1
|
Pitts SC, Schlom J, Donahue RN. Soluble immune checkpoints: implications for cancer prognosis and response to immune checkpoint therapy and conventional therapies. J Exp Clin Cancer Res 2024; 43:155. [PMID: 38822401 PMCID: PMC11141022 DOI: 10.1186/s13046-024-03074-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024] Open
Abstract
Longitudinal sampling of tumor tissue from patients with solid cancers, aside from melanoma and a few other cases, is often unfeasible, and thus may not capture the plasticity of interactions between the tumor and immune system under selective pressure of a given therapy. Peripheral blood analyses provide salient information about the human peripheral immunome while offering technical and practical advantages over traditional tumor biopsies, and should be utilized where possible alongside interrogation of the tumor. Some common blood-based biomarkers used to study the immune response include immune cell subsets, circulating tumor DNA, and protein analytes such as cytokines. With the recent explosion of immune checkpoint inhibitors (ICI) as a modality of treatment in multiple cancer types, soluble immune checkpoints have become a relevant area of investigation for peripheral immune-based biomarkers. However, the exact functions of soluble immune checkpoints and their roles in cancer for the most part remain unclear. This review discusses current literature on the production, function, and expression of nine soluble immune checkpoints - sPD-L1, sPD-1, sCTLA4, sCD80, sTIM3, sLAG3, sB7-H3, sBTLA, and sHVEM - in patients with solid tumors, and explores their role as biomarkers of response to ICI as well as to conventional therapies (chemotherapy, radiotherapy, targeted therapy, and surgery) in cancer patients.
Collapse
Affiliation(s)
- Stephanie C Pitts
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey Schlom
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Renee N Donahue
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Kurosaki T, Chamoto K, Suzuki S, Kanemura H, Mitani S, Tanaka K, Kawakami H, Kishimoto Y, Haku Y, Ito K, Sato T, Suminaka C, Yamaki M, Chiba Y, Yaguchi T, Omori K, Kobayashi T, Nakagawa K, Honjo T, Hayashi H. The combination of soluble forms of PD-1 and PD-L1 as a predictive marker of PD-1 blockade in patients with advanced cancers: a multicenter retrospective study. Front Immunol 2023; 14:1325462. [PMID: 38149256 PMCID: PMC10750355 DOI: 10.3389/fimmu.2023.1325462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 11/28/2023] [Indexed: 12/28/2023] Open
Abstract
Introduction The clinical relevance of soluble forms of programmed cell death-1 (sPD-1) and programmed cell death-ligand 1 (sPD-L1) remains unclear. We here investigated the relation between the efficacy of PD-1 blockade and pretreatment plasma levels of sPD-1 and sPD-L1 across a broad range of cancer types. Methods We retrospectively analyzed clinical data from 171 patients with advanced solid tumors who received nivolumab or pembrolizumab monotherapy regardless of treatment line. The concentrations of sPD-1 and sPD-L1 were measured with a fully automated immunoassay (HISCL system). Results The study subjects comprised patients with head and neck cancer (n = 50), urothelial cancer (n = 42), renal cell cancer (n = 37), gastric cancer (n = 20), esophageal cancer (n = 10), malignant pleural mesothelioma (n = 6), or microsatellite instability-high tumors (n = 6). High or low levels of sPD-1 or sPD-L1 were not significantly associated with progression-free survival (PFS) or overall survival (OS) for PD-1 blockade in the entire study population. Comparison of treatment outcomes according to combinations of high or low sPD-1 and sPD-L1 levels, however, revealed that patients with low sPD-1 and high sPD-L1 concentrations had a significantly poorer PFS (HR of 1.79 [95% CI, 1.13-2.83], p = 0.01) and a tendency toward poorer OS (HR of 1.70 [95% CI, 0.99-2.91], p = 0.05) compared with all other patients. Conclusion Our findings suggest that the combination of low sPD-1 and high sPD-L1 levels is a potential negative biomarker for PD-1 blockade therapy.
Collapse
Affiliation(s)
- Takashi Kurosaki
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kenji Chamoto
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Immuno-Oncology PDT, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shinichiro Suzuki
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Hiroaki Kanemura
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Seiichiro Mitani
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kaoru Tanaka
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Hisato Kawakami
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yo Kishimoto
- Department of Otolaryngology–Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuharu Haku
- Department of Otolaryngology–Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Katsuhiro Ito
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshiyuki Sato
- Central Research Laboratories, Sysmex Corporation, Kobe, Japan
| | | | - Mami Yamaki
- Business Strategy Development, Sysmex Corporation, Kobe, Japan
| | - Yasutaka Chiba
- Clinical Research Center, Kindai University Hospital, Osaka-Sayama, Japan
| | - Tomonori Yaguchi
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Immuno-Oncology PDT, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koichi Omori
- Department of Otolaryngology–Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Kobayashi
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Tasuku Honjo
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| |
Collapse
|
3
|
Mariotti FR, Ingegnere T, Landolina N, Vacca P, Munari E, Moretta L. Analysis of the mechanisms regulating soluble PD-1 production and function in human NK cells. Front Immunol 2023; 14:1229341. [PMID: 37638041 PMCID: PMC10449250 DOI: 10.3389/fimmu.2023.1229341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
NK cells represent important effectors that play a major role in innate defences against pathogens and display potent cytolytic activity against tumor cells. An array of surface receptors finely regulate their function and inhibitory checkpoints, such as PD-1, can dampen the immune response inducing an immunosuppressive state. Indeed, PD-1 expression in human NK cells correlated with impaired effector function and tumor immune evasion. Importantly, blockade of the PD-1/PD-L1 axis has been shown to reverse NK cell exhaustion and increase their cytotoxicity. Recently, soluble counterparts of checkpoint receptors, such as soluble PD-1 (sPD-1), are rising high interest due to their biological activity and ability to modulate immune responses. It has been widely demonstrated that sPD-1 can modulate T cell effector functions and tumor growth. Tumor-infiltrating T cells are considered the main source of circulating sPD-1. In addition, recently, also stimulated macrophages have been demonstrated to release sPD-1. However, no data are present on the role of sPD-1 in the context of other innate immune cell subsets and therefore this study is aimed to unveil the effect of sPD-1 on human NK cell function. We produced the recombinant sPD-1 protein and demonstrated that it binds PD-L1 and that its presence results in increased NK cell cytotoxicity. Notably, we also identified a pathway regulating endogenous sPD-1 synthesis and release in human NK cells. Secreted endogenous sPD-1, retained its biological function and could modulate NK cell effector function. Overall, these data reveal a pivotal role of sPD-1 in regulating NK-mediated innate immune responses.
Collapse
Affiliation(s)
| | - Tiziano Ingegnere
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Nadine Landolina
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Lymphoid Cells of Innate Immunity Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paola Vacca
- Lymphoid Cells of Innate Immunity Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Enrico Munari
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
4
|
Maebele LT, Mulaudzi TV, Yasasve M, Dlamini Z, Damane BP. Immunomodulatory Gene-Splicing Dysregulation in Tumorigenesis: Unmasking the Complexity. Molecules 2023; 28:5984. [PMID: 37630236 PMCID: PMC10458946 DOI: 10.3390/molecules28165984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer is a global health concern with rising incidence, morbidity, and mortality. The interaction between the tumor and immune cells within the tumor microenvironment is facilitated by signaling pathways driven by immunomodulatory proteins. Alternative splicing regulates the production of multiple immunomodulatory proteins with diverse functionality from a single mRNA transcript. Splicing factors are pivotal in modulating alternative splicing processes but are also subject to regulation. The dysregulation of alternative splicing may result from splicing factor (SF) abnormal expression levels and mutations in the cis and trans-acting elements and small nuclear RNA (snRNA) molecules. Aberrant splicing may generate abnormal mRNA transcripts encoding isoforms with altered functions that contribute to tumorigenesis or cancer progression. This review uncovers the complexity of immunomodulatory genes splicing dysregulation in oncogenesis. Identifying specific immunomodulatory splicing isoforms that contribute to cancer could be utilized to improve current immunotherapeutic drugs or develop novel therapeutic interventions for cancer.
Collapse
Affiliation(s)
| | - Thanyani Victor Mulaudzi
- Department of Surgery, Steve Biko Academic Hospital, University of Pretoria, Hatfield 0028, South Africa
| | - Madhavan Yasasve
- Department of Oral Medicine and Radiology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa
| | - Botle Precious Damane
- Department of Surgery, Steve Biko Academic Hospital, University of Pretoria, Hatfield 0028, South Africa
| |
Collapse
|
5
|
Shi W, Zhang Y, Hao C, Guo X, Yang Q, Du J, Hou Y, Cao G, Li J, Wang H, Fang W. The significance of PD-1/PD-L1 imbalance in ulcerative colitis. PeerJ 2023; 11:e15481. [PMID: 37273534 PMCID: PMC10239227 DOI: 10.7717/peerj.15481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 05/09/2023] [Indexed: 06/06/2023] Open
Abstract
Objectives To investigate the expression and significance of programmed cell death protein 1 (PD-1) and programmed cell death ligand-1 (PD-L1) in the mucosal tissues and peripheral blood of patients with ulcerative colitis (UC). Methods Eighty patients with UC were recruited from January 2021 to August 2022 from the Shanxi Province People's Hospital. PD-1 and PD-L1 expression was assessed by immunohistochemistry in mucosal tissues. An enzyme-linked immunosorbent assay was used to measure soluble PD-1 and PD-L1 levels in peripheral blood serum, and the membrane-bound forms of PD-1 (mPD-1), (T-helper cell) Th1 and Th17, in peripheral blood were determined by flow cytometry. Result PD-1 expression was observed only in the monocytes of the mucosal lamina propria of UC patients, while PD-L1 was mainly located in both epithelial cells and monocytes on the cell membrane. The expression level of PD-1/PD-L1 in the monocytes and epithelial cells of mucosal lamina propria increased with disease activity (P < 0.05). The percentages of PD-1/T and PD-1/CD4+T in the peripheral blood of moderate UC patients (PD-1/T 12.83 ± 6.15% and PD-1/CD4+T 19.67 ± 9.95%) and severe UC patients (PD-1/T 14.29 ± 5.71% and PD-1/CD4+T 21.63 ± 11.44%) were higher than in mild UC patients (PD-1/T 8.17 ± 2.80% and PD-1/CD4+T 12.44 ± 4.73%; P < 0.05). There were no significant differences in PD-1/CD8+T cells between mild and severe UC patients (P > 0.05). There was a statistically significant difference in the expression level of sPD-L1 between the UC groups and healthy controls, and the expression level of sPD-L1 increased with disease severity (P < 0.05); however, there was no statistically significant difference in sPD-1 expression levels between the UC groups and healthy controls (P > 0.05). The correlation coefficients between Th1 and sPD-L1, PD-1/T, PD-1/CD4+T and PD-1/CD8+T were 0.427, 0.589, 0.486, and 0.329, respectively (P < 0.001). The correlation coefficients between Th17 and sPD-L1, PD-1/T, PD-1/CD4+T and PD-1/CD8+T were 0.323, 0.452, 0.320, and 0.250, respectively (P < 0.05). Conclusion The expression level of PD-1/PD-L1 was correlated with UC disease activity, and two forms of PD-1 and PD-L1 may be used as a potential marker for predicting UC and assessing disease progression in UC patients. PD-1/PD-L1 imbalance was a significant phenomenon of UC immune dysfunction. Future research should focus on two forms of PD-1/PD-L1 signaling molecules to better understand the pathogenesis of UC and to identify potential drug therapies.
Collapse
Affiliation(s)
- Wei Shi
- Department of Clinical Laboratory, Shanxi Provincial People’s Hospital, Taiyuan, China
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Yu Zhang
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Chonghua Hao
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Xiaofeng Guo
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Qin Yang
- Department of Pathology, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Junfang Du
- Department of Clinical Laboratory, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Yabin Hou
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Gaigai Cao
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Jingru Li
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Haijiao Wang
- Shanxi Center for Disease Control and Prevention, Taiyuan, China
| | - Wei Fang
- Department of Clinical Laboratory, Shanxi Provincial People’s Hospital, Taiyuan, China
| |
Collapse
|
6
|
Saadi W, Fatmi A, Pallardó FV, García-Giménez JL, Mena-Molla S. Long Non-Coding RNAs as Epigenetic Regulators of Immune Checkpoints in Cancer Immunity. Cancers (Basel) 2022; 15:cancers15010184. [PMID: 36612180 PMCID: PMC9819025 DOI: 10.3390/cancers15010184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/19/2022] [Accepted: 12/25/2022] [Indexed: 12/30/2022] Open
Abstract
In recent years, cancer treatment has undergone significant changes, predominantly in the shift towards immunotherapeutic strategies using immune checkpoint inhibitors. Despite the clinical efficacy of many of these inhibitors, the overall response rate remains modest, and immunotherapies for many cancers have proved ineffective, highlighting the importance of knowing the tumor microenvironment and heterogeneity of each malignancy in patients. Long non-coding RNAs (lncRNAs) have attracted increasing attention for their ability to control various biological processes by targeting different molecular pathways. Some lncRNAs have a regulatory role in immune checkpoints, suggesting they might be utilized as a target for immune checkpoint treatment. The focus of this review is to describe relevant lncRNAs and their targets and functions to understand key regulatory mechanisms that may contribute in regulating immune checkpoints. We also provide the state of the art on super-enhancers lncRNAs (selncRNAs) and circular RNAs (circRNAs), which have recently been reported as modulators of immune checkpoint molecules within the framework of human cancer. Other feasible mechanisms of interaction between lncRNAs and immune checkpoints are also reported, along with the use of miRNAs and circRNAs, in generating new tumor immune microenvironments, which can further help avoid tumor evasion.
