101
|
Outh-Gauer S, Le Tourneau C, Broudin C, Scotte F, Roussel H, Hans S, Mandavit M, Tartour E, Badoual C. Actualités sur l’immunothérapie en pathologie des voies aérodigestives supérieures. Ann Pathol 2017; 37:79-89. [DOI: 10.1016/j.annpat.2016.12.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/07/2016] [Indexed: 02/06/2023]
|
102
|
Zhang X, Zhu S, Li T, Liu YJ, Chen W, Chen J. Targeting immune checkpoints in malignant glioma. Oncotarget 2017; 8:7157-7174. [PMID: 27756892 PMCID: PMC5351697 DOI: 10.18632/oncotarget.12702] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 10/12/2016] [Indexed: 12/31/2022] Open
Abstract
Malignant glioma is the most common and a highly aggressive cancer in the central nervous system (CNS). Cancer immunotherapy, strategies to boost the body's anti-cancer immune responses instead of directly targeting tumor cells, recently achieved great success in treating several human solid tumors. Although once considered "immune privileged" and devoid of normal immunological functions, CNS is now considered a promising target for cancer immunotherapy, featuring the recent progresses in neurobiology and neuroimmunology and a highly immunosuppressive state in malignant glioma. In this review, we focus on immune checkpoint inhibitors, specifically, antagonizing monoclonal antibodies for programmed cell death protein-1 (PD-1), cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), and indoleamine 2,3-dioxygenase (IDO). We discuss advances in the working mechanisms of these immune checkpoint molecules, their status in malignant glioma, and current preclinical and clinical trials targeting these molecules in malignant glioma.
Collapse
Affiliation(s)
- Xuhao Zhang
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Shan Zhu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Tete Li
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Yong-Jun Liu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
- Sanofi Research and Development, Cambridge, MA, USA
| | - Wei Chen
- ADC Biomedical Research Institute, Saint Paul, MN, USA
| | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| |
Collapse
|
103
|
Weichselbaum RR, Liang H, Deng L, Fu YX. Radiotherapy and immunotherapy: a beneficial liaison? Nat Rev Clin Oncol 2017; 14:365-379. [DOI: 10.1038/nrclinonc.2016.211] [Citation(s) in RCA: 564] [Impact Index Per Article: 70.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
104
|
Parra ER, Behrens C, Rodriguez-Canales J, Lin H, Mino B, Blando J, Zhang J, Gibbons DL, Heymach JV, Sepesi B, Swisher SG, Weissferdt A, Kalhor N, Izzo J, Kadara H, Moran C, Lee JJ, Wistuba II. Image Analysis-based Assessment of PD-L1 and Tumor-Associated Immune Cells Density Supports Distinct Intratumoral Microenvironment Groups in Non-small Cell Lung Carcinoma Patients. Clin Cancer Res 2016; 22:6278-6289. [PMID: 27252415 PMCID: PMC5558040 DOI: 10.1158/1078-0432.ccr-15-2443] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 04/21/2016] [Accepted: 05/21/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE We investigated the correlation between immunohistochemical PD-L1 expression and tumor-associated immune cells (TAICs) density in non-small cell lung carcinoma (NSCLC) and correlated them with clinicopathologic variables. EXPERIMENTAL DESIGN Tumor tissue specimens from 254 stage I-III NSCLCs [146 adenocarcinomas; 108 squamous cell carcinomas (SCCs)] were examined. PD-L1 expression in malignant cells and macrophages and the density of TAICs expressing CD3, CD4, CD8, CD57, granzyme B, CD45RO, PD-1, FOXP3, and CD68 were evaluated using immunohistochemistry and image analysis. RESULTS Malignant cells PD-L1 H-score > 5 was detected in 23% of adenocarcinomas and 31% of SCCs, and no significant differences were detected comparing both histologies; the median H-score in macrophages was significantly higher in SCC than in adenocarcinoma (P < 0.001). In adenocarcinoma, high malignant cells PD-L1 expression and high TAIC density correlated with solid histology, smoking history, and airflow limitation. Multivariate analysis demonstrated that high CD57-positive cell density correlated with better recurrence-free survival (RFS; P = 0.0236; HR, 0.457) and overall survival (OS; P = 0.0261; HR, 0.481) rates for SCC. High CD68-positive cell density in intratumoral compartment correlated with better RFS (P = 0.0436; HR, 0.553) for adenocarcinoma. The combination of low CD4/CD8/C68-positive cell density and PD-L1 H-score >5 in malignant cells identified small subset of adenocarcinomas with worse outcomes (RFS: P = 0.036; HR, 4.299; OS: P = 0.00034; HR, 5.632). CONCLUSIONS We detected different PD-L1 expression and TAIC density patterns in NSCLC. Distinct groups of tumor microenvironment correlated with NSCLC clinicopathologic features, including outcome. Clin Cancer Res; 22(24); 6278-89. ©2016 AACR.
Collapse
MESH Headings
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Aged
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- B7-H1 Antigen/metabolism
- Biomarkers, Tumor/metabolism
- CD57 Antigens/metabolism
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Disease-Free Survival
- Female
- Humans
- Immunohistochemistry/methods
- Kaplan-Meier Estimate
- Lung/metabolism
- Lung/pathology
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Male
- Tumor Microenvironment/physiology
Collapse
Affiliation(s)
- Edwin R Parra
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carmen Behrens
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jaime Rodriguez-Canales
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heather Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Barbara Mino
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jorge Blando
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephen G Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Annikka Weissferdt
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neda Kalhor
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Julie Izzo
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Humam Kadara
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cesar Moran
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jack J Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
105
|
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] [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
|
106
|
Davis RJ, Van Waes C, Allen CT. Overcoming barriers to effective immunotherapy: MDSCs, TAMs, and Tregs as mediators of the immunosuppressive microenvironment in head and neck cancer. Oral Oncol 2016; 58:59-70. [PMID: 27215705 PMCID: PMC4912416 DOI: 10.1016/j.oraloncology.2016.05.002] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 01/10/2023]
Abstract
A significant subset of head and neck cancers display a T-cell inflamed phenotype, suggesting that patients with these tumors should respond to therapeutic approaches aimed at strengthening anti-tumor immune responses. A major barrier to the development of an effective anti-tumor immune response, at baseline or in response to immunotherapy, is the development of an immunosuppressive tumor microenvironment. Several well described mechanisms of effector immune cell suppression in the head and neck cancer microenvironment are discussed here, along with updates on current trials designed to translate what we have learned from pre-clinical and correlative clinical studies into improved responses in patients with head and neck cancer following immune activating therapies.
