101
|
Ponce de León C, Angel López-Casado M, Lorite P, Palomeque T, Isabel Torres M. Dysregulation of the PD-1/PD-L1 pathway contributes to the pathogenesis of celiac disease. Cell Mol Immunol 2019; 16:777-779. [PMID: 31243357 DOI: 10.1038/s41423-019-0256-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 06/06/2019] [Indexed: 01/18/2023] Open
Affiliation(s)
| | | | - Pedro Lorite
- Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Teresa Palomeque
- Department of Experimental Biology, University of Jaén, Jaén, Spain
| | | |
Collapse
|
102
|
Wu X, Gu Z, Chen Y, Chen B, Chen W, Weng L, Liu X. Application of PD-1 Blockade in Cancer Immunotherapy. Comput Struct Biotechnol J 2019; 17:661-674. [PMID: 31205619 PMCID: PMC6558092 DOI: 10.1016/j.csbj.2019.03.006] [Citation(s) in RCA: 346] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/26/2019] [Accepted: 03/10/2019] [Indexed: 02/08/2023] Open
Abstract
The programmed cell death protein 1 (PD-1) pathway has received considerable attention due to its role in eliciting the immune checkpoint response of T cells, resulting in tumor cells capable of evading immune surveillance and being highly refractory to conventional chemotherapy. Application of anti-PD-1/PD-L1 antibodies as checkpoint inhibitors is rapidly becoming a promising therapeutic approach in treating tumors, and some of them have successfully been commercialized in the past few years. However, not all patients show complete responses and adverse events have been noted, suggesting a better understanding of PD-1 pathway mediated immunosuppression is needed to predict patient response and improve treatment efficacy. Here, we review the progresses on the studies of the mechanistic role of PD-1 pathway in the tumor immune evasion, recent clinical development and commercialization of PD-1 pathway inhibitors, the toxicities associated with PD-1 blockade observed in clinical trials as well as how to improve therapeutic efficacy and safety of cancer immunotherapy.
Collapse
Key Words
- 5-AZA-dC, 5-aza-2′-deoxycytidine
- ADCC, Antibody-dependent cellular cytotoxicity
- AEs, Adverse events
- AP1, Activator protein 1
- APCs, Antigen presenting cells
- ASCT, Autologous stem cell transplantation
- B2M, β2 microglobulin
- BATF, Basic leucine zipper transcriptional factor ATF-like
- BICR, Blinded Independent Central Review
- BV, Brentuximab vedotin
- CC, Cervical cancer
- CRC, Colorectal cancer
- CTLA-4, Cytotoxic T-lymphocyte–associated antigen 4
- CXCL9, C-X-C motif chemokine ligand 9
- Checkpoint blockade
- DCM, Dilated cardiomyopathy
- DCs, Dendritic cells
- DNMT, DNA methyltransferase
- DOR, Duration overall response
- DZNep, 3-Deazaneplanocin A
- ERK, Extracellular signal–regulated kinase
- EZH2, Enhancer of zeste homolog 2
- GC, Gastric cancer
- GEJ, GASTRIC or gastroesophageal junction
- HCC, Hepatocellular carcinoma
- HNSCC, Head and neck squamous cell carcinoma
- HR, Hazard ratio
- ICC, Investigator-choice chemotherapy
- ICOS, Inducible T-cell co-stimulator
- IFN, Interferon
- IHC, Immunohistochemistry
- ITIM, Immune-receptortyrosine-based inhibitory motif
- ITSM, Immune-receptortyrosine-based switch motif
- ITT, Intention-to-treat
- Immune surveillance
- Immunotherapy
- IrAEs, Immune related adverse events
- JMJD3, Jumonji Domain-Containing Protein 3
- LAG3, Lymphocyte-activation gene 3
- LCK, Tyrosine-protein kinase Lck
- MAP, Mitogen-activated protein
- MCC, Merkel cell carcinoma
- MHC, Major histocompatibility
- MSI-H, Microsatellite instability-high
- NF-κB, Nuclear factor-κB
- NFAT, Nuclear factor of activated T cells
- NSCLC, Non-small cell lung cancer
- ORR, Overall response rate
- OS, Overall survival
- PD-1
- PD-1, Programmed cell death 1
- PD-L1
- PD-L1, Programmed death-ligand 1
- PFS, Progression-free survival
- PI3K, Phosphoinositide 3-kinase
- PKC, Protein kinase C
- PMBCL, Primary mediastinal large B-cell lymphoma
- PRC2, Polycomb repressive complex 2
- PTEN, Phosphatase and tensin homolog
- PTPs, Protein tyrosine phosphatases
- RCC, Renal cell carcinoma
- SCLC, Small cell lung cancer
- SHP2, Src homology 2 domain-containing phosphatase 2
- SIRPα, Signal-regulatory protein alpha
- TCR, T-cell receptor
- TGF, Transforming growth factor
- TIICs, Tumor infiltrating immune cells
- TILs, Tumor-infiltrating lymphocytes
- TIM3, T-cell immunoglobulin and mucin-domain containing-3
- TMB, Tumor mutation burden
- TME, Tumor microenvironment
- UC, Urothelial carcinoma
- VEGF, Vascular endothelial growth factor
- ZAP70, Zeta-chain-associated protein kinase 70
- cHL, Classical Hodgkin lymphoma
- cTnI, Cardiac troponin I
- dMMR, DNA mismatch repair deficiency
Collapse
Affiliation(s)
- Xiaomo Wu
- Dermatology Institute of Fuzhou, Dermatology Hospital of Fuzhou, Xihong Road 243, Fuzhou 350025, PR China
- Department of Biomedicine, University of Basel, Klingelbergstr. 70, CH-4056 Basel, Switzerland
| | - Zhongkai Gu
- The Institute of Biomedical Sciences, Fudan University, Mingdao Building, Dongan Road 131, Shanghai 200032, PR China
| | - Yang Chen
- Dermatology Institute of Fuzhou, Dermatology Hospital of Fuzhou, Xihong Road 243, Fuzhou 350025, PR China
| | - Borui Chen
- Dermatology Institute of Fuzhou, Dermatology Hospital of Fuzhou, Xihong Road 243, Fuzhou 350025, PR China
| | - Wei Chen
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou 350005, PR China
| | - Liqiang Weng
- Dermatology Institute of Fuzhou, Dermatology Hospital of Fuzhou, Xihong Road 243, Fuzhou 350025, PR China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Xihong Road 312, Fuzhou 350025, PR China
| |
Collapse
|
103
|
Li X, Yuan Z, Chen J, Wang T, Shen Y, Chen L, Wen F. Microarray analysis reveals the changes of circular RNA expression and molecular mechanism in acute lung injury mouse model. J Cell Biochem 2019; 120:16658-16667. [PMID: 31106457 DOI: 10.1002/jcb.28924] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/19/2019] [Indexed: 02/05/2023]
Abstract
Acute lung injury (ALI) is a severe disease with sudden onset, rapid progression, poor treatment response, and high mortality. An increasing number of studies had found that circular RNAs (circRNAs) has significant functions in various diseases, while the role of circRNAs in ALI is not yet clear. The purpose of this study was to find circRNAs related to ALI and their mechanism of action. Expression profiles of lung circRNAs and messenger RNAs (mRNAs) were analyzed by microarray in the ALI mice models and healthy controlled mice. Differentially expressed RNAs were identified, function and pathways were analyzed by bioinformatics analysis. Moreover, the results of the microarray were verified by real-time PCR. We identified 2262 differentially expressed mRNAs and 581 circRNAs between ALI mice and control. Validation of candidate circRNAs by real-time PCR indicates that the majority of circRNAs identified by microarray are reliable and worthy of further study. ALI induced circRNAs primarily function in the metabolic regulatory process. Moreover, differentially expressed circRNAs were mainly involved in signaling pathways of mitogen-activated protein kinases, focal adhesion, FoxO, neurotrophin, and Wnt. In addition, a competitive endogenous RNA network was constructed to further interpret the molecular mechanism of ALI. This study observed significantly changed circRNAs profiles in LPS-induced mouse model and revealed a potential role of circRNAs in ALI.
