The blockade of immune checkpoints in cancer immunotherapy

Nature Reviews Cancer - Tập 12 Số 4 - Trang 252-264 - 2012
Drew M. Pardoll1
1Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center, CRB1 Room 444, 1650 Orleans Street, Baltimore, Maryland 21287, USA. [email protected]

Tóm tắt

Từ khóa


Tài liệu tham khảo

Greenwald, R. J., Freeman, G. J. & Sharpe, A. H. The B7 family revisited. Annu. Rev. Immunol. 23, 515–548 (2005).

Zou, W & Chen, L. Inhibitory B7-family molecules in the tumour microenvironment. Nature Rev. Immunol. 8, 467–477 (2008). A good review of the basic biology of the expanding B7 family of regulatory molecules in the context of cancer immunology.

Mellor, A. L., Keskin, D. B., Johnson, T., Chandler, P. & Munn, D. H. Cells expressing indoleamine 2,3-dioxygenase inhibit T cell responses. J. Immunol. 168, 3771–3776 (2002).

Friberg, M. et al. Indoleamine 2,3-dioxygenase contributes to tumor cell evasion of T cell-mediated rejection. J. Cancer 101, 151–155 (2002).

Hou, D. Y. et al. Inhibition of indoleamine 2,3-dioxygenase in dendritic cells by stereoisomers of 1-methyl-tryptophan correlates with antitumor responses. Cancer Res. 67, 792–801 (2007).

Munn, D. H. & Mellor, A. L. Indoleamine 2,3-dioxygenase and tumor-induced tolerance. J. Clin. Invest. 17, 1147–1154 (2007).

Bak, S. P., Alonso, A., Turk, M. J. & Berwin, B. Murine ovarian cancer vascular leukocytes require arginase-1 activity for T cell suppression. Mol. Immunol. 46, 258–268 (2008).

Ochoa, A. C., Zea, A. H., Hernandez, C. & Rodriguez, P. C. Arginase, prostaglandins, and myeloid-derived suppressor cells in renal cell carcinoma. Clin. Cancer Res. 13, 721–726 (2007).

Rodríguez, P. C. & Ochoa, A. C. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol. Rev. 222, 180–191 (2008).

Löb, S. et al. IDO1 and IDO2 are expressed in human tumors: levo- but not dextro-1-methyl tryptophan inhibits tryptophan catabolism. Cancer Immunol. Immunother. 58, 153–157 (2009).

Qian, F. et al. Efficacy of levo-1-methyl tryptophan and dextro-1-methyl tryptophan in reversing indoleamine-2, 3-dioxygenase-mediated arrest of T-cell proliferation in human epithelial ovarian cancer. Cancer Res. 69, 5498–5504 (2009).

Reisser, D., Onier-Cherix, N. & Jeannin, J. F. Arginase activity is inhibited by L-NAME, both in vitro and in vivo. J. Enzyme Inhib. Med. Chem. 17, 267–270 (2002).

Schwartz, R. H. Costimulation of T lymphocytes: the role of CD28, CTLA-4, and B7/BB1 in interleukin-2 production and immunotherapy. Cell 71, 1065–1068 (1992).

Lenschow, D. J., Walunas, T. L. & Bluestone, J. A. CD28/B7 system of T cell costimulation. Annu. Rev. Immunol. 14, 233–258 (1996).

Rudd, C. E., Taylor, A. & Schneider, H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol. Rev. 229, 12–26 (2009).

Hathcock, K. S. et al. Identification of an alternative CTLA-4 ligand costimulatory for T cell activation. Science 262, 905–907 (1993).

Freeman, G. J. et al. Cloning of B7–2: a CTLA-4 counter-receptor that costimulates human T cell proliferation. Science 262, 909–911 (1993).

Azuma, M. et al. B70 antigen is a second ligand for CTLA-4 and CD28. Nature 366, 76–79 (1993).

Linsley, P. S., Clark, E. A. & Ledbetter, J. A. T-cell antigen CD28 mediates adhesion with B cells by interacting with activation antigen B7/BB-1. Proc. Natl Acad. Sci. USA 87, 5031–5035 (1990).

Linsley, P. S. et al. CTLA-4 is a second receptor for the B cell activation antigen B7. J. Exp. Med. 174, 561–569 (1991).

Linsley, P. S. et al. Human B7–1 (CD80) and B7–2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity 1, 793–801 (1994).

