当前位置: 首页 > 医学版 > 期刊论文 > 基础医学 > 免疫学杂志 > 2005年 > 第11期 > 正文
编号:11256585
C5a-Mediated Leukotriene B4-Amplified Neutrophil Chemotaxis Is Essential in Tumor Immunotherapy Facilitated by Anti-Tumor Monoclonal Antibod
     Abstract

    Intravenous and orally administered -glucans promote tumor regression and survival by priming granulocyte and macrophage C receptor 3 (CR3, iC3bR and CD11b/CD18) to trigger the cytotoxicity of tumor cells opsonized with iC3b via anti-tumor Abs. Despite evidence for priming of macrophage CR3 by oral -glucan in vivo, the current study in C57BL/6 and BALB/c mice showed that granulocytes were the essential killer cells in mAb- and oral -glucan-mediated tumor regression, because responses were absent in granulocyte-depleted mice. Among granulocytes, neutrophils were the major effector cells, because tumor regression did not occur when C5a-dependent chemotaxis was blocked with a C5aR antagonist, whereas tumor regression was normal in C3aR–/– mice. Neutrophil recruitment by C5a in vivo required amplification via leukotriene B4, because both C5a-mediated leukocyte recruitment into the peritoneal cavity and tumor regression were suppressed in leukotriene B4R-deficient (BLT-1–/–) mice.

    Introduction

    The effectiveness of tumor immunotherapy with mAbs can be greatly enhanced by the combined administration of -glucan (1, 2, 3). Without -glucan, anti-tumor mAbs, such as Herceptin (Genetech) or Rituxan (Genetech), have limited effector mechanisms, including inhibition of growth factor receptor function (Herceptin) (4) or Ab-mediated cellular cytotoxicity, and there is little, if any, contribution from the C system (5). C-dependent cytotoxicity via the membrane attack complex is limited by C regulatory proteins, such as CD46, CD55, and CD59, that are frequently overexpressed on tumor cells (6, 7, 8). Moreover, C receptor (CR) 6-dependent phagocytosis or cytotoxicity is reserved for microbial pathogens and is not elicited by C3b/iC3b-opsonized tumor cells (9, 10). In particular, the iC3bR CR3 is normally only triggered by iC3b deposited onto yeast or fungal cell walls (11). This is because the activation of CR3 for phagocytosis or cytotoxic degranulation requires its dual ligation to both iC3b and cell wall -glucan (12). Tumor cells opsonized with iC3b bind to the iC3b-binding site located within the I domain of CD11b (13), but not to the lectin-like domain for -glucan located within the C terminus of CD11b (14, 15). However, soluble -glucan isolated from yeast cell walls overcomes the lack of endogenous tumor cell -glucan by binding to and priming the lectin site of CR3 to mediate cytotoxic degranulation in response to tumor cells opsonized with iC3b (9, 15). By using -glucan in combination with anti-tumor mAbs that activate C, the resulting coat of iC3b on tumor cells can trigger CR3-dependent cytotoxicity by granulocytes (neutrophils and eosinophils), monocytes, macrophages, and NK cells (5).

    In vitro experiments have shown that soluble yeast -glucan can prime CR3 of granulocytes, macrophages, and NK cells to kill iC3b-opsonized tumor cells. However, murine tumor therapy models incorporating i.v. anti-tumor mAbs and soluble yeast -glucan indicated that the major CR3+ effector cell was a granulocyte (2). Because i.v. administered -glucans are taken up rapidly by CR3+ blood granulocytes or liver Kupffer cells (16), little is likely to reach tissue macrophages that could migrate into tumors. Conversely, orally administered yeast -glucan particles or large soluble molecules of barley -glucan are taken up by gastrointestinal macrophages; shuttled to bone marrow, spleen, and lymph nodes; and then slowly degraded into smaller soluble -glucan fragments that prime the CR3 of the marginated pool of granulocytes within the bone marrow (3). Thioglycolate-elicited peritoneal neutrophils upon CR3 priming were able to kill iC3b-opsonized tumor cells in a CR3-dependent manner (3). Although bone marrow NK cells are also likely to be primed for killing, experiments with NK cell-depleted mice indicated no important role for NK cells in oral -glucan tumor therapy (17). In addition to granulocytes, splenic macrophages isolated from mice receiving oral -glucan were able to kill iC3b-opsonized tumor cells via CR3 in vitro (3). Thus, it was unclear whether granulocytes, macrophages, or both leukocyte types might be involved in killing iC3b-opsonized tumor cells during oral -glucan-mediated tumor immunotherapy.

