PD-L1 Distribution and Perspective for Cancer Immunotherapy-Blockade, Knockdown, or Inhibition

Yilun Wu, Weiyu Chen, Zhi Ping Xu, Wenyi Gu, Yilun Wu, Weiyu Chen, Zhi Ping Xu, Wenyi Gu

Abstract

Cancer immunotherapy involves blocking the interactions between the PD-1/PD-L1 immune checkpoints with antibodies. This has shown unprecedented positive outcomes in clinics. Particularly, the PD-L1 antibody therapy has shown the efficiency in blocking membrane PD-L1 and efficacy in treating some advanced carcinoma. However, this therapy has limited effects on many solid tumors, suspecting to be relevant to PD-L1 located in other cellular compartments, where they play additional roles and are associated with poor prognosis. In this review, we highlight the advances of 3 current strategies on PD-1/PD-L1 based immunotherapy, summarize cellular distribution of PD-L1, and review the versatile functions of intracellular PD-L1. The intracellular distribution and function of PD-L1 may indicate why not all antibody blockade is able to fully stop PD-L1 biological functions and effectively inhibit tumor growth. In this regard, gene silencing may have advantages over antibody blockade on suppression of PD-L1 sources and functions. Apart from cancer cells, PD-L1 silencing on host immune cells such as APC and DC can also enhance T cell immunity, leading to tumor clearance. Moreover, the molecular regulation of PD-L1 expression in cells is being elucidated, which helps identify potential therapeutic molecules to target PD-L1 production and improve clinical outcomes. Based on our understandings of PD-L1 distribution, regulation, and function, we prospect that the more effective PD-L1-based cancer immunotherapy will be combination therapies.

Keywords: PD-1/PD-L1 immune checkpoint; PD-L1 regulation; cancer immunotherapy; cellular PD-L1 distribution; combination therapy; gene silencing; signaling pathway inhibitor.

Figures

Figure 1
Figure 1
Immunotherapy based on PD-1/PD-L1 interaction. (A) The interaction of PD-1/PD-L1 causes tumor immune tolerance. The PD-1/PD-L1 interaction stimulates the downstream signals to suppress T cell activation, resulting in tumor cell survival. (B) Breakdown of the PD-1/PD-L1 interaction reactivates T cells and related immune responses. Without the PD-1/PD-L1 interaction, the suppression signal is removed, thus leading to T cell activation, proliferation, and cytokine generation and tumor cell elimination. KIR, killer-cell immunoglobulin-like receptor.
Figure 2
Figure 2
Illustration of different PD-L1 formats. (A) mPD-L1, located on the tumor cell membrane, is able to bind with PD-1 on T cells and response to tumor immune escape. PD-L1 antibody competitively binding to mPD-L1 breaks the tolerance, leading to tumor cell clearance. (B) cPD-L1 is located in cytoplasm, and potentiates to transfer to mPD-L1. (C) nPD-L1 is located in nuclei. Its aberrant upregulation is speculated to be associated with promoted cell chemo-resistance. (D) sPD-L1 refers to its soluble format in the serum, generated from either endogenous secretion or cleaved fraction of mPD-L1s. Both host cells (such as APC and DC) and tumor cells can be the source of sPD-L1. PD-L1 antibody therapeutic effect is limited to sPD-L1 consumption, and cannot modulate intracellular PD-L1.
Figure 3
Figure 3
Signaling pathways of PD-L1 regulation. The instinct PD-L1 expression is regulated by translational factors (HIF-1α, NF-κB, AP-1, and STATs) that binds to the gene promoter. The extracellular signals (hypoxia, cytokines, and EGF signals) will be transduced via different pathways (mainly through MAPK or PI3K/AKT) to regulate PD-L1 expression on transcriptional level. Some miRs are able to bind to 3′-UTR of PD-L1 mRNA for post transcriptional regulation.
Figure 4
Figure 4
The personalized PD-L1 combination therapy, a prospect of optimal PD-L1 immunotherapy guided by PD-L1 distribution and immune resistance of patients. Innate immune resistant cohort: (A) for cells with high constitutive mPD-L1, the regimen would mainly rely on antibody blockade while the combination of gene silencing or chemical inhibitor would benefit the treatment. (B) For cells with high intracellular PD-L1, the regimen should more rely on gene knockdown or inhibition method, supported by PD-L1 antibody. Adaptive immune resistant cohort: (C) the inducible PD-L1 is much easier to be controlled by gene silencing and chemical inhibitor combination therapy. Combined immune resistance cohort: (D) for cells with both high constitutive and inducible PD-L1, the regimen would be better to choose the combination of antibody + gene silencing or antibody + chemical inhibitor.

