负性免疫分子TIM-3和细胞因子IL-33在T细胞介导的肿瘤免疫中的作用及机制
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
肿瘤是一种慢性渐进性的常见严重危害健康的疾病,以其生长迅速,高转移性,治愈率低及易复发为特性。近几十年,尽管手术切除术、化学疗法和放射治疗技术,及其它治疗方法在不断改进和发展,但肿瘤的有效治疗仍然是个难题。
     近年来,肿瘤的免疫治疗为肿瘤治疗开拓新的途径。然而免疫治疗的最大障碍是肿瘤免疫抑制,尤其具有很强免疫抑制作用的肿瘤微环境。肿瘤细胞和浸润在肿瘤局部的免疫细胞,新生血管及其内皮细胞(BECs),肿瘤相关的成纤维细胞(CAFs)及细胞外基质,共同构成了肿瘤微环境。肿瘤微环境的一个主要作用是在肿瘤局部形成了一个免疫抑制的环境,消弱了免疫细胞对肿瘤细胞的杀伤,例如肿瘤局部大量调节性T细胞(Tregs)及髓系来源的抑制性细胞(MDSC)的聚集,各种抑制性细胞因子的产生,以及耗竭性T细胞的形成。这一切,使得肿瘤细胞逃逸了免疫系统的杀伤作用,最终造成了肿瘤细胞的免疫逃逸。因此,充分的认识肿瘤微环境的免疫抑制分子机制,有助于寻找能增强抗肿瘤免疫的分子靶点。
     TIM-3,属于T细胞免疫球蛋白粘蛋白分子(T cell immunoglobulin-and mucin-domain-containing molecules, Tim)家族的一员。人类TIM-3分子是由301个氨基酸组成的Ⅰ型膜蛋白,包括一个Ig样V区的N-端(富含半胱氨酸),一个富含丝氨酸和苏氨酸的粘蛋白区,一个跨膜区,以及一胞内区。TIM-3表达于辅助性Th1、Th17细胞,及CD8T细胞。是一个负性调节分子,在与其配体结合凝集素9(galectin-9)作用下,抑制Thl及Thl7免疫应答并引发外周免疫耐受。最近,TIM-3分子被认为可以作为耗竭性T细胞的表面标志之一,并且在肿瘤微环境内耗竭性T细胞中发挥重要作用。因此,本论文第一部分以TIM-3及人非小细胞肺癌(non-small cell lung cancer, NSCLC)为切入点,研究肿瘤微环境中的耗竭性T细胞及其对肿瘤的影响。
     肿瘤的免疫治疗可以通过对肿瘤微环境的改变,使得免疫细胞重新恢复对肿瘤细胞的杀伤作用。本论文第二部分以白细胞介素33(interleukin33,IL-33)为研究对象,揭示其在肿瘤微环境中的抗肿瘤作用。IL-33属于白细胞介素1(Interleukin1,IL-1)细胞因子家族成员。组成性表达于一些构成了我们机体第一道防御的上皮及内皮细胞。除此之外,还表达于一些坏死的细胞及活化的免疫细胞,如巨噬细胞在受到外源性感染的时候。尽管IL-33已经被证实诱导了Th2免疫应答,但其在Thl免疫应答中的作用仍不清楚。我们最近的研究发现,IL-33的受体ST2表达于效应性CD8+T细胞,并且受T-bet调控,且IL-33可提高效应性CD8+T细胞的功能。因此,IL-33具有潜在抗肿瘤作用。本论文第二部分初步探讨IL-33在体内的抗肿瘤作用。
     本论文分2个部分:(1)通过探讨TIM-3在非小细胞肺癌中的表达和作用,了解肿瘤微环境中的耗竭性T淋巴细胞及肿瘤微环境的免疫抑制机制。(2)以IL-33为工具,研究如何改变肿瘤微环境,为肿瘤的免疫治疗提供潜在的手段。
     第一部分TIM-3分子在人非小细胞肺癌肿瘤微环境中的作用
     [目的]以TIM-3分子为切入点,研究人非小细胞肺癌肿瘤微环境中的Treg和耗竭性T淋巴细胞及其抑制性微环境对肿瘤的影响。
     [方法]取非小细胞肺癌患者手术切除的肿瘤及癌旁组织,经Ⅳ型胶原酶消化及机械方法,获取单细胞悬液,并用密度梯度离心法分离出组织中浸润的淋巴细胞,同时分离患者外周血中的淋巴细胞,流式细胞仪检测T淋巴细胞上TIM-3的表达并分析其与临床病理因素之间的相关性。体外刺激肿瘤组织中分离出的淋巴细胞,用细胞内染色的方法分析以TIM-3为特征的耗竭性T细胞与正常T细胞在功能上的差异。
     [结果]与癌旁正常组织浸润淋巴细胞(TILs)及外周血T细胞相比,非小细胞肺癌患者肿瘤组织TILs表面TIM-3的表达明显上调,且TIM-3和PD-1在CD4+TILs和CD8+TILs呈现共表达。并且,TIM-3在NSCLC患者肿瘤浸润CD4+T细胞的表达和肿瘤淋巴结转移及患者分期相关。与TIM-3-CD4+和TIM-3-CD8+TILs相比,TIM-3+CD4+和TIM-3+CD8+TILs γ干扰素(IFN-γ)的分泌明显下降。我们还发现,在肿瘤组织中,大约70%的TIM-3+CD4+TILs表达FOXP3且大约60%的FOXP3+CD4+TILs表达TIM-3
     [结论]本研究发现TIM-3在非小细胞肺癌肿瘤中TILs上有较高的表达,并且其在CD4+T细胞上的表达程度与肿瘤的淋巴结转移及患者分期有关。TIM-3和PD1的共表达揭示了肿瘤局部大量无反应性T细胞的存在。此外,发现了以TIM-3+为表型特征的一群调节性T细胞(TIM-3+Treg)。使我们对肿瘤微环境得到进一步的认识,并且为免疫治疗提供了一个新的靶点。
     第二部分IL-33提高肿瘤微环境中Thl型免疫应答及其抗肿瘤作用