Collapse
Affiliation(s)
- Ruth J Davis
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Carter Van Waes
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Clint T Allen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States; Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
107
|
Xu S, Tao Z, Hai B, Liang H, Shi Y, Wang T, Song W, Chen Y, OuYang J, Chen J, Kong F, Dong Y, Jiang SW, Li W, Wang P, Yuan Z, Wan X, Wang C, Li W, Zhang X, Chen K. miR-424(322) reverses chemoresistance via T-cell immune response activation by blocking the PD-L1 immune checkpoint. Nat Commun 2016; 7:11406. [PMID: 27147225 PMCID: PMC4858750 DOI: 10.1038/ncomms11406] [Citation(s) in RCA: 231] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 03/23/2016] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint blockade of the inhibitory immune receptors PD-L1, PD-1 and CTLA-4 has emerged as a successful treatment strategy for several advanced cancers. Here we demonstrate that miR-424(322) regulates the PD-L1/PD-1 and CD80/CTLA-4 pathways in chemoresistant ovarian cancer. miR-424(322) is inversely correlated with PD-L1, PD-1, CD80 and CTLA-4 expression. High levels of miR-424(322) in the tumours are positively correlated with the progression-free survival of ovarian cancer patients. Mechanistic investigations demonstrated that miR-424(322) inhibited PD-L1 and CD80 expression through direct binding to the 3'-untranslated region. Restoration of miR-424(322) expression reverses chemoresistance, which is accompanied by blockage of the PD-L1 immune checkpoint. The synergistic effect of chemotherapy and immunotherapy is associated with the proliferation of functional cytotoxic CD8+ T cells and the inhibition of myeloid-derived suppressive cells and regulatory T cells. Collectively, our data suggest a biological and functional interaction between PD-L1 and chemoresistance through the microRNA regulatory cascade.
Collapse
Affiliation(s)
- Shaohua Xu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Zhen Tao
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer institute &Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Bo Hai
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Huagen Liang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Ying Shi
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Tao Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Wen Song
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Yong Chen
- Emergency Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jun OuYang
- Department of Gynecology, Changzhou Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Changzhou 213003, China
| | - Jinhong Chen
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Fanfei Kong
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Yishan Dong
- Department of Gynecology, Changzhou Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Changzhou 213003, China
| | - Shi-Wen Jiang
- Department of Biomedical Science, Mercer University School of Medicine, Savannah, Georgia 31404, USA
| | - Weiyong Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Ping Wang
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer institute &Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Zhiyong Yuan
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer institute &Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Xiaoping Wan
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Chenguang Wang
- Key Laboratory of Tianjin Radiation and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College &Chinese Academy of Medical Sciences, Tianjin 300308, China
| | - Wencheng Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Ke Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| |
Collapse
|
108
|
Ren R, Koti M, Hamilton T, Graham CH, Nayak JG, Singh J, Drachenberg DE, Siemens DR. A primer on tumour immunology and prostate cancer immunotherapy. Can Urol Assoc J 2016; 10:60-5. [PMID: 26977209 DOI: 10.5489/cuaj.3418] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
No abstract
Collapse
Affiliation(s)
- Runhan Ren
- Department of Urology, Queen's University, Kingston, ON, Canada
| | - Madhuri Koti
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Thomas Hamilton
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Charles H Graham
- Department of Urology, Queen's University, Kingston, ON, Canada;; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Jasmir G Nayak
- Section of Urology, University of Manitoba, Winnipeg, MB, Canada
| | - Jas Singh
- Section of Urology, University of Manitoba, Winnipeg, MB, Canada
| | | | - D Robert Siemens
- Department of Urology, Queen's University, Kingston, ON, Canada;; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
109
|
Zhang J, Wang CM, Zhang P, Wang X, Chen J, Yang J, Lu W, Zhou W, Yuan W, Feng Y. Expression of programmed death 1 ligand 1 on periodontal tissue cells as a possible protective feedback mechanism against periodontal tissue destruction. Mol Med Rep 2016; 13:2423-30. [PMID: 26847035 PMCID: PMC4768984 DOI: 10.3892/mmr.2016.4824] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 01/04/2016] [Indexed: 12/31/2022] Open
Abstract
Programmed death 1 ligand 1 (PD-L1) is a negative co-stimulatory molecule in immune responses. Previous reports have indicated that inflammatory cytokines can upregulate the expression of PD-L1 in tumor cells, which in turn suppresses host immune responses. Periodontitis is characterized by persistent inflammation of the periodontium, which is initiated by infection with oral bacteria and results in damage to cells and the matrices of the periodontal connective tissues. In the present study, the expression and function of PD-L1 in periodontal tissue destruction were examined. Periodontal ligament cells (PDLCs) were stimulated by inflammatory cytokines and periodontal pathogens. The expression and function of PD-L1 on the surface of PDLCs was investigated using flow cytometry in vitro. Periodontal disease was induced by the injection of Porphyromonas gingivalis in mouse models. The expression levels of PD-L1 in the periodontal tissues of the mice were analyzed using flow cytometry and immunohistochemistry. PD-L1 was inducibly expressed on the PDLCs by the inflammatory cytokines and periodontal pathogens. The inflammation-induced expression of PD-L1 was shown to cause the apoptosis of activated T lymphocytes and improve the survival of PDLCs. Furthermore, in the mouse model of experimental periodontitis, the expression of PD-L1 in severe cases of periodontitis was significantly lower, compared with that in mild cases. By contrast, no significant differences were observed between the healthy control and severe periodontitis groups. The results of the present study showed that the expression of PD-L1 may inhibit the destruction of periodontal tissues, indicating the involvement of a possible protective feedback mechanism against periodontal infection.