References

    1. Co M. Bavencio (Avelumab) Prescribing Information. (2017). Available online at:
    1. Co M. KEYTRUDA (Pembrolizumab) Prescribing Information. (2014). Available online at:
    1. Li K, Tian H. Development of small-molecule immune checkpoint inhibitors of PD-1/PD-L1 as a new therapeutic strategy for tumour immunotherapy. J Drug Target. (2018) 27:244–56. 10.1080/1061186X.2018.1440400
    1. Jiang X, Zhou J, Giobbie-Hurder A, Wargo J, Hodi FS. The activation of MAPK in melanoma cells resistant to BRAF inhibition promotes PD-L1 expression that is reversible by MEK and PI3K inhibition. Clin Cancer Res. (2013) 19:598–609. 10.1158/1078-0432.CCR-12-2731
    1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. . Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. (2015) 136:E359–E386. 10.1002/ijc.29210
    1. Stewart B, Wild CP. World Cancer Report 2014. Health (2017). Available online at:
    1. Sharma P, Wagner K, Wolchok JD, Allison JP. Novel cancer immunotherapy agents with survival benefit: recent successes and next steps. Nat Rev Cancer. (2011) 11:805. 10.1038/nrc3153
    1. Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov. (2015) 14:561. 10.1038/nrd4591
    1. Cao X. Immunology in China: the past, present and future. Nat Immunol. (2008) 9:339–42. 10.1038/ni0408-339
    1. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. (1996) 271:1734–6. 10.1126/science.271.5256.1734
    1. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, 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. (2000) 192:1027–34. 10.1084/jem.192.7.1027
    1. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al. . Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. (2002) 8:793–800. 10.1038/nm730
    1. Du S, McCall N, Park K, Guan Q, Fontina P, Ertel A, et al. . Blockade of tumor-expressed PD-1 promotes lung cancer growth. Oncoimmunology. (2018) 7:e1408747. 10.1080/2162402X.2017.1408747
    1. He R, Ding W, Viswanatha DS, Chen D, Shi M, Van Dyke D, et al. . PD-1 expression in chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) and large B-cell Richter transformation (DLBCL-RT). Am J Surg Pathol. (2018) 42:843–54. 10.1097/PAS.0000000000001077
    1. Xu J, Yao H, Wang H, Li C, Fang J-Y. Cancer cell-intrinsic PD-1 and implications in combinatorial immunotherapy. Front Immunol. (2018) 9:1774. 10.3389/fimmu.2018.01774
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. (2012) 12:252. 10.1038/nrc3239
    1. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. (2013) 39:1–10. 10.1016/j.immuni.2013.07.012
    1. Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, Sharfman WH, et al. . Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. (2014) 32:1020. 10.1200/JCO.2013.53.0105
    1. Frigola X, Inman BA, Krco CJ, Liu X, Harrington SM, Bulur PA, et al. . Soluble B7-H1: differences in production between dendritic cells and T cells. Immunol Lett. (2012) 142:78–82. 10.1016/j.imlet.2011.11.001
    1. Ukpo OC, Thorstad WL, Lewis JS, Jr. B7-H1 expression model for immune evasion in human papillomavirus-related oropharyngeal squamous cell carcinoma. Head Neck Pathol. (2013) 7:113–21. 10.1007/s12105-012-0406-z
    1. Tang Y, Fang W, Zhang Y, Hong S, Kang S, Yan Y, et al. . The association between PD-L1 and EGFR status and the prognostic value of PD-L1 in advanced non-small cell lung cancer patients treated with EGFR-TKIs. Oncotarget. (2015) 6:14209–19. 10.18632/oncotarget.3694
    1. Hua D, Sun J, Mao Y, Chen LJ, Wu YY, Zhang XG. B7-H1 expression is associated with expansion of regulatory T cells in colorectal carcinoma. World J Gastroenterol. (2012) 18:971–8. 10.3748/wjg.v18.i9.971