    Intratumoral release of the chemotactic factors C3a and C5a was hypothesized to be responsible for recruiting granulocytes and/or macrophages into tumors, thus expanding the importance of C activation beyond iC3b opsonization of tumors. Although receptors for C3a and C5a are present on all granulocytes and monocyte/macrophages (18), C5a primarily functions to recruit neutrophils and macrophages (19, 20), whereas C3a recruits eosinophils, basophils, and mast cells (18, 21). The lack of C3aR and C5aR (CD88) on NK cells probably also explains the absent requirement for NK cell function in mAb- and -glucan-mediated tumor immunotherapy (22, 23).

    C5a is a potent chemoattractant in vitro, with a short in vivo t1/2 due to serum carboxypeptidase N and intracellular degradation of C5a by C5aR-bearing cells (24). In addition, C5aR is widely distributed on leukocytes and nonleukocyte types, including epithelial and endothelial cells (25, 26, 27, 28). It was hypothesized that these cells, if stimulated by C5a, could play a role in amplifying the chemotactic responses of C5a by releasing leukotriene B4 (LTB4), a potent chemoattractant for neutrophils and macrophages (29, 30). LTB4 activates the G protein-coupled LTB4R type 1 (BLT-1) to mediate a diverse array of cellular responses in leukocytes, including chemotaxis, calcium mobilization, degranulation, and gene expression (31, 32, 33). LTB4 also activates respiratory burst and granule release from neutrophils (34). The current investigation sought to identify the major effector cell in i.v. and oral -glucan-mediated tumor therapy and to identify the chemotactic factors responsible for recruiting these effector leukocytes into C-opsonized tumors with i.v. or orally administered -glucan.

    Materials and Methods

    Graphing and statistical analysis of data

    Data were entered into PRISM 4.0 (GraphPad) to generate graphs of tumor regression or neutrophil chemotaxis, and Student’s t test was used from within the program to determine the significance of differences between two datasets. The Mann-Whitney rank-sum test was used to compare C5a-induced chemotaxis between BLT-1+/+ and BLT-1–/– mice. Survival curves were created using the Kaplan-Meier method, and statistical analyses of survival curves used a log-rank test.

    Results

    Granulocytes are required as killer cells for mAb- and oral -glucan-mediated immunotherapy

    Previous research has shown that orally administrated yeast WGP or barley -glucans are taken up by gastrointestinal macrophages that partially degraded the large -glucan molecules and released small soluble fragments of -glucan that function to prime the CR3 of marginated granulocytes and tissue macrophages in such a way that they were capable of mediating the cytotoxicity of iC3b-opsonized tumor cells in vitro (3). Although macrophages are essential for taking up the orally administrated -glucan and processing it into a form that could prime granulocytes and macrophage CR3, it appears possible that the more numerous granulocytes might play the major role in killing iC3b-opsonized tumor cells. To determine the requirement for granulocytes in oral -glucan tumor therapy, mice were injected i.p. with anti-Gr-1 rat anti-mouse granulocyte mAb before and during therapy to deplete peripheral granulocytes, but not monocytes, macrophages, or dendritic cells (2). Therapy of RMA-S-MUC1 tumors consisted of a combination of i.v. 14.G2a anti-GD2 ganglioside mAb and oral yeast WGP -glucan (Fig. 1). As observed previously with this model (3), combined mAb and oral yeast WGP -glucan induced significantly more tumor regression than did treatment with mAb alone (p < 0.01). However, granulocyte-depleted mice exhibited no tumor regression compared with untreated controls (Fig. 1). Moreover, depletion of granulocytes, despite combined therapy with oral WGP and anti-tumor mAb, resulted in significantly diminished survival compared with similarly treated animals that were not granulocyte depleted. Thus, tumor regression mediated by mAb and oral -glucan is dependent upon granulocytes.

