Releasing the brakes of tumor immunity with anti-PD-L1 and pushing its accelerator with L19-IL2 cures poorly immunogenic tumors when combined with radiotherapy

Veronica Olivo Pimentel, Damiënne Marcus, Alexander Ma van der Wiel, Natasja G Lieuwes, Rianne Biemans, Relinde Iy Lieverse, Dario Neri, Jan Theys, Ala Yaromina, Ludwig J Dubois, Philippe Lambin, Veronica Olivo Pimentel, Damiënne Marcus, Alexander Ma van der Wiel, Natasja G Lieuwes, Rianne Biemans, Relinde Iy Lieverse, Dario Neri, Jan Theys, Ala Yaromina, Ludwig J Dubois, Philippe Lambin

Abstract

Background: Poorly immunogenic tumors are hardly responsive to immunotherapies such as immune checkpoint blockade (ICB) and are, therefore, a therapeutic challenge. Combination with other immunotherapies and/or immunogenic therapies, such as radiotherapy (RT), could make these tumors more immune responsive. We have previously shown that the immunocytokine L19-IL2 combined with single-dose RT resulted in 75% tumor remission and a 20% curative abscopal effect in the T cell-inflamed C51 colon carcinoma model. This treatment schedule was associated with the upregulation of inhibitory immune checkpoint (IC) molecules on tumor-infiltrating T cells, leading to only tumor growth delay in the poorly immunogenic Lewis lung carcinoma (LLC) model.

Methods: We aimed to trigger curative therapeutic responses in three tumor models (LLC, C51 and CT26) by "pushing the accelerator" of tumor immunity with L19-IL2 and/or "releasing the brakes" with ICB, such as antibodies directed against cytotoxic T lymphocyte associated protein 4 (CTLA-4), programmed death 1 (PD-1) or its ligand (PD-L1), combined with single-dose RT (10 Gy or 5 Gy). Primary tumor endpoint was defined as time to reach four times the size of tumor volume at start of treatment (4T×SV). Multivariate analysis of 4T×SV was performed using the Cox proportional hazards model comparing each treatment group with controls. Causal involvement of T and natural killer (NK) cells in the anti-tumor effect was assessed by in vivo depletion of T, NK or both cell populations. Immune profiling was performed using flow cytometry on single cell suspensions from spleens, bone marrow, tumors and blood.

Results: Combining RT, anti-PD-L1 and L19-IL2 cured 38% of LLC tumors, which was both CD8+ T and NK cell dependent. LLC tumors were resistant to RT +anti-PD-L1 likely explained by the upregulation of other IC molecules and increased T regulatory cell tumor infiltration. RT+L19-IL2 outperformed RT+ICB in C51 tumors; effects were comparable in CT26 tumors. Triple combinations were not superior to RT+L19-IL2 in both these models.

Conclusions: This study demonstrated that combinatorial strategies rationally designed on biological effects can turn immunotherapy-resistant tumors into immunologically responsive tumors. This hypothesis is currently being tested in the international multicentric randomized phase 2 trial: ImmunoSABR (NCT03705403).

Keywords: immunotherapy; radiotherapy; tumor microenvironment.

Conflict of interest statement

Competing interests: PL reports, within the submitted work, an in kind donation of Philogen (L19-IL2) and a status of PI of the trial ImmunoSABR. DN reports, within the submitted work, his role as co-founder and president of the Scientific Advisory Board of Philogen. PL reports outside the submitted work grants/sponsored research agreements from Varian medical, Oncoradiomics, ptTheragnostic/DNAmito and Health Innovation Ventures. He received an advisor/presenter fee and/or reimbursement of travel costs/external grant writing fee and/or in kind manpower contribution from Oncoradiomics, BHV, Merck, Varian, Elekta, ptTheragnostic and Convert pharmaceuticals. PL has shares in the company Oncoradiomics SA, Convert pharmaceuticals SA and The Medical Cloud Company SPRL and is co-inventor of two issued patents with royalties on radiomics (PCT/NL2014/050248 and PCT/NL2014/050728) licensed to Oncoradiomics and one issued patent on mtDNA (PCT/EP2014/059089) licensed to ptTheragnostic/DNAmito, three non-patented invention (software) licensed to ptTheragnostic/DNAmito, Oncoradiomics and Health Innovation Ventures and three non-issued, non-licensed patents on Deep Learning-Radiomics and Lymphocytes Sparing Radiotherapy. LJD is co-inventor of a non-issued, non-licensed patent on Lymphocyte Sparing Radiotherapy.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY. Published by BMJ.