    1. Chowdhury S, Veyhl J, Jessa F, Polyakova O, Alenzi A, MacMillan C, et al. . Programmed death-ligand 1 overexpression is a prognostic marker for aggressive papillary thyroid cancer and its variants. Oncotarget. (2016) 7:32318–28. 10.18632/oncotarget.8698
    1. Satelli A, Batth IS, Brownlee Z, Rojas C, Meng QH, Kopetz S, et al. . Potential role of nuclear PD-L1 expression in cell-surface vimentin positive circulating tumor cells as a prognostic marker in cancer patients. Sci Rep. (2016) 6:28910. 10.1038/srep28910
    1. Ghebeh H, Lehe C, Barhoush E, Al-Romaih K, Tulbah A, Al-Alwan M, et al. . Doxorubicin downregulates cell surface B7-H1 expression and upregulates its nuclear expression in breast cancer cells: role of B7-H1 as an anti-apoptotic molecule. Breast Cancer Res. (2010) 12:R48. 10.1186/bcr2605
    1. Ruf M, Moch H, Schraml P. PD-L1 expression is regulated by hypoxia inducible factor in clear cell renal cell carcinoma. Int J Cancer. (2016) 139:396–403. 10.1002/ijc.30077
    1. Chen Y, Wang Q, Shi B, Xu P, Hu Z, Bai L, et al. . Development of a sandwich ELISA for evaluating soluble PD-L1 (CD274) in human sera of different ages as well as supernatants of PD-L1+ cell lines. Cytokine. (2011) 56:231–8. 10.1016/j.cyto.2011.06.004
    1. Qu Q-X, Xie F, Huang Q, Zhang X-G. Membranous and cytoplasmic expression of PD-L1 in ovarian cancer cells. Cell Physiol Biochem. (2017) 43:1893–906. 10.1159/000484109
    1. Zhu X, Lang J. Soluble PD-1 and PD-L1: predictive and prognostic significance in cancer. Oncotarget. (2017) 8:97671–82. 10.18632/oncotarget.18311
    1. Chen Y, Liu P, Gao F, Cheng H, Qi J, Gao GF. A dimeric structure of PD-L1: functional units or evolutionary relics? Protein Cell. (2010) 1:153–60. 10.1007/s13238-010-0022-1
    1. Chen J, Jiang CC, Jin L, Zhang XD. Regulation of PD-L1: a novel role of pro-survival signalling in cancer. Ann Oncol. (2016) 27:409–16. 10.1093/annonc/mdv615
    1. Mahoney KM, Sun H, Liao X, Hua P, Callea M, Greenfield EA, et al. . PD-L1 antibodies to its cytoplasmic domain most clearly delineate cell membranes in immunohistochemical staining of tumor cells. Cancer Immunol Res. (2015) 3:1308–15. 10.1158/2326-6066.CIR-15-0116
    1. Ott PA, Hodi FS, Robert C. CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefit in melanoma patients. Clin Cancer Res. (2013) 19:5300–9. 10.1158/1078-0432.CCR-13-0143
    1. Clark CA, Gupta HB, Curiel TJ. Tumor cell-intrinsic CD274/PD-L1: a novel metabolic balancing act with clinical potential. Autophagy. (2017) 13:987–8. 10.1080/15548627.2017.1280223
    1. He XH, Xu LH, Liu Y. Identification of a novel splice variant of human PD-L1 mRNA encoding an isoform-lacking Igv-like domain. Acta Pharmacol Sin. (2005) 26:462–8. 10.1111/j.1745-7254.2005.00086.x
    1. Apolo AB, Infante JR, Balmanoukian A, Patel MR, Wang D, Kelly K, et al. . Avelumab, an anti-programmed death-ligand 1 antibody, in patients with refractory metastatic urothelial carcinoma: results from a multicenter, phase Ib study. J Clin Oncol. (2017) 35:2117–24. 10.1200/JCO.2016.71.6795
    1. Massard C, Gordon MS, Sharma S, Rafii S, Wainberg ZA, Luke J, et al. . Safety and efficacy of durvalumab (MEDI4736), an anti-programmed cell death ligand-1 immune checkpoint inhibitor, in patients with advanced urothelial bladder cancer. J Clin Oncol. (2016) 34:3119–25. 10.1200/JCO.2016.67.9761