     [目的]研究细胞因子IL-33对肿瘤微环境的影响及其抗肿瘤作用。
     [方法]体外克隆并构建分泌型IL-33/pCDNA3.1重组表达载体,转染入黑色素瘤细胞系B16(B16-IL-33),并分别转入空载体pCDNA3.1(B16-mock)及IL-12/pCDNA3.1(B16-IL-12)作为对照。分别将2×105B16-moc、B16-IL-12及B16-IL33肿瘤细胞皮下接种于C57B/L6小鼠,观察肿瘤生长曲线,并分离肿瘤浸润淋巴细胞,流式检测相关免疫指标。分别将2×105B16-mock及B16-IL33肿瘤细胞皮下接种于正常C57B/L6(WT)或转录因子T-bet和eomes基因敲除(DKO)的小鼠,观察肿瘤生长曲线,并分离肿瘤浸润淋巴细胞,流式检测相关免疫指标。
     [结果]B16-IL-33肿瘤在C57B/L6小鼠生长速度显著减慢。与B16-mock肿瘤相比,B16-IL-33肿瘤浸润CD45+、CD8+T细胞、NK细胞比例明显增高。并且,B16-IL-33肿瘤浸润的CD8+T细胞,NK细胞分泌细胞因子IFN-γ的能力更高。在缺失了转录因子T-bet及Eomes的小鼠中,B16-IL33肿瘤的生长速度显著增快,并且,与接种了B16-IL-33肿瘤细胞的野生型(WT)小鼠相比,肿瘤组织中浸润的CD45+、CD8+T细胞、NK细胞比例明显减少,并且分泌细胞因子IFN-γ的能力降低。
     [结论]IL-33明显提高了肿瘤微环境中Th1型免疫应答,具有潜在的抗肿瘤作用,为肿瘤的免疫治疗提供了新的方法。
Tumors, especially malignant tumors are sorts of systemic disease. They are the major deadly diseases for human being because some unique feature, such as high speed of growth, high rate of metastasis, low cure rate and easy to relapse. Recent decades, although there's great improvement of surgery, chemo and radiation treatment, the therapy of tumor is still a difficult problem. We still need more understanding on tumor's occurring, developing as well as its therapy.
     Recently, immunotherapy became a new strategy of tumor therapy, and shows a great prospect. As we mentioned tumor immunotherapy, we have to give an introduction about tumor microenvironment. The malignant features of tumor cells cannot be manifested without an important interplay between cancer cells and their local environment. When there's occurring a tumor, meanwhile forms a local environment where tumor cells live in. Basically, tumor microenvironment consist of the tumor cells, infiltrating immune cells, blood endothelial cells (BECs), carcinoma-associated fibroblasts (CAFs), and the extracellular matrix. Tumor environment induces tumor cells tolerance, through many ways, forms a immune suppressive environment to avoid the killing of immune system, such as accumulation of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSC), production of some suppressive cytokines, and also the induce the T cell exhaustion. Therefore, the tumor immunotherapy need us to fully realize the tumor microenvironment and find effective target to change the immune suppressive situation.