Collapse
Affiliation(s)
- Wiam Saadi
- Department of Biology, Faculty of Nature, Life and Earth Sciences, University of Djillali Bounaama, Khemis Miliana 44225, Algeria
- Correspondence: (W.S.); (S.M.-M.)
| | - Ahlam Fatmi
- INCLIVA Health Research Institute, INCLIVA, 46010 Valencia, Spain
| | - Federico V. Pallardó
- INCLIVA Health Research Institute, INCLIVA, 46010 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, 46010 Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
| | - José Luis García-Giménez
- INCLIVA Health Research Institute, INCLIVA, 46010 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, 46010 Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
| | - Salvador Mena-Molla
- INCLIVA Health Research Institute, INCLIVA, 46010 Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
- Correspondence: (W.S.); (S.M.-M.)
| |
Collapse
|
7
|
Abstract
Gene therapy is a powerful biological tool that is reshaping therapeutic landscapes for several diseases. Researchers are using both non-viral and viral-based gene therapy methods with success in the lab and the clinic. In the cancer biology field, gene therapies are expanding treatment options and the possibility of favorable outcomes for patients. While cellular immunotherapies and oncolytic virotherapies have paved the way in cancer treatments based on genetic engineering, recombinant adeno-associated virus (rAAV), a viral-based module, is also emerging as a potential cancer therapeutic through its malleability, specificity, and broad application to common as well as rare tumor types, tumor microenvironments, and metastatic disease. A wide range of AAV serotypes, promoters, and transgenes have been successful at reducing tumor growth and burden in preclinical studies, suggesting more groundbreaking advances using rAAVs in cancer are on the horizon.
Collapse
Affiliation(s)
- Patrick L. Mulcrone
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University, Indianapolis, IN 46202, USA
| | - Roland W. Herzog
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Weidong Xiao
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| |
Collapse
|
8
|
Chen W, Huang Y, Pan W, Xu M, Chen L. Strategies for developing PD-1 inhibitors and future directions. Biochem Pharmacol 2022; 202:115113. [DOI: 10.1016/j.bcp.2022.115113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023]
|
9
|
Yuan J, Yang J, Wang R, Hao H, Li J. LncRNA NEAT1 regulate diffuse large B-cell lymphoma by targeting miR-495-3p/PD-L1 axis. Immunopharmacol Immunotoxicol 2022; 44:429-436. [PMID: 35352617 DOI: 10.1080/08923973.2022.2052896] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is a common lymphatic tumor in clinic. LncRNAs were reported to play a regulatory role in many cancers, including DLBCL. This study focused on the roles of NEAT1 in DLBCL. METHODS Real-time quantitative polymerase chain reaction (RT-qPCR) was carried out to detect mRNA expression. StarBase as well as TargetScan was used to predict targeting relationships, which was confirmed by the Dual Luciferase Reporter Assay and RNA pull-down assay. Cell Counting Kit 8 (CCK-8) were applied to measure cell viability. Flow cytometry assay was applied to detect cell apoptosis. Western blotting assay was conduct to determine protein expression. Lactate dehydrogenase (LDH) release assay were applied to evaluated cell cytotoxicity. RESULTS NEAT1 was overexpressed in DLBCL patients. Knockdown of NEAT1 reduced the viability while enhanced the apoptosis of tumor cells. However, overexpression of NEAT1 exhibited an opposite effect. miR-495-3p was a target of NEAT1 and was decreased in DLBCL cells. However, inhibiting miR-495-3p reversed the effect of NEAT1 knock-down on DLBCL cells and induced the malignant behaviors of DLBCL cells. Moreover, NEAT1 functioned as a sponge of miR-495-3p to upregulate PD-L1. CONCLUSION Our study demonstrated that a NEAT1/miR-495-3p/PD-L1 axis regulated the development of DLBCL. Therefore, NEAT1 may be a potential biomarker for DLBCL.
Collapse
Affiliation(s)
- Jun Yuan
- Blood Specialty, Hebei General Hospital, Shijiazhuang, China
| | - Jie Yang
- Blood Specialty, Hebei General Hospital, Shijiazhuang, China
| | - Ruicang Wang
- Blood Specialty, Hebei General Hospital, Shijiazhuang, China
| | - Hongling Hao
- Blood Specialty, Hebei General Hospital, Shijiazhuang, China
| | - Jie Li
- Blood Specialty, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
10
|
Niu M, Liu Y, Yi M, Jiao D, Wu K. Biological Characteristics and Clinical Significance of Soluble PD-1/PD-L1 and Exosomal PD-L1 in Cancer. Front Immunol 2022; 13:827921. [PMID: 35386715 PMCID: PMC8977417 DOI: 10.3389/fimmu.2022.827921] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
The immune checkpoint pathway consisting of the cell membrane-bound molecule programmed death protein 1 (PD-1) and its ligand PD-L1 has been found to mediate negative regulatory signals that effectively inhibit T-cell proliferation and function and impair antitumor immune responses. Considerable evidence suggests that the PD-1/PD-L1 pathway is responsible for tumor immune tolerance and immune escape. Blockage of this pathway has been found to reverse T lymphocyte depletion and restore antitumor immunity. Antagonists targeting this pathway have shown significant clinical activity in specific cancer types. Although originally identified as membrane-type molecules, several other forms of PD-1/PD-L1 have been detected in the blood of cancer patients, including soluble PD-1/PD-L1 (sPD-1/sPD-L1) and exosomal PD-L1 (exoPD-L1), increasing the composition and functional complications of the PD-1/PD-L1 signaling pathway. For example, sPD-1 has been shown to block the PD-1/PD-L immunosuppressive pathway by binding to PD-L1 and PD-L2, whereas the role of sPD-L1 and its mechanism of action in cancer remain unclear. In addition, many studies have investigated the roles of exoPD-L1 in immunosuppression, as a biomarker for tumor progression and as a predictive biomarker for response to immunotherapy. This review describes the molecular mechanisms underlying the generation of sPD-1/sPD-L1 and exoPD-L1, along with their biological activities and methods of detection. In addition, this review discusses the clinical importance of sPD-1/sPD-L1 and exoPD-L1 in cancer, including their predictive and prognostic roles and the effects of treatments that target these molecules.
Collapse
Affiliation(s)
- Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiming Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dechao Jiao
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Kongming Wu, ; Dechao Jiao,
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Kongming Wu, ; Dechao Jiao,
| |
Collapse
|
11
|
Xing Y, Liu J, Luo J, Ming T, Yang G, Sun S, Xu S, Li X, He E, Kong F, Yan S, Yang Y, Cai X. A Dual-Channel Intelligent Point-of-Care Testing System for Soluble Programmed Death-1 and Programmed Death-Ligand 1 Detection Based on Folding Paper-Based Immunosensors. ACS Sens 2022; 7:584-592. [PMID: 35060694 DOI: 10.1021/acssensors.1c02486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Both programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) are important proteins in cancer immunotherapy. Soluble forms (sPD-1 and sPD-L1) have potential for determining treatment and prognosis monitoring. However, there is a lack of detection methods for point-of-care testing (POCT) of these two proteins, so a low-cost rapid detection platform is urgently needed. To solve this problem, a dual-channel electrochemical platform, including a folding paper-based immunosensor and a POCT system for rapid simultaneous detection of these two proteins was designed and fabricated. The immunosensor consists of a three-electrode system and a reaction cell. The surface of the working electrode was modified with nanocomposites synthesized from amine-functionalized single-walled carbon nanotubes, new methylene blue, and gold nanoparticles. Antibodies to sPD-1 and sPD-L1 were also immobilized on the working electrode surface. A differential pulse voltammetry electrochemical method was adopted. The immunosensor was able to detect sPD-1 and sPD-L1 in the ranges of 50 pg/mL to 50 ng/mL and 5 pg/mL to 5 ng/mL, respectively. The limits of detection were 10 and 5 pg/mL. Using this detection platform, sPD-1 and sPD-L1 in plasma were detected by both enzyme-linked immunosorbent assay and the immunosensor, which has good application potential.
Collapse
Affiliation(s)
- Yu Xing
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, P.R. China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Juntao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, P.R. China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Jinping Luo
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, P.R. China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Tao Ming
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, P.R. China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Gucheng Yang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, P.R. China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Shuai Sun
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, P.R. China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Shengwei Xu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, P.R. China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Xinrong Li
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, P.R. China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Enhui He
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, P.R. China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Fanli Kong
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, P.R. China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Shi Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing 100142, P.R. China
| | - Yue Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing 100142, P.R. China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, P.R. China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| |
Collapse
|
12
|
Li W, Syed F, Yu R, Yang J, Xia Y, Relich RF, Russell PM, Zhang S, Khalili M, Huang L, Kacena MA, Zheng X, Yu Q. Soluble Immune Checkpoints Are Dysregulated in COVID-19 and Heavy Alcohol Users With HIV Infection. Front Immunol 2022; 13:833310. [PMID: 35281051 PMCID: PMC8904355 DOI: 10.3389/fimmu.2022.833310] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/28/2022] [Indexed: 01/08/2023] Open
Abstract
Immune checkpoints (ICPs) consist of paired receptor-ligand molecules that exert inhibitory or stimulatory effects on immune defense, surveillance, regulation, and self-tolerance. ICPs exist in both membrane and soluble forms in vivo and in vitro. Imbalances between inhibitory and stimulatory membrane-bound ICPs (mICPs) in malignant cells and immune cells in the tumor immune microenvironment (TIME) have been well documented. Blockades of inhibitory mICPs have emerged as an immense breakthrough in cancer therapeutics. However, the origin, structure, production regulation, and biological significance of soluble ICPs (sICPs) in health and disease largely remains elusive. Soluble ICPs can be generated through either alternative mRNA splicing and secretion or protease-mediated shedding from mICPs. Since sICPs are found in the bloodstream, they likely form a circulating immune regulatory system. In fact, there is increasing evidence that sICPs exhibit biological functions including (1) regulation of antibacterial immunity, (2) interaction with their mICP compartments to positively or negatively regulate immune responses, and (3) competition with their mICP compartments for binding to the ICP blocking antibodies, thereby reducing the efficacy of ICP blockade therapies. Here, we summarize current data of sICPs in cancer and infectious diseases. We particularly focus on sICPs in COVID-19 and HIV infection as they are the two ongoing global pandemics and have created the world's most serious public health challenges. A "storm" of sICPs occurs in the peripheral circulation of COVID-19 patients and is associated with the severity of COVID-19. Similarly, sICPs are highly dysregulated in people living with HIV (PLHIV) and some sICPs remain dysregulated in PLHIV on antiretroviral therapy (ART), indicating these sICPs may serve as biomarkers of incomplete immune reconstitution in PLHIV on ART. We reveal that HIV infection in the setting of alcohol misuse exacerbates sICP dysregulation as PLHIV with heavy alcohol consumption have significantly elevated plasma levels of many sICPs. Thus, both stimulatory and inhibitory sICPs are present in the bloodstream of healthy people and their balance can be disrupted under pathophysiological conditions such as cancer, COVID-19, HIV infection, and alcohol misuse. There is an urgent need to study the role of sICPs in immune regulation in health and disease.
Collapse
Affiliation(s)
- Wei Li
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Fahim Syed
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Richard Yu
- Department of Internal Medicine, School of Medicine, University of Nevada, Reno, NV, United States
| | - Jing Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ying Xia
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
- School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ryan F. Relich
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Patrick M. Russell
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Shanxiang Zhang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Mandana Khalili
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Laurence Huang
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Melissa A. Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiaoqun Zheng
- School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, China
- Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qigui Yu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
13
|
Doornebal EJ, Harris N, Riva A, Jagatia R, Pizanias M, Prachalias A, Menon K, Preziosi M, Zamalloa A, Miquel R, Zen Y, Orford MR, Eaton S, Heaton N, Ramage J, Palma E, Srirajaskanthan R, Chokshi S. Human Immunocompetent Model of Neuroendocrine Liver Metastases Recapitulates Patient-Specific Tumour Microenvironment. Front Endocrinol (Lausanne) 2022; 13:909180. [PMID: 35909511 PMCID: PMC9326114 DOI: 10.3389/fendo.2022.909180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Neuroendocrine liver metastases (LM-NEN) develop in a considerable proportion of patients with gastroenteropancreatic neuroendocrine neoplasms. There is a paucity of experimental models that accurately recapitulate this complex metastatic human liver microenvironment precluding scientific and clinical advancements. Here, we describe the development of a novel personalised immunocompetent precision cut tumour slice (PCTS) model for LM-NEN using resected human liver tissue. The histological assessment throughout the culture demonstrated that slices maintain viability for at least 7 days and retain the cellular heterogeneity of the original tumour. Essential clinical features, such as patient-specific histoarchitecture, tumour grade, neuroendocrine differentiation and metabolic capacity, are preserved in the slices. The PCTS also replicate the tumor-specific immunological profile as shown by the innate and adaptive immunity markers analysis. Furthermore, the study of soluble immune checkpoint receptors in the culture supernatants proves that these immunomodulators are actively produced by LM-NEN and suggests that this process is epithelium-dependent. This model can be employed to investigate these pathways and provides a powerful platform for mechanistic, immunological and pre-clinical studies.
