1
|
Paulsen EE, Kilvaer TK, Rakaee M, Richardsen E, Hald SM, Andersen S, Busund LT, Bremnes RM, Donnem T. CTLA-4 expression in the non-small cell lung cancer patient tumor microenvironment: diverging prognostic impact in primary tumors and lymph node metastases. Cancer Immunol Immunother 2017; 66:1449-1461. [PMID: 28707078 PMCID: PMC5645427 DOI: 10.1007/s00262-017-2039-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 07/07/2017] [Indexed: 12/11/2022]
Abstract
The immune checkpoint receptor CTLA-4 plays a crucial part in negatively regulating T cell activation and maintaining self-tolerance. It is frequently overexpressed in a variety of malignancies, yet its prognostic impact in non-small cell lung cancer (NSCLC) remains unclear. We constructed tissue microarrays from tumor tissue samples and evaluated the immunohistochemical expression of CTLA-4 in 536 patients with primary resected stage I-IIIA NSCLC. Expression of CTLA-4 was analyzed in tumor and stromal primary tumor tissue and in locoregional metastatic lymph nodes. CTLA-4 expression in neither tumor epithelial cells (T-CTLA-4) nor stromal cells (S-CTLA-4) of primary tumors was significantly associated with disease-specific survival (DSS) in all patients. However, high S-CTLA-4 expression independently predicted significantly improved DSS in the squamous cell carcinoma subgroup (HR 0.62, 95% CI 0.41-0.93, P = 0.021). In contrast, there was an independent negative prognostic impact of T-CTLA-4 expression in metastatic lymph nodes (HR 1.65, 95% CI 1.03-2.65, P = 0.039). Our results indicate that the expression of CTLA-4 has diverging prognostic impacts in metastatic NSCLC lymph nodes versus primary tumors. The presented results highlight important differences in the tumor microenvironments of primary and metastatic NSCLC tissues, and have potential to guide treatment and clinical sampling strategies.
Collapse
Affiliation(s)
- Erna-Elise Paulsen
- Department of Oncology, University Hospital of North Norway, Mailbox 13, 9038, Tromso, Norway.
- Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway.
- Translational Cancer Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9038, Tromso, Norway.
| | - Thomas K Kilvaer
- Department of Oncology, University Hospital of North Norway, Mailbox 13, 9038, Tromso, Norway
- Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Mehrdad Rakaee
- Department of Medical Biology, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Elin Richardsen
- Department of Clinical Pathology, University Hospital of North Norway, Mailbox 46, 9038, Tromso, Norway
- Department of Medical Biology, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Sigurd M Hald
- Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Sigve Andersen
- Department of Oncology, University Hospital of North Norway, Mailbox 13, 9038, Tromso, Norway
- Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Lill-Tove Busund
- Department of Clinical Pathology, University Hospital of North Norway, Mailbox 46, 9038, Tromso, Norway
- Department of Medical Biology, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Roy M Bremnes
- Department of Oncology, University Hospital of North Norway, Mailbox 13, 9038, Tromso, Norway
- Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Tom Donnem
- Department of Oncology, University Hospital of North Norway, Mailbox 13, 9038, Tromso, Norway
- Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| |
Collapse
|
2
|
Elevated regulatory T cells, surface and intracellular CTLA-4 expression and interleukin-17 in the lung cancer microenvironment in humans. Cancer Immunol Immunother 2016; 66:161-170. [PMID: 27866241 PMCID: PMC5281670 DOI: 10.1007/s00262-016-1930-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 11/14/2016] [Indexed: 12/16/2022]
Abstract
Regulatory T cells (Tregs) play an important role in the suppression of the immune response in lung cancer. Cytotoxic T-lymphocyte antigen 4 (CTLA-4) expressed on T lymphocytes is capable of downregulating cytotoxic T cells and is constitutively expressed on Tregs. Little is known about the population of Tregs with two forms of CTLA-4: surface (s) and intracellular (in) in the lung cancer environment. Th17 cells defined by production of IL-17 have pleiotropic functions in anticancer immune response. Our aim was to detect the elements of immune response regulation in lung cancer in three compartments: by analysis of bronchoalveolar lavage fluid (BALF) from the lung affected by cancer (clBALF), healthy symmetrical lung (hlBALF) and peripheral blood (PB) from the same patient. A total of 54 samples were collected. Tregs, (s)CTLA-4, (in)CTLA-4 were detected by flow cytometry with antibodies against CD4, CD25, Foxp3, CD127, CTLA-4, and concentration of IL-17 was estimated by ELISA. We observed a significantly higher proportion of Tregs in clBALF than in hlBALF or PB (8.5 vs. 5.0 vs. 5.1%, respectively, p < 0.05). The median proportion of (in)CTLA-4+ Tregs was higher in clBALF than in hlBALF or PB (89.0, 81.5, 56.0%, p < 0.05). IL-17 concentration was the highest in clBALF-6.6 pg/ml. We observed a significant correlation between the proportion of Tregs and (in)CTLA-4+ Tregs with IL-17A concentration in clBALF. We confirmed significant differences in the proportion of regulatory elements between cancerous lung and healthy lung and PB and the usefulness of BALF analysis in evaluation of immune response regulation in local lung cancer environment.