Riley, J. L. et al. Modulation of TCR-induced transcriptional profiles by ligation of CD28, ICOS, and CTLA-4 receptors. Proc. Natl Acad. Sci. USA 99, 11790–11795 (2002).

Schneider, H. et al. Reversal of the TCR stop signal by CTLA-4. Science 313, 1972–1975 (2006).

Egen, J. G. & Allison, J. P. Cytotoxic T lymphocyte antigen-4 accumulation in the immunological synapse is regulated by TCR signal strength. Immunity 16, 23–35 (2002).

Parry, R. V. et al. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol. Cell Biol. 25, 9543–9553 (2005).

Schneider, H. et al. Cutting edge: CTLA-4 (CD152) differentially regulates mitogen-activated protein kinases (extracellular signal-regulated kinase and c-Jun N-terminal kinase) in CD4+ T cells from receptor/ligand-deficient mice. J. Immunol. 169, 3475–3479 (2002).

Qureshi, O. S. et al. Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science 332, 600–603 (2011).

Tivol, E. A. et al. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 3, 541–547 (1995).

Waterhouse, P. et al. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science 270, 985–988 (1995). References 28 and 29 show the dramatic immune phenotype of Ctla4 -knockout mice, proving that CTLA4 is a crucial immune-checkpoint receptor in the immune system.

Wing, K. et al. CTLA-4 control over Foxp3+ regulatory T cell function. Science 322, 271–275 (2008). A T Reg cell-specific Ctla4 -knockout approach to prove that CTLA4 has a role in T Reg cell function that is independent of its role in modulating the amplitude of effector T cell activation.

Peggs, K. S., Quezada, S. A., Chambers, C. A., Korman, A. J. & Allison, J. P. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J. Exp. Med. 206, 1717–1725 (2009). A transgenic approach to prove that CTLA4 has independent roles in T Reg cell and effector T cell function.

Hori, S., Nomura, T. & Sakaguchi, S. Control of regulatory T cell development by the transcription factor Foxp3. Science 299, 1057–1061 (2003).

Fontenot, J. D., Gavin, M. A. & Rudensky, A. Y. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nature Immunol. 4, 330–336 (2003). References 32 and 33 define FOXP3 as a crucial transcription factor that mediates T Reg cell function.

Hill, J. A. et al. Foxp3 transcription-factor-dependent and -independent regulation of the regulatory T cell transcriptional signature. Immunity 27, 786–800 (2007).

Gavin, M. A. et al. Foxp3-dependent programme of regulatory T-cell differentiation. Nature 445, 771–775 (2007).

Leach, D. R., Krummel, M. F. & Allison, J. P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 271, 1734–1736 (1996). The first demonstration in mouse models of the ability of CTLA4 antibodies to induce therapeutic antitumour immunity.

van Elsas, A., Hurwitz, A. A. & Allison, J. P. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J. Exp. Med. 190, 355–366 (1999).

Hodi, F. S. et al. Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc. Natl Acad. Sci. USA 100, 4712–4717 (2003). The first clinical report of CTLA4 antibody treatment of cancer patients.

Phan, G. Q. et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc. Natl Acad. Sci. USA 100, 8372–8377 (2003). The first clear demonstration of tumour regressions induced by anti-CTLA4 therapy in patients with melanoma.

Ribas, A. et al. Antitumor activity in melanoma and anti-self responses in a phase I trial with the anti-cytotoxic T lymphocyte-associated antigen 4 monoclonal antibody CP-675,206. J. Clin. Oncol. 23, 8968–8977 (2005).

Beck, K. E. et al. Enterocolitis in patients with cancer after antibody blockade of cytotoxic T-lymphocyte-associated antigen 4. J. Clin. Oncol. 24, 2283–2289 (2006).

Ribas, A. Clinical development of the anti-CTLA-4 antibody tremelimumab. Semin. Oncol. 37, 450–454 (2010).

Downey, S. G. et al. Prognostic factors related to clinical response in patients with metastatic melanoma treated by CTL-associated antigen-4 blockade. Clin. Cancer Res. 13, 6681–6688 (2007).

Hodi, F. S. et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363, 711–723 (2010). A crucial Phase III trial demonstrating statistical survival benefit in patients with melanoma that were treated with the anti-CTLA4 therapy, ipilimumab.This was the first drug to demonstrate survival benefit in patients with advanced melanoma in a randomized trial.

