Está en la página 1de 14

Immune escape of tumors: apoptosis resistance and tumor counterattack

Frederik H. Igney and Peter H. Krammer Tumor Immunology Program, German Cancer Research Center (DKFZ), Heidelberg, Germany

Abstract: Interactions between the immune system and malignant cells play an important role in tumorigenesis. Failure of the immune system to detect and reject transformed cells may lead to cancer development. Tumors use multiple mechanisms to escape from immune-mediated rejection. Many of these mechanisms are now known on a cellular and molecular level. Despite this knowledge, cancer immunotherapy is still not an established treatment in the clinic. This review discusses the immune escape mechanisms used by tumors with an emphasis on mechanisms related to apoptosis. J. Leukoc. Biol. 71: 907920; 2002.
Key Words: CD95L immunosuppression cytotoxic T cells CD95 death receptor

TUMORS AND THE IMMUNE SYSTEM


The immune system has evolved in order to detect and eliminate pathogens that may do harm to the organism. Moreover, it serves as a watchdog against transformed cells that may lead to cancer [1, 2]. The key cells of the immune system for tumor surveillance are T cells, which are part of the adaptive immune response. After recognition of an antigen on a tumor cell via the T cell receptor (TCR), activated CD8 T cells can kill the tumor target cell and thus are called cytotoxic T cells (CTL). One subset of CD4 T cells, T helper cell type 1 (Th1), provides help for the activation of CD8 T cells. The other CD4 subset, Th2 cells, stimulates a humoral immune response and suppresses the development of a Th1 response. CD4 T cells can also display cytotoxic activity in some situations. CD8 and CD4 T cells recognize antigens presented as peptides by major histocompatibility complex (MHC) class I or class II molecules, respectively. Numerous tumor antigens have been identied that can be recognized by T cells [35]. Some of these antigens are expressed exclusively by tumors and thus are called tumorspecic antigens. These antigens arise from mutations or translocations of normal cellular genes (e.g., catenin, cdk4, ras). The mutations may be involved directly in carcinogenesis. Another group of antigens are the tumor-associated antigens that are not only expressed by tumor cells, but are also expressed by other cells of the body. Cancer-testes antigens are expressed on a variety of epithelial tumors as well as on testis and placental tissue (e.g., MAGE, BAGE, GAGE). Overexpressed nonmutated proteins (e.g., p53, Her2/neu) may also

serve as tumor antigens for T cells. In addition, tumor-inltrating lymphocytes have been identied that are reactive against differentiation antigens present on normal melanocytes as well as melanomas (e.g., MART-1/Melan-A, tyrosinase, gp100). Moreover, antigens from tumorigenic viruses are presented on tumor cells. The expression of tumor antigens may be heterogeneous within a tumor, and a single patient can develop immune reactions to multiple antigens [6, 7]. Two different models for the immune response to tumors have been proposed: the concept of immunosurveillance and the danger model. According to the immunosurveillance hypothesis, tumors expressing antigens are regarded as nonself by the immune system, and a major function of the immune system is to survey the body for the development of malignancy and to eliminate tumor cells as they arise [8]. To detect danger, the immune system uses professional antigen-presenting cells (APC) as sentinels of tissue damage. In the presence of danger signals, APCsuch as dendritic cells, activated macrophages, and B cellsstimulate the T cell response. The danger model proposes that cancer cells do not appear dangerous to the immune system, so that the response of T cells to tumors is not initiated [9]. Natural killer (NK) cells of the innate immune system also play an important role in immune surveillance of tumors [1]. NK cells kill MHC class I-decient cellsa phenomenon that is part of the missing self hypothesis [10, 11]. The activity of NK cells is controlled by a balance of positive and negative signals. Engagement of inhibitory receptors by MHC class I molecules blocks activation signals. Two families of inhibitory receptors have been identied in humans: the immunoglobulin-like killer cell inhibitory receptors and the lectin-like CD94-NKG2 receptors. Stimulatory receptors comprise receptors (e.g., CD16, CD94-NKG2C, natural cytotoxicity receptors) that are supposed to bind to constitutively expressed ligands [12] and NKG2d receptors, which bind to molecules that are induced by cellular stress [1315]. Ligands for NKG2d receptors are the MHC class I chain-related (MIC) glycoproteins MICA and MICB in humans and the minor histocompatibility antigen H60 and the retionic acid early inducible (Rae-1) family in mice.

Correspondence: Prof. Dr. Peter H. Krammer, Tumor Immunology Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany. E-mail: P.Krammer@dkfz-heidelberg.de Received February 3, 2002; revised March 18, 2002; accepted March 20, 2002.

Journal of Leukocyte Biology Volume 71, June 2002 907

Additional cells of the innate immune system involved in immunity against tumors are macrophages and neutrophils [16 19]. They can reject tumors by direct killing of the tumor cells, by destruction of tumor vessels and matrix, and by inhibition of angiogenesis. Moreover, they display tumor antigens and can stimulate other immune cells such as CTL, NK cells, or APC. In contrast, inammatory cells may also contribute to tumor progression by production of tumor growth factors and stimulation of angiogenesis [20]. Macrophages and neutrophils are recruited to the tumor site by expression of adhesion molecules on endothelial cells and by chemotactic proteins.

TUMOR IMMUNE ESCAPE MECHANISMS


Despite presentation of antigens by malignant cells and the presence of immune cells that could potentially react against these cells, in many cases the immune system does not get activated but ignores the tumor [2, 21]. According to the danger model, APC may not get activated in this situation due to of a lack of danger signals [9]. Other factors may also contribute to immunological ignorance. The immune system ignores tumor cells, which fail to migrate to lymph nodes and fail to activate T cells directly [22, 23]. In addition, tumors growing in immune privileged sites such as the brain or the eye are not surveilled by the immune system [24]. Down-regulation of adhesion molecules in malignant tissue may inhibit immune inltration and thus may also contribute to immunological ignorance [2527]. The tumor stroma has also been shown to play a critical role [23, 28]. It may serve as a physical barrier between the tumor and immune cells. Growth of antigenic tumors in the presence of potent immune cells cannot be explained by immunological ignorance alone. A major goal of cancer immunotherapy is to generate an anti-tumor immune response, e.g., by vaccination with cancer cells fused with APC or by transfer of anti-tumor T cells.

However, many of these approaches do not efciently stimulate immunity, or the tumors continue to grow despite the presence of an immune reaction [1, 3, 29]. Multiple mechanisms have been identied that tumors use to escape from rejection (Table 1) [30 32]. It is likely that malignant escape variants are selected by the immune system. One strategy to escape from the adaptive immune response is by impaired antigen presentation. In general, defects in antigen presentation are more pronounced in metastatic lesions than in the primary tumor. In some tumors, expression of the tumor antigen is down-regulated, leading to enhanced tumor incidence and metastasis [3335]. Moreover, mutations of the antigen can result in escape from the initial response and contribute to the heterogeneity of tumor lesions [36]. The heterogeneous expression of multiple antigens may hinder the establishment of an efcient specic immune response. In addition, reduced MHC-I expression prevents recognition of tumor cells by the immune system [37, 38]. Tumors frequently have a heterogeneous pattern of MHC-I expression. Total loss of MHC-I is mainly caused by mutations of the 2 microglobulin subunit [39]. Gene deletions or rearrangements, point mutations, and defects in transcriptional regulation lead to selective loss of an MHC haplotype, locus, or allele [27, 35, 40 43]. Total MHC-I down-regulation may target the tumor for NK cell attack. Therefore, the tumor needs further mechanisms to resist NK cell-mediated lysis, such as the expression of MHC-I surrogates [44, 45]. Reduced presentation of the tumor antigen is also achieved by defects in the antigen processing machinery. Down-regulation of the proteasome subunits LMP2 (low molecular mass polypeptide 2) and LMP7 changes the spectrum of peptides presented by MHC molecules [46 48]. Two proteins involved in loading antigenic peptides onto MHC-I molecules, TAP (transporter associated with antigen processing) and tapasin, are also frequently mutated or down-regulated in tumor cells [37, 38, 46 50]. TAP deciency results in loss of MHC-I expression and an increase in tumorigenesis. Moreover, some proteins of tumorigenic viruses are not efciently

TABLE 1. Strategy Ignorance

Tumor Immune Escape Mechanisms Mechanism Lack of danger signals Lack of tumor antigens in lymphoid organs Growth in immune privileged sites Lack of adhesion molecules Physical barrier by stroma Mutation or downregulation of tumor antigens Mutation or downregulation of MHC genes Defects in antigen processing (e.g., TAP, LMP deciency) Cytokines (TGF- , IL-10, VEGF, etc.) Prostaglandines RCAS1 Anergy induction (lack of costimulatory molecules) Immune deviation Regulatory T cells T cell deletion Expression of anti-apoptotic molecules Downregulation and mutation of pro-apoptotic molecules CD95L expression Expression of other death-receptor ligands

Impaired antigen presentation Expression of immunosuppressive factors and molecules Tolerance induction

Apoptosis resistance Counterattack (?)

908

Journal of Leukocyte Biology Volume 71, June 2002

http://www.jleukbio.org

presented because they interfere with their proteosomal proteolysis, e.g., by mutation of the clevage site [51]. Another strategy that tumors use to escape from immunemediated rejection is the expression of immunosuppressive factors [18, 52]. These factors may be expressed by the malignant cells themselves or by noncancerous cells present at the tumor site, such as immune, epithelial, or stromal cells. The most prominent of these factors is transforming growth factor (TGF- ) [5358]. TGF- is a cytokine that affects proliferation, activation, and differentiation of cells of innate and adaptive immunity and thus inhibits the anti-tumor immune response. Moreover, vascular endothelial growth factor (VEGF) is produced by many tumor cells [59]. Besides its angiogenic properties, it inhibits the differentiation of progenitors into dendritic cells. Further immunosuppressive factors expressed by malignant cells are prostaglandins [60 62], interleukin (IL)-10 [63], macrophage-colony stimulating factor [64, 65], and soluble tumor gangliosides [66]. The membrane protein RCAS1 (receptor-binding cancer antigen expressed on SiSo cells) inhibits proliferation and induces apoptosis in T cells in vitro, suggesting a role of this molecule in immune evasion of tumors [67]. The lack of a T cell response against tumor-associated antigens that are also expressed by other cells of the body or during development may be explained by tolerance [30, 68]. In healthy organisms, self-reactive T cells are tolerized mainly by deletion in the thymus, a process known as central tolerance. The mechanisms of peripheral tolerance induction prevent autoimmunity by tolerizing T cells that have escaped the process of central tolerance. Peripheral tolerance induction is a complex multistep process [69, 70], but in principle, four major mechanisms can be distinguished. One mechanism is the induction of anergy. T cell activation requires two signals, binding of a peptide-MHC complex to the TCR and binding of costimulatory molecules (e.g., B7) to their ligands (CD28) on the T cell surface [71, 72]. If a T cell binds via its TCR to a peptide-MHC complex on the target cell without sufcient costimulation, the T cell is rendered anergic and does not become activated when restimulated with antigen. Many tumor cells do not express costimulatory molecules and thus may induce anergy in anti-tumor lymphocytes [27, 7375]. Another process of tolerance induction that tumors exploit is immune deviation. In this process, the immune response is driven toward a Th2 humoral response away from a Th1 response required for efcient tumor rejection by cytotoxic T cells. The mechanism of immune deviation is not exactly understood, but it may depend on secretion of TGF- and IL-10 [76] or on the presentation of the tumor antigen by B cells to CD4 Th cells [77]. Tumors can also induce the generation of regulatory T cells [78]. Although the molecular mechanism is not clear, a subset of CD4 T cells seems to suppress the response of cytotoxic T cells against tumors in some settings [79, 80]. A further mechanism to establish peripheral tolerance to selfantigens is T cell deletion. Repetitive stimulation of T cells with the antigen induces apoptosis, a process referred to as activation-induced cell death (AICD). Thus, administration of antigens, such as superantigens, peptides, or allogeneic cells, or direct restimulation of the TCR by anti-CD3 antibodies has been shown to induce tolerance by T cell deletion [81 84].

This process is mainly mediated via the CD95/CD95L death system [85 88]. Costimulation via CD28 can rescue T cells from AICD [89], implicating another important role for expression of B7 on tumor cells. Tumors have been shown to induce tolerance by deleting anti-tumor T cells [90, 91]. AICD, as a result of chronic stimulation with the tumor antigen may contribute to immune escape, yet the signicance of this process has not directly been shown. Two further strategies used by tumors to evade rejection by the immune system are related to apoptosis. First, malignant cells have changes in the expression of molecules involved in apoptosis signaling, resulting in resistance of the tumor to the killing mechanisms of the immune system. Second, tumors may adopt a killing mechanism from cytotoxic immune cells to delete the attacking anti-tumor lymphocytes, a concept called tumor counterattack. These apoptosis-related immune-escape mechanisms will be discussed in detail below.