Figures

Figure 1
Figure 1
Therapeutic effect of RT combined with L19–IL2 and/or ICB treatment. (A) Treatment schedule. Each mouse was injected with tumor cells on the right flank on day 8 (C51 and CT26 models) or day 5 (LLC model). Blood was withdrawn 3 days before and 6 days after treatment start for flow cytometric analysis. Fraction of tumors not reaching four times start tumor volume in LLC (n=7–8 mice per treatment arm) (B), CT26 (n=7–9) (C) and C51 (n=7–9) (D) tumor models. C51 and CT26 tumors were irradiated with a single dose of 5 Gy and LLC tumors with an isoeffective single dose of 10 Gy. Treatments in the LLC and the CT26 models were performed in one single experiment, while in the C51 model two independent experiments were performed (see online supplemental figure 4). Combinations with anti-PD-1 in the C51 (n=6–8) and CT26 (n=5–6) were tested in an independent experiment. Differences between treatment groups for each tumor model are summarized in table 1. Treatments: blue: RT, red: RT + L19-IL2, green: RT + ICB, black: RT + L19-IL2 + ICB. ICB, immune checkpoint blockade; IgG, immunoglobulin G; LLC, Lewis lung carcinoma; PBS, phosphate-buffered saline; RT, radiotherapy.
Figure 2
Figure 2
RT+L19–IL2+anti-PD-L1 (trimodal) therapy effect is associated with high infiltration of NK and antigen-experienced CD8+ CD44+ T cells in the LLC model. (A) Treatment schedule for the evaluation of immunological parameters in the LLC model (n=5 mice per treatment arm), experiment was performed once. Stainings, flow cytometry data acquisition, and analysis of the samples were done in independent duplicates. (B) Representative flow cytometry dot plots of tumor-infiltrating CD8+ CD44+ (gated on CD3+ T cells) T cells 6 days after the start of treatment and quantification. (C) Representative flow cytometry dot plots of tumor-infiltrating T (CD3+ NK1.1-), NKT (CD3+ NK1.1+) and NK (CD3- NK1.1+) cells and quantifications of NKT and NK cell percentages. IgG, immunoglobulin G; LLC, Lewis lung carcinoma; PBS, phosphate-buffered saline; RT, radiotherapy.
Figure 3
Figure 3
Changes in pretreatment immunological blood parameters (expression of PD-1 and PD-L1 on CD4+ and CD8+ T cells) during triple therapy (RT+L19–IL2+anti-PD-L1). Blood samples were collected 3 days before treatment (day 3) and on day 6 after treatment start (n=8–9 mice per tumor model, see treatment scheme in figure 1A). Data are reported as the day 6/day -3 ratio of the MFI of PD-L1 on CD4+ (A) and CD8+ (B) T cells and of PD-1 on CD4+ (C) and CD8+ T cells (D). Ratio >1 indicates an increase and ratio <1 indicates a decrease in the parameter during therapy. (E) and (F) Representative flow cytometry histograms of (C) and (B), respectively. MFI, median fluorescence intensity.
Figure 4
Figure 4