     TIM-3belongs to T cell immunoglobulin and mucin domain containing molecules (TIM) family member. Human TIM-3is a301amino acid, type I membrane protein, includes an immunoglobulin V domain, a mucin domain, a trans-membrane domain and an intracellular tail with one tyrosine phosphorylation. TIM-3is expressed Thl, Th17cells, and also CD8+T cells that negatively regulate these cells by binding to its ligand Galectin-9, and also induce peripheral tolerance. Recently TIM-3was thought to be one of the exhausted T cell marker, and plays an important role in tumor microenvironment. So, the first part of the thesis is based on TIM-3and human non-small cell lung cancer (NSCLC), studying the exhausted T cell in tumor microenvironment and its influence on cancer patient.
     The most important of tumor immunotherapy is finding an efficient target to change the tumor microenvironment, make the tumor infiltrating immune cells exhibiting their killing function to tumor cells. The second part of the thesis focus on interleukin33(IL-33), disclosure its antitumor effect in tumor microenvironment. IL-33is a member of the interleukin1family (IL-1) of cytokines, and constitutively expressed in the nucleus of some endothelial and epithelial cells. In addition, it also released by some necrotic cells or activated innate immune cells such as macrophages during infection. Although IL-33is realized to induce the Th2response, but its role in Thl response is still not clear. Some results identified IL-33involves in Thl response, and also there are some literatures show development of tumor induced the production of IL-33. Recently, we found ST2, the receptor of IL-33was expressed in CD8+T cells cultured with Thl condition, and T-bet was required for this expression. And IL-33could enhance the CD8+T cell function. Hence, IL-33may have potent antitumor effect. The second part of the thesis is based on IL-33and tumor microenvironment, studying the antitumor effect of IL-33in vivo.
     This thesis is consist of two part:(1) studying the exhausted T cells in tumor microenvironment and the mechanism of the suppressive environment in tumor through TIM-3and NSCLC.(2) Use IL-33as a tool, study how to change the tumor microenvironment, to achieve the aim of tumor immunotherapy.
     Part I The Function of TIM-3in Human Non-Small Cell Lung Cancer Microenvironment
     Objective:Focus on TIM-3and exhausted T cell in tumor microenvironment, studying the effect of suppressive environment to tumor progress.
     Methods:Tumor tissue and adjacent normal tissue from the same patient were minced and digested with collagenase IV digestion solution. The pieces were then transferred to the steel mesh and single cell suspensions were obtained by mechanical grind. TILs were further purified by the gradient protocol, washed and re-suspended in Hank's media. Meanwhile, peripheral T cells were purified from fresh blood of patients, then use flow cytometry to test the expression of TIM-3in TILs, and analyze the correlation between TIM-3expression and clinical pathological parameters.
     Result:Compare to TILs from adjacent normal tissue and peripheral T cells, TIM-3were highly expressed in TILs from NSCLC tissue, and co-expressed with PD-1in CD4+/CD8+TILs. Also, the higher frequency of TIM-3+CD4+TILs showed significant association with lymph node metastasis and more advanced cancer stages. TIM-3+CD4+and TIM-3+CD8+TILs showed less IFN-γ production than TIM-3-CD4+and TIM-3-CD8+TILs. We also found that in tumor microenvironment, almost70%percent of TIM-3+CD4+TILs were FOXP3+and about60%percent FOXP3+CD4+TILs were TIM-3+.
     Conclusion:We found TIM-3was highly expressed in TILs of NSCLC, and the frequency of TIM-3+CD4+TILs was associating with lymph node metastasis and cancer stages. Co-expressing of TIM-3and PD-1showed the high amount of exhausted T cell in tumor microenvironment. In addition, the expression of TIM-3in Treg (Tim-3+Treg) cells characterized a new population of regulatory T cells in tumor tissue. This part of study improved our knowledge of the mechanisms of immunosuppression within tumor microenvironment, and provided a new target for tumor immunotherapy.
     Part Ⅱ IL-33signal enhances the Th1response and antitumor effect.
     Objective:Studying the antitumor effect of IL-33in tumor microenvironment.
     Methods:secretory IL-33was Cloned and constructed into pCDNA3.1vector (IL-33/pCDNA3.1), then transfected into melanoma cell line B16(B16-IL-33). Meanwhile, pCDNA3.1and IL-12/pCDNA3.1were also transfected into B16as control.2×105B16-mock、B16-IL-12or B16-IL33cells were intradermally (i.d.) injected into wild type C57B/L6mice, tumor size was observed and TILs were purified to test the immune parameters.2×105B16-mock、B16-IL-12or B16-IL33cells were injected i.d. into C57B/L6(WT) or transcription factor T-bet and Eomes double knockout mice(DKO), then tumor size was observed and TILs were purified to test the immune parameters.
     Result:Tumor growth dramatically delayed in B16-IL-33tumor. Compare to B16-mock tumor, the population of CD45+, CD8+TILs and NK cells in B16-IL33tumor were increased. Also, the IFN-Y production of CD8+TILs and NK cells were increased in B16-IL-33tumor. Without transcription factor T-bet and Eomes, B16-IL-33tumor grew faster, and compare to WT mice, tumor infiltrating CD45+, CD8+and NK cells were decreased, and also less amount of IFN-γ were produced by CD8+, and NK cells.
