1
|
Gu Y, Tang YY, Wan JX, Zou JY, Lu CG, Zhu HS, Sheng SY, Wang YF, Liu HC, Yang J, Hong H. Sex difference in the expression of PD-1 of non-small cell lung cancer. Front Immunol 2022; 13:1026214. [PMID: 36341395 PMCID: PMC9632486 DOI: 10.3389/fimmu.2022.1026214] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/27/2022] [Indexed: 12/03/2022] Open
Abstract
Evidence increasingly indicated that lung cancer incidence in female individuals continue to rise, and women have a higher risk to develop adenocarcinoma than men. Male and female individuals differ in their innate and adaptive immune responses, and there are sex differences in response to the PD-1/PD-L1-dependent blocking immunotherapy. Whether the differential expression of PD-1 between genders affect the response to blocking treatment is currently unknown. In this study, we examined sex differences in serum sPD-1, mPD-1 expression on T cells, and sex hormone levels in non-small cell lung cancer (NSCLC) patients. Our results revealed a higher level of sPD-1 and expression of PD-1 on CD4+T cell in female patients than in male patients; we identified that serum sPD-1 level and the expression of mPD-1 on T cells were significantly reduced in NSCLC; we also found that serum testosterone level increased in female patients compared with control subjects and that increased testosterone downregulated the expression of mPD-1 on T cell. These findings provide a better understanding of the differences in PD-1 expression between genders in NSCLC patients and the effect of sex hormones on PD-1 expression and supply evidence for early lung cancer diagnosis and responsiveness to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Yong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ying Y. Tang
- Key Laboratory of Tropical Disease Control of Sun Yat-Sen University, Ministry of Education, The Institute of Immunology of Zhong Shan Medical School, Sun Yat-Sen University, Guangzhou, China
| | - Jian X. Wan
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jian Y. Zou
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Chuan G. Lu
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Cardiothoracic Surgery, Sanya Central Hospital, Sanya, China
| | - Hao S. Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Si Y. Sheng
- Key Laboratory of Tropical Disease Control of Sun Yat-Sen University, Ministry of Education, The Institute of Immunology of Zhong Shan Medical School, Sun Yat-Sen University, Guangzhou, China
- Department of Basic Medicine, Xiangnan University, Chenzhou, China
| | - Yan F. Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hai Ch. Liu
- Key Laboratory of Tropical Disease Control of Sun Yat-Sen University, Ministry of Education, The Institute of Immunology of Zhong Shan Medical School, Sun Yat-Sen University, Guangzhou, China
| | - Jia Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hai Hong
- Key Laboratory of Tropical Disease Control of Sun Yat-Sen University, Ministry of Education, The Institute of Immunology of Zhong Shan Medical School, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Hai Hong,
| |
Collapse
|
2
|
Zhou L, Xu Q, Huang L, Zhan P, Jin J, Ye M, Liu H, Zhang F, Wang Z, Liu J, Chen C, Han H, Zhang Q, Zhu S, Ren J, Lv T, Song Y. Host STING is essential for the efficacy of anti-PD-1 inhibitors in non-small cell lung cancer. Immunol Suppl 2022; 167:495-507. [PMID: 35859099 DOI: 10.1111/imm.13549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/07/2022] [Indexed: 11/28/2022]
Abstract
The stimulator of interferon genes (STING) pathway is important for anticancer immune responses. However, the relative contributions of host and tumor STING in anti-PD-1 (programmed cell death protein 1) inhibitor responses in NSCLC are unknown. STING expression in tumor and blood was associated with anti-PD-1 therapy in non-small cell lung cancer (NSCLC) patients; Moreover, loss of PD-1 inhibitor therapeutic potency was demonstrated in STING KO splenocytes and STING KO mice. STING knockdown in tumor cells had no effect. STING on CD8+ T cells and host cells, not tumor cells, correlated with clinical effect of Anti-PD1 therapy in NSCLC patients. Finally, adoptive transfer of CD8+ T cells restored PD-1 inhibitor anticancer effects. STING in host cells but not in tumor cells, mediates anti-PD-1 inhibitor responses in cancer immunotherapy and could be used to select advantageous NSCLC patients from immunotherapy.