    1. McDermott DF, Sosman JA, Sznol M, Massard C, Gordon MS, Hamid O, et al. . Atezolizumab, an anti–programmed death-ligand 1 antibody, in metastatic renal cell carcinoma: long-term safety, clinical activity, and immune correlates from a phase Ia study. J Clin Oncol. (2016) 34:833–42. 10.1200/JCO.2015.63.7421
    1. Ning YM, Suzman D, Maher VE, Zhang L, Tang S, Ricks T, et al. . FDA approval summary: atezolizumab for the treatment of patients with progressive advanced urothelial carcinoma after platinum-containing chemotherapy. Oncologist. (2017) 22:743–9. 10.1634/theoncologist.2017-0087
    1. Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, et al. . Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. (2017) 389:255–65. 10.1016/S0140-6736(16)32517-X
    1. Ma WJ, Gilligan BM, Yuan JD, Li TH. Current status and perspectives in translational biomarker research for PD-1/PD-L1 immune checkpoint blockade therapy. J Hematol Oncol. (2016) 9:47. 10.1186/s13045-016-0277-y
    1. Gandini S, Massi D, Mandala M. PD-L1 expression in cancer patients receiving anti PD-1/PD-L1 antibodies: a systematic review and meta-analysis. Crit Rev Oncol Hemat. (2016) 100:88–98. 10.1016/j.critrevonc.2016.02.001
    1. Kato S, Goodman A, Walavalkar V, Barkauskas DA, Sharabi A, Kurzrock R. Hyperprogressors after immunotherapy: analysis of genomic alterations associated with accelerated growth rate. Clin Cancer Res. (2017) 23:4242–50. 10.1158/1078-0432.CCR-16-3133
    1. Baxi S, Yang A, Gennarelli RL, Khan N, Wang Z, Boyce L, et al. . Immune-related adverse events for anti-PD-1 and anti-PD-L1 drugs: systematic review and meta-analysis. BMJ. (2018) 360:k793. 10.1136/bmj.k793
    1. Abdel-Rahman O, Fouad M. Risk of pneumonitis in cancer patients treated with immune checkpoint inhibitors: a meta-analysis. Ther Adv Respir Dis. (2016) 10:183–93. 10.1177/1753465816636557
    1. Moslehi JJ, Salem J-E, Sosman JA, Lebrun-Vignes B, Johnson DB. Increased reporting of fatal immune checkpoint inhibitor-associated myocarditis. Lancet. (2018) 391:933. 10.1016/S0140-6736(18)30533-6
    1. Johnson DB, Balko JM, Compton ML, Chalkias S, Gorham J, Xu Y, et al. . Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med. (2016) 375:1749–55. 10.1056/NEJMoa1609214
    1. Luo X, Peng X, Hou J, Wu S, Shen J, Wang L. Folic acid-functionalized polyethylenimine superparamagnetic iron oxide nanoparticles as theranostic agents for magnetic resonance imaging and PD-L1 siRNA delivery for gastric cancer. Int J Nanomed. (2017) 12:5331. 10.2147/IJN.S137245
    1. Teo PY, Yang C, Whilding LM, Parente-Pereira AC, Maher J, George AJ, et al. . Ovarian cancer immunotherapy using PD-L1 siRNA targeted delivery from folic acid-functionalized polyethylenimine: strategies to enhance T cell killing. Adv Healthc Mater. (2015) 4:1180–9. 10.1002/adhm.201500089
    1. Li Y, Wang J, Li C, Ke X-Y. Contribution of PD-L1 to oncogenesis of lymphoma and its RNAi-based targeting therapy. Leuk Lymphoma. (2012) 53:2015–23. 10.3109/10428194.2012.673228
    1. Tang H, Liang Y, Anders RA, Taube JM, Qiu X, Mulgaonkar A, 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. Lin H, Wei S, Hurt EM, Green MD, Zhao L, Vatan L, 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. Karwacz K, Bricogne C, MacDonald D, Arce F, Bennett CL, Collins M, et al. . PD-L1 co-stimulation contributes to ligand-induced T cell receptor down-modulation on CD8+ T cells. EMBO Mol Med. (2011) 3:581–92. 10.1002/emmm.201100165
    1. Cubillos-Ruiz JR, Engle X, Scarlett UK, Martinez D, Barber A, Elgueta R, et al. . Polyethylenimine-based siRNA nanocomplexes reprogram tumor-associated dendritic cells via TLR5 to elicit therapeutic antitumor immunity. J Clin Invest. (2009) 119:2231–44. 10.1172/JCI37716