Collapse
Affiliation(s)
- Ewald Jan Doornebal
- Foundation for Liver Research, The Roger Williams Institute of Hepatology, London, United Kingdom
- King’s College London, Faculty of Life Sciences and Medicine, London, United Kingdom
| | - Nicola Harris
- Foundation for Liver Research, The Roger Williams Institute of Hepatology, London, United Kingdom
- King’s College London, Faculty of Life Sciences and Medicine, London, United Kingdom
| | - Antonio Riva
- Foundation for Liver Research, The Roger Williams Institute of Hepatology, London, United Kingdom
- King’s College London, Faculty of Life Sciences and Medicine, London, United Kingdom
| | - Ravi Jagatia
- Foundation for Liver Research, The Roger Williams Institute of Hepatology, London, United Kingdom
- King’s College London, Faculty of Life Sciences and Medicine, London, United Kingdom
| | - Michail Pizanias
- Institute of Liver Studies, King’s College Hospital and King’s College London, London, United Kingdom
| | - Andreas Prachalias
- Institute of Liver Studies, King’s College Hospital and King’s College London, London, United Kingdom
| | - Krishna Menon
- Institute of Liver Studies, King’s College Hospital and King’s College London, London, United Kingdom
| | - Melissa Preziosi
- Institute of Liver Studies, King’s College Hospital and King’s College London, London, United Kingdom
| | - Ane Zamalloa
- Institute of Liver Studies, King’s College Hospital and King’s College London, London, United Kingdom
| | - Rosa Miquel
- Liver Histopathology Laboratory, Institute of Liver Studies, King’s College Hospital, London, United Kingdom
| | - Yoh Zen
- Liver Histopathology Laboratory, Institute of Liver Studies, King’s College Hospital, London, United Kingdom
| | - Michael Robert Orford
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Simon Eaton
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Nigel Heaton
- Institute of Liver Studies, King’s College Hospital and King’s College London, London, United Kingdom
| | - John Ramage
- Institute of Liver Studies, King’s College Hospital and King’s College London, London, United Kingdom
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, King’s College Hospital, London, United Kingdom
| | - Elena Palma
- Foundation for Liver Research, The Roger Williams Institute of Hepatology, London, United Kingdom
- King’s College London, Faculty of Life Sciences and Medicine, London, United Kingdom
- *Correspondence: Shilpa Chokshi, ; Elena Palma,
| | - Rajaventhan Srirajaskanthan
- Institute of Liver Studies, King’s College Hospital and King’s College London, London, United Kingdom
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, King’s College Hospital, London, United Kingdom
| | - Shilpa Chokshi
- Foundation for Liver Research, The Roger Williams Institute of Hepatology, London, United Kingdom
- King’s College London, Faculty of Life Sciences and Medicine, London, United Kingdom
- *Correspondence: Shilpa Chokshi, ; Elena Palma,
| |
Collapse
|
14
|
Increased Plasma Soluble PD-1 Concentration Correlates with Disease Progression in Patients with Cancer Treated with Anti-PD-1 Antibodies. Biomedicines 2021; 9:biomedicines9121929. [PMID: 34944745 PMCID: PMC8698555 DOI: 10.3390/biomedicines9121929] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 12/21/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) confer remarkable therapeutic benefits to patients with various cancers. However, many patients are non-responders or develop resistance following an initial response to ICIs. There are no reliable biomarkers to predict the therapeutic effect of ICIs. Therefore, this study investigated the clinical implications of plasma levels of soluble anti-programmed death-1 (sPD-1) in patients with cancer treated with ICIs. In total, 22 patients (13 with non-small-cell lung carcinoma, 8 with gastric cancer, and 1 with bladder cancer) were evaluated for sPD-1 concentration using enzyme-linked immunosorbent assays for diagnostic and anti-PD-1 antibody analyses. sPD-1 levels were low before the administration of anti-PD-1 antibodies. After two and four cycles of anti-PD-1 antibody therapy, sPD-1 levels significantly increased compared with pretreatment levels (p = 0.0348 vs. 0.0232). We observed an increased rate of change in plasma sPD-1 concentrations after two and four cycles of anti-PD-1 antibody therapy that significantly correlated with tumor size progression (p = 0.024). sPD-1 may be involved in resistance to anti-PD-1 antibody therapy, suggesting that changes in sPD-1 levels can identify primary ICI non-responders early in treatment. Detailed analysis of each cancer type revealed the potential of sPD-1 as a predictive biomarker of response to ICI treatment in patients with cancer.
Collapse
|
15
|
Huyen PTM, Dung DTN, Weiß PJ, Hoan PQ, Giang DP, Uyen NT, Van Tuan N, Trung NT, Velavan TP, Song LH, Hoan NX. Circulating levels of sPD-1 and PD-1 genetic variants are associated with hepatitis B infection and related liver disease progression. Int J Infect Dis 2021; 115:229-236. [PMID: 34910956 DOI: 10.1016/j.ijid.2021.12.325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Programmed cell death-1 (PD-1) variants and circulating levels of soluble PD-1 are associated with susceptibility to malignant and infectious disease. This study aimed to examine the association of PD-1.5 and PD-1.9 variants, and plasma sPD-1 levels with HBV infection and disease progression. METHODS The study cohort consists of HBV-infected adults (n=513) stratified by clinical course, including chronic hepatitis B (CHB, n=173), liver cirrhosis (LC, n=134), hepatocellular carcinoma (HCC, n=206), and matched healthy controls (HC, n=196). The PD-1.5 (rs2227981 C/T) and PD-1.9 (rs2227982 C/T) genetic variants were genotyped by Sanger sequencing, and plasma sPD-1 levels were quantified by enzyme immunoassay. RESULTS The plasma sPD-1 levels were significantly high among HBV patients. The highest plasma sPD-1 levels were observed in CHB patients, followed by the LC and HCC groups. In addition, the plasma sPD-1 levels correlated positively with liver inflammation (aspartate transaminase, AST: rho=0.57, P<0.0001 and alanine aminotransferase, ALT: rho=0.57, P<0.0001) and were positively correlated with liver fibrosis (AST to Platelet Ratio Index, APRI score: rho=0.53, P<0.0001). The PD-1.9 TT genotype was less frequent in CHB patients compared to LC, HCC and HCC+LC patients in both codominant and recessive models (P<0.01) and was found to be a risk factor for HCC predisposition [HCC vs. non-HCC: OR=2.0 (95% CI: 1.13-3.7), Padj=0.017]. The PD-1.5 CT genotype was associated with a reduced risk of acquiring HCC [OR=0.6 (95%CI: 0.4-0.9), Padj=0.031]. CONCLUSION Our study concludes that sPD-1 levels are associated with liver inflammation and progression of liver fibrosis and the PD-1.5 and PD-1.9 variants are associated with HBV infection and progression of liver disease.
Collapse
Affiliation(s)
- Pham Thi Minh Huyen
- Department of Biochemistry, 108 Institute of Clinical Medical and Pharmaceutical Sciences, Hanoi, Vietnam; Faculty of Biochemistry, Hanoi Medical University, Hanoi, Vietnam; Faculty of Infectious Diseases, 108 Institute of Clinical Medical and Pharmaceutical Sciences, Hanoi, Vietnam
| | - Dang Thi Ngoc Dung
- Faculty of Biochemistry, Hanoi Medical University, Hanoi, Vietnam; Faculty of Infectious Diseases, 108 Institute of Clinical Medical and Pharmaceutical Sciences, Hanoi, Vietnam
| | - Peter Johann Weiß
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Phan Quoc Hoan
- Department of Molecular Biology, 108 Institute of Clinical Medical and Pharmaceutical Sciences, Hanoi, Vietnam
| | - Dao Phuong Giang
- Centre for Genetic Consultation and Cancer Screening, 108 Institute of Clinical Medical and Pharmaceutical Sciences, Hanoi, Vietnam; Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
| | - Ngo Thi Uyen
- Faculty of Biochemistry, Hanoi Medical University, Hanoi, Vietnam
| | | | - Ngo Tat Trung
- Centre for Genetic Consultation and Cancer Screening, 108 Institute of Clinical Medical and Pharmaceutical Sciences, Hanoi, Vietnam; Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
| | - Thirumalaisamy P Velavan
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany; Centre for Genetic Consultation and Cancer Screening, 108 Institute of Clinical Medical and Pharmaceutical Sciences, Hanoi, Vietnam
| | - Le Huu Song
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam; Faculty of Infectious Diseases, 108 Institute of Clinical Medical and Pharmaceutical Sciences, Hanoi, Vietnam
| | - Nghiem Xuan Hoan
- Department of Molecular Biology, 108 Institute of Clinical Medical and Pharmaceutical Sciences, Hanoi, Vietnam; Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam.
| |
Collapse
|
16
|
Yang W, Lei C, Song S, Jing W, Jin C, Gong S, Tian H, Guo T. Immune checkpoint blockade in the treatment of malignant tumor: current statue and future strategies. Cancer Cell Int 2021; 21:589. [PMID: 34727927 PMCID: PMC8565029 DOI: 10.1186/s12935-021-02299-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/24/2021] [Indexed: 01/08/2023] Open
Abstract
After being stagnant for decades, there has finally been a paradigm shift in the treatment of cancer with the emergence and application of immune checkpoint inhibitors (ICIs). The most extensively utilized ICIs are targeting the pathways involving programmed death-1 (PD-1) and cytotoxic T-lymphocyte associated protein 4 (CTLA-4). PD-1, as an crucial immune inhibitory molecule, by and large reasons the immune checkpoint response of T cells, making tumor cells get away from immune surveillance. Programmed cell death ligand-1 (PD-L1) is exceptionally expressed in most cancers cells and approves non-stop activation of the PD-1 pathway in the tumor microenvironment. PD-1/PD-L1 inhibitors can block the combination of PD-1 and PD-L1, inhibit hostile to regulatory signals, and restore the activity of T cells, thereby bettering immune response. The current researchers assume that the efficacy of these drugs is related to PD-L1 expression in tumor tissue, tumor mutation burden (TMB), and other emerging biomarkers. Although malignant tumors can benefit from the immunotherapy of PD-1/PD-L1 inhibitors, formulating a customized medication model and discovering biomarkers that can predict efficacy are the new trend in the new era of malignant tumor immunotherapy. This review summarizes the mechanism of action of PD-1/PD-L1 inhibitors, their clinical outcomes on various malignant tumors, their efficacy biomarkers, as well as predictive markers of irAEs.
Collapse
Affiliation(s)
- Wenwen Yang
- Department of Clinical Medicine, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China
| | - Caining Lei
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China
- The First Clinical Medicine College, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, People's Republic of China
| | - Shaoming Song
- Department of Clinical Medicine, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China
| | - Wutang Jing
- Department of Clinical Medicine, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China
| | - Chuanwei Jin
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China
- The First Clinical Medicine College, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, People's Republic of China
| | - Shiyi Gong
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China
| | - Hongwei Tian
- Department of Clinical Medicine, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China.
| | - Tiankang Guo
- Department of Clinical Medicine, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
17
|
Pawłowska A, Kwiatkowska A, Suszczyk D, Chudzik A, Tarkowski R, Barczyński B, Kotarski J, Wertel I. Clinical and Prognostic Value of Antigen-Presenting Cells with PD-L1/PD-L2 Expression in Ovarian Cancer Patients. Int J Mol Sci 2021; 22:11563. [PMID: 34768993 PMCID: PMC8583913 DOI: 10.3390/ijms222111563] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/21/2022] Open
Abstract
The latest literature demonstrates the predominant role of the programmed cell death axis (PD-1/PD-L1/PD-L2) in ovarian cancer (OC) pathogenesis. However, data concerning this issue is ambiguous. Our research aimed to evaluate the clinical importance of PD-L1/PD-L2 expression in OC environments. We evaluated the role of PD-L1/PD-L2 in OC patients (n = 53). The analysis was performed via flow cytometry on myeloid (mDCs) and plasmacytoid dendritic cells (pDCs) and monocytes/macrophages (MO/MA) in peripheral blood, peritoneal fluid (PF), and tumor tissue (TT). The data were correlated with clinicopathological characteristics and prognosis of OC patients. The concentration of soluble PD-L1 (sPD-L1) and PD-1 in the plasma and PF were determined by ELISA. We established an accumulation of PD-L1+/PD-L2+ mDCs, pDCs, and MA in the tumor microenvironment. We showed an elevated level of sPD-L1 in the PF of OC patients in comparison to plasma and healthy subjects. sPD-L1 levels in PF showed a positive relationship with Ca125 concentration. Moreover, we established an association between higher sPD-L1 levels in PF and shorter survival of OC patients. An accumulation of PD-L1+/PD-L2+ mDCs, pDCs, and MA in the TT and high sPD-L1 levels in PF could represent the hallmark of immune regulation in OC patients.
Collapse
Affiliation(s)
- Anna Pawłowska
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (D.S.); (A.C.); (I.W.)
| | - Agnieszka Kwiatkowska
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (D.S.); (A.C.); (I.W.)
| | - Dorota Suszczyk
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (D.S.); (A.C.); (I.W.)
| | - Agata Chudzik
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (D.S.); (A.C.); (I.W.)
| | - Rafał Tarkowski
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-081 Lublin, Poland; (R.T.); (B.B.); (J.K.)
| | - Bartłomiej Barczyński
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-081 Lublin, Poland; (R.T.); (B.B.); (J.K.)
| | - Jan Kotarski
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-081 Lublin, Poland; (R.T.); (B.B.); (J.K.)
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (D.S.); (A.C.); (I.W.)
| |
Collapse
|
18
|
Khan M, Arooj S, Wang H. Soluble B7-CD28 Family Inhibitory Immune Checkpoint Proteins and Anti-Cancer Immunotherapy. Front Immunol 2021; 12:651634. [PMID: 34531847 PMCID: PMC8438243 DOI: 10.3389/fimmu.2021.651634] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022] Open
Abstract
Co-inhibitory B7-CD28 family member proteins negatively regulate T cell responses and are extensively involved in tumor immune evasion. Blockade of classical CTLA-4 (cytotoxic T lymphocyte-associated antigen-4) and PD-1 (programmed cell death protein-1) checkpoint pathways have become the cornerstone of anti-cancer immunotherapy. New inhibitory checkpoint proteins such as B7-H3, B7-H4, and BTLA (B and T lymphocyte attenuator) are being discovered and investigated for their potential in anti-cancer immunotherapy. In addition, soluble forms of these molecules also exist in sera of healthy individuals and elevated levels are found in chronic infections, autoimmune diseases, and cancers. Soluble forms are generated by proteolytic shedding or alternative splicing. Elevated circulating levels of these inhibitory soluble checkpoint molecules in cancer have been correlated with advance stage, metastatic status, and prognosis which underscore their broader involvement in immune regulation. In addition to their potential as biomarker, understanding their mechanism of production, biological activity, and pathological interactions may also pave the way for their clinical use as a therapeutic target. Here we review these aspects of soluble checkpoint molecules and elucidate on their potential for anti-cancer immunotherapy.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Sumbal Arooj
- Department of Biochemistry, University of Sialkot, Sialkot, Pakistan
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| |
Collapse
|
19
|
Sukowati CHC, El-Khobar KE, Tiribelli C. Immunotherapy against programmed death-1/programmed death ligand 1 in hepatocellular carcinoma: Importance of molecular variations, cellular heterogeneity, and cancer stem cells. World J Stem Cells 2021; 13:795-824. [PMID: 34367478 PMCID: PMC8316870 DOI: 10.4252/wjsc.v13.i7.795] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/25/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a heterogeneous malignancy related to diverse etiological factors. Different oncogenic mechanisms and genetic variations lead to multiple HCC molecular classifications. Recently, an immune-based strategy using immune checkpoint inhibitors (ICIs) was presented in HCC therapy, especially with ICIs against the programmed death-1 (PD-1) and its ligand PD-L1. However, despite the success of anti-PD-1/PD-L1 in other cancers, a substantial proportion of HCC patients fail to respond. In this review, we gather current information on biomarkers of anti-PD-1/PD-L1 treatment and the contribution of HCC heterogeneity and hepatic cancer stem cells (CSCs). Genetic variations of PD-1 and PD-L1 are associated with chronic liver disease and progression to cancer. PD-L1 expression in tumoral tissues is differentially expressed in CSCs, particularly in those with a close association with the tumor microenvironment. This information will be beneficial for the selection of patients and the management of the ICIs against PD-1/PD-L1.