Collapse
Affiliation(s)
- Li Zhou
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Qiuli Xu
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Southeast University, Sch Med, Nanjing 210002, Nanjing, Jiangsu, China
| | - Litang Huang
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Southeast University, Sch Med, Nanjing 210002, Nanjing, Jiangsu, China
| | - Ping Zhan
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Jiajia Jin
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mingxiang Ye
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Hongbing Liu
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Fang Zhang
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Zimu Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Jiaxin Liu
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Cen Chen
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Hedong Han
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Qun Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Suhua Zhu
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Jianan Ren
- Research Institute of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Tangfeng Lv
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Mortensen JB, Monrad I, Enemark MB, Ludvigsen M, Kamper P, Bjerre M, d'Amore F. Soluble programmed cell death protein 1 (sPD-1) and the soluble programmed cell death ligands 1 and 2 (sPD-L1 and sPD-L2) in lymphoid malignancies. Eur J Haematol 2021; 107:81-91. [PMID: 33721375 DOI: 10.1111/ejh.13621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND The programmed cell death protein 1 (PD-1) and its ligand 1 and 2 (PD-L1/PD-L2) regulate the immune system, and the checkpoint pathway can be exploited by malignant cells to evade anti-tumor immune response. Soluble forms (sPD-1/sPD-L1/sPD-L2) exist in the peripheral blood, but their biological and clinical significance is unclear. METHOD Time-resolved immunofluorometric assay (TRIFMA) and enzyme-linked immunosorbent assay (ELISA) were used to measure sPD-1, sPD-L1, and sPD-L2 levels in serum from 131 lymphoma patients and 22 healthy individuals. RESULTS Patients had higher sPD-1 and sPD-L2 levels than healthy individuals. In diffuse large B-cell lymphoma, patients with high International Prognostic Index score had higher sPD-1 levels and sPD-L2 levels correlated with subtype according to cell of origin. Compared to other lymphoma types, follicular lymphoma displayed higher sPD-1 and lower sPD-L1 levels along with lower ligand/receptor ratios. CONCLUSION This is the first study to simultaneously characterize pretherapeutic sPD-1, sPD-L1, and sPD-L2 in a variety of lymphoma subtypes. The relation between higher sPD-1 levels and adverse prognostic factors suggests a possible biological role and potential clinical usefulness of sPD-1. Moreover, the reverse expression pattern in follicular lymphoma and T-cell lymphoma/leukemia may reflect biological information relevant for immunotherapy targeting the PD-1 pathway.
Collapse
Affiliation(s)
- Julie B Mortensen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Ida Monrad
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Marie B Enemark
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Maja Ludvigsen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Peter Kamper
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Mette Bjerre
- Medical/SDCA Research Laboratory, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Francesco d'Amore
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
4
|
Goto M, Chamoto K, Higuchi K, Yamashita S, Noda K, Iino T, Miura M, Yamasaki T, Ogawa O, Sonobe M, Date H, Hamanishi J, Mandai M, Tanaka Y, Chikuma S, Hatae R, Muto M, Minamiguchi S, Minato N, Honjo T. Analytical performance of a new automated chemiluminescent magnetic immunoassays for soluble PD-1, PD-L1, and CTLA-4 in human plasma. Sci Rep 2019; 9:10144. [PMID: 31300681 PMCID: PMC6626008 DOI: 10.1038/s41598-019-46548-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/29/2019] [Indexed: 12/25/2022] Open
Abstract
Current clinically approved biomarkers for the PD-1 blockade cancer immunotherapy are based entirely on the properties of tumour cells. With increasing awareness of clinical responses, more precise biomarkers for the efficacy are required based on immune properties. In particular, expression levels of immune checkpoint-associated molecules such as PD-1, PD-L1, and CTLA-4 would be critical to evaluate the immune state of individuals. Although quantification of their soluble form leased from the membrane will provide quick evaluation of patients’ immune status, available methods such as enzyme-linked immunosorbent assays to measure these soluble factors have limitations in sensitivity and reproducibility for clinical use. To overcome these problems, we developed a rapid and sensitive immunoassay system based on chemiluminescent magnetic technology. The system is fully automated, providing high reproducibility. Application of this system to plasma of patients with several types of tumours demonstrated that soluble PD-1, PD-L1, and CTLA-4 levels were increased compared to those of healthy controls and varied among tumour types. The sensitivity and detection range were sufficient for evaluating plasma concentrations before and after the surgical ablation of cancers. Therefore, our newly developed system shows potential for accurate detection of soluble PD-1, PD-L1, and CTLA-4 levels in the clinical practice.