    Granulocyte chemotaxis into tumors is mediated by C5a, but not by C3a

    C activation by naturally occurring tumor-reactive Abs or exogenous anti-tumor mAbs not only targets tumor cells with covalently bound iC3b, but also releases the chemotactic factors C3a and C5a (1, 2, 3, 38). Receptors for C3a and C5a are expressed on all myeloid-derived leukocytes, such as neutrophils, eosinophils, monocytes, and macrophages. Neutrophils, however, express far more C5aR (CD88) than C3aR, whereas eosinophils express more C3aR than C5aR (18). To examine whether C3a or C5a plays a critical role in the recruitment of neutrophils, which are required for combined anti-tumor mAbs and -glucan tumor immunotherapy, protocols were conducted using a C5aR antagonist peptide to block C5aR in wild-type mice or mice deficient in C3aR (Figs. 2 and 3). Blockade of C5aR completely inhibited (p < 0.05) the tumor regression mediated by either i.v. (Fig. 2A) or orally administered (Fig. 2B) -glucan. In contrast, in the tumor model that compared wild-type to C3aR–/– BALB/c mice, there was no significant difference in tumor regression mediated by i.v. yeast -glucan (Fig. 3A) or oral barley or yeast -glucan (Fig. 3B). In addition, tumor histology displayed similar levels of tumor-infiltrating granulocytes in these animals (not shown). These findings in combination with the observation that tumors did not contain significantly increased numbers of eosinophils relative to neutrophils indicate that neutrophils recruited via C5a are probably responsible for the tumor regression mediated by anti-tumor mAb and -glucan therapy.

    Discussion

    These studies demonstrated the essential role of C-mediated chemotaxis of neutrophils in tumor regression mediated by mAb and -glucan. Among granulocytes, the chemoattractants C5a and C3a preferentially recruit neutrophils and eosinophils, respectively. Because tumor regression mediated by mAb and -glucan depended on C5aR, but was independent of C3aR, a role for neutrophils, rather than other granulocytes, was suggested. The loss of tumor regression after selective depletion of granulocytes indicated that macrophages, although sensitive to C5a, could not mediate tumor regression independently from neutrophils. Lastly, the functional activity of C5a in vivo was shown to require amplification by LTB4, because rC5a-mediated neutrophil recruitment to the peritoneal cavity and tumor regression were suppressed in mice deficient in BLT-1. Indeed, many fewer infiltrating neutrophils were observed in the tumors of BLT-1–/– compared with wild-type animals (Fig. 5). In a zymosan-induced peritonitis model, in which the alternative pathway of C is activated, LTB4 was shown to be important in neutrophil recruitment (30, 44). Although a second C5aR as well as a low affinity LTB4R (BLT-2) were recently identified (45, 46), they did not seem to influence neutrophil recruitment in these experiments. These data support the hypothesis that C5a initiates a chemokine cascade in which C5a induces LTB4, making LTB4 the effective chemoattractant mediating inflammatory events initiated by C activation. Moreover, these studies provide additional molecular mechanisms for the effectiveness of combined -glucan and anti-tumor mAb immunotherapy and show a dual role for C activation in the opsonization of tumors with iC3b and the C5a-mediated recruitment of neutrophils to the tumor.

    The tumor microenvironment suppresses effective immune responses and may promote host immune tolerance. Altering the tumor milieu to promote inflammation enhances the recruitment of immune cells, particularly APCs and T cells, to tumors, leading to clinical benefit (47). In contrast, chronic inflammation can increase mutagenesis and promote tumor development (48, 49). Indeed, DNA damage resulting from chronic inflammation is believed to be the mechanism by which infection can cause cancer (50). Therefore, manipulation of inflammation toward therapeutic benefit would significantly improve the efficacy of tumor immunotherapy. Our data expand those observations to include the significant contribution of neutrophil chemotaxis in response to C5a and LTB4 to mediate tumor regression by combined oral -glucan and anti-tumor mAb therapy. Notably, an abundant neutrophil infiltrate in the regressing tumors of treated animals implies that the recruited -glucan-primed neutrophils were efficient killers. Thus, it is hypothesized that strategies to improve the quantity of infiltrating primed neutrophils should improve therapeutic efficacy.

