Blockade of PD-1/PD-L1 promotes adoptive T-cell immunotherapy in a tolerogenic environment

Stephen J P Blake, Alan L H Ching, Tony J Kenna, Ryan Galea, Justin Large, Hideo Yagita, Raymond J Steptoe, Stephen J P Blake, Alan L H Ching, Tony J Kenna, Ryan Galea, Justin Large, Hideo Yagita, Raymond J Steptoe

Abstract

Adoptive cellular immunotherapy using in vitro expanded CD8+ T cells shows promise for tumour immunotherapy but is limited by eventual loss of function of the transferred T cells through factors that likely include inactivation by tolerogenic dendritic cells (DC). The co-inhibitory receptor programmed death-1 (PD-1), in addition to controlling T-cell responsiveness at effector sites in malignancies and chronic viral diseases is an important modulator of dendritic cell-induced tolerance in naive T cell populations. The most potent therapeutic capacity amongst CD8+ T cells appears to lie within Tcm or Tcm-like cells but memory T cells express elevated levels of PD-1. Based on established trafficking patterns for Tcm it is likely Tcm-like cells interact with lymphoid-tissue DC that present tumour-derived antigens and may be inherently tolerogenic to develop therapeutic effector function. As little is understood of the effect of PD-1/PD-L1 blockade on Tcm-like CD8+ T cells, particularly in relation to inactivation by DC, we explored the effects of PD-1/PD-L1 blockade in a mouse model where resting DC tolerise effector and memory CD8+ T cells. Blockade of PD-1/PD-L1 promoted effector differentiation of adoptively-transferred Tcm-phenotype cells interacting with tolerising DC. In tumour-bearing mice with tolerising DC, effector activity was increased in both lymphoid tissues and the tumour-site and anti-tumour activity was promoted. Our findings suggest PD-1/PD-L1 blockade may be a useful adjunct for adoptive immunotherapy by promoting effector differentiation in the host of transferred Tcm-like cells.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Blockade of PD-1/PD-L1 impairs induction…
Fig 1. Blockade of PD-1/PD-L1 impairs induction and partially reverses tolerance in naïve CD8+ T cells.
A) CD45.1+ OT-I T cells were transferred to 11c.OVA or C57BL/6 (non-Tg) mice treated with αPD-1, αPD-L1 or isotype control mAb at transfer and every subsequent three days. At the indicated time points after transfer spleens were collected and OT-I T cell number determined by a flow-cytometric counting assay. B-E): CD45.1+ OT-I T cells were transferred to C57BL/6 (non-Tg) or 11c.OVA mice and 21 days later mice were treated with αPD-1, αPD-L1 or isotype control mAb every three days. Six days after commencement of mAb treatment some mice were challenged with OVA/QuilA immunization and a further five days later mice were euthanized and tissues collected for analysis using a flow-cytometric counting assay and intracellular cytokine staining. (A) ***: for 11c.OVA recipients, αPD-L1 significantly greater than isotype control at day 3, 7, 14 (p<0.001), αPD-L1 significantly greater than αPD-1 at d7 (p<0.001) and d14 (p<0.05), αPD-1 greater than isotype at day 3 (p<0.001) and day 7, 14 (p<0.05). Data comprise: (A) four to seven mice for each time point (mean ± SEM) pooled from two or more experiments with 2–3 mice per group, (B-E) data pooled from 2 or 3 individual experiments with 1–2 mice per group (n = 4 for all groups) with values for individual mice shown.
Fig 2. Blockade of PD-1/PD-L1 impairs tolerance…
Fig 2. Blockade of PD-1/PD-L1 impairs tolerance in memory CD8+ T cells.
