Genomics of NSCLC patients both affirm PD-L1 expression and predict their clinical responses to anti-PD-1 immunotherapy

Kim A Brogden, Deepak Parashar, Andrea R Hallier, Terry Braun, Fang Qian, Naiyer A Rizvi, Aaron D Bossler, Mohammed M Milhem, Timothy A Chan, Taher Abbasi, Shireen Vali, Kim A Brogden, Deepak Parashar, Andrea R Hallier, Terry Braun, Fang Qian, Naiyer A Rizvi, Aaron D Bossler, Mohammed M Milhem, Timothy A Chan, Taher Abbasi, Shireen Vali

Abstract

Background: Programmed Death Ligand 1 (PD-L1) is a co-stimulatory and immune checkpoint protein. PD-L1 expression in non-small cell lung cancers (NSCLC) is a hallmark of adaptive resistance and its expression is often used to predict the outcome of Programmed Death 1 (PD-1) and PD-L1 immunotherapy treatments. However, clinical benefits do not occur in all patients and new approaches are needed to assist in selecting patients for PD-1 or PD-L1 immunotherapies. Here, we hypothesized that patient tumor cell genomics influenced cell signaling and expression of PD-L1, chemokines, and immunosuppressive molecules and these profiles could be used to predict patient clinical responses.

Methods: We used a recent dataset from NSCLC patients treated with pembrolizumab. Deleterious gene mutational profiles in patient exomes were identified and annotated into a cancer network to create NSCLC patient-specific predictive computational simulation models. Validation checks were performed on the cancer network, simulation model predictions, and PD-1 match rates between patient-specific predicted and clinical responses.

Results: Expression profiles of these 24 chemokines and immunosuppressive molecules were used to identify patients who would or would not respond to PD-1 immunotherapy. PD-L1 expression alone was not sufficient to predict which patients would or would not respond to PD-1 immunotherapy. Adding chemokine and immunosuppressive molecule expression profiles allowed patient models to achieve a greater than 85.0% predictive correlation among predicted and reported patient clinical responses.

Conclusions: Our results suggested that chemokine and immunosuppressive molecule expression profiles can be used to accurately predict clinical responses thus differentiating among patients who would and would not benefit from PD-1 or PD-L1 immunotherapies.

Trial registration: ClinicalTrials.gov NCT01295827.

Keywords: Computational modeling; Immunotherapy; NSCLC; PD-1; PD-L1.

Conflict of interest statement

Ethics approval and consent to participate

All patients had stage IV non-small cell lung cancer (NSCLC) and were treated at Memorial Sloan Kettering Cancer Center (n = 29) or the University of California at Los Angeles (n = 5) on protocol NCT01295827. All patients had consented to the Memorial Sloan Kettering Cancer Center Institutional Review Board-approved protocols permitting tissue collection and sequencing.

All patient related research was Memorial Sloan Kettering Cancer Center Institutional Review Board-approved and treated under protocol NCT0129827. Written informed consent was obtained from all patients.

Consent for publication

Not applicable.