Collapse
Affiliation(s)
- Xiaoou Li
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Zhicheng Yuan
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Jun Chen
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Tao Wang
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Yongchun Shen
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Lei Chen
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Fuqiang Wen
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| |
Collapse
|
104
|
Tumangelova-Yuzeir K, Naydenov E, Ivanova-Todorova E, Krasimirova E, Vasilev G, Nachev S, Kyurkchiev D. Mesenchymal Stem Cells Derived and Cultured from Glioblastoma Multiforme Increase Tregs, Downregulate Th17, and Induce the Tolerogenic Phenotype of Monocyte-Derived Cells. Stem Cells Int 2019; 2019:6904638. [PMID: 31191680 PMCID: PMC6525812 DOI: 10.1155/2019/6904638] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/08/2019] [Accepted: 02/24/2019] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess immunosuppressive properties and have been described in the tumor microenvironment of glioblastoma multiforme (GBM). This manuscript has two major topics-first, to describe isolated and cultured MSCs derived from GBM (GB-MSCs) and second, to examine their in vitro immunosuppressive capacity. Our results display cells with morphology and phenotype, clonogenic ability, and osteogenic potential, typical for MSCs. Furthermore, the cultured cells show intracellular expression of the neural markers Nestin and GFAP. They express PD-L1 and secrete TGFβ, CCL-2, PGE2, IL-6, and sVEGF. Coculturing of GB-MSCs with PBMCs isolated from healthy donors results in a decreased percentage of Th17 lymphocytes and an increased percentage of Tregs. Regarding the impact of GB-MSCs on monocytes, we establish an augmented expression of CD14 and CD86 along with diminished expression of HLA-DR and CD80, which is associated with tolerogenic phenotype monocyte-derived cells. In conclusion, our results describe in detail GBM-derived and cultured cells that meet the criteria for MSCs but at the same time express Nestin and GFAP. GB-MSCs express and secrete suppressive molecules, influencing in vitro T cells and monocytes, and are probably another factor involved in the immune suppression exerted by GBM.
Collapse
Affiliation(s)
- Kalina Tumangelova-Yuzeir
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Emanuil Naydenov
- Clinic of Neurosurgery, University Hospital “St. Ivan Rilski,” Medical University Sofia, 15 “Acad. Ivan Geshov” Str., 1431 Sofia, Bulgaria
| | - Ekaterina Ivanova-Todorova
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Ekaterina Krasimirova
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Georgi Vasilev
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Sevdalin Nachev
- Laboratory of Clinical Pathology, University Hospital “St. Ivan Rilski,” Medical University Sofia, 15 “Acad. Ivan Geshov” Str., 1431 Sofia, Bulgaria
| | - Dobroslav Kyurkchiev
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| |
Collapse
|
105
|
de Castro LL, Lopes-Pacheco M, Weiss DJ, Cruz FF, Rocco PRM. Current understanding of the immunosuppressive properties of mesenchymal stromal cells. J Mol Med (Berl) 2019; 97:605-618. [PMID: 30903229 DOI: 10.1007/s00109-019-01776-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/17/2019] [Accepted: 03/11/2019] [Indexed: 12/14/2022]
Abstract
Several studies have demonstrated the anti-inflammatory potential of mesenchymal stromal cells (MSCs) isolated from bone marrow, adipose tissue, placenta, and other sources. Nevertheless, MSCs may also induce immunosuppression when administered systemically or directly to injured environments, as shown in different preclinical disease models. MSCs express certain receptors, including toll-like receptors and the aryl-hydrocarbon receptor, that are activated by the surrounding environment, thus leading to modulation of their immunosuppressive activity. Once MSCs are activated, they can affect a wide range of immune cells (e.g., neutrophils, monocytes/macrophages, dendritic cells, natural killer cells, T and B lymphocytes), a phenomenon that has been correlated to secretion of several mediators (e.g., indolamine 2,3-dioxygenase, galectins, prostaglandin E2, nitric oxide, and damage- and pathogen-associated molecular patterns) and stimulation of certain signaling pathways (e.g., protein kinase R, signal transducer and activator of transcription-1, nuclear factor-κB). Additionally, MSC manipulation and culture conditions, as well as the number of passages, duration of cryopreservation, and O2 content available, can significantly affect the immunosuppressive properties of MSCs. This review sheds light on current knowledge regarding the mechanisms by which MSCs exert immunosuppressive effects both in vitro and in vivo, focusing on the receptors expressed by MSCs, the correlation between soluble factors secreted by MSCs and their immunosuppressive effects, and interactions between MSCs and immune cells.
Collapse
Affiliation(s)
- Ligia Lins de Castro
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Daniel Jay Weiss
- Department of Medicine, College of Medicine, University of Vermont, Burlington, VT, USA
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Patricia Rieken Macêdo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil. .,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.
| |
Collapse
|
106
|
Zhao Y, Han P, Liu L, Wang X, Xu P, Wang H, Yu T, Sun Y, Li L, Sun T, Liu X, Zhou H, Qiu J, Wang L, Peng J, Xu S, Hou M. Indirubin modulates CD4 + T-cell homeostasis via PD1/PTEN/AKT signalling pathway in immune thrombocytopenia. J Cell Mol Med 2019; 23:1885-1898. [PMID: 30609280 PMCID: PMC6378207 DOI: 10.1111/jcmm.14089] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 12/12/2022] Open
Abstract
Immune thrombocytopenia (ITP) is an acquired autoimmune disease characterized by an immune mediated decrease in platelet number. Disturbance of CD4+ T-cell homeostasis with simultaneous decrease of CD4+ CD25+ Foxp3+ regulatory T cells (Tregs) as well as unrestricted proliferation and activation of peripheral CD4+ effector T cells underpin the pathophysiology of ITP. Indirubin is an active ingredient of a traditional Chinese herb called Indigofera tinctoria L. which is clinically used for the treatment of ITP patients. Whether indirubin targets the Tregs/effector T cell-axis to restore platelet number is unknown. In our in vitro studies, Indirubin could significantly enhance the number and function of Tregs and meanwhile dampen the activation of effector T cells in a dose-dependent manner. Indirubin was observed to restore the expression of programmed cell-death 1 (PD1) and phosphatase and tensin homolog (PTEN) on the CD4+ T cells of ITP patients, leading to the subsequent attenuation of the AKT/mTOR pathway. Furthermore, these observations were recapitulated in an active murine model of ITP with a prominent platelet response. Thus, our results identified a potentially novel mechanism of the therapeutic action of indirubin in the treatment of ITP through regulating the homeostasis of CD4+ T cells in a PD1/PTEN/AKT signalling pathway.
Collapse
Affiliation(s)
- Yajing Zhao
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Panpan Han
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Lei Liu
- Department of UrologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Xiaojie Wang
- Department of DermatologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Pengcheng Xu
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Haoyi Wang
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Tianshu Yu
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Yunqi Sun
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Lizhen Li
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Tao Sun
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Xinguang Liu
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Hai Zhou
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Jihua Qiu
- Department of Geriatric MedicineQilu Hospital, Shandong UniversityJinanShandongChina
| | - Liang Wang
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Jun Peng
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
- Shandong Provincial Key Laboratory of ImmunohaematologyQilu Hospital, Shandong UniversityJinanShandongChina
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong UniversityJinanShandongChina
| | - Shuqian Xu
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
| | - Ming Hou
- Department of HaematologyQilu Hospital, Shandong UniversityJinanShandongChina
- Shandong Provincial Key Laboratory of ImmunohaematologyQilu Hospital, Shandong UniversityJinanShandongChina
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong UniversityJinanShandongChina
- Leading Research Group of Scientific Innovation, Department of Science and Technology of Shandong ProvinceQilu Hospital, Shandong UniversityJinanShandongChina
| |
Collapse
|
107
|
Predictive Value of Soluble Programmed Death-1 for Severe Sepsis and Septic Shock During the First Week in an Intensive Care Unit. Shock 2019; 51:289-297. [DOI: 10.1097/shk.0000000000001171] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
108
|
Mihailovic PM, Lio WM, Herscovici R, Chyu KY, Yano J, Zhao X, Zhou J, Zhou B, Freeman MR, Yang W, Shah PK, Cercek B, Dimayuga PC. Keratin 8 is a potential self-antigen in the coronary artery disease immunopeptidome: A translational approach. PLoS One 2019; 14:e0213025. [PMID: 30811493 PMCID: PMC6392305 DOI: 10.1371/journal.pone.0213025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 02/13/2019] [Indexed: 12/31/2022] Open
Abstract
Background Inflammation is an important risk factor in atherosclerosis, the underlying cause of coronary artery disease (CAD). Unresolved inflammation may result in maladaptive immune responses and lead to immune reactivity to self-antigens. We hypothesized that inflammation in CAD patients would manifest in immune reactivity to self-antigens detectable in soluble HLA-I/peptide complexes in the plasma. Methods Soluble HLA-I/peptide complexes were immuno-precipitated from plasma of male acute coronary syndrome (ACS) patients or age-matched controls and eluted peptides were subjected to mass spectrometry to generate the immunopeptidome. Self-peptides were ranked according to frequency and signal intensity, then mouse homologs of selected peptides were used to test immunologic recall in spleens of male apoE-/- mice fed either normal chow or high fat diet. The peptide detected with highest frequency in patient plasma samples and provoked T cell responses in mouse studies was selected for use as a self-antigen to stimulate CAD patient peripheral blood mononuclear cells (PBMCs). Results The immunopeptidome profile identified self-peptides unique to the CAD patients. The mouse homologs tested showed immune responses in apoE-/- mice. Keratin 8 was selected for further study in patient PBMCs which elicited T Effector cell responses in CAD patients compared to controls, associated with reduced PD-1 mRNA expression. Conclusion An immunopeptidomic strategy to search for self-antigens potentially involved in CAD identified Keratin 8. Self-reactive immune response to Keratin 8 may be an important factor in the inflammatory response in CAD.
