Abstract
Recent clinical data support ideas of Programmed death receptor-ligand 1 (PD-L1; also called B7-H1, CD274) playing an important role in immune evasion of tumor cells. Expression of PD-L1 on tumors strongly correlates with the survival of cancer patients. PD-L1 on tumors interacts with the co-inhibitory molecule Programmed death receptor-1 (PD-1, CD279) on T cells mediating decreased TCR-mediated proliferation and cytokine production. In animal tumor models, blockade of PD-L1/PD-1 interactions resulted in an improved tumor control. In addition, exhausted T cells during chronic viral infections could be revived by PD-L1 blockade. Thus, targeting PD-L1/PD-1 interactions might improve the efficacy of adoptive cell therapies (ACT) of chronic infections as well as cancers. Obstacles for a general blockade of PD-L1 might be its role in mediating peripheral tolerance. This review discusses the currently available data concerning the role of PD-L1 in tumor immune evasion and envisions possibilities for implementation into ACT for cancer patients.
Keywords: Immune Evasion, Peripheral Tolerance, Chronic Viral Infection, Adoptive Cell Therapy, Recent Clinical Data
Introduction
Cancer immunotherapies, either based on vaccination with tumor associated antigens (TAA) or adoptive cell therapies (ACT) using TAA-specific T cells, have attracted immunologists as a possible alternative or as an addition to conventional therapies (surgery, chemotherapy and radiation). Despite sufficient homing to tumor sites [1], tumor associated antigen (TAA)-specific T cells so far seldom control tumor growth in a clinical setting [2-4].
The reason for this is the generally accepted notion that the tumor microenvironment can protect tumor cells from immune destruction. Soluble factors as well as membrane-bound molecules, including transforming growth factor β (TGF-β), interleukin (IL)-10, prostaglandin E2, FAS, CTLA-4 ligands and tumor necrosis factor related inducing ligand (TRAIL) have been found to be expressed by tumors and have been postulated to mediate immune evasion [5-7].
Recently, various molecules have been identified, that can modulate TCR signals [8, 9]. These include besides the well known CD28 and CTLA-4, the recently described molecules PD-1, ICOS and BTLA. Except the latter one, all interact with B7 family ligands. Besides B7-1 and -2, PD-L1 (B7-H1), PD-L2 (B7-DC), ICOS-L (B7-H2, B7 h), B7-H3 and B7-H4 (B7x, B7-S1) have been identified so far.
Within this group of molecules, PD-L1 and B7-H4 are thought to predominately mediate inhibitory signals towards T cells. The strong expression of PD-L1 on various tumors led us and other groups to the hypothesis of this ligand playing an important role in immune evasion of cancer cells [7, 10-12]. Support for our ideas evolves from recent clinical data on long-term follow-up of renal cell carcinoma (RCC) patients [13], and data from esophageal, gastric and breast cancer patients indicate a correlation of the prognosis with an increased expression of PD-L1 on the cancer cells [14-16]. Thus, a PD-L1/PD-1 blockade might revert the immuno-compromised state of TAA-specific T cells allowing an increase in the efficacy of ACT.
PD-1 and PD-L1/PD-L2
PD-1 is a transmembrane receptor of the Ig superfamily that lacks the relevant motif for binding to B7-1 and B7-2 [17]. It is found to be expressed on thymocytes [18, 19], mature T and B cells following activation [8, 17, 20] and on myeloid cells [18]. Compared to the restricted expression of CTLA-4, this wide expression of PD-1 suggests a broader role in immune regulation [21, 22].
Two ligands for PD-1 (CD279), both belonging to the B7 family, have been identified: PD-L1 (B7-H1, CD274) and PD-L2 (B7-DC, CD273) [23, 24]. Interaction of PD-1 with PD-L1 or PD-L2 has been described to negative regulate cytokine production and proliferation of T cells [23, 25-27]. This inhibitory effect has been shown for CD4+ as well as for CD8+ T cells [23, 26]. Whereas Carter et al. found that murine CD8+ T cells are more susceptible to PD-L1 inhibition, we found human CD4+ T cells transfected with TAA-specific TCR more susceptible compared to the CD8+ counterparts [28]. In addition, some other reports identified co-stimulatory functions of these ligands, possibly mediated via an unidentified receptor different from PD-1 [29-33]. The discordant signaling effects might result from differences in the surface expression of PD-1, a different yet unidentified receptor, or from interfering signals from PD-L2 that has been also shown to mediate inhibitory or co-stimulatory effects [24, 30, 32, 34]. In addition, PD-1 is known to induce tolerance by resting dendritic cells [35]. Whereas many of the tumor models examined the direct interaction of the T-cell with the tumor, recent data indicate that the blockade of PD-L1 also led to improved control of the tumor growth in the presence of antigen presentation by DCs [12].