Collapse
|
3
|
McLaughlin J, Han G, Schalper KA, Carvajal-Hausdorf D, Pelekanou V, Rehman J, Velcheti V, Herbst R, LoRusso P, Rimm DL. Quantitative Assessment of the Heterogeneity of PD-L1 Expression in Non-Small-Cell Lung Cancer. JAMA Oncol 2016; 2:46-54. [PMID: 26562159 DOI: 10.1001/jamaoncol.2015.3638] [Citation(s) in RCA: 655] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
IMPORTANCE Early-phase trials with monoclonal antibodies targeting PD-1 (programmed cell death protein 1) and PD-L1 (programmed cell death 1 ligand 1) have demonstrated durable clinical responses in patients with non-small-cell lung cancer (NSCLC). However, current assays for the prognostic and/or predictive role of tumor PD-L1 expression are not standardized with respect to either quantity or distribution of expression. OBJECTIVE To demonstrate PD-L1 protein distribution in NSCLC tumors using both conventional immunohistochemistry (IHC) and quantitative immunofluorescence (QIF) and compare results obtained using 2 different PD-L1 antibodies. DESIGN, SETTING, AND PARTICIPANTS PD-L1 was measured using E1L3N and SP142, 2 rabbit monoclonal antibodies, in 49 NSCLC whole-tissue sections and a corresponding tissue microarray with the same 49 cases. Non-small-cell lung cancer biopsy specimens from 2011 to 2012 were collected retrospectively from the Yale Thoracic Oncology Program Tissue Bank. Human melanoma Mel 624 cells stably transfected with PD-L1 as well as Mel 624 parental cells, and human term placenta whole tissue sections were used as controls and for antibody validation. PD-L1 protein expression in tumor and stroma was assessed using chromogenic IHC and the AQUA (Automated Quantitative Analysis) method of QIF. Tumor-infiltrating lymphocytes (TILs) were scored in hematoxylin-eosin slides using current consensus guidelines. The association between PD-L1 protein expression, TILs, and clinicopathological features were determined. MAIN OUTCOMES AND MEASURES PD-L1 expression discordance or heterogeneity using the diaminobenzidine chromogen and QIF was the main outcome measure selected prior to performing the study. RESULTS Using chromogenic IHC, both antibodies showed fair to poor concordance. The PD-L1 antibodies showed poor concordance (Cohen κ range, 0.124-0.340) using conventional chromogenic IHC and showed intra-assay heterogeneity (E1L3N coefficient of variation [CV], 6.75%-75.24%; SP142 CV, 12.17%-109.61%) and significant interassay discordance using QIF (26.6%). Quantitative immunofluorescence showed that PD-L1 expression using both PD-L1 antibodies was heterogeneous. Using QIF, the scores obtained with E1L3N and SP142 for each tumor were significantly different according to nonparametric paired test (P < .001). Assessment of 588 serial section fields of view from whole tissue showed discordant expression at a frequency of 25%. Expression of PD-L1 was correlated with high TILs using both E1L3N (P = .007) and SP142 (P = .02). CONCLUSIONS AND RELEVANCE Objective determination of PD-L1 protein levels in NSCLC reveals heterogeneity within tumors and prominent interassay variability or discordance. This could be due to different antibody affinities, limited specificity, or distinct target epitopes. Efforts to determine the clinical value of these observations are under way.