    1. Hobo W, Maas F, Adisty N, de Witte T, Schaap N, van der Voort R, et al. . siRNA silencing of PD-L1 and PD-L2 on dendritic cells augments expansion and function of minor histocompatibility antigen–specific CD8+ T cells. Blood. (2010) 116:4501–11. 10.1182/blood-2010-04-278739
    1. Hobo W, Novobrantseva TI, Fredrix H, Wong J, Milstein S, Epstein-Barash H, et al. . Improving dendritic cell vaccine immunogenicity by silencing PD-1 ligands using siRNA-lipid nanoparticles combined with antigen mRNA electroporation. Cancer Immunol Immunother. (2013) 62:285–97. 10.1007/s00262-012-1334-1
    1. Roeven MW, Hobo W, van der Voort R, Fredrix H, Norde WJ, Teijgeler K, et al. . Efficient nontoxic delivery of PD-L1 and PD-L2 siRNA into dendritic cell vaccines using the cationic lipid SAINT-18. J Immunother. (2015) 38:145–54. 10.1097/CJI.0000000000000071
    1. Van Der Waart AB, Fredrix H, Van Der Voort R, Schaap N, Hobo W, Dolstra H. siRNA silencing of PD-1 ligands on dendritic cell vaccines boosts the expansion of minor histocompatibility antigen-specific CD8(+) T cells in NOD/SCID/IL2Rg(null) mice. Cancer Immunol Immunother. (2015) 64:645–54. 10.1007/s00262-015-1668-6
    1. Wang X, Li J, Dong K, Lin F, Long M, Ouyang Y, et al. . Tumor suppressor miR-34a targets PD-L1 and functions as a potential immunotherapeutic target in acute myeloid leukemia. Cell Signal. (2015) 27:443–52. 10.1016/j.cellsig.2014.12.003
    1. Xu S, Tao Z, Hai B, Liang H, Shi Y, Wang T, et al. . miR-424 (322) reverses chemoresistance via T-cell immune response activation by blocking the PD-L1 immune checkpoint. Nat Commun. (2016) 7:11406. 10.1038/ncomms11406
    1. Zhao L, Yu H, Yi S, Peng X, Su P, Xiao Z, et al. . The tumor suppressor miR-138-5p targets PD-L1 in colorectal cancer. Oncotarget. (2016) 7:45370. 10.18632/oncotarget.9659
    1. Jia L, Xi Q, Wang H, Zhang Z, Liu H, Cheng Y, et al. . miR-142-5p regulates tumor cell PD-L1 expression and enhances anti-tumor immunity. Biochem Biophys Res Commun. (2017) 488:425–31. 10.1016/j.bbrc.2017.05.074
    1. Chen L, Gibbons DL, Goswami S, Cortez MA, Ahn YH, Byers LA, et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumor cell PD-L1 expression and intratumoral immunosuppression. Nat Commun. (2014) 5:5241 10.1038/ncomms6241
    1. Noman MZ, Janji B, Abdou A, Hasmim M, Terry S, Tan TZ, et al. . The immune checkpoint ligand PD-L1 is upregulated in EMT-activated human breast cancer cells by a mechanism involving ZEB-1 and miR-200. Oncoimmunology. (2017) 6:e1263412. 10.1080/2162402X.2016.1263412
    1. Zhu J, Chen L, Zou L, Yang P, Wu R, Mao Y, et al. . MiR-20b,−21, and−130b inhibit PTEN expression resulting in B7-H1 over-expression in advanced colorectal cancer. Hum Immunol. (2014) 75:348–53. 10.1016/j.humimm.2014.01.006
    1. Fujita Y, Yagishita S, Hagiwara K, Yoshioka Y, Kosaka N, Takeshita F, et al. . The clinical relevance of the miR-197/CKS1B/STAT3-mediated PD-L1 network in chemoresistant non-small-cell lung cancer. Mol Ther. (2015) 23:717–27. 10.1038/mt.2015.10
    1. Epple M, Ganesan K, Heumann R, Klesing J, Vikman A, Neumann S, et al. Application of calcium phosphate nanoparticles in biomedicine. J Mater Chem. (2010) 20:18–23. 10.1039/B910885H
    1. Wu Y, Gu W, Tang J, Xu ZP. Devising new lipid-coated calcium phosphate/carbonate hybrid nanoparticles for controlled release in endosomes for efficient gene delivery. J Mater Chem B. (2017) 5:7194–203. 10.1039/C7TB01635B