The cytoplasmatic tail of PD-1 contains an immuno-receptor tyrosine-based inhibitory motif (ITIM) and an immunoreceptor tyrosine switch motif (ITSM). Whereas the former is thought to mediate inhibitory signals, mutational studies indicate that the latter motif is responsible for signaling after PD-1 ligation [36]. This result opens a second explanation for the contrary results after PD-L1/PD-1 interaction. As the ITSM-motif has been shown to be capable of mediating co-stimulatory as well as co-inhibitory signals, depending on the presence or absence of another adaptor molecule (SH2D1A), PD-1 signals might depend on the activation status of the T cell. Indeed, we found WT CD8+ T cells to be superior to PD-1 gene-deficient CD8+ T cells concerning cytokine production in a naïve but not activated status (Blank et al. unpublished data), which, however, does not exclude the theory so far of a second receptor signaling early in activation.
At least in vivo, the status of the antigen-presenting cell (APC) will also alter the outcome of the TCR ligation, as it seems that the inhibitory effects of PD-L1 are mediated predominantly from resting or virus-treated DC, but not from activated DC [35, 37], which might reconcile at least some of the contrary findings.
In vivo models examining the graft survival and diabetes onset using either PD1-/- NOD mice, PD-L1Ig in islet transplantation or PD-L1tg over-expression in pancreatic islets resulted in discordant conclusions [33, 38, 39]. Besides the discussed different activation settings of the T cells in those models, different strain background of the animal models could also play a role for the discordant findings [40].
The variable expression of PD-1 on the T-cell surface, the restricted availability of agonistic PD-1 monoclonal antibodies and the low affinity of the ligand fusion protein hinders so far definitive experiments to delineate the co-stimulatory and/or co-inhibitory effects.
PD-L1 and peripheral tolerance/autoimmunity
The co-inhibitory effects of PD-1 were initially suggested because PD-1-/- mice developed, depending on the strain background, spontaneous autoimmune diseases, such as lupus-like glomerulonephritis, arthritis, gastritis or dilatative cardiomyopathy [41-44]. The effect of PD-L1/PD-1 interaction on immunological tolerance has been examined in 2C and HY TCR transgenic mice. It was found that over-expression of PD-1 inhibits, or in the absence of PD-1 increases, the positive selection [45, 46]. In contrast, we found that on an RAG-/- background, the absence of PD-1 altered the percentage of death by neglect allowing increased numbers of DN T cells to occur in the periphery, but also increased the negative selection, which was not observed in the H–Y system [19]. Using PD-1-/- mice crossed to PD-L1-/- or PD-L2-/- clearly demonstrated the role of PD-L1/PD-1 interaction during thymic maturation. The perception that the total numbers of 2C T cells is not changed on an autoreactive background by PD-1 deficiency leads to the conclusion of a normal central tolerance in PD-1 sufficient mice [37]. Under the light of the above-mentioned data showing alterations of the positive selection, the negative selection and death by neglect by PD-1, the previous conclusion needs to be examined further.
Independent of these considerations, the main function of the PD-L1/PD-1 pathway seems to be the regulation of peripheral tolerance [18]. This notion is supported by the various autoimmune diseases found in PD-1 gene-deficient mice. In addition, the observed expression of PD-L1 on human and murine endothelia [47-49], at sites of inflammation in experimental autoimmune encephalomyelitis [50] and in muscle biopsies from patients with inflammatory myopathies [51], supports its postulated role in regulation of peripheral tolerance. The absence of PD-L1 indeed increased the induction of experimental autoimmune hepatitis [52], which could be mediated by the absence of PD-L1, not only on Kupfer cells and monocyte-derived cells within the liver [11, 53], but also by PD-L1 on the liver parechym itself [54]. A definitive proof of the role of peripheral tolerance is the role of PD-L1 in the feto-maternal tolerance. It has been earlier shown that PD-L1 is strongly expressed at this immunological important barrier [55], but just recently data proved the functional relevance in preventing abortion in a mouse model [56].
PD-L1 and T-cell exhaustion during chronic viral infections
During chronic viral infections, functional virus-specific T cells are induced; however, they gradually lose their function during the course of the infection. IFN-γ is a key player in host defences against viral infections, but also has been shown to up-regulate PD-L1 expression predominantly on non-lymphoid tissues, due to the PD-L1 promotor region containing several IFN-γ-responsive elements [11, 49].
Thus, it has been self-evident to examine the role of the PDL-1/PD-1 pathway during chronic viral infections. Recently, R. Ahmed et al. elegantly showed that antiviral immune functions could be restored in vivo by a blockade of PD-L1 in the LCMV infection model. By doing so, they also proved that PD-L1/PD-1 interaction plays a pivotal role during CD8+ T-cell exhaustion from chronic antigenic stimuli [57].
This mechanism might be extended to other chronic viral infections in humans, such as human immunodeficiency (HIV). Indeed, PD-L1 has been shown to be up-regulated during HIV-infection [58] and several recently published studies suggest a role for the PD-1-PD-L1 pathway in exhaustion of virus-specific CD8+ T cells during HIV infection [59-61]. In addition, these groups could show that the PD-1 expression correlates with disease progression and blockade of PD-L1/PD-1 pathway could restore immune functions of the T cells.