Collapse
Affiliation(s)
| | | | - Claudio Tiribelli
- Centro Studi Fegato, Fondazione Italiana Fegato ONLUS, Trieste 34149, Italy
| |
Collapse
|
20
|
Cunha Pereira T, Rodrigues-Santos P, Almeida JS, Rêgo Salgueiro F, Monteiro AR, Macedo F, Soares RF, Domingues I, Jacinto P, Sousa G. Immunotherapy and predictive immunologic profile: the tip of the iceberg. Med Oncol 2021; 38:51. [PMID: 33788049 DOI: 10.1007/s12032-021-01497-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/14/2021] [Indexed: 12/14/2022]
Abstract
The interplay between cancer and the immune system has been under investigation for more than a century. Immune checkpoint inhibitors have changed the outcome of several tumors; however, there is a significant percentage of patients presenting resistance to immunotherapy. Besides the action mechanism, it is essential to unravel this complex interplay between host immune system and tumorigenesis to determine an immune profile as a predictive factor to immune checkpoint blockade agents. Tumor expression of programmed death-ligand 1 (PD-L1), tumor mutational burden, or mismatch repair deficiency are recognized predictive biomarkers to immunotherapy but are insufficient to explain the response rates and heterogeneity across tumor sites. Therefore, it is crucial to explore the role of the tumor microenvironment in the diversity and clonality of tumor-infiltrating immune cells since different checkpoint molecules play an influential role in cytotoxic T cell activation. Moreover, cytokines, chemokines, and growth factors regulated by epigenetic factors play a complex part. Peripheral immune cells expressing PD-1/PD-L1 and the biologic roles of soluble immune checkpoint molecules are the subject of new lines of investigation. This article addresses some of the new molecules and mechanisms studied as possible predictive biomarkers to immunotherapy, linked with the concept of immune dynamics monitoring.
Collapse
Affiliation(s)
- Tatiana Cunha Pereira
- Medical Oncology Department, Portuguese Oncolology Institute of Coimbra Francisco Gentil, Avenida Bissaya Barreto, 98, 3000-075, Coimbra, Portugal.
| | - Paulo Rodrigues-Santos
- Immunology Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Laboratory of Immunology and Oncology, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Jani Sofia Almeida
- Immunology Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Laboratory of Immunology and Oncology, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Fábio Rêgo Salgueiro
- Medical Oncology Department, Portuguese Oncolology Institute of Coimbra Francisco Gentil, Avenida Bissaya Barreto, 98, 3000-075, Coimbra, Portugal
| | - Ana Raquel Monteiro
- Medical Oncology Department, Portuguese Oncolology Institute of Coimbra Francisco Gentil, Avenida Bissaya Barreto, 98, 3000-075, Coimbra, Portugal
| | - Filipa Macedo
- Medical Oncology Department, Portuguese Oncolology Institute of Coimbra Francisco Gentil, Avenida Bissaya Barreto, 98, 3000-075, Coimbra, Portugal
| | - Rita Félix Soares
- Medical Oncology Department, Portuguese Oncolology Institute of Coimbra Francisco Gentil, Avenida Bissaya Barreto, 98, 3000-075, Coimbra, Portugal
| | - Isabel Domingues
- Medical Oncology Department, Portuguese Oncolology Institute of Coimbra Francisco Gentil, Avenida Bissaya Barreto, 98, 3000-075, Coimbra, Portugal
| | - Paula Jacinto
- Medical Oncology Department, Portuguese Oncolology Institute of Coimbra Francisco Gentil, Avenida Bissaya Barreto, 98, 3000-075, Coimbra, Portugal
| | - Gabriela Sousa
- Medical Oncology Department, Portuguese Oncolology Institute of Coimbra Francisco Gentil, Avenida Bissaya Barreto, 98, 3000-075, Coimbra, Portugal
| |
Collapse
|
21
|
Khan M, Zhao Z, Arooj S, Fu Y, Liao G. Soluble PD-1: Predictive, Prognostic, and Therapeutic Value for Cancer Immunotherapy. Front Immunol 2020; 11:587460. [PMID: 33329567 PMCID: PMC7710690 DOI: 10.3389/fimmu.2020.587460] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022] Open
Abstract
Programmed death protein 1 (PD-1) interaction with PD-L1 deliver immunosuppressive environment for tumor growth, and its blockade with directed monoclonal antibodies (anti-PD-1/anti-PD-L1) has shown remarkable clinical outcome. Lately, their soluble counterparts, sPD-1 and sPD-L1, have been detected in plasma, and elevated levels have been associated with advanced disease, clinical stages, and worst prognosis for cancer patients. Elevated plasma levels of sPD-L1 have been correlated with worst prognosis in several studies and has displayed a persistent outlook. On the other hand, sPD-1 levels have been inconsistent in their predictive and prognostic ability. Pretherapeutic higher sPD-1 plasma levels have shown to predict advanced disease state and to a lesser extent worst prognosis. Any increase in sPD-1 plasma level post therapeutically have been correlated with improved survival for various cancers. In vitro and in vivo studies have shown sPD-1 ability to bind PD-L1 and PD-L2 and block PD-1/PD-L1 interaction. Local delivery of sPD-1 in cancer tumor microenvironment through local gene therapy have demonstrated an increase in tumor specific CD8+ T cell immunity and tumor growth reduction. It had also exhibited enhancement of T cell immunity induced by vaccination and other gene therapeutic agents. Furthermore, it may also lessen the inhibitory effect of circulating sPD-L1 and enhance the effects of mAb-based immunotherapy. In this review, we highlight various aspects of sPD-1 role in cancer prediction, prognosis, and anti-cancer immunity, as well as, its therapeutic value for local gene therapy or systemic immunotherapy in blocking the PD-1 and PD-L1 checkpoint interactions.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China.,Department of Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhihong Zhao
- Department of Nephrology, Shenzhen People's Hospital, Second Clinical Medicine Centre, Jinan University, Shenzhen, China
| | - Sumbal Arooj
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China.,Department of Biochemistry, University of Sialkot, Sialkot, Pakistan
| | - Yuxiang Fu
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Guixiang Liao
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
22
|
Quatrini L, Mariotti FR, Munari E, Tumino N, Vacca P, Moretta L. The Immune Checkpoint PD-1 in Natural Killer Cells: Expression, Function and Targeting in Tumour Immunotherapy. Cancers (Basel) 2020; 12:E3285. [PMID: 33172030 PMCID: PMC7694632 DOI: 10.3390/cancers12113285] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022] Open
Abstract
In the last years, immunotherapy with antibodies against programmed cell death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) has shown remarkable efficacy in the treatment of different types of tumours, representing a true revolution in oncology. While its efficacy has initially been attributed only to unleashing T cell responses, responsivity to PD-1/PD-L1 blockade was observed in some tumours with low Human Leukocyte Antigen (HLA) I expression and increasing evidence has revealed PD-1 surface expression and inhibitory function also in natural killer (NK) cells. Thus, the contribution of anti-PD-1/PD-L1 therapy to the recovery of NK cell anti-tumour response has recently been appreciated. Here, we summarize the studies investigating PD-1 expression and function in NK cells, together with the limitations and perspectives of immunotherapies. A better understanding of checkpoint biology is needed to design next-generation therapeutic strategies and to improve the clinical protocols of current therapies.
Collapse
Affiliation(s)
- Linda Quatrini
- Department of Immunology, IRCCS Bambino Gesù Children’s Hospital, 00146 Rome, Italy; (F.R.M.); (N.T.); (P.V.); (L.M.)
| | - Francesca Romana Mariotti
- Department of Immunology, IRCCS Bambino Gesù Children’s Hospital, 00146 Rome, Italy; (F.R.M.); (N.T.); (P.V.); (L.M.)
| | - Enrico Munari
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy;
| | - Nicola Tumino
- Department of Immunology, IRCCS Bambino Gesù Children’s Hospital, 00146 Rome, Italy; (F.R.M.); (N.T.); (P.V.); (L.M.)
| | - Paola Vacca
- Department of Immunology, IRCCS Bambino Gesù Children’s Hospital, 00146 Rome, Italy; (F.R.M.); (N.T.); (P.V.); (L.M.)
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Bambino Gesù Children’s Hospital, 00146 Rome, Italy; (F.R.M.); (N.T.); (P.V.); (L.M.)
| |
Collapse
|
23
|
Pawłowska A, Suszczyk D, Tarkowski R, Paduch R, Kotarski J, Wertel I. Programmed Death-1 Receptor (PD-1) as a Potential Prognosis Biomarker for Ovarian Cancer Patients. Cancer Manag Res 2020; 12:9691-9709. [PMID: 33116828 PMCID: PMC7548235 DOI: 10.2147/cmar.s263010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/03/2020] [Indexed: 12/25/2022] Open
Abstract
Aim Ovarian cancer (OC) is one of the most lethal gynecological malignancies. Recent studies suggest a crucial role of the PD-1/PD-L1 pathway in OC pathogenesis. Therefore, our study aimed at evaluation of the clinical importance of PD-1 expression in ovarian cancer patients. Patients and Methods In this study, we investigated the role of PD-1 in OC patients (n=50) by analyzing its expression on CD4+ and CD8+ T cells in three OC environments: peripheral blood (PB), peritoneal fluid (PF), and tumor (TT) as well as soluble PD-1 (sPD-1) in plasma and PF in terms of their clinical and prognostic significance. T cells with PD-1 expression were analyzed using flow cytometry. The concentration of sPD-1 was determined with the use of ELISA. Our research demonstrated differences in PD-1 expression on CD4+ and CD8+ T cells in the OC environments. Results We found an elevated level of CD4+PD-1+ T cells in tumor and PF, compared to PB. Additionally, we found the highest percentage of CD8+ PD-1+ in tumor, compared to PB and PF. The levels of sPD-1 were higher (p<0.0001) in plasma than in PF. For the first time, we discovered that the higher level of CD4+PD-1+ T cells in the circulation and the higher sPD-1 level in plasma predict poor survival of OC patients. Conclusion We suggest that PD-1 could be a predictive biomarker for OC patients and successful immunotherapy.
Collapse
Affiliation(s)
- Anna Pawłowska
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin 20-081, Poland
| | - Dorota Suszczyk
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin 20-081, Poland
| | - Rafał Tarkowski
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin 20-081, Poland
| | - Roman Paduch
- Department of Virology and Immunology, Maria Curie-Sklodowska University, Lublin 20-033, Poland
| | - Jan Kotarski
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin 20-081, Poland
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin 20-081, Poland
| |
Collapse
|
24
|
Zeng Z, Yang B, Liao ZY. Current progress and prospect of immune checkpoint inhibitors in hepatocellular carcinoma. Oncol Lett 2020; 20:45. [PMID: 32802167 PMCID: PMC7412709 DOI: 10.3892/ol.2020.11909] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023] Open
Abstract
In recent years, the incidence of liver cancer has increased and is currently the sixth most common tumor and the second leading cause of cancer-associated mortality worldwide. Most cases of liver cancer are hepatocellular carcinoma (HCC). Surgery, including liver transplantation or resection, and radiofrequency ablation therapies are all considered to be the curative treatment options for early-stage HCC. However, most patients have advanced HCC at the time of diagnosis, contributing to a poor prognosis. Therefore, improved treatment for late-stage HCC is needed. Immune checkpoint inhibitors (ICIs), among which programmed death receptor 1 (PD-1)/PD-ligand 1 and cytotoxic T lymphocyte-associated protein 4 are the representative immunological checkpoints, have shown great promise and progress for HCC treatment. The present review summarizes recent studies that have focused on ICIs and discusses the present limitations affecting the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Zhu Zeng
- Department of Abdominal Oncology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Biao Yang
- Department of Gastroenterology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zheng-Yin Liao
- Department of Abdominal Oncology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
25
|
Tannig P, Peter AS, Lapuente D, Klessing S, Schmidt A, Damm D, Tenbusch M, Überla K, Temchura V. Genetic Co-Administration of Soluble PD-1 Ectodomains Modifies Immune Responses against Influenza A Virus Induced by DNA Vaccination. Vaccines (Basel) 2020; 8:vaccines8040570. [PMID: 33019546 PMCID: PMC7712647 DOI: 10.3390/vaccines8040570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 01/08/2023] Open
Abstract
Due to the low efficacy and the need for seasonal adaptation of currently licensed influenza A vaccines, the importance of alternative vaccination strategies is increasingly recognized. Considering that DNA vaccines can be rapidly manufactured and readily adapted with novel antigen sequences, genetic vaccination is a promising immunization platform. However, the applicability of different genetic adjuvants to this approach still represents a complex challenge. Immune checkpoints are a class of molecules involved in adaptive immune responses and germinal center reactions. In this study, we immunized mice by intramuscular electroporation with a DNA-vaccine encoding hemagglutinin (HA) and nucleoprotein (NP) of the influenza A virus. The DNA-vaccine was applied either alone or in combination with genetic adjuvants encoding the soluble ectodomains of programmed cell death protein-1 (sPD-1) or its ligand (sPD-L1). Co-administration of genetic checkpoint adjuvants did not significantly alter immune responses against NP. In contrast, sPD-1 co-electroporation elevated HA-specific CD4+ T cell responses, decreased regulatory CD4+ T cell pools, and modulated the IgG2a-biased HA antibody pattern towards an isotype-balanced IgG response with a trend to higher influenza neutralization in vitro. Taken together, our data demonstrate that a genetic DNA-adjuvant encoding soluble ectodomains of sPD-1 was able to modulate immune responses induced by a co-administered influenza DNA vaccine.