    1. Kong L, Wu Y, Alves CS, Shi X. Efficient delivery of therapeutic siRNA into glioblastoma cells using multifunctional dendrimer-entrapped gold nanoparticles. Nanomedicine. (2016) 12:3103–15. 10.2217/nnm-2016-0240
    1. Yu M, Niu Y, Zhang J, Zhang H, Yang Y, Taran E, et al. Size-dependent gene delivery of amine-modified silica nanoparticles. Nano Res. (2016) 9:291–305. 10.1007/s12274-015-0909-5
    1. Pantarotto D, Singh R, McCarthy D, Erhardt M, Briand JP, Prato M, et al. . Functionalized carbon nanotubes for plasmid DNA gene delivery. Angew Chem Int Ed. (2004) 43:5242–6. 10.1002/anie.200460437
    1. Li L, Gu W, Chen J, Chen W, Xu ZP. Co-delivery of siRNAs and anti-cancer drugs using layered double hydroxide nanoparticles. Biomaterials. (2014) 35:3331–9. 10.1016/j.biomaterials.2013.12.095
    1. Zou SM, Erbacher P, Remy JS, Behr JP. Systemic linear polyethylenimine (L-PEI)-mediated gene delivery in the mouse. J Gene Med. (2000) 2:128–34. 10.1002/(SICI)1521-2254(200003/04)2:2<128::AID-JGM95>;2-W
    1. Lv H, Zhang S, Wang B, Cui S, Yan J. Toxicity of cationic lipids and cationic polymers in gene delivery. J Control Release. (2006) 114:100–9. 10.1016/j.jconrel.2006.04.014
    1. Beg MS, Brenner AJ, Sachdev J, Borad M, Kang YK, Stoudemire J, et al. . Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Invest New Drugs. (2017) 35:180–8. 10.1007/s10637-016-0407-y
    1. Ebbesen M, Jensen TG, Andersen S, Pedersen FS. Ethical perspectives on RNA interference therapeutics. Int J Med Sci. (2008) 5:159–68. 10.7150/ijms.5.159
    1. Ghafouri-Fard S, Ghafouri-Fard S. siRNA and cancer immunotherapy. Immunotherapy. (2012) 4:907–17. 10.2217/imt.12.87
    1. Qin X, Liu C, Zhou Y, Wang G. Cisplatin induces programmed death-1-ligand 1(PD-L1) over-expression in hepatoma H22 cells via Erk /MAPK signaling pathway. Cell Mol Biol. (2010) 56 Suppl:OL1366–72. 10.1170/156
    1. Coelho MA, de Carné Trécesson S, Rana S, Zecchin D, Moore C, Molina-Arcas M, et al. . Oncogenic RAS signaling promotes tumor immunoresistance by stabilizing PD-L1 mRNA. Immunity. (2017) 47:1083–99. 10.1016/j.immuni.2017.11.016
    1. Shi J, Qin X, Zhao L, Wang G, Liu C. Human immunodeficiency virus type 1 Tat induces B7-H1 expression via ERK/MAPK signaling pathway. Cell Immunol. (2011) 271:280–5. 10.1016/j.cellimm.2011.07.005
    1. Lopez-Bergami P, Huang C, Goydos JS, Yip D, Bar-Eli M, Herlyn M, et al. . Rewired ERK-JNK signaling pathways in melanoma. Cancer Cell. (2007) 11:447–60. 10.1016/j.ccr.2007.03.009
    1. Green MR, Rodig S, Juszczynski P, Ouyang J, Sinha P, O'Donnell E, et al. . Constitutive AP-1 activity and EBV infection induce PD-L1 in Hodgkin lymphomas and posttransplant lymphoproliferative disorders: implications for targeted therapy. Clin Cancer Res. (2012) 18:1611–8. 10.1158/1078-0432.CCR-11-1942
    1. Crane CA, Panner A, Murray JC, Wilson SP, Xu H, Chen L, et al. . PI(3) kinase is associated with a mechanism of immunoresistance in breast and prostate cancer. Oncogene. (2008) 28:306–12. 10.1038/onc.2008.384
    1. Parsa AT, Waldron JS, Panner A, Crane CA, Parney IF, Barry JJ, et al. . Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med. (2006) 13:84. 10.1038/nm1517
    1. Lastwika KJ, Wilson W, Li QK, Norris J, Xu H, Ghazarian SR, et al. . Control of PD-L1 expression by oncogenic activation of the AKT–mTOR pathway in non–small cell lung cancer. Cancer Res. (2016) 76:227–38. 10.1158/0008-5472.CAN-14-3362
    1. Song M, Chen D, Lu B, Wang C, Zhang J, Huang L, et al. . PTEN loss increases PD-L1 protein expression and affects the correlation between PD-L1 expression and clinical parameters in colorectal cancer. PLoS ONE. (2013) 8:e65821. 10.1371/journal.pone.0065821