It seems that there is a distinct hierarchy of exhaustion, as that with the duration of the infection, the level of antigen exposure increases, the CD4 help decreases and PD-1 expression on antigen-specific T cells increases [62]. Whether these ideas also could be transferred on chronic antigen exposure during ACT in cancer patients needs to be further evaluated.
PD-L1 and immune evasion of tumor cells
Whereas the initial broad detection of PD-L1 was based on mRNA levels, its surface expression was detected only on T- and B-cells, dendritic cells, macrophages, endothelia, epithelia, cardiac myocard, pancreatic islet cells, glial cells, inflamed muscle, keratinocytes and the feto-maternal barrier [20]. Interestingly, PD-L1 surface expression can also be found on almost all tumor entities. It has been detected by immunohistochemistry, for example, on the surface of human cancers of larynx, lung, stomach, colon, breast, cervix, ovary, renal cell, bladder, liver, glioma and melanoma [11, 25, 63]. In vitro experiments indicate that many tumor cell lines also express PD-L1 and/or up-regulate PD-L1 surface expression upon exposure to IFN-γ [11, 20, 63]. This expression is in strong contrast to the expression to B7-1 and B7-2, which are seldom found on tumors. The attributed inhibitory effect of PD-L1/PD-1 interaction earlier led to the hypothesis that tumors escape from the host immune system by negative attenuation of tumor-specific T-cell responses via the PD-L1/PD-1 pathway [7, 64].
In vitro experiments using PD-L1 over-expressing murine tumor cell lines and blocking antibodies against PD-L1 and PD-1 clearly demonstrated that PD-L1 on tumor cells suppresses the cytolytic activity of CD8+ T cells [7, 12]. In addition, others and we could show that endogenous PD-L1 expression on tumor cells are also capable of suppressing T-cell functions, e.g., proliferation and cytokine production [11, 20].
Transfers to in vivo experiments using blocking antibodies of PD-1, PD-L1 or TCR-transgenic mice crossed to PD-1-/- mice revealed accelerated tumor eradication in the absence of PD-L1/PD-1 interaction [7, 12, 20, 65]. In addition, PD-L1 blockade has been shown to suppress tumor metastasis using melanoma or colon cancer cell lines [66].
Similar to murine studies, others and we recently found PD-L1 to be expressed on human tumor tissues and human tumor lines and to be up-regulated upon IFN-γ exposure. Over-expression of human PD-L1 or interference using anti-PD-L1 blocking antibodies support the notion of tumor-expressed PD-L1 being capable of suppressing T-cell immune functions [11, 28, 63, 67].
Whereas some studies did not indicate a significant relevance of PD-L1 expression for the patients-prognosis in non-small lung cancer [68] or even did not detect PD-L1 at all on acute myeloid leukemia [69], we found in contrast to the latter, frequent expression of PD-L1 on leukemic blasts (Blank et al. unpublished data). Furthermore, a recent paper revealed prognostic value of PD-L1 expression for breast cancer [16], esopgageal cancer [14] and gastric carcinomas [15]. The strongest correlation between cancer patients-survival and tumor-associated PD-L1 expression has been shown within a group of over 300 patients that underwent nephrectomy for clear cell renal carcinoma [13, 70]. Survival rates at 1, 5 and 10 years post-nephrectomy in patients expressing PD-L1 within the primary tumor (compared to PD-L1 negative tumor, in brackets) were 77.3% (94.6%), 41.9% (82.9%) and 36.7% (77.4%). Univariate analysis of these data indicate that patients with a PD-L1-positive tumor nearly four times more likely die from RCC compared to patients with a PD-L1-negative tumor [13].
Based on these in vitro data and the correlation of the clinical prognosis of patients with PD-L1 expression in the tumor microenvironment, it is very likely that the PD-1/PD-L1 pathway plays a pivotal role in tumor-immune evasion from the endogenous immune system, as well as from adoptively transferred tumor-specific or vaccination-induced T cells.
Considerations concerning PD-L1 blockade and ACT
Tumor-specific T cells have been shown to be dysfunctional or anergised [71] and just recently Blankenstein et al. have elegantly shown that immunogenic tumors indeed induced T-cell anergy allowing immune evasion [72]. Continuous exposure to antigen in tumor patients might therefore exhaust T cells, similar to chronic viral infections [57]. Despite differences between chronic antigen stimulation during viral infections and malignancies (tumors seldom express very immunogenic antigens, IFN-γ induction is more likely during viral infections, connective tissues barriers in tumors, etc.) it is tempting to speculate that immunogenic tumors might induce anergy of tumor-specific T cells by expressing PD-L1 on their surface [37]. In the light of many immunotherapy protocols aiming at induction of a type-I immune response [73, 74], even IFN-γ-induced PD-L1 up-regulation on tumor cells might be accountable for the so far often disappointing outcome of tumor immunotherapies. The blockade of the PD-L1/PD-1 pathway might therefore either revert an immuno-compromised state of the cancer patients or prevent inhibition of adoptively transferred T cells during ACT and allow the immune system to eradicate the tumors.