Collapse
Affiliation(s)
- Jiehua Zhang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Chieh-Mei Wang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ping Zhang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaoqian Wang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jiao Chen
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jun Yang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Wanlu Lu
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Wenjie Zhou
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Wenwen Yuan
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yun Feng
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
110
|
Josefsson A, Nedrow JR, Park S, Banerjee SR, Rittenbach A, Jammes F, Tsui B, Sgouros G. Imaging, Biodistribution, and Dosimetry of Radionuclide-Labeled PD-L1 Antibody in an Immunocompetent Mouse Model of Breast Cancer. Cancer Res 2015; 76:472-9. [PMID: 26554829 DOI: 10.1158/0008-5472.can-15-2141] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/28/2015] [Indexed: 12/16/2022]
Abstract
The programmed cell death ligand 1 (PD-L1) participates in an immune checkpoint system involved in preventing autoimmunity. PD-L1 is expressed on tumor cells, tumor-associated macrophages, and other cells in the tumor microenvironment. Anti-PD-L1 antibodies are active against a variety of cancers, and combined anti-PD-L1 therapy with external beam radiotherapy has been shown to increase therapeutic efficacy. PD-L1 expression status is an important indicator of prognosis and therapy responsiveness, but methods to precisely capture the dynamics of PD-L1 expression in the tumor microenvironment are still limited. In this study, we developed a murine anti-PD-L1 antibody conjugated to the radionuclide Indium-111 ((111)In) for imaging and biodistribution studies in an immune-intact mouse model of breast cancer. The distribution of (111)In-DTPA-anti-PD-L1 in tumors as well as the spleen, liver, thymus, heart, and lungs peaked 72 hours after injection. Coinjection of labeled and 100-fold unlabeled antibody significantly reduced spleen uptake at 24 hours, indicating that an excess of unlabeled antibody effectively blocked PD-L1 sites in the spleen, thus shifting the concentration of (111)In-DTPA-anti-PD-L1 into the blood stream and potentially increasing tumor uptake. Clearance of (111)In-DTPA-anti-PD-L1 from all organs occurred at 144 hours. Moreover, dosimetry calculations revealed that radionuclide-labeled anti-PD-L1 antibody yielded tolerable projected marrow doses, further supporting its use for radiopharmaceutical therapy. Taken together, these studies demonstrate the feasibility of using anti-PD-L1 antibody for radionuclide imaging and radioimmunotherapy and highlight a new opportunity to optimize and monitor the efficacy of immune checkpoint inhibition therapy.
Collapse
Affiliation(s)
- Anders Josefsson
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Jessie R Nedrow
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Sunju Park
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Andrew Rittenbach
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Fabien Jammes
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Benjamin Tsui
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - George Sgouros
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, Maryland.
| |
Collapse
|
111
|
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
|
112
|
Chowdhury F, Dunn S, Mitchell S, Mellows T, Ashton-Key M, Gray JC. PD-L1 and CD8+PD1+ lymphocytes exist as targets in the pediatric tumor microenvironment for immunomodulatory therapy. Oncoimmunology 2015. [DOI: 10.1080/2162402x.2015.1029701] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Ferdousi Chowdhury
- Cancer Sciences Unit; Faculty of Medicine; University of Southampton; Southampton, UK
| | - Stuart Dunn
- Cancer Sciences Unit; Faculty of Medicine; University of Southampton; Southampton, UK
| | - Simon Mitchell
- Cancer Sciences Unit; Faculty of Medicine; University of Southampton; Southampton, UK
| | - Toby Mellows
- Cellular Pathology; University Hospitals Southampton; Southampton, UK
| | - Margaret Ashton-Key
- Cancer Sciences Unit; Faculty of Medicine; University of Southampton; Southampton, UK
- Cellular Pathology; University Hospitals Southampton; Southampton, UK
| | - Juliet C Gray
- Cancer Sciences Unit; Faculty of Medicine; University of Southampton; Southampton, UK
| |
Collapse
|
113
|
Shrimali RK, Janik JE, Abu-Eid R, Mkrtichyan M, Khleif SN. Programmed death-1 & its ligands: promising targets for cancer immunotherapy. Immunotherapy 2015; 7:777-92. [PMID: 26250412 DOI: 10.2217/imt.15.49] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Novel strategies for cancer treatment involving blockade of immune inhibitors have shown significant progress toward understanding the molecular mechanism of tumor immune evasion. The preclinical findings and clinical responses associated with programmed death-1 (PD-1) and PD-ligand pathway blockade seem promising, making these targets highly sought for cancer immunotherapy. In fact, the anti-PD-1 antibodies, pembrolizumab and nivolumab, were recently approved by the US FDA for the treatment of unresectable and metastatic melanoma resistant to anticytotoxic T-lymphocyte antigen-4 antibody (ipilimumab) and BRAF inhibitor. Here, we discuss strategies of combining PD-1/PD-ligand interaction inhibitors with other immune checkpoint modulators and standard-of-care therapy to break immune tolerance and induce a potent antitumor activity, which is currently a research area of key scientific pursuit.
Collapse
Affiliation(s)
| | - John E Janik
- Georgia Regents University Cancer Center, Augusta, GA 30912, USA
| | - Rasha Abu-Eid
- Georgia Regents University Cancer Center, Augusta, GA 30912, USA
| | | | - Samir N Khleif
- Georgia Regents University Cancer Center, Augusta, GA 30912, USA
| |
Collapse
|
114
|
Harnessing the PD-1 pathway in renal cell carcinoma: current evidence and future directions. BioDrugs 2015; 28:513-26. [PMID: 25445176 DOI: 10.1007/s40259-014-0111-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Programmed cell death-1 (PD-1) is a recognized immune checkpoint. It is frequently upregulated on the T cells that infiltrate tumors, providing an inhibitory signal, which may facilitate immune escape. Blocking antibodies have been developed to interrupt the interaction of PD-1 with its ligands PD-L1/PD-L2, with the goal of increasing the host antitumor immune response. Initial results have been encouraging, with durable responses in both treatment-naive and pretreated patients, along with an acceptable toxicity profile. This tolerability makes PD-1 blockade an excellent potential partner for combination strategies with the approved targeted agents, such as tyrosine kinase inhibitors (TKIs) and anti-vascular endothelial growth factor (anti-VEGF) antibodies, as well as other investigational immune checkpoint inhibitors or agonist antibodies that may costimulate an immune response. PD-L1 expression on tumor cells and tumor-infiltrating immune cells is also being evaluated as a predictive biomarker of response to treatment. This review summarizes the biological basis, preclinical studies, ongoing trials, and future challenges associated with targeting the PD-1 pathway in renal cell carcinoma.