KILLING MECHANISMS OF THE IMMUNE SYSTEM


T cells and NK cells use two major mechanisms to kill tumor cells: the death receptor pathway and the granule exocytosis pathway [92]. In the death receptor pathway, the lymphocyte displays the death ligand CD95L on the cell surface, triggering apoptosis via the death receptor CD95 on the target cell [9395]. Moreover, for immune surveillance of tumors and metastases, NK cells also use the death ligand TRAIL [tumor necrosis factor (TNF)-related apoptosis-inducing ligand], which triggers apoptosis via the death receptors TRAIL-R1 or TRAIL-R2 [96 98]. Death receptors are members of the TNF receptor superfamily and comprise a subfamily characterized by an intracellular domain, the death domain [99, 100]. The so-called decoy receptors are closely related to the death receptors and lack a functional death domain [101, 102]. Death receptors are activated by their natural ligands, co-evolved as a death ligand family, called the TNF family (Fig. 1). When the respective ligand binds to the death receptor, the death domains attract intracellular adaptor proteins, which, in turn, recruit the proform of the initiator caspase 8. (Caspase-10 may also be an initiator caspase, but this is still controversial) [103107]. The resulting protein complex is called deathinducing signaling complex (DISC). At the DISC, procaspase-8 is cleaved autocatalytically and yields the active initiator caspase-8. In some cells, so-called type I cells, the amount of active caspase-8 formed at the DISC is sufcient to initiate apoptosis directly. In type II cells, the amount is too small, and mitochondria are used as ampliers of the apoptotic signal [108]. Activation of mitochondria is mediated by the Bcl-2 homology (BH)3-only Bcl-2 family member Bid, which is cleaved by active caspase-8 and translocates to the mitochondria. After activation, mitochondria release cytochrome c, apoptosis-inducing factor, and other apoptogenic factors from the intermembrane space to the cytosol [109, 110]. Concomitantly, the mitochondrial transmembrane potential m drops. In the cytosol, cytochrome c forms a complex with Apaf-1, adenosine 5 -triphosphate, and procaspase-9. This complex is called
Igney and Krammer Apoptosis and tumor immune escape 909

Fig. 1. Apoptosis signaling via death receptors. Binding of death ligands (shown here for CD95L) leads to formation of the DISC. In the DISC, the initiator caspase-8 is activated. (Whether caspase-10 is a true initiator caspase is still controversial.) Cleavage of Bid by caspase-8 activates the mitochondrial pathway and can be used to amplify the apoptotic signal. Activation of mitochondria leads to cytochrome c release into the cytosol and formation of the apoptosome. At the apoptosome, caspase-9 is activated. Caspase-8 and -9 activate executioner caspases, which in turn cleave the death substrates, eventually resulting in apoptosis. Apoptosis can be inhibited on different levels by antiapoptotic proteins (shown in red).

apoptosome. Within the apoptosome, the initiator caspase-9 is activated. Activated initiator caspases cleave and activate executioner caspases, mainly caspase-3, -6, and -7 [111]. The active executioner caspases then cleave each other, and thus start an amplifying proteolytic cascade of caspase activation. The active executioner caspases cleave cellular substrates, the death substrates, leading to the biochemical and morphological changes characteristic of apoptosis [111]. Various proteins regulate the apoptotic pathway at different levels. FLIPs (FLICE-inhibitory proteins) interfere with the initiation of apoptosis directly at the level of death receptors [112]. Two splice varians have been identied in human cells, a long form (cFLIPL) and a short form (cFLIPS). They share structural homology with procaspase-8, but lack its catalytic site. This structure enables them to bind to the DISC, thereby inhibiting the processing and activation of the initiator caspases. A major class of regulatory proteins are members of the Bcl-2 family that regulate apoptosis at the mitochondrial level [109, 110]. According to their function, Bcl-2 family members can be divided into antiapoptotic and proapoptotic proteins. Bcl-2 family proteins inuence the permeability of the mitochondrial membrane; however, the biochemical mechanism of their function is not entirely clear. A third class of regulatory proteins are the IAPs (inhibitor of apoptosis proteins), which bind to and inhibit caspases [113]. They may also function as ubiquitin ligases, promoting the degradation of the caspases bound. However, not all IAPs have been shown to suppress apoptosis, and some of them may also have functions other than caspase inhibition. IAPs themselves are inhibited by a protein, Smac/DIABLO [114, 115], which is
910 Journal of Leukocyte Biology Volume 71, June 2002

released from mitochondria along with cytochrome c during apoptosis and promotes caspase activation by binding to IAPs. In the calcium-dependent granule exocytosis pathway, lymphocytes secrete perforin and granzymes from cytotoxic granules toward the target cell. In the presence of calcium, perforin polymerizes and initiates as yet ill-dened changes in the target cell membrane, which allow granzymes to pass into the cell [116 118]. Granzymes are neutral serine proteases that can activate caspases in the target cell [119 121]. In addition, granzyme B may directly cleave the Bcl-2 family member Bid, initiating the mitochondrial death pathway [122]. Perforindecient mice are very susceptible to some carcinogens and oncogenic viruses [123] and develop spontaneous lymphomas with age [124]. Although it is clear that granzymes are indispensable effector molecules in a granule exocytosis-mediated host defense against viral pathogens [125], their contribution to tumor rejection remains controversial [126]. Thus, mice decient for granzymes A and B are capable of rejecting tumors in an efcient way. Macrophages and neutrophils use totally different killing mechanisms. In principle, they use four kinds of cytotoxic molecules [127]. The rst effector mechanism is the oxidative burst consisting of the release of reactive oxygen species (superoxide anion, hydrogen peroxide, and derivatives) produced by the phagocytic reduced nicotinamide adenine dinucleotide phosphate oxidase (NADPH) [128 130]. Reaction of peroxide via the myeloperoxidase pathway can yield hypochlorous acid and chloramines. A further cytotoxicity mechanism is the production of nitric oxide (NO) by the inducible NO synthase [129, 131]. The toxicity of NO is enhanced greatly by reacting with superoxide to form peroxynitrite. The molecular targets of reactive oxygen species, NO, and derivatives inside
http://www.jleukbio.org

the target cells are not fully dened yet, but may include enzymes essential for cellular survival. Phagocytes also release antimicrobial peptides, defensins and cathelicidins, which have afnity for bacterial and eukaryotic membranes and may lyse cells by disrupting the phospholipid bilayer [132]. Other mechanisms include the production and release of a variety of proteases including elastase, proteinase 3, and metalloproteases [127]. These proteases degrade extracellular matrix components and other proteins.

RESISTANCE TO APOPTOSIS AND IMMUNE ESCAPE


Resistance of tumor cells to the effector mechanisms of the immnue system leads not only to escape of the tumors from immunosurveillance, but may also dramatically inuence the efcacy of immunotherapy. In an immune-competent host, tumor cells are selected for resistance against the effector mechanisms of the immune system, a concept known as immunoselection or immunoediting. This concept has been conrmed by several experiments. Tumor cells derived from T cell-decient RAG2 / mice grew progressively in RAG2 / mice, but were rejected in wild-type mice [133]. Tumors derived from wild-type mice grew at a similar rate in both mouse strains. Moreover, a signicant proportion of aging mice with a mutated CD95L (gld mice) develop B cell malignancies [134]. These B cell lymphomas grew and metastasized in immmunodecient mice but were rejected by immunocompetent mice. Furthermore, neutraliziation of TRAIL promoted tumor development in mice inoculated with a chemical carcinogen [98]. Tumor cells derived from these animals were sensitive to TRAIL-mediated apoptosis, whereas those from control mice were not. Similarly, spontaneous lymphomas from perforin knockout mice were rejected by T cells when transplanted into wild-type animals, but grew in perforin knockout mice [124]. Although many mechanisms of tumor resistance to apoptosis have been identied (Fig. 1), only a few of them have directly been shown to be involved in immune escape. One strategy tumors use to acquire apoptosis resistance is the overexpression of antiapoptotic molecules. The antiapoptotic proteins FLIPL,S interfere with apoptosis induction at the level of death receptors, but do not prevent apoptosis by perforin/granzyme [112, 135, 136]. Nevertheless, overexpression of FLIP mediates the immune escape of tumors in mouse models. Tumors with high expression levels of FLIPL were shown to escape from T cell-mediated immunity in vivo despite the presence of the perforin/granzyme pathway [137]. In vivo tumor cells were selected for elevated FLIPL levels. FLIPL overepxression also prevents rejection of tumors by perforin-decient NK cells [138]. Human melanomas were shown to express high levels of FLIP [135, 139, 140]. Moreover, in Epstein-Barr virus-positive Burkitts lymphoma cell lines, an increased ratio of FLIP to caspase-8 correlated with resistance to CD95-mediated apoptosis [141]. Viral analogs of FLIP, viral FLIPs (v-FLIPs), are encoded by some tumorigenic viruses, such as Kaposi sarcoma-associated herpesvirus (KSHV) [142144]. KSHV-FLIP promotes tumor establishment and growth in immunocompetent mice by prevention of death receptor-induced apoptosis

triggered by T cells [145]. Therefore, v-FLIPs may contribute to immune escape of v-FLIP-encoding viruses. Further antiapoptotic proteins are involved in apoptosis resistance of tumors, although the signicance for immune escape is less clear. Enhanced Bcl-2 expression is found in follicular B-cell lymphoma (Bcl) with the chromosomal translocation t(14;18) that couples the Bcl-2 gene to the immunoglobulin heavy chain locus [146 149]. In cooperation with the oncogenes c-Myc or promyelocytic leukemia retinoic acid receptor , Bcl-2 contributes to tumorigenesis [150 153]. In some studies, high Bcl-2 expression correlates with the grade of malignancy of human tumors [154 156]. Moreover, it has been shown in in vitro and in vivo models that Bcl-2 expression confers resistance to CD95L and other apoptosis stimuli [154, 157159]. In some types of tumors, high Bcl-2 expression appears to be predictive of a poor disease-free survival [155, 156, 160]. The tumor-associated viruses Epstein-Barr virus and human KSHV encode proteins that are homologs of Bcl-2. Both proteins, BHRF1 or KSbcl-2 (vBcl-2), respectively, have an antiapoptotic function and enhance survival of the infected cells [161164]. Thus, they may contribute to apoptosis resistance of virus-induced tumors. In addition, the antiapoptotic Bcl-2 family members Bcl-xL and Mcl-1 are up-regulated in tumors and can confer resistance to multiple apoptosis-inducing pathways [159, 165168]. The IAP family member Survivin is expressed in a highly tumor-specic manner [169]. It is found in the vast majority of human tumors, but not in normal adult tissues [170]. In neuroblastomas, expression correlates with a more aggressive and unfavorable disease [171]. Besides its antiapoptotic activity, Survivin also has an apparent role in the cell cycle [169]. In transgenic mice expressing Survivin in the skin, its antiapoptotic function was more prominent than its role in cell division; Survivin, however, did not affect CD95-induced cell death [172]. Expression of a mutant of Survivin that could not be phosphorylated induces cytochrome c release and cell death. In xenograft tumor models, this mutant suppressed tumor growth and reduced intraperitoneal tumor dissemination [173, 174]. The tumor-suppressing activity has been observed in immune-decient mice and thus does not seem to depend on the immune system but on other apoptosis stimuli. Another IAP family member, cIAP2, is affected by the translocation t(11;18)(q21;q21) that is found in about 50% of marginal cell lymphomas of the mucosa-associated lymphoid tissue (MALT) [175]. This suggests a role of cIAP2 in the oncogenesis of MALT lymphoma. Tumor cells cannot only resist killing by cytotoxic lymphocytes through mechanisms blocking the death receptor pathway, but also through direct interference with the perforin/ granzyme pathway. The serine protease inhibitor PI-9/SPI-6 that inhibits granzyme B is expressed in a variety of human and murine tumors. Overexpressions result in resistance of tumor cells to cytotoxic lymphocytes and to immune escape [176 178]. A different strategy of tumors to escape from the effector mechanisms of the immune system may be to neutralize or impair the death-inducing stimuli. Thus, the expression of soluble receptors that act as decoys for death ligands may interfere with apoptosis induction via death receptors. To date,
Igney and Krammer Apoptosis and tumor immune escape 911

two distinct soluble receptors, soluble CD95 (sCD95) and decoy receptor 3 (DcR3), have been shown to inhibit CD95 signaling competitively. sCD95 is expressed in various malignancies, and elevated levels can be found in the sera of cancer patients [179 182]. High sCD95 serum levels were associated with poor prognosis in melanoma patients. DcR3 is amplied genetically in a number of lung and colon carcinomas and is overepxressed in several adenocarcinomas, glioma cell lines, and glioblastomas [101, 102, 183, 184]. Ectopic expression of DcR3 in a rat gliosarcoma model resulted in a decrease of immune cell inltration, suggesting an involvement of DcR3 in immune evasion of malignant glioma [184]. Moreover, a mechanism for resistance toward the perforin/granzyme pathway may be that binding of perforin to the tumor cell membrane is impaired [185]. Acute myeloid leukemia cells that failed to bind perforin on their surface were completely resistant toward NK cell-mediated cytotoxicity. Tumors can also acquire apoptosis resistance by downregulation or inactivation of proapoptotic molecules. In comparison to their normal counterparts, some tumor cells show a decreased expression of the death receptor CD95. This has been demonstrated for hepatocellular carcinomas, neoplastic colon epithelium, melanomas, and other tumors [186 190]. Loss of CD95 may contribute to chemoresistance and immune evasion. Transcriptional mechanisms may account for this. Oncogenic Ras seems to down-regulate CD95 [191], and in hepatocellular carcinomas, loss of CD95 expression was accompanied by p53 aberrations [190]. Several CD95 gene mutations have been demonstrated in primary samples of myeloma and T cell leukemia [192194]. The mutations include point mutations in the cytoplasmic death domain of CD95 and a deletion leading to a truncated form of the death receptor. These mutated forms of CD95 may interfere in a dominantnegative way with apoptosis induction via CD95. In families with germline CD95 mutations, usually resulting in autoimmune lymphoproliferative syndrome, the risk for the development of lymphomas is increased [195]. Deletions and mutations of the death receptors TRAIL-R1 and TRAIL-R2 have also been observed in tumors [196 199]. The frequent deletion of the chromosomal region 8p21-22 in head and neck cancer and non-small cell lung cancers affects the TRAIL-R2 gene. Mutations have been found in the ectodomain or the death domain of TRAIL-R1 or TRAIL-R2. Further mutations result in a truncated form or other antiapoptotic forms. Down-regulation or mutation of death receptors may impair tumor immunosurveillance by NK and T cells. In neuroblastomas with amplication of the oncogene MYCN, the gene for the initiator caspase-8 is frequently inactivated by DNA methylation and gene deletion [200]. Caspase8-decient neuroblastoma cells are resistant to death receptorand doxorubicin-mediated apoptosis. In certain types of cancer, the proapoptotic Bcl-2 family member Bax is mutated [201203]. Common mutations comprise frameshift mutations leading to loss of expression and mutations in the BH domains resulting in loss of function. Tumor cell lines with frameshift mutations are more resistant to apoptosis. Reduced Bax expression is associated with a poor response rate to chemotherapy and shorter survival in some situations [204]. Several mouse studies conrmed the function
912 Journal of Leukocyte Biology Volume 71, June 2002

of Bax as a tumor suppressor [205207]. However, Bax may be relevant primarily for apoptosis stimuli such as chemotherapy or p53 and not for apoptosis triggered by the immune system. Metastatic melanomas often do not express Apaf-1, which forms an integral part of the apoptosome [208]. A high rate of allelic loss of the Apaf-1 locus can be observed. The remaining allele is transcriptionally inactivated by gene methylation. Apaf-1-negative melanomas fail to respond to chemotherapy, a situation found commonly in this type of tumor. Taken together, tumor cells use many mechanisms to acquire apoptosis resistance. Although a direct role of these mechanisms for immune escape has only been shown in a few studies, it is likely that apoptosis resistance is not only relevant for tumorigenesis and resistance to chemotherapy, but also inuences immunosurveillance and immunotherapy.