Collapse
Affiliation(s)
- Peter M. Mihailovic
- Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Wai Man Lio
- Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Romana Herscovici
- Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Kuang-Yuh Chyu
- Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Juliana Yano
- Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Xiaoning Zhao
- Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Jianchang Zhou
- Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Bo Zhou
- Division of Cancer Biology and Therapeutics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Michael R. Freeman
- Division of Cancer Biology and Therapeutics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Wei Yang
- Division of Cancer Biology and Therapeutics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Prediman K. Shah
- Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Bojan Cercek
- Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Paul C. Dimayuga
- Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
109
|
Mohammadi S, Saghaeian Jazi M, Zare Ebrahimabad M, Eghbalpour F, Abdolahi N, Tabarraei A, Yazdani Y. Interleukin 10 gene promoter polymorphisms (rs1800896, rs1800871 and rs1800872) and haplotypes are associated with the activity of systemic lupus erythematosus and IL10 levels in an Iranian population. Int J Immunogenet 2019; 46:20-30. [PMID: 30430731 DOI: 10.1111/iji.12407] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/21/2018] [Accepted: 09/30/2018] [Indexed: 02/05/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with unknown aetiology. According to the role of interleukin 10 (IL10) in SLE pathogenesis, the genetic alterations in its promoter region could be associated with elevated IL10 levels and exacerbated disease. Here, we investigated the association of genotype and haplotype frequencies of three IL10 gene promoter polymorphisms with susceptibility to SLE, IL10 plasma levels and disease activity of patients in an Iranian population. A total of 116 SLE patients and 131 healthy subjects were enrolled. The PCR-RFLP technique was used to detect IL10 promoter genotypes at the positions of -1082 (G/A), -819 (C/T) and -592 (C/A) in association with IL10 plasma levels and SLEDAI scores. The GG genotype of -1082 polymorphism was associated with the increased risk of SLE [OR = 2.65, 95% CI (1.21-5.82), p-value = 0.046]. The CC genotype in -819 region was associated with SLE susceptibility [OR = 3.38, 95% CI (1.26-9.07), p-value = 0.034] and C allele was introduced as risk allele [OR = 1.86, 95% CI (1.15-3.01), p-value = 0.009] in this region. IL10 plasma levels were overexpressed in CC genotype carriers of -592 SNP and decreased in AA genotype carriers of -1082. IL10 was also increased in SLE patients with CGT (-592/-1082/-819) haplotype. The SLEDAI score was higher among CC genotype carriers at the position of -592 and TT genotype carriers at the region of -819. SLEDAI was also elevated among patients with CGC (-592/-1082/-819) and CAC (p = 0.011) haplotypes. The present study suggests that the IL10 -819(C/T), -1082(G/A) and -592(C/A) polymorphisms and the haplotypes are associated with SLE susceptibility, increased disease activity and elevated IL10 levels. While this is the first time to report such an association in an Iranian population, further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Saeed Mohammadi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Marie Saghaeian Jazi
- Biochemistry and Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mojtaba Zare Ebrahimabad
- Department of Biochemistry, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Farnaz Eghbalpour
- Department of Molecular medicine, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Nafiseh Abdolahi
- Golestan Rheumatology Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Alijan Tabarraei
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Yaghoub Yazdani
- Infectious Diseases Research Center and Laboratory Science Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
110
|
Yaghoubi N, Soltani A, Ghazvini K, Hassanian SM, Hashemy SI. PD-1/ PD-L1 blockade as a novel treatment for colorectal cancer. Biomed Pharmacother 2019; 110:312-318. [DOI: 10.1016/j.biopha.2018.11.105] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/10/2018] [Accepted: 11/25/2018] [Indexed: 12/14/2022] Open
|
111
|
Tajiri K, Ieda M. Cardiac Complications in Immune Checkpoint Inhibition Therapy. Front Cardiovasc Med 2019; 6:3. [PMID: 30729114 PMCID: PMC6351438 DOI: 10.3389/fcvm.2019.00003] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/07/2019] [Indexed: 12/31/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have changed the treatment landscape of advanced cancers. Unfortunately, these agents can induce a wide spectrum of immune-related adverse events (irAEs) through activation of immune responses in non-target organs, including the heart. As the clinical use of ICI therapy increases rapidly, management of irAEs is becoming extremely important. The most commonly presented cardiac irAE is myocarditis. Histopathologically, T-cell (with a predominance of CD8+ cells) and macrophage infiltration in the myocardium is typically observed in ICI-associated myocarditis. Other presentations of cardiac irAEs include congestive heart failure, Takotsubo cardiomyopathy, pericardial disease, arrhythmias, and conduction disease. Although cardiac irAEs are relatively rare, they can be life-threatening. Hence, cardiologists and oncologists should be vigilant for these presentations.
Collapse
Affiliation(s)
- Kazuko Tajiri
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masaki Ieda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
112
|
Yu J, Lin Y, Xiong X, Li K, Yao Z, Dong H, Jiang Z, Yu D, Yeung SCJ, Zhang H. Detection of Exosomal PD-L1 RNA in Saliva of Patients With Periodontitis. Front Genet 2019; 10:202. [PMID: 30923536 PMCID: PMC6426748 DOI: 10.3389/fgene.2019.00202] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 02/26/2019] [Indexed: 02/05/2023] Open
Abstract
Periodontitis is the most prevalent inflammatory disease of the periodontium, and is related to oral and systemic health. Exosomes are emerging as non-invasive biomarker for liquid biopsy. We here evaluated the levels of programmed death-ligand 1 (PD-L1) mRNA in salivary exosomes from patients with periodontitis and non-periodontitis controls. The purposes of this study were to establish a procedure for isolation and detection of mRNA in exosomes from saliva of periodontitis patients, to characterize the level of salivary exosomal PD-L1, and to illustrate its clinical relevance. Bioinformatics analysis suggested that periodontitis was associated with an inflammation gene expression signature, that PD-L1 expression positively correlated with inflammation in periodontitis based on gene set enrichment analysis (GSEA) and that PD-L1 expression was remarkably elevated in periodontitis patients versus control subjects. Exosomal RNAs were successfully isolated from saliva of 61 patients and 30 controls and were subjected to qRT-PCR. Levels of PD-L1 mRNA in salivary exosomes were higher in periodontitis patients than controls (P < 0.01). Salivary exosomal PD-L1 mRNA showed significant difference between the stages of periodontitis. In summary, the protocols for isolating and detecting exosomal RNA from saliva of periodontitis patients were, for the first time, characterized. The current study suggests that assay of exosomes-based PD-L1 mRNA in saliva has potential to distinguish periodontitis from the healthy, and the levels correlate with the severity/stage of periodontitis.
Collapse
Affiliation(s)
- Jialiang Yu
- Department of Stomatology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yusheng Lin
- Cancer Research Center, Shantou University Medical College, Shantou, China
| | - Xiao Xiong
- Cancer Research Center, Shantou University Medical College, Shantou, China
| | - Kai Li
- Cancer Research Center, Shantou University Medical College, Shantou, China
| | - Zhimeng Yao
- Cancer Research Center, Shantou University Medical College, Shantou, China
| | - Hongmei Dong
- Cancer Research Center, Shantou University Medical College, Shantou, China
- Institute of Precision Cancer and Pathology, Jinan University Medical College, Guangzhou, China
| | - Zuojie Jiang
- Cancer Research Center, Shantou University Medical College, Shantou, China
| | - Dan Yu
- Department of Stomatology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hao Zhang
- Institute of Precision Cancer and Pathology, Jinan University Medical College, Guangzhou, China
- Research Center of Translational Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Hao Zhang,
| |
Collapse
|
113
|
Furubayashi N, Negishi T, Uozumi T, Takamatsu D, Shiraishi K, Hirose D, Nakamura M. Isolated adrenocorticotropic hormone deficiency potentially induced by nivolumab following pseudo-progression in clear cell renal cell carcinoma: A case report. Mol Clin Oncol 2018; 10:304-308. [PMID: 30680212 DOI: 10.3892/mco.2018.1781] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/27/2018] [Indexed: 01/05/2023] Open
Abstract
Nivolumab is a monoclonal immunoglobulin G antibody blocking programmed death receptor-1 (PD-1) that promotes the restoration of the natural T-cell-mediated immune response against cancer cells; however, it also causes a number of autoimmune-related adverse events (irAEs) that often involve the endocrine system. The present report describes a 71-year-old man with clear cell renal cell carcinoma metastasis in the lung. Following the 14th course of nivolumab therapy, the patient complained of general malaise, loss of appetite and mild consciousness disturbance. Laboratory tests revealed a severely elevated eosinophil ratio (26.2%) and low sodium value (122 mmol/l). Endocrine system tests revealed that the patient's adrenocorticotropic hormone (ACTH; 4.5 pg/ml) and cortisol (0.1 µg/dl) levels were lower than normal, while those of other pituitary hormones were higher than normal. This case was therefore diagnosed as isolated ACTH deficiency induced by nivolumab. Magnetic resonance imaging (MRI) showed normal pituitary glands. Hydrocortisone replacement therapy improved the clinical symptoms early and enabled the patient to restart nivolumab therapy. Isolated ACTH deficiency due to nivolumab, a PD-1 immune checkpoint inhibitor antibody, is a rare occurrence. This report may be useful for avoiding delays in the diagnosis and treatment of this life-threatening irAE even if no pituitary abnormalities are identified via MRI.