Collapse
|
115
|
Abstract
INTRODUCTION Despite improvements in treatment, survival rates of head and neck squamous cell carcinoma (HNSCC) are stagnant. The existing chemotherapeutic agents are non-selective and associated with toxicities. Combinations of the only the US FDA-approved epidermal growth factor receptor (EGFR)-targeted agent, cetuximab, with chemotherapy or radiation improves overall survival. However, the response rates to cetuximab are modest. Thus, there is an urgent need for new agents that can be safely integrated into current treatment regimens to improve outcome. AREAS COVERED Current EGFR-targeted drugs under clinical development include mAbs and tyrosine kinase inhibitors. The modest efficacy of these drugs implicates intrinsic or acquired resistance. Novel molecular agents inhibiting alternative targets to overcome anti-EGFR resistance in HNSCC are under investigation. Gene therapy and immunotherapy are also promising strategies to improve efficacy and reduce toxicity. EXPERT OPINION To date, only six drugs have been FDA-approved for the treatment of head and neck cancer. Cetuximab is the only approved molecular targeting agent for HNSCC and despite ubiquitous expression of EGFR in HNSCC tumors, clinical responses are limited. Genetic and epigenetic characterization of HNSCC tumors, coupled with improved preclinical models, should facilitate the development of more effective drugs.
Collapse
Affiliation(s)
- Yihui Wen
- Sun Yat-Sen University and The First Affiliated Hospital of Sun Yat-Sen University, Department of Otolaryngology, Guangzhou, Guangzhou, PR China
- Sun Yat-Sen University and Otolaryngology Institute of Sun Yat-Sen University, Guangzhou, Guangzhou, PR China
- University of Pittsburgh and University of Pittsburgh School of Medicine, Department of Otolaryngology, Pittsburgh, PA, USA
| | - Jennifer R Grandis
- University of Pittsburgh and University of Pittsburgh School of Medicine, Department of Otolaryngology, Pittsburgh, PA, USA
- University of Pittsburgh and University of Pittsburgh School of Medicine, Department of Pharmacology, Pittsburgh, PA, USA
- Eye and Ear Institute, 200 Lothrop St., Suite 500, Pittsburgh, PA, USA
- University of California, Clinical and Translational Science Institute, Department of Otolaryngology-Head and Neck Surgery, San Francisco, CA, USA
| |
Collapse
|
116
|
Datta J, Xu S, Rosemblit C, Smith JB, Cintolo JA, Powell DJ, Czerniecki BJ. CD4(+) T-Helper Type 1 Cytokines and Trastuzumab Facilitate CD8(+) T-cell Targeting of HER2/neu-Expressing Cancers. Cancer Immunol Res 2015; 3:455-63. [PMID: 25791067 PMCID: PMC4556111 DOI: 10.1158/2326-6066.cir-14-0208] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 03/12/2015] [Indexed: 12/17/2022]
Abstract
Vaccination strategies incorporating the immunodominant HLA-A2-restricted HER2/neu-derived peptide 369-377 (HER2369-377) are increasingly utilized in HER2/neu-expressing cancer patients. The failure of postvaccination HER2369-377-specific CD8(+) T cells to recognize HLA-A2(pos)HER2/neu-expressing cells in vitro, however, has been attributed to impaired MHC class I/HLA-A2 presentation observed in HER2/neu-overexpressing tumors. We reconcile this controversy by demonstrating that HER2369-377 is directly recognized by high functional-avidity HER2369-377-specific CD8(+) T cells-either genetically modified to express a novel HER2369-377 TCR or sensitized using HER2369-377-pulsed type 1-polarized dendritic cells (DC1)-on class I-abundant HER2(low), but not class I-deficient HER2(high), cancer cells. Importantly, a critical cooperation between CD4(+) T-helper type-1 (Th1) cytokines IFNγ/TNFα and HER2/neu-targeted antibody trastuzumab is necessary to restore class I expression in HER2(high) cancers, thereby facilitating recognition and lysis of these cells by HER2369-377-specific CD8(+) T cells. Concomitant induction of PD-L1 on HER2/neu-expressing cells by IFNγ/TNF and trastuzumab, however, has minimal impact on DC1-sensitized HER2369-377-CD8(+) T-cell-mediated cytotoxicity. Although activation of EGFR and HER3 signaling significantly abrogates IFNγ/TNFα and trastuzumab-induced class I restoration, EGFR/HER3 receptor blockade rescues class I expression and ensuing HER2369-377-CD8(+) cytotoxicity of HER2/neu-expressing cells. Thus, combinations of CD4(+) Th1 immune interventions and multivalent targeting of HER family members may be required for optimal anti-HER2/neu CD8(+) T-cell-directed immunotherapy.
Collapse
Affiliation(s)
- Jashodeep Datta
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Shuwen Xu
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Cinthia Rosemblit
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jenessa B Smith
- Department of Pathology and Laboratory Medicine; University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jessica A Cintolo
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Daniel J Powell
- Department of Pathology and Laboratory Medicine; University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Brian J Czerniecki
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania. Rena Rowen Breast Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
117
|
Bellucci R, Martin A, Bommarito D, Wang K, Hansen SH, Freeman GJ, Ritz J. Interferon-γ-induced activation of JAK1 and JAK2 suppresses tumor cell susceptibility to NK cells through upregulation of PD-L1 expression. Oncoimmunology 2015; 4:e1008824. [PMID: 26155422 PMCID: PMC4485824 DOI: 10.1080/2162402x.2015.1008824] [Citation(s) in RCA: 238] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 01/13/2015] [Accepted: 01/14/2015] [Indexed: 01/07/2023] Open
Abstract
Inhibition of JAK1 or JAK2 in human tumor cells was previously shown to increase susceptibility of these cells to NK cell lysis. In the present study, we examined the cellular mechanisms that mediate this effect in hematopoietic tumor cell lines and primary tumor cells. Incubation of tumor cells with supernatant from activated NK cells or interferon-gamma (IFNγ)-induced activation of pSTAT1 and increased expression of PD-L1 without altering expression of other activating or inhibitory NK cell ligands. These functional effects were blocked by chemical JAK inhibition or shRNAs targeting JAK1, JAK2 or STAT1. Inhibition of IFNγ signaling also prevented the upregulation of PD-L1 and blocking PD-L1 resulted in increased tumor lysis by NK cells. These results show that NK cell activation and secretion of IFNγ results in activation of JAK1, JAK2 and STAT1 in tumor cells, resulting in rapid up-regulation of PD-L1 expression. Increased expression of PD-L1 results in increased resistance to NK cell lysis. Blockade of JAK pathway activation prevents increased PD-L1 expression resulting in increased susceptibility of tumor cells to NK cell activity. These observations suggest that JAK pathway inhibitors as well as PD-1 and PD-L1 antibodies may work synergistically with other immune therapies by preventing IFN-induced inhibition of NK cell-mediated tumor cell lysis.