A, B) In vitro generated CD45.1+ OT-I memory T cells were transferred to 11c.OVA or C57BL/6 (non-Tg) mice treated with αPD-1, αPD-L1 or isotype control mAb at transfer and every subsequent three days. At the indicated time points spleens were collected and OT-I T cell number (A) and total IFN-γ-producing OT-I number (B) determined by a flow-cytometric counting assay and intracellular cytokine staining. C, D) In vitro generated CD45.1+ OT-I Tcm-phenotype cells were transferred to 11c.OVA or C57BL/6 (non-Tg) mice treated with αPD-1, αPD-L1 or isotype control mAb at transfer and every subsequent 3 days and seven (C) or 14 (D) days later CTL activity determined in vivo. Data comprise: (A) 4–6 mice for each time point (mean ± SEM) pooled from two or more experiments of 2–3 mice per group, ** 11c.OVA + αPD-L1 significantly greater than 11c.OVA + isotype and 11c.OVA + αPD-1 at day 7 (p<0.01), (B) 4–6 mice for each time point (mean ± SEM) pooled from more than two experiments of 2 mice per group, * 11c.OVA + αPD-L1 significantly greater than 11c.OVA + isotype and 11c.OVA + αPD-1 at day 3 (p<0.05), ** 11c.OVA + αPD-L1 significantly greater than 11c.OVA + isotype and 11c.OVA + αPD-1 at day 7 (p<0.01), (C, D) data pooled from 2 individual experiments of 2 mice per group with values for individual mice shown.
Fig 3. Blockade of PD-1/PD-L1 partially reverses…
Fig 3. Blockade of PD-1/PD-L1 partially reverses non-responsiveness of tolerised memory CD8+ T cells.
A, B) In vitro generated memory CD45.1+ OT-I T cells were transferred to 11c.OVA or C57BL/6 mice. Twenty eight days later mice were treated with αPD-1, αPD-L1 or isotype control mAb every subsequent three days. Nine days after commencement of mAb treatment some mice were challenged with OVA/QuilA immunization and a further five days later mice were euthanized and tissues collected for analysis using a flow-cytometric counting assay and intracellular cytokine staining. Data comprise: (A) four mice pooled from 4 experiments with n = 1 for each treatment set. Values for individual mice shown.
Fig 4. Anti-PD-1 and anti-PD-L1 synergises with…
Fig 4. Anti-PD-1 and anti-PD-L1 synergises with Tcm-phenotype cell transfer to a tolerogenic environment.
A) B16.mOVA cells were analysed for expression of PD-1 and PD-1 ligands. Histogram plots show specific antibody (solid line) and isotype control (dashed line) stained cells. Data are from a single analysis representative of 3 separate analyses. B) B16.mOVA cells (105) were injected s.c. to 11c.OVA mice. Mice were left untreated (●) or 3 days later OT-I Tcm-phenotype cells (5 x 106) transferred i.v. OT-I recipients were injected on the day of OT-I transfer and every subsequent 3 days with isotype control (◯), αPD-1 (▲) or αPD-L1 (▽) mAb. C-E) B16.mOVA cells (105, C; 0.5 x 105, D; 0.25 x 105, E) were injected s.c. to C57BL/6 (C) or 11c.OVA mice (D, E). Mice were left untreated (●) or 3 days later OT-I Tcm-phenotype cells (5 x 106) transferred i.v. OT-I recipients were injected on the day of OT-I transfer and every subsequent 3 days with isotype control (◯), αPD-1 (▲) or αPD-L1 (▽) mAb. Data show survival curves (left) or cumulative mean tumour area ± SEM (right).
Fig 5. Anti-PD-L1 treatment increases tumour-specific CD8…
Fig 5. Anti-PD-L1 treatment increases tumour-specific CD8+ T cell infiltration and promotes effector function within tumours.
A-G) B16.mOVA cells (105) were injected s.c. to 11c.OVA mice and 3 days later OT-I Tcm-phenotype cells (5 x 106) transferred i.v. OT-I recipients were injected on the day of OT-I transfer and every subsequent 3 days with isotype control or αPD-L1 mAb as indicated. Twelve days after OT-I Tcm-phenotype cell transfer, spleen (A, E), tumour draining LN (TDLN) (B), and tumour (C, D, F, G) were harvested and OT-I (CD45.1+/CD8+/Vα2+) number and cytokine production determined using a flow-cytometric counting assay (A, B, C, D) and intracellular cytokine staining (E, F) or a combination of both. Data represent individual mice pooled from 2 experiments of 2–3 mice per group.