Interfering into the PD-L1/PD-1 pathway by, e.g., an anti-PD-L1 monoclonal antibody has been even in our considerations [10]. However, PD-L1’s broad expression on endothelia and its important role in regulation of peripheral tolerance and thus prevention of autoimmunity [18, 37, 75] will require a more specific blockade of the PD-L1/PD-1 pathway or at least extensive testing of binding to normal tissues before in vivo applications.
In addition, the difference between PD-L1 and PD-L2, which share many features, but with contrary outcomes resulting from their blockade, is still poorly understood. Thus, it could be problematic blocking selectively PD-L1 and in doing so allowing an unrestricted PD-L2-mediated, possibly co-stimulatory pathway to act on a broad range of resting peripheral T cells.
Therefore, we would attribute a preference for the blockade of PD-1 on selected tumor- or virus-specific T cells. However, in this case, in advance of such approaches, the bidirectional roles of PD-1 toward co-stimulatory versus co-inhibitory effects have to be dissected clearly.
Acknowledgment
Dr. Christian Blank is supported by the Deutsche Krebshilfe e.V. Grant No. 10-2204-Bl 1.
Footnotes
This article is a symposium paper from the conference “Cancer Immunotherapy 2006 Meets Strategies for Immune Therapy,-held in Mainz, Germany, on 4- May 2006.
References
- 1.Meidenbauer N, Marienhagen J, Laumer M, Vogl S, Heymann J, Andreesen R, Mackensen A. Survival and tumor localization of adoptively transferred melan-a-specific T cells in melanoma patients. J Immunol. 2003;170:2161–2169. doi: 10.4049/jimmunol.170.4.2161. [DOI] [PubMed] [Google Scholar]
- 2.Dudley ME, Wunderlich JR, Robbins PF, Yang JC, Hwu P, Schwartzentruber DJ, Topalian SL, Sherry R, Restifo NP, Hubicki AM, Robinson MR, Raffeld M, Duray P, Seipp CA, Rogers-Freezer L, Morton KE, Mavroukakis SA, White DE, Rosenberg SA. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science. 2002;298:850–854. doi: 10.1126/science.1076514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, Royal RE, Topalian SL, Kammula US, Restifo NP, Zheng Z, Nahvi A, de Vries CR, Rogers-Freezer LJ, Mavroukakis SA, Rosenberg SA. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006;314:126–129. doi: 10.1126/science.1129003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Mackensen A, Meidenbauer N, Vogl S, Laumer M, Berger J, Andreesen R. Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T cells for treatment of patients with metastatic melanoma. J Clin Oncol. 2006;24:5060–5069. doi: 10.1200/JCO.2006.07.1100. [DOI] [PubMed] [Google Scholar]
- 5.Smyth MJ, Godfrey DI, Trapani JA. A fresh look at tumor immunosurveillance and immunotherapy. Nat Immunol. 2001;2:293–299. doi: 10.1038/86297. [DOI] [PubMed] [Google Scholar]
- 6.Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3:991–998. doi: 10.1038/ni1102-991. [DOI] [PubMed] [Google Scholar]
- 7.Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. 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. 2002;99:12293–12297. doi: 10.1073/pnas.192461099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Carreno BM, Collins M. The B7 family of ligands and its receptors: new pathways for costimulation and inhibition of immune responses. Annu Rev Immunol. 2002;20:29–53. doi: 10.1146/annurev.immunol.20.091101.091806. [DOI] [PubMed] [Google Scholar]
- 9.Carreno BM, Collins M. BTLA: a new inhibitory receptor with a B7-like ligand. Trends Immunol. 2003;24:524–527. doi: 10.1016/j.it.2003.08.005. [DOI] [PubMed] [Google Scholar]
- 10.Blank C, Gajewski TF, Mackensen A. Interaction of PD-L1 on tumor cells with PD-1 on tumor-specific T cells as a mechanism of immune evasion: implications for tumor immunotherapy. Cancer Immunol Immunother. 2005;54:307–314. doi: 10.1007/s00262-004-0593-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, Lennon VA, Celis E, Chen L. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800. doi: 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
- 12.Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, Rietz C, Flies DB, Lau JS, Zhu G, Tamada K, Chen L. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005;65:1089–1096. [PubMed] [Google Scholar]