     Conclusion:IL-33can dramatically enhance the Thl response in tumor environment and has the potent antitumor effect, which may provide us a novel therapeutic approach for tumor immunotherapy.
引文
1. Karagiannis, G.S., et al., Cancer-associated fibroblasts drive the progression of metastasis through both paracrine and mechanical pressure on cancer tissue. Mol Cancer Res,2012.10(11):p.1403-18.
    2. Hanahan, D. and L.M. Coussens, Accessories to the crime:functions of cells recruited to the tumor microenvironment. Cancer Cell,2012.21(3):p.309-22.
    3. Hanahan, D. and R.A. Weinberg, Hallmarks of cancer:the next generation. Cell, 2011.144(5):p.646-74.
    4. Nishikawa, H. and S. Sakaguchi, Regulatory T cells in tumor immunity. Int J Cancer,2010.127(4):p.759-67.
    5. Gondek, D.C., et al., Cutting edge:contact-mediated suppression by CD4+CD25+ regulatory cells involves a granzyme B-dependent, perforin-independent. mechanism. J Immunol,2005.174(4):p.1783-6.
    6. Serafini, P., I. Borrello, and V. Bronte, Myeloid suppressor cells in cancer: recruitment, phenotype, properties, and mechanisms of immune suppression. Semin Cancer Biol,2006.16(1):p.53-65.
    7. Gallina, G, et al., Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells. J Clin Invest,2006.116(10):p. 2777-90.
    8. Zea, A.H., et al., L-Arginine modulates CD3zeta expression and T cell function in activated human T lymphocytes. Cell Immunol,2004.232(1-2):p.21-31.
    9. Bronte, V., et al., Apoptotic death of CD8+ T lymphocytes after immunization: induction of a suppressive population of Mac-1+/Gr-1+cells. J Immunol,1998. 161(10):p.5313-20.
    10. Fujimura, T., et al., Regulatory T cells stimulate B7-H1 expression in myeloid-derived suppressor cells in ret melanomas. J Invest Dermatol,2012. 132(4):p.1239-46.
    11. Obermajer, N., et al., PGE(2)-induced CXCL12 production and CXCR4 expression controls the accumulation of human MDSCs in ovarian cancer environment. Cancer Res,2011.71(24):p.7463-70.
    12. Fujita, M., et al., COX-2 blockade suppresses gliomagenesis by inhibiting myeloid-derived suppressor cells. Cancer Res,2011.71(7):p.2664-74.
    13. Pan, P.Y., et al., Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res,2010.70(1):p.99-108.
    14. Yang, R., et al., CD80 in immune suppression by mouse ovarian carcinoma-associated Gr-1+CD11b+myeloid cells. Cancer Res,2006.66(13):p. 6807-15.
    15. Cerundolo, V., et al., Harnessing invariant NKT cells in vaccination strategies. Nat Rev Immunol,2009.9(1):p.28-38.
    16. Park, J.M., et al., Natural immunosurveillance against spontaneous, autochthonous breast cancers revealed and enhanced by blockade of IL-13-mediated negative regulation. Cancer Immunol Immunother,2008.57(6):p.907-12.
    17. Terabe, M., et al., Transforming growth factor-beta production and myeloid cells are an effector mechanism through which CD1d-restricted T cells block cytotoxic T lymphocyte-mediated tumor immunosurveillance:abrogation prevents tumor recurrence. J Exp Med,2003.198(11):p.1741-52.
    18. Terabe, M., J.M. Park, and J.A. Berzofsky, Role of IL-13 in regulation of anti-tumor immunity and tumor growth. Cancer Immunol Immunother,2004.53(2): p.79-85.
    19. Gorelik, L. and R.A. Flavell, Transforming growth factor-beta in T-cell biology. Nat Rev Immunol,2002.2(1):p.46-53.
    20. Letterio, J.J. and A.B. Roberts, Regulation of immune responses by TGF-beta. Annu Rev Immunol,1998.16:p.137-61.
    21. Li, M.O. and R.A. Flavell, TGF-beta:a master of all T cell trades. Cell,2008. 134(3):p.392-404.
    22. Geissmann, F., et al., TGF-beta 1 prevents the noncognate maturation of human dendritic Langerhans cells. J Immunol,1999.162(8):p.4567-75.
    23. Borkowski, T.A., et al., A role for endogenous transforming growth factor beta 1 in Langerhans cell biology:the skin of transforming growth factor beta 1 null mice is devoid of epidermal Langerhans cells. J Exp Med,1996.184(6):p.2417-22.