Fig 6. Anti-PD-L1 treatment reduces exhaustion marker…
Fig 6. Anti-PD-L1 treatment reduces exhaustion marker expression on tumour-specific CD8+ T cells.
A-G) B16.mOVA cells (105) were injected s.c. to 11c.OVA mice and 3 days later OT-I Tcm-phenotype cells (5 x 106) transferred i.v. OT-I recipients were injected on the day of OT-I transfer and every subsequent 3 days with isotype control or αPD-L1 mAb as indicated. Twelve days after OT-I Tcm-phenotype cells transfer, spleen, tumour draining LN (TDLN), and tumour sites were harvested for flow-cytometric analysis. A) PD-L1 expression was determined in tumour, spleen and TDLN (A) gated on the total bulk population for isotype control mAb-injected (solid line) mice. Isotype-control staining of tumour is shown (dashed line). B, C) the proportion of DC expressing PD-L1 (B) and PD-L1 expression level on DC (C) was determined (gated on CD11c+I-Ab+ DC). CD62L and CD44 (D), PD-1 (E, F) and LAG-3 (G) expression was determined on OT-I T cells in spleen, TDLN and tumour. Data are representative of 3 mice/group in 2 separate experiments (A) or individual mice pooled from 2 separate experiments of 2–3 mice per group (B-G) except TDLN in (B, C) which is 3 mice from a single experiment of the 2 performed.

References

    1. Wolint P, Betts MR, Koup RA, Oxenius A. Immediate cytotoxicity but not degranulation distinguishes effector and memory subsets of CD8+ T cells. J Exp Med. 2004;199: 92–936. 10.1084/jem.20031799
    1. Klebanoff CA, Gattinoni L, Torabi-Parizi P, Kerstann K, Cardones AR, Finkelstein SE, et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc Natl Acad Sci U S A. 2005;102: 9571–9576. 10.1073/pnas.0503726102
    1. Klebanoff CA, Gattinoni L, Restifo NP. Sorting through subsets: which T-cell populations mediate highly effective adoptive immunotherapy? J Immunother. 2012;35: 651–660. 10.1097/CJI.0b013e31827806e6
    1. Wang A, Chandran S, Shah SA, Chiu Y, Paria BC, Aghamolla T, et al. The stoichiometric production of IL-2 and IFN-γ mRNA defines memory T cells that can self-renew after adoptive transfer in humans. Sci Transl Med. 2012;4 10.1126/scitranslmed.3004306
    1. Yang S, Gattinoni L, Liu F, Ji Y, Yu Z, Restifo NP, et al. In vitro generated anti-tumor T lymphocytes exhibit distinct subsets mimicking in vivo antigen-experienced cells. Cancer Immunol Immunother. 2011;60: 739–746. 10.1007/s00262-011-0977-7
    1. Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat Rev Immunol. 2012;12: 269–281. 10.1038/nri3191
    1. Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, Restifo NP, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol. 2005;23: 2346–2357. 10.1200/JCO.2005.00.240
    1. Zippelius A, Batard P, Rubio-Godoy V, Bioley G, Liénard D, Lejeune F, et al. Effector function of human tumor-specific CD8 T cells in melanoma lesions: a state of local functional tolerance. Cancer Res. 2004;64: 2865–2873. 10.1158/0008-5472.CAN-03-3066
    1. Yang C, Robbins PD. The Roles of Tumor-Derived Exosomes in Cancer Pathogenesis. Clin Dev Immunol. 2011;2011: 842849 10.1155/2011/842849
    1. McDonnell AM, Robinson BWS, Currie AJ. Tumor Antigen Cross-Presentation and the Dendritic Cell: Where it All Begins? Clin Dev Immunol. 2010;2010:539519 10.1155/2010/539519