Collapse
Key Words
- ADCC, Antibody dependent cellular cytotoxicity
- AKT, Ak strain transforming
- APC, Allophycocyanin
- CTRL, Control
- DMSO, Dimethyl sulfoxide
- ERK, extracellular-signal-regulated kinases
- IFNγ
- JAK1/JAK2
- MACS, Magnetic cell separation
- MAPK, Mitogen-activated protein kinases
- NK cells
- PD-1/PD-L1
- RAS, Rat sarcoma
- STAT, signal transducer and activator of transcription
Collapse
Affiliation(s)
- Roberto Bellucci
- Department of Medical Oncology; Dana-Farber Cancer Institute ; Boston, MA, USA ; Department of Medicine; Brigham and Woman's Hospital ; Boston, MA, USA ; Harvard Medical School; Harvard University ; Boston, MA, USA
| | - Allison Martin
- Department of Medical Oncology; Dana-Farber Cancer Institute ; Boston, MA, USA
| | - Davide Bommarito
- Department of Medical Oncology; Dana-Farber Cancer Institute ; Boston, MA, USA
| | - Kathy Wang
- Department of Medical Oncology; Dana-Farber Cancer Institute ; Boston, MA, USA
| | - Steen H Hansen
- Department of Medicine; Brigham and Woman's Hospital ; Boston, MA, USA ; Harvard Medical School; Harvard University ; Boston, MA, USA ; GI Cell Biology Research Laboratory; Children's Hospital Boston ; Boston, MA, USA
| | - Gordon J Freeman
- Department of Medical Oncology; Dana-Farber Cancer Institute ; Boston, MA, USA ; Department of Medicine; Brigham and Woman's Hospital ; Boston, MA, USA ; Harvard Medical School; Harvard University ; Boston, MA, USA ; Cancer Vaccine Center; Dana-Farber Cancer Institute ; Boston, MA, USA
| | - Jerome Ritz
- Department of Medical Oncology; Dana-Farber Cancer Institute ; Boston, MA, USA ; Department of Medicine; Brigham and Woman's Hospital ; Boston, MA, USA ; Harvard Medical School; Harvard University ; Boston, MA, USA ; Cancer Vaccine Center; Dana-Farber Cancer Institute ; Boston, MA, USA
| |
Collapse
|
118
|
Reuben A, Godin-Ethier J, Santos MM, Lapointe R. T lymphocyte-derived TNF and IFN-γ repress HFE expression in cancer cells. Mol Immunol 2015; 65:259-66. [PMID: 25700349 DOI: 10.1016/j.molimm.2015.01.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 01/26/2015] [Accepted: 01/30/2015] [Indexed: 01/20/2023]
Abstract
The immune system and tumors are closely intertwined initially upon tumor development. During this period, tumors evolve to promote self-survival through immune escape, including by targeting crucial components involved in the presentation of antigens to the immune system in order to avoid recognition. Accordingly, components involved in MHC I presentation of tumor antigens are often mutated and down-regulated targets in tumors. On the other hand, the immune system has been shown to influence tumors through production of immunosuppressive cytokines, recruitment and polarization of cells favoring or impeding tumor escape or through production of anti-tumor cytokines promoting tumor rejection. We previously discovered that the hemochromatosis protein HFE, a negative regulator of iron absorption, dampens classical MHC I antigen presentation. In this study, we evaluated the impact of activated T lymphocytes purified from peripheral blood mononuclear cells (PBMC) on HFE expression in tumor cell lines. We co-cultured tumor cell lines from melanoma, lung, and kidney cancers with anti-CD3-activated PBMC and established that HFE expression is increased in tumor cell lines compared to healthy tissues, whilst being down-regulated significantly upon exposure to activated PBMC. HFE down-regulation was mediated by both CD4 and CD8 T lymphocytes, through production of soluble mediators, namely TNF and IFN-γ. These results suggest that the immune system may modulate tumor HFE expression in inflammatory conditions in order to regulate MHC I antigen presentation and promote tumor clearance.
Collapse
Affiliation(s)
- Alexandre Reuben
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, Québec, Canada H2X 0A9; Département de Médecine, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | - Jessica Godin-Ethier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, Québec, Canada H2X 0A9; Département de Médecine, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | - Manuela M Santos
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, Québec, Canada H2X 0A9; Département de Médecine, Université de Montréal, Montréal, Québec, Canada H3C 3J7.
| | - Réjean Lapointe
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, Québec, Canada H2X 0A9; Département de Médecine, Université de Montréal, Montréal, Québec, Canada H3C 3J7.
| |
Collapse
|
119
|
Clinicopathological analysis of PD-L1 and PD-L2 expression in pulmonary squamous cell carcinoma: Comparison with tumor-infiltrating T cells and the status of oncogenic drivers. Lung Cancer 2015; 88:24-33. [PMID: 25662388 DOI: 10.1016/j.lungcan.2015.01.016] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/09/2015] [Accepted: 01/16/2015] [Indexed: 12/13/2022]
Abstract
PURPOSE Programmed cell death-1 (PD-1)/programmed cell death-ligand-1 (PD-L1) pathway-targeted immunotherapy has beneficial therapeutic effects in pulmonary squamous cell carcinoma (SqCC) patients. However, the expression patterns of PD-1 and PD-1 ligands (PD-Ls) in pulmonary SqCC remain unclear. Moreover, the association between the PD-1/PD-Ls pathway and the status of oncogenic drivers in pulmonary SqCC is unknown. METHODS PD-L1 and PD-L2 expression in tumor cells and the numbers of PD-1(+) and CD8(+) tumor-infiltrating lymphocytes (TILs) were examined in 331 resected SqCC tumors along with matched lymph node metastases from 77 cases using immunohistochemistry. EGFR and FGFR1 and MET expression and genetic status were also examined. RESULTS PD-L1 and PD-L2 expression was detected in 26.9% and 23.9% of the pulmonary SqCC samples, respectively. PD-L1 and PD-L2 expression was maintained or increased in the metastatic lymph node tumors in 81.1% and 93.5% of the 77 cases, respectively. The numbers of PD-1(+) and CD8(+) TILs were significantly positively correlated (P<0.001). Cases displaying high PD-L1 expression exhibited consistently high CD8(+) T cell infiltration (P<0.001), even in subgroup analyses according to age, smoking status, tumor size, lymph node metastasis, stage, and the EGFR, MET and FGFR1 status. Moreover, MET expression in the tumors was significantly correlated with high PD-L2 expression and increased PD-1(+) TILs (P=0.001 for both). Increased numbers of CD8(+) or PD-1(+) TILs were significantly associated with prolonged disease-free survival of these patients, whereas PD-L1 and PD-L2 expression had no significant prognostic implications. CONCLUSION PD-L1 and PD-L2 expression in pulmonary SqCC is associated with an increased number of CD8(+) TILs and increased MET expression, which might provide therapeutic insight into targeting the PD-1/PD-Ls pathway in pulmonary SqCC.
