51
|
Abstract
OPINION STATEMENT Biliary tract cancers (BTCs) are rare aggressive neoplasms with a poor prognosis and a median survival of less than 1 year in the locally advanced or metastatic setting. Among the few patients who undergo curative resection the recurrence rates are high. About 90% of patients are detected at advanced stages, and systemic chemotherapy is the mainstay of their treatment. The treatment options for these patients are limited and multiple modalities of therapy from targeted therapy to immunotherapy and combination therapies (immunotherapy, targeted therapy, and chemotherapy) have been tested in this disease. Targeted therapies have failed to show a survival benefit. The deregulation of the immune system plays a significant role in the pathogenesis of BTCs. Therefore, immunotherapy, especially, immune checkpoint inhibitors hold great promise for this group of cancers. Numerous trials of immunotherapy in BTC are currently ongoing. In this review, we will discuss the available data and evidence for immunotherapy in BTC.
Collapse
Affiliation(s)
- Urvi A Shah
- Department of Medical Oncology, Montefiore Medical Center, 1695 Eastchester Road, 2nd Floor, Bronx, NY, 10461, USA
| | - Amara G Nandikolla
- Department of Medical Oncology, Montefiore Medical Center, 1695 Eastchester Road, 2nd Floor, Bronx, NY, 10461, USA
| | - Lakshmi Rajdev
- Department of Medical Oncology, Montefiore Medical Center, 1695 Eastchester Road, 2nd Floor, Bronx, NY, 10461, USA.
| |
Collapse
|
52
|
Yin P, Liu X, Mansfield AS, Harrington SM, Li Y, Yan Y, Dong H. CpG-induced antitumor immunity requires IL-12 in expansion of effector cells and down-regulation of PD-1. Oncotarget 2018; 7:70223-70231. [PMID: 27602959 PMCID: PMC5342548 DOI: 10.18632/oncotarget.11833] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 08/24/2016] [Indexed: 12/29/2022] Open
Abstract
CpG oligodeoxynucleotides, as a ligand of toll-like receptor (TLR)-9, have demonstrated promising antitumor effects in some clinical trials; however, its toxicity and low efficacy as a systemic therapy has limited its therapeutic applications. In order to improve its therapeutic efficacy, we investigated the mechanisms of CpG-induced antitumor immunity in the context of CD8+ T cell responses. We show that IL-12 is required for the expansion of IFN-γ producing tumor-reactive CD8+ T cells capable of rejecting tumors. In addition, CpGs reduced PD-1 expression by effector CD8+ T cells via the IL-12 pathway. The combination of CpG and PD-1 blockade show a synergistic effect in generation of systemic antitumor immunity. Our studies define a critical role of IL-12 in CpG-induced antitumor immunity and provide a rationale for combined therapy with TLR agonists and immune checkpoint blockade in cancer treatment.
Collapse
Affiliation(s)
- Peng Yin
- Department of Urology, Mayo Clinic, Rochester, MN, USA
| | - Xin Liu
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Yinghua Li
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Yiyi Yan
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Haidong Dong
- Department of Urology, Mayo Clinic, Rochester, MN, USA.,Department of Immunology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
53
|
Wang Y, Zhang L, Xu Z, Miao L, Huang L. mRNA Vaccine with Antigen-Specific Checkpoint Blockade Induces an Enhanced Immune Response against Established Melanoma. Mol Ther 2018; 26:420-434. [PMID: 29249397 PMCID: PMC5835019 DOI: 10.1016/j.ymthe.2017.11.009] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/14/2017] [Accepted: 11/15/2017] [Indexed: 12/25/2022] Open
Abstract
We reported a preclinical cancer vaccine that simultaneously introduced an mRNA antigen and an immune checkpoint blocking siRNA into the antigen-presenting cells. This was achieved by formulating both nucleic acid-based immunotherapeutics into a lipid-coated calcium phosphate (LCP) nanoparticle (NP) as a carrier to address the delivery challenge. The PEGylated lipid NPs were functionalized with mannose as the targeting ligand to facilitate the preferential uptake by the dendritic cells (DCs) in the lymph nodes after subcutaneous administration. The calcium phosphate core allowed acid-mediated dissolution in the endo-lysosomal compartment, which prompted rapid release of cargoes after cellular internalization of NP. LCP mRNA vaccine encoding TRP2 elicited a robust antigen-specific cytotoxic T cell response and a humoral immune response in a C57BL/6 mouse model of B16F10 melanoma. The immune responses efficaciously inhibited the melanoma growth. Moreover, co-delivery of PD-L1 siRNA and mRNA vaccine resulted in the downregulation of PD-L1 in the DCs that presented tumor antigens, significantly prompting T cell activation and proliferation. The enhanced T cell response had a profound inhibitory effect on tumor growth and metastasis. Generally, the work provided a paradigm for the development of an mRNA vaccine carrier to boost the anticancer immune response.
Collapse
Affiliation(s)
- Yuhua Wang
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lu Zhang
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zhenghong Xu
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lei Miao
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
54
|
Cubillos-Zapata C, Avendaño-Ortiz J, Hernandez-Jimenez E, Toledano V, Casas-Martin J, Varela-Serrano A, Torres M, Almendros I, Casitas R, Fernández-Navarro I, Garcia-Sanchez A, Aguirre LA, Farre R, López-Collazo E, García-Rio F. Hypoxia-induced PD-L1/PD-1 crosstalk impairs T-cell function in sleep apnoea. Eur Respir J 2017; 50:50/4/1700833. [PMID: 29051270 DOI: 10.1183/13993003.00833-2017] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/23/2017] [Indexed: 12/16/2022]
Abstract
Obstructive sleep apnoea (OSA) is associated with higher cancer incidence, tumour aggressiveness and cancer mortality, as well as greater severity of infections, which have been attributed to an immune deregulation. We studied the expression of programmed cell death (PD)-1 receptor and its ligand (PD-L1) on immune cells from patients with OSA, and its consequences on immune-suppressing activity. We report that PD-L1 was overexpressed on monocytes and PD-1 was overexpressed on CD8+ T-cells in a severity-dependent manner. PD-L1 and PD-1 overexpression were induced in both the human in vitro and murine models of intermittent hypoxia, as well as by hypoxia-inducible factor-1α transfection. PD-L1/PD-1 crosstalk suppressed T-cell proliferation and activation of autologous T-lymphocytes and impaired the cytotoxic activity of CD8+ T-cells. In addition, monocytes from patients with OSA exhibited high levels of retinoic acid related orphan receptor, which might explain the differentiation of myeloid-derived suppressor cells. Intermittent hypoxia upregulated the PD-L1/PD-1 crosstalk in patients with OSA, resulting in a reduction in CD8+ T-cell activation and cytotoxicity, providing biological plausibility to the increased incidence and aggressiveness of cancer and the higher risk of infections described in these patients.
Collapse
Affiliation(s)
- Carolina Cubillos-Zapata
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain.,Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Jose Avendaño-Ortiz
- The Innate Immune Response Group, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Enrique Hernandez-Jimenez
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain.,The Innate Immune Response Group, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Victor Toledano
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain.,The Innate Immune Response Group, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Jose Casas-Martin
- The Innate Immune Response Group, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | | | - Marta Torres
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain.,Sleep Laboratory, Pneumology Service, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Isaac Almendros
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain.,Biophysics and Bioengineering Unit, Faculty of Medicine, University of Barcelona-IDIBAPS, Barcelona, Spain
| | - Raquel Casitas
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain.,Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Isabel Fernández-Navarro
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Aldara Garcia-Sanchez
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Luis A Aguirre
- The Innate Immune Response Group, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Ramón Farre
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain.,Biophysics and Bioengineering Unit, Faculty of Medicine, University of Barcelona-IDIBAPS, Barcelona, Spain
| | - Eduardo López-Collazo
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain.,The Innate Immune Response Group, La Paz University Hospital, IdiPAZ, Madrid, Spain.,Joint principal investigators
| | - Francisco García-Rio
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain .,Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain.,Dept of Medicine, Autonomous University of Madrid, Madrid, Spain.,Joint principal investigators
| |
Collapse
|
55
|
CD80 Expressed by CD8 + T Cells Contributes to PD-L1-Induced Apoptosis of Activated CD8 + T Cells. J Immunol Res 2017; 2017:7659462. [PMID: 29181416 PMCID: PMC5664331 DOI: 10.1155/2017/7659462] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/13/2017] [Accepted: 09/24/2017] [Indexed: 12/24/2022] Open
Abstract
Tumor cells are capable of limiting antitumor CD8+ T cell responses through their cell surface expression of PD-L1. In addition to PD-1 expressed by CD8+ T cells, PD-L1 also binds to CD80 expressed by CD8+ T cells. The influence of the PD-L1/CD80 interaction on CD8+ T cell function has not been fully characterized, so we sought to investigate the impact of the PD-L1/CD80 interaction on PD-L1-induced apoptosis of activated CD8+ T cells. We found that CD8+ T cells that lacked CD80 expression got activated to the same extent as wild-type CD8+ T cells, but when cultured with anti-CD3 and PD-L1/Fc protein, activated CD8+ T cells that lacked CD80 expression survived better than activated wild-type CD8+ T cells. These findings indicate that PD-L1 induces apoptosis in activated CD8+ T cells in part by signaling through CD80. Thus, in the design and implementation of checkpoint blockade therapies that target PD-L1, it is essential that both binding partners for PD-L1, PD-1, and CD80 are considered.
Collapse
|
56
|
Téglási V, Reiniger L, Fábián K, Pipek O, Csala I, Bagó AG, Várallyai P, Vízkeleti L, Rojkó L, Tímár J, Döme B, Szállási Z, Swanton C, Moldvay J. Evaluating the significance of density, localization, and PD-1/PD-L1 immunopositivity of mononuclear cells in the clinical course of lung adenocarcinoma patients with brain metastasis. Neuro Oncol 2017; 19:1058-1067. [PMID: 28201746 PMCID: PMC5570158 DOI: 10.1093/neuonc/now309] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Management of lung cancer patients who suffer from brain metastases represents a major challenge. Considering the promising results with immune checkpoint inhibitor treatment, evaluating the status of immune cell (IC) infiltrates in the prognosis of brain metastasis may lead to better therapeutic strategies with these agents. The aim of this study was to characterize the distribution of ICs and determine the expression of the checkpoint molecules programmed death protein 1 (PD-1) and its ligand, PD-L1, in brain metastasis of lung adenocarcinoma (LUAD) patients and to analyze their clinicopathological correlations. METHODS We determined the presence of peritumoral mononuclear cells (mononuclear ring) and the density of intratumoral stromal mononuclear cells on brain metastasis tissue sections of 208 LUAD patients. PD-L1/PD-1 expressions were analyzed by immunohistochemistry. RESULTS Mononuclear rings were significantly associated with better survival after brain metastasis surgery. Cases with massive stromal IC infiltration also showed a tendency for better overall survival. Lower expression of PD-1 and PD-L1 was associated with better survival in patients who underwent surgery for the primary tumor and had multiple brain metastases. Steroid administration and chemotherapy appear not to influence the density of IC in brain metastasis. CONCLUSION This is the first study demonstrating the independent prognostic value of mononuclear rings in LUAD cases with brain metastasis. Our results also suggest that the density of tumor-associated ICs in addition to PD-L1 expression of tumor cells and ICs as well as PD-1 expression of ICs may hold relevant information for the appropriate selection of patients who might benefit from anti-PD-L1 or anti-PD-1 therapy.