TUMOR COUNTERATTACK
Tumor cells may not only resist destruction by the immune system passively. They may also kill tumor-inltrating lymphocytes actively to suppress the anti-tumor immune response, a phenomenon called tumor counterattack [209 211]. The weapon tumors may use to delete CD95-sensitive immune cells is CD95L. Many publications have supported the idea of tumor counterattack as an immune escape mechanism. CD95/CD95L interactions are discussed as being an important mechanism for the maintenance of immune privilege. CD95L is expressed in immune-privileged sites, e.g., the eye and the testis [212214]. Thus, in the eye a high percentage of human corneal transplants are accepted without tissue matching or immunosuppressive therapy. In a mouse model, CD95L-positive corneal allografts were accepted at a rate of about 45%, whereas all grafts from mice with a mutated, nonfunctional CD95L (gld) were rejected [214]. Moreover, inammatory cells entering the anterior chamber of the eye in response to viral infection underwent apoptosis dependent on CD95/CD95L interactions and did not produce any tissue damage [213]. In contrast, viral infection in gld mice, which lack functional CD95L, resulted in inammation and invasion of ocular tissue by cells without signs of apoptosis. CD95-mediated apoptosis of lymphoid cells was necessary for tolerance induction following antigen injection into the anterior chamber of the eye [215]. Data along similar lines were also found with murine testis grafts. Grafts expressing wild-type CD95L survived indenitely when transplanted under the kidney capsule of allogeneic animals, whereas testis grafts from gld mice were rejected [216]. However, attempts to reproduce these observations by another group were not successful [217]. Because neurons and astrocytes also express CD95L [212, 218, 219], immune privilege in the central nervous system may also involve CD95L. These ndings suggest that CD95L may be used to render a transplanted tissue an immune privileged site. Indeed, it was demonstrated that cotransplantation of syngeneic CD95Ltransfected myoblasts protected allogeneic islets of Langerhans grafts from immune rejection [220]. However, other authors have found exactly opposite results using a similar technology [221]. Numerous studies have shown that tumor counterattack
http://www.jleukbio.org

may be a relevant immune escape mechanism of tumors. Many CD95-resistant tumors express CD95L constitutively or after induction by chemotherapy [211, 218, 222227]. Such tumor cells killed CD95-positive, apoptosis-sensitive cells in vitro. Moreover, apoptosis of tumor-inltrating lymphocytes has been found in situ within CD95L-expressing human tumors [186, 224]. In esophageal cancer, the number of inltrated lymphocytes was reduced concomitantly with increased lymphocyte apoptosis within CD95L-expressing areas of the tumors [223]. Various animal models have been used to demonstrate the ability of CD95L expressed on tumors to down-regulate antitumor immune responses. Tumor growth of a subcutaneously injected CD95L-positive murine melanoma cell line was slightly faster in wild-type or gld mice than in mice with a mutated or down-regulated CD95 receptor (lpr mice) [224]. Another study in syngeneic mice showed that growth of tumors of murine CD95L-transfected cells was signicantly better than that of control cells when implanted under the kidney capsule [228]. Immunosuppression in vivo was directly demonstrated in allogeneic mice injected with CD95L-transfected colon carcinoma cells. Alloantibodies were virtually completely abolished and allospecic cytotoxic T lymphocytes and helper T cells were reduced [229]. Despite the wealth of data accumulated in support of the tumor counterattack hypothesis, many contradictory studies have also been published [230]. In contrast to the above ndings, it has been shown that CD95L expression by grafts or by tumor cells targeted the cells for rapid destruction by neutrophils. CD95L expression on pancreatic islets transplanted into allogeneic hosts resulted in a massive inltration of neutrophils and in islet destruction. Similarly, transgenic mice expressing CD95L in pancreatic cells developed a massive inltration of neutrophils and diabetes [231]. CD95Lexpressing islet cells transplanted under the kidney capsule of syngeneic or allogeneic animals were not protected from rejection [232, 233]. In addition, CD95L-expressing hearts from transgenic mice transplanted into sygneneic and allogeneic recipients were more rapidly rejected than control grafts and showed massive neutrophil inltration as early as 1 day after transplantation [234]. Various studies have shown that the overexpression of CD95L in murine tumor cells resistant to CD95-mediated apoptosis did not affect growth in vitro, but caused rejection by neutrophils in vivo [235239]. CD8 T cell-mediated protective immunity against subsequent challenge with the parental tumor cells was elicited. Rejection of the CD95L-expressing tumor has even been observed in the study mentioned above, demonstrating immunosuppression by the tumor in an allogeneic mouse model [229]. In addition, when control tumor cells were co-implanted with the CD95L-expressing cells at the same site, a bystander rejection of the CD95L-negative cells was found [236,237]. Infection of a subcutaneously growing CD95-negative tumor by an adenoviral vector encoding CD95L resulted in rapid elimination of the tumor [240]. These studies indicate that CD95L has a proinammatory function and that gene transfer of CD95L may be used in tumor eradication. Several mechanisms for the recruitment of the graft- or tumor-rejecting neutrophils have been proposed. Two studies suggested that soluble CD95L is directly chemotactic for neu-

trophils implicating that soluble CD95L promotes rejection of CD95L-expressing grafts [241, 242]. However, others have not found a chemotactic activity of soluble CD95L in vitro or in vivo [235, 243]. Moreover, tumor cells expressing only soluble CD95L did not elicit a neutrophilic response [243, 244]. Conversely, tumor cells expressing a noncleavable membranebound form of CD95L were rapidly rejected [243245]. The extent of inammation induced by the various transfectants seemed to correlate with the cytotoxic activity of CD95L. Alternatively, it has been suggested that CD95L acts on surrounding cells to induce the production of granulocyte chemoattractants. Thus, it has been demonstrated that CD95L induces the processing and release of IL-1 , which may be responsible for the inltration by neutrophils [246]. CD95L may act on resident macrophages, leading to increased production of IL-1 and macrophage inammatory proteins [247]. Moreover, engagement of CD95 on dendritic cells may induce the secretion of proinammatory cytokines [248]. These data support an indirect mechanism for the inammatory effect of CD95L. Nevertheless, the exact mechanism of how neutrophils are recruited to CD95L-expressing tumors and how CD95Lexpressing tumors are rejected is still unclear. Many studies of tumor counterattack have been published, but the results are contradictory, and therefore do not clarify whether tumor counterattack is a relevant immune escape mechanism in vivo. No study has demonstrated conclusively that a tumor (or graft), by CD95L expression, deleted antitumor-specic lymphocytes, escaped the immune response, and thus had a growth advantage in vivo. Additional factors may inuence the outcome of CD95L expression on tumors in vivo and thus may account for the controversial situation. Level and time-point of CD95L expression may be particularly relevant. In most animal experiments, CD95L-transfected tumor cell lines have been used. These cells may express different, probably higher levels of CD95L than naturally occurring tumors. Overexpression of CD95L may lead to rejection by neutrophils, whereas physiological levels may not induce neutrophilic inltration but may still sufce to delete anti-tumor lymphocytes. Moreover, tumors growing in situ may express CD95L at late stages of tumorigenesis, e.g., after induction by chemotherapy [249, 250]. Therefore, the time-point of CD95L expression may directly inuence tumor counterattack. Furthermore, the sensitivity of T cells to CD95-mediated apoptosis varies considerably with respect to the activation status of the T cells [84, 89, 251253]. Therefore, it may be crucial when T cells encounter CD95L and how the T cells have been activated and costimulated. The consequences of CD95L expression may also depend on the microenvironment. It has been suggested that TGF- is necessary as a cofactor for promoting immunologic tolerance [254]. CD95L-expressing tumor cells were rejected by neutrophils when injected subcutaneously. When TGF- was simultaneously provided to the subcutaneous sites, protection against tumor rejection was observed. However, it is not clear whether in vivo TGF- and CD95L together are necessary for immune escape and whether CD95L has an immunosuppressive effect on T cells in the presence of TGF- . Despite the controversial situation concerning CD95L and tumor counterattack, other molecules have also been impliIgney and Krammer Apoptosis and tumor immune escape 913

cated in T cell deletion by tumors. Thus, the death ligand TRAIL has been suggested to suppress tumor-specic T cell responses in a similar manner as CD95L [255]. Moreover, the membrane protein RCAS1 [67] not only inhibits proliferation, but also induces apoptosis in T cells in vitro. Chemokine production by tumor cells may also sensitize T cells to apoptosis [256].

CONCLUSIONS
Research in tumor immunology has provided a wealth of information about the interactions between tumors and the immune system. Many of these interactions are now not only known on a cellular, but also on a molecular level. Despite this knowledge, cancer immunotherapy still is not an established treatment in the clinic. Many approaches may fail because tumors use multiple mechanisms to become resistant to apoptosis or to counterattack the immune system. These mechanisms render tumor cells insensitive to the effector mechanisms of the immune system, yet the signicance for immune escape has only been shown in few studies. It is the present aim of research in this area to understand these events further and to use this insight to resensitize tumor cells to apoptosis. Recently, it has been shown that low doses of chemotherapy or irradiation sensitized resistant cells to TRAIL-induced apoptosis in vitro and in vivo [257, 258]. Therefore, these treatments might enhance the susceptibility of a tumor to immune attack. A future therapeutic strategy may also involve down-regulation of antiapoptotic molecules such as FLIP or Bcl-2 by antisense oligonucleotides or dsRNA interference [259, 260]. However, a macroscopic tumor is heterogeneous, and different cells within the tumor may also use different immune escape mechanisms including apoptosis resistance, impaired antigen presentation, secretion of immunosuppressive factors, and other strategies (Table 1). Moreover, multiple mechanisms may develop in a single tumor cell. Therefore, it is questionable whether a single, predominant immune-escape mechanism can be identied in a tumor and whether therapeutic targeting of one mechanism alone is promising. Deeper insight into the molecular mechanisms underlying tumor immune escape may nally lead to novel therapeutic approaches that will be used for the benet of cancer patients.

ACKNOWLEDGMENTS
This work was supported by grants from the Deutsche Forschungsgemeinschaft, the European Community, the Deutsche Krebshilfe, the Sander Stiftung, the Tumor Center Heidelberg/ Mannheim, the BMBF, and the German-Israeli Cooperation in Cancer Research.

REFERENCES
1. Smyth, M. J., Godfrey, D. I., Trapani, J. A. (2001) A fresh look at tumor immunosurveillance and immunotherapy. Nat. Immunol. 2, 293299.