Competing interests

KAB has had a Cooperative Research and Development Agreement with Cellworks Group Inc., San Jose, CA. TA and SV work for Cellworks Group Inc., San Jose, California. DP, NKS, and UM work for Cellworks Research India Ltd., Whitefield, Bangalore, India. TAC is a co-founder of Gritstone Oncology and holds equity in the company. All other authors declare no competing financial interests in the findings of this study or with Cellworks Group Inc., San Jose, CA or Cellworks Research India Ltd., Whitefield, Bangalore, India.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
The schema for creating predictive computational simulation models to predict molecule responses and identify patients that would respond or not respond to PD-1 immunotherapy treatment using patient SA97V5 as a model example. Exome information from patient SA97V5 (a) contained 1192 total mutations with 36 deleterious mutations. This profile (b) was converted from a mutational profile to a computational format and annotated into the computational workflow to convert (c) a nontransformed model in the cancer network into (d) a patient SA97V5-specific simulation model. The patient SA97V5-specific simulation model was used to predict PD-L1 expression (e.g., 67.0% with respect to control), dendritic cell (DC) infiltration index (e.g., 23.8% with respect to control); and an immunosuppressive molecule expression profile (e.g., range − 1.9% to 56.5% with respect to controls) (e). Predicted expression responses were all used (f) to sort patients into groups that would respond or not respond to PD-1 immunotherapy treatment. SA97V5 was identified as a patient who would respond to PD-1 immunotherapy treatment. Numerous validation checks (g) occurred on the cancer network, the simulation model predictions, and the PD-1 match rates between the predicted responses and the patient clinical responses
Fig. 2
Fig. 2
A decision tree was used to identify PD-1 drug responder status. At step 1, 9 patients with PD-L1 expression below 29.0% were identified as PD-1 drug non-responders. The remaining 16 patients (including patient SA97V5) with PD-L1 expression equal to or greater than 29.0% proceeded to step 2. At Steps 2a and 2b, 2 patients with dendritic cell infiltration index values below 20.0% were identified as non-responders and 2 patients with index values greater than 60.0% were identified as PD-1 drug responders. Twelve patients with index values greater than 20.0% (including patient SA97V5), but less than 60.0% proceeded to step 3. At Step 3, 4 patients with immunosuppressive molecule (ISM) values higher than that of their PD-L1 expression with a margin of greater than 5.0%, were identified as non-responders (Step 3a) and 8 patient-specific models with values lower than that of their PD-L1 expression with a margin of greater than 5.0% were identified as responders (Step 3b, including patient SA97V5). Mismatch patients GI7AGZ, 2FCOH7, F3FK2W were not listed
Fig. 3
Fig. 3
Patient-specific simulation models were used to predict the expression of PD-L1 (a) and at step 1 of the decision tree, 9 patients (black bars) with predicted PD-L1 expression below 29.0% (bold line) were identified as PD-1 drug non-responders. Patient SA97V5 had a predicted PD-L1 expression of 67.0%. Patient-specific simulation models were used to predict the expression of chemokines used to create a dendritic cell (DC) infiltration index (b). At step 2a of the decision tree, 2 patients (black bars) with index values greater that 60.0% (bold line) were identified as PD-1 drug responders and 2 patients (black bars) with index values less than 20.0% (black line) were identified as PD-1 drug non-responders. Patient SA97V5 had a predicted DC infiltration index of 23.9%
Fig. 4
Fig. 4
Patient-specific simulation models were used to predict the expression of 14 immunosuppressive molecules. At step 3 of the decision tree, patients with immunosuppressive molecule predictions higher than that of PD-L1 with a margin of greater than 5.0% (bold line), were considered to be non-responders and patient-specific models with predictions lower than that of PD-L1 with a margin of greater than 5.0% were considered to be responders. Eight remaining patients were identified as responders and 4 remaining patients were identified as non-responders. Patient SA97V5 (a) had all 14 molecules below the threshold and was identified as a PD-1 drug responder. Patient QIA43T (b) had 2 molecules above the threshold and was identified as a PD-1 drug non-responder
Fig. 5
Fig. 5
The expression of PD-L1 was influenced via a number of signaling pathways. Activating signals were processed via the ERK signaling pathway (via EGFR; B-Raf proto-oncogene, serine/threonine kinase, BRAF-V600E; mitogen-activated protein kinase kinase 1/2, MEK1/2; mitogen-activated protein kinase kinase 1, MAP2K1; MAP2K2; ERK1/2; mitogen-activated protein kinase 3, MAPK3; mitogen-activated protein kinase 1, MAPK1; and Jun proto-oncogene, c-Jun). Activating signals were processed via the EGFR signaling pathway (via neuroblastoma RAS viral oncogene homolog, NRAS; phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha, PIK3CA; V-akt murine thymoma viral oncogene homolog, AKT; mechanistic target of rapamycin, MTOR; and STAT3). Also, activating signals were processed via the interferon gamma (IFNG) pathway (via IFNG; interferon gamma receptor 1, IFNGR1; signal transducer and activator of transcription 1, STAT1; and interferon regulatory factor 1, IRF1). Pathway signals converge to activation factors Activator protein 1 (AP1), STAT1, STAT3, and IRF1 leading to transcription of PD-L1 genes. Common pathways were utilized among a number patient-specific simulation models. Patient C9TGAJ (KRAS mutation), patient RDD2UW (KRAS mutation), patient M9GYO4 (MAP2K2 mutation), and patient DFZLO2 (MAP3K1 mutation) involved the ERK activation pathway. Patient P90A0O (BRAF1, TP53 mutations) and patient L8MTGU (KRAS, TP53 mutations) involved the ERK activation and apoptotic pathways