- 13.Thompson RH, Kuntz SM, Leibovich BC, Dong H, Lohse CM, Webster WS, Sengupta S, Frank I, Parker AS, Zincke H, Blute ML, Sebo TJ, Cheville JC, Kwon ED. Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. Cancer Res. 2006;66:3381–3385. doi: 10.1158/0008-5472.CAN-05-4303. [DOI] [PubMed] [Google Scholar]
- 14.Ohigashi Y, Sho M, Yamada Y, Tsurui Y, Hamada K, Ikeda N, Mizuno T, Yoriki R, Kashizuka H, Yane K, Tsushima F, Otsuki N, Yagita H, Azuma M, Nakajima Y. Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand-2 expression in human esophageal cancer. Clin Cancer Res. 2005;11:2947–2953. doi: 10.1158/1078-0432.CCR-04-1469. [DOI] [PubMed] [Google Scholar]
- 15.Wu C, Zhu Y, Jiang J, Zhao J, Zhang XG, Xu N. Immunohistochemical localization of programmed death-1 ligand-1 (PD-L1) in gastric carcinoma and its clinical significance. Acta Histochem. 2006;108:19–24. doi: 10.1016/j.acthis.2006.01.003. [DOI] [PubMed] [Google Scholar]
- 16.Ghebeh H, Mohammed S, Al-Omair A, Qattan A, Lehe C, Al-Qudaihi G, Elkum N, Alshabanah M, Bin Amer S, Tulbah A, Ajarim D, Al-Tweigeri T, Dermime S. 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. 2006;8:190–198. doi: 10.1593/neo.05733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubata T, Yagita H, Honjo T. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 1996;8:765–772. doi: 10.1093/intimm/8.5.765. [DOI] [PubMed] [Google Scholar]
- 18.Nishimura H, Honjo T. PD-1: an inhibitory immunoreceptor involved in peripheral tolerance. Trends Immunol. 2001;22:265–268. doi: 10.1016/S1471-4906(01)01888-9. [DOI] [PubMed] [Google Scholar]
- 19.Blank C, Brown I, Marks R, Nishimura H, Honjo T, Gajewski TF. Absence of programmed death receptor 1 alters thymic development and enhances generation of CD4/CD8 double-negative TCR-transgenic T cells. J Immunol. 2003;171:4574–4581. doi: 10.4049/jimmunol.171.9.4574. [DOI] [PubMed] [Google Scholar]
- 20.Blank C, Brown I, Peterson AC, Spiotto M, Iwai Y, Honjo T, Gajewski TF. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res. 2004;64:1140–1145. doi: 10.1158/0008-5472.CAN-03-3259. [DOI] [PubMed] [Google Scholar]
- 21.Okazaki T, Iwai Y, Honjo T. New regulatory co-receptors: inducible co-stimulator and PD-1. Curr Opin Immunol. 2002;14:779–782. doi: 10.1016/S0952-7915(02)00398-9. [DOI] [PubMed] [Google Scholar]
- 22.Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol. 2005;23:515–548. doi: 10.1146/annurev.immunol.23.021704.115611. [DOI] [PubMed] [Google Scholar]
- 23.Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, Horton HF, Fouser L, Carter L, Ling V, Bowman MR, Carreno BM, Collins M, Wood CR, Honjo T. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027–1034. doi: 10.1084/jem.192.7.1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, Iwai Y, Long AJ, Brown JA, Nunes R, Greenfield EA, Bourque K, Boussiotis VA, Carter LL, Carreno BM, Malenkovich N, Nishimura H, Okazaki T, Honjo T, Sharpe AH, Freeman GJ. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2:261–268. doi: 10.1038/85330. [DOI] [PubMed] [Google Scholar]
- 25.Brown JA, Dorfman DM, Ma FR, Sullivan EL, Munoz O, Wood CR, Greenfield EA, Freeman GJ. Blockade of programmed death-1 ligands on dendritic cells enhances T cell activation and cytokine production. J Immunol. 2003;170:1257–1266. doi: 10.4049/jimmunol.170.3.1257. [DOI] [PubMed] [Google Scholar]
- 26.Carter L, Fouser LA, Jussif J, Fitz L, Deng B, Wood CR, Collins M, Honjo T, Freeman GJ, Carreno BM. PD-1:PD-L inhibitory pathway affects both CD4(+) and CD8(+) T cells and is overcome by IL-2. Eur J Immunol. 2002;32:634–643. doi: 10.1002/1521-4141(200203)32:3<634::AID-IMMU634>3.0.CO;2-9. [DOI] [PubMed] [Google Scholar]
- 27.Selenko-Gebauer N, Majdic O, Szekeres A, Hofler G, Guthann E, Korthauer U, Zlabinger G, Steinberger P, Pickl WF, Stockinger H, Knapp W, Stockl J. B7-h1 (programmed death-1 ligand) on dendritic cells is involved in the induction and maintenance of T cell anergy. J Immunol. 2003;170:3637–3644. doi: 10.4049/jimmunol.170.7.3637. [DOI] [PubMed] [Google Scholar]