Yuan, J. et al. Integrated NY-ESO-1 antibody and CD8+ T-cell responses correlate with clinical benefit in advanced melanoma patients treated with ipilimumab. Proc. Natl Acad. Sci. USA. 108, 16723–16728 (2011).

Liakou, C. I. et al. CTLA-4 blockade increases IFNγ-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc. Natl Acad. Sci. USA 105, 14987–14992 (2008).

Ji, R. R et al. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol. Immunother. 07 Dec 2011 (doi:10.1007/s00262-011-1172-6).

Hoos, A. et al. Improved endpoints for cancer immunotherapy trials. J. Natl Cancer Inst. 102, 1388–1397 (2010).

Ishida, Y., Agata, Y., Shibahara, K. & Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 11, 3887–3895 (1992).

Freeman, G. J. et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 192, 1027–1034 (2000). The demonstration that PD1 is the receptor for PDL1.

Keir, M. E. et al. Tissue expression of PD-L1 mediates peripheral T cell tolerance. J. Exp. Med. 203, 883–895 (2006).

Nishimura, H. et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 291, 319–322 (2001).

Nishimura, H., Nose, M., Hiai, H., Minato, N. & Honjo, T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 11, 141–151 (1999). The first demonstration that PD1 downmodulates immune responses.

Okazaki, T. & Honjo, T. PD-1 and PD-1 ligands: from discovery to clinical application. Int. Immunol. 19, 813–824 (2007).

Keir, M. E., Butte, M. J., Freeman, G. J. & Sharpe, A. H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 26, 677–704 (2008).

Dong, H. et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nature Med. 8, 793–800 (2002). The crucial demonstration that PDL1 is upregulated on tumour cells and that PDL1 expression on tumour cells mediates immune resistance.

Blank, C. et al. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res. 64, 1140–1145 (2004).

Taube, J. M. et al. B7-H1 expression co-localizes with inflammatory infiltrates in benign and malignant melanocytic lesions: implications for immunotherapy. Sci. Transl. Med. (In the press, 2012). Introduction of the adaptive immune resistance hypothesis that proposes that the expression of immune-checkpoint ligand by tumours can be induced by an antitumour immune response as an adaptive immune-protective mechanism.

Fife, B. T. et al. Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal. Nature Immunol. 10, 1185–1192 (2009).

Francisco, L. M. et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J. Exp. Med. 206, 3015–3029 (2009).

Dong, H., Zhu, G., Tamada, K. & Chen, L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nature Med. 5, 1365–1369 (1999). The discovery of PDL1.

Latchman, Y. et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nature Immunol. 2, 261–268 (2001).

Tseng, S. Y. et al. B7-DC, a new dendritic cell molecule with potent costimulatory properties for T cells. J. Exp. Med. 193, 839–846 (2001).

Butte, M. J., Keir, M. E., Phamduy, T. B., Sharpe, A. H. & Freeman, G. J. Programmed death-1 ligand 1 interacts specifically with the B7–1 costimulatory molecule to inhibit T cell responses. Immunity 27, 111–122 (2007).

Paterson, A. M. et al. The programmed death-1 ligand 1:B7–1 pathway restrains diabetogenic effector T cells in vivo. J. Immunol. 187, 1097–1105 (2011).

Park, J. J. et al. B7-H1/CD80 interaction is required for the induction and maintenance of peripheral T-cell tolerance. Blood 116, 1291–1298 (2010).

Shin, T. et al. In vivo costimulatory role of B7-DC in tuning T helper cell 1 and cytotoxic T lymphocyte responses. J. Exp. Med. 201, 1531–1541 (2005).

Terme, M. et al. IL-18 induces PD-1-dependent immunosuppression in cancer. Cancer Res. 71, 5393–5399 (2011).

Fanoni, D. et al. New monoclonal antibodies against B-cell antigens: possible new strategies for diagnosis of primary cutaneous B-cell lymphomas. Immunol. Lett. 134, 157–160 (2011).

Velu, V. et al. Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature 458, 206–210 (2009).

Barber, D. L. et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439, 682–687 (2006). The demonstration that the PD1 pathway mediates T cell 'exhaustion'. This paper investigated chronic viral infection but the mechanism is probably applicable to tumour-induced immune tolerance.