CD8+ T and NK cells play a key role in the anti-tumor effect of triple therapy. (A) Treatment schedule of the depletion study. (B) Flow cytometric analysis of CD8+ T cell and NK cell presence in the blood to confirm their depletion after 3 doses of depleting antibodies. (C) Fraction of tumors not reaching four times start tumor volume in LLC tumor-bearing mice (n=9 mice per treatment arm) treated with 10 Gy+L19–IL2+anti-PD-L1 depleted of CD8+ T (red), NK (green) or both cell types (blue) or IgG-treated (black) according to schedule in (A). This depletion study was performed once. IgG, immunoglobulin G; LLC, Lewis lung carcinoma.
Figure 5
Figure 5
PD-L1 blockade upregulates the expression of other co-inhibitory receptors on tumor-infiltrating T cells. Representative flow cytometry histograms and quantifications of PD-1+ (A), Tim-3+ (B) and CD39+ (C) tumor-infiltrating CD4+ and CD8+ T cells from LLC tumors at day 6 after the start of treatment depicted in figure 2A. (D) Representative flow cytometry histograms of CD45− PD-L1+ LLC tumor cells and CD3− NK1.1− CD11b+ LLC tumor-infiltrating cells and quantifications. Experiment was performed once. Stainings, flow cytometry data acquisition and analysis of the samples were done in independent duplicates. LLC, Lewis lung carcinoma; RT, radiotherapy.
Figure 6
Figure 6
RT+anti-PD-L1 increases tumor-infiltrating Tregs. (A) Representative flow cytometry dot plots of tumor-infiltrating Tregs and quantification of CD4+ CD25+ FoxP3+ Tregs, CD4+ CD25- FoxP3+ Tregs and CD8+ CD44+ T cell to CD25+ Treg ratio in LLC tumors at day six after the start of treatment depicted in figure 2A. (B) Representative flow cytometry histograms of PD-1+ and Tim-3+ tumor-infiltrating CD25+ Tregs and quantifications. Experiment was performed once. Stainings, flow cytometry data acquisition and analysis of the samples were done in independent duplicates. LLC, Lewis lung carcinoma; MFI, median fluorescence intensity; RT, radiotherapy; Tregs, regulatory T cells.
Figure 7
Figure 7
Anti-PD-L1-based trimodal therapy causes an accumulation of memory T cell subsets in lymphoid organs of cured LLC tumor-bearing mice. Representative flow cytometry dot plots and quantification of CD8+ CD44+ CD127+ memory T cells (A) and CD44+ CD62L+ central memory CD8+ T cells (B) from spleen and bone marrow of LLC tumor-bearing mice treated with 10 Gy+L19–IL2+anti-PD-L1 harvested 43±2.1 days after tumor cure (12 days after tumor rechallenge) or endpoint (T4×SV) had been reached. Cells were obtained from one experiment performed in figure 1B. Stainings, flow cytometry data acquisition, and analysis of the samples were done in independent duplicates. (C) Representative flow cytometry dot plots and quantifications of CD8+ CD44+ T cells expressing IFNγ and granzyme B from splenocytes of cured or non-cured LLC tumor-bearing mice treated with 10 Gy+L19–IL2+anti-PD-L1, or from naïve mice after co-culture with irradiated LLC target cells or GL261 as non-specific target cells. Cells were obtained from the same experiment as in (A) and (B). Co-culture assay, stainings, flow cytometry data acquisition, and analysis of the samples were done in independent duplicates. LLC, Lewis lung carcinoma.