Collapse
Affiliation(s)
- Nobuki Furubayashi
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Takahito Negishi
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Tomoharu Uozumi
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Dai Takamatsu
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Koichi Shiraishi
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Daisuke Hirose
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Motonobu Nakamura
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| |
Collapse
|
114
|
Tio M, Rai R, Ezeoke OM, McQuade JL, Zimmer L, Khoo C, Park JJ, Spain L, Turajlic S, Ardolino L, Yip D, Goldinger SM, Cohen JV, Millward M, Atkinson V, Kane AY, Ascierto PA, Garbe C, Gutzmer R, Johnson DB, Rizvi HA, Joshua AM, Hellmann MD, Long GV, Menzies AM. Anti-PD-1/PD-L1 immunotherapy in patients with solid organ transplant, HIV or hepatitis B/C infection. Eur J Cancer 2018; 104:137-144. [PMID: 30347289 PMCID: PMC10176037 DOI: 10.1016/j.ejca.2018.09.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 09/20/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Anti-programmed cell death protein 1/programmed death ligand 1 (PD-1/PD-L1) immunotherapy is now routinely used to treat several cancers. Clinical trials have excluded several populations, including patients with solid organ transplant, HIV infection and hepatitis B/C infection. We examined the safety outcomes of these populations treated with anti-PD-1/PD-L1 treatment in a multicentre retrospective study. METHODS Patients from 16 centres with advanced cancer and solid organ transplant, HIV infection or hepatitis B/C infection were included. Demographic, tumour, treatment, toxicity and outcome data were recorded. RESULTS Forty-six patients were included for analysis, with a median age of 60 years, and the majority of patients diagnosed with melanoma (72%). Among six patients with solid organ transplants, two graft rejections occurred, with one resulting in death, whereas two patients achieved partial responses. There were four responses in 12 patients with HIV infection. In 14 patients with hepatitis B, there were three responses, and similarly, there were three responses in 14 patients with hepatitis C. There was no unexpected toxicity in any viral infection group or an increase in viral load. CONCLUSION Patients with HIV or hepatitis B/C infections treated with anti-PD-1/PD-L1 immunotherapy may respond to treatment without increased toxicity. Given the risk of graft rejection in solid organ transplant patients and also the potential for response, the role of anti-PD-1/PD-L1 immunotherapy needs to be carefully considered.
Collapse
Affiliation(s)
- Martin Tio
- Melanoma Institute Australia, Sydney, Australia.
| | - Rajat Rai
- Melanoma Institute Australia, Sydney, Australia
| | | | | | - Lisa Zimmer
- University of Duisburg-Essen Hospital, Heidelberg, Germany
| | - Chloe Khoo
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | - John J Park
- Crown Princess Mary Cancer Centre, Sydney, Australia; Westmead Hospital, Sydney, Australia
| | - Lavinia Spain
- Skin and Renal Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Samra Turajlic
- Skin and Renal Unit, The Royal Marsden NHS Foundation Trust, London, UK; Francis Crick Institute, London, UK
| | | | - Desmond Yip
- The Canberra Hospital, Canberra, Australia; ANU Medical School, Australian National University, Canberra, Australia
| | | | | | | | | | - Alisa Y Kane
- Liverpool Hospital, Sydney, Australia; Garvan Institute, Sydney, Australia
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | | | | | | | - Hira A Rizvi
- Memorial Sloan Kettering Cancer Center, New York, USA
| | - Anthony M Joshua
- Melanoma Institute Australia, Sydney, Australia; St Vincents Hospital, Sydney, Australia; University of New South Wales, Sydney, Australia
| | | | - Georgina V Long
- Melanoma Institute Australia, Sydney, Australia; The University of Sydney, Sydney, Australia; Royal North Shore Hospital, Sydney, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, Sydney, Australia; The University of Sydney, Sydney, Australia; Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|
115
|
Gianchecchi E, Fierabracci A. Inhibitory Receptors and Pathways of Lymphocytes: The Role of PD-1 in Treg Development and Their Involvement in Autoimmunity Onset and Cancer Progression. Front Immunol 2018; 9:2374. [PMID: 30386337 PMCID: PMC6199356 DOI: 10.3389/fimmu.2018.02374] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
Regulatory T (Treg) cells represent a subpopulation of suppressor CD4+ T cells critically involved in the establishment of peripheral tolerance through the inhibition of effector T (Teff) cells and the suppression of the immune-mediated tissue destruction toward self-antigens. Treg generation, their suppressive properties and also Treg-Teff cell interactions could be modulated at least in part by programmed cell death-1 (PD-1) expression on their surface and through binding between PD-1 and programmed cell death ligand-1 (PD-L1). Defects involving PD-1 and Tregs can lead to the development of pathological conditions, including autoimmune disorders or promote cancer progression by favoring tumor evasion from the host immune response. At the same time, PD-1 and Tregs could represent attractive targets for treatment, as demonstrated by the therapeutic blockade of PD-L1 applied for the management of different cancer conditions in humans. In the present Review, we focus specifically the role of PD-1/PD-L1 on Treg development and activity.
Collapse
Affiliation(s)
- Elena Gianchecchi
- Infectivology and Clinical Trials Research Department, Children's Hospital Bambino Gesù, Rome, Italy
- VisMederi S.r.l., Siena, Italy
| | - Alessandra Fierabracci
- Infectivology and Clinical Trials Research Department, Children's Hospital Bambino Gesù, Rome, Italy
| |
Collapse
|
116
|
Grywalska E, Pasiarski M, Góźdź S, Roliński J. Immune-checkpoint inhibitors for combating T-cell dysfunction in cancer. Onco Targets Ther 2018; 11:6505-6524. [PMID: 30323625 PMCID: PMC6177399 DOI: 10.2147/ott.s150817] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Under normal conditions, the immune system responds effectively to both external and internal threats without damaging healthy tissues. Cells undergoing a neoplastic transformation are one such threat. An efficient activation of T cells is enabled by T-cell receptor (TCR) interactions with antigen-presenting class I and class II molecules of the major histocompatibility complex (MHC), co-stimulatory molecules, and cytokines. After threatening stimuli are removed from the body, the host's immune response ceases, which prevents tissue damage or chronic inflammation. The recognition of foreign antigens is highly selective, which requires multistep regulation to avoid reactions against the antigens of healthy cells. This multistep regulation includes central and peripheral tolerance toward the body's own antigens. Here, we discuss T-cell dysfunction, which leads to poor effector function against foreign antigens, including cancer. We describe selected cellular receptors implicated in T-cell dysfunction and discuss how immune-checkpoint inhibitors can help overcome T-cell dysfunction in cancer treatment.
Collapse
Affiliation(s)
- Ewelina Grywalska
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, Lublin, Poland,
| | - Marcin Pasiarski
- Department of Hematology, Holy Cross Oncology Center of Kielce, Kielce, Poland.,Faculty of Health Sciences, Jan Kochanowski University, Kielce, Poland
| | - Stanisław Góźdź
- Faculty of Health Sciences, Jan Kochanowski University, Kielce, Poland.,Department of Oncology, Holy Cross Oncology Center of Kielce, Kielce, Poland
| | - Jacek Roliński
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, Lublin, Poland,
| |
Collapse
|
117
|
LaFleur MW, Muroyama Y, Drake CG, Sharpe AH. Inhibitors of the PD-1 Pathway in Tumor Therapy. THE JOURNAL OF IMMUNOLOGY 2018; 200:375-383. [PMID: 29311378 DOI: 10.4049/jimmunol.1701044] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/05/2017] [Indexed: 12/13/2022]
Abstract
The programmed death 1 (PD-1) pathway delivers inhibitory signals that function as a brake for immune responses. This pathway limits the initiation and duration of immune responses, thereby protecting tissues from immune-mediated damage and autoimmune diseases. However, the PD-1 pathway also inhibits immune responses to tumors. The critical role of PD-1 in preventing antitumor immunity is demonstrated by the transformative effects of PD-1 pathway blockade in a broad range of cancers with the hallmark of durability of response. Despite this success, most patients do not respond to PD-1 monotherapy, and some patients experience adverse events. In this review, we discuss the functions of the PD-1 pathway and its translation to cancer immunotherapy. We also consider current challenges and opportunities for PD-1 cancer immunotherapy, including mechanisms of response and resistance, identification of biomarkers of response to PD-1 therapy, characterization and treatment of PD-1 therapy-related adverse events, and development of safe and effective combination therapies.