    1. Wölfle SJ, Strebovsky J, Bartz H, Sähr A, Arnold C, Kaiser C, et al. . PD-L1 expression on tolerogenic APCs is controlled by STAT-3. Eur J Immunol. (2011) 41:413–24. 10.1002/eji.201040979
    1. Carbotti G, Barisione G, Airoldi I, Mezzanzanica D, Bagnoli M, Ferrero S, et al. . IL-27 induces the expression of IDO and PD-L1 in human cancer cells. Oncotarget. (2015) 6:43267–80. 10.18632/oncotarget.6530
    1. Liu J, Hamrouni A, Wolowiec D, Coiteux V, Kuliczkowski K, Hetuin D, et al. . Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-γ and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood. (2007) 110:296–304. 10.1182/blood-2006-10-051482
    1. Liang M, Yang H, Fu J. Nimesulide inhibits IFN-gamma-induced programmed death-1-ligand 1 surface expression in breast cancer cells by COX-2 and PGE2 independent mechanisms. Cancer Lett. (2009) 276:47–52. 10.1016/j.canlet.2008.10.028
    1. Abiko K, Matsumura N, Hamanishi J, Horikawa N, Murakami R, Yamaguchi K, et al. . IFN-gamma from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer. Br J Cancer. (2015) 112:1501–9. 10.1038/bjc.2015.101
    1. Chen J, Feng Y, Lu L, Wang H, Dai L, Li Y, et al. . Interferon-gamma-induced PD-L1 surface expression on human oral squamous carcinoma via PKD2 signal pathway. Immunobiology. (2012) 217:385–93. 10.1016/j.imbio.2011.10.016
    1. Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, et al. . Interferon receptor signaling pathways regulating PD-L1 and PD-L2 expression. Cell Rep. (2017) 19:1189–201. 10.1016/j.celrep.2017.04.031
    1. Kondo A, Yamashita T, Tamura H, Zhao W, Tsuji T, Shimizu M, et al. . Interferon-gamma and tumor necrosis factor-alpha induce an immunoinhibitory molecule, B7-H1, via nuclear factor-kappaB activation in blasts in myelodysplastic syndromes. Blood. (2010) 116:1124–31. 10.1182/blood-2009-12-255125
    1. Gowrishankar K, Gunatilake D, Gallagher SJ, Tiffen J, Rizos H, Hersey P. Inducible but not constitutive expression of PD-L1 in human melanoma cells is dependent on activation of NF-kappa B. PLoS ONE. (2015) 10:e0123410. 10.1371/journal.pone.0123410
    1. Bellucci R, Martin A, Bommarito D, Wang K, Hansen SH, Freeman GJ, et al. . Interferon-gamma-induced activation of JAK1 and JAK2 suppresses tumor cell susceptibility to NK cells through upregulation of PD-L1 expression. Oncoimmunology. (2015) 4:e1008824. 10.1080/2162402X.2015.1008824
    1. Okita R, Maeda A, Shimizu K, Nojima Y, Saisho S, Nakata M. PD-L1 overexpression is partially regulated by EGFR/HER2 signaling and associated with poor prognosis in patients with non-small-cell lung cancer. Cancer Immunol Immunother. (2017) 66:865–76. 10.1007/s00262-017-1986-y
    1. Zhang N, Zeng Y, Du W, Zhu J, Shen D, Liu Z, et al. . The EGFR pathway is involved in the regulation of PD-L1 expression via the IL-6/JAK/STAT3 signaling pathway in EGFR-mutated non-small cell lung cancer. Int J Oncol. (2016) 49:1360–8. 10.3892/ijo.2016.3632
    1. Zhang W, Pang Q, Yan C, Wang Q, Yang J, Yu S, et al. . Induction of PD-L1 expression by epidermal growth factor receptor-mediated signaling in esophageal squamous cell carcinoma. Onco Targets Ther. (2017) 10:763–71. 10.2147/OTT.S118982
    1. Akbay EA, Koyama S, Carretero J, Altabef A, Tchaicha JH, Christensen CL, et al. . Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Disc. (2013) 3:1355–63. 10.1158/-13-0310
    1. Barsoum IB, Smallwood CA, Siemens DR, Graham CH. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res. (2014) 74:665–74. 10.1158/0008-5472.CAN-13-0992