Collapse
|
26
|
Zhang A, Sun Y, Wang S, Du J, Gao X, Yuan Y, Zhao L, Yang Y, Xu L, Lei Y, Duan L, Xu C, Ma L, Wang J, Hu G, Chen H, Wang Q, Hu L, Zhang B. Secretion of human soluble programmed cell death protein 1 by chimeric antigen receptor-modified T cells enhances anti-tumor efficacy. Cytotherapy 2020; 22:734-743. [PMID: 32684339 DOI: 10.1016/j.jcyt.2020.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/18/2020] [Accepted: 05/31/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND AIMS Chimeric antigen receptor (CAR) T cells have achieved favorable responses in patients with hematologic malignancies, but the outcome has been far from satisfactory in the treatment of tumors with high expression of immunosuppressive molecules. To overcome this limitation, we modified CAR T cells to secrete types of human soluble programmed cell death protein 1 (PD-1) called sPD-1 CAR T cells. METHODS To compare the effector function between second (conventional second-generation CAR targeting CD19) and sPD-1 CAR T cells, we measured cytotoxicity, cytokine secretion and activation markers incubated with or without tumor cells expressing CD19 and/or programmed cell death ligand 1 (PD-L1). Furthermore, the anti-tumor efficacy of second and sPD-1 CAR T cells was determined using an NSG mouse model bearing NALM-6-PD-L1. Finally, the underlying mechanism was investigated by metabolic parameters and RNA sequencing analysis of different CAR T cells. RESULTS Compared with second CAR T cells, sPD-1 CAR T cells enhanced killing efficiency toward CD19+PD-L1+ tumor cells in vitro. Furthermore, sPD-1 CAR T cells reduced the tumor burden and prolonged overall survival of the NSG (NOD-SCID-IL2rg) mice bearing NALM-6-PD-L1. To explore the effect of soluble PD-1 on CAR T cells, we found that sPD-1 CAR T cells exhibited higher levels of activation and ameliorative profiles of differentiation, exhaustion, glycolysis and apoptosis. CONCLUSIONS With constitutive soluble PD-1 secretion, sPD-1 CAR T cells have tended to eradicate tumors with a high expression of PD-L1 more effectively than second CAR T cells. This may be due to soluble PD-1 enhancing apoptosis resistance, aerobic metabolism and a more "stem" differentiation of CAR T cells. Overall, our study presents a feasible strategy to increase the efficacy of CAR T cells.
Collapse
Affiliation(s)
- Ang Zhang
- Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China; Department of Hematopoietic Stem Cell Transplantation, the Cell and Gene Therapy Center, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Beijing Key Laboratory of Stem Cell Therapy and Transformation Research, Beijing, China
| | - Yao Sun
- Department of Hematopoietic Stem Cell Transplantation, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shenyu Wang
- Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China; Department of Hematopoietic Stem Cell Transplantation, the Cell and Gene Therapy Center, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Beijing Key Laboratory of Stem Cell Therapy and Transformation Research, Beijing, China
| | - Jie Du
- SAFE Pharmaceutical Research Institute Co., Ltd
| | | | - Ye Yuan
- National Beijing Center for Drug Safety Evaluation and Research, State Key, Laboratory of Medical Countermeasures and Toxicology, Institute of Pharmacology and Toxicology, Academy of Military Sciences, Beijing, China
| | - Long Zhao
- Department of Hematopoietic Stem Cell Transplantation, the Cell and Gene Therapy Center, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Beijing Key Laboratory of Stem Cell Therapy and Transformation Research, Beijing, China
| | - Yang Yang
- Department of Hematopoietic Stem Cell Transplantation, the Cell and Gene Therapy Center, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Beijing Key Laboratory of Stem Cell Therapy and Transformation Research, Beijing, China
| | - Lei Xu
- Department of Hematopoietic Stem Cell Transplantation, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yangyang Lei
- Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Lian Duan
- Department of Neurosurgery, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chen Xu
- Department of Hematopoietic Stem Cell Transplantation, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lei Ma
- Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jinyu Wang
- Peking University International Hospital, Beijing, China
| | - Guoliang Hu
- Department of Hematopoietic Stem Cell Transplantation, the Cell and Gene Therapy Center, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Beijing Key Laboratory of Stem Cell Therapy and Transformation Research, Beijing, China
| | - Hu Chen
- Department of Hematopoietic Stem Cell Transplantation, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Department of Hematopoietic Stem Cell Transplantation, the Cell and Gene Therapy Center, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Beijing Key Laboratory of Stem Cell Therapy and Transformation Research, Beijing, China
| | - Quanjun Wang
- National Beijing Center for Drug Safety Evaluation and Research, State Key, Laboratory of Medical Countermeasures and Toxicology, Institute of Pharmacology and Toxicology, Academy of Military Sciences, Beijing, China.
| | - Liangding Hu
- Department of Hematopoietic Stem Cell Transplantation, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; National Beijing Center for Drug Safety Evaluation and Research, State Key, Laboratory of Medical Countermeasures and Toxicology, Institute of Pharmacology and Toxicology, Academy of Military Sciences, Beijing, China.
| | - Bin Zhang
- Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China; Department of Hematopoietic Stem Cell Transplantation, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Department of Hematopoietic Stem Cell Transplantation, the Cell and Gene Therapy Center, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Beijing Key Laboratory of Stem Cell Therapy and Transformation Research, Beijing, China.
| |
Collapse
|
27
|
Mariotti FR, Quatrini L, Munari E, Vacca P, Tumino N, Pietra G, Mingari MC, Moretta L. Inhibitory checkpoints in human natural killer cells: IUPHAR Review 28. Br J Pharmacol 2020; 177:2889-2903. [PMID: 32335915 PMCID: PMC7279970 DOI: 10.1111/bph.15081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors have revolutionized cancer therapy leading to exceptional success. However, there is still the need to improve their efficacy in non‐responder patients. Natural killer (NK) cells represent the first line of defence against tumours, due to their ability to release immunomodulatory cytokines and kill target cells that have undergone malignant transformation. Harnessing NK cell response will open new possibilities to improve control of tumour growth. In this respect inhibitory checkpoints expressed on these innate lymphocytes represents a promising target for next‐generation immunotherapy. In this review, we will summarize recent evidences on the expression of NK cells receptors in cancer, with a focus on the inhibitory checkpoint programmed cell death protein 1 (PD‐1). We will also highlight the strength and limitations of the blockade of PD‐1 inhibitory pathway and suggest new combination strategies that may help to unleash more efficiently NK cell anti‐tumour response.
Collapse
Affiliation(s)
- F R Mariotti
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - L Quatrini
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - E Munari
- Department of Pathology, Sacro Cuore Don Calabria, Negrar, Italy
| | - P Vacca
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - N Tumino
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - G Pietra
- Laboratory of Immunology, Department of Integrated Oncological Therapies, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Experimental Medicine (DIMES), Università di Genova, Genoa, Italy
| | - M C Mingari
- Laboratory of Immunology, Department of Integrated Oncological Therapies, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Experimental Medicine (DIMES), Center of Excellence for Biomedical Research, Università di Genova, Genoa, Italy
| | - L Moretta
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
28
|
Zhang S, Wang L, Li M, Zhang F, Zeng X. The PD-1/PD-L pathway in rheumatic diseases. J Formos Med Assoc 2020; 120:48-59. [PMID: 32334916 DOI: 10.1016/j.jfma.2020.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND/PURPOSE Autoimmune diseases are diseases in which the body produces an abnormal immune response to self-antigens and damages its own tissues. Programmed death-1 (PD-1) and its ligands (PD-Ls) have been discovered to be important negative regulators of the immune system, playing crucial roles in autoimmunity. METHODS We analyzed the existing scientific literature dealing with this issue. In this review, the PD-1/PD-L pathway in the genetic susceptibility to and pathogenesis of rheumatic diseases is discussed. The PD-1/PD-L pathway might be helpful for diagnosing, evaluating the disease activity of and treating rheumatic diseases. RESULTS PD-1/PD-L gene polymorphisms are associated with a genetic predisposition to rheumatic disorders, which can provide reference information for diagnosis and disease activity. The conclusion of the crucial role of the PD-1/PD-L pathway in the pathogenesis of rheumatic diseases is consistent, but the details remain controversial. In some animal models, manipulating the PD-1/PD-L pathway could decrease disease severity. PD-1/PD-Ls may enable us to develop new therapeutics for patients with rheumatic diseases in the future. CONCLUSION The PD-1/PD-L pathway plays crucial roles in rheumatic disease. More work is needed to provide a better mechanistic understanding of the PD-1/PD-L pathway and to facilitate the precise therapeutic manipulation of this pathway.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Li Wang
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Mengtao Li
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Fengchun Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiaofeng Zeng
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| |
Collapse
|
29
|
Regulations on Messenger RNA: Wires and Nodes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:251-263. [PMID: 32185714 DOI: 10.1007/978-981-15-3266-5_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Somatic cells of an organism virtually share the same DNA but it is the timely expression of specific genes that determine their phenotype and cellular identity. A series of complex molecular machinery allows for the regulated process of RNA transcription, splicing, and translation. In addition, microRNAs and specialized RNA binding proteins can trigger the degradation of mRNAs. Long non-coding RNAs can also regulate mRNA fate in multiple ways. In this chapter, we reviewed the RNA processing mechanisms directly regulating immune checkpoint genes. We also cover RNA-based therapeutic strategies aiming at restoring immunity by targeting immune checkpoint genes.
Collapse
|
30
|
Tiako Meyo M, Jouinot A, Giroux-Leprieur E, Fabre E, Wislez M, Alifano M, Leroy K, Boudou-Rouquette P, Tlemsani C, Khoudour N, Arrondeau J, Thomas-Schoemann A, Blons H, Mansuet-Lupo A, Damotte D, Vidal M, Goldwasser F, Alexandre J, Blanchet B. Predictive Value of Soluble PD-1, PD-L1, VEGFA, CD40 Ligand and CD44 for Nivolumab Therapy in Advanced Non-Small Cell Lung Cancer: A Case-Control Study. Cancers (Basel) 2020; 12:cancers12020473. [PMID: 32085544 PMCID: PMC7072584 DOI: 10.3390/cancers12020473] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/05/2020] [Accepted: 02/13/2020] [Indexed: 02/07/2023] Open
Abstract
A large interindividual variability has been observed in anti Programmed cell Death 1 (anti-PD1) therapies efficacy. The aim of this study is to assess the correlation of soluble PD-1 (sPD-1), soluble Programmed cell Death Ligand 1 (sPD-L1), Vascular Endothelial Growth Factor A (VEGFA), soluble CD40 ligand (sCD40L) and soluble CD44 (sCD44), with survival in nivolumab-treated metastatic non-small cell lung cancer (NSCLC) patients. Plasma biomarkers were assayed at baseline and after two cycles of nivolumab. A cut-off of positivity for sPD-1, sPD-L1 and sCD40L expressions was defined as a plasma level above the lower limit of quantification. Baseline sPD-1 and sPD-L1 levels were subsequently analyzed in a control group of EGFR-mutated (Epidermal Growth Factor Receptor) NSCLC patients. Association between survival and biomarkers was investigated using Cox proportional hazard regression model. Eighty-seven patients were included (51 nivolumab-treated patients, 36 in EGFR-mutated group). In nivolumab group, baseline sPD-1, sPD-L1 and sCD40L were positive for 15(29.4%), 27(52.9%) and 18(50%) patients, respectively. We defined a composite criteria (sCombo) corresponding to sPD-1 and/or sPD-L1 positivity for each patient. In nivolumab group, baseline sCombo positivity was associated with shorter median progression-free survival (PFS) (78 days 95%CI (55–109) vs. 658 days (222-not reached); HR: 4.12 (1.95–8.71), p = 0.0002) and OS (HR: 3.99(1.63–9.80), p = 0.003). In multivariate analysis, baseline sCombo independently correlated with PFS (HR: 2.66 (1.17–6.08), p = 0.02) but not OS. In EGFR-mutated group, all patients were baseline sCombo positive; therefore this factor was not associated with survival. After two cycles of nivolumab, an increased or stable sPD-1 level independently correlated with longer PFS (HR: 0.49, 95%CI (0.30–0.80), p = 0.004) and OS (HR: 0.39, 95%CI (0.21–0.71), p = 0.002). VEGFA, sCD40L and sCD44 did not correlate with survival. We propose a composite biomarker using sPD-1and sPDL-1 to predict nivolumab efficacy in NSCLC patients. A larger validation study is warranted.
Collapse
Affiliation(s)
- Manuela Tiako Meyo
- Drug Biology–Toxicology, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France; (N.K.); (M.V.); (B.B.)
- UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, Paris Descartes University, PRES Sorbonne Paris Cité, 75006 Paris, France;
- Department of Medical Oncology, Cochin Hospital, AP-HP, Paris Descartes University, CARPEM, 75014 Paris, France; (A.J.); (P.B.-R.); (C.T.); (J.A.); (F.G.); (J.A.)