References

    1. Haslam A, Prasad V. Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw Open 2019;2:e192535–e. 10.1001/jamanetworkopen.2019.2535
    1. Sharma P, Hu-Lieskovan S, Wargo JA, et al. . Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 2017;168:707–23. 10.1016/j.cell.2017.01.017
    1. Herbst RS, Soria J-C, Kowanetz M, et al. . Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014;515:563–7. 10.1038/nature14011
    1. Peggs KS, Quezada SA, Allison JP. Cell intrinsic mechanisms of T-cell inhibition and application to cancer therapy. Immunol Rev 2008;224:141–65. 10.1111/j.1600-065X.2008.00649.x
    1. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov 2018;8:1069–86. 10.1158/-18-0367
    1. Sanmamed MF, Chen L. A paradigm shift in cancer immunotherapy: from enhancement to normalization. Cell 2019;176:677. 10.1016/j.cell.2019.01.008
    1. Bentzen SM. Preventing or reducing late side effects of radiation therapy: radiobiology meets molecular pathology. Nat Rev Cancer 2006;6:702–13. 10.1038/nrc1950
    1. Barker HE, Paget JTE, Khan AA, et al. . The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer 2015;15:409–25. 10.1038/nrc3958
    1. Rosental B, Appel MY, Yossef R, et al. . The effect of chemotherapy/radiotherapy on cancerous pattern recognition by NK cells. Curr Med Chem 2012;19:1780–91. 10.2174/092986712800099730
    1. Eckert F, Jelas I, Oehme M, et al. . Tumor-targeted IL-12 combined with local irradiation leads to systemic tumor control via abscopal effects in vivo. Oncoimmunology 2017;6:e1323161. 10.1080/2162402X.2017.1323161
    1. Eckert F, Schmitt J, Zips D, et al. . Enhanced binding of necrosis-targeting immunocytokine NHS-IL12 after local tumour irradiation in murine xenograft models. Cancer Immunol Immunother 2016;65:1003–13. 10.1007/s00262-016-1863-0
    1. Fallon J, Tighe R, Kradjian G, et al. . The immunocytokine NHS-IL12 as a potential cancer therapeutic. Oncotarget 2014;5:1869–84. 10.18632/oncotarget.1853
    1. Golden EB, Chhabra A, Chachoua A, et al. . Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial. Lancet Oncol 2015;16:795–803. 10.1016/S1470-2045(15)00054-6
    1. Morris ZS, Guy EI, Francis DM, et al. . In situ tumor vaccination by combining local radiation and tumor-specific antibody or immunocytokine treatments. Cancer Res 2016;76:3929–41. 10.1158/0008-5472.CAN-15-2644
    1. Seung SK, Curti BD, Crittenden M, et al. . Phase 1 study of stereotactic body radiotherapy and interleukin-2--tumor and immunological responses. Sci Transl Med 2012;4:137ra74. 10.1126/scitranslmed.3003649
    1. van den Heuvel MM, Verheij M, Boshuizen R, et al. . NHS-IL2 combined with radiotherapy: preclinical rationale and phase Ib trial results in metastatic non-small cell lung cancer following first-line chemotherapy. J Transl Med 2015;13:32. 10.1186/s12967-015-0397-0
    1. Neri D. Antibody-Cytokine fusions: versatile products for the modulation of anticancer immunity. Cancer Immunol Res 2019;7:348–54. 10.1158/2326-6066.CIR-18-0622
    1. Neri D, Sondel PM. Immunocytokines for cancer treatment: past, present and future. Curr Opin Immunol 2016;40:96–102. 10.1016/j.coi.2016.03.006
    1. Rekers NH, Olivo Pimentel V, Yaromina A, et al. . The immunocytokine L19-IL2: an interplay between radiotherapy and long-lasting systemic anti-tumour immune responses. Oncoimmunology 2018;7:e1414119. 10.1080/2162402X.2017.1414119
    1. Zegers CML, Rekers NH, Quaden DHF, et al. . Radiotherapy combined with the immunocytokine L19-IL2 provides long-lasting antitumor effects. Clin Cancer Res 2015;21:1151–60. 10.1158/1078-0432.CCR-14-2676