2. Sogn, J. A. (1998) Tumor immunology: the glass is half full. Immunity 9, 757763. 3. Rosenberg, S. A. (2001) Progress in human tumour immunology and immunotherapy. Nature 411, 380 384. 4. Houghton, A. N., Gold, J. S., Blachere, N. E. (2001) Immunity against cancer: lessons learned from melanoma. Curr. Opin. Immunol. 13, 134 140. 5. Boon, T., van der Bruggen, P. (1996) Human tumor antigens recognized by T lymphocytes. J. Exp. Med. 183, 725729. 6. Lee, P. P., Yee, C., Savage, P. A., Fong, L., Brockstedt, D., Weber, J. S., Johnson, D., Swetter, S., Thompson, J., Greenberg, P. D., Roederer, M., Davis, M. M. (1999) Characterization of circulating T cells specic for tumor-associated antigens in melanoma patients. Nat. Med. 5, 677 685. 7. Wortzel, R. D., Philipps, C., Schreiber, H. (1983) Multiple tumourspecic antigens expressed on a single tumour cell. Nature 304, 165 167. 8. Burnet, F. M. (1970) The concept of immunological surveillance. Prog. Exp. Tumor Res. 13, 127. 9. Fuchs, E. J., Matzinger, P. (1996) Is cancer dangerous to the immune system? Semin. Immunol. 8, 271280. 10. Watzl, C., Long, E. O. (2000) Exposing tumor cells to killer cell attack. Nat. Med. 6, 867 868. 11. Pardoll, D. M. (2001) Immunology. Stress, NK receptors, and immune surveillance. Science 294, 534 536. 12. Moretta, A., Biassoni, R., Bottino, C., Mingari, M. C., Moretta, L. (2000) Natural cytotoxicity receptors that trigger human NK-cell-mediated cytolysis. Immunol. Today 21, 228 234. 13. Cerwenka, A., Bakker, A. B., McClanahan, T., Wagner, J., Wu, J., Phillips, J. H., Lanier, L. L. (2000) Retinoic acid early inducible genes dene a ligand family for the activating NKG2D receptor in mice. Immunity 12, 721727. 14. Bauer, S., Groh, V., Wu, J., Steinle, A., Phillips, J. H., Lanier, L. L., Spies, T. (1999) Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 285, 727729. 15. Wu, J., Song, Y., Bakker, A. B., Bauer, S., Spies, T., Lanier, L. L., Phillips, J. H. (1999) An activating immunoreceptor complex formed by NKG2D and DAP10. Science 285, 730 732. 16. Lollini, P. L., Forni, G. (1999) Specic and nonspecic immunity in the prevention of spontaneous tumours. Immunol Today 20, 347350. 17. Di Carlo, E., Forni, G., Lollini, P., Colombo, M. P., Modesti, A., Musiani, P. (2001) The intriguing role of polymorphonuclear neutrophils in antitumor reactions. Blood 97, 339 345. 18. Elgert, K. D., Alleva, D. G., Mullins, D. W. (1998) Tumor-induced immune dysfunction: the macrophage connection. J. Leukoc. Biol. 64, 275290. 19. Bonnotte, B., Larmonier, N., Favre, N., Fromentin, A., Moutet, M., Martin, M., Gurbuxani, S., Solary, E., Chauffert, B., Martin, F. (2001) Identication of tumor-inltrating macrophages as the killers of tumor cells after immunization in a rat model system. J. Immunol. 167, 5077 5083. 20. Lin, E. Y., Nguyen, A. V., Russell, R. G., Pollard, J. W. (2001) Colonystimulating factor 1 promotes progression of mammary tumors to malignancy. J. Exp. Med. 193, 727740. 21. Chen, L. (1998) Immunological ignorance of silent antigens as an explanation of tumor evasion. Immunol. Today 19, 2730. 22. Ochsenbein, A. F., Klenerman, P., Karrer, U., Ludewig, B., Pericin, M., Hengartner, H., Zinkernagel, R. M. (1999) Immune surveillance against a solid tumor fails because of immunological ignorance. Proc. Natl. Acad. Sci. USA 96, 22332238. 23. Ochsenbein, A. F., Sierro, S., Odermatt, B., Pericin, M., Karrer, U., Hermans, J., Hemmi, S., Hengartner, H., Zinkernagel, R. M. (2001) Roles of tumour localization, second signals and cross priming in cytotoxic T-cell induction. Nature 411, 1058 1064. 24. Rocha, G., Deschenes, J., Rowsey, J. J. (1998) The immunology of corneal graft rejection. Crit. Rev. Immunol. 18, 305325. 25. Onrust, S. V., Hartl, P. M., Rosen, S. D., Hanahan, D. (1996) Modulation of L-selectin ligand expression during an immune response accompanying tumorigenesis in transgenic mice. J. Clin. Investig. 97, 54 64. 26. Piali, L., Fichtel, A., Terpe, H. J., Imhof, B. A., Gisler, R. H. (1995) Endothelial vascular cell adhesion molecule 1 expression is suppressed by melanoma and carcinoma. J. Exp. Med. 181, 811 816. 27. Ishido, S., Choi, J. K., Lee, B. S., Wang, C., DeMaria, M., Johnson, R. P., Cohen, G. B., Jung, J. U. (2000) Inhibition of natural killer cell-mediated cytotoxicity by Kaposis sarcoma-associated herpesvirus K5 protein. Immunity 13, 365374. 28. Singh, S., Ross, S. R., Acena, M., Rowley, D. A., Schreiber, H. (1992) Stroma is critical for preventing or permitting immunological destruction of antigenic cancer cells. J. Exp. Med. 175, 139 146.

914

Journal of Leukocyte Biology Volume 71, June 2002

http://www.jleukbio.org

29. Wick, M., Dubey, P., Koeppen, H., Siegel, C. T., Fields, P. E., Chen, L., Bluestone, J. A., Schreiber, H. (1997) Antigenic cancer cells grow progressively in immune hosts without evidence for T cell exhaustion or systemic anergy. J. Exp. Med. 186, 229 238. 30. Antonia, S. J., Extermann, M., Flavell, R. A. (1998) Immunologic nonresponsiveness to tumors. Crit. Rev. Oncog. 9, 35 41. 31. Villunger, A., Strasser, A. (1999) The great escape: is immune evasion required for tumor progression? Nat. Med. 5, 874 875. 32. Gilboa, E. (1999) How tumors escape immune destruction and what we can do about it. Cancer Immunol. Immunother. 48, 382385. 33. Maeurer, M. J., Gollin, S. M., Martin, D., Swaney, W., Bryant, J., Castelli, C., Robbins, P., Parmiani, G., Storkus, W. J., Lotze, M. T. (1996) Tumor escape from immune recognition: lethal recurrent melanoma in a patient associated with downregulation of the peptide transporter protein TAP-1 and loss of expression of the immunodominant MART-1/Melan-A antigen. J. Clin. Investig. 98, 16331641. 34. Kim, U., Baumler, A., Carruthers, C., Bielat, K. (1975) Immunological escape mechanism in spontaneously metastasizing mammary tumors. Proc. Natl. Acad. Sci. USA 72, 10121016. 35. Vasmel, W. L., Sijts, E. J., Leupers, C. J., Matthews, E. A., Melief, C. J. (1989) Primary virus-induced lymphomas evade T cell immunity by failure to express viral antigens. J. Exp. Med. 169, 12331254. 36. Stackpole, C. W., Cremona, P., Leonard, C., Stremmel, P. (1980) Antigenic modulation as a mechanism for tumor escape from immune destruction: identication of modulation-positive and modulation-negative mouse lymphomas with xenoantisera to murine leukemia virus gp70. J. Immunol. 125, 17151723. 37. Hicklin, D. J., Marincola, F. M., Ferrone, S. (1999) HLA class I antigen downregulation in human cancers: T-cell immunotherapy revives an old story. Mol. Med. Today 5, 178 186. 38. Garrido, F., Ruiz-Cabello, F., Cabrera, T., Perez-Villar, J. J., LopezBotet, M., Duggan-Keen, M., Stern, P. L. (1997) Implications for immunosurveillance of altered HLA class I phenotypes in human tumours. Immunol. Today 18, 89 95. 39. Bicknell, D. C., Rowan, A., Bodmer, W. F. (1994) Beta 2-microglobulin gene mutations: a study of established colorectal cell lines and fresh tumors. Proc. Natl. Acad. Sci. USA 91, 4751 4756. 40. Seung, S., Urban, J. L., Schreiber, H. (1993) A tumor escape variant that has lost one major histocompatibility complex class I restriction element induces specic CD8 T cells to an antigen that no longer serves as a target. J. Exp. Med. 178, 933940. 41. Natali, P. G., Nicotra, M. R., Bigotti, A., Venturo, I., Marcenaro, L., Giacomini, P., Russo, C. (1989) Selective changes in expression of HLA class I polymorphic determinants in human solid tumors. Proc. Natl. Acad. Sci. USA 86, 6719 6723. 42. Blanchet, O., Bourge, J. F., Zinszner, H., Israel, A., Kourilsky, P., Dausset, J., Degos, L., Paul, P. (1992) Altered binding of regulatory factors to HLA class I enhancer sequence in human tumor cell lines lacking class I antigen expression. Proc. Natl. Acad. Sci. USA 89, 3488 3492. 43. Bosshart, H., Jarrett, R. F. (1998) Decient major histocompatibility complex class II antigen presentation in a subset of Hodgkins disease tumor cells. Blood 92, 22522259. 44. Farrell, H. E., Vally, H., Lynch, D. M., Fleming, P., Shellam, G. R., Scalzo, A. A., Davis-Poynter, N. J. (1997) Inhibition of natural killer cells by a cytomegalovirus MHC class I homologue in vivo. Nature 386, 510 514. 45. Cretney, E., Degli-Esposti, M. A., Densley, E. H., Farrell, H. E., DavisPoynter, N. J., Smyth, M. J. (1999) m144, a murine cytomegalovirus (MCMV)-encoded major histocompatibility complex class I homologue, confers tumor resistance to natural killer cell-mediated rejection. J. Exp. Med. 190, 435 444. 46. Seliger, B., Maeurer, M. J., Ferrone, S. (1997) TAP offtumors on. Immunol. Today 18, 292299. 47. Restifo, N. P., Esquivel, F., Kawakami, Y., Yewdell, J. W., Mule, J. J., Rosenberg, S. A., Bennink, J. R. (1993) Identication of human cancers decient in antigen processing. J. Exp. Med. 177, 265272. 48. Rotem-Yehudar, R., Groettrup, M., Soza, A., Kloetzel, P. M., Ehrlich, R. (1996) LMP-associated proteolytic activities and TAP-dependent peptide transport for class 1 MHC molecules are suppressed in cell lines transformed by the highly oncogenic adenovirus 12. J. Exp. Med. 183, 499 514. 49. Cromme, F. V., Airey, J., Heemels, M. T., Ploegh, H. L., Keating, P. J., Stern, P. L., Meijer, C. J., Walboomers, J. M. (1994) Loss of transporter protein, encoded by the TAP-1 gene, is highly correlated with loss of HLA expression in cervical carcinomas. J. Exp. Med. 179, 335340. 50. Johnsen, A. K., Templeton, D. J., Sy, M., Harding, C. V. (1999) Deciency of transporter for antigen presentation (TAP) in tumor cells allows

51.

52.

53.

54. 55.

56.

57.

58.

59.

60.

61.

62.

63.

64.

65.

66. 67.

68.

69. 70.

evasion of immune surveillance and increases tumorigenesis. J. Immunol. 163, 4224 4231. Ossendorp, F., Eggers, M., Neisig, A., Ruppert, T., Groettrup, M., Sijts, A., Mengede, E., Kloetzel, P. M., Neefjes, J., Koszinowski, U., Melief, C. (1996) A single residue exchange within a viral CTL epitope alters proteasome-mediated degradation resulting in lack of antigen presentation. Immunity 5, 115124. Chouaib, S., Asselin-Paturel, C., Mami-Chouaib, F., Caignard, A., Blay, J. Y. (1997) The host-tumor immune conict: from immunosuppression to resistance and destruction. Immunol. Today 18, 493 497. Loefer, C. M., Smyth, M. J., Longo, D. L., Kopp, W. C., Harvey, L. K., Tribble, H. R., Tase, J. E., Urba, W. J., Leonard, A. S., Young, H. A., et. al. (1992) Immunoregulation in cancer-bearing hosts. Down-regulation of gene expression and cytotoxic function in CD8 T cells. J. Immunol. 149, 949 956. Pasche, B. (2001) Role of transforming growth factor beta in cancer. J. Cell. Physiol. 186, 153168. Gorelik, L., Flavell, R. A. (2001) Immune-mediated eradication of tumors through the blockade of transforming growth factor-beta signaling in T cells. Nat. Med. 7, 1118 1122. Tada, T., Ohzeki, S., Utsumi, K., Takiuchi, H., Muramatsu, M., Li, X. F., Shimizu, J., Fujiwara, H., Hamaoka, T. (1991) Transforming growth factor-beta-induced inhibition of T cell function. Susceptibility difference in T cells of various phenotypes and functions and its relevance to immunosuppression in the tumor-bearing state. J. Immunol. 146, 1077 1082. Ranges, G. E., Figari, I. S., Espevik, T., Palladino Jr., M. A. (1987) Inhibition of cytotoxic T cell development by transforming growth factor beta and reversal by recombinant tumor necrosis factor alpha. J. Exp. Med. 166, 991998. Torre-Amione, G., Beauchamp, R. D., Koeppen, H., Park, B. H., Schreiber, H., Moses, H. L., Rowley, D. A. (1990) A highly immunogenic tumor transfected with a murine transforming growth factor type beta 1 cDNA escapes immune surveillance. Proc. Natl. Acad. Sci. USA 87, 1486 1490. Gabrilovich, D. I., Chen, H. L., Girgis, K. R., Cunningham, H. T., Meny, G. M., Nadaf, S., Kavanaugh, D., Carbone, D. P. (1996) Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat. Med. 2, 1096 1103. Plescia, O. J., Smith, A. H., Grinwich, K. (1975) Subversion of immune system by tumor cells and role of prostaglandins. Proc. Natl. Acad. Sci. USA 72, 1848 1851. Young, M. R., Knies, S. (1984) Prostaglandin E production by Lewis lung carcinoma: mechanism for tumor establishment in vivo. J. Natl. Cancer Inst. 72, 919 922. McLemore, T. L., Hubbard, W. C., Litterst, C. L., Liu, M. C., Miller, S., McMahon, N. A., Eggleston, J. C.,Boyd, M. R. (1988) Proles of prostaglandin biosynthesis in normal lung and tumor tissue from lung cancer patients. Cancer Res. 48, 3140 3147. Matsuda, M., Salazar, F., Petersson, M., Masucci, G., Hansson, J., Pisa, P., Zhang, Q. J., Masucci, M. G., Kiessling, R. (1994) Interleukin 10 pretreatment protects target cells from tumor- and allo-specic cytotoxic T cells and downregulates HLA class I expression. J. Exp. Med. 180, 23712376. Oghiso, Y., Yamada, Y., Ando, K., Ishihara, H., Shibata, Y. (1993) Differential induction of prostaglandin E2-dependent and -independent immune suppressor cells by tumor-derived GM-CSF and M-CSF. J. Leukoc. Biol. 53, 86 92. Sotomayor, E. M., Fu, Y. X., Lopez-Cepero, M., Herbert, L., Jimenez, J. J., Albarracin, C., Lopez, D. M. (1991) Role of tumor-derived cytokines on the immune system of mice bearing a mammary adenocarcinoma. II. Down-regulation of macrophage-mediated cytotoxicity by tumor-derived granulocyte-macrophage colony-stimulating factor. J. Immunol. 147, 2816 2823. McKallip, R., Li, R., Ladisch, S. (1999) Tumor gangliosides inhibit the tumor-specic immune response. J. Immunol. 163, 3718 3726. Nakashima, M., Sonoda, K., Watanabe, T. (1999) Inhibition of cell growth and induction of apoptotic cell death by the human tumorassociated antigen RCAS1. Nat. Med. 5, 938 942. Kiessling, R., Wasserman, K., Horiguchi, S., Kono, K., Sjoberg, J., Pisa, P., Petersson, M. (1999) Tumor-induced immune dysfunction. Cancer Immunol. Immunother. 48, 353362. Arnold, B., Schonrich, G., Hammerling, G. J. (1993) Multiple levels of peripheral tolerance. Immunol. Today 14, 1214. Ganss, R., Limmer, A., Sacher, T., Arnold, B., Hammerling, G. J. (1999) Autoaggression and tumor rejection: it takes more than self-specic T-cell activation. Immunol. Rev. 169, 263272.