- Correspondence: ; Tel.: +331-5841-2313; Fax: +331-5841-2315
| | - Anne Jouinot
- Department of Medical Oncology, Cochin Hospital, AP-HP, Paris Descartes University, CARPEM, 75014 Paris, France; (A.J.); (P.B.-R.); (C.T.); (J.A.); (F.G.); (J.A.)
- Institut Cochin, INSERM U1016, 75014 Paris, France
| | - Etienne Giroux-Leprieur
- Department of Respiratory Diseases and Thoracic Oncology, APHP-AmbroiseParé Hospital and EA 4340 University Versailles-Saint Quentin en Yvelines, 92100 Boulogne, France;
| | - Elizabeth Fabre
- Department of Thoracic Oncology, Georges Pompidou European Hospital, AP-HP, 75015 Paris, France;
| | - Marie Wislez
- Department of Pneumology, Cochin Hospital, APHP, 75014 Paris, France;
| | - Marco Alifano
- Department of Thoracic Surgery, Cochin Hospital, APHP, 75014 Paris, France;
| | - Karen Leroy
- Department of Cyto-pathology, Cochin Hospital, AP-HP, 75014 Paris, France; (K.L.); (A.M.-L.); (D.D.)
| | - Pascaline Boudou-Rouquette
- Department of Medical Oncology, Cochin Hospital, AP-HP, Paris Descartes University, CARPEM, 75014 Paris, France; (A.J.); (P.B.-R.); (C.T.); (J.A.); (F.G.); (J.A.)
| | - Camille Tlemsani
- Department of Medical Oncology, Cochin Hospital, AP-HP, Paris Descartes University, CARPEM, 75014 Paris, France; (A.J.); (P.B.-R.); (C.T.); (J.A.); (F.G.); (J.A.)
| | - Nihel Khoudour
- Drug Biology–Toxicology, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France; (N.K.); (M.V.); (B.B.)
| | - Jennifer Arrondeau
- Department of Medical Oncology, Cochin Hospital, AP-HP, Paris Descartes University, CARPEM, 75014 Paris, France; (A.J.); (P.B.-R.); (C.T.); (J.A.); (F.G.); (J.A.)
| | - Audrey Thomas-Schoemann
- UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, Paris Descartes University, PRES Sorbonne Paris Cité, 75006 Paris, France;
- Department of Medical Oncology, Cochin Hospital, AP-HP, Paris Descartes University, CARPEM, 75014 Paris, France; (A.J.); (P.B.-R.); (C.T.); (J.A.); (F.G.); (J.A.)
| | - Hélène Blons
- Department of Cyto-pathology, Georges Pompidou European Hospital, AP-HP, 75015 Paris, France;
| | - Audrey Mansuet-Lupo
- Department of Cyto-pathology, Cochin Hospital, AP-HP, 75014 Paris, France; (K.L.); (A.M.-L.); (D.D.)
| | - Diane Damotte
- Department of Cyto-pathology, Cochin Hospital, AP-HP, 75014 Paris, France; (K.L.); (A.M.-L.); (D.D.)
| | - Michel Vidal
- Drug Biology–Toxicology, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France; (N.K.); (M.V.); (B.B.)
- UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, Paris Descartes University, PRES Sorbonne Paris Cité, 75006 Paris, France;
| | - François Goldwasser
- Department of Medical Oncology, Cochin Hospital, AP-HP, Paris Descartes University, CARPEM, 75014 Paris, France; (A.J.); (P.B.-R.); (C.T.); (J.A.); (F.G.); (J.A.)
- Institut Cordeliers, INSERM U1147, 75006 Paris, France
| | - Jérôme Alexandre
- Department of Medical Oncology, Cochin Hospital, AP-HP, Paris Descartes University, CARPEM, 75014 Paris, France; (A.J.); (P.B.-R.); (C.T.); (J.A.); (F.G.); (J.A.)
- Institut Cochin, INSERM U1016, 75014 Paris, France
- Institut Cordeliers, INSERM U1147, 75006 Paris, France
| | - Benoit Blanchet
- Drug Biology–Toxicology, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France; (N.K.); (M.V.); (B.B.)
- UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, Paris Descartes University, PRES Sorbonne Paris Cité, 75006 Paris, France;
| |
Collapse
|
31
|
Modulation of Vaccine-Induced HIV-1-Specific Immune Responses by Co-Electroporation of PD-L1 Encoding DNA. Vaccines (Basel) 2020; 8:vaccines8010027. [PMID: 31947643 PMCID: PMC7157229 DOI: 10.3390/vaccines8010027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/31/2022] Open
Abstract
The importance of a balanced TH1/TH2 humoral immune response against the HIV-1 envelope protein (Env) for antibody-mediated HIV-1 control is increasingly recognized. However, there is no defined vaccination strategy to raise it. Since immune checkpoints are involved in the induction of adoptive immunity and their inhibitors (monoclonal antibodies) are licensed for cancer therapy, we investigated the effect of checkpoint blockade after HIV-1 genetic vaccination on enhancement and modulation of antiviral antibody responses. By intraperitoneal administration of checkpoint antibodies in mice we observed an induction of anti-drug antibodies which may interfere with immunomodulation by checkpoint inhibitors. Therefore, we blocked immune checkpoints locally by co-electroporation of DNA vaccines encoding the active soluble ectodomains of programmed cell death protein-1 (PD-1) or its ligand (PD-L1), respectively. Plasmid-encoded immune checkpoints did not elicit a detectable antibody response, suggesting no interference with their immunomodulatory effects. Co-electroporation of a HIV-1 DNA vaccine formulation with soluble PD-L1 ectodomain increased HIV-1 Env-specific TH1 CD4 T cell and IgG2a antibody responses. The overall antibody response was hereby shifted towards a more TH1/TH2 balanced subtype pattern. These findings indicate that co-electroporation of soluble checkpoint ectodomains together with DNA-based vaccines has modulatory effects on vaccine-induced immune responses that could improve vaccine efficacies.
Collapse
|
32
|
Mohammadzadeh S, Khanahmad H, Esmaeil N, Eskandari N, Rahimmanesh I, Rezaei A, Andalib A. Producing Soluble Human Programmed Cell Death Protein-1: A Natural Supporter for CD4+T Cell Cytotoxicity and Tumor Cells Apoptosis. IRANIAN JOURNAL OF BIOTECHNOLOGY 2019; 17:e2104. [PMID: 32671122 PMCID: PMC7357696 DOI: 10.30498/ijb.2019.85180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Programmed cell death protein-1 (PD-1)/PD-L1 pathway is one of the immune checkpoint pathways involved in the regulation of the immune responses and the suppression of anti-tumor defense. PD-1/PD-L1 blocking antibodies improve immune responses such as cytotoxic activity of CD8+/CD4+T cells and increase mortality of tumor cells as well; however, their use is accompanied by adverse side effects. OBJECTIVES We aimed to produce a native blocker of human PD-1/PD-L1, for developing T cells cytotoxicity and tumor cells apoptosis. MATERIALS AND METHODS We designed and cloned soluble human PD-1-GFP-pcDNA3.1/hygro construct in Escherichia coli strain TOP10 cells and then transfected this construct into the HEK cells. The concentration of the secreted shPD-1 in the supernatant was measured and the supernatant was used for blocking PD-L1 on the MDA-MB-231 cells. The cytotoxicity of CD8+/CD4+T cells and the apoptosis of MDA-MB-231 cells, under the influence of shPD-1 in the co-culture of T cells with the MDA-MB-231 cells, were evaluated using flow cytometry technique. RESULTS The GFP expression in the transfected cells illustrated the successful designing, transfection, and production of shPD-1. Soluble human PD-1 concentration in the supernatant of the transfected HEK cells was significantly higher than the untransfected cells. In addition, shPD-1 significantly blocked PD-L1 on the MDA- MB-231 cells, improved the cytotoxicity of CD4+T cells, and increased the apoptosis of MDA-MB-231 cells. CONCLUSION Overall, increased CD4+T cell cytotoxicity and tumor cells apoptosis under the influence of shPD-1, confirmed the effectiveness of shPD-1 as a natural blocker of PD-L1and as an augmenter of the anti-tumorimmune responses.
Collapse
Affiliation(s)
- Samane Mohammadzadeh
- Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad
- Genetics and Molecular Biology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Esmaeil
- Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nahid Eskandari
- Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ilnaz Rahimmanesh
- Genetics and Molecular Biology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbas Rezaei
- Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Andalib
- Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
33
|
Xue Q, Li X, Gu Y, Wang X, Wang M, Tian J, Duan X, Gao H, Ji X, Yan X, Dong W, Fang Q, Zhang X. Unbalanced Expression of ICOS and PD-1 in Patients with Neuromyelitis Optica Spectrum Disorder. Sci Rep 2019; 9:14130. [PMID: 31575949 PMCID: PMC6773714 DOI: 10.1038/s41598-019-50479-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 09/13/2019] [Indexed: 12/22/2022] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) likely results from humoral immune abnormalities. The role that helper T cells play in the pathogenesis of this disease is not fully understood. To ascertain the clinical significance of two important costimulatory molecules required for T-cell activation in the peripheral blood of patients with NMOSD, we examined the expression levels of a membrane- and soluble-type inducible costimulatory molecule (ICOS), its ligand (ICOSL), programmed death-1 (PD-1), and its ligand (PD-L1) in the peripheral blood of 30 patients with NMOSD and compared these levels with those in patients with longitudinally extensive transverse myelitis (LETM), those with optic neuritis (ON), and healthy controls (HCs). Our results showed that the ICOS/ICOSL and PD-1/PD-L1 pathways may play important roles in the early stages of NMOSD pathogenesis. ICOS and PD-1 are potential therapeutic targets and valuable biomarkers for the differential diagnosis of early-stage NMOSD.
Collapse
Affiliation(s)
- Qun Xue
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China. .,Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China. .,Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu, 215004, China.
| | - Xiaoping Li
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.,Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Yanzheng Gu
- Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.,Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu, 215004, China
| | - Xiaozhu Wang
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Mingyuan Wang
- Suzhou Red Cross Central Blood Station, Suzhou, Jiangsu, 215006, China
| | - Jingluan Tian
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xiaoyu Duan
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Hanqing Gao
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xiaopei Ji
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xiaoming Yan
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Wanli Dong
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Qi Fang
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China. .,Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
| | - Xueguang Zhang
- Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China. .,Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu, 215004, China.
| |
Collapse
|
34
|
Chakrabarti R, Kapse B, Mukherjee G. Soluble immune checkpoint molecules: Serum markers for cancer diagnosis and prognosis. Cancer Rep (Hoboken) 2019; 2:e1160. [PMID: 32721130 PMCID: PMC7941475 DOI: 10.1002/cnr2.1160] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/18/2018] [Accepted: 01/09/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND With the recent advances in the understanding of the interaction of the immune system with developing tumor, it has become imperative to consider the immunological parameters for both cancer diagnosis and disease prognosis. Additionally, in the era of emerging immunotherapeutic strategies in cancer, it is very important to follow the treatment outcome and also to predict the correct immunotherapeutic strategy in individual patients. There being enormous heterogeneity among tumors at different sites or between primary and metastatic tumors in the same individual, or interpatient heterogeneity, it is very important to study the tumor-immune interaction in the tumor microenvironment and beyond. Importantly, molecular tools and markers identified for such studies must be suitable for monitoring in a noninvasive manner. RECENT FINDINGS Recent studies have shown that the immune checkpoint molecules play a key role in the development and progression of tumors. In-depth studies of these molecules have led to the development of most of the cancer immunotherapeutic reagents that are currently either in clinical use or under different phases of clinical trials. Interestingly, many of these cell surface molecules undergo alternative splicing to produce soluble isoforms, which can be tracked in the serum of patients. CONCLUSIONS Several studies demonstrate that the serum levels of these soluble isoforms could be used as noninvasive markers for cancer diagnosis and disease prognosis or to predict patient response to specific therapeutic strategies.
Collapse
Affiliation(s)
- Rituparna Chakrabarti
- School of Medical Science and TechnologyIndian Institute of Technology KharagpurKharagpurIndia
| | - Bhavya Kapse
- Department of BiotechnologyIndian Institute of Technology KharagpurKharagpurIndia
| | - Gayatri Mukherjee
- School of Medical Science and TechnologyIndian Institute of Technology KharagpurKharagpurIndia
| |
Collapse
|
35
|
Yan X, Gu Y, Wang C, Sun S, Wang X, Tian J, Wang M, Ji X, Duan X, Gao H, Fang Q, Dong W, Zhang X, Xue Q. Unbalanced expression of membrane-bound and soluble inducible costimulator and programmed cell death 1 in patients with myasthenia gravis. Clin Immunol 2019; 207:68-78. [PMID: 31374257 DOI: 10.1016/j.clim.2019.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/23/2019] [Accepted: 07/23/2019] [Indexed: 02/01/2023]
Abstract
This study aimed to investigate the possible functions and mechanisms of positive and negative costimulatory molecules in the pathological process of myasthenia gravis (MG). The expression levels of membrane-bound inducible costimulator (ICOS) and programmed cell death 1 (PD-1) in peripheral blood T cells, their corresponding ligands ICOSL and PDL-1 on B cells, and their soluble forms (sICOS, sPD-1, sICOSL, and sPDL-1) in plasma were detected in patients with untreated-stage MG (USMG) and remission-stage MG (RSMG). The results showed that the expression levels of membrane-bound ICOS and PD-1 in the peripheral blood T cells of the USMG group and their corresponding ligands ICOSL and PD-L1 on B cells were significantly increased compared to those in the RSMG group and healthy controls (HCs). The levels of sICOSL and sPD-1 were significantly upregulated in USMG patients compared to those in the RSMG and HC groups, while the levels of sICOS and sPD-L1 were not different. The expression of PD-L1 on CD19+ B cells was positively correlated with the concentrations of AchR Ab in the USMG group. The expression of ICOS and PD-1 in CD4+ T cells and the expression of ICOSL and PD-L1 on CD19+ B cells were positively correlated with the quantitative myasthenia gravis (QMG) scores in the USMG group. Also, in the USMG group, the plasma levels of sICOSL and sPD-1 were positively correlated with the QMG scores. In addition, the percentage of peripheral blood follicular helper T (Tfh) cells in the USMG group was positively correlated with ICOS and PD-1 expression on CD4+ T cells and ICOSL and PD-L1 expression on CD19+ B cells. There were positive correlations between sICOSL and sPD-1 levels and the percentage of peripheral blood Tfh cells and plasma interleukin-21 (IL-21) levels in the USMG group. The results suggest that the positive ICOS/ICOSL and negative PD-1/PD-L1 costimulatory molecule pairs participate in the pathological process of MG. Abnormal sICOSL and sPD-1 expression might interfere with the normal signal transduction of ICOS and PD-1 on Tfh cells, causing excessive activation of Tfh cells and promotion of disease progression. sICOSL and sPD-1 have potential value in monitoring MG disease states.