    1. Kenna TJ, Thomas R, Steptoe RJ. Steady-state dendritic cells expressing cognate antigen terminate memory CD8+ T-cell responses. Blood. 2008;111: 2091–2100. 10.1182/blood-2007-07-103200
    1. Kenna TJ, Waldie T, McNally A, Thomson M, Yagita H, Thomas R, et al. Targeting antigen to diverse APCs inactivates memory CD8+ T cells without eliciting tissue-destructive effector function. Journal of Immunology. 2010;184: 598–606. 10.4049/jimmunol.0900032
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nature Reviews Cancer. 2012;12: 252–264. 10.1038/nrc3239
    1. Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8: 239–245. 10.1038/ni1443
    1. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26: 677–704. 10.1146/annurev.immunol.26.021607.090331
    1. Barber DL, Wherry EJ, Masupost D, Zhu B, Allison JP, Sharpe AH, et al. Restoring function in exhausted CD8+ T cells during chronic viral infection. Nature. 2006;439: 682–687. 10.1038/nature04444
    1. Tsushima F, Yao S, Shin T, Flies A, Flies S, Xu H, et al. Interaction between B7–H1 and PD-1 determines initiation and reversal of T-cell anergy. Blood. 2007;110: 180–185. 10.1182/blood-2006-11-060087
    1. Goldberg MV, Maris CH, Hipkiss EL, Flies AS, Zhen L, Tuder RM, et al. Role of PD-1 and its ligand, B7–H1, in early fate decisions of CD8 T cells. Blood. 2007;110: 186–192. 10.1182/blood-2006-12-062422
    1. Blank C, Brown I, Peterson AC, Spiotto M, Iwai Y, Honjo T, et al. 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. 10.1158/0008-5472.CAN-03-3259
    1. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366: 2455–2465. 10.1056/NEJMoa1200694
    1. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366: 2443–2454. 10.1056/NEJMoa1200690
    1. Zhou Q, Xiao H, Liu Y, Peng Y, Hong Y, Yagita H, et al. Blockade of programmed death-1 pathway rescues the effector function of tumor-infiltrating T cells and enhances the antitumor efficacy of lentivector immunization. J Immunol. 2010;185: 5082–5092. 10.4049/jimmunol.1001821
    1. Okazaki T, Chikuma S, Iwai Y, Fagarasan S, Honjo T. A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application. Nat Immunol. 2013;14: 1212–1218. 10.1038/ni.2762
    1. Ansari MJ, Salama AD, Chitnis T, Smith RN, Yagita H, Akiba H, et al. The programmed death-1 (PD-1) pathway regulates autoimmune diabetes in nonobese diabetic (NOD) mice. J Exp Med. 2003;198: 63–69. 10.1084/jem.20022125
    1. 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. 10.1038/ni1165
    1. Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 1999;401: 708–712. 10.1038/44385
    1. Weninger W, Crowley MA, Manjunath N, von Andrian UH. Migratory properties of naive, effector, and memory CD8(+) T cells. J Exp Med. 2001;194: 953–966. 10.1084/jem.194.7.953
    1. Hogquist KA, Jameson SC, Heath WR, Howard JL, Bevan MJ, Carbone FR. T cell receptor antagonist peptides induce positive selection. Cell. 1994;76: 17–27. 10.1016/0092-8674(94)90169-4
    1. Steptoe RJ, Ritchie JM, Wilson NS, Villadangos JA, Lew AM, Harrison LC. Cognate CD4+ help elicited by resting dendritic cells does not impair the induction of peripheral tolerance in CD8+ T cells. JI. 2007;178: 2094–2103. 10.4049/jimmunol.178.4.2094
    1. Fiorenza S, Kenna TJ, Comerford I, McColl S, Steptoe RJ, Leggatt GR, et al. A combination of local inflammation and central memory T cells potentiates immunotherapy in the skin J Immunol 2012;189: 5622–5631.