    Tumor immunotherapy mediated by anti-tumor mAbs and -glucan is particularly novel, in that it recruits neutrophils as effector cells and can use humanized mAbs already in clinical use (e.g., Herceptin, Erbitux, Campath, (ImCLONE Systems) and Rituxan (Berlex)). The critical feature required of such therapeutic mAbs is that they activate C, thus requiring a human IgG1 or IgG3 framework, allowing iC3b opsonization of tumors and C5a-mediated recruitment of neutrophils (17). Immunotherapy with mAb and -glucan can be coupled easily to vaccine therapies that elicit immune CTL, exploiting the concurrent production of anti-tumor Abs. C5a-recruited neutrophils and immune CTL would probably kill tumors more effectively than would CTL alone. Tumor escape from CTL through down-regulation of MHC class I would also be prevented, because neutrophils are targeted to the iC3b on tumors and not to MHC class I.

    Footnotes

    The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

    1 This work was supported by grants from the National Institutes of Health (R01CA86412; to G.D.R. and J.Y.) and the U.S. Army Breast Cancer Research Program (DAMD17-02-1-0445; to G.D.R.), the Kentucky Lung Cancer Research Board (to J.Y.), a gift fund from Biopolymer Engineering, Inc. (Eagan, MN; to G.D.R. and J.Y.), a National Cancer Institute supplement to promote research collaboration (to G.D.R. and B.H.), and Grants R29AI43184 (to B.H.) and R01AI52381 (to B.H.).

    2 Address correspondence and reprint requests to Dr. Jun Yan, James Graham Brown Cancer Center, 580 South Preston Street, Room 119A, Louisville, KY 40202. E-mail address: jun.yan{at}louisville.edu

    3 Deceased.

    4 Current address: Department of Clinical Immunology, Polish-American Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland.

    5 Current address: Division of Protein Sciences, Aurigene Discoveries Technologies, 39-40 KIADB Industrial Area, Electronic City Phase II, Hosur Road, Bangalore 560100, India.

    6 Abbreviations used in this paper: CR, C receptor; anti-Gr-1, rat anti-mouse mAb RB6-8C5 specific for Ly-6G; BLT-1, leukotriene B4 receptor type 1; LTB4, leukotriene B4; MMTV, murine mammary tumor virus; NSG, neutral soluble glucan; WGP, whole glucan particle.

    Received for publication December 17, 2004. Accepted for publication March 30, 2005.

    References

    ]

    Cheung, N. K., S. Modak, A. Vickers, B. Knuckles. 2002. Orally administered -glucans enhance anti-tumor effects of monoclonal antibodies. Cancer Immunol. Immunother. 51: 557-564.

    Hong, F., R. D. Hansen, J. Yan, D. J. Allendorf, J. T. Baran, G. R. Ostroff, G. D. Ross. 2003. -Glucan functions as an adjuvant for monoclonal antibody immunotherapy by recruiting tumoricidal granulocytes as killer cells. Cancer Res. 63: 9023-9031.

    Hong, F., J. Yan, J. T. Baran, D. J. Allendorf, R. D. Hansen, G. R. Ostroff, P. X. Xing, N. K. Cheung, G. D. Ross. 2004. Mechanism by which orally administered -1,3-glucans enhance the tumoricidal activity of antitumor monoclonal antibodies in murine tumor models. J. Immunol. 173: 797-806.

    Wang, S. C., L. Zhang, G. N. Hortobagyi, M. C. Hung. 2001. Targeting HER2: recent developments and future directions for breast cancer patients. Semin. Oncol. 28: 21-29.

    Gelderman, K. A., S. Tomlinson, G. D. Ross, A. Gorter. 2004. Complement function in mAb-mediated cancer immunotherapy. Trends Immunol. 25: 158-164.

    Niehans, G. A., D. L. Cherwitz, N. A. Staley, D. J. Knapp, A. P. Dalmasso. 1996. Human carcinomas variably express the complement inhibitory proteins CD46 (membrane cofactor protein), CD55 (decay-accelerating factor), and CD59 (protectin). Am. J. Pathol. 149: 129-142.

    Yu, J., T. Caragine, S. Chen, B. P. Morgan, A. B. Frey, S. Tomlinson. 1999. Protection of human breast cancer cells from complement-mediated lysis by expression of heterologous CD59. Clin. Exp. Immunol. 115: 13-18.

    Li, L., I. Spendlove, J. Morgan, L. G. Durrant. 2001. CD55 is over-expressed in the tumour environment. Br. J. Cancer 84: 80-86.(Daniel J. Allendorf*,, Ju)