Sfanos, K. S. et al. Human prostate-infiltrating CD8+ T lymphocytes are oligoclonal and PD-1+. Prostate 69, 1694–1703 (2009).

Ahmadzadeh, M. et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 114, 1537–1544 (2009).

Iwai, Y. et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc. Natl Acad. Sci. USA 99, 12293–12297 (2002).

Konishi, J. et al. B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression. Clin. Cancer Res. 10, 5094–5100 (2004).

Brown, J. A. et al. Blockade of programmed death-1 ligands on dendritic cells enhances T cell activation and cytokine production. J. Immunol. 170, 1257–1266 (2003).

Curiel, T. J. et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nature Med. 9, 562–567 (2003).

Kuang, D. M. et al. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J. Exp. Med. 206, 1327–1337 (2009).

Liu, Y., Zeng, B., Zhang, Z., Zhang, Y. & Yang, R. B7-H1 on myeloid-derived suppressor cells in immune suppression by a mouse model of ovarian cancer. Clin. Immunol. 129, 471–481 (2008).

Thompson, R. H. et al. Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target. Proc. Natl Acad. Sci. USA 101, 17174–17179 (2004).

Hamanishi, J. et al. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc. Natl Acad. Sci. USA 104, 3360–3365 (2007).

Ohigashi, Y. et al. Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand-2 expression in human esophageal cancer. Clin. Cancer Res. 11, 2947–2953 (2005).

Wu, C. et al. Immunohistochemical localization of programmed death-1 ligand-1 (PD-L1) in gastric carcinoma and its clinical significance. Acta Histochem. 108, 19–24 (2006).

Ghebeh, H. et al. The B7-H1 (PD-L1) T lymphocyte-inhibitory molecule is expressed in breast cancer patients with infiltrating ductal carcinoma: correlation with important high-risk prognostic factors. Neoplasia 8, 190–198 (2006).

Hino, R. et al. Tumor cell expression of programmed cell death-1 ligand 1 is a prognostic factor for malignant melanoma. Cancer 116, 1757–1766 (2010).

Rosenwald, A. et al. Molecular diagnosis of primary mediastinal B cell lymphoma identifies a clinically favorable subgroup of diffuse large B cell lymphoma related to Hodgkin lymphoma. J. Exp. Med. 198, 851–862 (2003).

Steidl, C. et al. MHC class II transactivator CIITA is a recurrent gene fusion partner in lymphoid cancers. Nature 471, 377–381 (2011).

Parsa, A. T. et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nature Med. 13, 84–88 (2007). The first evidence of an oncogenic pathway causing the induction of PDL1 expression on tumour cells.

Marzec, M. et al. Oncogenic kinase NPM/ALK induces through STAT3 expression of immunosuppressive protein CD274 (PD-L1, B7-H1). Proc. Natl Acad. Sci. USA 105, 20852–20857 (2008).

Kim, J. et al. Constitutive and inducible expression of b7 family of ligands by human airway epithelial cells. Am. J. Respir. Cell. Mol. Biol. 33, 280–289 (2005).

Lee, S. K. et al. IFN-γ regulates the expression of B7-H1 in dermal fibroblast cells. J. Dermatol. Sci. 40, 95–103 (2005).

Wilke, C. M. et al. Dual biological effects of the cytokines interleukin-10 and interferon-γ. Cancer Immunol. Immunother. 60, 1529–1541 (2011).

Gajewski, T. F., Louahed, J. & Brichard, V. G. Gene signature in melanoma associated with clinical activity: a potential clue to unlock cancer immunotherapy. Cancer J. 16, 399–403 (2010).

Brahmer, J. R. et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J. Clin. Oncol. 28, 3167–3175 (2010). The first report of the treatment of patients and preliminary clinical efficacy of PD1 antibodies in multiple cancer histologies.

Sznol, M. et al. Safety and antitumor activity of biweekly MDX 1106 (Anti PD 1) in patients with advanced refractory malignancies. J. Clin. Oncol. 28 (Suppl.), Abstract 2506 (2010).

McDermott, D. F. et al. A phase I study to evaluate safety and antitumor activity of biweekly MDX 1106 (Anti PD 1) in patients with RCC and other advanced refractory malignancies. J. Clin. Oncol. 29 (Suppl. 7), Abstract 331 (2011).

Yi, K. H. & Chen, L. Fine tuning the immune response through B7-H3 and B7-H4. Immunol. Rev. 229, 145–151 (2009).