Igney and Krammer Apoptosis and tumor immune escape

915

71. Chen, L., Ashe, S., Brady, W. A., Hellstrom, I., Hellstrom, K. E., Ledbetter, J. A., McGowan, P., Linsley, P. S. (1992) Costimulation of antitumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-4. Cell 71, 10931102. 72. Guinan, E. C., Gribben, J. G., Boussiotis, V. A., Freeman, G. J., Nadler, L. M. (1994) Pivotal role of the B7:CD28 pathway in transplantation tolerance and tumor immunity. Blood 84, 32613282. 73. Staveley-OCarroll, K., Sotomayor, E., Montgomery, J., Borrello, I., Hwang, L., Fein, S., Pardoll, D., Levitsky, H. (1998) Induction of antigen-specic T cell anergy: an early event in the course of tumor progression. Proc. Natl. Acad. Sci. USA 95, 1178 1183. 74. Becker, J. C., Brabletz, T., Czerny, C., Termeer, C., Brocker, E. B. (1993) Tumor escape mechanisms from immunosurveillance: induction of unresponsiveness in a specic MHC-restricted CD4 human T cell clone by the autologous MHC class II melanoma. Int. Immunol. 5, 1501 1508. 75. Shrikant, P., Khoruts, A., Mescher, M. F. (1999) CTLA-4 blockade reverses CD8 T cell tolerance to tumor by a CD4 T cell- and IL-2-dependent mechanism. Immunity 11, 483 493. 76. Maeda, H., Shiraishi, A. (1996) TGF-beta contributes to the shift toward Th2-type responses through direct and IL-10-mediated pathways in tumor-bearing mice. J. Immunol. 156, 7378. 77. Qin, Z., Richter, G., Schuler, T., Ibe, S., Cao, X., Blankenstein, T. (1998) B cells inhibit induction of T cell-dependent tumor immunity. Nat. Med. 4, 627 630. 78. Sakaguchi, S. (2000) Regulatory T cells: key controllers of immunologic self-tolerance. Cell 101, 455 458. 79. Matsui, S., Ahlers, J. D., Vortmeyer, A. O., Terabe, M., Tsukui, T., Carbone, D. P., Liotta, L. A., Berzofsky, J. A. (1999) A model for CD8 CTL tumor immunosurveillance and regulation of tumor escape by CD4 T cells through an effect on quality of CTL. J. Immunol. 163, 184 193. 80. Takahashi, K., Ono, K., Hirabayashi, Y., Taniguchi, M. (1988) Escape mechanisms of melanoma from immune system by soluble melanoma antigen. J. Immunol. 140, 3244 3248. 81. Russell, J. H., White, C. L., Loh, D. Y., Meleedy-Rey, P. (1991) Receptor-stimulated death pathway is opened by antigen in mature T cells. Proc. Natl. Acad. Sci. USA 88, 21512155. 82. Radvanyi, L. G., Mills, G. B., Miller, R. G. (1993) Religation of the T cell receptor after primary activation of mature T cells inhibits proliferation and induces apoptotic cell death. J. Immunol. 150, 5704 5715. 83. Martin, D. R., Miller, R. G. (1989) In vivo administration of histoincompatible lymphocytes leads to rapid functional deletion of cytotoxic T lymphocyte precursors. J. Exp. Med. 170, 679 690. 84. Kirchhoff, S., Muller, W. W., Krueger, A., Schmitz, I., Krammer, P. H. (2000) TCR-mediated up-regulation of c-FLIPshort correlates with resistance toward CD95-mediated apoptosis by blocking death-inducing signaling complex activity. J. Immunol. 165, 6293 6300. 85. Singer, G. G., Abbas, A. K. (1994) The fas antigen is involved in peripheral but not thymic deletion of T lymphocytes in T cell receptor transgenic mice. Immunity 1, 365371. 86. Dhein, J., Walczak, H., Baumler, C., Debatin, K. M., Krammer, P. H. (1995) Autocrine T-cell suicide mediated by APO-1/(Fas/CD95). Nature 373, 438 441. 87. Brunner, T., Mogil, R. J., LaFace, D., Yoo, N. J., Mahboubi, A., Echeverri, F., Martin, S. J., Force, W. R., Lynch, D. H., Ware, C. F., et al. (1995) Cell-autonomous Fas (CD95)/Fas-ligand interaction mediates activation-induced apoptosis in T-cell hybridomas. Nature 373, 441 444. 88. Li-Weber, M., Laur, O., Hekele, A., Coy, J., Walczak, H., Krammer, P. H. (1998) A regulatory element in the CD95 (APO-1/Fas) ligand promoter is essential for responsiveness to TCR-mediated activation. Eur. J. Immunol. 28, 23732383. 89. Kirchhoff, S., Muller, W. W., Li-Weber, M., Krammer, P. H. (2000) Up-regulation of c-FLIPshort and reduction of activation-induced cell death in CD28-costimulated human T cells. Eur. J. Immunol. 30, 2765 2774. 90. Bogen, B. (1996) Peripheral T cell tolerance as a tumor escape mechanism: deletion of CD4 T cells specic for a monoclonal immunoglobulin idiotype secreted by a plasmacytoma. Eur. J. Immunol. 26, 2671 2679. 91. Lauritzsen, G. F., Hofgaard, P. O., Schenck, K., Bogen, B. (1998) Clonal deletion of thymocytes as a tumor escape mechanism. Int. J. Cancer 78, 216 222. 92. Shresta, S., Pham, C. T., Thomas, D. A., Graubert, T. A., Ley, T. J. (1998) How do cytotoxic lymphocytes kill their targets? Curr. Opin. Immunol. 10, 581587. 93. Rouvier, E., Luciani, M. F., Golstein, P. (1993) Fas involvement in Ca(2 )-independent T cell-mediated cytotoxicity. J. Exp. Med. 177, 195200.

94. Walsh, C. M., Glass, A. A., Chiu, V., Clark, W. R. (1994) The role of the Fas lytic pathway in a perforin-less CTL hybridoma. J. Immunol. 153, 2506 2514. 95. Li, J. H., Rosen, D., Ronen, D., Behrens, C. K., Krammer, P. H., Clark, W. R., Berke, G. (1998) The regulation of CD95 ligand expression and function in CTL. J. Immunol. 161, 39433949. 96. Smyth, M. J., Cretney, E., Takeda, K., Wiltrout, R. H., Sedger, L. M., Kayagaki, N., Yagita, H., Okumura, K. (2001) Tumor necrosis factorrelated apoptosis-inducing ligand (TRAIL) contributes to interferon gammadependent natural killer cell protection from tumor metastasis. J. Exp. Med. 193, 661 670. 97. Takeda, K., Hayakawa, Y., Smyth, M. J., Kayagaki, N., Yamaguchi, N., Kakuta, S., Iwakura, Y., Yagita, H., Okumura, K. (2001) Involvement of tumor necrosis factor-related apoptosis-inducing ligand in surveillance of tumor metastasis by liver natural killer cells. Nat. Med. 7, 94 100. 98. Takeda, K., Smyth, M. J., Cretney, E., Hayakawa, Y., Kayagaki, N., Yagita, H., Okumura, K. (2002) Critical role for tumor necrosis factorrelated apoptosis-inducing ligand in immune surveillance against tumor development. J. Exp. Med. 195, 161169. 99. Krammer, P. H. (1999) CD95(APO-1/Fas)-mediated apoptosis: live and let die. Adv. Immunol. 71, 163210. 100. Schmitz, I., Kirchhoff, S., Krammer, P. H. (2000) Regulation of death receptor-mediated apoptosis pathways. Int. J. Biochem. Cell Biol. 32, 11231136. 101. Pitti, R. M., Marsters, S. A., Lawrence, D. A., Roy, M., Kischkel, F. C., Dowd, P., Huang, A., Donahue, C. J., Sherwood, S. W., Baldwin, D. T., Godowski, P. J., Wood, W. I., Gurney, A. L., Hillan, K. J., Cohen, R. L., Goddard, A. D., Botstein, D., Ashkenazi, A. (1998) Genomic amplication of a decoy receptor for Fas ligand in lung and colon cancer. Nature 396, 699 703. 102. Yu, K. Y., Kwon, B., Ni, J., Zhai, Y., Ebner, R., Kwon, B. S. (1999) A newly identied member of tumor necrosis factor receptor superfamily (TR6) suppresses LIGHT-mediated apoptosis. J. Biol. Chem. 274, 1373313736. 103. Kischkel, F. C., Hellbardt, S., Behrmann, I., Germer, M., Pawlita, M., Krammer, P. H., Peter, M. E. (1995) Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO J. 14, 5579 5588. 104. Muzio, M., Chinnaiyan, A. M., Kischkel, F. C., ORourke, K., Shevchenko, A., Ni, J., Scafdi, C., Bretz, J. D., Zhang, M., Gentz, R., Mann, M., Krammer, P. H., Peter, M. E., Dixit, V. M. (1996) FLICE, a novel FADD-homologous ICE/CED-3-like protease, is recruited to the CD95 (Fas/APO-1) deathinducing signaling complex. Cell 85, 817 827. 105. Sprick, M. R., Weigand, M. A., Rieser, E., Rauch, C. T., Juo, P., Blenis, J., Krammer, P. H., Walczak, H. (2000) FADD/MORT1 and caspase-8 are recruited to TRAIL receptors 1 and 2 and are essential for apoptosis mediated by TRAIL receptor 2. Immunity 12, 599 609. 106. Kischkel, F. C., Lawrence, D. A., Chuntharapai, A., Schow, P., Kim, K. J., Ashkenazi, A. (2000) Apo2L/TRAIL-dependent recruitment of endogenous FADD and caspase-8 to death receptors 4 and 5. Immunity 12, 611 620. 107. Kischkel, F. C., Lawrence, D. A., Tinel, A., LeBlanc, H., Virmani, A., Schow, P., Gazdar, A., Blenis, J., Arnott, D., Ashkenazi, A. (2001) Death receptor recruitment of endogenous caspase-10 and apoptosis initiation in the absence of caspase-8. J. Biol. Chem. 276, 46639 46646. 108. Scafdi, C., Fulda, S., Srinivasan, A., Friesen, C., Li, F., Tomaselli, K. J., Debatin, K. M., Krammer, P. H., Peter, M. E. (1998) Two CD95 (APO1/Fas) signaling pathways. EMBO J. 17, 16751687. 109. Zamzami, N., Kroemer, G. (2001) The mitochondrion in apoptosis: how Pandoras box opens. Nat. Rev. Mol. Cell. Biol. 2, 6771. 110. Martinou, J. C., Green, D. R. (2001) Breaking the mitochondrial barrier. Nat. Rev. Mol. Cell. Biol. 2, 63 67. 111. Rathmell, J. C., Thompson, C. B. (1999) The central effectors of cell death in the immune system. Annu. Rev. Immunol. 17, 781 828. 112. Krueger, A., Baumann, S., Krammer, P. H., Kirchhoff, S. (2001) FLICEinhibitory proteins: regulators of death receptor-mediated apoptosis. Mol. Cell. Biol. 21, 8247 8254. 113. Deveraux, Q. L., Reed, J. C. (1999) IAP family proteinssuppressors of apoptosis. Genes Dev. 13, 239 252. 114. Verhagen, A. M., Ekert, P. G., Pakusch, M., Silke, J., Connolly, L. M., Reid, G. E., Moritz, R. L., Simpson, R. J., Vaux, D. L. (2000) Identication of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 102, 4353. 115. Du, C., Fang, M., Li, Y., Li, L., Wang, X. (2000) Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 102, 33 42.