Collapse
Affiliation(s)
- Vanda Téglási
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Lilla Reiniger
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Katalin Fábián
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Orsolya Pipek
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Irén Csala
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Attila G Bagó
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Péter Várallyai
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Laura Vízkeleti
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Lívia Rojkó
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - József Tímár
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Balázs Döme
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Zoltán Szállási
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Charles Swanton
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Judit Moldvay
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| |
Collapse
|
57
|
Gong Y, Zhang X, Chen R, Wei Y, Zou Z, Chen X. Cytoplasmic expression of C-MYC protein is associated with risk stratification of mantle cell lymphoma. PeerJ 2017. [PMID: 28626618 PMCID: PMC5472035 DOI: 10.7717/peerj.3457] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aim To investigate the association of C-MYC protein expression and risk stratification in mantle cell lymphoma (MCL), and to evaluate the utility of C-MYC protein as a prognostic biomarker in clinical practice. Methods We conducted immunohistochemical staining of C-MYC, Programmed cell death ligand 1 (PD-L1), CD8, Ki-67, p53 and SRY (sex determining region Y) -11 (SOX11) to investigate their expression in 64 patients with MCL. The staining results and other clinical data were evaluated for their roles in risk stratification of MCL cases using ANOVA, Chi-square, and Spearman’s Rank correlation coefficient analysis. Results Immunohistochemical staining in our study indicated that SOX11, Ki-67 and p53 presented nuclear positivity of tumor cells, CD8 showed membrane positivity in infiltrating T lymphocytes while PD-L1 showed membrane and cytoplasmic positivity mainly in macrophage cells and little in tumor cells. We observed positive staining of C-MYC either in the nucleus or cytoplasm or in both subcellular locations. There were significant differences in cytoplasmic C-MYC expression, Ki-67 proliferative index of tumor cells, and CD8 positive tumor infiltrating lymphocytes (CD8+TIL) among three risk groups (P = 0.000, P = 0.037 and P=0.020, respectively). However, no significant differences existed in the expression of nuclear C-MYC, SOX11, p53, and PD-L1 in MCL patients with low-, intermediate-, and high risks. In addition, patient age and serum LDH level were also significantly different among 3 groups of patients (P = 0.006 and P = 0.000, respectively). Spearman’s rank correlation coefficient analysis indicated that cytoplasmic C-MYC expression, Ki-67 index, age, WBC, as well as LDH level had significantly positive correlations with risk stratification (P = 0.000, 0.015, 0.000, 0.029 and 0.000, respectively), while CD8+TIL in tumor microenvironment negatively correlated with risk stratification of patients (P = 0.006). Patients with increased positive cytoplasmic expression of C-MYC protein and decreased CD8+TIL appeared to be associated with a poor response to chemotherapy, but the correlation was not statistically significant. Conclusion Our study suggested that assessment of cytoplasmic C-MYC overexpression and cytotoxic T lymphocytes (CTLs) by immunohistochemical staining might be helpful for MCL risk stratification and outcome prediction. However, large cohort studies of MCL patients with complete follow up are needed to validate our speculation.
Collapse
Affiliation(s)
- Yi Gong
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China.,Department of Hematology-oncology, Chongqing Cancer Institute/Hospital, Chongqing, China
| | - Xi Zhang
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Rui Chen
- Department of Pathology, Chongqing Cancer Institute/Hospital, Chongqing, China
| | - Yan Wei
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Zhongmin Zou
- Institute of Toxicology, School of Preventive Medicine, The Third Military Medical University, Chongqing, China
| | - Xinghua Chen
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| |
Collapse
|
58
|
Cellular and molecular targets for the immunotherapy of hepatocellular carcinoma. Mol Cell Biochem 2017; 437:13-36. [PMID: 28593566 DOI: 10.1007/s11010-017-3092-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/01/2017] [Indexed: 02/06/2023]
|
59
|
Nanovaccines for remodeling the suppressive tumor microenvironment: New horizons in cancer immunotherapy. Front Chem Sci Eng 2017. [DOI: 10.1007/s11705-017-1640-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
60
|
Balar AV, Weber JS. PD-1 and PD-L1 antibodies in cancer: current status and future directions. Cancer Immunol Immunother 2017; 66:551-564. [PMID: 28213726 PMCID: PMC11028560 DOI: 10.1007/s00262-017-1954-6] [Citation(s) in RCA: 233] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/06/2017] [Indexed: 12/20/2022]
Abstract
Immunotherapy has moved to the center stage of cancer treatment with the recent success of trials in solid tumors with PD-1/PD-L1 axis blockade. Programmed death-1 or PD-1 is a checkpoint molecule on T cells that plays a vital role in limiting adaptive immune responses and preventing autoimmune and auto-inflammatory reactivity in the normal host. In cancer patients, PD-1 expression is very high on T cells in the tumor microenvironment, and PD-L1, its primary ligand, is variably expressed on tumor cells and antigen-presenting cells within tumors, providing a potent inhibitory influence within the tumor microenvironment. While PD-L1 expression on tumors is often regarded as a negative prognostic factor, it is clearly associated with a positive outcome for treatment with PD-1/PD-L1 blocking antibodies, and has been used to select patients for this therapy. Responses of long duration, a minority of patients with atypical responses in which progression may precede tumor shrinkage, and a pattern of autoimmune side effects often seen with this class of drugs characterize therapy with PD-1/PD-L1 blocking drugs. While excellent efficacy has been seen with a limited number of tumor types, most epithelial cancers do not show responses of long duration with these agents. In the current review, we will briefly summarize the scientific background data supporting the development of PD-1/PD-L1 blockade, and then describe the track record of these antibodies in multiple different histologies ranging from melanoma and lung cancer to less common tumor types as well as discuss biomarkers that may assist in patient selection.
Collapse
Affiliation(s)
- Arjun Vasant Balar
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, 522 First Avenue, 1310 Smilow Research Building, New York, NY, 10016, USA
| | - Jeffrey S Weber
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, 522 First Avenue, 1310 Smilow Research Building, New York, NY, 10016, USA.
| |
Collapse
|
61
|
Burova E, Hermann A, Waite J, Potocky T, Lai V, Hong S, Liu M, Allbritton O, Woodruff A, Wu Q, D’Orvilliers A, Garnova E, Rafique A, Poueymirou W, Martin J, Huang T, Skokos D, Kantrowitz J, Popke J, Mohrs M, MacDonald D, Ioffe E, Olson W, Lowy I, Murphy A, Thurston G. Characterization of the Anti–PD-1 Antibody REGN2810 and Its Antitumor Activity in Human PD-1 Knock-In Mice. Mol Cancer Ther 2017; 16:861-870. [DOI: 10.1158/1535-7163.mct-16-0665] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 12/05/2016] [Accepted: 01/30/2017] [Indexed: 11/16/2022]
|
62
|
Li D, Xu J, Wang Z, Gong Z, Liu J, Zheng Y, Li J, Li J. Epitope mapping reveals the binding mechanism of a functional antibody cross-reactive to both human and murine programmed death 1. MAbs 2017; 9:628-637. [PMID: 28300475 PMCID: PMC5419084 DOI: 10.1080/19420862.2017.1296612] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/12/2017] [Accepted: 02/14/2017] [Indexed: 10/20/2022] Open
Abstract
Of the inhibitory checkpoints in the immune system, programmed death 1 (PD-1) is one of the most promising targets for cancer immunotherapy. The anti-PD-1 antibodies currently approved for clinical use or under development bind to human PD-1 (hPD-1), but not murine PD-1. To facilitate studies in murine models, we developed a functional antibody against both human and murine PD-1, and compared the epitopes of such antibody to a counterpart that only bound to hPD-1. To quickly identify the epitopes of the 2 antibodies, we used alanine scanning and mammalian cell expression cassette. The epitope identification was based on PD-1-binding ELISA and supported by affinity ranking of surface plasmon resonance results. The hPD-1 epitopes of the 2 functional antibodies were also compared with the binding region on hPD-1 that is responsible for PD-L1 interaction. In silico modeling were conducted to explain the different binding modes of the 2 antibodies, suggesting a potential mechanism of the antibody cross-species binding.
Collapse
Affiliation(s)
- Dong Li
- Biologics Discovery, WuXi Biologics, Waigaoqiao Free Trade Zone, Shanghai, China
| | - Jianqing Xu
- Biologics Discovery, WuXi Biologics, Waigaoqiao Free Trade Zone, Shanghai, China
| | - Zhuozhi Wang
- Biologics Discovery, WuXi Biologics, Waigaoqiao Free Trade Zone, Shanghai, China
| | - Zhen Gong
- State Key Laboratory of Lead Compound Research, WuXi AppTec, Waigaoqiao Free Trade Zone, Shanghai, China
| | - Jieying Liu
- Biologics Discovery, WuXi Biologics, Waigaoqiao Free Trade Zone, Shanghai, China
| | - Yong Zheng
- Biologics Discovery, WuXi Biologics, Waigaoqiao Free Trade Zone, Shanghai, China
| | - Jian Li
- State Key Laboratory of Lead Compound Research, WuXi AppTec, Waigaoqiao Free Trade Zone, Shanghai, China
| | - Jing Li
- Biologics Discovery, WuXi Biologics, Waigaoqiao Free Trade Zone, Shanghai, China
| |
Collapse
|
63
|
de Groot AE, Roy S, Brown JS, Pienta KJ, Amend SR. Revisiting Seed and Soil: Examining the Primary Tumor and Cancer Cell Foraging in Metastasis. Mol Cancer Res 2017; 15:361-370. [PMID: 28209759 DOI: 10.1158/1541-7786.mcr-16-0436] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/18/2017] [Accepted: 02/03/2017] [Indexed: 12/24/2022]
Abstract
Metastasis is the consequence of a cancer cell that disperses from the primary tumor, travels throughout the body, and invades and colonizes a distant site. On the basis of Paget's 1889 hypothesis, the majority of modern metastasis research focuses on the properties of the metastatic "seed and soil," but the implications of the primary tumor "soil" have been largely neglected. The rare lethal metastatic "seed" arises as a result of the selective pressures in the primary tumor. Optimal foraging theory describes how cancer cells adopt a mobile foraging strategy to balance predation risk and resource reward. Further selection in the dispersal corridors leading out of the primary tumor enhances the adaptive profile of the potentially metastatic cell. This review focuses on the selective pressures of the primary tumor "soil" that generate lethal metastatic "seeds" which is essential to understanding this critical component of prostate cancer metastasis.Implication: Elucidating the selective pressures of the primary tumor "soil" that generate lethal metastatic "seeds" is essential to understand how and why metastasis occurs in prostate cancer. Mol Cancer Res; 15(4); 361-70. ©2017 AACR.
Collapse
Affiliation(s)
- Amber E de Groot
- The James Buchanan Brady Urological Institute at the Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sounak Roy
- The James Buchanan Brady Urological Institute at the Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joel S Brown
- Department of Biological Sciences and UIC Cancer Center, University of Illinois at Chicago, Chicago, Illinois.,Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Kenneth J Pienta
- The James Buchanan Brady Urological Institute at the Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sarah R Amend
- The James Buchanan Brady Urological Institute at the Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
64
|
Xia H, Shen J, Hu F, Chen S, Huang H, Xu Y, Ma H. PD-L1 over-expression is associated with a poor prognosis in Asian non-small cell lung cancer patients. Clin Chim Acta 2017; 469:191-194. [PMID: 28188721 DOI: 10.1016/j.cca.2017.02.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 02/05/2017] [Accepted: 02/06/2017] [Indexed: 01/12/2023]
Abstract
BACKGROUND The prognostic role of programmed cell death-ligand 1 (PD-L1) in non-small cell lung cancer (NSCLC) remains controversial. This meta-analysis was conducted to clarify the association of PD-L1 with survival in NSCLC patients. METHODS Relevant studies were collected from PubMed, Embase and Web of Science. Only studies in which PD-L1 expression was detected by immunohistochemical staining in NSCLC patients correlated with patient survival data were included. Stata 12.0 was performed in this meta-analysis. RESULTS Fifteen studies with 3116 patients were included in this meta-analysis. The combined hazard ratio (HR) of 1.18 (95%CI, 0.90-1.56; P>0.05) shows that PD-L1 over-expression in NSCLC patients didn't associate with overall survival (OS). However, subgroup analysis indicated that PD-L1 over-expression was correlated with poor OS in Asian patients (HR=1.84, 95%CI: 1.14-2.28; P<0.001). Moreover, the results suggested that a significant relationship between PD-L1 expression and OS was also showed in studies with late stage (HR=1.27, 95%CI: 1.06-1.48; P=0.031), poor tumor differentiation (HR=1.86, 95%CI: 1.35-2.64; P=0.001) and anaplastic lymphoma kinase (ALK) translocation (HR=2.58, 95%CI: 1.03-5.78; P=0.028). CONCLUSIONS PD-L1 over-expression is correlated with a poor prognosis in Asian NSCLC patients.