References

    1. Shih K, Arkenau HT, Infante JR. Clinical impact of checkpoint inhibitors as novel cancer therapies. Drugs. 2014;74(17):1993–2013. doi: 10.1007/s40265-014-0305-6.
    1. Sunshine J, Taube JM. PD-1/PD-L1 inhibitors. Curr Opin Pharmacol. 2015;23:32–38. doi: 10.1016/j.coph.2015.05.011.
    1. He J, Hu Y, Hu M, Li B. Development of PD-1/PD-L1 pathway in tumor immune microenvironment and treatment for non-small cell lung cancer. Sci Rep. 2015;5:13110. doi: 10.1038/srep13110.
    1. Shukuya T, Carbone DP. Predictive markers for the efficacy of anti-PD-1/PD-L1 antibodies in lung cancer. J Thorac Oncol. 2016;11(7):976–988. doi: 10.1016/j.jtho.2016.02.015.
    1. Carbognin L, Pilotto S, Milella M, Vaccaro V, Brunelli M, Calio A, Cuppone F, Sperduti I, Giannarelli D, Chilosi M, et al. Differential activity of Nivolumab, Pembrolizumab and MPDL3280A according to the tumor expression of programmed death-ligand-1 (PD-L1): sensitivity analysis of trials in melanoma, lung and genitourinary cancers. PLoS One. 2015;10(6):e0130142. doi: 10.1371/journal.pone.0130142.
    1. McLaughlin J, Han G, Schalper KA, Carvajal-Hausdorf D, Pelekanou V, Rehman J, Velcheti V, Herbst R, LoRusso P, Rimm DL. Quantitative assessment of the heterogeneity of PD-L1 expression in non-small-cell lung cancer. JAMA Oncology. 2016;2(1):46–54. doi: 10.1001/jamaoncol.2015.3638.
    1. Kerr KM, Hirsch FR. Programmed death ligand 1 immunohistochemistry: friend or foe? Arch Pathol Lab Med. 2016;140(4):326–331. doi: 10.5858/arpa.2015-0522-SA.
    1. Yu H, Boyle TA, Zhou C, Rimm DL, Hirsch FR. PD-L1 expression in lung cancer. J Thorac Oncol. 2016;11(7):964–975. doi: 10.1016/j.jtho.2016.04.014.
    1. Brunnstrom H, Johansson A, Westbom-Fremer S, Backman M, Djureinovic D, Patthey A, Isaksson-Mettavainio M, Gulyas M, Micke P. PD-L1 immunohistochemistry in clinical diagnostics of lung cancer: inter-pathologist variability is higher than assay variability. Mod Pathol. 2017;30(10):1411–1421. doi: 10.1038/modpathol.2017.59.
    1. Hirsch FR, McElhinny A, Stanforth D, Ranger-Moore J, Jansson M, Kulangara K, Richardson W, Towne P, Hanks D, Vennapusa B, et al. PD-L1 immunohistochemistry assays for lung cancer: results from phase 1 of the blueprint PD-L1 IHC assay comparison project. J Thorac Oncol. 2017;12(2):208–222. doi: 10.1016/j.jtho.2016.11.2228.
    1. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, McDermott DF, Powderly JD, Gettinger SN, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515(7528):563–567. doi: 10.1038/nature14011.
    1. Chae YK, Pan A, Davis AA, Raparia K, Mohindra NA, Matsangou M, Giles FJ. Biomarkers for PD-1/PD-L1 blockade therapy in non-small-cell lung cancer: is PD-L1 expression a good marker for patient selection? Clinical Lung Cancer. 2016;17(5):350–361. doi: 10.1016/j.cllc.2016.03.011.
    1. Bhaijee F, Anders RA. PD-L1 expression as a predictive biomarker: is absence of proof the same as proof of absence? JAMA Oncology. 2016;2(1):54–55. doi: 10.1001/jamaoncol.2015.3782.
    1. Blank CU, Haanen JB, Ribas A, Schumacher TN. The “cancer immunogram”. Science. 2016;352(6286):658–660. doi: 10.1126/science.aaf2834.