He, C., Qiao, H., Jiang, H. & Sun, X. The inhibitory role of B7-H4 in antitumor immunity: association with cancer progression and survival. Clin. Dev. Immunol. 2011, 695834 (2011).

Huang, C. T. et al. Role of LAG-3 in regulatory T cells. Immunity 21, 503–513 (2004). The identification of LAG3 as an immune-checkpoint receptor.

Triebel, F. et al. LAG-3, a novel lymphocyte activation gene closely related to CD4. J. Exp. Med. 171, 1393–1405 (1990).

Goldberg, M. V. & Drake, C. G. LAG-3 in cancer immunotherapy. Curr. Top. Microbiol. Immunol. 344, 269–278 (2011).

Grosso, J. F. et al. LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. J. Clin. Invest. 117, 3383–3392 (2007).

Blackburn, S. D. et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nature Immunol. 10, 29–37 (2009).

Grosso, J. F. et al. Functionally distinct LAG-3 and PD-1 subsets on activated and chronically stimulated CD8 T cells. J. Immunol. 182, 6659–6669 (2009).

Woo, S. R. et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 72, 917–927 (2012).

Zhu, C. et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nature Immunol. 6, 1245–1252 (2005). The demonstration that TIM3 is an immune-checkpoint receptor.

Ngiow, S. F. et al. Anti-TIM3 antibody promotes T cell IFN-γ-mediated antitumor immunity and suppresses established tumors. Cancer Res. 71, 3540–3551 (2011).

Sakuishi, K. et al. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J. Exp. Med. 207, 2187–2194 (2010).

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. 207, 2175–2186 (2010).

Baitsch, L. et al. Extended co-expression of inhibitory receptors by human CD8 T-cells depending on differentiation, antigen-specificity and anatomical localization. PLoS ONE 7, e30852. (2012).

Watanabe, N. et al. BTLA is a lymphocyte inhibitory receptor with similarities to CTLA-4 and PD-1. Nature Immunol. 4, 670–679 (2003).

Sedy, J. R. et al. B and T lymphocyte attenuator regulates T cell activation through interaction with herpesvirus entry mediator. Nature Immunol. 6, 90–98 (2005).

Lasaro, M. O. et al. Active immunotherapy combined with blockade of a coinhibitory pathway achieves regression of large tumor masses in cancer-prone mice. Mol. Ther. 19, 1727–1736 (2011).

Fourcade, J. et al. CD8+ T cells specific for tumor antigens can be rendered dysfunctional by the tumor microenvironment through upregulation of the inhibitory receptors BTLA and PD-1. Cancer Res. 72, 887–896 (2012).

Zarek, P. E. et al. A2A receptor signaling promotes peripheral tolerance by inducing T-cell anergy and the generation of adaptive regulatory T cells. Blood 111, 251–259 (2008).

Deaglio, S. et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J. Exp. Med. 204, 1257–1265 (2007).

Waickman, A. T. et al. Enhancement of tumor immunotherapy by deletion of the A(2A) adenosine receptor. Cancer Immunol. Immunother. 25 Nov 2011 (doi:10.1007/s00262-011-1155-7).

Lanier, L. L. Up on the tightrope: natural killer cell activation and inhibition. Nature Immunol. 9, 495–502 (2008).

Plougastel, B. F. & Yokoyama, W. M. Extending missing-self? Functional interactions between lectin-like NKrp1 receptors on NK cells with lectin-like ligands. Curr. Top. Microbiol. Immunol. 298, 77–89 (2006).

Raulet, D. H. Missing self recognition and self tolerance of natural killer (NK) cells. Semin. Immunol. 18, 145–150 (2006).

Mingari, M. C., Pietra, G. & Moretta, L. Human cytolytic T lymphocytes expressing HLA class-I-specific inhibitory receptors. Curr. Opin. Immunol. 17, 312–319 (2005).

Li, B. et al. Anti-programmed death-1 synergizes with granulocyte macrophage colony-stimulating factor—secreting tumor cell immunotherapy providing therapeutic benefit to mice with established tumors. Clin. Cancer Res. 15, 1623–1634 (2009).

Vanneman, M. & Dranoff, G. Combining immunotherapy and targeted therapies in cancer treatment. Nature Rev. Cancer 12, 237–251 (2012).