Quantitative Assessment of the Heterogeneity of PD-L1 Expression in Non-Small-Cell Lung Cancer.

IMPORTANCE Early-phase trials with monoclonal antibodies targeting PD-1 (programmed cell death protein 1) and PD-L1 (programmed cell death 1 ligand 1) have demonstrated durable clinical responses in patients with non-small-cell lung cancer (NSCLC). However, current assays for the prognostic and/or predictive role of tumor PD-L1 expression are not standardized with respect to either quantity or distribution of expression. OBJECTIVE To demonstrate PD-L1 protein distribution in NSCLC tumors using both conventional immunohistochemistry (IHC) and quantitative immunofluorescence (QIF) and compare results obtained using 2 different PD-L1 antibodies. DESIGN, SETTING, AND PARTICIPANTS PD-L1 was measured using E1L3N and SP142, 2 rabbit monoclonal antibodies, in 49 NSCLC whole-tissue sections and a corresponding tissue microarray with the same 49 cases. Non-small-cell lung cancer biopsy specimens from 2011 to 2012 were collected retrospectively from the Yale Thoracic Oncology Program Tissue Bank. Human melanoma Mel 624 cells stably transfected with PD-L1 as well as Mel 624 parental cells, and human term placenta whole tissue sections were used as controls and for antibody validation. PD-L1 protein expression in tumor and stroma was assessed using chromogenic IHC and the AQUA (Automated Quantitative Analysis) method of QIF. Tumor-infiltrating lymphocytes (TILs) were scored in hematoxylin-eosin slides using current consensus guidelines. The association between PD-L1 protein expression, TILs, and clinicopathological features were determined. MAIN OUTCOMES AND MEASURES PD-L1 expression discordance or heterogeneity using the diaminobenzidine chromogen and QIF was the main outcome measure selected prior to performing the study. RESULTS Using chromogenic IHC, both antibodies showed fair to poor concordance. The PD-L1 antibodies showed poor concordance (Cohen κ range, 0.124-0.340) using conventional chromogenic IHC and showed intra-assay heterogeneity (E1L3N coefficient of variation [CV], 6.75%-75.24%; SP142 CV, 12.17%-109.61%) and significant interassay discordance using QIF (26.6%). Quantitative immunofluorescence showed that PD-L1 expression using both PD-L1 antibodies was heterogeneous. Using QIF, the scores obtained with E1L3N and SP142 for each tumor were significantly different according to nonparametric paired test (P < .001). Assessment of 588 serial section fields of view from whole tissue showed discordant expression at a frequency of 25%. Expression of PD-L1 was correlated with high TILs using both E1L3N (P = .007) and SP142 (P = .02). CONCLUSIONS AND RELEVANCE Objective determination of PD-L1 protein levels in NSCLC reveals heterogeneity within tumors and prominent interassay variability or discordance. This could be due to different antibody affinities, limited specificity, or distinct target epitopes. Efforts to determine the clinical value of these observations are under way.

[1]  D. Rimm,et al.  Automated subcellular localization and quantification of protein expression in tissue microarrays , 2002, Nature Medicine.

[2]  Haidong Dong,et al.  Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion , 2002, Nature Medicine.

[3]  P. Loke,et al.  PD-L1 and PD-L2 are differentially regulated by Th1 and Th2 cells , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[4]  M. Nishimura,et al.  B7-H1 Expression on Non-Small Cell Lung Cancer Cells and Its Relationship with Tumor-Infiltrating Lymphocytes and Their PD-1 Expression , 2004, Clinical Cancer Research.

[5]  A. Qattan,et al.  The B7-H1 (PD-L1) T lymphocyte-inhibitory molecule is expressed in breast cancer patients with infiltrating ductal carcinoma: correlation with important high-risk prognostic factors. , 2006, Neoplasia.

[6]  G. Freeman,et al.  Tissue expression of PD-L1 mediates peripheral T cell tolerance , 2006, The Journal of experimental medicine.

[7]  N. Xu,et al.  Immunohistochemical localization of programmed death-1 ligand-1 (PD-L1) in gastric carcinoma and its clinical significance. , 2006, Acta histochemica.