Collapse
Affiliation(s)
- Haifeng Xia
- Department of Cardiothoracic Surgery, Suzhou Dushuhu Public Hospital (Soochow University Multi-Disciplinary Polyclinic), Suzhou 215006, Jiangsu Province, People's Republic of China
| | - Ji Shen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, People's Republic of China; Department of Orthopedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200000, People's Republic of China
| | - Fang Hu
- Department of Hematology, Suzhou Dushuhu Public Hospital (Soochow University Multi-Disciplinary Polyclinic), Suzhou 215006, Jiangsu Province, People's Republic of China
| | - Shaomu Chen
- Department of thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, People's Republic of China
| | - Haitao Huang
- Department of thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, People's Republic of China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, People's Republic of China.
| | - Haitao Ma
- Department of thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, People's Republic of China
| |
Collapse
|
65
|
Chajon E, Castelli J, Marsiglia H, De Crevoisier R. The synergistic effect of radiotherapy and immunotherapy: A promising but not simple partnership. Crit Rev Oncol Hematol 2017; 111:124-132. [PMID: 28259287 DOI: 10.1016/j.critrevonc.2017.01.017] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/21/2016] [Accepted: 01/25/2017] [Indexed: 12/20/2022] Open
Abstract
Radiotherapy (RT) is one of the main components in the treatment of cancer. The better understanding of the immune mechanisms associated with tumor establishment and how RT affects inflammation and immunity has led to the development of novel treatment strategies. Several preclinical studies support the use of RT in combination with immunotherapy obtaining better local and systemic tumor control. Current ongoing studies will provide information about the optimal RT approach, but the development of reliable predictors of the response from the preclinical and the early phases of clinical studies is necessary to avoid discarding treatment strategies with significant clinical benefit. This review summarize the current concepts of the synergism between RT and immunotherapy, the molecular effects of RT in the tumor microenvironment, their impact on immune activation and its potential clinical applications in trials exploring this important therapeutic opportunity. Finally, the potential predictors of clinical response are discussed.
Collapse
Affiliation(s)
- Enrique Chajon
- Department of Radiation Oncology, Centre Eugene Marquis, Rennes, F-35000, France.
| | - Joël Castelli
- Department of Radiation Oncology, Centre Eugene Marquis, Rennes, F-35000, France; Université de Rennes 1, LTSI, INSERM, Rennes U1099, France
| | - Hugo Marsiglia
- Department of Radiation Oncology, Instituto Oncologico Fundacion Arturo Lopez Perez, Santiago de Chile, 7500921, Chile
| | - Renaud De Crevoisier
- Department of Radiation Oncology, Centre Eugene Marquis, Rennes, F-35000, France; Université de Rennes 1, LTSI, INSERM, Rennes U1099, France
| |
Collapse
|
66
|
Lin CF, Lin CM, Lee KY, Wu SY, Feng PH, Chen KY, Chuang HC, Chen CL, Wang YC, Tseng PC, Tsai TT. Escape from IFN-γ-dependent immunosurveillance in tumorigenesis. J Biomed Sci 2017; 24:10. [PMID: 28143527 PMCID: PMC5286687 DOI: 10.1186/s12929-017-0317-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/19/2017] [Indexed: 12/24/2022] Open
Abstract
Immune interferon (IFN), also known as IFN-γ, promotes not only immunomodulation but also antimicrobial and anticancer activity. After IFN-γ binds to the complex of IFN-γ receptor (IFNGR) 1-IFNGR2 and subsequently activates its downstream signaling pathways, IFN-γ immediately causes transcriptional stimulation of a variety of genes that are principally involved in its biological activities. Regarding IFN-γ-dependent immunosurveillance, IFN-γ can directly suppress tumorigenesis and infection and/or can modulate the immunological status in both cancer cells and infected cells. Regarding the anticancer effects of IFN-γ, cancer cells develop strategies to escape from IFN-γ-dependent cancer immunosurveillance. Immune evasion, including the recruitment of immunosuppressive cells, secretion of immunosuppressive factors, and suppression of cytotoxic T lymphocyte responses, is speculated to be elicited by the oncogenic microenvironment. All of these events effectively downregulate IFN-γ-expressing cells and IFN-γ production. In addition to these extrinsic pathways, cancer cells may develop cellular tolerance that manifests as hyporesponsiveness to IFN-γ stimulation. This review discusses the potential escape mechanisms from IFN-γ-dependent immunosurveillance in tumorigenesis.
Collapse
Affiliation(s)
- Chiou-Feng Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan. .,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.
| | - Chih-Ming Lin
- Department of Thoracic Surgery, Lotung Poh-Ai Hospital, Yilan, 265, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, 110, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Szu-Yuan Wu
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.,Department of Radiation Oncology, Wan Fang Hospital, Taipei Medical University, Taipei Medical University, Taipei, 110, Taiwan.,Department of Biotechnology, Hung Kuang University, Taichung, 433, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, 110, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, 110, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Chia-Ling Chen
- Translational Research Center, Taipei Medical University, Taipei, 110, Taiwan
| | - Yu-Chih Wang
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Po-Chun Tseng
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Tsung-Ting Tsai
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| |
Collapse
|
67
|
Kumar SR, Kim DY, Henry CJ, Bryan JN, Robinson KL, Eaton AM. Programmed death ligand 1 is expressed in canine B cell lymphoma and downregulated by MEK inhibitors. Vet Comp Oncol 2017; 15:1527-1536. [PMID: 28111882 DOI: 10.1111/vco.12297] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 11/03/2016] [Accepted: 11/11/2016] [Indexed: 12/21/2022]
Abstract
Programmed death ligand 1 (PD-L1) expression in antigen-presenting cells and tumors can inhibit T cell-mediated immunity. In this study, PD-L1 mRNA and protein expression was evaluated in canine B cell lymphoma (CLL17-71), large T-cell leukemia (CLGL-90), B cell leukemia (GL-1) and primitive leukocyte round cell neoplasia (CLL-1390). Variable PD-L1 mRNA and protein were observed in these cells with high endogenous expression present in CLL17-71 cells. PD-L1 protein was also observed in canine patient B cell lymphoma tissues using immunostaining. PD-L1 and signal transducer and activator of transcription 1 ( STAT1 ) mRNA expression were reduced in the presence of mitogen-activated protein kinase kinase 1.2 (MEK1/2) inhibitors RDEA119 and AZD6244 in CLL 17-71 cells. RDEA119 had similar effect on PD-L1 and STAT-1 in IFN-γ activated CLL-1390 cells. Overall, these results indicate that PD-L1 is expressed in canine B cell lymphoma. Its inhibition by MEK1/2 inhibitors suggests a possible treatment strategy using targeted drugs which likely could enhance antitumor immune response.
Collapse
Affiliation(s)
- S R Kumar
- Comparative Oncology, Radiopharmaceutical and Epigenetics Laboratory, School of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - D Y Kim
- Veterinary Pathobiology, School of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - C J Henry
- Comparative Oncology, Radiopharmaceutical and Epigenetics Laboratory, School of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - J N Bryan
- Comparative Oncology, Radiopharmaceutical and Epigenetics Laboratory, School of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - K L Robinson
- Comparative Oncology, Radiopharmaceutical and Epigenetics Laboratory, School of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - A M Eaton
- Comparative Oncology, Radiopharmaceutical and Epigenetics Laboratory, School of Veterinary Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
68
|
Berzaghi R, Maia VSC, Pereira FV, Melo FM, Guedes MS, Origassa CST, Scutti JB, Matsuo AL, Câmara NOS, Rodrigues EG, Travassos LR. SOCS1 favors the epithelial-mesenchymal transition in melanoma, promotes tumor progression and prevents antitumor immunity by PD-L1 expression. Sci Rep 2017; 7:40585. [PMID: 28079159 PMCID: PMC5227698 DOI: 10.1038/srep40585] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/02/2016] [Indexed: 01/05/2023] Open
Abstract
Silencing of SOCS1 protein with shRNAi lentivirus (shR-SOCS1) led to partial reversion of the tumorigenic phenotype of B16F10-Nex2 melanoma cells. SOCS1 silencing inhibited cell migration and invasion as well as in vitro growth by cell cycle arrest at S phase with increased cell size and nuclei. Down-regulation of SOCS1 decreased the expression of epidermal growth factor receptor, Ins-Rα, and fibroblast growth factor receptors. The present work aimed at analyzing the SOCS1 cell signaling and expression of proteins relevant to tumor development. An RNA microarray analysis of B16F10-Nex2 melanoma cells with SOCS1 silenced by shRNAi-SOCS1 was undertaken in comparison with cells transduced with the empty vector. Among 609 differentially expressed genes, c-Kit, Met and EphA3 cytokine/tyrosine-kinase (TK) receptors were down regulated. A significant decrease in the expression of TK receptors, the phosphorylation of mediators of ERK1/2 and p38 pathways and STAT3 (S727) were observed. Subcutaneous immunization with shR-SOCS1-transduced viable tumor cells rendered protection against melanoma in a syngeneic model, with decreased expression of PD-L1 and of matrix metallo-proteinases (MMPs) and CD-10 in those cells. The present work shows the role of SOCS1 in murine melanoma development and the potential of SOCS1-silenced tumor cells in raising an effective anti-melanoma immune response.
Collapse
Affiliation(s)
- R. Berzaghi
- Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, University of São Paulo, São Paulo, Brazil
| | | | - F. V. Pereira
- Laboratory of Cancer Immunobiology, University of São Paulo, São Paulo, Brazil
| | - F. M. Melo
- Immunology Department, Federal University of São Paulo, São Paulo, Brazil
| | - M. S. Guedes
- Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, University of São Paulo, São Paulo, Brazil
| | - C. S. T. Origassa
- Laboratory of Cancer Immunobiology, University of São Paulo, São Paulo, Brazil
| | - J. B. Scutti
- Immunotherapy Platform, Department of Immunology, MD Anderson Cancer Center, Houston Texas, USA
| | - A. L. Matsuo
- Interdepartmental Group of Health Economics (Grides), Federal University of São Paulo, SP, Brazil
| | - N. O. S. Câmara
- Immunology Department, Biomedical Sciences Institute IV, University of São Paulo, São Paulo, Brazil
| | - E. G. Rodrigues
- Laboratory of Cancer Immunobiology, University of São Paulo, São Paulo, Brazil
| | - L. R. Travassos
- Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, University of São Paulo, São Paulo, Brazil
- Recepta Biopharma São Paulo, Brazil
| |
Collapse
|
69
|
Dong H, Yan Y, Dronca RS, Markovic SN. A T cell equation as a conceptual model of T cell responses for maximizing the efficacy of cancer immunotherapy. ACTA ACUST UNITED AC 2017; 5:1-5. [PMID: 29629436 DOI: 10.15226/2372-0948/5/1/00155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Following antigen stimulation, the net outcomes of a T cell response are shaped by integrated signals from both positive co-stimulatory and negative regulatory molecules. Recently, the blockade of negative regulatory molecules (i.e. immune checkpoint signals) demonstrates promising therapeutic effects in treatment of human cancers, but only in a fraction of cancer patients. Since this therapy is aimed to enhance T cell responses to cancers, here we devised a conceptual model by integrating both positive and negative signals in addition to antigen stimulation that can evaluate strategies to enhance T cell responses. A digital range of adjustment of each signal is formulated in our model for prediction of a final T cell response. Our model provides a rational combination strategy for maximizing the therapeutic effects of cancer immunotherapy.