    24. Johnson, B., et al., Physiology and therapeutics of vascular endothelial growth factor in tumor immunosuppression. Curr Mol Med,2009.9(6):p.702-7.
    25. Jordan, J.T., et al., Preferential migration of regulatory T cells mediated by glioma-secreted chemokines can be blocked with chemotherapy. Cancer Immunol Immunother,2008.57(1):p.123-31.
    26. Ueno, T., et al., Significance of macrophage chemoattractant protein-1 in macrophage recruitment, angiogenesis, and survival in human breast cancer. Clin Cancer Res,2000.6(8):p.3282-9.
    27. Hu, K., et al., Recombined CC chemokine ligand 2 into B16 cells induces production of Th2-dominant [correction of dominanted] cytokines and inhibits melanoma metastasis. Immunol Lett,2007.113(1):p.19-28.
    28. Fuller, M.J., et al., Maintenance, loss, and resurgence of T cell responses during acute, protracted, and chronic viral infections. J Immunol,2004.172(7):p. 4204-14.
    29. Wherry, E.J., et al., Viral persistence alters CD8 T-cell immunodominance and, tissue distribution and results in distinct stages of functional impairment. J Virol, 2003.77(8):p.4911-27.
    30. Fuller, M.J. and A.J. Zajac, Ablation of CD8 and CD4 T cell responses by high viral loads. J Immunol,2003.170(1):p.477-86.
    31. Richter, K., P. Agnellini, and A. Oxenius, On the role of the inhibitory receptor LAG-3 in acute and chronic LCMV infection. Int Immunol,2010.22(1):p.13-23.
    32. Jones, R.B., et al., Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. J Exp Med, 2008.205(12):p.2763-79.
    33. Barber, D.L., et al., Restoring function in exhausted CD8 T cells during chronic viral infection. Nature,2006.439(7077):p.682-7.
    34. Mumprecht, S., et al., Programmed death 1 signaling on chronic myeloid leukemia-specific T cells results in T-cell exhaustion and disease progression. Blood,2009.114(8):p.1528-36.
    35. Ladell, K., et al., Central memory CD8+T cells appear to have a shorter lifespan and reduced abundance as a function of HIV disease progression. J Immunol,2008. 180(12):p.7907-18.
    36. Moskophidis, D., et al., Virus persistence in acutely infected immunocompetent mice by exhaustion of antiviral cytotoxic effector T cells. Nature,1993.362(6422): p.758-61.
    37. Schwartz, R.H., T cell anergy. Annu Rev Immunol,2003.21:p.305-34.
    38. Monney, L., et al., Thl-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature,2002.415(6871):p. 536-41.
    39. Sabatos, C.A., et al., Interaction of Tim-3 and Tim-3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat Immunol,2003.4(11):p. 1102-10.
    40. Nakae, S., et al., Phenotypic differences between Thl and Thl7 cells and negative regulation of Thl cell differentiation by IL-17. J Leukoc Biol,2007.81(5):p. 1258-68.
    41. Anderson, A.C., et al., Promotion of tissue inflammation by the immune receptor Tim-3 expressed on innate immune cells. Science,2007.318(5853):p.1141-3.
    42. Nakae, S., et al., TIM-1 and TIM-3 enhancement of Th2 cytokine production by mast cells. Blood,2007.110(7):p.2565-8.
    43. Oikawa, T., et al., Preferential involvement of Tim-3 in the regulation of hepatic CD8+T cells in murine acute graft-versus-host disease. J Immunol,2006.177(7): p.4281-7.
    44. Frisancho-Kiss, S., et al., Cutting edge:T cell Ig mucin-3 reduces inflammatory heart disease by increasing CTLA-4 during innate immunity. J Immunol,2006. 176(11):p.6411-5.
    45. Khademi, M., et al., T Cell Ig-and mucin-domain-containing molecule-3 (TIM-3) and TIM-1 molecules are differentially expressed on human Thl and Th2 cells and in cerebrospinal fluid-derived mononuclear cells in multiple sclerosis. J Immunol, 2004.172(11):p.7169-76.
    46. Zhou, Q., et al., Coexpression of Tim-3 and PD-1 identifies a CD8+T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia. Blood,2011.117(17):p.4501-10.
    47. Sakuishi, K., et al., Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med,2010.207(10):p. 2187-94.
    48. Fourcade, J., et al., Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med, 2010.207(10):p.2175-86.
    49. Dardalhon, V., et al., Tim-3/galectin-9 pathway:regulation of Thl immunity through promotion of CD11b+Ly-6G+myeloid cells. J Immunol,2010.185(3):p. 1383-92.
    50. Jan, M., et al., Prospective separation of normal and leukemic stem cells based on differential expression of TIM3, a human acute myeloid leukemia stem cell marker. Proc Natl Acad Sci U S A,2011.108(12):p.5009-14.