Collapse
Affiliation(s)
- Martin W LaFleur
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115.,Department of Pediatric Oncology, Dana Farber Cancer Institute, Boston, MA 02115
| | - Yuki Muroyama
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231.,Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032
| | - Charles G Drake
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; .,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032; and
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115; .,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115
| |
Collapse
|
118
|
Cardiac Complications Associated With Checkpoint Inhibition: A Systematic Review of the Literature in an Important Emerging Area. Can J Cardiol 2018; 34:1059-1068. [DOI: 10.1016/j.cjca.2018.03.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/06/2018] [Accepted: 03/16/2018] [Indexed: 01/22/2023] Open
|
119
|
Immunometabolism of T cells and NK cells: metabolic control of effector and regulatory function. Inflamm Res 2018; 67:813-828. [PMID: 30066126 DOI: 10.1007/s00011-018-1174-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 01/10/2023] Open
Abstract
Metabolic flux can dictate cell fate, including immune cell effector and regulatory function. The metabolic regulation of cell function is well characterized with respect to effector, memory, and regulatory T cells. This knowledge may allow for manipulation of T cell metabolic pathways that set the stage for more effective T cell therapy. Natural Killer (NK) and T-lymphocytes have complementary roles in the defense against pathogens. However, studies of NK cell metabolism are only beginning to emerge and there is comparatively little knowledge on the metabolic regulation of NK-cell activation and effector function. Given their common lymphoid lineage, effector functions and cellular memory potential our current knowledge on T cell metabolism could inform investigation of metabolic reprogramming in NK cells. In this review, we compare the current knowledge of metabolic regulation in T cell and NK cell development, activation, effector and memory function. Commonalties in glucose transport, hypoxia-inducible factors and mTOR highlight metabolic control points in both cells types. Contrasting the glycolytic and oxidative nodes of metabolic regulation in T cells versus NK cells may provide insight into the contribution of specific immune responses to disease and promote the development of immunotherapeutic approaches targeting both innate and adaptive immune responses.
Collapse
|
120
|
Andersen MH. The T-win® technology: immune-modulating vaccines. Semin Immunopathol 2018; 41:87-95. [PMID: 29968045 DOI: 10.1007/s00281-018-0695-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 06/14/2018] [Indexed: 12/25/2022]
Abstract
The T-win® technology is an innovative investigational approach designed to activate the body's endogenous anti-regulatory T cells (anti-Tregs) to target regulatory as well as malignant cells. Anti-Tregs are naturally occurring T cells that can directly react against regulatory immune cells because they recognize proteins that these targets express, including indoleamine 2,3-dioxygenase (IDO), tryptophan 2,6-dioxygenase, arginase, and programmed death ligand 1 (PD-L1). The T-win® technology is characterized by therapeutic vaccination with long peptide epitopes derived from these antigens and therefore offers a novel way to target genetically stable cells with regular human leukocyte antigen expression in the tumor microenvironment. The T-win® technology thus also represents a novel way to attract pro-inflammatory cells to the tumor microenvironment where they can directly affect immune inhibitory pathways, potentially altering tolerance to tumor antigens. The modification of an immune regulatory environment into a pro-inflammatory milieu potentiates effective anti-tumor T cell responses. Many regulatory immune cells may be reverted into effector cells given the right stimulus. Because T-win® technology is based on the immune-modulatory function of the vaccines, the vaccines activate both CD4 and CD8 anti-Tregs. Of importance, in clinical trials, vaccinations against IDO or PD-L1 to potentiate anti-Tregs have so far proved to be safe, with minimal toxicity.
Collapse
Affiliation(s)
- Mads Hald Andersen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, DK-2730, Herlev, Denmark. .,Department of Immunology and Microbiology, University of Copenhagen, DK-2200, Copenhagen, Denmark. .,IO Biotech ApS, DK-2200, Copenhagen, Denmark.
| |
Collapse
|
121
|
Zhang Y, Liu Z, Tian M, Hu X, Wang L, Ji J, Liao A. The altered PD-1/PD-L1 pathway delivers the 'one-two punch' effects to promote the Treg/Th17 imbalance in pre-eclampsia. Cell Mol Immunol 2018; 15:710-723. [PMID: 28890543 PMCID: PMC6123412 DOI: 10.1038/cmi.2017.70] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 06/27/2017] [Accepted: 06/30/2017] [Indexed: 02/07/2023] Open
Abstract
The programmed cell death-1 (PD-1)/PD-ligand 1 (PD-L1) pathway is critical for normal pregnancy by promoting regulatory T (Treg) cell development and inhibiting the Th17 response. However, the relationship between the PD-1/PD-L1 pathway and the Treg/Th17 imbalance in pre-eclampsia (PE) is an enigma. In this study, decreased PD-1 and PD-L1 expression and a Treg/Th17 imbalance were observed at the maternal-fetal interface in PE. The regulatory effects of the PD-1/PD-L1 pathway on the Treg and Th17 cell quantities were determined in vitro by targeting T-cell proliferation, differentiation and transdifferentiation. First, decreased PD-1 expression might contribute to a higher Th17 cell frequency by promoting proliferation in PE. Second, the percentages of Treg but not Th17 cells differentiated from peripheral naive CD4+ T cells were increased by PD-L1 Fc administration. This effect was accompanied by decreased PI3K/AKT/m-TOR and increased PTEN mRNA expression and was completely reversed by PD-1 blockade. Finally, the percentage of IL-17-producing Treg cells increased and was positively associated with the Th17 cell frequency in PE. Increased RORγt and IL-17 but not Foxp3 and IL-10 mRNA expression by Treg cells was observed with PD-1 blockade. Similar findings occurred when Treg cells were exposed to IL-6/IL-23/IL-1β and were reversed by PD-L1 Fc. Taken together, our findings indicate that the PD-1/PD-L1 pathway contributes to the Treg/Th17 imbalance via 'one-two punch' approaches: (i) promoting Th17 cell proliferation, (ii) inhibiting Treg cell differentiation and (iii) enhancing Treg cell plasticity into Th17 cells in PE. The therapeutic value of PD-L1 Fc for PE treatment will be explored in the future.
Collapse
Affiliation(s)
- Yonghong Zhang
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Zhaozhao Liu
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Mei Tian
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Xiaohui Hu
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Liling Wang
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Jinlu Ji
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Aihua Liao
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.
| |
Collapse
|
122
|
Matsuda K, Miyoshi H, Hiraoka K, Hamada T, Yoshida S, Ishibashi Y, Haraguchi T, Shiba N, Ohshima K. Clinicopathological value of programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1) expression in synovium of patients with rheumatoid arthritis. Clin Exp Med 2018; 18:487-494. [DOI: 10.1007/s10238-018-0515-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 06/24/2018] [Indexed: 12/31/2022]
|
123
|
Du J, Qin Y, Wu Y, Zhao W, Zhai W, Qi Y, Wang C, Gao Y. The design of high affinity human PD-1 mutants by using molecular dynamics simulations (MD). Cell Commun Signal 2018; 16:25. [PMID: 29879980 PMCID: PMC5992718 DOI: 10.1186/s12964-018-0239-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/28/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Programmed cell death protein 1 (PD-1), a negative co-stimulatory molecule, plays crucial roles in immune escape. Blockade of the interaction between PD-1 and PD-L1 shows exciting clinical responses in a fraction of cancer patients and the success makes PD-1 as a valuable target in immune checkpoint therapy. For the rational design of PD-1 targeting modulators, the ligand binding mechanism of PD-1 should be well understood in prior. METHODS In this study, we applied 50 ns molecular dynamics simulations to observe the structural properties of PD-1 molecule in both apo and ligand bound states, and we studied the structural features of PD-1 in human and mouse respectively. RESULTS The results showed that the apo hPD-1 was more flexible than that in PD-L1 bound state. We unexpectedly found that K135 was important for binding energy although it was not at the binding interface. Moreover, the residues which stabilized the interactions with PD-L1 were distinguished. Taking the dynamic features of these residues into account, we identified several residual sites where mutations may gain the function of ligand binding. The in vitro binding experiments revealed the mutants M70I, S87 W, A129L, A132L, and K135 M were better in ligand binding than the wild type PD-1. CONCLUSIONS The structural information from MD simulation combined with in silico mutagenesis provides guidance to design engineered PD-1 mutants to modulate the PD-1/PD-L1 pathway.