Collapse
Affiliation(s)
- Haidong Dong
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.,Department of Urology, Mayo Clinic, Rochester, MN, USA
| | - Yiyi Yan
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA.,Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Roxana S Dronca
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Svetomir N Markovic
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.,Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
70
|
Van Der Kraak L, Goel G, Ramanan K, Kaltenmeier C, Zhang L, Normolle DP, Freeman GJ, Tang D, Nason KS, Davison JM, Luketich JD, Dhupar R, Lotze MT. 5-Fluorouracil upregulates cell surface B7-H1 (PD-L1) expression in gastrointestinal cancers. J Immunother Cancer 2016; 4:65. [PMID: 27777774 PMCID: PMC5067917 DOI: 10.1186/s40425-016-0163-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 09/13/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Resistance to chemotherapy is a major obstacle in the effective treatment of cancer patients. B7-homolog 1, also known as programmed death ligand-1 (PD-L1), is an immunoregulatory protein that is overexpressed in several human cancers. Interaction of B7-H1 with programmed death 1 (PD-1) prevents T-cell activation and proliferation, sequestering the T-cell receptor from the cell membrane, inducing T-cell apoptosis, thereby leading to cancer immunoresistance. B7-H1 upregulation contributes to chemoresistance in several types of cancer, but little is known with respect to changes associated with 5-fluorouracil (5-FU) or gastrointestinal cancers. METHODS HCT 116 p53+/+, HCT 116 p53-/- colorectal cancer (CRC) and OE33 esophageal adenocarcinoma (EAC) cells were treated with increasing doses of 5-FU (0.5 uM, 5 uM, 50 uM, 500 uM) or interferon gamma (IFN-γ, 10 ng/mL) in culture for 24 h and B7-H1 expression was quantified using flow cytometry and western blot analysis. We also evaluated B7-H1 expression, by immunohistochemistry, in tissue collected prior to and following neoadjuvant therapy in 10 EAC patients. RESULTS B7-H1 expression in human HCT 116 p53+/+ and HCT 116 p53-/- CRC cells lines, while low at baseline, can be induced by treatment with 5-FU. OE33 baseline B7-H1 expression exceeded CRC cell maximal expression and could be further increased in a dose dependent manner following 5-FU treatment in the absence of immune cells. We further demonstrate tumor B7-H1 expression in esophageal adenocarcinoma patient-derived pre-treatment biopsies. While B7-H1 expression was not enhanced in post-treatment esophagectomy specimens, this may be due to the limits of immunohistochemical quantification. CONCLUSIONS B7-H1/PD-L1 expression can be increased following treatment with 5-FU in gastrointestinal cancer cell lines, suggesting alternative mechanisms to classic immune-mediated upregulation. This suggests that combining 5-FU treatment with PD-1/B7-H1 blockade may improve treatment in patients with gastrointestinal adenocarcinoma.
Collapse
Affiliation(s)
- Lauren Van Der Kraak
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA USA
| | - Gaurav Goel
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh, Pittsburgh, PA USA
- Current address: Division of Medical Oncology, University of Kentucky Markey Cancer Center, Lexington, KY USA
| | | | | | - Lin Zhang
- Department of Pharmacology & Chemical Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Daniel P. Normolle
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA USA
| | - Gordon J. Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA USA
| | - Daolin Tang
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA USA
| | - Katie S. Nason
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA USA
| | - Jon M. Davison
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA USA
| | - James D. Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA USA
| | - Rajeev Dhupar
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA USA
| | - Michael T. Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA USA
| |
Collapse
|
71
|
Lanzel EA, Paula Gomez Hernandez M, Bates AM, Treinen CN, Starman EE, Fischer CL, Parashar D, Guthmiller JM, Johnson GK, Abbasi T, Vali S, Brogden KA. Predicting PD-L1 expression on human cancer cells using next-generation sequencing information in computational simulation models. Cancer Immunol Immunother 2016; 65:1511-1522. [PMID: 27688163 DOI: 10.1007/s00262-016-1907-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 09/23/2016] [Indexed: 12/31/2022]
Abstract
PURPOSE Interaction of the programmed death-1 (PD-1) co-receptor on T cells with the programmed death-ligand 1 (PD-L1) on tumor cells can lead to immunosuppression, a key event in the pathogenesis of many tumors. Thus, determining the amount of PD-L1 in tumors by immunohistochemistry (IHC) is important as both a diagnostic aid and a clinical predictor of immunotherapy treatment success. Because IHC reactivity can vary, we developed computational simulation models to accurately predict PD-L1 expression as a complementary assay to affirm IHC reactivity. METHODS Multiple myeloma (MM) and oral squamous cell carcinoma (SCC) cell lines were modeled as examples of our approach. Non-transformed cell models were first simulated to establish non-tumorigenic control baselines. Cell line genomic aberration profiles, from next-generation sequencing (NGS) information for MM.1S, U266B1, SCC4, SCC15, and SCC25 cell lines, were introduced into the workflow to create cancer cell line-specific simulation models. Percentage changes of PD-L1 expression with respect to control baselines were determined and verified against observed PD-L1 expression by ELISA, IHC, and flow cytometry on the same cells grown in culture. RESULT The observed PD-L1 expression matched the predicted PD-L1 expression for MM.1S, U266B1, SCC4, SCC15, and SCC25 cell lines and clearly demonstrated that cell genomics play an integral role by influencing cell signaling and downstream effects on PD-L1 expression. CONCLUSION This concept can easily be extended to cancer patient cells where an accurate method to predict PD-L1 expression would affirm IHC results and improve its potential as a biomarker and a clinical predictor of treatment success.
Collapse
Affiliation(s)
- Emily A Lanzel
- Department of Oral Pathology, Radiology and Medicine, College of Dentistry, University of Iowa, Iowa City, IA, USA
| | - M Paula Gomez Hernandez
- Iowa Institute for Oral Health Research, N423 DSB, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA
| | - Amber M Bates
- Iowa Institute for Oral Health Research, N423 DSB, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA
| | - Christopher N Treinen
- Iowa Institute for Oral Health Research, N423 DSB, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA
| | - Emily E Starman
- Iowa Institute for Oral Health Research, N423 DSB, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA
| | - Carol L Fischer
- Iowa Institute for Oral Health Research, N423 DSB, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA
| | | | - Janet M Guthmiller
- College of Dentistry, University of Nebraska Medical Center, 40th and Holdrege, Lincoln, NE, USA
| | - Georgia K Johnson
- Department of Periodontics, College of Dentistry, The University of Iowa, Iowa City, IA, USA
| | - Taher Abbasi
- Cellworks Research India Ltd, Whitefield, Bangalore, India
- Cellworks Group Inc, 2033 Gateway Place Suite 500, San Jose, CA, USA
| | - Shireen Vali
- Cellworks Research India Ltd, Whitefield, Bangalore, India
- Cellworks Group Inc, 2033 Gateway Place Suite 500, San Jose, CA, USA
| | - Kim A Brogden
- Iowa Institute for Oral Health Research, N423 DSB, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA.
- Department of Periodontics, College of Dentistry, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
72
|
Immune infiltration and PD-L1 expression in the tumor microenvironment are prognostic in osteosarcoma. Sci Rep 2016; 6:30093. [PMID: 27456063 PMCID: PMC4960483 DOI: 10.1038/srep30093] [Citation(s) in RCA: 212] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/10/2016] [Indexed: 12/11/2022] Open
Abstract
Osteosarcoma patient survival has remained stagnant for 30 years. Novel therapeutic approaches are needed to improve outcomes. We examined the expression of Programmed Death Ligand 1 (PD-L1) and defined the tumor immune microenvironment to assess the prognostic utility in osteosarcoma. PD-L1 expression in osteosarcoma was examined in two patient cohorts using immunohistochemistry (IHC) (n = 48, n = 59) and expression was validated using quantitative real time PCR (n = 21) and western blotting (n = 9). IHC was used to determine the presence of tumor infiltrating lymphocytes and antigen-presenting cells (APCs) in the tumor. Expression of PD-L1 was correlated with immune cell infiltration and event-free-survival (EFS). The 25% of primary osteosarcoma tumors that express PD-L1 were more likely to contain cells that express PD-1 than PD-L1 negative tumors (91.7% vs 47.2%, p = 0.002). Expression of PD-L1 was significantly associated with the presence of T cells, dendritic cells, and natural killer cells. Although all immune cell types examined were present in osteosarcoma samples, only infiltration by dendritic cells (28.3% vs. 83.9%, p = 0.001) and macrophages (45.5% vs. 84.4%, p = 0.031) were associated with worse five-year-EFS. PD-L1 expression was significantly associated with poorer five-year-EFS (25.0%. vs. 69.4%, p = 0.014). Further studies in osteosarcoma are needed to determine if targeting the PD-L1:PD-1 axis improves survival.
Collapse
|
73
|
A meta-analysis reveals prognostic role of programmed death ligand-1 in Asian patients with non-small cell lung cancer. ACTA ACUST UNITED AC 2016; 36:313-320. [PMID: 27376797 DOI: 10.1007/s11596-016-1585-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/12/2016] [Indexed: 12/25/2022]
Abstract
Accumulating studies explored the clinicopathologic and prognostic value of programmed death ligand-1 (PD-L1) in non-small cell lung cancer (NSCLC), but the results were controversial. We therefore conducted a meta-analysis to evaluate the predictive role of PD-L1 in NSCLC patients. We systematically collected relevant studies from PubMed, Embase, Web of Science and China National Knowledge Infrastructure. The pooled hazard ratios (HRs) with 95% confidence intervals (CIs) for overall survival (OS), and odd ratios (ORs) with 95% CIs for clinicopathologic factors were calculated. A total of 15 studies involving 3605 patients were included in this meta-analysis. The results showed no prognostic role of PD-L1 in the whole patients (HR=1.60, 95% CI: 0.88-2.89, P=0.123). Subgroup analysis showed that PD-L1 was associated with decreased OS in Asian patients (HR=2.00, 95% CI: 1.55-2.57, P<0.001). Among all the clinicopathologic factors, PD-L1 overexpression was significantly in relevance with poor tumor cell differentiation (HR=1.84, 95% CI: 1.49-2.28, P<0.001), late stage (HR=1.21, 95% CI: 1.02-1.43, P=0.026) and anaplastic lymphoma kinase (ALK) translocation (HR=2.63, 95% CI: 1.08-6.40, P=0.034), but not with other factors. In conclusion, our meta-analysis demonstrated that PD-L1 has a prognostic role in Asian patients with NSCLC.
Collapse
|
74
|
Safaei HR, Rostamzadeh A, Rahmani O, Mohammadi M, Ghaderi O, Yahaghi H, Ahmadi K. Prognostic investigations of B7-H1 and B7-H4 expression levels as independent predictor markers of renal cell carcinoma. Tumour Biol 2016; 37:7583-7. [PMID: 26687644 DOI: 10.1007/s13277-015-4652-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 12/14/2015] [Indexed: 12/31/2022] Open
Abstract
In order to evaluate the correlation of B7-H4 and B7-H1 with renal cell carcinoma (RCC), we analyzed B7-H1 and B7-H4 expressions and their clinical significance by immunohistochemical method. Our result indicated that B7-H4-positive staining was detected in 58.13 % of RCC tissues (25 tissues tumors), and there were 18 tissues of patients without detectable B7-H4. Furthermore, 21 cases (48.83 %) were B7-H1-positive. Positive tumor expressions of B7-H4 and B7-H1 were markedly related to advanced TNM stage (P = 0.001; P = 0.014), high grade (P = 0.001; P = 002), and larger tumor size (P = 0.002; P = 024) in RCC tissues than patients with B7-H4-negative and B7-H1-negative in RCC tissues. The patients with B7-H1 and B7-H4-positive expressions were found to be markedly correlated with the overall survival of the patients (P < 0.05) and tended to have an increased risk of death when compared with negative expression groups. Univariate analysis showed that B7-H4 and B7-H1 expressions, TNM stage, high grade, and tumor size were significantly related to the prognosis of RCC. Furthermore, multivariate analysis showed that B7-H4 and B7-H1 expressions decreased overall survival. The adjusted HR for B7-H1 was 2.83 (95 % CI 1.210-2.971; P = 0.031) and also was 2.918 (95 % CI 1.243-3.102; P = 0.006) for B7-H4 that showed these markers were independent prognostic factors in RCC patients. The expressions of B7-H1 and B7-H4 in RCC patients indicate that these markers may be as a predictor of tumor development and death risk. Further investigations can be helpful to confirm B7-H1 and B7-H4 roles as an independent predictor of clinical RCC outcome.