    1. Noman MZ, Chouaib S. Targeting hypoxia at the forefront of anticancer immune responses. Oncoimmunology. (2014) 3:e954463. 10.4161/21624011.2014.954463
    1. Koh J, Jang JY, Keam B, Kim S, Kim MY, Go H, et al. . EML4-ALK enhances programmed cell death-ligand 1 expression in pulmonary adenocarcinoma via hypoxia-inducible factor (HIF)-1alpha and STAT3. Oncoimmunology. (2016) 5:e1108514. 10.1080/2162402X.2015.1108514
    1. Chen TC, Wu CT, Wang CP, Hsu WL, Yang TL, Lou PJ, et al. . Associations among pretreatment tumor necrosis and the expression of HIF-1alpha and PD-L1 in advanced oral squamous cell carcinoma and the prognostic impact thereof. Oral Oncol. (2015) 51:1004–10. 10.1016/j.oraloncology.2015.08.011
    1. Luke JJ, Hodi FS. Vemurafenib and BRAF inhibition: a new class of treatment for metastatic melanoma. Clin Cancer Res. (2012) 18:9–14. 10.1158/1078-0432.CCR-11-2197
    1. Zou H, Li L, Carcedo IG, Xu ZP, Monteiro M, Gu W. Synergistic inhibition of colon cancer cell growth with nanoemulsion-loaded paclitaxel and PI3K/mTOR dual inhibitor BEZ235 through apoptosis. Int J Nanomed. (2016) 11:1947. 10.2147/IJN.S100744
    1. Goswami S, Basu S, Sharma P. A potential biomarker for anti-PD-1 immunotherapy. Nat Med. (2018) 24:123. 10.1038/nm.4489
    1. Yin N, Ma W, Pei J, Ouyang Q, Tang C, Lai L. Synergistic and antagonistic drug combinations depend on network topology. PLoS ONE. (2014) 9:e93960. 10.1371/journal.pone.0093960
    1. Miller WH, Kim TM, Lee CB, Flaherty KT, Reddy S, Jamal R, et al. Atezolizumab (A)+ cobimetinib (C) in metastatic melanoma (mel): updated safety and clinical activity. J Clin Oncol. (2017) 35:3057 10.1200/JCO.2017.35.15_suppl.3057
    1. Bendell JC, Kim TW, Goh BC, Wallin J, Oh D-Y, Han S-W, et al. Clinical activity and safety of cobimetinib (cobi) and atezolizumab in colorectal cancer (CRC). J Clin Oncol. (2016) 34:3502 10.1200/JCO.2016.34.15_suppl.3502
    1. Liao Y, Chen L, Feng Y, Shen J, Gao Y, Cote G, et al. . Targeting programmed cell death ligand 1 by CRISPR/Cas9 in osteosarcoma cells. Oncotarget. (2017) 8:30276–87. 10.18632/oncotarget.16326
    1. Hughes PE, Caenepeel S, Wu LC. Targeted therapy and checkpoint immunotherapy combinations for the treatment of cancer. Trends Immunol. (2016) 37:462–76. 10.1016/j.it.2016.04.010
    1. Peng J, Hamanishi J, Matsumura N, Abiko K, Murat K, Baba T, et al. . Chemotherapy induces programmed cell death-ligand 1 overexpression via the nuclear factor-κB to foster an immunosuppressive tumor microenvironment in ovarian cancer. Cancer Res. (2015) 75:5034–45. 10.1158/0008-5472.CAN-14-3098
    1. Chae YK, Arya A, Iams W, Cruz MR, Chandra S, Choi J, et al. . Current landscape and future of dual anti-CTLA4 and PD-1/PD-L1 blockade immunotherapy in cancer; lessons learned from clinical trials with melanoma and non-small cell lung cancer (NSCLC). J Immunother Cancer. (2018) 6:39. 10.1186/s40425-018-0349-3
    1. Melero I, Berman DM, Aznar MA, Korman AJ, Gracia JLP, Haanen J. Evolving synergistic combinations of targeted immunotherapies to combat cancer. Nat Rev Cancer. (2015) 15:457. 10.1038/nrc3973
    1. Yang Z, Tam KY. Combination strategies using EGFR-TKi in NSCLC therapy: learning from the gap between pre-clinical results and clinical outcomes. Int J Biol Sci. (2018) 14:204. 10.7150/ijbs.22955
    1. Manegold C, Dingemans AC, Gray JE, Nakagawa K, Nicolson M, Peters S, et al. . The potential of combined immunotherapy and antiangiogenesis for the synergistic treatment of advanced NSCLC. J Thorac Oncol. (2017) 12:194–207. 10.1016/j.jtho.2016.10.003

Source: PubMed

Подписаться