Collapse
Affiliation(s)
- Megumi Goto
- Clinical Innovation, Sysmex Corporation, Hyogo, Japan
| | - Kenji Chamoto
- Department of Immunology and Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiko Higuchi
- Clinical Innovation, Sysmex Corporation, Hyogo, Japan
| | - Saya Yamashita
- Technology Development, Sysmex Corporation, Hyogo, Japan
| | - Kenta Noda
- Technology Development, Sysmex Corporation, Hyogo, Japan
| | - Takuya Iino
- Central Research Laboratories, Sysmex Corporation, Hyogo, Japan
| | - Masahiro Miura
- Central Research Laboratories, Sysmex Corporation, Hyogo, Japan
| | - Toshinari Yamasaki
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Osamu Ogawa
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Makoto Sonobe
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshimasa Tanaka
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Shunsuke Chikuma
- Department of Microbiology and Immunology, Keio University School of Medicine, Kyoto, Japan
| | - Ryusuke Hatae
- Department of Immunology and Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sachiko Minamiguchi
- Department of Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nagahiro Minato
- DSK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tasuku Honjo
- Kyoto University Institute for Advanced Study, Kyoto, Japan.
| |
Collapse
|
5
|
Critical reagent screening and characterization: benefits and approaches for protein biomarker assays by hybrid LC–MS. Bioanalysis 2019; 11:785-795. [DOI: 10.4155/bio-2018-0277] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In recent years, hybrid ligand-binding assays (LBAs)/LC–MS assays have been increasingly used for quantitation of protein biomarkers in biological matrices. However, unlike in LBAs where the importance of critical reagent screening and characterization is well understood and widely reported, benefits of well-characterized hybrid LC–MS assay reagents are frequently underestimated. Two groups of analyte-specific reagents, binding reagents and assay calibrators, are considered the critical reagents for biomarker assays. In this article, we summarize the similarities and differences of critical reagents used in LBAs and hybrid LC–MS assays, overview the benefits and approaches of critical reagent screening, characterization, antibody conjugation and discuss bioanalytical considerations in hybrid LC–MS assay development for robust measurements of protein biomarkers in biological matrices.
Collapse
|
6
|
Mizutani K, Horie K, Kato T, Nakane K, Kawakami K, Fujita Y, Ito M. Serum PD-1 levels measured by ELISA using Nivolumab increased in advanced RCC patients: novel approach to develop companion diagnostics for antibody therapy. J Cancer Res Clin Oncol 2018; 145:1661-1663. [PMID: 30515569 PMCID: PMC6527537 DOI: 10.1007/s00432-018-2806-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 11/27/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Kosuke Mizutani
- Department of Urology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan.
| | - Kengo Horie
- Department of Urology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan
| | - Taku Kato
- Department of Urology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan
| | - Keita Nakane
- Department of Urology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Gifu, 501-1194, Japan
| | - Kyojiro Kawakami
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Yasunori Fujita
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Masafumi Ito
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan.
| |
Collapse
|
7
|
Cowan KJ, Amaravadi L, Cameron MJ, Fink D, Jani D, Kamat M, King L, Neely RJ, Ni Y, Rhyne P, Riffon R, Zhu Y. Recommendations for Selection and Characterization of Protein Biomarker Assay Calibrator Material. AAPS JOURNAL 2017; 19:1550-1563. [DOI: 10.1208/s12248-017-0146-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/20/2017] [Indexed: 11/30/2022]
|
8
|
Cole EL, Kim J, Donnelly DJ, Smith RA, Cohen D, Lafont V, Morin PE, Huang RYC, Chow PL, Hayes W, Bonacorsi S. Radiosynthesis and preclinical PET evaluation of 89Zr-nivolumab (BMS-936558) in healthy non-human primates. Bioorg Med Chem 2017; 25:5407-5414. [PMID: 28803798 DOI: 10.1016/j.bmc.2017.07.066] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 07/25/2017] [Accepted: 07/31/2017] [Indexed: 11/30/2022]
Abstract
Cancer immunotherapy, unlike traditional cytotoxic chemotherapeutic treatments, engages the immune system to identify cancer cells and stimulate immune responses. The Programmed Death-1 (PD-1) protein is an immunoinhibitory receptor expressed by activated cytotoxic T-lymphocytes (CTL) that seek out and destroy cancer cells. Multiple cancer types express and upregulate the Programmed Death-Ligand 1 (PD-L1) and 2 (PD-L2) which bind to PD-1 as an immune escape mechanism. Nivolumab is a fully human IgG4 anti-PD-1 monoclonal antibody (mAb) approved for treatment of multiple cancer types. This study reports the preparation and in vivo evaluation of 89Zr labeled nivolumab in healthy non-human primates (NHP) as a preliminary study of biodistribution and clearance. The radiochemical and in vivo stabilities of the 89Zr complex were shown to be acceptable for imaging. Three naïve NHPs were intravenously injected with tracer only or tracer co-injected with nivolumab followed by co-registered by positron emission tomography (PET) and magnetic resonance imaging (MRI), acquired for eight days following injection. Image-derived standardized uptake values (SUV) were quantified by region of interest (ROI) analysis. Radioactivity in the spleen was significantly reduced by addition of excess nivolumab compared to the tracer only study at all imaging time points. Liver uptake of the radiotracer was consistent as a clearance organ with minimal signal from other tissues: lung, muscle, brain, heart, and kidney. The results indicate specific biodistribution to the spleen, which can be blocked by co-administration of excess nivolumab. Distribution to other organs is consistent with elimination pathways of antibodies, with primary clearance through the liver.