Collapse
Affiliation(s)
- Joseph McLaughlin
- Department of Medical Oncology, Yale University School of Medicine, New Haven, Connecticut2Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Gang Han
- Department of Biostatistics, Yale University School of Public Health, New Haven, Connecticut
| | - Kurt A Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | | | - Vasiliki Pelekanou
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Jamaal Rehman
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | | | - Roy Herbst
- Department of Medical Oncology, Yale University School of Medicine, New Haven, Connecticut
| | - Patricia LoRusso
- Department of Medical Oncology, Yale University School of Medicine, New Haven, Connecticut
| | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
4
|
Sundar R, Soong R, Cho BC, Brahmer JR, Soo RA. Immunotherapy in the treatment of non-small cell lung cancer. Lung Cancer 2014; 85:101-9. [PMID: 24880938 PMCID: PMC4332778 DOI: 10.1016/j.lungcan.2014.05.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/07/2014] [Indexed: 12/12/2022]
Abstract
Advances in the understanding of the role of the immune system in tumor immunosurveillance have resulted in the recognition that tumors can evade immune destruction via the dysregulation of co-inhibitory or checkpoint signals. This has led to the development of a generation immunotherapeutic agents targeting the immune checkpoint pathway. Recent early phase studies of immune checkpoint modulators, such as CTLA-4, PD-1 and PD-L1 inhibitors in NSCLC have reported promising results with prolonged clinical responses and tolerable toxicity. This article provides an overview of co-stimulatory and inhibitory molecules that regulate the immune response to tumors, recent therapies that have been developed to exploit these interactions and the role of predictive biomarkers in treatment selection.
Collapse
Affiliation(s)
- Raghav Sundar
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Richie Soong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pathology, National University Health System, Singapore
| | - Byoung-Chul Cho
- Division of Medical Oncology, Yonsei Cancer Center, Seoul, South Korea
| | - Julie R Brahmer
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, United States
| | - Ross A Soo
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Surgery, University of Western Australia, Australia.
| |
Collapse
|
5
|
Ctla-4 expression and polymorphisms in lung tissue of patients with diagnosed non-small-cell lung cancer. BIOMED RESEARCH INTERNATIONAL 2013; 2013:576486. [PMID: 23936819 PMCID: PMC3722891 DOI: 10.1155/2013/576486] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 06/19/2013] [Accepted: 06/23/2013] [Indexed: 12/04/2022]
Abstract
Cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) is a potent immunoregulatory molecule that downregulates T-cell activation and thus influences the antitumor immune response. CTLA-4 polymorphisms are associated with various cancers, and CTLA-4 mRNA/protein increased expression is found in several tumor types. However, most of the studies are based on peripheral blood mononuclear cells, and much less is known about the relationship between CTLA-4 expression, especially gene expression, and its polymorphic variants in cancer tissue. In our study we assessed the distribution of CTLA-4 two polymorphisms (+49A/G and −318C/T), using TaqMan probes (rs231775 and rs5742909, resp.), and CTLA-4 gene expression in real-time PCR assay in non-small-cell lung cancer (NSCLC) tissue samples. The increased CTLA-4 expression was observed in the majority of NSCLC patients, and it was significantly correlated with TT genotype (−318C/T) and with tumor size (T2 versus T3 + T4). The presence of G allele and GG genotype in cancer tissue (+49A/G) was significantly associated with the increased NSCLC risk. Additionally, we compared genotype distributions in the corresponding tumor and blood samples and found statistically significant differences. The shift from one genotype in the blood to another in the tumor may confirm the complexity of gene functionality in cancer tissue.
Collapse
|