916

Journal of Leukocyte Biology Volume 71, June 2002

http://www.jleukbio.org

116. van den Broek, M. E., Kagi, D., Ossendorp, F., Toes, R., Vamvakas, S., Lutz, W. K., Melief, C. J., Zinkernagel, R. M., Hengartner, H. (1996) Decreased tumor surveillance in perforin-decient mice. J. Exp. Med. 184, 17811790. 117. Kagi, D., Ledermann, B., Burki, K., Seiler, P., Odermatt, B., Olsen, K. J., Podack, E. R., Zinkernagel, R. M., Hengartner, H. (1994) Cytotoxicity mediated by T cells and natural killer cells is greatly impaired in perforin-decient mice. Nature 369, 3137. 118. Lowin, B., Beermann, F., Schmidt, A., Tschopp, J. (1994) A null mutation in the perforin gene impairs cytolytic T lymphocyte- and natural killer cell-mediated cytotoxicity. Proc. Natl. Acad. Sci. USA 91, 11571 11575. 119. Heusel, J. W., Wesselschmidt, R. L., Shresta, S., Russell, J. H., Ley, T. J. (1994) Cytotoxic lymphocytes require granzyme B for the rapid induction of DNA fragmentation and apoptosis in allogeneic target cells. Cell 76, 977987. 120. Sarin, A., Williams, M. S., Alexander-Miller, M. A., Berzofsky, J. A., Zacharchuk, C. M., Henkart, P. A. (1997) Target cell lysis by CTL granule exocytosis is independent of ICE/Ced-3 family proteases. Immunity 6, 209 215. 121. Medema, J. P., Toes, R. E., Scafdi, C., Zheng, T. S., Flavell, R. A., Melief, C. J., Peter, M. E., Offringa, R., Krammer, P. H. (1997) Cleavage of FLICE (caspase-8) by granzyme B during cytotoxic T lymphocyteinduced apoptosis. Eur. J. Immunol. 27, 34923498. 122. Heibein, J. A., Goping, I. S., Barry, M., Pinkoski, M. J., Shore, G. C., Green, D. R., Bleackley, R. C. (2000) Granzyme B-mediated cytochrome c release is regulated by the Bcl-2 family members Bid and Bax. J. Exp. Med. 192, 13911402. 123. van den Broek, M. E., Kagi, D., Ossendorp, F., Toes, R., Vamvakas, S., Lutz, W. K., Melief, C. J., Zinkernagel, R. M., Hengartner, H. (1996) Decreased tumor surveillance in perforin-decient mice. J. Exp. Med. 184, 17811790. 124. Smyth, M. J., Thia, K. Y., Street, S. E., MacGregor, D., Godfrey, D. I., Trapani, J. A. (2000) Perforin-mediated cytotoxicity is critical for surveillance of spontaneous lymphoma. J. Exp. Med. 192, 755760. 125. Mullbacher, A., Waring, P., Tha Hla, R., Tran, T., Chin, S., Stehle, T., Museteanu, C., Simon, M. M. (1999) Granzymes are the essential downstream effector molecules for the control of primary virus infections by cytolytic leukocytes. Proc. Natl. Acad. Sci. USA 96, 13950 13955. 126. Davis, J. E., Smyth, M. J., Trapani, J. A. (2001) Granzyme A- and B-decient killer lymphocytes are defective in eliciting DNA fragmentation but retain potent in vivo anti-tumor capacity. Eur. J. Immunol. 31, 39 47. 127. Witko-Sarsat, V., Rieu, P., Descamps-Latscha, B., Lesavre, P., Halbwachs-Mecarelli, L. (2000) Neutrophils: molecules, functions and pathophysiological aspects. Lab. Investig. 80, 617 653. 128. Babior, B. M. (1999) NADPH oxidase: an update. Blood 93, 1464 1476. 129. Bastian, N. R., Hibbs Jr., J. B. (1994) Assembly and regulation of NADPH oxidase and nitric oxide synthase. Curr. Opin. Immunol. 6, 131139. 130. Hampton, M. B., Kettle, A. J., Winterbourn, C. C. (1998) Inside the neutrophil phagosome: oxidants, myeloperoxidase, and bacterial killing. Blood 92, 30073017. 131. Bogdan, C., Rollinghoff, M., Diefenbach, A. (2000) The role of nitric oxide in innate immunity. Immunol. Rev. 173, 1726. 132. Gudmundsson, G. H., Agerberth, B. (1999) Neutrophil antibacterial peptides, multifunctional effector molecules in the mammalian immune system. J. Immunol. Methods 232, 4554. 133. Shankaran, V., Ikeda, H., Bruce, A. T., White, J. M., Swanson, P. E., Old, L. J., Schreiber, R. D. (2001) IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 410, 11071111. 134. Davidson, W. F., Giese, T., Fredrickson, T. N. (1998) Spontaneous development of plasmacytoid tumors in mice with defective Fas-Fas ligand interactions. J. Exp. Med. 187, 18251838. 135. Irmler, M., Thome, M., Hahne, M., Schneider, P., Hofmann, K., Steiner, V., Bodmer, J. L., Schroter, M., Burns, K., Mattmann, C., Rimoldi, D., French, L. E., Tschopp, J. (1997) Inhibition of death receptor signals by cellular FLIP. Nature 388, 190 195. 136. Kataoka, T., Schroter, M., Hahne, M., Schneider, P., Irmler, M., Thome, M., Froelich, C. J., Tschopp, J. (1998) FLIP prevents apoptosis induced by death receptors but not by perforin/granzyme B, chemotherapeutic drugs, and gamma irradiation. J. Immunol. 161, 3936 3942. 137. Medema, J. P., de Jong, J., van Hall, T., Melief, C. J., Offringa, R. (1999) Immune escape of tumors in vivo by expression of cellular FLICEinhibitory protein. J. Exp. Med. 190, 10331038. 138. Taylor, M. A., Chaudhary, P. M., Klem, J., Kumar, V., Schatzle, J. D., Bennett, M. (2001) Inhibition of the death receptor pathway by cFLIP

139.

140.

141.

142.

143.

144.

145.

146.

147. 148.

149.

150.

151.

152. 153.

154.

155.

156.

157.

158.

159.

confers partial engraftment of MHC class I-decient stem cells and reduces tumor clearance in perforin-decient mice. J. Immunol. 167, 4230 4237. Grifth, T. S., Chin, W. A., Jackson, G. C., Lynch, D. H., Kubin, M. Z. (1998) Intracellular regulation of TRAIL-induced apoptosis in human melanoma cells. J. Immunol. 161, 28332840. Mueller, C. M., Scott, D. W. (2000) Distinct molecular mechanisms of Fas resistance in murine B lymphoma cells. J. Immunol. 165, 1854 1862. Tepper, C. G., Seldin, M. F. (1999) Modulation of caspase-8 and FLICEinhibitory protein expression as a potential mechanism of Epstein-Barr virus tumorigenesis in Burkitts lymphoma. Blood 94, 17271737. Thome, M., Schneider, P., Hofmann, K., Fickenscher, H., Meinl, E., Neipel, F., Mattmann, C., Burns, K., Bodmer, J. L., Schroter, M., Scafdi, C., Krammer, P. H., Peter, M. E., Tschopp, J. (1997) Viral FLICEinhibitory proteins (FLIPs) prevent apoptosis induced by death receptors. Nature 386, 517521. Hu, S., Vincenz, C., Ni, J., Gentz, R., Dixit, V. M. (1997) I-FLICE, a novel inhibitor of tumor necrosis factor receptor-1- and CD-95-induced apoptosis. J. Biol. Chem. 272, 1725517257. Bertin, J., Armstrong, R. C., Ottilie, S., Martin, D. A., Wang, Y., Banks, S., Wang, G. H., Senkevich, T. G., Alnemri, E. S., Moss, B., Lenardo, M. J., Tomaselli, K. J., Cohen, J. I. (1997) Death effector domaincontaining herpesvirus and poxvirus proteins inhibit both Fas- and TNFR1-induced apoptosis. Proc. Natl. Acad. Sci. USA 94, 11721176. Djerbi, M., Screpanti, V., Catrina, A. I., Bogen, B., Biberfeld, P., Grandien, A. (1999) The inhibitor of death receptor signaling, FLICE-inhibitory protein denes a new class of tumor progression factors. J. Exp. Med. 190, 10251032. Stamatopoulos, K., Kosmas, C., Belessi, C., Stavroyianni, N., Kyriazopoulos, P., Papadaki, T. (2000) Molecular insights into the immunopathogenesis of follicular lymphoma. Immunol. Today 21, 298 305. Tsujimoto, Y., Cossman, J., Jaffe, E., Croce, C. M. (1985) Involvement of the bcl-2 gene in human follicular lymphoma. Science 228, 1440 1443. McDonnell, T. J., Deane, N., Platt, F. M., Nunez, G., Jaeger, U., McKearn, J. P., Korsmeyer, S. J. (1989) bcl-2-immunoglobulin transgenic mice demonstrate extended B cell survival and follicular lymphoproliferation. Cell 57, 79 88. Reed, J. C., Cuddy, M., Slabiak, T., Croce, C. M., Nowell, P. C. (1988) Oncogenic potential of bcl-2 demonstrated by gene transfer. Nature 336, 259 261. Strasser, A., Harris, A. W., Bath, M. L., Cory, S. (1990) Novel primitive lymphoid tumours induced in transgenic mice by cooperation between myc and bcl-2. Nature 348, 331333. Vaux, D. L., Cory, S., Adams, J. M. (1988) Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature 335, 440 442. Fanidi, A., Harrington, E. A., Evan, G. I. (1992) Cooperative interaction between c-myc and bcl-2 proto-oncogenes. Nature 359, 554 556. Kogan, S. C., Brown, D. E., Shultz, D. B., Truong, B. T., LallemandBreitenbach, V., Guillemin, M. C., Lagasse, E., Weissman, I. L., Bishop, J. M. (2001) BCL-2 cooperates with promyelocytic leukemia retinoic acid receptor alpha chimeric protein (PMLRARalpha) to block neutrophil differentiation and initiate acute leukemia. J. Exp. Med. 193, 531543. Weller, M., Malipiero, U., Aguzzi, A., Reed, J. C., Fontana, A. (1995) 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. Investig. 95, 26332643. Campos, L., Rouault, J. P., Sabido, O., Oriol, P., Roubi, N., Vasselon, C., Archimbaud, E., Magaud, J. P., Guyotat, D. (1993) High expression of bcl-2 protein in acute myeloid leukemia cells is associated with poor response to chemotherapy. Blood 81, 30913096. Hermine, O., Haioun, C., Lepage, E., dAgay, M. F., Briere, J., Lavignac, C., Fillet, G., Salles, G., Marolleau, J. P., Diebold, J., Reyas, F., Gaulard, P. (1996) Prognostic signicance of bcl-2 protein expression in aggressive non-Hodgkins lymphoma. Groupe dEtude des Lymphomes de lAdulte (GELA). Blood 87, 265272. Miyashita, T., Reed, J. C. (1992) bcl-2 Gene transfer increases relative resistance of S49.1 and WEHI7.2 lymphoid cells to cell death and DNA fragmentation induced by glucocorticoids and multiple chemotherapeutic drugs. Cancer Res. 52, 54075411. Schmitt, C. A., Rosenthal, C. T., Lowe, S. W. (2000) Genetic analysis of chemoresistance in primary murine lymphomas. Nat. Med. 6, 1029 1035. Findley, H. W., Gu, L., Yeager, A. M., Zhou, M. (1997) Expression and regulation of Bcl-2, Bcl-xl, and Bax correlate with p53 status and

Igney and Krammer Apoptosis and tumor immune escape

917

160.

161.

162.

163.

164.

165.

166.

167.

168.

169. 170.

171.

172.

173.

174.

175.

176.

177.

178.