    1. Rekers NH, Zegers CML, Yaromina A, et al. . Combination of radiotherapy with the immunocytokine L19-IL2: additive effect in a NK cell dependent tumour model. Radiother Oncol 2015;116:438–42. 10.1016/j.radonc.2015.06.019
    1. Lechner MG, Karimi SS, Barry-Holson K, et al. . Immunogenicity of murine solid tumor models as a defining feature of in vivo behavior and response to immunotherapy. J Immunother 2013;36:477–89. 10.1097/01.cji.0000436722.46675.4a
    1. Mosely SIS, Prime JE, Sainson RCA, et al. . Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery. Cancer Immunol Res 2017;5:29–41. 10.1158/2326-6066.CIR-16-0114
    1. Van Limbergen EJ, De Ruysscher DK, Olivo Pimentel V, et al. . Combining radiotherapy with immunotherapy: the past, the present and the future. Br J Radiol 2017;90:20170157. 10.1259/bjr.20170157
    1. Amend SR, Valkenburg KC, Pienta KJ. Murine hind limb long bone dissection and bone marrow isolation. J Vis Exp 2016;110:e53936. 10.3791/53936
    1. Allard B, Longhi MS, Robson SC, et al. . The ectonucleotidases CD39 and CD73: novel checkpoint inhibitor targets. Immunol Rev 2017;276:121–44. 10.1111/imr.12528
    1. De Luca R, Neri D. Potentiation of PD-L1 blockade with a potency-matched dual cytokine-antibody fusion protein leads to cancer eradication in BALB/c-derived tumors but not in other mouse strains. Cancer Immunol Immunother 2018;67:1381–91. 10.1007/s00262-018-2194-0
    1. Puca E, Probst P, Stringhini M, et al. . The antibody-based delivery of interleukin-12 to solid tumors boosts NK and CD8+ T cell activity and synergizes with immune checkpoint inhibitors. Int J Cancer 2020;146:2518-2530. 10.1002/ijc.32603
    1. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov 2019;18:197–218. 10.1038/s41573-018-0007-y
    1. Sevenich L. Turning “cold” into “hot” tumors—opportunities and challenges for radio-immunotherapy against primary and metastatic brain cancers. Front Oncol 2019;9. 10.3389/fonc.2019.00163
    1. Maleki Vareki S. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J Immunother Cancer 2018;6:157. 10.1186/s40425-018-0479-7
    1. Cai DL, Jin L-P. Immune cell population in ovarian tumor microenvironment. J Cancer 2017;8:2915–23. 10.7150/jca.20314
    1. Heong V, Ngoi N, Tan DSP. Update on immune checkpoint inhibitors in gynecological cancers. J Gynecol Oncol 2017;28:0. 10.3802/jgo.2017.28.e20
    1. Johnson DB, Rioth MJ, Horn L. Immune checkpoint inhibitors in NSCLC. Curr Treat Options Oncol 2014;15:658–69. 10.1007/s11864-014-0305-5
    1. Rausch MP HK. Immune Checkpoint Inhibitors in the Treatment of Melanoma: From Basic Science to Clinical Application. : Ward WH, Farma JM, . Cutaneous melanoma: etiology and therapy [Internet]. Brisbane (AU): Codon Publications, 2017.
    1. Azuma T, Yao S, Zhu G, et al. . B7-H1 is a ubiquitous antiapoptotic receptor on cancer cells. Blood 2008;111:3635–43. 10.1182/blood-2007-11-123141
    1. Diskin B, Adam S, Cassini MF, et al. . Pd-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer. Nat Immunol 2020;21:442–54. 10.1038/s41590-020-0620-x
    1. Hartley GP, Chow L, Ammons DT, et al. . Programmed cell death ligand 1 (PD-L1) signaling regulates macrophage proliferation and activation. Cancer Immunol Res 2018;6:1260–73. 10.1158/2326-6066.CIR-17-0537
    1. Bar N, Costa F, Das R, et al. . Differential effects of PD-L1 versus PD-1 blockade on myeloid inflammation in human cancer. JCI Insight 2020;5. 10.1172/jci.insight.129353. [Epub ahead of print: 18 Jun 2020].
    1. De Sousa Linhares A, Battin C, Jutz S, et al. . Therapeutic PD-L1 antibodies are more effective than PD-1 antibodies in blocking PD-1/PD-L1 signaling. Sci Rep 2019;9:11472. 10.1038/s41598-019-47910-1
