localization of transglutaminase. Effect of collagen.
Biochem J
1988
;
50
:
421
–7.
86
Savinov AY, Wong FS, Stonebraker AC, Chervonsky AV. Presentation of antigen by endothelial cells and chemoattraction are required for homing of insulin-specific CD8+ T cells.
J Exp Med
2003
;
97
:
643
–56.
87
Spiotto MT, Yu P, Rowley DA, et al. Increasing tumor antigen expression overcomes “ignorance” to solid tumors via crosspresentation by bone marrow-derived stromal cells.
Immunity
2002
;
7
:
737
–47.
88
Fonteneau JF, Kavanagh DG, Lirvall M, et al. Characterization of the MHC class I cross-presentation pathway for cell-associated antigens by human dendritic cells.
Blood
2003
;
102
:
4448
–55.
89
Buttiglieri S, Galetto A, Forno S, De Andrea M, Matera L. Influence of drug-induced apoptotic death on processing and presentation of tumor antigens by dendritic cells.
Int J Cancer
2003
;
106
:
516
–20.
90
Dengjel J, Schoor O, Fischer R, et al. Autophagy promotes MHC class II presentation of peptides from intracellular source proteins.
Proc Natl Acad Sci U S A
2005
;
102
:
7922
–7.
91
Strauss L, Volland D, Kunkel M, Reichert TE. Dual role of VEGF family members in the pathogenesis of head and neck cancer (HNSCC): possible link between angiogenesis and immune tolerance.
Med Sci Monit
2005
;
11
:
BR280
–92.
92
Ishida T, Oyama T, Carbone DP, Gabrilovich DI. Defective function of Langerhans cells in tumor-bearing animals is the result of defective maturation from hemopoietic progenitors.
J Immunol
1998
;
161
:
4842
–51.
93
Lee CG, Heijn M, di Tomaso E, et al. Anti-vascular endothelial growth factor treatment augments tumor radiation response under normoxic or hypoxic conditions.
Cancer Res
2000
;
60
:
5565
–70.
94
Hurwitz HI, Fehrenbacher L, Hainsworth JD, et al. Bevacizumab in combination with fluorouracil and leucovorin: an active regimen for first-line metastatic colorectal cancer.
J Clin Oncol
2005
;
23
:
3502
–8.
95
Johnson DH, Fehrenbacher L, Novotny WF, et al. Randomized phase II trial comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and paclitaxel alone in previously untreated locally advanced or metastatic non-small-cell lung cancer.
J Clin Oncol
2004
;
22
:
2184
–91.
96
Rugo HS. Bevacizumab in the treatment of breast cancer: rationale and current data.
Oncologist
2004
;
9
:
43
–9.
97
Wiemann B, Starnes CO. Coley's toxins, tumor necrosis factor and cancer research: a historical perspective.
Pharmacol Ther
1994
;
64
:
529
–64.
98
Krieg AM, Yi AK, Matson S, et al. CpG motifs in bacterial DNA trigger direct B-cell activation.
Nature
1995
;
374
:
546
–9.
99
Hemmi H, Takeuchi O, Kawai T, et al. A Toll-like receptor recognizes bacterial DNA.
Nature
2000
;
408
:
740
–5.
100
Speiser DE, Lienard D, Rufer N, et al. Rapid and strong human CD8+ T cell responses to vaccination with peptide, IFA, and CpG oligodeoxynucleotide 7909.
J Clin Invest
2005
;
115
:
739
–46.
101
Weigel BJ, Rodeberg DA, Krieg AM, Blazar BR. CpG oligodeoxynucleotides potentiate the antitumor effects of chemotherapy or tumor resection in an orthotopic murine model of rhabdomyosarcoma.
Clin Cancer Res
2003
;
9
:
3105
–14.
102
Milas L, Mason KA, Ariga H, et al. CpG oligodeoxynucleotide enhances tumor response to radiation.
Cancer Res
2004
;
64
:
5074
–7.
103
Emi M, Kim R, Tanabe K, Uchida Y, Toge T. Targeted therapy against Bcl-2-related proteins in breast cancer cells.
Breast Cancer Res
2005
;
7
:
R940
–52.
104
Badros AZ, Goloubeva O, Rapoport AP, et al. Phase II study of G3139, a Bcl-2 antisense oligonucleotide, in combination with dexamethasone and thalidomide in relapsed multiple myeloma patients.
J Clin Oncol
2005
;
23
:
4089
–99.
105
Gianni L, Zambetti M, Clark K, et al. Gene expression profiles in paraffin-embedded core biopsy tissue predict response to chemotherapy in women with locally advanced breast cancer.
J Clin Oncol
2005
;
23
:
7265
–77.
106
Chen CN, Lin JJ, Chen JJ, et al. Gene expression profile predicts patient survival of gastric cancer after surgical resection.
J Clin Oncol
2005
;
23
:
7286
–95.