Collapse
Affiliation(s)
- Hamid Reza Safaei
- Department of Pediatric Nephrology, AJA University of Medical Sciences, Tehran, Iran
| | - Ayoob Rostamzadeh
- Department of Anatomical Sciences, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Omid Rahmani
- Department of Pathology, Be'sat Hospital, AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Mohammadi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Omar Ghaderi
- Department of Pharmaceutical Biotechnology, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Yahaghi
- Department of Molecular Biology, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Koroosh Ahmadi
- Department of Emergency Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
75
|
Colli LM, Machiela MJ, Myers TA, Jessop L, Yu K, Chanock SJ. Burden of Nonsynonymous Mutations among TCGA Cancers and Candidate Immune Checkpoint Inhibitor Responses. Cancer Res 2016; 76:3767-72. [PMID: 27197178 DOI: 10.1158/0008-5472.can-16-0170] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/20/2016] [Indexed: 02/07/2023]
Abstract
Immune checkpoint inhibitor treatment represents a promising approach toward treating cancer and has been shown to be effective in a subset of melanoma, non-small cell lung cancer (NSCLC), and kidney cancers. Recent studies have suggested that the number of nonsynonymous mutations (NsM) can be used to select melanoma and NSCLC patients most likely to benefit from checkpoint inhibitor treatment. It is hypothesized that a higher burden of NsM generates novel epitopes and gene products, detected by the immune system as foreign. We conducted an assessment of NsM across 7,757 tumor samples drawn from 26 cancers sequenced in the Cancer Genome Atlas (TCGA) Project to estimate the subset of cancers (both types and fractions thereof) that fit the profile suggested for melanoma and NSCLC. An additional independent set of 613 tumors drawn from 5 cancers were analyzed for replication. An analysis of the receiver operating characteristic curves of published data on checkpoint inhibitor response in melanoma and NSCLC data estimates a cutoff of 192 NsM with 74% sensitivity and 59.3% specificity to discriminate potential clinical benefit. Across the 7,757 samples of TCGA, 16.2% displayed an NsM count that exceeded the threshold of 192. It is notable that more than 30% of bladder, colon, gastric, and endometrial cancers have NsM counts above 192, which was also confirmed in melanoma and NSCLC. Our data could inform the prioritization of tumor types (and subtypes) for possible clinical trials to investigate further indications for effective use of immune checkpoint inhibitors, particularly in adult cancers. Cancer Res; 76(13); 3767-72. ©2016 AACR.
Collapse
Affiliation(s)
- Leandro M Colli
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Timothy A Myers
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lea Jessop
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
76
|
Lipowska-Bhalla G, Fagnano E, Illidge TM, Cheadle EJ. Improving therapeutic activity of anti-CD20 antibody therapy through immunomodulation in lymphoid malignancies. Leuk Lymphoma 2016; 57:1269-80. [PMID: 27050042 DOI: 10.3109/10428194.2016.1157874] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Nearly two decades ago rituximab heralded a new era in management of B cell malignancies significantly increasing response rates and survival. However, despite clear therapeutic advantage, significant numbers of patients become refractory to anti-CD20 mAb therapy, suggesting urgent improvements are required. It is now well recognized that the suppressive tumor microenvironment plays an important role in the outcome of anti-CD20 mAb therapy and that manipulation of this environment may improve the efficacy and produce long-term tumor control. The past few years have seen a surge of interest in immunomodulatory agents capable of overwriting immune suppressive networks into favorable clinical outcome. Currently, a number of such combinations with anti-CD20 mAb is under evaluation and some have produced encouraging outcomes in rituximab refractory disease. In this review, we give an outline of anti-CD20 mAbs and explore the combinations with immunomodulatory agents that enhance antitumor immunity through targeting stimulatory or inhibitory pathways and have proven potential to synergize with anti-CD20 mAb therapy. These agents, primarily mAbs, target CTLA-4, PD-1/PD-L1, and CD40.
Collapse
MESH Headings
- Animals
- Antigens, CD20
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- B7-H1 Antigen/antagonists & inhibitors
- CD40 Antigens/antagonists & inhibitors
- CTLA-4 Antigen/antagonists & inhibitors
- Cytotoxicity, Immunologic/drug effects
- Humans
- Immunologic Factors/pharmacology
- Immunologic Factors/therapeutic use
- Immunomodulation/drug effects
- Leukemia, B-Cell/drug therapy
- Leukemia, B-Cell/immunology
- Leukemia, B-Cell/metabolism
- Leukemia, B-Cell/pathology
- Lymphoma, B-Cell/drug therapy
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/metabolism
- Lymphoma, B-Cell/pathology
- Molecular Targeted Therapy
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Rituximab/pharmacology
- Rituximab/therapeutic use
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Grazyna Lipowska-Bhalla
- a Targeted Therapy Group, Institute of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre , Manchester , UK
| | - Ester Fagnano
- a Targeted Therapy Group, Institute of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre , Manchester , UK
| | - Timothy M Illidge
- a Targeted Therapy Group, Institute of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre , Manchester , UK
| | - Eleanor J Cheadle
- a Targeted Therapy Group, Institute of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre , Manchester , UK
| |
Collapse
|
77
|
Abstract
Biliary tract cancers (BTCs), which encompass intra- and extrahepatic cholangiocarcinomas as well as gallbladder carcinomas, are one of the most aggressive malignancies. Although the development of systemic chemotherapy approaches has made progress, the prognosis of BTC remains poor. Chronic inflammation plays an important role in the carcinogenesis of BTC, highlighting the immune etiology of this disease. Immunotherapy has emerged as a promising new modality of treatment for BTC. Here, we summarize the relevant tumor immunology of BTC and recently completed and ongoing clinical trials of immunotherapy for BTC.
Collapse
Affiliation(s)
- Yi Chai
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
78
|
Leng C, Li Y, Qin J, Ma J, Liu X, Cui Y, Sun H, Wang Z, Hua X, Yu Y, Li H, Zhang J, Zheng Y, Wang W, Zhu J, Wang Q. Relationship between expression of PD-L1 and PD-L2 on esophageal squamous cell carcinoma and the antitumor effects of CD8⁺ T cells. Oncol Rep 2015; 35:699-708. [PMID: 26718132 DOI: 10.3892/or.2015.4435] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 08/18/2015] [Indexed: 11/06/2022] Open
Abstract
The programmed death-1 (PD-1)/programmed death-ligands (PD-Ls) signal pathway has been implicated as a potential immune escape mechanism in several human cancers. However, the studies of PD‑1/PD‑Ls pathway in esophageal squamous cell carcinoma (ECSS) are not yet sufficient. The current study investigated the expression of PD‑L1, PD‑L2 and PD‑1 in ESCC tissues. The correlations between the expression of these proteins and clinical histopathological parameters were analyzed. Then the stable transfected Ec109 cell lines overexpressing PD‑L1/PD‑L2 were established by plasmid transfection successfully. Ec109 and CD8+ T cells were co‑cultured to analyze the effects of PD‑1/PD‑Ls signal pathway on the function of CD8+ T cells including proliferation, apoptosis and interferon‑γ production. We found that PD‑L1-positive patients had significantly poorer prognosis than the negative patients, while their prognosis was not related to PD‑L2 expression. The count of PD‑1+ TILs (tumor‑infiltrating lymphocytes) was negatively correlated with both PD‑L1 and PD‑L2 expression. In functional studies, we found that PD‑1/PD‑Ls signal pathway was able to downregulate the function of CD8+ T lymphocyte and its function could be restored by blocking the signal pathway. This indicates that PD‑1/PD‑Ls may prevent effective antitumor immunity, which provides important evidence to delineate the cellular immune deficiency mechanism in ESCC. Therefore, PD-1/PD-Ls are predicted to become novel targets for ESCC immunotherapy.
Collapse
Affiliation(s)
- Changsen Leng
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Yin Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Jianjun Qin
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Jun Ma
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Xianben Liu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Yingying Cui
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Haibo Sun
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Zongfei Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Xionghuai Hua
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Yongkui Yu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Haomiao Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Jun Zhang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Yan Zheng
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Wei Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Junwei Zhu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Qiuming Wang
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| |
Collapse
|
79
|
The increase of circulating PD-L1-expressing CD68(+) macrophage in ovarian cancer. Tumour Biol 2015; 37:5031-7. [PMID: 26541760 DOI: 10.1007/s13277-015-4066-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 04/02/2014] [Indexed: 12/29/2022] Open
Abstract
Tumor-associated macrophages (TAMs) have been characterized as a critical population of immunosuppressive cells in a variety of tumor types. PD-L1 (also termed B7-H1) has been described to exert co-inhibitory and immune regulatory functions. Here, in ovarian cancer, PD-L1 is selectively overexpressed on some TAM compared that of benign ovarian disease. When expanding the data in peripheral blood, the proportion of PD-L1(+)CD68(+) cell among CD68(+) cells and the intensity of PD-L1 staining on CD68(+) cell in healthy group were similar to that observed in ovarian cyst group; instead, these two measures were significantly higher in ovarian cancer group, thereafter related to TNM stage. Interestingly, intracellular levels of IL-10, IL-6, TNF-α, and IFN-γ in PD-L1(+)CD68(+) macrophage were higher than those in PD-L1(-)CD68(+) macrophage, especially IL-6 expression. Based on the PD-L1 receptor PD-1 expression on tumor-infiltrating cytotoxic cells, our data supported that expression of PD-L1 on TAM promoted apoptosis of T cells via interaction with PD-1 on CD8(+)T cells. Taken together, these results suggested that PD-L1-expressing macrophage represents a novel suppressor cell population in ovarian cancer, which contributes immune escape of ovarian cancer.
Collapse
|
80
|
Xiong HY, Ma TT, Wu BT, Lin Y, Tu ZG. IL-12 regulates B7-H1 expression in ovarian cancer-associated macrophages by effects on NF-κB signalling. Asian Pac J Cancer Prev 2015; 15:5767-72. [PMID: 25081699 DOI: 10.7314/apjcp.2014.15.14.5767] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND AND AIM B7-H1, a co-inhibitory molecule of the B7 family, is found aberrantly expressed in ovarian cancer cells and infiltrating macrophage/dendritic-like cells, and plays a critical role in immune evasion by ovarian cancer. IL-12, an inducer of Th1 cell development, exerts immunomodulatory effects on ovarian cancer. However, whether IL-12 regulates B7-H1 expression in human ovarian cancer associated-macrophages has not been clarified. Therefore, we investigated the effects of IL-12 on the expression of B7-H1 in ovarian cancer-associated macrophages and possible mechanisms. METHODS PMA induced THP-1-derived macrophages or human monocyte-derived macrophages were treated with recombinant IL-12 (rIL-12) or infected with adenovirus carrying human IL-12 gene (Ad-IL-12-GFP) for 24 h, then cocultured with the SKOV3 ovarian cancer cell line for another 24 h. Macrophages were collected for real-time PCR and Western blot to detect the expression of B7-H1, and activation of the NF-κB signaling pathway. Moreover, supernatants were collected to assay for IL-12, IFN-γ and IL-10 by ELISA. In addition, monocyte-derived macrophages treated with IFN-γ were cocultured with SKOV3 and determined for the expression of B7-H1. Furthermore, the expression of B7-H1 in monocyte-derived macrophages was also evaluated after blocking NF-κB signaling. RESULTS The expression of B7-H1 was significantly upregulated in monocyte-derived macrophages treated with rIL-12 or Ad-IL-12-GFP compared with the control groups (p<0.05), accompanied by a remarkable upregulation of IFN-γ (p<0.05), a marked downregulation of IL-10 (p<0.05) and activation of NF-κB signaling. However, the upregulation of B7- H1 was inhibited by blocking the NF-κB signaling pathway (p<0.05). Expression of B7-H1 was also increased (p<0.05) in monocyte-derived macrophages treated with IFN-γ and cocultured with SKOV3. By contrast, the expression of B7-H1 in THP-1-derived macrophages was significantly decreased when treated in the same way as monocyte-derived macrophages (p<0.05), and IL-10 was also significantly decreased but IFN-γ was almost absent. CONCLUSIONS IL-12 upregulates the expression of B7-H1 in monocyte-derived macrophages, which is possible though inducing the secretion of IFN-γ and further activating the NF-κB signal pathway. However, IL-12 downregulates the expression of B7-H1 in THP-1-derived macrophages, associated with a lack of IFN-γ and inhibition of expression of IL-10.