    1. Butte MJ, Keir ME, Phamduy TB, et al. . Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity 2007;27:111–22. 10.1016/j.immuni.2007.05.016
    1. Baaten BJG, Tinoco R, Chen AT, et al. . Regulation of antigen-experienced T cells: lessons from the Quintessential memory marker CD44. Front Immunol 2012;3:23. 10.3389/fimmu.2012.00023
    1. Ali HR, Provenzano E, Dawson S-J, et al. . Association between CD8+ T-cell infiltration and breast cancer survival in 12,439 patients. Ann Oncol 2014;25:1536–43. 10.1093/annonc/mdu191
    1. Ovarian Tumor Tissue Analysis (OTTA) Consortium, Goode EL, Block MS, et al. . Dose-response association of CD8+ tumor-infiltrating lymphocytes and survival time in high-grade serous ovarian cancer. JAMA Oncol 2017;3:e173290. 10.1001/jamaoncol.2017.3290
    1. Gooden MJM, de Bock GH, Leffers N, et al. . The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis. Br J Cancer 2011;105:93–103. 10.1038/bjc.2011.189
    1. Liu S, Lachapelle J, Leung S, et al. . Cd8+ lymphocyte infiltration is an independent favorable prognostic indicator in basal-like breast cancer. Breast Cancer Res 2012;14:R48. 10.1186/bcr3148
    1. Ménétrier-Caux C, Ray-Coquard I, Blay J-Y, et al. . Lymphopenia in cancer patients and its effects on response to immunotherapy: an opportunity for combination with cytokines? J Immunother Cancer 2019;7:85. 10.1186/s40425-019-0549-5
    1. López-Soto A, Gonzalez S, Smyth MJ, et al. . Control of metastasis by NK cells. Cancer Cell 2017;32:135–54. 10.1016/j.ccell.2017.06.009
    1. Terabe M, Berzofsky JA. Tissue-specific roles of NKT cells in tumor immunity. Front Immunol 2018;9:9. 10.3389/fimmu.2018.01838
    1. Woo S-R, Corrales L, Gajewski TF. Innate immune recognition of cancer. Annu Rev Immunol 2015;33:445–74. 10.1146/annurev-immunol-032414-112043
    1. Mortara L, Balza E, Bruno A, et al. . Anti-cancer therapies employing IL-2 cytokine tumor targeting: contribution of innate, adaptive and immunosuppressive cells in the anti-tumor efficacy. Front Immunol 2018;9:2905. 10.3389/fimmu.2018.02905
    1. Barry KC, Hsu J, Broz ML, et al. . A natural killer-dendritic cell axis defines checkpoint therapy-responsive tumor microenvironments. Nat Med 2018;24:1178–91. 10.1038/s41591-018-0085-8
    1. Hsu J, Hodgins JJ, Marathe M, et al. . Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J Clin Invest 2018;128:4654–68. 10.1172/JCI99317
    1. Dong W, Wu X, Ma S, et al. . The mechanism of Anti–PD-L1 antibody efficacy against PD-L1–Negative tumors identifies NK cells expressing PD-L1 as a cytolytic effector. Cancer Discov 2019;9:1422–37. 10.1158/-18-1259
    1. Mariotti FR, Petrini S, Ingegnere T, et al. . Pd-1 in human NK cells: evidence of cytoplasmic mRNA and protein expression. Oncoimmunology 2019;8:1557030. 10.1080/2162402X.2018.1557030
    1. Pesce S, Greppi M, Grossi F, et al. . PD/1-PD-Ls checkpoint: insight on the potential role of NK cells. Front Immunol 2019;10:1242. 10.3389/fimmu.2019.01242
    1. Dahan R, Sega E, Engelhardt J, et al. . FcγRs modulate the anti-tumor activity of antibodies targeting the PD-1/PD-L1 axis. Cancer Cell 2015;28:285–95. 10.1016/j.ccell.2015.08.004
    1. Boyerinas B, Jochems C, Fantini M, et al. . Antibody-Dependent cellular cytotoxicity activity of a novel anti-PD-L1 antibody Avelumab (MSB0010718C) on human tumor cells. Cancer Immunol Res 2015;3:1148–57. 10.1158/2326-6066.CIR-15-0059
    1. Juliá EP, Amante A, Pampena MB, et al. . Avelumab, an IgG1 anti-PD-L1 immune checkpoint inhibitor, triggers NK cell-mediated cytotoxicity and cytokine production against triple negative breast cancer cells. Front Immunol 2018;9:2140. 10.3389/fimmu.2018.02140