[8]  A. Mackensen,et al.  Contribution of the PD-L1/PD-1 pathway to T-cell exhaustion: an update on implications for chronic infections and tumor evasion , 2007, Cancer Immunology, Immunotherapy.

[9]  J. Allison,et al.  The B7 Family and Cancer Therapy: Costimulation and Coinhibition , 2007, Clinical Cancer Research.

[10]  野見 武男 Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer , 2007 .

[11]  Yoshimasa Tanaka,et al.  Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer , 2007, Proceedings of the National Academy of Sciences.

[12]  G. Freeman,et al.  PD-1 and its ligands in tolerance and immunity. , 2008, Annual review of immunology.

[13]  Lieping Chen,et al.  Inhibitory B7-family molecules in the tumour microenvironment , 2008, Nature Reviews Immunology.

[14]  R. Nurieva,et al.  Yin–Yang of costimulation: crucial controls of immune tolerance and function , 2009, Immunological reviews.

[15]  Yun Li,et al.  gp96与免疫相关基因CTLA-4、CD8在肺癌组织芯片中的表达及意义 , 2010, Zhongguo fei ai za zhi = Chinese journal of lung cancer.

[16]  T. Okazaki,et al.  Tumor cell expression of programmed cell death‐1 ligand 1 is a prognostic factor for malignant melanoma , 2010, Cancer.

[17]  Amal Hasan,et al.  Therapeutic targeting of B7-H1 in breast cancer , 2011, Expert opinion on therapeutic targets.

[18]  C. Mu,et al.  High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor infiltrating dendritic cells maturation , 2011, Medical oncology.

[19]  R. Schreiber,et al.  Cancer Immunoediting: Integrating Immunity’s Roles in Cancer Suppression and Promotion , 2011, Science.

[20]  C. Sautès-Fridman,et al.  The immune contexture in human tumours: impact on clinical outcome , 2012, Nature Reviews Cancer.

[21]  Drew M. Pardoll,et al.  The blockade of immune checkpoints in cancer immunotherapy , 2012, Nature Reviews Cancer.

[22]  David C. Smith,et al.  Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. , 2012, The New England journal of medicine.

[23]  C. Drake,et al.  Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. , 2012, The New England journal of medicine.

[24]  Antoni Ribas,et al.  Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. , 2013, The New England journal of medicine.

[25]  J. Sosman,et al.  A study of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic tumors. , 2013 .

[26]  I. Mellman,et al.  Oncology meets immunology: the cancer-immunity cycle. , 2013, Immunity.

[27]  Lieping Chen,et al.  Molecular mechanisms of T cell co-stimulation and co-inhibition , 2013, Nature Reviews Immunology.

[28]  David C. Smith,et al.  Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[29]  R. Herbst,et al.  Programmed death ligand-1 expression in non-small cell lung cancer , 2014, Laboratory Investigation.

[30]  M. Dolled-Filhart,et al.  Safety and clinical activity of MK-3475 in previously treated patients (pts) with non-small cell lung cancer (NSCLC). , 2014 .

[31]  H. Kohrt,et al.  Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients , 2014, Nature.

[32]  J. Taube,et al.  Association of PD-1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti–PD-1 Therapy , 2014, Clinical Cancer Research.

[33]  R. Herbst,et al.  Objective measurement and clinical significance of TILs in non-small cell lung cancer. , 2015, Journal of the National Cancer Institute.

[34]  J. Madore,et al.  PD‐L1 expression in melanoma shows marked heterogeneity within and between patients: implications for anti‐PD‐1/PD‐L1 clinical trials , 2015, Pigment cell & melanoma research.

[35]  T. Nielsen,et al.  The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. , 2015, Annals of oncology : official journal of the European Society for Medical Oncology.

[36]  J. Ahn,et al.  Pembrolizumab for the treatment of non-small cell lung cancer , 2016, Expert opinion on biological therapy.

[37]  M. Jakopović,et al.  Immunotherapy in the treatment of non-small cell lung cancer , 2017 .