Collapse
Affiliation(s)
- Hai-Yu Xiong
- College of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics of Education Ministry, Chongqing Medical University, Chongqing, China E-mail :
| | | | | | | | | |
Collapse
|
81
|
Qing Y, Li Q, Ren T, Xia W, Peng Y, Liu GL, Luo H, Yang YX, Dai XY, Zhou SF, Wang D. Upregulation of PD-L1 and APE1 is associated with tumorigenesis and poor prognosis of gastric cancer. Drug Des Devel Ther 2015; 9:901-909. [PMID: 25733810 PMCID: PMC4338255 DOI: 10.2147/dddt.s75152] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Gastric cancer is a fatal malignancy with a rising incidence rate. Effective methods for early diagnosis, monitoring metastasis, and prognosis are currently unavailable for gastric cancer. In this study, we examined the association of programmed death ligand-1 (PD-L1) and apurinic/apyrimidinic endonuclease 1 (APE1) expression with the prognosis of gastric cancer. METHODS The expressions of PD-L1 and APE1 were detected by immunohistochemistry in 107 cases of human gastric carcinoma. The correlation of PD-L1 and APE1 expression with the clinicopathologic features of gastric carcinoma was analyzed by SPSS version 19.0. RESULTS The positive expression rates of PD-L1 and APE1 in gastric cancer tissues were 50.5% (54/107) and 86.9% (93/107), respectively. PD-L1 and APE1 positive expressions were significantly associated with depth of invasion, lymph node metastasis, pathological type, overall survival, and higher T stage. Furthermore, the expression of PD-L1 in highly differentiated gastric cancers was higher than that in poorly differentiated cancers (P=0.008). Moreover, the expression of APE1 and PD-L1 in gastric cancers was positively correlated (r=0.336, P<0.01). Multivariate analysis showed that the depth of invasion was a significant prognostic factor (risk ratio 19.91; P=0.000), but there was no significant relationship with PD-L1, APE1, prognosis, and other characteristics. CONCLUSION The deregulation of PD-L1 and APE1 might contribute to the development and the poor prognosis of gastric cancer. Our findings suggest that high expression of PD-L1 and APE1 is a risk factor of gastric cancer and a new biomarker to predict the prognosis of gastric cancer. Furthermore, our findings suggest that targeting the PD-L1 and APE1 signaling pathways may be a new strategy for cancer immune therapy and targeted therapy for gastric cancer, especially in patients with deep invasion and lymph node metastasis.
Collapse
Affiliation(s)
- Yi Qing
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| | - Qing Li
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| | - Tao Ren
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| | - Wei Xia
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| | - Yu Peng
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| | - Gao-Lei Liu
- Urological Surgery, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| | - Hao Luo
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| | - Yu-Xin Yang
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| | - Xiao-Yan Dai
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Dong Wang
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
82
|
Bracarda S, Altavilla A, Hamzaj A, Sisani M, Marrocolo F, Del Buono S, Danielli R. Immunologic checkpoints blockade in renal cell, prostate, and urothelial malignancies. Semin Oncol 2015; 42:495-505. [PMID: 25965369 DOI: 10.1053/j.seminoncol.2015.02.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Genitourinary (GU) tumors, and in particular renal cell and prostate cancer, represent one of the most dynamic areas in oncology from the scientific point of view. One of the most recent treatment approaches for GU tumors has focused on a series of molecules known as immune checkpoints and the possibility of manipulating immune responses against tumor cells by blocking these molecules with monoclonal antibodies (mAbs). Cytotoxic T lymphocyte antigen-4 (CTLA-4), and the immune checkpoint inhibitor mAbs ipilimumab and tremelimumab, represent the prototypes of this new growing class of agents called immunomodulating antibodies, while programmed death/ligand 1 (PD-1/PD-L1) also has garnered a significant interest as a new immune checkpoints to target in urothelial cancer, with the anti-PD-1/PD-L1 inhibitor mAbs nivolumab, MPDL-3280, and BMS-936559 as the first agents tested. Here we report the encouraging initial data observed in GU cancers with this new class of agents, which have reinforced the interest of investigating the therapeutic potential of the immune checkpoint modulators in large controlled trials.
Collapse
Affiliation(s)
- Sergio Bracarda
- Medical Oncology Unit, Department of Oncology, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy.
| | - Amelia Altavilla
- Medical Oncology Unit, Department of Oncology, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy
| | - Alketa Hamzaj
- Medical Oncology Unit, Department of Oncology, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy
| | - Michele Sisani
- Medical Oncology Unit, Department of Oncology, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy
| | - Francesca Marrocolo
- Medical Oncology Unit, Department of Oncology, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy
| | - Sabrina Del Buono
- Medical Oncology Unit, Department of Oncology, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy
| | - Riccardo Danielli
- Medical Oncology and Immunotherapy Unit, Azienda Ospedaliera Senese, University of Siena, Istituto Toscano Tumori (ITT), Siena, Italy
| |
Collapse
|
83
|
Drakes ML, Stiff PJ. Harnessing immunosurveillance: current developments and future directions in cancer immunotherapy. Immunotargets Ther 2014; 3:151-65. [PMID: 27471706 PMCID: PMC4918242 DOI: 10.2147/itt.s37790] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Despite improved methods of cancer detection and disease management over the last few decades, cancer remains a major public health problem in many societies. Conventional therapies, such as chemotherapy, radiation, and surgery, are not usually sufficient to prevent disease recurrence. Therefore, efforts have been focused on developing novel therapies to manage metastatic disease and to prolong disease-free and overall survival, by modulating the immune system to alleviate immunosuppression, and to enhance antitumor immunity. This review discusses protumor mechanisms in patients that circumvent host immunosurveillance, and addresses current immunotherapy modalities designed to target these mechanisms. Given the complexity of cancer immunosuppressive mechanisms, we propose that identification of novel disease biomarkers will drive the development of more targeted immunotherapy. Finally, administration of different classes of immunotherapy in combination regimens, will be the ultimate route to impact low survival rates in advanced cancer patients.
Collapse
Affiliation(s)
- Maureen L Drakes
- Department of Medicine, Division of Hematology and Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Patrick J Stiff
- Department of Medicine, Division of Hematology and Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
84
|
Fusion protein of mutant B7-DC and Fc enhances the antitumor immune effect of GM-CSF-secreting whole-cell vaccine. J Immunother 2014; 37:147-54. [PMID: 24598447 DOI: 10.1097/cji.0000000000000025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
B7-DC [also known as programmed death ligand 2 (PD-L2)] is a costimulatory molecule expressed predominantly on dendritic cells (DCs) and macrophages. In addition to its coinhibitory receptor, programmed death receptor 1 (PD-1), evidence suggests that B7-DC interacts with an unidentified costimulatory receptor on T cells. B7-DC mutants with selective binding capacity for the costimulatory receptor may be effective in stimulating antitumor immune responses, while avoiding the inhibitory effects of PD-1. In this study, we concomitantly administered a GM-CSF-secreting whole-cell vaccine together with a fusion protein of mutant B7-DC and Fc portion (mB7-DC-Fc), which binds selectively to the costimulatory receptor. This lead to an increased number of tumor antigen-specific cytotoxic T lymphocytes both in the spleen and at the tumor site and complete elimination of established tumors in vivo. In addition, mB7-DC-Fc increased IFN-γ and IL-2 production and decreased IL-4 and IL-10 production in vitro, indicating that mB7-DC-Fc tips the Th1/Th2 balance toward Th1 dominance, which is more favorable for antitumor immunity. Furthermore, mB7-DC-Fc decreased the PD-1(+) proportion of CD8(+) T cells in vitro and tumor-infiltrating CD8(+) T cells in vivo, suggesting that mB7-DC-Fc may maintain tumor-infiltrating CD8(+) T cells in a nonexhausted state. In conclusion, mB7-DC-Fc administration during the T-cell priming phase enhances antitumor effects of vaccine by generating more tumor antigen-specific cytotoxic T lymphocytes and leading to their accumulation at the tumor site. We suggest that this combination approach may be a promising strategy for antitumor immunotherapy.
Collapse
|
85
|
Bazhin AV, von Ahn K, Maier C, Soltek S, Serba S, Diehl L, Werner J, Karakhanova S. Immunological in vivo effects of B7-H1 deficiency. Immunol Lett 2014; 162:273-86. [PMID: 25173046 DOI: 10.1016/j.imlet.2014.08.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 08/11/2014] [Accepted: 08/19/2014] [Indexed: 11/27/2022]
Abstract
B7-H1 regulatory protein, a member of the B7-H family, plays a crucial role in the modulation of immune response in healthy steady-state conditions as well as in different pathologies. B7-H1 knockout mice represent an important model to elucidate further molecular and cellular mechanisms involved, among others, in autoimmunity development and cancer progression. However, a deep immunologic characterization of this model is not complete yet. This study examined the role of B7-H1 in vivo further by direct comparison of specifically phenotyped spleen immune-cell subpopulations and their activation and naïve/memory state as well as cytokine profile in wild-type and B7-H1 knockout mice. Our results demonstrated that B7-H1 deficiency in vivo modulates several immunological parameters, including the amount and composition of Gr1(+)CD11b(+) myeloid population, the composition and activation state of the DC compartment, the frequency and status of NK and NKT cells, B-cells, naïve/memory state of CD8 T-cells and production of IL-2 and IL-10 cytokines. Moreover, we observed an increase in the PD-1 expression in the immune cells in B7-H1 knockout mice compared to the wild-type animals. Valuing the importance of B7-H1 knockout mice for their use in disease models, these data underline the role of B7-H1 in vivo also in healthy state and should be taken into account in future studies on this immunosuppressive molecule.
Collapse
Affiliation(s)
- Alexandr V Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany.
| | - Katharina von Ahn
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Caroline Maier
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Sabine Soltek
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Susanne Serba
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Linda Diehl
- Institutes of Molecular Medicine, University of Bonn, Bonn, Germany; Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | | |
Collapse
|
86
|
Seo SK, Seo DI, Park WS, Jung WK, Lee DS, Park SG, Choi JS, Kang MS, Choi YH, Choi I, Yu BC, Choi IW. Attenuation of IFN-γ-induced B7-H1 expression by 15-deoxy-delta(12,14)-prostaglandin J2 via downregulation of the Jak/STAT/IRF-1 signaling pathway. Life Sci 2014; 112:82-9. [PMID: 25072357 DOI: 10.1016/j.lfs.2014.07.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 07/07/2014] [Accepted: 07/16/2014] [Indexed: 01/22/2023]
Abstract
AIM B7-H1, which belongs to the B7 family of costimulatory molecules, is implicated in the ability of tumors to evade the host immune response. The development of evasion mechanisms within the tumor microenvironment may be responsible for poor therapeutic responses. In this manuscript, we report that the 15-deoxy-δ(12,14)-prostaglandin J2 (15d-PGJ2), peroxisome proliferator-activated receptor gamma (PPARγ) activator leads to the downregulation of the cancer-associated expression of B7-H1 in response to interferon-gamma (IFN-γ) and the associated signaling cascades. MAIN METHODS The expression of B7-H1 from IFN-γ-induced B16F10 melanoma cells was measured with flow cytometric analysis. The regulatory mechanisms of 15d-PGJ2 on cellular signaling pathways were examined using Western blot and electrophoretic mobility shift assays. KEY FINDINGS The flow cytometric analysis revealed that the B7-H1 costimulatory molecule is significantly upregulated in B16F10 melanoma cells by stimulation with IFN-γ. However, 15d-PGJ2 strongly downregulates B7-H1 expression in IFN-γ-stimulated B16F10 melanoma cells. Furthermore, the significant damping effect of 15d-PGJ2 on B7-H1 expression involves the inhibition of the tyrosine phosphorylation of Janus kinase (Jak) and signal transducer(s) and activator(s) of transcription (STAT) and, thereby, the interferon regulatory factor-1 (IRF-1) trans-activation of STAT. These effects of 15d-PGJ2 were not abrogated by the PPARγ antagonist GW9662, indicating that they occur through a PPARγ-independent mechanism. SIGNIFICANCE In this study, we demonstrate that 15d-PGJ2 suppresses the IFN-γ-elicited expression of B7-H1 by the inhibition of IRF-1 transcription via the Jak/STAT signaling pathway through a PPARγ-independent mechanism in mouse melanoma cells.