    51. Kikushige, Y., et al., TIMS is a promising target to selectively kill acute myeloid leukemia stem cells. Cell Stem Cell,2010.7(6):p.708-17.
    52. Huang, X., et al., Lymphoma endothelium preferentially expresses Tim-3 and facilitates the progression of lymphoma by mediating immune evasion. J Exp Med, 2010.207(3):p.505-20.
    53. Wada, J. and Y.S. Kanwar, Identification and characterization of galectin-9, a novel beta-galactoside-binding mammalian lectin. J Biol Chem,1997.272(9):p. 6078-86.
    54. Zhu, C., et al., The Tim-3 ligand galectin-9 negatively regulates T helper type I immunity. Nat Immunol,2005.6(12):p.1245-52.
    55. Imaizumi, T., et al., Interferon-gamma stimulates the expression of galectin-9 in cultured human endothelial cells. J Leukoc Biol,2002.72(3):p.486-91.
    56. Sanchez-Fueyo, A., et al., Tim-3 inhibits T helper type 1-mediated auto-and alloimmune responses and promotes immunological tolerance. Nat Immunol,2003. 4(11):p.1093-101.
    57. Koguchi, K., et al., Dysregulated T cell expression of TIM3 in multiple sclerosis. J Exp Med,2006.203(6):p.1413-8.
    58. Hastings, W.D., et al., TIM-3 is expressed on activated human CD4+T cells and regulates Thl and Th17 cytokines. Eur J Immunol,2009.39(9):p.2492-501.
    59. Seki, M., et al., Galectin-9 suppresses the generation of Thl 7, promotes the induction of regulatory T cells, and regulates experimental autoimmune arthritis. Clin Immunol,2008.127(1):p.78-88.
    60. Golden-Mason, L., et al., Negative immune regulator Tim-3 is overexpressed on T cells in hepatitis C virus infection and its blockade rescues dysfunctional CD4+ and CD8+ T cells. J Virol,2009.83(18):p.9122-30.
    61. Wherry, E.J., T cell exhaustion. Nat Immunol,2011.12(6):p.492-9.
    62. Ahmadzadeh, M., et al., Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood,2009.114(8):p. 1537-44.
    63. Gehring, A.J., et al., Profile of tumor antigen-specific CD8 T cells in patients with hepatitis B virus-related hepatocellular carcinoma. Gastroenterology,2009.137(2): p.682-90.
    64. Yamamoto, R., et al., PD-1-PD-1 ligand interaction contributes to immunosuppressive microenvironment of Hodgkin lymphoma. Blood,2008.111(6): p.3220-4.
    65. Blank, C., et al., Blockade of PD-L1 (B7-H1) augments human tumor-specific T cell responses in vitro. Int J Cancer,2006.119(2):p.317-27.
    66. Baitsch, L., et al., Exhaustion of tumor-specific CD8(+) T cells in metastases from melanoma patients. J Clin Invest,2011.121(6):p.2350-60.
    67. Schmitz, J., et al., IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity,2005.23(5):p.479-90.
    68. Moussion, C., N. Ortega, and J.P. Girard, The IL-1-like cytokine IL-33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo:a novel 'alarmin'? PLoS One,2008.3(10):p. e3331.
    69. Yanagawa, Y., et al., Prostaglandin E(2) enhances IL-33 production by dendritic cells. Immunol Lett,2011.141(1):p.55-60.
    70. Haraldsen, G., et al., Interleukin-33-cytokine of dual function or novel alarmin? Trends Immunol,2009.30(5):p.227-33.
    71. Chackerian, A.A., et al., IL-1 receptor accessory protein and ST2 comprise the IL-33 receptor complex. J Immunol,2007.179(4):p.2551-5.
    72. Palmer, G., et al., The IL-1 receptor accessory protein (AcP) is required for IL-33 signaling and soluble AcP enhances the ability of soluble ST2 to inhibit IL-33. Cytokine,2008.42(3):p.358-64.
    73. Ali, S., et al., IL-1 receptor accessory protein is essential for IL-33-induced activation of T lymphocytes and mast cells. Proc Natl Acad Sci U S A,2007. 104(47):p.18660-5.
    74. Tago, K., et al., Tissue distribution and subcellular localization of a variant form of the human ST2 gene product, ST2V. Biochem Biophys Res Commun,2001.285(5): p.1377-83.
    75. Li, H., et al., The cloning and nucleotide sequence of human ST2L cDNA. Genomics,2000.67(3):p.284-90.
    76. Bergers, G, et al., Alternative promoter usage of the Fos-responsive gene Fit-1 generates mRNA isoforms coding for either secreted or membrane-bound proteins related to the IL-1 receptor. EMBO J,1994.13(5):p.1176-88.