Collapse
Affiliation(s)
- Jiangfeng Du
- School of Life Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, China
| | - Yaping Qin
- School of Life Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, China
| | - Yahong Wu
- School of Life Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, China
| | - Wenshan Zhao
- School of Life Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, China
| | - Wenjie Zhai
- School of Life Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, China
| | - Yuanming Qi
- School of Life Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, China
| | - Chuchu Wang
- School of Life Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, China
| | - Yanfeng Gao
- School of Life Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, China.
| |
Collapse
|
124
|
Xi X, Liu JM, Guo JY. Correlation of PD-1/PD-L1 Signaling Pathway with Treg/Th17 Imbalance from Asthmatic Children. Int Arch Allergy Immunol 2018; 176:255-267. [PMID: 29874664 DOI: 10.1159/000489338] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 04/17/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The balance between T helper 17 (Th17) and regulatory T cells (Treg) is a new paradigm in asthma pathogenesis, but no therapeutic targets could modulate the Th17/Treg balance specifically for asthma. Since previous studies have shown the programmed cell death-1(PD-1)/PD-ligand 1 (PD-L1) pathway is critical to immune homeostasis in this disease, we hypothesized that the PD-1/PD-L1 pathway might be involved in the regulation of Treg/Th17 imbalance in asthmatic children. METHODS The percentage of Treg and Th17 cells and the expression of PD-1 and PD-L1 were detected by flow cytometry in children with asthma and healthy controls. CD4+ T cells were stimulated with Th17 and Treg differentiating factors, and treated with anti-PD-1. Then cells were harvested and measured for Th17 and Treg percentages and Foxp3 and RORγt levels using RT-PCR. RESULTS We observed an inverse correlation between the percentages of Treg and Th17 cells, and the expression of PD-1 and PD-L1 in the two subsets also changed in the mild persistent and moderate to severe persistent groups compared with healthy controls. In vitro, administration of anti-PD-1 could decrease Th17 percentages and RORγt mRNA, and increase Treg percentages and Foxp3 mRNA in CD4+ T cells of children with asthma in the mild persistent and moderate to persistent groups. Additionally, the role played by anti-PD-1 in regulating Treg/Th17 balance was further confirmed in an asthmatic mouse model. CONCLUSION Alteration of the PD-1/PD-L1 pathway can modulate Treg/Th17 balance in asthmatic children. Treatment with anti-PD-1 posed protective effects on asthma models, providing a novel theoretical target for asthma.
Collapse
Affiliation(s)
- Xia Xi
- Department of Paediatrics, Dongying People's Hospital, Dongying, China
| | - Jing-Mei Liu
- Department of Clinical Laboratory, Dongying People's Hospital, Dongying, China
| | - Jun-Ying Guo
- Department of Paediatrics, Dongying People's Hospital, Dongying, China
| |
Collapse
|
125
|
Nguyen T, Avci NG, Shin DH, Martinez-Velez N, Jiang H. Tune Up In Situ Autovaccination against Solid Tumors with Oncolytic Viruses. Cancers (Basel) 2018; 10:E171. [PMID: 29857493 PMCID: PMC6025332 DOI: 10.3390/cancers10060171] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/17/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023] Open
Abstract
With the progress of immunotherapy in cancer, oncolytic viruses (OVs) have attracted more and more attention during the past decade. Due to their cancer-selective and immunogenic properties, OVs are considered ideal candidates to be combined with immunotherapy to increase both specificity and efficacy in cancer treatment. OVs preferentially replicate in and lyse cancer cells, resulting in in situ autovaccination leading to adaptive anti-virus and anti-tumor immunity. The main challenge in OV approaches is how to redirect the host immunity from anti-virus to anti-tumor and optimize the clinical outcome of cancer patients. Here, we summarize the conceptual updates on oncolytic virotherapy and immunotherapy in cancer, and the development of strategies to enhance the virus-mediated anti-tumor immune response, including: (1) arm OVs with cytokines to modulate innate and adaptive immunity; (2) combining OVs with immune checkpoint inhibitors to release T cell inhibition; (3) combining OVs with immune co-stimulators to enhance T cell activation. Future studies need to be enforced on developing strategies to augment the systemic effect on metastasized tumors.
Collapse
Affiliation(s)
- Teresa Nguyen
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 6767 Bertner St., Houston, TX 77030, USA.
| | - Naze G Avci
- Neurosurgery Research, Houston Methodist Research Institute, Houston, TX 77030, USA.
| | - Dong Ho Shin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 6767 Bertner St., Houston, TX 77030, USA.
| | | | - Hong Jiang
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 6767 Bertner St., Houston, TX 77030, USA.
| |
Collapse
|
126
|
Greisen S, Kunder R, Deleuran B. T cell co-stimulatory factors. Rheumatology (Oxford) 2018; 56:861-862. [PMID: 27402872 DOI: 10.1093/rheumatology/kew276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2016] [Indexed: 11/13/2022] Open
Affiliation(s)
- Stinne Greisen
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | - Rebecca Kunder
- Department of Pediatric Rheumatology, Stanford Hospital and Clinics, Palo Alto, CA, USA
| | - Bent Deleuran
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
127
|
Freitas CMT, Johnson DK, Weber KS. T Cell Calcium Signaling Regulation by the Co-Receptor CD5. Int J Mol Sci 2018; 19:E1295. [PMID: 29701673 PMCID: PMC5983667 DOI: 10.3390/ijms19051295] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/19/2018] [Accepted: 04/24/2018] [Indexed: 12/21/2022] Open
Abstract
Calcium influx is critical for T cell effector function and fate. T cells are activated when T cell receptors (TCRs) engage peptides presented by antigen-presenting cells (APC), causing an increase of intracellular calcium (Ca2+) concentration. Co-receptors stabilize interactions between the TCR and its ligand, the peptide-major histocompatibility complex (pMHC), and enhance Ca2+ signaling and T cell activation. Conversely, some co-receptors can dampen Ca2+ signaling and inhibit T cell activation. Immune checkpoint therapies block inhibitory co-receptors, such as cytotoxic T-lymphocyte associated antigen 4 (CTLA-4) and programmed death 1 (PD-1), to increase T cell Ca2+ signaling and promote T cell survival. Similar to CTLA-4 and PD-1, the co-receptor CD5 has been known to act as a negative regulator of T cell activation and to alter Ca2+ signaling and T cell function. Though much is known about the role of CD5 in B cells, recent research has expanded our understanding of CD5 function in T cells. Here we review these recent findings and discuss how our improved understanding of CD5 Ca2+ signaling regulation could be useful for basic and clinical research.
Collapse
Affiliation(s)
- Claudia M Tellez Freitas
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84604, USA.
| | - Deborah K Johnson
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84604, USA.
| | - K Scott Weber
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84604, USA.
| |
Collapse
|
128
|
Lee NP, Wu H, Ng KT, Luo R, Lam TW, Lo CM, Man K. Transcriptome Analysis of Acute Phase Liver Graft Injury in Liver Transplantation. Biomedicines 2018; 6:biomedicines6020041. [PMID: 29642405 PMCID: PMC6027418 DOI: 10.3390/biomedicines6020041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Liver transplantation remains the treatment of choice for a selected group of hepatocellular carcinoma (HCC) patients. However, the long-term benefit is greatly hampered by post-transplant HCC recurrence. Our previous studies have identified liver graft injury as an acute phase event leading to post-transplant tumor recurrence. METHODS To re-examine this acute phase event at the molecular level and in an unbiased way, RNA sequencing (RNA-Seq) was performed on liver graft biopsies obtained from the transplant recipients two hours after portal vein reperfusion with an aim to capture frequently altered pathways that account for post-transplant tumor recurrence. Liver grafts from recurrent recipients (n = 6) were sequenced and compared with those from recipients without recurrence (n = 5). RESULTS RNA expression profiles comparison pointed to several frequently altered pathways, among which pathways related to cell adhesion molecules were the most involved. Subsequent validation using quantitative polymerase chain reaction confirmed the differential involvement of two cell adhesion molecules HFE (hemochromatosis) and CD274 and their related molecules in the acute phase event. CONCLUSION This whole transcriptome strategy unravels the molecular landscape of liver graft gene expression alterations, which can identify key pathways and genes that are involved in acute phase liver graft injury that may lead to post-transplant tumor recurrence.
Collapse
Affiliation(s)
- Nikki P. Lee
- Department of Surgery, The University of Hong Kong, Hong Kong, China; (K.T.P.N.); (C.-M.L.); (K.M.)
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China
- Correspondence: ; Tel: +852-3917-9652; Fax: +852-3917-9634
| | - Haiyang Wu
- Department of Computer Science, The University of Hong Kong, Hong Kong, China; (H.W.); (R.L.); (T.-W.L.)
| | - Kevin T.P. Ng
- Department of Surgery, The University of Hong Kong, Hong Kong, China; (K.T.P.N.); (C.-M.L.); (K.M.)
| | - Ruibang Luo
- Department of Computer Science, The University of Hong Kong, Hong Kong, China; (H.W.); (R.L.); (T.-W.L.)
| | - Tak-Wah Lam
- Department of Computer Science, The University of Hong Kong, Hong Kong, China; (H.W.); (R.L.); (T.-W.L.)
| | - Chung-Mau Lo
- Department of Surgery, The University of Hong Kong, Hong Kong, China; (K.T.P.N.); (C.-M.L.); (K.M.)
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China
| | - Kwan Man
- Department of Surgery, The University of Hong Kong, Hong Kong, China; (K.T.P.N.); (C.-M.L.); (K.M.)