Collapse
|
120
|
Ritprajak P, Azuma M. Intrinsic and extrinsic control of expression of the immunoregulatory molecule PD-L1 in epithelial cells and squamous cell carcinoma. Oral Oncol 2014; 51:221-8. [PMID: 25500094 DOI: 10.1016/j.oraloncology.2014.11.014] [Citation(s) in RCA: 254] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 11/13/2014] [Accepted: 11/20/2014] [Indexed: 01/01/2023]
Abstract
Recent clinical results for PD-1 blockade therapy have demonstrated durable tumor control with minimal immune-related adverse effects. PD-L1 is induced in non-lymphoid tissue cells and tumor cells, in addition to tissue-recruiting immune cells, under inflammatory conditions triggered by several cytokines, especially IFN-γ, and exogenous stimuli delivered by pathogen-associated molecular patterns. Receptor-mediated signaling molecules that affect the cell cycle, proliferation, apoptosis, and survival (including NF-κB, MAPK, PI3K, mTOR, and JAK/STAT) are involved in PD-L1 induction. PD-L1 expression in tumor cells is also triggered by the signals described above, but in some instances, intrinsic cell alteration associated with carcinogenesis contributes to PD-L1 induction. The tumor suppressor genes PTEN and Lkb1 and epithelial-mesenchymal transition-related molecules are also involved in the regulation of PD-L1 expression. Notably, squamous cell carcinoma of the head and neck (SCCHN) often exhibits both host immunosuppression and cytogenetic alternations of tumor cells. Precise understanding of how PD-L1 expression is controlled will allow the development of effective approaches to PD-1 blockade therapy for patients with SCCHN.
Collapse
Affiliation(s)
- Patcharee Ritprajak
- Department of Microbiology and Immunology and DRU of Oral Microbiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Miyuki Azuma
- Department of Molecular Immunology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
121
|
Harshman LC, Drake CG, Choueiri TK. PD-1 blockade in renal cell carcinoma: to equilibrium and beyond. Cancer Immunol Res 2014; 2:1132-41. [PMID: 25480555 PMCID: PMC4695990 DOI: 10.1158/2326-6066.cir-14-0193] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The past several years have witnessed a resurgence of interest in cancer immunotherapy. The development of blocking antibodies against the inhibitory programmed death-1 (PD-1) pathway represents a clinical breakthrough in the treatment of solid tumors such as melanoma, and these agents show great promise in renal cell carcinoma (RCC). The early data have been surprising in that they demonstrate that blockade of a single immune checkpoint can elicit objective responses in patients with RCC, despite the recognized complexity of the immunosuppressive tumor microenvironment. Reinvigorating the patient's own immune cells to reactivate and to target the tumor has the potential advantages of more selective killing and thus decreased toxicity. In addition, checkpoint blockade immunotherapy has the advantage of inducing a memory response that is unattainable with our current cytotoxic and targeted therapies. This Crossroads overview will highlight the emerging investigation of PD-1 blockade in RCC and how this T cell-targeted strategy may thwart the tumor's escape mechanisms and shift the immune system/tumor balance back to a state of equilibrium and even to tumor elimination.
Collapse
Affiliation(s)
- Lauren C Harshman
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Charles G Drake
- Department of Oncology and the Brady Urological Institute, Johns Hopkins University, Baltimore, Maryland
| | - Toni K Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
122
|
Peng W, Lizée G, Hwu P. Blockade of the PD-1 pathway enhances the efficacy of adoptive cell therapy against cancer. Oncoimmunology 2014; 2:e22691. [PMID: 23524510 PMCID: PMC3601154 DOI: 10.4161/onci.22691] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The blockade of the PD-1 pathway can increase the production of interferon γ by tumor-specific T cells located within or in the proximity of the malignant lesion, thereby increasing the chemokine-dependent trafficking of immune effector cells. This can boost the efficacy of adoptive T-cell therapy to achieve superior antitumor responses.
Collapse
Affiliation(s)
- Weiyi Peng
- Department of Melanoma Medical Oncology; The Center for Cancer Immunology Research; The University of Texas MD Anderson Cancer Center; Houston, TX USA
| | | | | |
Collapse
|
123
|
Zhu Y, Knolhoff BL, Meyer MA, Nywening TM, West BL, Luo J, Wang-Gillam A, Goedegebuure SP, Linehan DC, DeNardo DG. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res 2014; 74:5057-69. [PMID: 25082815 PMCID: PMC4182950 DOI: 10.1158/0008-5472.can-13-3723] [Citation(s) in RCA: 959] [Impact Index Per Article: 87.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer immunotherapy generally offers limited clinical benefit without coordinated strategies to mitigate the immunosuppressive nature of the tumor microenvironment. Critical drivers of immune escape in the tumor microenvironment include tumor-associated macrophages and myeloid-derived suppressor cells, which not only mediate immune suppression, but also promote metastatic dissemination and impart resistance to cytotoxic therapies. Thus, strategies to ablate the effects of these myeloid cell populations may offer great therapeutic potential. In this report, we demonstrate in a mouse model of pancreatic ductal adenocarcinoma (PDAC) that inhibiting signaling by the myeloid growth factor receptor CSF1R can functionally reprogram macrophage responses that enhance antigen presentation and productive antitumor T-cell responses. Investigations of this response revealed that CSF1R blockade also upregulated T-cell checkpoint molecules, including PDL1 and CTLA4, thereby restraining beneficial therapeutic effects. We found that PD1 and CTLA4 antagonists showed limited efficacy as single agents to restrain PDAC growth, but that combining these agents with CSF1R blockade potently elicited tumor regressions, even in larger established tumors. Taken together, our findings provide a rationale to reprogram immunosuppressive myeloid cell populations in the tumor microenvironment under conditions that can significantly empower the therapeutic effects of checkpoint-based immunotherapeutics.
