Programmed Cell Death Ligand 1 Expression in Osteosarcoma

Shen, Cote, and colleagues developed a quantitative RNA-based PD-L1 assay and report PD-L1 expression in 84% of human osteosarcomas, of which 24% were at high levels and correlated with TILs, indicating that this subset of patients may benefit from anti-PD-L1 immunotherapy. Programmed cell death ligand 1 (PDL1, also known as B7H1) is a cell-surface protein that suppresses the cytotoxic CD8+ T-cell–mediated immune response. PDL1 expression and its clinical relevance in sarcomas are not well understood. Therefore, we sought to measure RNA expression levels for PDL1 in 38 clinically annotated osteosarcoma tumor samples and aimed to determine if PDL1 expression correlates with clinical features and tumor-infiltrating lymphocytes (TIL). Quantitative real-time RT-PCR for PDL1 was optimized in 18 cell lines, of which 5 were osteosarcoma derived. qRT-PCR results were validated via flow cytometry and immunohistochemistry (IHC) in select cell lines. Total RNA was isolated from 38 human osteosarcoma samples for qRT-PCR analysis. Clinical data were sorted, and significance was determined by the Student t test. TILs were examined in patient samples by tissue microarray hematoxylin–eosin staining. We confirmed the constitutive PDL1 mRNA expression in cell lines by qRT-PCR, flow cytometry, and IHC. Across human osteosarcoma samples, PDL1 mRNA gene expression ranged over 4 log (>5,000-fold difference). Relative expression levels were evaluated against clinical factors such as age/gender, metastasis, recurrence, chemotherapy, percentage of necrosis, and survival; no significant associations were identified. The presence of TILs was associated with high PDL1 expression (R2 = 0.37; P = 0.01). In summary, we developed an RNA-based assay to determine PDL1 expression levels, and we show, for the first time, that high levels of PDL1 are expressed in a subset of osteosarcoma, and PDL1 expression is positively correlated with TILs. Multiple agents targeting PD1/PDL1 are in clinical development, and this may be a novel immunotherapeutic strategy for osteosarcoma clinical trials. Cancer Immunol Res; 2(7); 690–8. ©2014 AACR.

[1]  H. Mankin,et al.  Tissue Microarray Immunohistochemical Detection of Brachyury Is Not a Prognostic Indicator in Chordoma , 2013, PloS one.

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

[3]  L. Terracciano,et al.  Clinical impact of programmed cell death ligand 1 expression in colorectal cancer. , 2013, European journal of cancer.

[4]  R. Herbst,et al.  Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic non-small cell lung cancer (NSCLC). , 2013 .

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

[6]  J. Sosman,et al.  Biomarkers and associations with the clinical activity of PD-L1 blockade in a MPDL3280A study. , 2013 .

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

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

[9]  Alison P. Klein,et al.  Colocalization of Inflammatory Response with B7-H1 Expression in Human Melanocytic Lesions Supports an Adaptive Resistance Mechanism of Immune Escape , 2012, Science Translational Medicine.

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

[11]  B. Ksander,et al.  Tumor Cell Programmed Death Ligand 1-Mediated T Cell Suppression Is Overcome by Coexpression of CD80 , 2011, The Journal of Immunology.

[12]  P. Tien,et al.  PD‐1 and PD‐L1 upregulation promotes CD8+ T‐cell apoptosis and postoperative recurrence in hepatocellular carcinoma patients , 2011, International journal of cancer.

[13]  Israel Lowy,et al.  Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[14]  L. Mirabello,et al.  Osteosarcoma incidence and survival rates from 1973 to 2004 , 2009, Cancer.

[15]  R. Gorlick,et al.  Novel therapeutic agents for osteosarcoma , 2009, Expert review of anticancer therapy.

[16]  S. Qiu,et al.  Overexpression of PD-L1 Significantly Associates with Tumor Aggressiveness and Postoperative Recurrence in Human Hepatocellular Carcinoma , 2009, Clinical Cancer Research.

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

[18]  S. Dermime,et al.  FOXP3+ Tregs and B7-H1+/PD-1+ T lymphocytes co-infiltrate the tumor tissues of high-risk breast cancer patients: Implication for immunotherapy , 2008, BMC Cancer.

[19]  M. van Glabbeke,et al.  Improvement in histologic response but not survival in osteosarcoma patients treated with intensified chemotherapy: a randomized phase III trial of the European Osteosarcoma Intergroup. , 2007, Journal of the National Cancer Institute.

[20]  B. Seddon,et al.  Management of osteosarcoma , 2006, Current treatment options in oncology.

[21]  J. Cheville,et al.  Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. , 2006, Cancer research.

[22]  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.

[23]  M. Azuma,et al.  Clinical Significance of Programmed Death-1 Ligand-1 and Programmed Death-1 Ligand-2 Expression in Human Esophageal Cancer , 2005, Clinical Cancer Research.

[24]  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.

[25]  M. Gebhardt,et al.  Biology and therapeutic advances for pediatric osteosarcoma. , 2004, The oncologist.

[26]  D. Hawkins,et al.  Pattern of disease recurrence and prognostic factors in patients with osteosarcoma treated with contemporary chemotherapy , 2003, Cancer.

[27]  M. Weller,et al.  Expression of the B7-related molecule B7-H1 by glioma cells: a potential mechanism of immune paralysis. , 2003, Cancer research.

[28]  J. Cheville,et al.  B7-H1 blockade augments adoptive T-cell immunotherapy for squamous cell carcinoma. , 2003, Cancer research.

[29]  G. Freeman,et al.  Blockade of Programmed Death-1 Ligands on Dendritic Cells Enhances T Cell Activation and Cytokine Production 1 , 2003, The Journal of Immunology.

[30]  A. Huvos,et al.  Intensification of preoperative chemotherapy for osteogenic sarcoma: results of the Memorial Sloan-Kettering (T12) protocol. , 1998, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

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

[32]  C. Fletcher,et al.  WHO classification of tumours of soft tissue and bone , 2013 .

[33]  C. Beauchamp Second and Subsequent Recurrences of Osteosarcoma: Presentation, Treatment, and Outcomes of 249 Consecutive Cooperative Osteosarcoma Study Group Patients , 2009 .

[34]  G. Ottaviani,et al.  The epidemiology of osteosarcoma. , 2009, Cancer treatment and research.

[35]  J. Cheville,et al.  Costimulatory B7-H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

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