- 28.Blank C, Kuball J, Voelkl S, Wiendl H, Becker B, Walter B, Majdic O, Gajewski TF, Theobald M, Andreesen R, Mackensen A. Blockade of PD-L1 (B7-H1) augments human tumor-specific T cell responses in vitro. Int J Cancer. 2006;119:317–327. doi: 10.1002/ijc.21775. [DOI] [PubMed] [Google Scholar]
- 29.Tamura H, Dong H, Zhu G, Sica GL, Flies DB, Tamada K, Chen L. B7-H1 costimulation preferentially enhances CD28-independent T-helper cell function. Blood. 2001;97:1809–1816. doi: 10.1182/blood.V97.6.1809. [DOI] [PubMed] [Google Scholar]
- 30.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. Nat Med. 1999;5:1365–1369. doi: 10.1038/70932. [DOI] [PubMed] [Google Scholar]
- 31.Youngnak P, Kozono Y, Kozono H, Iwai H, Otsuki N, Jin H, Omura K, Yagita H, Pardoll DM, Chen L, Azuma M. Differential binding properties of B7-H1 and B7-DC to programmed death-1. Biochem Biophys Res Commun. 2003;307:672–677. doi: 10.1016/S0006-291X(03)01257-9. [DOI] [PubMed] [Google Scholar]
- 32.Wang S, Bajorath J, Flies DB, Dong H, Honjo T, Chen L. Molecular modeling and functional mapping of B7-H1 and B7-DC uncouple costimulatory function from PD-1 interaction. J Exp Med. 2003;197:1083–1091. doi: 10.1084/jem.20021752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Subudhi SK, Zhou P, Yerian LM, Chin RK, Lo JC, Anders RA, Sun Y, Chen L, Wang Y, Alegre ML, Fu YX. Local expression of B7-H1 promotes organ-specific autoimmunity and transplant rejection. J Clin Invest. 2004;113:694–700. doi: 10.1172/JCI19210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Tseng SY, Otsuji M, Gorski K, Huang X, Slansky JE, Pai SI, Shalabi A, Shin T, Pardoll DM, Tsuchiya H. B7-DC, a new dendritic cell molecule with potent co-stimulatory properties for T cells. J Exp Med. 2001;193:839–846. doi: 10.1084/jem.193.7.839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Probst HC, McCoy K, Okazaki T, Honjo T, van den Broek M. Resting dendritic cells induce peripheral CD8+ T cell tolerance through PD-1 and CTLA-4. Nat Immunol. 2005;6:280–286. doi: 10.1038/ni1165. [DOI] [PubMed] [Google Scholar]
- 36.Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol. 2004;173:945–954. doi: 10.4049/jimmunol.173.2.945. [DOI] [PubMed] [Google Scholar]
- 37.Okazaki T, Honjo T. The PD-1-PD-L pathway in immunological tolerance. Trends Immunol. 2006;27:195–201. doi: 10.1016/j.it.2006.02.001. [DOI] [PubMed] [Google Scholar]
- 38.Wang J, Yoshida T, Nakaki F, Hiai H, Okazaki T, Honjo T. Establishment of NOD-Pdcd1-/- mice as an efficient animal model of type I diabetes. Proc Natl Acad Sci USA. 2005;102:11823–11828. doi: 10.1073/pnas.0505497102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Gao W, Demirci G, Strom TB, Li XC. Stimulating PD-1-negative signals concurrent with blocking CD154 co-stimulation induces long-term islet allograft survival. Transplantation. 2003;76:994–999. doi: 10.1097/01.TP.0000085010.39567.FB. [DOI] [PubMed] [Google Scholar]
- 40.Khoury SJ, Sayegh MH. The roles of the new negative T cell costimulatory pathways in regulating autoimmunity. Immunity. 2004;20:529–538. doi: 10.1016/S1074-7613(04)00116-5. [DOI] [PubMed] [Google Scholar]
- 41.Nishimura H, Minato N, Nakano T, Honjo T. Immunological studies on PD-1 deficient mice: implication of PD-1 as a negative regulator for B cell responses. Int Immunol. 1998;10:1563–1572. doi: 10.1093/intimm/10.10.1563. [DOI] [PubMed] [Google Scholar]
- 42.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. 1999;11:141–151. doi: 10.1016/S1074-7613(00)80089-8. [DOI] [PubMed] [Google Scholar]
- 43.Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, Matsumori A, Sasayama S, Mizoguchi A, Hiai H, Minato N, Honjo T. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science. 2001;291:319–322. doi: 10.1126/science.291.5502.319. [DOI] [PubMed] [Google Scholar]
- 44.Okazaki T, Tanaka Y, Nishio R, Mitsuiye T, Mizoguchi A, Wang J, Ishida M, Hiai H, Matsumori A, Minato N, Honjo T. Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat Med. 2003;9:1477–1483. doi: 10.1038/nm955. [DOI] [PubMed] [Google Scholar]