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
129
|
Ishibashi N, Watanabe T, Kanehira M, Watanabe Y, Hoshikawa Y, Notsuda H, Noda M, Sakurada A, Ohkouchi S, Kondo T, Okada Y. Bone marrow mesenchymal stromal cells protect allograft lung transplants from acute rejection via the PD-L1/IL-17A axis. Surg Today 2018; 48:726-734. [PMID: 29546496 DOI: 10.1007/s00595-018-1643-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 02/06/2018] [Indexed: 12/13/2022]
Abstract
PURPOSE Using a rat model of allograft lung transplantation, we investigated the effectiveness of mesenchymal stromal cells (MSCs) as prophylactic and therapeutic agents against the acute rejection of lung grafts. METHODS Lung grafts were harvested from donor rats and transplanted orthotopically into major histocompatibility complex-mismatched rats. MSCs were administered to the recipients once (on day 0) or twice (on days 0 and 3) after transplantation. The grade of acute rejection was evaluated both macroscopically and microscopically 6 days after transplantation. To elucidate the related mechanism, mRNA levels of inflammatory cytokines and immunomodulatory receptors in the transplanted grafts were measured using quantitative RT-PCR. RESULTS The lung graft tissue from the rats that received MSCs post-surgically was protected from acute rejection significantly better than that from the untreated controls. Notably, the rats administered MSCs twice after surgery exhibited the least signs of rejection, with a markedly upregulated mRNA level of PD-L1 and a downregulated mRNA level of IL-17A. CONCLUSION This study assessed MSC protection of lung allografts from acute rejection by modulating T cell activity via enforced expression of PD-L1 in transplants and downregulation of IL-17A.
Collapse
Affiliation(s)
- Naoya Ishibashi
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.,Department of Thoracic Surgery, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Tatsuaki Watanabe
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Masahiko Kanehira
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Yui Watanabe
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yasushi Hoshikawa
- Department of Thoracic Surgery, Fujita Health University, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Hirotsugu Notsuda
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Masafumi Noda
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Akira Sakurada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Shinya Ohkouchi
- Department of Occupational Health, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Takashi Kondo
- Department of Thoracic Surgery, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Yoshinori Okada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
130
|
Gomez GVB, Rinck-Junior JA, Oliveira C, Silva DHL, Mamoni RL, Lourenço GJ, Moraes AM, Lima CSP. PDCD1 gene polymorphisms as regulators of T-lymphocyte activity in cutaneous melanoma risk and prognosis. Pigment Cell Melanoma Res 2018; 31:308-317. [PMID: 29090522 DOI: 10.1111/pcmr.12665] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 10/07/2017] [Indexed: 01/16/2023]
Abstract
This study aimed to evaluate whether PD1.1 (c.-606G>A), PD1 (c.627 + 252C>T), PD1.5 (c.804C>T), and PD1.9 (c.644C>T) single nucleotide polymorphisms of PDCD1 gene influence the risk, clinicopathological aspects, and survival of cutaneous melanoma (CM). Individuals with phototype I or II and PD1 CC genotype were under 5.89-fold increased risk of developing CM. PD1.5 TT genotype increased PDCD1 expression (2.49 versus 1.28 arbitrary units, p = .03) and PD1.5 CT or TT genotype and allele T increased PD1 expression in TCD4+ lymphocytes (16.6 versus 12.5%, p = .01; 17.0 versus 13.1%, p = .006). At 60 months of follow-up, short recurrence-free survival was seen in patients with PD1.1 AA genotype (33.3 versus 71.8%, p = .03). Patients with PD1.1 AA and PD1.5 CC genotype had 4.21 and 2.62 more chances of presenting relapse and evolving death by disease in Cox analyses, respectively. Our data provide preliminary evidence that abnormalities in regulation of T lymphocyte alter CM risk, clinical aspects, and prognosis.
Collapse
Affiliation(s)
- Gabriela V B Gomez
- Clinical Oncology Service, Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - José A Rinck-Junior
- Clinical Oncology Service, Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Cristiane Oliveira
- Clinical Oncology Service, Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Dennis H L Silva
- Department of Clinical Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Ronei L Mamoni
- Department of Clinical Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Gustavo J Lourenço
- Laboratory of Cancer Genetics, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Aparecida M Moraes
- Clinical Oncology Service, Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Carmen S P Lima
- Clinical Oncology Service, Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
131
|
PD-1 and its ligands are important immune checkpoints in cancer. Oncotarget 2018; 8:2171-2186. [PMID: 27974689 PMCID: PMC5356790 DOI: 10.18632/oncotarget.13895] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 11/21/2016] [Indexed: 12/31/2022] Open
Abstract
Checkpoint programmed death-1 (PD-1)/programmed cell death ligands (PD-Ls) have been identified as negative immunoregulatory molecules that promote immune evasion of tumor cells. The interaction of PD-1 and PD-Ls inhibits the function of T cells and tumor-infiltrating lymphocytes (TIL) while increasing the function of immunosuppressive regulatory T cells (Tregs). This condition causes the tumor cells to evade immune response. Thus, the blockade of PD-1/PD-L1 enhances anti-tumor immunity by reducing the number and/or the suppressive activity of Tregs and by restoring the activity of effector T cells. Furthermore, some monoclonal antibodies blockading PD-1/PD-Ls axis have achieved good effect and received Food and Drug Administration approval. The role of PD-1/PD-Ls in tumors has been well studied, but little is known on the mechanism by which PD-1 blocks T-cell activation. In this study, we provide a brief overview on the discovery and regulatory mechanism of PD-1 and PD-L1 dysregulation in tumors, as well as the function and signaling pathway of PD-1 and its ligands; their roles in tumor evasion and clinical treatment were also studied.
Collapse
|
132
|
Irregular antibodies in no hemolytic autoimmune diseases are able to induce erythrophagocytosis. Immunol Res 2018; 65:410-418. [PMID: 27561786 DOI: 10.1007/s12026-016-8853-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Irregular antibodies are produced by alloimmunization because of pregnancies or blood transfusions. They are called "irregular" due to target erythrocyte antigens from "rare blood systems," those different from the ABO system. Irregular antibodies have been widely investigated in immunohematology since their presence in blood donors may lead to difficulties in blood typing and in blood cross-matching, or to induce hemolytic transfusion reactions. Nevertheless, their incidence and participation in the physiopathology of autoimmune diseases have not been thoroughly studied. In this work, we analyzed the presence and pro-hemolytic capabilities of irregular antibodies in patients with different autoimmune diseases lacking signs of hemolytic anemia, in comparison with healthy multiparous women. Five of 141 autoimmune patients (3.5 %) and two of 77 multiparous women (2.6 %) were positive. Although frequency was relatively low and similar in both populations, the targeted antigens were Kell (k, Kpb, Jsb) and Luth (Lub) in multiparous women, and the same plus Duffy (Fya), Kidd (Jka) and MNS (M, s) in autoimmune patients. Irregular antibodies from autoimmune patients did not induce complement-mediated hemolysis (intravascular), but they were able to induce macrophages-mediated phagocytosis (extravascular hemolysis) in vitro. It is the first approach exploring the presence of irregular antibodies associated with the loss of immune tolerance and demonstrating their hemolytic potential in autoimmune patients without hemolytic manifestations. The presence of irregular antibodies targeted to Duffy (Fya), Kidd (Jka) and MNS (M, s) antigens only in autoimmune patients suggests a loss of immune tolerance to these erythrocyte antigens.
Collapse
|
133
|
Ochoa CE, Joseph RW. Nivolumab in Renal Cell Carcinoma: Current Trends and Future Perspectives. J Kidney Cancer VHL 2018; 5:15-18. [PMID: 29468108 PMCID: PMC5806048 DOI: 10.15586/jkcvhl.2018.102] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 01/10/2018] [Indexed: 01/05/2023] Open
Abstract
Targeted agents form the backbone of most therapeutic strategies in advanced renal cell carcinoma (aRCC) but ultimately resistance develops and toxicity often leads to discontinuation of treatment, limiting the clinical benefits of these treatments. Nivolumab, a fully human IgG4 anti-PD-1 antibody, selectively blocks the interaction between PD-1 and its ligands PD-L1 and PD-L2 and provides a novel therapy option for patients with aRCC. In 2015, the pivotal phase III study CheckMate 025 led to the Food and Drug Administration approval of nivolumab in patients with aRCC who had received prior anti-angiogenic therapy, and in 2017, the phase III study CheckMate 214 showed that combined immunotherapy with nivolumab plus ipilimumab resulted in greater objective response rate and prolonged progression-free survival when compared with sunitinib in intermediate- and poor-risk patients with previously untreated aRCC. Early studies of nivolumab in association with anti-angiogenic therapy have generated enthusiasm and multiple combination trials are ongoing.