    77. Jovanovic, I.P., et al., IL-33/ST2 axis in innate and acquired immunity to tumors. Oncoimmunology,2012.1(2):p.229-231.
    78. Roussel, L., et al., Molecular mimicry between IL-33 and KSHV for attachment to chromatin through the H2A-H2B acidic pocket. EMBO Rep,2008.9(10):p. 1006-12.
    79. Carriere, V., et al., IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo. Proc Natl Acad Sci U S A,2007. 104(1):p.282-7.
    80. Joshi, A.D., et al., Interleukin-33 contributes to both Ml and M2 chemokine marker expression in human macrophages. BMC Immunol,2010.11:p.52.
    81. Mun, S.H., et al., Interleukin-33 stimulates formation of functional osteoclasts from human CD14(+) monocytes. Cell Mol Life Sci,2010.67(22):p.3883-92.
    82. Ho, L.H., et al., IL-33 induces IL-13 production by mouse mast cells independently of IgE-FcepsilonRI signals. J Leukoc Biol,2007.82(6):p.1481-90.
    83. Komai-Koma, M., et al., IL-33 is a chemoattractant for human Th2 cells. Eur J Immunol,2007.37(10):p.2779-86.
    84. Iikura, M., et al., IL-33 can promote survival, adhesion and cytokine production in human mast cells. Lab Invest,2007.87(10):p.971-8.
    85. Coyle, A.J., et al., Crucial role of the interleukin 1 receptor family member T1/ST2 in T helper cell type 2-mediated lung mucosal immune responses. J Exp Med,1999. 190(7):p.895-902.
    86. Lohning, M., et al., T1/ST2 is preferentially expressed on murine Th2 cells, independent of interleukin 4, interleukin 5, and interleukin 10, and important for Th2 effeetor function.Proc Natl Acad Sci U S A,1998.95(12):p.6930-5. (?)
    87. Kurowska-Stolarska, M., et al., IL-33 amplifies the polarization of alternatively activated macrophages that contribute to airway inflammation. J Immunol,2009. 183(10):p.6469-77.
    88. Yang, Z., et al., Association of increased serum IL-33 levels with clinical and laboratory characteristics of systemic lupus erythematosus in Chinese population. Clin Exp Med,2011.11(2):p.75-80.
    89. Mok, M.Y., et al., Serum levels of IL-33 and soluble ST2 and their association with disease activity in systemic lupus erythematosus. Rheumatology (Oxford),2010. 49(3):p.520-7.
    90. Matsuyama, Y., et al., Increased levels of interleukin 33 in sera and synovial fluid frompatients with active rheumatoid arthritis. J Rheumatol,2010.37(1):p.18-25.
    91. Espinassous, Q., et al., IL-33 enhances lipopolysaccharide-induced inflammatory cytokine production from mouse macrophages by regulating lipopolysaccharide receptor complex. J Immunol,2009.183(2):p.1446-55.
    92. Yang, Q., et al., IL-33 synergizes with TCR and IL-12 signaling to promote the effector function of CD8+ T cells. Eur J Immunol,2011.41(11):p.3351-60.
    93. Lee, S., et al., Profiling of transcripts and proteins modulated by K-ras oncogene in the lung tissues of K-ras transgenic mice by omics approaches. Int J Oncol, 2009.34(1):p.161-72.
    94. Wu, J., et al., Identification and functional analysis of 9p24 amplified genes in human breast cancer. Oncogene,2012.31(3):p.333-41.
    95. Sun, P., et al., Serum interleukin-33 levels in patients with gastric cancer. Dig Dis Sci,2011.56(12):p.3596-601.
    96. Sabates-Bellver, J., et al., Transcriptome profile of human colorectal adenomas. Mol Cancer Res,2007.5(12):p.1263-75.
    97. Hong, Y, et al., A susceptibility gene set for early onset colorectal cancer that integrates diverse signaling pathways:implication for tumorigenesis. Clin Cancer Res,2007.13(4):p.1107-14.
    1. Galon, J., et al., Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science,2006.313(5795):p.1960-4.
    2. Zou, W., Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer,2005.5(4):p.263-74.
    3. Zou, W., Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol,2006.6(4):p.295-307.
    4. Fourcade, J., et al., Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+T cell dysfunction in melanoma patients. J Exp Med, 2010.207(10):p.2175-86.
    5. Sakuishi, K., et al., Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med,2010.207(10):p. 2187-94.
    6. Hastings, W.D., et al., TIM-3 is expressed on activated human CD4+T cells and regulates Thl and Th17 cytokines. Eur J Immunol,2009.39(9):p.2492-501.