    1. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348(6230):124–128. doi: 10.1126/science.aaa1348.
    1. Martelotto LG, Ng CK, De Filippo MR, Zhang Y, Piscuoglio S, Lim RS, Shen R, Norton L, Reis-Filho JS, Weigelt B. Benchmarking mutation effect prediction algorithms using functionally validated cancer-related missense mutations. Genome Biol. 2014;15(10):484. doi: 10.1186/s13059-014-0484-1.
    1. Reva B, Antipin Y, Sander C. Predicting the functional impact of protein mutations: application to cancer genomics. Nucleic Acids Res. 2011;39(17):e118. doi: 10.1093/nar/gkr407.
    1. Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, Kondrashov AS, Sunyaev SR. A method and server for predicting damaging missense mutations. Nat Methods. 2010;7(4):248–249. doi: 10.1038/nmeth0410-248.
    1. Choi Y, Sims GE, Murphy S, Miller JR, Chan AP. Predicting the functional effect of amino acid substitutions and indels. PLoS One. 2012;7(10):e46688. doi: 10.1371/journal.pone.0046688.
    1. Sim NL, Kumar P, Hu J, Henikoff S, Schneider G, Ng PC: SIFT web server: predicting effects of amino acid substitutions on proteins. In: Nucleic Acids Res. vol. 40 (Web Server issue); 2012: W452–7. doi: 10.1093/nar/gks539.
    1. Kholodenko BN, Demin OV, Moehren G, Hoek JB. Quantification of short term signaling by the epidermal growth factor receptor. J Biol Chem. 1999;274(42):30169–30181. doi: 10.1074/jbc.274.42.30169.
    1. Kaushik P, Gorin F, Vali S. Dynamics of tyrosine hydroxylase mediated regulation of dopamine synthesis. J Comput Neurosci. 2007;22(2):147–160. doi: 10.1007/s10827-006-0004-8.
    1. Hairer E, Wanner G. Stiff differential equations solved by Radau methods. J Comput Appl Math. 1999;111(1–2):93–111. doi: 10.1016/S0377-0427(99)00134-X.
    1. Lanzel EA, Hernandez MPG, Bates AM, Treinen CN, Starman EE, Fischer CL, Parashar D, Guthmiller JM, Johnson GK, Abbasi T, et al. Predicting PD-L1 expression on human cancer cells using next-generation sequencing information in computational simulation models. Cancer Immunol Immunother. 2016;65(12):1511–1522. doi: 10.1007/s00262-016-1907-5.
    1. Sagar D, Lamontagne A, Foss CA, Khan ZK, Pomper MG, Jain P. Dendritic cell CNS recruitment correlates with disease severity in EAE via CCL2 chemotaxis at the blood-brain barrier through paracellular transmigration and ERK activation. J Neuroinflammation. 2012;9:245. doi: 10.1186/1742-2094-9-245.
    1. Charmoy M, Brunner-Agten S, Aebischer D, Auderset F, Launois P, Milon G, Proudfoot AE, Tacchini-Cottier F. Neutrophil-derived CCL3 is essential for the rapid recruitment of dendritic cells to the site of Leishmania major inoculation in resistant mice. PLoS Pathog. 2010;6(2):e1000755. doi: 10.1371/journal.ppat.1000755.
    1. Nobile C, Lind M, Miro F, Chemin K, Tourret M, Occhipinti G, Dogniaux S, Amigorena S, Hivroz C. Cognate CD4+ T-cell-dendritic cell interactions induce migration of immature dendritic cells through dissolution of their podosomes. Blood. 2008;111(7):3579–3590. doi: 10.1182/blood-2007-08-107755.