- 45.Nishimura H, Honjo T, Minato N. Facilitation of beta election and modification of positive selection in the thymus of PD-1-deficient mice. J Exp Med. 2000;191:891–898. doi: 10.1084/jem.191.5.891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Keir ME, Latchman YE, Freeman GJ, Sharpe AH. Programmed death-1 (PD-1):PD-ligand 1 interactions inhibit TCR-mediated positive selection of thymocytes. J Immunol. 2005;175:7372–7379. doi: 10.4049/jimmunol.175.11.7372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Eppihimer MJ, Gunn J, Freeman GJ, Greenfield EA, Chernova T, Erickson J, Leonard JP. Expression and regulation of the PD-L1 immunoinhibitory molecule on microvascular endothelial cells. Microcirculation. 2002;9:133–145. doi: 10.1038/sj/mn/7800123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Iwai Y, Terawaki S, Ikegawa M, Okazaki T, Honjo T. PD-1 inhibits antiviral immunity at the effector phase in the liver. J Exp Med. 2003;198:39–50. doi: 10.1084/jem.20022235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Mazanet MM, Hughes CC. B7-H1 is expressed by human endothelial cells and suppresses T cell cytokine synthesis. J Immunol. 2002;169:3581–3588. doi: 10.4049/jimmunol.169.7.3581. [DOI] [PubMed] [Google Scholar]
- 50.Saint-Ruf C, Ungewiss K, Groettrup M, Bruno L, Fehling HJ, von Boehmer H. Analysis and expression of a cloned pre-T cell receptor gene. Science. 1994;266:1208–1212. doi: 10.1126/science.7973703. [DOI] [PubMed] [Google Scholar]
- 51.Wiendl H, Mitsdoerffer M, Schneider D, Chen L, Lochmuller H, Melms A, Weller M. Human muscle cells express a B7-related molecule, B7-H1, with strong negative immune regulatory potential: a novel mechanism of counterbalancing the immune attack in idiopathic inflammatory myopathies. FASEB J. 2003;17:1892–1894. doi: 10.1096/fj.03-0039fje. [DOI] [PubMed] [Google Scholar]
- 52.Dong H, Zhu G, Tamada K, Flies DB, van Deursen JM, Chen L. B7-H1 determines accumulation and deletion of intrahepatic CD8(+) T lymphocytes. Immunity. 2004;20:327–336. doi: 10.1016/S1074-7613(04)00050-0. [DOI] [PubMed] [Google Scholar]
- 53.Yu MC, Chen CH, Liang X, Wang L, Gandhi CR, Fung JJ, Lu L, Qian S. Inhibition of T-cell responses by hepatic stellate cells via B7-H1-mediated T-cell apoptosis in mice. Hepatology. 2004;40:1312–1321. doi: 10.1002/hep.20488. [DOI] [PubMed] [Google Scholar]
- 54.Muhlbauer M, Fleck M, Schutz C, Weiss T, Froh M, Blank C, Scholmerich J, Hellerbrand C. PD-L1 is induced in hepatocytes by viral infection and by interferon-alpha and gamma and mediates T cell apoptosis. J Hepatol. 2006;45:520–528. doi: 10.1016/j.jhep.2006.05.007. [DOI] [PubMed] [Google Scholar]
- 55.Petroff MG, Chen L, Phillips TA, Azzola D, Sedlmayr P, Hunt JS. B7 family molecules are favorably positioned at the human maternal-fetal interface. Biol Reprod. 2003;68:1496–1504. doi: 10.1095/biolreprod.102.010058. [DOI] [PubMed] [Google Scholar]
- 56.Guleria I, Khosroshahi A, Ansari MJ, Habicht A, Azuma M, Yagita H, Noelle RJ, Coyle A, Mellor AL, Khoury SJ, Sayegh MH. A critical role for the programmed death ligand 1 in fetomaternal tolerance. J Exp Med. 2005;202:231–237. doi: 10.1084/jem.20050019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, Ahmed R. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439:682–687. doi: 10.1038/nature04444. [DOI] [PubMed] [Google Scholar]
- 58.Trabattoni D, Saresella M, Biasin M, Boasso A, Piacentini L, Ferrante P, Dong H, Maserati R, Shearer GM, Chen L, Clerici M. B7-H1 is up-regulated in HIV infection and is a novel surrogate marker of disease progression. Blood. 2003;101:2514–2520. doi: 10.1182/blood-2002-10-3065. [DOI] [PubMed] [Google Scholar]
- 59.Trautmann L, Janbazian L, Chomont N, Said EA, Gimmig S, Bessette B, Boulassel MR, Delwart E, Sepulveda H, Balderas RS, Routy JP, Haddad EK, Sekaly RP. Upregulation of PD-1 expression on HIV-specific CD8(+) T cells leads to reversible immune dysfunction. Nat Med. 2006;12:1198–1202. doi: 10.1038/nm1482. [DOI] [PubMed] [Google Scholar]