    1. Koyama S, Akbay EA, Li YY, et al. . Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun 2016;7:10501. 10.1038/ncomms10501
    1. Syn NL, Teng MWL, Mok TSK, et al. . De-Novo and acquired resistance to immune checkpoint targeting. Lancet Oncol 2017;18:e731–41. 10.1016/S1470-2045(17)30607-1
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. . Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med 2015;373:23–34. 10.1056/NEJMoa1504030
    1. Motzer RJ, Tannir NM, McDermott DF, et al. . Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med 2018;378:1277–90. 10.1056/NEJMoa1712126
    1. Davis AA, Patel VG. The role of PD-L1 expression as a predictive biomarker: an analysis of all US food and drug administration (FDA) approvals of immune checkpoint inhibitors. J Immunother Cancer 2019;7:278. 10.1186/s40425-019-0768-9
    1. Lin H, Wei S, Hurt EM, et al. . Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade-mediated tumor regression. J Clin Invest 2018;128:805–15. 10.1172/JCI96113
    1. Tang H, Liang Y, Anders RA, et al. . Pd-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression. J Clin Invest 2018;128:580–8. 10.1172/JCI96061
    1. Boyman O, Sprent J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol 2012;12:180–90. 10.1038/nri3156
    1. Malek TR, Castro I. Interleukin-2 receptor signaling: at the interface between tolerance and immunity. Immunity 2010;33:153–65. 10.1016/j.immuni.2010.08.004
    1. Whiteside TL. Induced regulatory T cells in inhibitory microenvironments created by cancer. Expert Opin Biol Ther 2014;14:1411–25. 10.1517/14712598.2014.927432
    1. Jacquelot N, Roberti MP, Enot DP, et al. . Predictors of responses to immune checkpoint blockade in advanced melanoma. Nat Commun 2017;8:592. 10.1038/s41467-017-00608-2
    1. Twyman-Saint Victor C, Rech AJ, Maity A, et al. . Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015;520:373–7. 10.1038/nature14292
    1. Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors in cancer therapy: a focus on T-regulatory cells. Immunol Cell Biol 2018;96:21–33. 10.1111/imcb.1003
    1. Oweida A, Hararah MK, Phan A, et al. . Resistance to radiotherapy and PD-L1 blockade is mediated by Tim-3 upregulation and regulatory T-cell infiltration. Clin Cancer Res 2018;24:5368–80. 10.1158/1078-0432.CCR-18-1038
    1. Liu Z, McMichael EL, Shayan G, et al. . Novel effector phenotype of Tim-3+ regulatory T cells leads to enhanced suppressive function in head and neck cancer patients. Clin Cancer Res 2018;24:4529–38. 10.1158/1078-0432.CCR-17-1350
    1. Selby MJ, Engelhardt JJ, Quigley M, et al. . Anti-Ctla-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res 2013;1:32–42. 10.1158/2326-6066.CIR-13-0013
    1. Wing K, Onishi Y, Prieto-Martin P, et al. . Ctla-4 control over Foxp3+ regulatory T cell function. Science 2008;322:271–5. 10.1126/science.1160062
    1. Schwager K, Hemmerle T, Aebischer D, et al. . The immunocytokine L19-IL2 eradicates cancer when used in combination with CTLA-4 blockade or with L19-TNF. J Invest Dermatol 2013;133:751–8. 10.1038/jid.2012.376
    1. Hannani D, Vétizou M, Enot D, et al. . Anticancer immunotherapy by CTLA-4 blockade: obligatory contribution of IL-2 receptors and negative prognostic impact of soluble CD25. Cell Res 2015;25:208–24. 10.1038/cr.2015.3
    1. Maker AV, Phan GQ, Attia P, et al. . Tumor regression and autoimmunity in patients treated with cytotoxic T lymphocyte-associated antigen 4 blockade and interleukin 2: a phase I/II study. Ann Surg Oncol 2005;12:1005–16. 10.1245/ASO.2005.03.536

Source: PubMed

3
Suscribir