    1. Chabot V, Reverdiau P, Iochmann S, Rico A, Senecal D, Goupille C, Sizaret PY, Sensebe L. CCL5-enhanced human immature dendritic cell migration through the basement membrane in vitro depends on matrix metalloproteinase-9. J Leukoc Biol. 2006;79(4):767–778. doi: 10.1189/jlb.0804464.
    1. Beaulieu S, Robbiani DF, Du X, Rodrigues E, Ignatius R, Wei Y, Ponath P, Young JW, Pope M, Steinman RM, et al. Expression of a functional eotaxin (CC chemokine ligand 11) receptor CCR3 by human dendritic cells. J Immunol. 2002;169(6):2925–2936. doi: 10.4049/jimmunol.169.6.2925.
    1. Demedts IK, Bracke KR, Van Pottelberge G, Testelmans D, Verleden GM, Vermassen FE, Joos GF, Brusselle GG. Accumulation of dendritic cells and increased CCL20 levels in the airways of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2007;175(10):998–1005. doi: 10.1164/rccm.200608-1113OC.
    1. Johnson LA, Jackson DG. The chemokine CX3CL1 promotes trafficking of dendritic cells through inflamed lymphatics. J Cell Sci. 2013;126(Pt 22):5259–5270. doi: 10.1242/jcs.135343.
    1. Hall M, Frank E, Holmes G, Pfahringer B, Reutemann P, Witten IH: The WEKA data mining software: an update. In: ACM SIGKDD Explorations Newsletter. 2009;11(1):10–18.
    1. Dong ZY, Zhong WZ, Zhang XC, Su J, Xie Z, Liu SY, Tu HY, Chen HJ, Sun YL, Zhou Q, et al. Potential predictive value of TP53 and KRAS mutation status for response to PD-1 blockade immunotherapy in lung adenocarcinoma. Clin Cancer Res. 2017;23(12):3012–3024. doi: 10.1158/1078-0432.CCR-16-2554.
    1. Skoulidis F, Byers LA, Diao L, Papadimitrakopoulou VA, Tong P, Izzo J, Behrens C, Kadara H, Parra ER, Canales JR, et al. Co-occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities. Cancer Discov. 2015;5(8):860–877. doi: 10.1158/-14-1236.
    1. Zitvogel L, Kroemer G. Targeting PD-1/PD-L1 interactions for cancer immunotherapy. OncoImmunology. 2012;1(8):1223–1225. doi: 10.4161/onci.21335.
    1. Malm IJ, Bruno TC, Fu J, Zeng Q, Taube JM, Westra W, Pardoll D, Drake CG, Kim YJ. Expression profile and in vitro blockade of programmed death-1 in human papillomavirus-negative head and neck squamous cell carcinoma. Head Neck. 2015;37(8):1088–1095. doi: 10.1002/hed.23706.
    1. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8(8):793–800. doi: 10.1038/nm730.
    1. 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 costimulatory properties for T cells. J Exp Med. 2001;193(7):839–846. doi: 10.1084/jem.193.7.839.
    1. Jiang Y, Li Y, Zhu B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis. 2015;6:e1792. doi: 10.1038/cddis.2015.162.
    1. Borghaei H, Brahmer JR, Horn L, Ready N, Steins M, Felip E, Paz-Ares LG, Arrieta O, Barlesi F, Antonia SJ, et al. Nivolumab (nivo) vs docetaxel (doc) in patients (pts) with advanced NSCLC: CheckMate 017/057 2-y update and exploratory cytokine profile analyses. J Clin Oncol. 2016;34(15_suppl):9025.
    1. Balkwill F. Cancer and the chemokine network. Nat Rev Cancer. 2004;4(7):540–550. doi: 10.1038/nrc1388.
    1. Perrot I, Blanchard D, Freymond N, Isaac S, Guibert B, Pacheco Y, Lebecque S. Dendritic cells infiltrating human non-small cell lung cancer are blocked at immature stage. J Immunol. 2007;178(5):2763–2769. doi: 10.4049/jimmunol.178.5.2763.