    7. Anderson, A.C., et al., Promotion of tissue inflammation by the immune receptor Tim-3 expressed on innate immune cells. Science,2007.318(5853):p.1141-3.
    8. Monney, L., et al., Thl-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature,2002.415(6871):p. 536-41.
    9. Zhu, C., et al., The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol,2005.6(12):p.1245-52.
    10. Sabatos, C.A., et al., Interaction of Tim-3 and Tim-3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat Immunol,2003.4(11):p. 1102-10.
    11. Sanchez-Fueyo, A., et al., Tim-3 inhibits T helper type 1-mediated auto-and alloimmune responses and promotes immunological tolerance. Nat Immunol,2003. 4(11):p.1093-101.
    12. Jones, R.B., et al., Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HTV-1 infection. J Exp Med, 2008.205(12):p.2763-79.
    13. Wang, L., et al., Programmed death 1 ligand signaling regulates the generation of adaptive Foxp3+ CD4+ regulatory T cells. Proc Natl Acad Sci U S A,2008. 105(27):p.9331-6.
    14. Boenisch, O., et al., TIM-3 a novel regulatory molecule of alloimmune activation. J Immunol,2010.185(10):p.5806-19.
    15. Seki, M., et al., Galectin-9 suppresses the generation of Th17, promotes the induction of regulatory T cells, and regulates experimental autoimmune arthritis. Clin Immunol,2008.127(1):p.78-88.
    1. Galon, J., et al., Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science,2006.313(5795):p.1960-4.
    2. Zou, W., Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer,2005.5(4):p.263-74.
    3. Zou, W., Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol,2006.6(4):p.295-307.
    4. Frey, A.B. and N. Monu, Signaling defects in anti-tumor T cells. Immunol Rev, 2008.222:p.192-205.
    5. Liew, F.Y., N.I. Pitman, and I.B. McInnes, Disease-associated functions of IL-33: the new kid in the IL-1 family. Nat Rev Immunol,2010.10(2):p.103-10.
    6. Yanagawa, Y., et al., Prostaglandin E(2) enhances IL-33 production by dendritic cells. Immunol Lett,2011.141(1):p.55-60.
    7. Haraldsen, G., et al., Interleukin-33-cytokine of dual function or novel alarmin? Trends Immunol,2009.30(5):p.227-33.
    8. Schmitz, J., et al., IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity,2005.23(5):p.479-90.
    9. Chackerian, A.A., et al., IL-1 receptor accessory protein and ST2 comprise the IL-33 receptor complex. J Immunol,2007.179(4):p.2551-5.
    10. Ali, S., et al., IL-1 receptor accessory protein is essential for IL-33-induced activation of T lymphocytes and mast cells. Proc Natl Acad Sci U S A,2007. 104(47):p.18660-5.
    11. Palmer, G, et al., The IL-1 receptor accessory protein (AcP) is required for IL-33 signaling and soluble AcP enhances the ability of soluble ST2 to inhibit IL-33. Cytokine,2008.42(3):p.358-64.
    12. Jovanovic, I.P., et al., IL-33/ST2 axis in innate and acquired immunity to tumors. Oncoimmunology,2012.1(2):p.229-231.
    13. Joshi, A.D., et al., Interleukin-33 contributes to both M1 and M2 chemokine marker expression in human macrophages. BMC Immunol,2010.11:p.52.
    14. Espinassous, Q., et al., IL-33 enhances lipopolysaccharide-induced inflammatory cytokine production from mouse macrophages by regulating lipopolysaccharide receptor complex. J Immunol,2009.183(2):p.1446-55.
    15. Yang, Q., et al., IL-33 synergizes with TCR and IL-12 signaling to promote the effector function of CD8+ T cells. Eur J Immunol,2011.41(11):p.3351-60.
    16. Wu, J., et al., Identification and functional analysis of 9p24 amplified genes in human breast cancer. Oncogene,2012.31(3):p.333-41.
    17. Sun, P., et al., Serum interleukin-33 levels in patients with gastric cancer. Dig Dis Sci,2011.56(12):p.3596-601.
    18. Sabates-Bellver, J., et al., Transcriptome profile of human colorectal adenomas. Mol Cancer Res,2007.5(12):p.1263-75.
    19. Hong, Y., et al., A susceptibility gene set for early onset colorectal cancer that integrates diverse signaling pathways:implication for tumorigenesis. Clin Cancer Res,2007.13(4):p.1107-14.
    20. Lee, S., et al., Profiling of transcripts and proteins modulated by K-ras oncogene in the lung tissues of K-ras transgenic mice by omics approaches. Int J Oncol, 2009.34(1):p.161-72.
    21. Yibei Zhu, et al., T-bet and eomesodermin are required for T cell-mediated antitumor immune response. J. Immunol.2010.185:3174-3183.