Collapse
Affiliation(s)
- Xiaoming Yan
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yanzheng Gu
- Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu 215004, China
| | - Caiqin Wang
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Simao Sun
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaozhu Wang
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jingluan Tian
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Mingyuan Wang
- Suzhou Red Cross Central Blood Station, Suzhou, Jiangsu 215006, China
| | - Xiaopei Ji
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaoyu Duan
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Hanqing Gao
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Qi Fang
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu 215004, China
| | - Wanli Dong
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xueguang Zhang
- Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Qun Xue
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu 215004, China.
| |
Collapse
|
36
|
Mariotti FR, Quatrini L, Munari E, Vacca P, Moretta L. Innate Lymphoid Cells: Expression of PD-1 and Other Checkpoints in Normal and Pathological Conditions. Front Immunol 2019; 10:910. [PMID: 31105707 PMCID: PMC6498986 DOI: 10.3389/fimmu.2019.00910] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/09/2019] [Indexed: 12/14/2022] Open
Abstract
Innate lymphoid cells (ILCs) belong to a family of immune cells. Recently, ILCs have been classified into five different groups that mirror the function of adaptive T cell subsets counterparts. In particular, NK cells mirror CD8+ cytotoxic T cells while ILC1, ILC2, ILC3, and Lymphoid tissue inducer (LTi)-like cells reflect the function of CD4+T helper (Th) cells (Th1, Th2, and Th17 respectively). ILCs are involved in innate host defenses against pathogens and tumors, in lymphoid organogenesis, and in tissue remodeling/repair. In recent years, important molecular inducible checkpoints (PD-1, TIM3, and TIGIT) were shown to control/inactivate different immune cell types. The expression of many of these receptors has been detected on NK cells and subsets of tissue-resident ILCs in both physiological and pathological conditions, including cancer. In particular, it has been demonstrated that the interaction between PD-1+ immune cells and PD-L1/PD-L2+ tumor cells may compromise the anti-tumor effector function leading to tumor immune escape. However, while the effector function of NK cells in tumor is well-established, limited information exists on the other ILC subsets. We will summarize what is known to date on the expression and function of these checkpoint receptors on NK cells and ILCs, with a particular focus on the recent data that reveal an essential contribution of the blockade of PD-1 and TIGIT on NK cells to the immunotherapy of cancer. A better information regarding the presence and the function of different ILCs and of the inhibitory checkpoints in pathological conditions may offer important clues for the development of new immune therapeutic strategies.
Collapse
Affiliation(s)
| | - Linda Quatrini
- Department of Immunology, IRCSS Bambino Gesù Children's Hospital, Rome, Italy
| | - Enrico Munari
- Department of Pathology, Sacro Cuore Don Calabria Hospital, Negrar, Italy
| | - Paola Vacca
- Department of Immunology, IRCSS Bambino Gesù Children's Hospital, Rome, Italy
| | - Lorenzo Moretta
- Department of Immunology, IRCSS Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
37
|
Mariotti FR, Petrini S, Ingegnere T, Tumino N, Besi F, Scordamaglia F, Munari E, Pesce S, Marcenaro E, Moretta A, Vacca P, Moretta L. PD-1 in human NK cells: evidence of cytoplasmic mRNA and protein expression. Oncoimmunology 2018; 8:1557030. [PMID: 30723590 PMCID: PMC6350684 DOI: 10.1080/2162402x.2018.1557030] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/30/2018] [Accepted: 12/05/2018] [Indexed: 01/01/2023] Open
Abstract
Under physiological conditions, PD-1/PD-L1 interactions regulate unwanted over-reactions of immune cells and contribute to maintain peripheral tolerance. However, in tumor microenvironment, this interaction may greatly compromise the immune-mediated anti-tumor activity. PD-1+ NK cells have been detected in high percentage in peripheral blood and ascitic fluid of ovarian carcinoma patients. To acquire information on PD-1 expression and physiology in human NK cells, we analyzed whether PD-1 mRNA and protein are present in resting, surface PD-1−, NK cells from healthy donors. Both different splicing isoforms of PD-1 mRNA and a cytoplasmic pool of PD-1 protein were detected. Similar results were obtained also from both in vitro-activated and tumor-associated NK cells. PD-1 mRNA and protein were higher in CD56dim than in CD56bright NK cells. Confocal microscopy analyses revealed that PD-1 protein is present in virtually all NK cells analyzed. The present findings are compatible with a rapid surface expression of PD-1 in NK cells in response to appropriate, still undefined, stimuli.
Collapse
Affiliation(s)
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Research Center, IRCSS Bambino Gesù Children's Hospital, Rome, Italy
| | - Tiziano Ingegnere
- Department of Immunology, IRCSS Bambino Gesù Children's Hospital, Rome, Italy
| | - Nicola Tumino
- Department of Immunology, IRCSS Bambino Gesù Children's Hospital, Rome, Italy
| | - Francesca Besi
- Department of Immunology, IRCSS Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Enrico Munari
- Department of Pathology, Sacro Cuore Don Calabria Hospital, Negrar, Italy.,Department of Pathology AOUI, University of Verona, Verona, Italy
| | - Silvia Pesce
- Department of Experimental Medicine (DIMES), University of Genova, Genoa, Italy
| | - Emanuela Marcenaro
- Department of Experimental Medicine (DIMES), University of Genova, Genoa, Italy.,Center of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Alessandro Moretta
- Department of Experimental Medicine (DIMES), University of Genova, Genoa, Italy.,Center of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Paola Vacca
- Department of Immunology, IRCSS Bambino Gesù Children's Hospital, Rome, Italy
| | - Lorenzo Moretta
- Department of Immunology, IRCSS Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
38
|
Liang L, Ge K, Zhang F, Ge Y. The suppressive effect of co-inhibiting PD-1 and CTLA-4 expression on H22 hepatomas in mice. Cell Mol Biol Lett 2018; 23:58. [PMID: 30564277 PMCID: PMC6295075 DOI: 10.1186/s11658-018-0122-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 11/22/2018] [Indexed: 12/31/2022] Open
Abstract
Objective We investigated the suppressive effect of siRNA-mediated co-inhibition of PD-1 and CTLA-4 expression on H22 hepatomas in mice. Methods Murine H22 cells were cultured in vivo in ICR mice. An allograft tumor model was also established in another ICR mouse group. The tumor-bearing mice were randomly divided into four groups: control, single PD-1 siRNA, single CTLA-4 siRNA, and double PD-1 + CTLA-4 siRNAs. The survival time and physiological condition of the mice were observed after the injection of the siRNAs and placebo. The volume and weight of the solid tumor were measured to assess the inhibition of the tumor. To assess the effects of siRNAs on mouse immune function, the protein levels of IFN-γ and IL-10 in the blood and PD-L1 in the tumor and liver were determined using ELISA, and the mRNA levels of IFN-γ, PD-L1, PD-1, CTLA-4, IL-6 and Survivin in the tumor, liver and spleen were determined using quantitative RT-PCR. The ratios of Bax and Bcl-2 protein were determined via western blot to analyze the effect of siRNAs on tumor cell apoptosis. Results The anti-tumor effect appeared in all groups with siRNA-mediated inhibition. The tumor growth suppression was stronger in the group with double inhibition. The weight and volume of the tumors were significantly lower and the survival rate improved in the three siRNA groups. IFN-γ levels increased but IL-10 levels decreased in the blood of the siRNA group mice compared with the results for the control group. In the tumor and spleen tissue, the IFN-γ levels significantly increased, but in the liver tissue they significantly decreased in the three siRNA groups. The results of quantitative RT-PCR showed that the mRNAs for PD-1 and CTLA-4 were downregulated in spleen tissue in the three siRNA groups, while the PD-L1 mRNA and protein levels increased significantly in the tumor, but decreased in the liver. Survivin and IL-6 mRNA levels decreased in the tumor. Western blot results showed that ratio of Bax and Bcl-2 had significantly increased. These results indicated that downregulating PD-1 and CTLA-4 could increase the body’s immune response and promote apoptosis of tumor cells. Conclusion Co-inhibiting the expressions of PD-1 and CTLA-4 can effectively suppress the growth of H22 hepatoma and promote the apoptosis of tumor cells in mice. Blocking PD-1 and CTLA-4 can improve the vitality of T cells, and improve the immune environment and response.
Collapse
Affiliation(s)
- Leilei Liang
- 1Department of Biochemistry and Molecular Biology, Medical College, Qingdao University, 38 Dengzhou Road, Qingdao, 266021 Shandong China.,2Central Laboratory, Binzhou People's Hospital, Binzhou, 256610 Shandong China
| | - Keli Ge
- 3Integrative Medicine Research Center, Medical College, Qingdao University, Qingdao, 266021 Shandong China
| | - Fengying Zhang
- 4Department of Biochemistry and Molecular Biology, Heze Medical College, Heze, 274000 Shandong China
| | - Yinlin Ge
- 1Department of Biochemistry and Molecular Biology, Medical College, Qingdao University, 38 Dengzhou Road, Qingdao, 266021 Shandong China
| |
Collapse
|
39
|
Chang B, Huang T, Wei H, Shen L, Zhu D, He W, Chen Q, Zhang H, Li Y, Huang R, Li W, Wu P. The correlation and prognostic value of serum levels of soluble programmed death protein 1 (sPD-1) and soluble programmed death-ligand 1 (sPD-L1) in patients with hepatocellular carcinoma. Cancer Immunol Immunother 2018; 68:353-363. [PMID: 30506460 PMCID: PMC6426820 DOI: 10.1007/s00262-018-2271-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 11/01/2018] [Indexed: 02/07/2023]
Abstract
Background Blocking the programmed death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) pathway in hepatocellular carcinoma (HCC) is a very promising approach in immunotherapy. However, the correlation and prognostic values of serum soluble PD-1 and PD-L1 (sPD-1/sPD-L1) have not been explored conjointly in HCC patients. Methods This study retrospectively included 120 HCC patients receiving radical resection. The serum levels of sPD-1/sPD-L1 and inflammatory cytokines were measured by antibody array assay. Immunohistochemistry was applied to assess both the expression of membrane-bound PD-L1, and the number of CD4+ tumor-infiltrating lymphocytes (TILs) and CD8+ TILs. Results The best cut-off values of sPD-1 and sPD-L1 for predicting disease-free survival (DFS) were 33.0 µg/ml and 11.2 µg/ml, respectively. Multivariable analysis showed that sPD-L1 was a negative independent prognostic factor [DFS, Hazard Ratio (HR) 2.58, 95% CI 1.14–5.84, P = 0.023; overall survival (OS), HR 1.77, 95% CI 1.01–3.12, P = 0.048], while sPD-1 was a favorable independent prognostic factor (DFS, HR 0.32, 95% CI 0.14–0.74, P = 0.007; OS, HR 0.54, 95% CI 0.30–0.98, P = 0.044) in HCC patients. We also observed some similar associations between inflammatory cytokines (IL-10, IL-17, TNF-α) and sPD-1 or sPD-L1, as well as a close positive association between sPD-1 and sPD-L1. No significant associations of sPD-1/sPD-L1 with either intra-tumoral PD-L1 expression, or the numbers of CD4+ TILs and CD8+ TILs were determined. Conclusions Our findings indicate that sPD-1 and sPD-L1 are independent prognostic factors with opposite prognostic roles in predicting both DFS and OS in HCC patients. Electronic supplementary material The online version of this article (10.1007/s00262-018-2271-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Boyang Chang
- Department of Vascular Interventional Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, People's Republic of China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Tao Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China.,Department of Medical Imaging and Interventional Radiology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Huajun Wei
- Department of Medical Oncology, Gaozhou People's Hospital, Gaozhou, 525200, Guangdong, People's Republic of China
| | - Lujun Shen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China.,Department of Medical Imaging and Interventional Radiology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Duo Zhu
- Department of Vascular Interventional Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Wenjun He
- Department of Medical Statistic and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Qifeng Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China.,Department of Medical Imaging and Interventional Radiology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Huihua Zhang
- RayBiotech, Inc, Guangzhou, 510600, Guangdong, People's Republic of China.,South China Biochip Research Center, Guangzhou, 510600, Guangdong, People's Republic of China
| | - Yunjian Li
- RayBiotech, Inc, Guangzhou, 510600, Guangdong, People's Republic of China.,South China Biochip Research Center, Guangzhou, 510600, Guangdong, People's Republic of China
| | - Ruopan Huang
- South China Biochip Research Center, Guangzhou, 510600, Guangdong, People's Republic of China.,RayBiotech, Inc, Norcross, GA, 30092, USA
| | - Wang Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China. .,Department of Medical Imaging and Interventional Radiology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
| | - Peihong Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China. .,Department of Medical Imaging and Interventional Radiology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
40
|
Yang B, Liu T, Qu Y, Liu H, Zheng SG, Cheng B, Sun J. Progresses and Perspectives of Anti-PD-1/PD-L1 Antibody Therapy in Head and Neck Cancers. Front Oncol 2018; 8:563. [PMID: 30547012 PMCID: PMC6279860 DOI: 10.3389/fonc.2018.00563] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/12/2018] [Indexed: 12/14/2022] Open
Abstract
Head and neck cancer is the 6th most common malignancy worldwide and urgently requires novel therapy methods to change the situation of low 5-years survival rate and poor prognosis. Targeted therapy provides more precision, higher efficiency while lower adverse effects than traditional treatments like surgery, radiotherapy, and chemotherapy. Blockade of PD-1 pathway with antibodies against PD-1 or PD-L1 is such a typical targeted therapy which reconstitutes anti-tumor activity of T cell in treatments of cancers, especially those highly expressing PD-L1, including head and neck cancers. There are many clinical trials all over the world and FDA has approved anti-PD-1/PD-L1 drugs for head and neck cancers. However, with the time going, the dark side of this therapy has emerged, including some serious side effects and drug resistance. Novel materials like nanoparticles and combination therapy have been developed to improve the efficacy. At the same time, standards for evaluation of activity and safety are to be established for this new therapy. Here we provide a systematic review with comprehensive depth on the application of anti-PD1/PD-L1 antibodies in head and neck cancer treatment: mechanism, drugs, clinical studies, influencing factors, adverse effects and managements, and the potential future developments.