    1. Mukaida N, Sasaki S, Baba T. Chemokines in cancer development and progression and their potential as targeting molecules for cancer treatment. Mediat Inflamm. 2014;2014:170381. doi: 10.1155/2014/170381.
    1. Inoshima N, Nakanishi Y, Minami T, Izumi M, Takayama K, Yoshino I, Hara N. The influence of dendritic cell infiltration and vascular endothelial growth factor expression on the prognosis of non-small cell lung cancer. Clin Cancer Res. 2002;8(11):3480–3486.
    1. Menetrier-Caux C, Montmain G, Dieu MC, Bain C, Favrot MC, Caux C, Blay JY. Inhibition of the differentiation of dendritic cells from CD34(+) progenitors by tumor cells: role of interleukin-6 and macrophage colony-stimulating factor. Blood. 1998;92(12):4778–4791.
    1. Hegde S, Pahne J, Smola-Hess S. Novel immunosuppressive properties of interleukin-6 in dendritic cells: inhibition of NF-kappaB binding activity and CCR7 expression. FASEB J. 2004;18(12):1439–1441. doi: 10.1096/fj.03-0969fje.
    1. Ishii H, Tanaka S, Masuyama K. Therapeutic strategy for cancer immunotherapy in head and neck cancer. Adv Cell Mol Otolaryngol. 2015;3:27690. doi: 10.3402/acmo.v3.27690.
    1. Pinzon-Charry A, Maxwell T, Lopez JA. Dendritic cell dysfunction in cancer: a mechanism for immunosuppression. Immunol Cell Biol. 2005;83(5):451–461. doi: 10.1111/j.1440-1711.2005.01371.x.
    1. Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol. 2007;25:267–296. doi: 10.1146/annurev.immunol.25.022106.141609.
    1. Sauter ER, Nesbit M, Watson JC, Klein-Szanto A, Litwin S, Herlyn M. Vascular endothelial growth factor is a marker of tumor invasion and metastasis in squamous cell carcinomas of the head and neck. Clin Cancer Res. 1999;5(4):775–782.
    1. Moutsopoulos NM, Wen J, Wahl SM. TGF-beta and tumors--an ill-fated alliance. Curr Opin Immunol. 2008;20(2):234–240. doi: 10.1016/j.coi.2008.04.003.
    1. Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, Kavanaugh D, Carbone DP. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med. 1996;2(10):1096–1103. doi: 10.1038/nm1096-1096.
    1. Munn DH, Zhou M, Attwood JT, Bondarev I, Conway SJ, Marshall B, Brown C, Mellor AL. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science. 1998;281(5380):1191–1193. doi: 10.1126/science.281.5380.1191.
    1. Lob S, Konigsrainer A, Rammensee HG, Opelz G, Terness P. Inhibitors of indoleamine-2,3-dioxygenase for cancer therapy: can we see the wood for the trees? Nat Rev Cancer. 2009;9(6):445–452. doi: 10.1038/nrc2639.
    1. Spranger S, Spaapen RM, Zha Y, Williams J, Meng Y, Ha TT, Gajewski TF. Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells. Sci Transl Med. 2013;5(200):200ra116. doi: 10.1126/scitranslmed.3006504.
    1. Iversen TZ, Andersen MH, Svane IM. The targeting of indoleamine 2,3 dioxygenase -mediated immune escape in cancer. Basic Clin Pharmacol Toxicol. 2015;116(1):19–24. doi: 10.1111/bcpt.12320.
    1. Pilotte L, Larrieu P, Stroobant V, Colau D, Dolusic E, Frederick R, De Plaen E, Uyttenhove C, Wouters J, Masereel B, et al. Reversal of tumoral immune resistance by inhibition of tryptophan 2,3-dioxygenase. Proc Natl Acad Sci U S A. 2012;109(7):2497–2502. doi: 10.1073/pnas.1113873109.