Collapse
Affiliation(s)
- Su-Kil Seo
- Department of Microbiology, College of Medicine, Inje University, Busan, Republic of Korea
| | - Dae-Il Seo
- Department of Preventive Medicine, College of Medicine Kosin University, Busan, Republic of Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Won-Kyo Jung
- Department of Biomedical Engineering, and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan, Republic of Korea
| | - Dae-Sung Lee
- Marine Biodiversity Institute of Korea, Sejong, Republic of Korea
| | - Sae-Gwang Park
- Department of Microbiology, College of Medicine, Inje University, Busan, Republic of Korea
| | - Jung Sik Choi
- Department of Internal Medicine, Pusan Paik Hospital, College of Medicine Inje University, Busan, Republic of Korea
| | - Mi-Seon Kang
- Department of Pathology, Inje University College of Medicine, Busan, Republic of Korea
| | - Young Hyun Choi
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan, Republic of Korea
| | - Inhak Choi
- Department of Microbiology, College of Medicine, Inje University, Busan, Republic of Korea
| | - Byeng Chul Yu
- Department of Preventive Medicine, College of Medicine Kosin University, Busan, Republic of Korea.
| | - Il-Whan Choi
- Department of Microbiology, College of Medicine, Inje University, Busan, Republic of Korea.
| |
Collapse
|
87
|
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 2014; 5:200ra116. [PMID: 23986400 DOI: 10.1126/scitranslmed.3006504] [Citation(s) in RCA: 1393] [Impact Index Per Article: 126.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tumor escape from immune-mediated destruction has been associated with immunosuppressive mechanisms that inhibit T cell activation. Although evidence for an active immune response, including infiltration with CD8(+) T cells, can be found in a subset of patients, those tumors are nonetheless not immunologically rejected. In the current report, we show that it is the subset of T cell-inflamed tumors that showed high expression of three defined immunosuppressive mechanisms: indoleamine-2,3-dioxygenase (IDO), PD-L1/B7-H1, and FoxP3(+) regulatory T cells (T(regs)), suggesting that these inhibitory pathways might serve as negative feedback mechanisms that followed, rather than preceded, CD8(+) T cell infiltration. Mechanistic studies in mice revealed that up-regulated expression of IDO and PD-L1, as well as recruitment of T(regs), in the tumor microenvironment depended on the presence of CD8(+) T cells. The former was driven by interferon-γ and the latter by a production of CCR4-binding chemokines along with a component of induced proliferation. Our results argue that these major immunosuppressive pathways are intrinsically driven by the immune system rather than being orchestrated by cancer cells, and imply that cancer immunotherapy approaches targeting negative regulatory immune checkpoints might be preferentially beneficial for patients with a preexisting T cell-inflamed tumor microenvironment.
Collapse
Affiliation(s)
- Stefani Spranger
- Department of Pathology, Section of Hematology/Oncology, University of Chicago, IL 60637, USA
| | | | | | | | | | | | | |
Collapse
|
88
|
Merelli B, Massi D, Cattaneo L, Mandalà M. Targeting the PD1/PD-L1 axis in melanoma: Biological rationale, clinical challenges and opportunities. Crit Rev Oncol Hematol 2014; 89:140-65. [DOI: 10.1016/j.critrevonc.2013.08.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/10/2013] [Accepted: 08/15/2013] [Indexed: 12/16/2022] Open
|
89
|
Song X, Liu J, Lu Y, Jin H, Huang D. Overexpression of B7-H1 correlates with malignant cell proliferation in pancreatic cancer. Oncol Rep 2013; 31:1191-8. [PMID: 24378899 DOI: 10.3892/or.2013.2955] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 12/12/2013] [Indexed: 12/13/2022] Open
Abstract
B7-H1, a member of the B7 family of proteins, is hypothesised to play an important role in the immune escape of tumours through its binding to the PD-1 receptor on activated T and B cells. By inducing T lymphocyte apoptosis, tumour cells can suppress an effective antitumour immune response. Although the immunosuppressive effect of B7-H1 has been studied in many tumours, its other biological functions remain unclear. We previously demonstrated a high expression level of this molecule in pancreatic cancer patient samples and its antiapoptotic effect in pancreatic ductal adenocarcinoma (PDA) cells. The aim of the present study was to investigate the possible role of B7-H1 in the proliferation of PDA cells. Functional studies were performed using pancreatic cell lines that were genetically engineered to express high or low levels of B7-H1, and we found that the overexpression of B7-H1 through plasmid transfection in PDA cells promoted cell proliferation. Conversely, the short-hairpin RNA (shRNA) knockdown of B7-H1 inhibited PDA cell proliferation. Further analyses of the cell cycle and cell cycle-related molecules confirmed this result. Taken together, our results indicate that the upregulation of B7-H1 in pancreatic cancer cells promotes proliferation and accelerates carcinogenesis; these data, therefore, provide insights into the effects of B7-H1 overexpression on pancreatic tumour cells.
Collapse
Affiliation(s)
- Xiao Song
- Department of General Surgery, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310016, P.R. China
| | - Junwei Liu
- Department of General Surgery, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310016, P.R. China
| | - Yi Lu
- Department of General Surgery, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310016, P.R. China
| | - Hongchuan Jin
- Biomedical Research Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Dongsheng Huang
- Department of General Surgery, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
90
|
Gong W, Song Q, Lu X, Gong W, Zhao J, Min P, Yi X. Paclitaxel Induced B7-H1 Expression in Cancer Cells via the MAPK Pathway. J Chemother 2013; 23:295-9. [DOI: 10.1179/joc.2011.23.5.295] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
91
|
Krönig H, Kremmler L, Haller B, Englert C, Peschel C, Andreesen R, Blank CU. Interferon-induced programmed death-ligand 1 (PD-L1/B7-H1) expression increases on human acute myeloid leukemia blast cells during treatment. Eur J Haematol 2013; 92:195-203. [PMID: 24175978 DOI: 10.1111/ejh.12228] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2013] [Indexed: 12/27/2022]
Abstract
INTRODUCTION While current treatment for acute myeloid leukemia is characterized by high response rates, patients' long-term outcome is still disappointing, due to frequent relapse and ineligibility of the often elderly patients for stem cell transplantation approaches. Considerable efforts have, thus, been made to incorporate immunotherapeutic approaches in the acute myeloid leukemia (AML) consolidation, with so far disappointing clinical benefit. The B7 family ligand programmed-death receptor-ligand 1 (PD-L1, B7-H1, CD274) has been recently described (with conflicting results) to be expressed on AML blast cells, and interaction with its receptor on T cells, programmed death receptor-1 (PD-1, CD279), has been shown to suppress T-cell functions and to allow survival of dormant AML cells in animal models. DESIGN AND METHODS In this work, we analyzed freshly isolated myeloid precursor cells from healthy donors and from AML patients for PD-L1 expression with or without interferon-γ exposure at different time points during their treatment. RESULTS While without IFN exposure, only minor differences were observed, we found IFN-γ-induced PD-L1 expression most prominent after initial treatment and independent of treatment outcome. CONCLUSIONS Our observations support the recently suggested PD-L1-mediated adaptive immune resistance and argue for a targeting of the PD-L1/PD-1 pathway during the consolidation phase of AML treatment.
Collapse
Affiliation(s)
- Holger Krönig
- Department of Hematology and Medical Oncology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
92
|
Baglio F, Saresella M, Preti MG, Cabinio M, Griffanti L, Marventano I, Piancone F, Calabrese E, Nemni R, Clerici M. Neuroinflammation and brain functional disconnection in Alzheimer's disease. Front Aging Neurosci 2013; 5:81. [PMID: 24324435 PMCID: PMC3838994 DOI: 10.3389/fnagi.2013.00081] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/11/2013] [Indexed: 11/29/2022] Open
Abstract
Neuroinflammation and brain functional disconnection result from β-amyloid (Aβ) accumulation and play fundamental roles in the pathogenesis of Alzheimer’s disease (AD). We investigated possible correlations between these two AD-associated phenomena using DTI-based tractography and immunologic analyses in people with amnestic mild cognitive impairment (aMCI) and AD. DTI-Analyses focused on corpus callosum (CC). We found that frontal CC regions were preserved with respect to the posterior ones in aMCI; in these individuals significant correlations were seen between DTI-derived metrics in frontal-parietal CC areas and Aβ42-stimulated BDNF-producing CD4+ T lymphocytes and PDL-1-expressing CD14+ cells. These associations were lost in AD where DTI data involving the same CC areas correlated instead with Aβ42-stimulated interleukin (IL)-21 producing CD4+ T lymphocytes. Higher susceptibility to PDL-1-mediated apoptosis of Aβ42-specific lymphocytes and BDNF-associated survival of existing neurons could contribute to the relative CC structure preservation seen in aMCI. These potentially protective mechanisms are lost in frank AD, when severe alterations in the CC are mirrored in peripheral blood by proinflammatory cytokines-producing T cells. Monitoring of immune cells in peripheral blood could have a prognostic value in AD.
Collapse
|
93
|
Shi SJ, Wang LJ, Wang GD, Guo ZY, Wei M, Meng YL, Yang AG, Wen WH. B7-H1 expression is associated with poor prognosis in colorectal carcinoma and regulates the proliferation and invasion of HCT116 colorectal cancer cells. PLoS One 2013; 8:e76012. [PMID: 24124529 PMCID: PMC3790819 DOI: 10.1371/journal.pone.0076012] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 08/19/2013] [Indexed: 12/31/2022] Open
Abstract
Background And Objective The investigation concerning the B7-H1 expression in colorectal cancer cells is at an early stage. It is unclear whether B7-H1 expression may have diagnostic or prognostic value in colorectal carcinoma. Additionally, how B7-H1 is associated with the clinical features of colorectal carcinoma is not known. In order to investigate the relationship between B7-H1 and colorectal cancer, we analyzed B7-H1 expression and its effect in clinical specimens and HCT116 cells. Methods Paraffin-embedded specimens from 143 eligible patients were used to investigate the expression of CD274 by immunohistochemistry. We also examined whether B7-H1 itself may be related to cell proliferation, apoptosis, migration and invasion in colon cancer HCT116 cells. Results Our results show that B7-H1 was highly expressed in colorectal carcinoma and was significantly associated with cell differentiation status and TNM (Tumor Node Metastasis) stage. Patients with positive B7-H1 expression showed a trend of shorter survival time. Using multivariate analysis, we demonstrate that positive B7-H1 expression is an independent predictor of colorectal carcinoma prognosis. Our results indicate that B7-H1 silencing with siRNA inhibits cell proliferation, migration and invasion. Furthermore, cell apoptosis was also increased by B7-H1 inhibition. Conclusions Positive B7-H1 expression is an independent predictor for colorectal carcinoma prognosis. Moreover, knockdown of B7-H1 can inhibit cell proliferation, migration and invasion.
Collapse
Affiliation(s)
- Sheng-Jia Shi
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
| | - Li-Juan Wang
- Department of Oncology, the First Affiliated Hospital, College of Medicine, Xi’an Jiaotong University, Xi’an, China
| | - Guo-Dong Wang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Department of Comprehensive Medicine, 323 Hospital of the Chinese People’s Liberation Army, Xi’an, China
| | - Zhang-Yan Guo
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
| | - Ming Wei
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yan-Ling Meng
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
| | - An-Gang Yang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
- * E-mail: (WW); (AY)
| | - Wei-Hong Wen
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
- * E-mail: (WW); (AY)
| |
Collapse
|
94
|
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. [PMID: 23986400 DOI: 10.1126/scitranslmed.3006504.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Tumor escape from immune-mediated destruction has been associated with immunosuppressive mechanisms that inhibit T cell activation. Although evidence for an active immune response, including infiltration with CD8(+) T cells, can be found in a subset of patients, those tumors are nonetheless not immunologically rejected. In the current report, we show that it is the subset of T cell-inflamed tumors that showed high expression of three defined immunosuppressive mechanisms: indoleamine-2,3-dioxygenase (IDO), PD-L1/B7-H1, and FoxP3(+) regulatory T cells (T(regs)), suggesting that these inhibitory pathways might serve as negative feedback mechanisms that followed, rather than preceded, CD8(+) T cell infiltration. Mechanistic studies in mice revealed that up-regulated expression of IDO and PD-L1, as well as recruitment of T(regs), in the tumor microenvironment depended on the presence of CD8(+) T cells. The former was driven by interferon-γ and the latter by a production of CCR4-binding chemokines along with a component of induced proliferation. Our results argue that these major immunosuppressive pathways are intrinsically driven by the immune system rather than being orchestrated by cancer cells, and imply that cancer immunotherapy approaches targeting negative regulatory immune checkpoints might be preferentially beneficial for patients with a preexisting T cell-inflamed tumor microenvironment.