Collapse
MESH Headings
- Adenocarcinoma/immunology
- Adenocarcinoma/pathology
- Animals
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/therapy
- Cohort Studies
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Female
- Humans
- Immunotherapy/methods
- Lectins, C-Type/biosynthesis
- Lectins, C-Type/immunology
- Macrophage Colony-Stimulating Factor/antagonists & inhibitors
- Macrophage Colony-Stimulating Factor/biosynthesis
- Macrophage Colony-Stimulating Factor/immunology
- Macrophages/immunology
- Mannose Receptor
- Mannose-Binding Lectins/biosynthesis
- Mannose-Binding Lectins/immunology
- Mice
- Mice, Inbred C57BL
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- Random Allocation
- Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors
- Receptor, Macrophage Colony-Stimulating Factor/immunology
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/immunology
- Signal Transduction
- T-Lymphocytes/immunology
- Tissue Array Analysis
- Tumor Microenvironment
- Gemcitabine
Collapse
Affiliation(s)
- Yu Zhu
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri. BRIGHT Institute, Washington University School of Medicine, St Louis, Missouri
| | - Brett L Knolhoff
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri. BRIGHT Institute, Washington University School of Medicine, St Louis, Missouri
| | - Melissa A Meyer
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri. BRIGHT Institute, Washington University School of Medicine, St Louis, Missouri
| | - Timothy M Nywening
- Department of Surgery, Washington University School of Medicine, St Louis, Missouri. Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
| | | | - Jingqin Luo
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri. Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri
| | - Andrea Wang-Gillam
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - S Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, St Louis, Missouri. Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
| | - David C Linehan
- Department of Surgery, Washington University School of Medicine, St Louis, Missouri. Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
| | - David G DeNardo
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri. BRIGHT Institute, Washington University School of Medicine, St Louis, Missouri. Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri. Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri.
| |
Collapse
|
124
|
Bernstein MB, Garnett CT, Zhang H, Velcich A, Wattenberg MM, Gameiro SR, Kalnicki S, Hodge JW, Guha C. Radiation-induced modulation of costimulatory and coinhibitory T-cell signaling molecules on human prostate carcinoma cells promotes productive antitumor immune interactions. Cancer Biother Radiopharm 2014; 29:153-61. [PMID: 24693958 DOI: 10.1089/cbr.2013.1578] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We sought to determine if single-dose external beam radiation therapy (EBRT) could modulate the expression signature of T-cell costimulatory and coinhibitory molecules in human prostate cancer (PCa) cell lines in vitro. We investigated the functional impact of irradiated PCa cells with a modulated costimulatory profile on responder T-cell activity. We used three PCa cell lines (DU145, PC3, and LNCaP) and two epithelial cell lines from noncancerous prostate and lung tissue. After 72 hours of EBRT, surface expression of four immunostimulatory molecules (CD70, CD275/ICOSL, CD134L/OX40L, and CD137L/41BBL) and two immunosuppressive markers (CTLA-4/CD152 and PD-L1/CD274) were evaluated by flow cytometry. We evaluated the impact of several radiation doses and the longevity of modulated expression. We examined the functional impact of radiation-induced modulation of cancer cells by cytotoxic T cells (CTL) cytotoxicity and ELISPOT assay for interferon-gamma (IFN-γ) production. Last, we evaluated whether IFN-γ-induced PD-L1 expression could be reversed by EBRT. After 10 Gy EBRT, expression of OX40L and 41BBL increased in all three PCa cell lines; expression of CD70 and ICOSL increased in PC3 cells. Conversely, a decrease in PD-L1 expression in DU145 and PC3 cells was detectable up to 144 hours after EBRT. No PD-L1 was detected in LNCaP. Epithelial cells from normal prostate were not modulated by radiation. CTL cytolytic activity and IFN-γ production were enhanced by interaction with irradiated PCa cells. Finally, EBRT failed to prevent IFN-γ-induced upregulation of PD-L1. We demonstrate that a single dose of EBRT increased surface expression of costimulatory molecules and decreased the expression of coinhibitory molecules in human PCa cell lines. Changes in irradiated tumor cells led to functional enhancement of T-cell activity, despite EBRT failing to reduce IFN-γ-induced expression of PD-L1. These data suggest that combining radiotherapy with T-cell stimulating immunotherapy may be an attractive strategy for cancer treatment.
Collapse
Affiliation(s)
- Michael B Bernstein
- 1 Department of Radiation Oncology, Montefiore Medical Center , Albert Einstein College of Medicine, Bronx, New York
| | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Varilla V, Atienza J, Dasanu CA. Immune alterations and immunotherapy prospects in head and neck cancer. Expert Opin Biol Ther 2013; 13:1241-56. [PMID: 23789839 DOI: 10.1517/14712598.2013.810716] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Several literature sources have suggested that subjects with head and neck squamous cell carcinoma (HNSCC) display significant abnormalities of immunocompetent cells and cytokine secretion. Serious side effects and only a limited success with traditional therapies in HNSCC dictate the need for newer therapies. AREAS COVERED This article comprehensively reviews the immune system alterations in HNSCC and the rationale behind various experimental immunotherapies, aiming at keeping this disease under control. Relevant publications were identified through the PubMed database search. The ongoing clinical trials regarding experimental immunotherapy agents in HNSCC were accessed at www.clinicaltrials.gov . The obtained information was thoroughly analyzed and systematized. EXPERT OPINION Important and severe immune defects including T-cell dysfunction, cytokine alterations and antigen presentation defects are present in patients with HNSCC. In addition, tumor microenvironment was shown to play a critical role in the HNSCC progression. These discoveries have triggered a growing interest in immunotherapy as a potential treatment strategy for HNSCC. Effective immunotherapy could avoid the toxic side effects plaguing the current management of HNSCC. It is also hoped that immunotherapy will have long-lasting effects due to induction of immunologic memory. Promising directions include nonspecific immune stimulation, targeting specific HNSCC tumor antigens and therapeutic vaccines among others. These new agents may expand the existing therapy options for HNSCC in future.