    1. Agata A, Kawasaki Y, Nishimura H, Ishida Y, Tsubata T, Yagita H, et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 1996; 8: 765–772. 10.1093/intimm/8.5.765
    1. Rodig N, Ryan T, Allen JA, Pang H, Grabie N, Chernova T, et al. Endothelial expression of PD-L1 and PD-L2 down-regulates CD8+ T cell activation and cytolysis. Eur J Immunol. 2003;33: 3117–3126. 10.1002/eji.200324270
    1. Takamura S, Tsuji-Kawahara S, Yagita H, Akiba H, Sakamoto M, Chikaishi T, et al. Premature terminal exhaustion of Friend virus-specific effector CD8+ T cells by rapid induction of multiple inhibitory receptors. J Immunol. 2010;184: 4696–4707. 10.4049/jimmunol.0903478
    1. Huang CT, Huso DL, Lu Z, Wang T, Zhou G, Kennedy EP, et al. CD4+ T cells pass through an effector phase during the process of in vivo tolerance induction. J Immunol. 2003;170: 3945–3953. 10.4049/jimmunol.170.8.3945
    1. Hernandez J, Aung S, Redmond WL, Sherman LA. Phenotypic and functional analysis of CD8(+) T cells undergoing peripheral deletion in response to cross-presentation of self-antigen. J Exp Med. 2001;194: 707–717. 10.1084/jem.194.6.707
    1. Huang X, Yang Y. Transient gain of effector function by CD8+ T cells undergoing peripheral tolerance to high-dose self-antigen. Eur J Immunol. 2004;34: 1351–1360. 10.1002/eji.200324734
    1. Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7–1 costimulatory molecule to inhibit T cell responses. Immunity. 2007;27(1):111–22: 111–122. 10.1016/j.immuni.2007.05.016
    1. Xu D, Fu HH, Obar JJ, Park JJ, Tamada K, Yagita H, et al. A potential new pathway for PD-L1 costimulation of the CD8-T cell response to Listeria monocytogenes infection. PLoS One. 2013;8: e56539 10.1371/journal.pone.0056539
    1. Park JJ, Omiya R, Matsumura Y, Sakoda Y, Kuramasu A, Augustine MM, et al. B7–H1/CD80 interaction is required for the induction and maintenance of peripheral T-cell tolerance. Blood. 2010;116: 1291–1298. 10.1182/blood-2010-01-265975
    1. Latchman YE, Liang SC, Wu Y, Chernova T, Sobel RA, Klemm M, et al. PD-L1-deficient mice show that PD-L1 on T cells, antigen-presenting cells, and host tissues negatively regulates T cells. Proc Natl Acad Sci U S A. 2004;101: 10691–10696. 10.1073/pnas.0307252101
    1. Paterson AM, Brown KE, Keir ME, Vanguri VK, Riella LV, Chandraker A, et al. The programmed death-1 ligand 1:B7–1 pathway restrains diabetogenic effector T cells in vivo. J Immunol. 2011;187: 1097–1105. 10.4049/jimmunol.1003496
    1. Kuipers H, Muskens F, Willart M, Hijdra D, van Assema FB, Coyle AJ, et al. Contribution of the PD-1 ligands/PD-1 signaling pathway to dendritic cell-mediated CD4+ T cell activation. Eur J Immunol. 2006;36: 2472–2482. 10.1002/eji.200635978
    1. Mueller SN, Matloubian M, Clemens DM, Sharpe AH, Freeman GJ, Gangappa S, et al. Viral targeting of fibroblastic reticular cells contributes to immunosuppression and persistence during chronic infection. Proc Natl Acad Sci U S A. 2007;104: 15430–15435. 10.1073/pnas.0702579104
    1. Mühlbauer M, Fleck M, Schütz C, Weiss T, Froh M, Blank C, et al. 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. 10.1016/j.jhep.2006.05.007