179.

sensitivity to apoptosis in childhood acute lymphoblastic leukemia. Blood 89, 2986 2993. Coustan-Smith, E., Kitanaka, A., Pui, C. H., McNinch, L., Evans, W. E., Raimondi, S. C., Behm, F. G., Arico, M., Campana, D. (1996) Clinical relevance of BCL-2 overexpression in childhood acute lymphoblastic leukemia. Blood 87, 1140 1146. Henderson, S., Huen, D., Rowe, M., Dawson, C., Johnson, G., Rickinson, A. (1993) Epstein-Barr virus-coded BHRF1 protein, a viral homologue of Bcl-2, protects human B cells from programmed cell death. Proc. Natl. Acad. Sci. USA 90, 8479 8483. Pearson, G. R., Luka, J., Petti, L., Sample, J., Birkenbach, M., Braun, D., Kieff, E. (1987) Identication of an Epstein-Barr virus early gene encoding a second component of the restricted early antigen complex. Virology 160, 151161. Tarodi, B., Subramanian, T., Chinnadurai, G. (1994) Epstein-Barr virus BHRF1 protein protects against cell death induced by DNA-damaging agents and heterologous viral infection. Virology 201, 404 407. Sarid, R., Sato, T., Bohenzky, R. A., Russo, J. J., Chang, Y. (1997) Kaposis sarcoma-associated herpesvirus encodes a functional bcl-2 homologue. Nat. Med. 3, 293298. Boise, L. H., Gonzalez-Garcia, M., Postema, C. E., Ding, L., Lindsten, T., Turka, L. A., Mao, X., Nunez, G., Thompson, C. B. (1993) bcl-x, a bcl-2-Related gene that functions as a dominant regulator of apoptotic cell death. Cell 74, 597 608. Dole, M. G., Jasty, R., Cooper, M. J., Thompson, C. B., Nunez, G., Castle, V. P. (1995) Bcl-xL is expressed in neuroblastoma cells and modulates chemotherapy-induced apoptosis. Cancer Res. 55, 2576 2582. Minn, A. J., Rudin, C. M., Boise, L. H., Thompson, C. B. (1995) Expression of bcl-xL can confer a multidrug resistance phenotype. Blood 86, 19031910. Zhou, P., Qian, L., Kozopas, K. M., Craig, R. W. (1997) Mcl-1, a Bcl-2 family member, delays the death of hematopoietic cells under a variety of apoptosis-inducing conditions. Blood 89, 630 643. Reed, J. C. (2001) The Survivin saga goes in vivo. J. Clin. Investig. 108, 965969. Ambrosini, G., Adida, C., Altieri, D. C. (1997) A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat. Med. 3, 917 921. Adida, C., Berrebi, D., Peuchmaur, M., Reyes-Mugica, M., Altieri, D. C. (1998) Anti-apoptosis gene, survivin, and prognosis of neuroblastoma. Lancet 351, 882 883. Grossman, D., Kim, P. J., Blanc-Brude, O. P., Brash, D. E., Tognin, S., Marchisio, P. C., Altieri, D. C. (2001) Transgenic expression of survivin in keratinocytes counteracts UVB-induced apoptosis and cooperates with loss of p53. J. Clin. Investig. 108, 991999. Grossman, D., Kim, P. J., Schechner, J. S., Altieri, D. C. (2001) Inhibition of melanoma tumor growth in vivo by survivin targeting. Proc. Natl. Acad. Sci. USA 98, 635 640. Mesri, M., Wall, N. R., Li, J., Kim, R. W., Altieri, D. C. (2001) Cancer gene therapy using a survivin mutant adenovirus. J. Clin. Investig. 108, 981990. Dierlamm, J., Baens, M., Wlodarska, I., Stefanova-Ouzounova, M., Hernandez, J. M., Hossfeld, D. K., De Wolf-Peeters, C., Hagemeijer, A., Van den Berghe, H., Marynen, P. (1999) The apoptosis inhibitor gene API2 and a novel 18q gene, MLT, are recurrently rearranged in the t(11; 18)(q21;q21)p6ssociated with mucosa-associated lymphoid tissue lymphomas. Blood 93, 36013609. Bird, C. H., Sutton, V. R., Sun, J., Hirst, C. E., Novak, A., Kumar, S., Trapani, J. A., Bird, P. I. (1998) 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. 18, 6387 6398. Medema, J. P., de Jong, J., Peltenburg, L. T., Verdegaal, E. M., Gorter, A., Bres, S. A., Franken, K. L., Hahne, M., Albar, J. P., Melief, C. J., Offringa, R. (2001) 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 98, 1151511520. Bladergroen, B. A., Meijer, C. J., ten Berge, R. L., Hack, C. E., Muris, J. J., Dukers, D. F., Chott, A., Kazama, Y., Oudejans, J. J., van Berkum, O., Kummer, J. A. (2002) Expression of the granzyme B inhibitor, protease inhibitor 9, by tumor cells in patients with non-Hodgkin and Hodgkin lymphoma: a novel protective mechanism for tumor cells to circumvent the immune system? Blood 99, 232237. Cheng, J., Zhou, T., Liu, C., Shapiro, J. P., Brauer, M. J., Kiefer, M. C., Barr, P. J., Mountz, J. D. (1994) Protection from Fas-mediated apoptosis by a soluble form of the Fas molecule. Science 263, 1759 1762.

180. Midis, G. P., Shen, Y., Owen-Schaub, L. B. (1996) Elevated soluble Fas (sFas) levels in nonhematopoietic human malignancy. Cancer Res. 56, 3870 3874. 181. Ugurel, S., Rappl, G., Tilgen, W., Reinhold, U. (2001) Increased soluble CD95 (sFas/CD95) serum level correlates with poor prognosis in melanoma patients. Clin. Cancer Res. 7, 12821286. 182. Gerharz, C. D., Ramp, U., Dejosez, M., Mahotka, C., Czarnotta, B., Bretschneider, U., Lorenz, I., Muller, M., Krammer, P. H., Gabbert, H. E. (1999) Resistance to CD95 (APO-1/Fas)-mediated apoptosis in human renal cell carcinomas: an important factor for evasion from negative growth control. Lab. Investig. 79, 15211534. 183. Bai, C., Connolly, B., Metzker, M. L., Hilliard, C. A., Liu, X., Sandig, V., Soderman, A., Galloway, S. M., Liu, Q., Austin, C. P., Caskey, C. T. (2000) Overexpression of M68/DcR3 in human gastrointestinal tract tumors independent of gene amplication and its location in a four-gene cluster. Proc. Natl. Acad. Sci. USA 97, 1230 1235. 184. Roth, W., Isenmann, S., Nakamura, M., Platten, M., Wick, W., Kleihues, P., Bahr, M., Ohgaki, H., Ashkenazi, A., Weller, M. (2001) Soluble decoy receptor 3 is expressed by malignant gliomas and suppresses CD95 ligand-induced apoptosis and chemotaxis. Cancer Res. 61, 2759 2765. 185. Lehmann, C., Zeis, M., Schmitz, N., Uharek, L. (2000) Impaired binding of perforin on the surface of tumor cells is a cause of target cell resistance against cytotoxic effector cells. Blood 96, 594 600. 186. Strand, S., Hofmann, W. J., Hug, H., Muller, M., Otto, G., Strand, D., Mariani, S. M., Stremmel, W., Krammer, P. H., Galle, P. R. (1996) Lymphocyte apoptosis induced by CD95 (APO-1/Fas) ligand-expressing tumor cellsa mechanism of immune evasion? Nat. Med. 2, 13611366. 187. Higaki, K., Yano, H., Kojiro, M. (1996) Fas antigen expression and its relationship with apoptosis in human hepatocellular carcinoma and noncancerous tissues. Am. J. Pathol. 149, 429 437. 188. Moller, P., Koretz, K., Leithauser, F., Bruderlein, S., Henne, C., Quentmeier, A., Krammer, P. H. (1994) Expression of APO-1 (CD95), a member of the NGF/TNF receptor superfamily, in normal and neoplastic colon epithelium. Int. J. Cancer 57, 371377. 189. Leithauser, F., Dhein, J., Mechtersheimer, G., Koretz, K., Bruderlein, S., Henne, C., Schmidt, A., Debatin, K. M., Krammer, P. H., Moller, P. (1993) Constitutive and induced expression of APO-1, a new member of the nerve growth factor/tumor necrosis factor receptor superfamily, in normal and neoplastic cells. Lab. Investig. 69, 415 429. 190. Volkmann, M., Schiff, J. H., Hajjar, Y., Otto, G., Stilgenbauer, F., Fiehn, W., Galle, P. R., Hofmann, W. J. (2001) Loss of CD95 expression is linked to most but not all p53 mutants in European hepatocellular carcinoma. J. Mol. Med. 79, 594 600. 191. Peli, J., Schroter, M., Rudaz, C., Hahne, M., Meyer, C., Reichmann, E., Tschopp, J. (1999) Oncogenic Ras inhibits Fas ligand-mediated apoptosis by downregulating the expression of Fas. EMBO J. 18, 1824 1831. 192. Maeda, T., Yamada, Y., Moriuchi, R., Sugahara, K., Tsuruda, K., Joh, T., Atogami, S., Tsukasaki, K., Tomonaga, M., Kamihira, S. (1999) Fas gene mutation in the progression of adult T cell leukemia. J. Exp. Med. 189, 10631071. 193. Landowski, T. H., Qu, N., Buyuksal, I., Painter, J. S., Dalton, W. S. (1997) Mutations in the Fas antigen in patients with multiple myeloma. Blood 90, 4266 4270. 194. Cascino, I., Papoff, G., De Maria, R., Testi, R., Ruberti, G. (1996) Fas/Apo-1 (CD95) receptor lacking the intracytoplasmic signaling domain protects tumor cells from Fas-mediated apoptosis. J. Immunol. 156, 1317. 195. Straus, S. E., Jaffe, E. S., Puck, J. M., Dale, J. K., Elkon, K. B., Rosen-Wolff, A., Peters, A. M., Sneller, M. C., Hallahan, C. W., Wang, J., Fischer, R. E., Jackson, C. M., Lin, A. Y., Baumler, C., Siegert, E., Marx, A., Vaishnaw, A. K., Grodzicky, T., Fleisher, T. A., Lenardo, M. J. (2001) The development of lymphomas in families with autoimmune lymphoproliferative syndrome with germline Fas mutations and defective lymphocyte apoptosis. Blood 98, 194 200. 196. Shin, M. S., Kim, H. S., Lee, S. H., Park, W. S., Kim, S. Y., Park, J. Y., Lee, J. H., Lee, S. K., Lee, S. N., Jung, S. S., Han, J. Y., Kim, H., Lee, J. Y., Yoo, N. J. (2001) Mutations of tumor necrosis factor-related apoptosis-inducing ligand receptor 1 (TRAIL-R1) and receptor 2 (TRAIL-R2) genes in metastatic breast cancers. Cancer Res. 61, 4942 4946. 197. Fisher, M. J., Virmani, A. K., Wu, L., Aplenc, R., Harper, J. C., Powell, S. M., Rebbeck, T. R., Sidransky, D., Gazdar, A. F., El-Deiry, W. S. (2001) Nucleotide substitution in the ectodomain of trail receptor DR4 is associated with lung cancer and head and neck cancer. Clin. Cancer Res. 7, 1688 1697. 198. Pai, S. I., Wu, G. S., Ozoren, N., Wu, L., Jen, J., Sidransky, D., El-Deiry, W. S. (1998) Rare loss-of-function mutation of a death receptor gene in head and neck cancer. Cancer Res 58, 35133518.

918

Journal of Leukocyte Biology Volume 71, June 2002

http://www.jleukbio.org

199. Lee, S. H., Shin, M. S., Kim, H. S., Lee, H. K., Park, W. S., Kim, S. Y., Lee, J. H., Han, S. Y., Park, J. Y., Oh, R. R., Jang, J. J., Han, J. Y., Lee, J. Y., Yoo, N. J. (1999) Alterations of the DR5/TRAIL receptor 2 gene in non-small cell lung cancers. Cancer Res. 59, 56835686. 200. Teitz, T., Wei, T., Valentine, M. B., Vanin, E. F., Grenet, J., Valentine, V. A., Behm, F. G., Look, A. T., Lahti, J. M., Kidd, V. J. (2000) Caspase 8 is deleted or silenced preferentially in childhood neuroblastomas with amplication of MYCN. Nat. Med. 6, 529 535. 201. Rampino, N., Yamamoto, H., Ionov, Y., Li, Y., Sawai, H., Reed, J. C., Perucho, M. (1997) Somatic frameshift mutations in the BAX gene in colon cancers of the microsatellite mutator phenotype. Science 275, 967969. 202. Meijerink, J. P., Mensink, E. J., Wang, K., Sedlak, T. W., Sloetjes, A. W., de Witte, T., Waksman, G., Korsmeyer, S. J. (1998) Hematopoietic malignancies demonstrate loss-of-function mutations of BAX. Blood 91, 29912997. 203. Molenaar, J. J., Gerard, B., Chambon-Pautas, C., Cave, H., Duval, M., Vilmer, E., Grandchamp, B. (1998) Microsatellite instability and frameshift mutations in BAX and transforming growth factor-beta RII genes are very uncommon in acute lymphoblastic leukemia in vivo but not in cell lines. Blood 92, 230 233. 204. Krajewski, S., Blomqvist, C., Franssila, K., Krajewska, M., Wasenius, V. M., Niskanen, E., Nordling, S., Reed, J. C. (1995) Reduced expression of proapoptotic gene BAX is associated with poor response rates to combination chemotherapy and shorter survival in women with metastatic breast adenocarcinoma. Cancer Res. 55, 4471 4478. 205. Yin, C., Knudson, C. M., Korsmeyer, S. J., Van Dyke, T. (1997) Bax suppresses tumorigenesis and stimulates apoptosis in vivo. Nature 385, 637 640. 206. Bargou, R. C., Wagener, C., Bommert, K., Mapara, M. Y., Daniel, P. T., Arnold, W., Dietel, M., Guski, H., Feller, A., Royer, H. D., Dorken, B. (1996) Overexpression of the death-promoting gene bax-alpha which is downregulated in breast cancer restores sensitivity to different apoptotic stimuli and reduces tumor growth in SCID mice. J. Clin. Investig. 97, 26512659. 207. Ionov, Y., Yamamoto, H., Krajewski, S., Reed, J. C., Perucho, M. (2000) Mutational inactivation of the proapoptotic gene BAX confers selective advantage during tumor clonal evolution. Proc. Natl. Acad. Sci. USA 97, 1087210877. 208. Soengas, M. S., Capodieci, P., Polsky, D., Mora, J., Esteller, M., OpitzAraya, X., McCombie, R., Herman, J. G., Gerald, W. L., Lazebnik, Y. A., Cordon-Cardo, C., Lowe, S. W. (2001) Inactivation of the apoptosis effector Apaf-1 in malignant melanoma. Nature 409, 207211. 209. Krammer, P. H. (1997) The tumor strikes back. Cell Death Differ. 4, 362364. 210. Igney, F. H., Behrens, C. K., Krammer, P. H. (2000) Tumor counterattack concept and reality. Eur. J. Immunol. 30, 725731. 211. Walker, P. R., Saas, P., Dietrich, P. Y. (1998) Tumor expression of Fas ligand (CD95L) and the consequences. Curr. Opin. Immunol. 10, 564 572. 212. French, L. E., Hahne, M., Viard, I., Radlgruber, G., Zanone, R., Becker, K., Muller, C., Tschopp, J. (1996) Fas and Fas ligand in embryos and adult mice: ligand expression in several immune-privileged tissues and coexpression in adult tissues characterized by apoptotic cell turnover. J. Cell Biol. 133, 335343. 213. Grifth, T. S., Brunner, T., Fletcher, S. M., Green, D. R., Ferguson, T. A. (1995) Fas ligand-induced apoptosis as a mechanism of immune privilege. Science 270, 1189 1192. 214. Stuart, P. M., Grifth, T. S., Usui, N., Pepose, J., Yu, X., Ferguson, T. A. (1997) CD95 ligand (FasL)-induced apoptosis is necessary for corneal allograft survival. J. Clin. Investig. 99, 396 402. 215. Grifth, T. S., Yu, X., Herndon, J. M., Green, D. R., Ferguson, T. A. (1996) CD95-induced apoptosis of lymphocytes in an immune privileged site induces immunological tolerance. Immunity 5, 716. 216. Bellgrau, D., Gold, D., Selawry, H., Moore, J., Franzusoff, A., Duke, R. C. (1995) A role for CD95 ligand in preventing graft rejection. Nature 377, 630 632. 217. Allison, J., Georgiou, H. M., Strasser, A., Vaux, D. L. (1997) Transgenic expression of CD95 ligand on islet beta cells induces a granulocytic inltration but does not confer immune privilege upon islet allografts. Proc. Natl. Acad. Sci. USA 94, 39433947. 218. Saas, P., Walker, P. R., Hahne, M., Quiquerez, A. L., Schnuriger, V., Perrin, G., French, L., Van Meir, E. G., de Tribolet, N., Tschopp, J., Dietrich, P. Y. (1997) Fas ligand expression by astrocytoma in vivo: maintaining immune privilege in the brain? J. Clin. Investig. 99, 1173 1178. 219. Choi, C., Park, J. Y., Lee, J., Lim, J. H., Shin, E. C., Ahn, Y. S., Kim, C. H., Kim, S. J., Kim, J. D., Choi, I. S., Choi, I. H. (1999) Fas ligand and