- 60.Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, Mackey EW, Miller JD, Leslie AJ, DePierres C, Mncube Z, Duraiswamy J, Zhu B, Eichbaum Q, Altfeld M, Wherry EJ, Coovadia HM, Goulder PJ, Klenerman P, Ahmed R, Freeman GJ, Walker BD. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature. 2006;443:350–354. doi: 10.1038/nature05115. [DOI] [PubMed] [Google Scholar]
- 61.Petrovas C, Casazza JP, Brenchley JM, Price DA, Gostick E, Adams WC, Precopio ML, Schacker T, Roederer M, Douek DC, Koup RA. PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection. J Exp Med. 2006;203:2281–2292. doi: 10.1084/jem.20061496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Freeman GJ, Wherry EJ, Ahmed R, Sharpe AH. Reinvigorating exhausted HIV-specific T cells via PD-1-PD-1 ligand blockade. J Exp Med. 2006;203:2223–2227. doi: 10.1084/jem.20061800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wintterle S, Schreiner B, Mitsdoerffer M, Schneider D, Chen L, Meyermann R, Weller M, Wiendl H. Expression of the B7-related molecule B7-H1 by glioma cells: a potential mechanism of immune paralysis. Cancer Res. 2003;63:7462–7467. [PubMed] [Google Scholar]
- 64.Dong H, Chen L. B7-H1 pathway and its role in the evasion of tumor immunity. J Mol Med. 2003;81:281–287. doi: 10.1007/s00109-003-0430-2. [DOI] [PubMed] [Google Scholar]
- 65.Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, Krzysiek R, Knutson KL, Daniel B, Zimmermann MC, David O, Burow M, Gordon A, Dhurandhar N, Myers L, Berggren R, Hemminki A, Alvarez RD, Emilie D, Curiel DT, Chen L, Zou W. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med. 2003;9:562–567. doi: 10.1038/nm863. [DOI] [PubMed] [Google Scholar]
- 66.Iwai Y, Terawaki S, Honjo T (2004) PD-1 blockade inhibits hematogenous spread of poorly immunogenic tumor cells by enhanced recruitment of effector T cells. Int Immunol [DOI] [PubMed]
- 67.Strome SE, Dong H, Tamura H, Voss SG, Flies DB, Tamada K, Salomao D, Cheville J, Hirano F, Lin W, Kasperbauer JL, Ballman KV, Chen L. B7-H1 blockade augments adoptive T-cell immunotherapy for squamous cell carcinoma. Cancer Res. 2003;63:6501–6505. [PubMed] [Google Scholar]
- 68.Konishi J, Yamazaki K, Azuma M, Kinoshita I, Dosaka-Akita H, Nishimura M. 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. 2004;10:5094–5100. doi: 10.1158/1078-0432.CCR-04-0428. [DOI] [PubMed] [Google Scholar]
- 69.Tamura H, Dan K, Tamada K, Nakamura K, Shioi Y, Hyodo H, Wang SD, Dong H, Chen L, Ogata K. Expression of functional B7-H2 and B7.2 costimulatory molecules and their prognostic implications in de novo acute myeloid leukemia. Clin Cancer Res. 2005;11:5708–5717. doi: 10.1158/1078-0432.CCR-04-2672. [DOI] [PubMed] [Google Scholar]
- 70.Thompson RH, Gillett MD, Cheville JC, Lohse CM, Dong H, Webster WS, Krejci KG, Lobo JR, Sengupta S, Chen L, Zincke H, Blute ML, Strome SE, Leibovich BC, Kwon ED. Costimulatory B7-H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. Proc Natl Acad Sci USA. 2004;101:17174–17179. doi: 10.1073/pnas.0406351101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Gajewski TF, Meng Y, Harlin H. Immune suppression in the tumor microenvironment. J Immunother. 2006;29:233–240. doi: 10.1097/01.cji.0000199193.29048.56. [DOI] [PubMed] [Google Scholar]
- 72.Willimsky G, Blankenstein T. Sporadic immunogenic tumours avoid destruction by inducing T-cell tolerance. Nature. 2005;437:141–146. doi: 10.1038/nature03954. [DOI] [PubMed] [Google Scholar]
- 73.Peterson AC, Harlin H, Gajewski TF. Immunization with melan-A peptide-pulsed peripheral blood mononuclear cells plus recombinant human interleukin-12 induces clinical activity and T-cell responses in advanced melanoma. J Clin Oncol. 2003;21:2342–2348. doi: 10.1200/JCO.2003.12.144. [DOI] [PubMed] [Google Scholar]
- 74.Spiotto MT, Yu P, Rowley DA, Nishimura MI, Meredith SC, Gajewski TF, Fu YX, Schreiber H. Increasing tumor antigen expression overcomes ``ignorance’-to solid tumors via crosspresentation by bone marrow-derived stromal cells. Immunity. 2002;17:737–747. doi: 10.1016/S1074-7613(02)00480-6. [DOI] [PubMed] [Google Scholar]
- 75.Dong H, Chen X. Immunoregulatory role of B7-H1 in chronicity of inflammatory responses. Cell Mol Immunol. 2006;3:179–187. [PMC free article] [PubMed] [Google Scholar]