Collapse
Affiliation(s)
- Vincent Varilla
- University of Connecticut Medical Center, Department of Internal Medicine, Hartford, CT 06106, USA.
| | | | | |
Collapse
|
126
|
Lu W, Lu L, Feng Y, Chen J, Li Y, Kong X, Chen S, Li X, Chen Q, Zhang P. Inflammation promotes oral squamous carcinoma immune evasion via induced programmed death ligand-1 surface expression. Oncol Lett 2013; 5:1519-1526. [PMID: 23761816 PMCID: PMC3678870 DOI: 10.3892/ol.2013.1238] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 03/01/2013] [Indexed: 12/13/2022] Open
Abstract
The association between inflammation and cancer provides a new target for tumor biotherapy. The inflammatory cells and molecules within the tumor microenvironment have decisive dual roles in antitumor immunity and immune evasion. In the present study, phytohemagglutinin (PHA) was used to stimulate peripheral blood mononuclear cells (PBMCs) to simulate the tumor inflammatory microenvironment. The effect of immune cells and inflammatory cytokines on the surface expression of programmed cell death-1 ligand 1 (PD-L1) and tumor immune evasion was investigated using flow cytometry (FCM) and an in vivo xenotransplantation model. Based on the data, PHA-activated, but not resting, immune cells were able to promote the surface expression of PD-L1 in Tca8113 oral squamous carcinoma cells via the secretion of inflammatory cytokines, but not by cell-cell contact. The majority of the inflammatory cytokines had no significant effect on the proliferation, cell cycle progression and apoptosis of the Tca8113 cells, although they each induced the expression of PD-L1 in a dose-dependent manner. In total, 99% of the Tca8113 cells expressed PD-L1 following treatment with the supernatant of PHA-stimulated PBMCs. The PHA-supernatant pretreated Tca8113 cells unusually induced Tca8113 antigen-specific CD8+ T cell apoptosis in vitro and the evasion of antigen-specific T cell attraction in a nude mouse tumor-bearing model. These results indicate a new mechanism for the promotion of tumor immune evasion by the tumor inflammatory microenvironment
Collapse
Affiliation(s)
- Wanlu Lu
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Brusa D, Serra S, Coscia M, Rossi D, D'Arena G, Laurenti L, Jaksic O, Fedele G, Inghirami G, Gaidano G, Malavasi F, Deaglio S. The PD-1/PD-L1 axis contributes to T-cell dysfunction in chronic lymphocytic leukemia. Haematologica 2013; 98:953-63. [PMID: 23300177 DOI: 10.3324/haematol.2012.077537] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic lymphocytic leukemia is marked by profound defects in T-cell function. Programmed death-1 is a receptor involved in tumor-mediated immunosuppression through binding of the PD-L1 ligand. Multiparametric flow cytometry and immunohistochemistry were used to study PD-1/PD-L1 expression. Functional assays were used to determine the involvement of the PD-1/PD-L1 axis in T-cell responses. PD-1 expression by CD4(+) and CD8(+) T lymphocytes was significantly higher in 117 chronic lymphocytic leukemia patients than in 33 donors of a comparable age. CD4(+) and CD8(+) T lymphocytes from chronic lymphocytic leukemia patients displayed increased numbers of effector memory and terminally differentiated cells, respectively, when compared to controls. The number of effector memory CD4(+) and terminally differentiated CD8(+) lymphocytes positively associated with a more advanced stage of disease, treatment requirements and unfavorable genomic aberrations. Furthermore, leukemic lymphocytes expressed higher levels of PD-L1 than circulating B lymphocytes from normal donors. PD-1 and PD-L1 surface expression spiked in proliferating T and B lymphocytes, suggesting that this interaction works efficiently in activated environments. Within chronic lymphocytic leukemia proliferation centers in the lymph node, CD4(+)/PD-1(+) T lymphocytes were found to be in close contact with PD-L1(+) chronic lymphocytic leukemia cells. Lastly, functional experiments using recombinant soluble PD-L1 and blocking antibodies indicated that this axis contributes to the inhibition of IFN-γ production by CD8(+) T cells. These observations suggest that pharmacological manipulation of the PD-1/PD-L1 axis may contribute to restoring T-cell functions in the chronic lymphocytic leukemia microenvironment.
Collapse
Affiliation(s)
- Davide Brusa
- Human Genetics Foundation (HuGeF) and Department of Medical Sciences, University of Turin, Turin, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
|
129
|
Murphy KA, Lechner MG, Popescu FE, Bedi J, Decker SA, Hu P, Erickson JR, O'Sullivan MG, Swier L, Salazar AM, Olin MR, Epstein AL, Ohlfest JR. An in vivo immunotherapy screen of costimulatory molecules identifies Fc-OX40L as a potent reagent for the treatment of established murine gliomas. Clin Cancer Res 2012; 18:4657-68. [PMID: 22781551 DOI: 10.1158/1078-0432.ccr-12-0990] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE We tested the combination of a tumor lysate vaccine with a panel of costimulatory molecules to identify an immunotherapeutic approach capable of curing established murine gliomas. EXPERIMENTAL DESIGN Glioma-bearing mice were primed with a tumor lysate vaccine, followed by systemic administration of the following costimulatory ligands: OX40L, CD80, 4-1BBL, and GITRL, which were fused to the Fc portion of human immunoglobulin. Lymphocytes and mRNA were purified from the brain tumor site for immune monitoring studies. Numerous variations of the vaccine and Fc-OX40L regimen were tested alone or in combination with temozolomide. RESULTS Lysate vaccinations combined with Fc-OX40L led to the best overall survival, yielding cure rates of 50% to 100% depending on the timing, regimen, and combination with temozolomide. Cured mice that were rechallenged with glioma cells rejected the challenge, showing immunologic memory. Lymphocytes isolated from the draining lymph nodes of vaccine/Fc-OX40L-treated mice had superior tumoricidal function relative to all other groups. Vaccine/Fc-OX40L-treated mice exhibited a significant increase in proliferation of brain-infiltrating CD4 and CD8 T cells, as indicated by Ki67 staining. Fc-OX40L had single-agent activity in transplanted and spontaneous glioma models, and the pattern of inflammatory gene expression in the tumor predicted the degree of therapeutic response. CONCLUSIONS These data show that Fc-OX40L has unique and potent activity against experimental gliomas and warrants further testing.
Collapse
Affiliation(s)
- Katherine A Murphy
- Departments of Pediatrics and Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|