    1. Lee SJ, Jang BC, Lee SW, Yang YI, Suh SI, Park YM, et al. Interferon regulatory factor-1 is prerequisite to the constitutive expression and IFN-gamma-induced upregulation of B7–H1 (CD274). FEBS Lett. 2006;580: 755–762. 10.1016/j.febslet.2005.12.093
    1. Benedict CA, Loewendorf A, Garcia Z, Blazar BR, Janssen EM. Dendritic cell programming by cytomegalovirus stunts naive T cell responses via the PD-L1/PD-1 pathway. J Immunol. 2008;180: 4836–4847. 10.4049/jimmunol.180.7.4836
    1. Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10: 29–37. 10.1038/ni.1679
    1. Probst HC, Lagnel J, Kollias G, van den Broek M. Inducible transgenic mice reveal resting dendritic cells as potent inducers of CD8+ T cell tolerance. Immunity. 2003;18: 713–720. 10.1016/S1074-7613(03)00120-1
    1. Zou W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer. 2005;5: 263–274. 10.1038/nrc1586
    1. Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med. 2010;207: 2187–2194. 10.1084/jem.20100643
    1. Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A. 2010;107: 4275–4280. 10.1073/pnas.0915174107
    1. Berrien-Elliott MM, Jackson SR, Meyer JM, Rouskey CJ, Nguyen TL, Yagita H, et al. Durable adoptive immunotherapy for leukemia produced by manipulation of multiple regulatory pathways of CD8+ T-cell tolerance. Cancer Research. 2013;73: 605–616. 10.1158/0008-5472.CAN-12-2179
    1. Pilon-Thomas S, Mackay A, Vohra N, J.J. M. Blockade of programmed death ligand 1 enhances the therapeutic efficacy of combination immunotherapy against melanoma. J Immunol. 2010;184: 3442–3449. 10.4049/jimmunol.0904114
    1. Currie AJ, Prosser A, McDonnell A, Cleaver AL, Robinson BW, Freeman GJ, et al. Dual control of antitumor CD8 T cells through the programmed death-1/programmed death-ligand 1 pathway and immunosuppressive CD4 T cells: regulation and counterregulation. J Immunol. 2009;183: 7898–7908. 10.4049/jimmunol.0901060
    1. Sierro SR, Donda A, Perret R, Guillaume P, Yagita H, Levy F, et al. Combination of lentivector immunization and low-dose chemotherapy or PD-1/PD-L1 blocking primes self-reactive T cells and induces anti-tumor immunity. Eur J Immunol. 2011;41: 221–2228. 10.1002/eji.201041235
    1. Peng W, Liu C, Xu C, Lou Y, Chen J, Yang Y, et al. PD-1 blockade enhances T-cell migration to tumors by elevating IFN-γ inducible chemokines. Cancer Research. 2012;72: 5209–5218. 10.1158/0008-5472.CAN-12-1187
    1. Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, et al. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. 2014;124: 687–695. 10.1172/JCI67313
    1. Zhou Q, Munger ME, Highfill SL, Tolar J, Weigel BJ, Riddle M, et al. Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia. Blood. 2010;116: 2484–2493. 10.1182/blood-2010-03-275446
    1. John LB, Devaud C, Duong CP, Yong CS, Beavis PA, Haynes NM, et al. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin Cancer Res. 2013;19(20):. 5636–5646. 10.1158/1078-0432.CCR-13-0458
    1. Reynoso ED, Elpek KG, Francisco L, Bronson R, Bellemare-Pelletier A, Sharpe AH, et al. Intestinal tolerance is converted to autoimmune enteritis upon PD-1 ligand blockade. J Immunol. 2009;182: 2102–2112. 10.4049/jimmunol.0802769
    1. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with Ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363: 711–723. 10.1056/NEJMoa1003466

Source: PubMed

3
Se inscrever