Collapse
Affiliation(s)
- Bo Yang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Tingjun Liu
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yang Qu
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Hangbo Liu
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Song Guo Zheng
- Division of Rheumatology, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Bin Cheng
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jianbo Sun
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
41
|
Gu D, Ao X, Yang Y, Chen Z, Xu X. Soluble immune checkpoints in cancer: production, function and biological significance. J Immunother Cancer 2018; 6:132. [PMID: 30482248 PMCID: PMC6260693 DOI: 10.1186/s40425-018-0449-0] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoints play important roles in immune regulation, and blocking immune checkpoints on the cell membrane is a promising strategy in the treatment of cancer. Based on this, monoclonal antibodies are having much rapid development, such as those against CTLA-4 (cytotoxic T lymphocyte antigen 4) and PD-1 (programmed cell death protein 1).But the cost of preparation of monoclonal antibodies is too high and the therapeutic effect is still under restrictions. Recently, a series of soluble immune checkpoints have been found such as sCTLA-4 (soluble CTLA-4) and sPD-1 (soluble PD-1). They are functional parts of membrane immune checkpoints produced in different ways and can be secreted by immune cells. Moreover, these soluble checkpoints can diffuse in the serum. Much evidence has demonstrated that these soluble checkpoints are involved in positive or negative immune regulation and that changes in their plasma levels affect the development, prognosis and treatment of cancer. Since they are endogenous molecules, they will not induce immunological rejection in human beings, which might make up for the deficiencies of monoclonal antibodies and enhance the utility value of these molecules. Therefore, there is an increasing need for investigating novel soluble checkpoints and their functions, and it is promising to develop relevant therapies in the future. In this review, we describe the production mechanisms and functions of various soluble immune checkpoint receptors and ligands and discuss their biological significance in regard to biomarkers, potential candidate drugs, therapeutic targets, and other topics.
Collapse
Affiliation(s)
- Daqian Gu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, 400042 People’s Republic of China
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| | - Xiang Ao
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, 400042 People’s Republic of China
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| | - Yu Yang
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, 400042 People’s Republic of China
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| | - Zhuo Chen
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, 400042 People’s Republic of China
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| | - Xiang Xu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, 400042 People’s Republic of China
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
42
|
Chen Z, Lin H, Hu K, Su R, Lai N, Yang Z, Kang S. [Soluble PD-1 over-expression enhances the anti-tumor effect of senescence tumor cell vaccine against breast cancer cell growth in tumor-bearing mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:20-26. [PMID: 33177023 DOI: 10.3969/j.issn.1673-4254.2018.01.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate whether soluble PD-1 overexpression in 4T1 senescence tumor cells enhances the antitumor effect of senescence tumor cell vaccine (STCV) against breast tumor cells in a tumor-bearing mouse model. METHODS 4T1 cells were treated with interferon-γ (IFN-γ) and the expression of PD-L1 was detected by flow cytometry. CCK8 assay was used to compare the cell proliferation activity between 4T1 cells and 4T1 cells infected by a lentiviral vector of sPD-1 (4T1/sPD-1 cells), and the expressions of sPD-1 mRNA and protein in 4T1/sPD-1 cells were detected using qPCR and Western blotting. The culture supernatant of 4T1/sPD-1 cells was added in 4T1 cells pre-treated with IFN-γ, and PD-1-positive 4T1 cells were detected with flow cytometry. Senescence β-galactosidase staining kit was used to detect the senescent 4T1 and 4T1/sPD-1 cells following exposure to X-ray radiation and Veliparib. Balb/c mice bearing subcutaneous 4T1 tumor xenografts were treated with injections of PBS, 4T1 STCV, or 4T1/sPD-1 STCV, and tumor growth was observed. RESULTS Stimulation with IFN-γ concentration-dependently up-regulated PD-L1 expression by as much as (84.80 ± 1.03)% in 4T1 cells (P < 0.001). sPD-1 overexpression in 4T1 cells did not significantly affect the cell proliferation. Treatment of 4T1 cells with 4T1/sPD-1 cell culture supernatant significantly increased the proportion of PD-1 + cells from (6.893 ± 0.271)% to (55.450 ± 0.555)% (P < 0.001). X-ray irradiation combined with Veliparib caused obvious senescence in 4T1 and 4T1/sPD-1 cells. In the tumor-preventing experiment, tumor formation occurred in all the mice in PBS group; 28.787% of the mice in 4T1 STCV group and 55.556% in 4T1/sPD-1 STCV group showed no tumor formation. In the tumor treatment experiment, tumor formation occurred in all the mice in PBS groups while in 4T1 STCV and 4T1/sPD-1 STCV groups, 11.111% and 38.89% of the mice were tumor-free during the observation period, respectively. CONCLUSIONS Senescence tumor cells vaccine has antitumor effect against breast cancer in mice, and sPD-1 overexpression can enhance this effect of the vaccine.
Collapse
Affiliation(s)
- Zehong Chen
- Department of Oncology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Huiwen Lin
- Department of Oncology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Kang Hu
- Department of Oncology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Ruxiong Su
- Department of Pharmacy, Puning Peoples' Hospital, Southern Medical University, Puning 515300, China
| | - Nan Lai
- Department of Oncology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Zike Yang
- Department of Oncology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Shijun Kang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
43
|
Soluble PD-1 and PD-L1: predictive and prognostic significance in cancer. Oncotarget 2017; 8:97671-97682. [PMID: 29228642 PMCID: PMC5722594 DOI: 10.18632/oncotarget.18311] [Citation(s) in RCA: 245] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 05/22/2017] [Indexed: 12/16/2022] Open
Abstract
The membrane-bound molecules programmed death 1 (PD-1) and its ligand PD-L1 (PD-1/PD-L1) belong to the immune checkpoint pathway. PD-1 pathway downregulates effector T cells in immune response, thereby causing immune suppression. Recent studies have revealed that membrane-bound PD-1 and PD-L1 also have soluble forms. These soluble forms increase the complexity and diversity of the composition and function of the PD-1/PD-L1 signaling pathway. However, the exact roles of these molecules remain unknown. The objective of this systematic review was to elucidate the biological significance of soluble PD-1/PD-L1 in human cancers and evaluate whether they are potential diagnostic, therapeutic, or prognostic biomarkers. We expect to provide new clues for future research on soluble PD-1/PD-L1 pathway in human malignant tumors.
Collapse
|
44
|
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
|
45
|
Xiong HY, Ma TT, Wu BT, Lin Y, Tu ZG. IL-12 regulates B7-H1 expression in ovarian cancer-associated macrophages by effects on NF-κB signalling. Asian Pac J Cancer Prev 2015; 15:5767-72. [PMID: 25081699 DOI: 10.7314/apjcp.2014.15.14.5767] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND AND AIM B7-H1, a co-inhibitory molecule of the B7 family, is found aberrantly expressed in ovarian cancer cells and infiltrating macrophage/dendritic-like cells, and plays a critical role in immune evasion by ovarian cancer. IL-12, an inducer of Th1 cell development, exerts immunomodulatory effects on ovarian cancer. However, whether IL-12 regulates B7-H1 expression in human ovarian cancer associated-macrophages has not been clarified. Therefore, we investigated the effects of IL-12 on the expression of B7-H1 in ovarian cancer-associated macrophages and possible mechanisms. METHODS PMA induced THP-1-derived macrophages or human monocyte-derived macrophages were treated with recombinant IL-12 (rIL-12) or infected with adenovirus carrying human IL-12 gene (Ad-IL-12-GFP) for 24 h, then cocultured with the SKOV3 ovarian cancer cell line for another 24 h. Macrophages were collected for real-time PCR and Western blot to detect the expression of B7-H1, and activation of the NF-κB signaling pathway. Moreover, supernatants were collected to assay for IL-12, IFN-γ and IL-10 by ELISA. In addition, monocyte-derived macrophages treated with IFN-γ were cocultured with SKOV3 and determined for the expression of B7-H1. Furthermore, the expression of B7-H1 in monocyte-derived macrophages was also evaluated after blocking NF-κB signaling. RESULTS The expression of B7-H1 was significantly upregulated in monocyte-derived macrophages treated with rIL-12 or Ad-IL-12-GFP compared with the control groups (p<0.05), accompanied by a remarkable upregulation of IFN-γ (p<0.05), a marked downregulation of IL-10 (p<0.05) and activation of NF-κB signaling. However, the upregulation of B7- H1 was inhibited by blocking the NF-κB signaling pathway (p<0.05). Expression of B7-H1 was also increased (p<0.05) in monocyte-derived macrophages treated with IFN-γ and cocultured with SKOV3. By contrast, the expression of B7-H1 in THP-1-derived macrophages was significantly decreased when treated in the same way as monocyte-derived macrophages (p<0.05), and IL-10 was also significantly decreased but IFN-γ was almost absent. CONCLUSIONS IL-12 upregulates the expression of B7-H1 in monocyte-derived macrophages, which is possible though inducing the secretion of IFN-γ and further activating the NF-κB signal pathway. However, IL-12 downregulates the expression of B7-H1 in THP-1-derived macrophages, associated with a lack of IFN-γ and inhibition of expression of IL-10.
Collapse
Affiliation(s)
- Hai-Yu Xiong
- College of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics of Education Ministry, Chongqing Medical University, Chongqing, China E-mail :
| | | | | | | | | |
Collapse
|
46
|
Luo J, Luo Y, Sun J, Zhou Y, Zhang Y, Yang X. Adeno-associated virus-mediated cancer gene therapy: current status. Cancer Lett 2014; 356:347-56. [PMID: 25444906 DOI: 10.1016/j.canlet.2014.10.045] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/30/2014] [Accepted: 10/31/2014] [Indexed: 01/18/2023]
Abstract
Gene therapy is one of the frontiers of modern medicine. Adeno-associated virus (AAV)-mediated gene therapy is becoming a promising approach to treat a variety of diseases and cancers. AAV-mediated cancer gene therapies have rapidly advanced due to their superiority to other gene-carrying vectors, such as the lack of pathogenicity, the ability to transfect both dividing and non-dividing cells, low host immune response, and long-term expression. This article reviews and provides up to date knowledge on AAV-mediated cancer gene therapy.
Collapse
Affiliation(s)
- Jingfeng Luo
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Qingchun Road NO.3, Hangzhou, Zhejiang, China
| | - Yuxuan Luo
- Department of Nephrology, Zhuji People's Hospital, Zhuji, Zhejiang, China
| | - Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Qingchun Road NO.3, Hangzhou, Zhejiang, China
| | - Yurong Zhou
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Qingchun Road NO.3, Hangzhou, Zhejiang, China
| | - Yajing Zhang
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Qingchun Road NO.3, Hangzhou, Zhejiang, China
| | - Xiaoming Yang
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Qingchun Road NO.3, Hangzhou, Zhejiang, China; Image-Guided Bio-Molecular Intervention Research, Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
47
|
Dai S, Jia R, Zhang X, Fang Q, Huang L. The PD-1/PD-Ls pathway and autoimmune diseases. Cell Immunol 2014; 290:72-9. [PMID: 24908630 DOI: 10.1016/j.cellimm.2014.05.006] [Citation(s) in RCA: 278] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 04/17/2014] [Accepted: 05/07/2014] [Indexed: 12/27/2022]
Abstract
The programmed death (PD)-1/PD-1 ligands (PD-Ls) pathway, is a new member of the B7/CD28 family, and consists of the PD-1 receptor and its ligands PD-L1 (B7-H1, CD274) and PD-L2 (B7-DC, CD273). Recently, it is reported that PD-1, PD-L1 and PD-L2 also have soluble forms aside from their membrane bound forms. The soluble forms increase the diversity and complexity of PD-1/PD-Ls pathway in both composition and function. The PD-1/PD-Ls pathway is broadly expressed and exerts a wider range of immunoregulatory roles in T-cell activation and tolerance compared with other B7/CD28 family members. Studies show that the PD-1/PD-Ls pathway regulates the induction and maintenance of peripheral tolerance and protects tissues from autoimmune attack in physiological conditions. In addition, it is also involved in various diseases mediated by T cells, such as autoimmunity, tumor immunity, chronic viral infections, and transplantation immunity. In this review, we will summarize the relevance of the soluble forms and the latest researches on the role of PD-1/PD-Ls pathway in autoimmune diseases.
Collapse
Affiliation(s)
- Suya Dai
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People's Republic of China
| | - Ru Jia
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People's Republic of China
| | - Xiao Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People's Republic of China
| | - Qiwen Fang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People's Republic of China
| | - Lijuan Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People's Republic of China.
| |
Collapse
|
48
|
Wang YH, Cao YW, Yang XC, Niu HT, Sun LJ, Wang XS, Liu J. Effect of TLR4 and B7-H1 on Immune Escape of Urothelial Bladder Cancer and its Clinical Significance. Asian Pac J Cancer Prev 2014; 15:1321-6. [DOI: 10.7314/apjcp.2014.15.3.1321] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|