220.

221.

222.

223.

224.

225.

226.

227.

228.

229. 230. 231.

232.

233.

234.

235.

236. 237.

238.

239. 240.

241.

Fas are expressed constitutively in human astrocytes and the expression increases with IL-1, IL-6, TNF-alpha, or IFN-gamma. J. Immunol. 162, 1889 1895. Lau, H. T., Yu, M., Fontana, A., Stoeckert Jr., C. J. (1996) Prevention of islet allograft rejection with engineered myoblasts expressing FasL in mice. Science 273, 109 112. Kang, S. M., Hoffmann, A., Le, D., Springer, M. L., Stock, P. G., Blau, H. M. (1997) Immune response and myoblasts that express Fas ligand. Science 278, 13221324. Strand, S., Galle, P. R. (1998) Immune evasion by tumours: involvement of the CD95 (APO-1/Fas) system and its clinical implications. Mol. Med. Today 4, 63 68. Bennett, M. W., OConnell, J., OSullivan, G. C., Brady, C., Roche, D., Collins, J. K., Shanahan, F. (1998) The Fas counterattack in vivo: apoptotic depletion of tumor-inltrating lymphocytes associated with Fas ligand expression by human esophageal carcinoma. J. Immunol. 160, 5669 5675. Hahne, M., Rimoldi, D., Schroter, M., Romero, P., Schreier, M., French, L. E., Schneider, P., Bornand, T., Fontana, A., Lienard, D., Cerottini, J., Tschopp, J. (1996) Melanoma cell expression of Fas(Apo-1/CD95) ligand: implications for tumor immune escape. Science 274, 13631366. Mariani, S. M., Krammer, P. H. (1998) Differential regulation of TRAIL and CD95 ligand in transformed cells of the T and B lymphocyte lineage. Eur. J. Immunol. 28, 973982. OConnell, J., OSullivan, G. C., Collins, J. K., Shanahan, F. (1996) The Fas counterattack: Fas-mediated T cell killing by colon cancer cells expressing Fas ligand. J. Exp. Med. 184, 10751082. Villunger, A., Egle, A., Marschitz, I., Kos, M., Bock, G., Ludwig, H., Geley, S., Koer, R., Greil, R. (1997) Constitutive expression of Fas (Apo-1/CD95) ligand on multiple myeloma cells: a potential mechanism of tumor-induced suppression of immune surveillance. Blood 90, 1220. Nishimatsu, H., Takeuchi, T., Ueki, T., Kajiwara, T., Moriyama, N., Ishida, T., Li, B., Kakizoe, T., Kitamura, T. (1999) CD95 ligand expression enhances growth of murine renal cell carcinoma in vivo. Cancer Immunol. Immunother. 48, 56 61. Arai, H., Chan, S. Y., Bishop, D. K., Nabel, G. J. (1997) Inhibition of the alloantibody response by CD95 ligand. Nat. Med. 3, 843 848. Restifo, N. P. (2001) Countering the counterattack hypothesis. Nat. Med. 7, 259. Kang, S. M., Schneider, D. B., Lin, Z., Hanahan, D., Dichek, D. A., Stock, P. G., Baekkeskov, S. (1997) Fas ligand expression in islets of Langerhans does not confer immune privilege and instead targets them for rapid destruction. Nat. Med. 3, 738 743. Allison, J., Georgiou, H. M., Strasser, A., Vaux, D. L. (1997) Transgenic expression of CD95 ligand on islet beta cells induces a granulocytic inltration but does not confer immune privilege upon islet allografts. Proc. Natl. Acad. Sci. USA 94, 39433947. Judge, T. A., Desai, N. M., Yang, Z., Rostami, S., Alonso, L., Zhang, H., Chen, Y., Markman, J. F., DeMateo, R. P., Barker, C. F., Naji, A., Turka, L. A. (1998) Utility of adenoviral-mediated Fas ligand gene transfer to modulate islet allograft survival. Transplantation 66, 426 434. Takeuchi, T., Ueki, T., Nishimatsu, H., Kajiwara, T., Ishida, T., Jishage, K., Ueda, O., Suzuki, H., Li, B., Moriyama, N., Kitamura, T. (1999) Accelerated rejection of Fas ligand-expressing heart grafts. J. Immunol. 162, 518 522. Behrens, C. K., Igney, F. H., Arnold, B., Moller, P., Krammer, P. H. (2001) CD95 ligand-expressing tumors are rejected in anti-tumor TCR transgenic perforin knockout mice. J. Immunol. 166, 3240 3247. Seino, K., Kayagaki, N., Okumura, K., Yagita, H. (1997) Antitumor effect of locally produced CD95 ligand. Nat. Med. 3, 165170. Shimizu, M., Fontana, A., Takeda, Y., Yagita, H., Yoshimoto, T., Matsuzawa, A. (1999) Induction of antitumor immunity with Fas/APO-1 ligand (CD95L)-transfected neuroblastoma neuro-2a cells. J. Immunol. 162, 7350 7357. Okamoto, S., Takamizawa, S., Bishop, W., Wen, J., Kimura, K., Sandler, A. (1999) Overexpression of Fas ligand does not confer immune privilege to a pancreatic beta tumor cell line (betaTC-3). J. Surg. Res. 84, 77 81. Yagita, H., Seino, K., Kayagaki, N., Okumura, K. (1996) CD95 ligand in graft rejection. Nature 379, 682. Arai, H., Gordon, D., Nabel, E. G., Nabel, G. J. (1997) Gene transfer of Fas ligand induces tumor regression in vivo. Proc. Natl. Acad. Sci. USA 94, 1386213867. Seino, K., Iwabuchi, K., Kayagaki, N., Miyata, R., Nagaoka, I., Matsuzawa, A., Fukao, K., Yagita, H., Okumura, K. (1998) Chemotactic activity of soluble Fas ligand against phagocytes. J. Immunol. 161, 4484 4488.

Igney and Krammer Apoptosis and tumor immune escape

919

242. Ottonello, L., Tortolina, G., Amelotti, M., Dallegri, F. (1999) Soluble Fas ligand is chemotactic for human neutrophilic polymorphonuclear leukocytes. J. Immunol. 162, 36013606. 243. Shudo, K., Kinoshita, K., Imamura, R., Fan, H., Hasumoto, K., Tanaka, M., Nagata, S., Suda, T. (2001) The membrane-bound but not the soluble form of human Fas ligand is responsible for its inammatory activity. Eur. J. Immunol. 31, 2504 2511. 244. Hohlbaum, A. M., Moe, S., Marshak-Rothstein, A. (2000) Opposing effects of transmembrane and soluble Fas ligand expression on inammation and tumor cell survival. J. Exp. Med. 191, 1209 1220. 245. Kang, S. M., Braat, D., Schneider, D. B., ORourke, R. W., Lin, Z., Ascher, N. L., Dichek, D. A., Baekkeskov, S., Stock, P. G. (2000) A non-cleavable mutant of Fas ligand does not prevent neutrophilic destruction of islet transplants. Transplantation 69, 18131817. 246. Miwa, K., Asano, M., Horai, R., Iwakura, Y., Nagata, S., Suda, T. (1998) Caspase 1-independent IL-1beta release and inammation induced by the apoptosis inducer Fas ligand. Nat. Med. 4, 12871292. 247. Hohlbaum, A. M., Gregory, M. S., Ju, S. T., Marshak-Rothstein, A. (2001) Fas ligand engagement of resident peritoneal macrophages in vivo induces apoptosis and the production of neutrophil chemotactic factors. J. Immunol. 167, 6217 6224. 248. Rescigno, M., Piguet, V., Valzasina, B., Lens, S., Zubler, R., French, L., Kindler, V., Tschopp, J., Ricciardi-Castagnoli, P. (2000) Fas engagement induces the maturation of dendritic cells (DCs), the release of interleukin (IL)-1beta, and the production of interferon gamma in the absence of IL-12 during DC-T cell cognate interaction: a new role for Fas ligand in inammatory responses. J. Exp. Med. 192, 16611668. 249. Muller, M., Strand, S., Hug, H., Heinemann, E. M., Walczak, H., Hofmann, W. J., Stremmel, W., Krammer, P. H., Galle, P. R. (1997) Drug-induced apoptosis in hepatoma cells is mediated by the CD95 (APO-1/Fas) receptor/ligand system and involves activation of wild-type p53. J. Clin. Investig. 99, 403 413. 250. Friesen, C., Herr, I., Krammer, P. H., Debatin, K. M. (1996) Involvement of the CD95 (APO-1/FAS) receptor/ligand system in drug-induced apoptosis in leukemia cells. Nat. Med. 2, 574 577.

251. Scafdi, C., Kirchhoff, S., Krammer, P. H., Peter, M. E. (1999) Apoptosis signaling in lymphocytes. Curr. Opin. Immunol. 11, 277285. 252. Klas, C., Debatin, K. M., Jonker, R. R., Krammer, P. H. (1993) Activation interferes with the APO-1 pathway in mature human T cells. Int. Immunol. 5, 625 630. 253. Ehl, S., Hoffmann-Rohrer, U., Nagata, S., Hengartner, H., Zinkernagel, R. (1996) Different susceptibility of cytotoxic T cells to CD95 (Fas/ Apo-1) ligand-mediated cell death after activation in vitro versus in vivo. J. Immunol. 156, 23572360. 254. Chen, J. J., Sun, Y., Nabel, G. J. (1998) Regulation of the proinammatory effects of Fas ligand (CD95L). Science 282, 1714 1717. 255. Giovarelli, M., Musiani, P., Garotta, G., Ebner, R., Di Carlo, E., Kim, Y., Cappello, P., Rigamonti, L., Bernabei, P., Novelli, F., Modesti, A., Coletti, A., Ferrie, A. K., Lollini, P. L., Ruben, S., Salcedo, T., Forni, G. (1999) A stealth effect: adenocarcinoma cells engineered to express TRAIL elude tumor-specic and allogeneic T cell reactions. J. Immunol. 163, 4886 4893. 256. Mellado, M., de Ana, A. M., Moreno, M. C., Martinez, C., RodriguezFrade, J. M. (2001) A potential immune escape mechanism by melanoma cells through the activation of chemokine-induced T cell death. Curr. Biol. 11, 691 696. 257. Keane, M. M., Ettenberg, S. A., Nau, M. M., Russell, E. K., Lipkowitz, S. (1999) Chemotherapy augments TRAIL-induced apoptosis in breast cell lines. Cancer Res. 59, 734 741. 258. Chinnaiyan, A. M., Prasad, U., Shankar, S., Hamstra, D. A., Shanaiah, M., Chenevert, T. L., Ross, B. D., Rehemtulla, A. (2000) Combined effect of tumor necrosis factor-related apoptosis-inducing ligand and ionizing radiation in breast cancer therapy. Proc. Natl. Acad. Sci. USA 97, 1754 1759. 259. Jansen, B., Schlagbauer-Wadl, H., Brown, B. D., Bryan, R. N., van Elsas, A., Muller, M., Wolff, K., Eichler, H. G., Pehamberger, H. (1998) bcl-2 Antisense therapy chemosensitizes human melanoma in SCID mice. Nat. Med. 4, 232234. 260. Zamore, P. D. (2001) RNA interference: listening to the sound of silence. Nat. Struct. Biol. 8, 746 750.

920

Journal of Leukocyte Biology Volume 71, June 2002

http://www.jleukbio.org

También podría gustarte