Collapse
Affiliation(s)
- Stefani Spranger
- Department of Pathology, Section of Hematology/Oncology, University of Chicago, IL 60637, USA
| | | | | | | | | | | | | |
Collapse
|
95
|
Thomas AA, Fisher JL, Ernstoff MS, Fadul CE. Vaccine-based immunotherapy for glioblastoma. CNS Oncol 2013; 2:331-49. [PMID: 25054578 PMCID: PMC6166520 DOI: 10.2217/cns.13.29] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma remains the most lethal human brain tumor, despite the advent of multimodal treatment approaches. Because immune tolerance plays an important role in tumor progression, adding immunotherapy has become an attractive and innovative treatment approach for these aggressive tumors. Several early-phase clinical trials have demonstrated that vaccine-based immunotherapies, including dendritic cell therapy, peptide-based vaccines and vaccines containing autologous tumor lysates, are feasible and well tolerated. These trials have revealed promising trends in overall survival and progression-free survival for patients with glioblastoma, and have paved the way for ongoing randomized controlled trials.
Collapse
Affiliation(s)
- Alissa A Thomas
- Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Jan L Fisher
- Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Marc S Ernstoff
- Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Camilo E Fadul
- Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, 1 Medical Center Drive, Lebanon, NH 03756, USA
| |
Collapse
|
96
|
Stiff PJ, Czerlanis C, Drakes ML. Dendritic cell immunotherapy in ovarian cancer. Expert Rev Anticancer Ther 2013; 13:43-53. [PMID: 23259426 DOI: 10.1586/era.12.153] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is one of the most frequent gynecological malignancies. However, as there is no effective screening method to detect early disease, it is usually only diagnosed when already widespread in the abdomen. The majority of patients diagnosed with advanced-stage disease will relapse and require additional therapy. In the search for additional effective treatments for the management of recurrent disease, researchers have focused on the potential usefulness of immunotherapeutic modulation by administering autologous immune cells, such as dendritic cells (DCs), to stimulate antitumor host responses. With the ultimate goal of improved survival, this review addresses mechanisms in ovarian cancer that may limit the expansion of antitumor immunity, discusses the parameters to be considered for optimal DC immunotherapy, outlines evaluation methodology used to monitor the success of treatment regimens and reviews reported DC immunotherapy trials in ovarian cancer.
Collapse
Affiliation(s)
- Patrick J Stiff
- Department of Medicine, Division of Hematology & Oncology, Cardinal Bernardin Cancer Center, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | | | | |
Collapse
|
97
|
Liu X, Gibbons RM, Harrington SM, Krco CJ, Markovic SN, Kwon ED, Dong H. Endogenous tumor-reactive CD8 + T cells are differentiated effector cells expressing high levels of CD11a and PD-1 but are unable to control tumor growth. Oncoimmunology 2013; 2:e23972. [PMID: 23894697 PMCID: PMC3716732 DOI: 10.4161/onci.23972] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 02/11/2013] [Accepted: 02/12/2013] [Indexed: 12/28/2022] Open
Abstract
Immunotherapies aimed at enhancing natural or endogenous antitumor T-cell immunity in patients affected by advanced malignancies are currently being implemented in the clinic with promising results. In order to optimize therapeutic protocols and monitor the effectiveness of such therapies, reliable biomarkers are needed. We used CD11a, an integrin that is upregulated on the surface of effector and memory CD8+ T cells, and PD-1, an immunoregulatory receptor expressed by activated T cells, as biomarkers to identify, quantify and monitor endogenous tumor-reactive cytotoxic T lymphocytes (CTLs) in two mouse tumor models and in the peripheral blood of 12 patients affected by Stage IV melanoma. High expression levels of CD11a and PD-1 were detected among CD8+ T cells residing within primary and metastatic murine tumor sites, as well as in spontaneous murine breast cancer tissues. In the peripheral blood of melanoma patients, tumor antigen-specific CD8+ T cells were associated with a population of CD11ahigh CD8+ T cells that co-expressed high levels of PD-1. Healthy donors exhibited a comparatively much lower frequency of such PD-1+CD11ahighCD8+ T cells. Phenotypic analyses demonstrated that CD11ahighCD8+ T cells are proliferating (Ki67+) and activated (CD62L-CD69+). Increased CD11ahighCD8+ T cells and delayed tumor growth were observed in PD-1 deficient mice, suggesting that the antitumor effector functions of CD8+ T cells is compromised by an elevated expression of PD-1. The CD11ahighCD8+ T-cell population expresses high levels of PD-1 and presumably constitutes the cellular target of PD-1 blockade therapy. The expression level of CD11a and PD-1 by CD8+ T cells may therefore represent a novel biomarker to identify and monitor endogenous tumor-reactive CTLs. This may not only provide an immunological readout for evaluating the efficacy of immunotherapy but also contribute to the selection of cancer patients who are likely to benefit from anti-PD-1 therapy.
Collapse
Affiliation(s)
- Xin Liu
- Department of Urology; College of Medicine; Mayo Clinic; Rochester, MN USA
| | - Rachel M. Gibbons
- Department of Immunology; College of Medicine; Mayo Clinic; Rochester, MN USA
| | | | | | - Svetomir N. Markovic
- Department of Hematology and Oncology; College of Medicine; Mayo Clinic; Rochester, MN USA
| | - Eugene D. Kwon
- Department of Urology; College of Medicine; Mayo Clinic; Rochester, MN USA
- Department of Immunology; College of Medicine; Mayo Clinic; Rochester, MN USA
| | - Haidong Dong
- Department of Urology; College of Medicine; Mayo Clinic; Rochester, MN USA
- Department of Immunology; College of Medicine; Mayo Clinic; Rochester, MN USA
| |
Collapse
|
98
|
Elhag OAO, Hu XJ, Wen-Ying Z, Li X, Yuan YZ, Deng LF, Liu DL, Liu YL, Hui G. Reconstructed adeno-associated virus with the extracellular domain of murine PD-1 induces antitumor immunity. Asian Pac J Cancer Prev 2013; 13:4031-6. [PMID: 23098512 DOI: 10.7314/apjcp.2012.13.8.4031] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The negative signaling provided by interactions of the co-inhibitory molecule, programmed death-1 (PD-1), and its ligands, B7-H1 (PD-L1) and B7-DC (PD-L2), is a critical mechanism contributing to tumor evasion; blockade of this pathway has been proven to enhance cytotoxic activity and mediate antitumor therapy. Here we evaluated the anti-tumor efficacy of AAV-mediated delivery of the extracellular domain of murine PD-1 (sPD-1) to a tumor site. MATERIAL AND METHODS An rAAV vector was constructed in which the expression of sPD-1, a known negative regulator of TCR signals, is driven by human cytomegalovirus immediate early promoter (CMV-P), using a triple plasmid transfection system. Tumor-bearing mice were then treated with the AAV/sPD1 construct and expression of sPD-1 in tumor tissues was determined by semi quantitative RT-PCR, and tumor weights and cytotoxic activity of splenocytes were measured. RESULTS Analysis of tumor homogenates revealed sPD-1 mRNA to be significantly overexpressed in rAAV/sPD-1 treated mice as compared with control levels. Its use for local gene therapy at the inoculation site of H22 hepatoma cells could inhibit tumor growth, also enhancing lysis of tumor cells by lymphocytes stimulated specifically with an antigen. In addition, PD-1 was also found expressed on the surfaces of activated CD8+ T cells. CONCLUSION This study confirmed that expression of the soluble extracellular domain of PD-1 molecule could reduce tumor microenvironment inhibitory effects on T cells and enhance cytotoxicity. This suggests that it might be a potential target for development of therapies to augment T-cell responses in patients with malignancies.
Collapse
Affiliation(s)
- Osama A O Elhag
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Hubei, China
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Cancer immunotherapy strategies based on overcoming barriers within the tumor microenvironment. Curr Opin Immunol 2013; 25:268-76. [PMID: 23579075 DOI: 10.1016/j.coi.2013.02.009] [Citation(s) in RCA: 337] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 02/11/2013] [Accepted: 02/15/2013] [Indexed: 12/22/2022]
Abstract
For tumor antigen-specific T cells to effectively control the growth of cancer cells in vivo, they must gain access to, and function within, the tumor microenvironment. While tumor antigen-based vaccines and T cell adoptive transfer strategies can result in clinical benefit in a subset of patients, most of the patients do not respond clinically. Even for tumor-infiltrating lymphocyte (TIL)-based adoptive transfer for patients with metastatic melanoma, which can provide tumor shrinkage in around 50% of treated individuals, many patients are not eligible, in part because there are not sufficient TIL present in the resected tumor. Thus, the denominator is in fact larger, and it has been suggested that absence of TIL may be a marker for poor efficacy of immunotherapies in general. While qualitative and/or quantitative features of the T cells are important considerations for efficacy, a major component of primary resistance likely can be attributed to the tumor microenvironment. Data are accumulating suggesting that two major categories of immune resistance within the tumor microenvironment may exist: failure of T cell trafficking due to low levels of inflammation and lack of chemokines for migration, and dominant suppression through immune inhibitory mechanisms. New therapeutic interventions are being guided by these observations, and preliminary clinical success is validating this working model.
Collapse
|
100
|
Ott PA, Henry T, Baranda SJ, Frleta D, Manches O, Bogunovic D, Bhardwaj N. Inhibition of both BRAF and MEK in BRAF(V600E) mutant melanoma restores compromised dendritic cell (DC) function while having differential direct effects on DC properties. Cancer Immunol Immunother 2013; 62:811-22. [PMID: 23306863 PMCID: PMC11028975 DOI: 10.1007/s00262-012-1389-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 12/13/2012] [Indexed: 01/08/2023]
Abstract
PURPOSE Dendritic cells (DCs) can induce strong tumor-specific T-cell immune responses. Constitutive upregulation of the mitogen-activated protein kinase (MAPK) pathway by a BRAF(V600) mutation, which is present in about 50 % of metastatic melanomas, may be linked to compromised function of DCs in the tumor microenvironment. Targeting both MEK and BRAF has shown efficacy in BRAF(V600) mutant melanoma. METHODS We co-cultured monocyte-derived human DCs with melanoma cell lines pretreated with the MEK inhibitor U0126 or the BRAF inhibitor vemurafenib. Cytokine production (IL-12 and TNF-α) and surface marker expression (CD80, CD83, and CD86) in DCs matured with the Toll-like receptor 3/Melanoma Differentiation-Associated protein 5 agonist polyI:C was examined. Additionally, DC function, viability, and T-cell priming capacity were assessed upon direct exposure to U0126 and vemurafenib. RESULTS Cytokine production and co-stimulation marker expression were suppressed in polyI:C-matured DCs exposed to melanoma cells in co-cultures. This suppression was reversed by MAPK blockade with U0126 and/or vemurafenib only in melanoma cell lines carrying a BRAF(V600E) mutation. Furthermore, when testing the effect of U0126 directly on DCs, marked inhibition of function, viability, and DC priming capacity was observed. In contrast, vemurafenib had no effect on DC function across a wide range of dose concentrations. CONCLUSIONS BRAF(V600E) mutant melanoma cells modulate DC through the MAPK pathway as its blockade can reverse suppression of DC function. MEK inhibition negatively impacts DC function and viability if applied directly. In contrast, vemurafenib does not have detrimental effects on important functions of DCs and may therefore be a superior candidate for combination immunotherapy approaches in melanoma patients.
Collapse
Affiliation(s)
- Patrick A Ott
- New York University Cancer Institute, New York University School of Medicine, 522 First Avenue, SRB 1303, New York, NY, 10016, USA.
| | | | | | | | | | | | | |
Collapse
|