Collapse
Affiliation(s)
- Erin L Cole
- Radiochemistry Group, Bristol-Myers Squibb Company, Princeton, NJ, USA.
| | - Joonyoung Kim
- Imaging Group, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - David J Donnelly
- Radiochemistry Group, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - R Adam Smith
- Imaging Group, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Daniel Cohen
- Protein Science Group, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Virginie Lafont
- Protein Science Group, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Paul E Morin
- Protein Science Group, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Richard Y-C Huang
- Bioanalytical and Discovery Analytical Sciences, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Patrick L Chow
- Imaging Group, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Wendy Hayes
- Imaging Group, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Samuel Bonacorsi
- Radiochemistry Group, Bristol-Myers Squibb Company, Princeton, NJ, USA
| |
Collapse
|
9
|
Fit-for-purpose biomarker immunoassay qualification and validation: three case studies. Bioanalysis 2016; 8:2329-2340. [PMID: 27712082 DOI: 10.4155/bio-2016-0184] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
AIM To improve on the efficiency of biomarker assay readiness, and for reliable biomarker data to support three drug programs, we implemented a fit-for-purpose approach, qualifying two biomarker assays and validating a third. Results/methodology: The qualification strategy and selection of experiments for two exploratory biomarkers (CXCL1, CCL19) was determined by the intended use of the biomarker data. The third biomarker, IL-6, was validated as the data would be used in monitoring patient safety during dose-escalation studies in a Phase I trial. All three assays passed a priori acceptance criteria. CONCLUSION These assays highlight strategies and methodologies for a fit-for-purpose approach. Minimum qualification, full qualification and validation were chosen and supported programs at different stages of drug development.
Collapse
|
10
|
Sorensen SF, Demuth C, Weber B, Sorensen BS, Meldgaard P. Increase in soluble PD-1 is associated with prolonged survival in patients with advanced EGFR-mutated non-small cell lung cancer treated with erlotinib. Lung Cancer 2016; 100:77-84. [PMID: 27597284 DOI: 10.1016/j.lungcan.2016.08.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/23/2016] [Accepted: 08/06/2016] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The central immune co-inhibitory programmed cell death receptor/ligand 1 (PD-1/PD-L1) pathway plays a key role in tumor immune evasion in non-small cell lung cancer (NSCLC). Soluble PD-1 (sPD-1) can be detected in the blood, and preclinical evidence suggests that sPD-1 blocks PD-1/-L1 interaction and improves anti-tumor immunity. The present study compares the concentration of sPD-1 in the serum of advanced NSCLC patients with Epidermal Growth Factor Receptor (EGFR) mutation prior to erlotinib treatment and at the time of progression and correlates these results to patient outcome. MATERIALS AND METHODS Blood samples from 38 patients with EGFR-mutated advanced NSCLC treated with erlotinib were analyzed for sPD-1 by sandwich ELISA. EGFR mutational status was assessed in circulating tumor DNA (ctDNA) and tumor biopsies. RESULTS sPD-1 could be detected in 21% of patients prior to erlotinib treatment, and at disease progression in 37% (p=0.015). An increase in sPD-1 during erlotinib therapy was found in 34%, a decrease in 8% and no change in 58% of patients. An increase in sPD-1 during treatment was associated with prolonged progression-free (adjusted HR 0.32, p=0.013) and overall survival (adjusted HR 0.33, p=0.006), but not associated with the emergence of EGFR T790M mutation in ctDNA at progression or any clinicopathological factors. CONCLUSION Patients with an increase in sPD-1 during erlotinib treatment have a more favorable outcome. Our results emphasize the vast clinical impact of the PD-1/PD-L1 axis, and support the existing preclinical evidence in the bioactive function of sPD-1.
Collapse
Affiliation(s)
| | - Christina Demuth
- Department of Clinical Biochemistry, Aarhus University Hospital, Noerrebrogade 44, 8000 Aarhus C, Denmark
| | - Britta Weber
- Department of Oncology, Aarhus University Hospital, Noerrebrogade 44, 8000 Aarhus C, Denmark
| | - Boe Sandahl Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Noerrebrogade 44, 8000 Aarhus C, Denmark
| | - Peter Meldgaard
- Department of Oncology, Aarhus University Hospital, Noerrebrogade 44, 8000 Aarhus C, Denmark
| |
Collapse
|
11
|
Implementing fit-for-purpose biomarker assay approaches: a bioanalytical perspective. Bioanalysis 2016; 8:1221-3. [DOI: 10.4155/bio-2016-0070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|