    1. Holt D, Ma X, Kundu N, Fulton A. Prostaglandin E(2) (PGE (2)) suppresses natural killer cell function primarily through the PGE(2) receptor EP4. Cancer Immunol Immunother. 2011;60(11):1577–1586. doi: 10.1007/s00262-011-1064-9.
    1. Li H, Wu K, Tao K, Chen L, Zheng Q, Lu X, Liu J, Shi L, Liu C, Wang G, et al. Tim-3/galectin-9 signaling pathway mediates T-cell dysfunction and predicts poor prognosis in patients with hepatitis B virus-associated hepatocellular carcinoma. Hepatology. 2012;56(4):1342–1351. doi: 10.1002/hep.25777.
    1. Avogadri F, Yuan J, Yang A, Schaer D, Wolchok JD. Modulation of CTLA-4 and GITR for cancer immunotherapy. Curr Top Microbiol Immunol. 2011;344:211–244.
    1. Mocellin S, Nitti D. CTLA-4 blockade and the renaissance of cancer immunotherapy. Biochim Biophys Acta. 2013;1836(2):187–196.
    1. Grosso JF, Jure-Kunkel MN. CTLA-4 blockade in tumor models: an overview of preclinical and translational research. Cancer Immun. 2013;13:5.
    1. Peguet-Navarro J, Sportouch M, Popa I, Berthier O, Schmitt D, Portoukalian J. Gangliosides from human melanoma tumors impair dendritic cell differentiation from monocytes and induce their apoptosis. J Immunol. 2003;170(7):3488–3494. doi: 10.4049/jimmunol.170.7.3488.
    1. Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity. 2016;44(5):989–1004. doi: 10.1016/j.immuni.2016.05.001.
    1. Monney L, Sabatos CA, Gaglia JL, Ryu A, Waldner H, Chernova T, Manning S, Greenfield EA, Coyle AJ, Sobel RA, et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature. 2002;415(6871):536–541. doi: 10.1038/415536a.
    1. Anderson AC. Tim-3: an emerging target in the cancer immunotherapy landscape. Cancer Immunol Res. 2014;2(5):393–398. doi: 10.1158/2326-6066.CIR-14-0039.
    1. Pedoeem A, Azoulay-Alfaguter I, Strazza M, Silverman GJ, Mor A. Programmed death-1 pathway in cancer and autoimmunity. Clin Immunol. 2014;153(1):145–152. doi: 10.1016/j.clim.2014.04.010.
    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(3):409–416. doi: 10.1093/annonc/mdv615.
    1. Ritprajak P, Azuma M. Intrinsic and extrinsic control of expression of the immunoregulatory molecule PD-L1 in epithelial cells and squamous cell carcinoma. Oral Oncol. 2015;51(3):221–228. doi: 10.1016/j.oraloncology.2014.11.014.
    1. Parsa AT, Waldron JS, Panner A, Crane CA, Parney IF, Barry JJ, Cachola KE, Murray JC, Tihan T, Jensen MC, et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med. 2007;13(1):84–88. doi: 10.1038/nm1517.
    1. Lastwika KJ, Wilson W, 3rd, Li QK, Norris J, Xu H, Ghazarian SR, Kitagawa H, Kawabata S, Taube JM, Yao S, 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(2):227–238. doi: 10.1158/0008-5472.CAN-14-3362.
    1. Casey SC, Tong L, Li Y, Do R, Walz S, Fitzgerald KN, Gouw AM, Baylot V, Gutgemann I, Eilers M, et al. MYC regulates the antitumor immune response through CD47 and PD-L1. Science. 2016;352(6282):227–231. doi: 10.1126/science.aac9935.

Source: PubMed

3
Předplatit