Collapse
Affiliation(s)
- Cesar E Ochoa
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Richard W Joseph
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
134
|
Chen H, Peng Q, Yang H, Yin L, Shi J, Zhang Y, Wang G. Increased Levels of Soluble Programmed Death Ligand 1 Associate with Malignancy in Patients with Dermatomyositis. J Rheumatol 2018; 45:835-840. [DOI: 10.3899/jrheum.170544] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2017] [Indexed: 01/01/2023]
Abstract
Objective.To investigate the levels of soluble programmed death ligand 1 (sPD-L1) and evaluate its association with malignancy in patients with dermatomyositis (DM).Methods.Levels of sPD-L1 were measured in serum from 88 DM patients without malignancies (sDM), 40 with cancer-related DM (CRDM), and 30 healthy controls (HC) using ELISA. The CRDM subjects were divided into new-onset cancers (nCRDM) and stable cancers (sCRDM). Receiver-operating characteristic (ROC) curve analysis was performed to determine the cutoff sPD-L1 value that distinguished patients with nCRDM from those who were sDM. Serum antitranscriptional intermediary factor 1-γ (TIF1-γ) antibodies were detected using immunoblot, and the diagnostic values for malignancy were compared with sPD-L1 levels in patients with DM.Results.Serum sPD-L1 levels were significantly higher in sDM [median 12.3 ng/ml, interquartile range (IQR) 8.4–16.2] than in HC (median 1.3 ng/ml, IQR 0.4–2.2, p = 0.0001). Extremely high sPD-L1 levels were seen in nCRDM (median 18.5 ng/ml, IQR 13.8–22.4), much higher than those in sCRDM (median 8.5 ng/ml, IQR 6.8–11.8, p = 0.0001). The sPD-L1 levels in 4 patients with nCRDM decreased after curative cancer treatment (p = 0.013). ROC curve analysis revealed that the sPD-L1 value distinguishing nCRDM from sDM was 16.1 ng/ml, with an area under the curve value of 0.72 ± 0.04 (p = 0.0001). The combination of sPD-L1 and anti-TIF1-γ antibodies yielded greater specificity and positive predictive value in diagnosing cancer, reaching values of 95% and 70%, respectively.Conclusion.Serum sPD-L1 levels increased significantly in sDM, and markedly high sPD-L1 levels could be a diagnostic indicator for malignancies in patients with DM, especially in those with anti-TIF1-γ antibodies.
Collapse
|
135
|
Lidar M, Giat E, Garelick D, Horowitz Y, Amital H, Steinberg-Silman Y, Schachter J, Shapira-Frommer R, Markel G. Rheumatic manifestations among cancer patients treated with immune checkpoint inhibitors. Autoimmun Rev 2018; 17:284-289. [PMID: 29341936 DOI: 10.1016/j.autrev.2018.01.003] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND The use of immune checkpoint inhibitors (ICI) has grown incessantly since they were first approved in 2014. These monoclonal antibodies inhibit T cell activation, yielding a dramatic tumor response with improved survival. However, immunotherapy is frequently hampered by immune adverse events (iAE) such as hypophysitis, colitis, hepatitis, pneumonitis and rash. Until recently, rheumatic side effects were only infrequently reported. AIM To describe the rheumatic manifestations encountered among patients treated with ICIs in a large tertiary cancer center in Israel METHODS: The cancer center's patient registry was screened for patients who had ever been treated with ipilimumab, pembrolizumab and/or nivolumab with relevant data gathered from clinical charts. RESULTS Rheumatic manifestations were encountered in 14 of 400 patients (3.5%) who had received immunotherapy between January 1st 2013 and April 30th, 2017. The most common rheumatic manifestation was inflammatory arthritis (85%) for which a third (4/11) had a clear cut predisposing factor such as a personal or family history of psoriasis, a prior episode of uveitis or ACPA positivity. Pulmonary sarcoidosis and biopsy-proven eosinophilic fasciitis were diagnosed in two additional patients. Treatment with NSAIDS was mostly unsuccessful while steroid therapy was beneficial in doses ≥20 mg/d. Methotrexate enabled steroid tapering without an excess of side effects or tumor progression in the short follow-up available. Overall, rheumatic manifestations tended to occur later in the course of immunotherapy as compared to other iAE. CONCLUSIONS Our findings underscore that rheumatic iAE are part of the side effect profile of ICIs and require heightened awareness as these therapies are becoming the standard of care for various malignancies. We show that these appear later in the course of iAEs and respond preferentially to high dose steroids. MTX appears effective as a steroid sparing agent.
Collapse
Affiliation(s)
- Merav Lidar
- Rheumatology Unit, Sheba Medical Center, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Eitan Giat
- Rheumatology Unit, Sheba Medical Center, Israel
| | | | | | - Howard Amital
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Internal Medicine B, Sheba Medical Center, Israel
| | | | - Jacob Schachter
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Israel
| | | | - Gal Markel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Israel.
| |
Collapse
|
136
|
Virus-stimulated neutrophils in the tumor microenvironment enhance T cell-mediated anti-tumor immunity. Oncotarget 2018; 7:42195-42207. [PMID: 27259252 PMCID: PMC5173127 DOI: 10.18632/oncotarget.9743] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 05/22/2016] [Indexed: 12/17/2022] Open
Abstract
The tumor microenvironment (TME) fosters tumors by attenuating anti-tumor immunity, reinforcing tumor cell survival and increasing angiogenesis. Among the constituents of the TME, here, we focused on tumor-associated neutrophils (TANs). First, we found that the combination of poly I:C and inactivated Sendai virus particles (hemagglutinating virus of Japan envelope; HVJ-E) synergistically suppressed tumor growth in the B16-F10 melanoma mouse model. In this model, poly I:C contributed to the recruitment of CD11b+Ly6G+ neutrophils to the TME, and co-injection of poly I:C and HVJ-E increased CD11b+Ly6G+FAS+ TAN in the TME. Depletion of neutrophils abolished the synergistic anti-tumor effect of HVJ-E and poly I:C in B16-F10 tumors. We revealed that C-X-C motif chemokine ligand 2 (CXCL2) is produced in the TME by poly I:C, but HVJ-E enhanced neutrophil infiltration of the TME does not occur. An anti-CXCL2 antibody inhibited the tumor suppression by HVJ-E+poly I:C. HVJ-E in combination with recombinant CXCL2 protein or CXCL2 pDNA suppressed mouse melanoma by increasing cytotoxic T lymphocyte activity against B16-F10 melanoma, which was abolished by an anti-Ly6G antibody. HVJ-E directly and indirectly increased FAS and ICAM-1 expression in cultured bone marrow-derived naïve neutrophils. Thus, HVJ-E activates anti-tumor immunity via anti-tumorigenic neutrophils in the TME. An HVJ-E vector containing the CXCL2 gene may be applicable as a novel cancer gene therapy strategy.
Collapse
|
137
|
Dimitrov V, Bouttier M, Boukhaled G, Salehi-Tabar R, Avramescu RG, Memari B, Hasaj B, Lukacs GL, Krawczyk CM, White JH. Hormonal vitamin D up-regulates tissue-specific PD-L1 and PD-L2 surface glycoprotein expression in humans but not mice. J Biol Chem 2017; 292:20657-20668. [PMID: 29061851 DOI: 10.1074/jbc.m117.793885] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 09/28/2017] [Indexed: 12/14/2022] Open
Abstract
PD-L1 (programmed death ligand 1) and PD-L2 are cell-surface glycoproteins that interact with programmed death 1 (PD-1) on T cells to attenuate inflammation. PD-1 signaling has attracted intense interest for its role in a pathophysiological context: suppression of anti-tumor immunity. Similarly, vitamin D signaling has been increasingly investigated for its non-classical actions in stimulation of innate immunity and suppression of inflammatory responses. Here, we show that hormonal 1,25-dihydroxyvitamin D (1,25D) is a direct transcriptional inducer of the human genes encoding PD-L1 and PD-L2 through the vitamin D receptor, a ligand-regulated transcription factor. 1,25D stimulated transcription of the gene encoding PD-L1 in epithelial and myeloid cells, whereas the gene encoding the more tissue-restricted PD-L2 was regulated only in myeloid cells. We identified and characterized vitamin D response elements (VDREs) located in both genes and showed that 1,25D treatment induces cell-surface expression of PD-L1 in epithelial and myeloid cells. In co-culture experiments with primary human T cells, epithelial cells pretreated with 1,25D suppressed activation of CD4+ and CD8+ cells and inhibited inflammatory cytokine production in a manner that was abrogated by anti-PD-L1 blocking antibody. Consistent with previous observations of species-specific regulation of immunity by vitamin D, the VDREs are present in primate genes, but neither the VDREs nor the regulation by 1,25D is present in mice. These findings reinforce the physiological role of 1,25D in controlling inflammatory immune responses but may represent a double-edged sword, as they suggest that elevated vitamin D signaling in humans could suppress anti-tumor immunity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Benedeta Hasaj
- Microbiology and Immunology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | | | - Connie M Krawczyk
- From the Departments of Physiology, .,Microbiology and Immunology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - John H White
- From the Departments of Physiology, .,Medicine, and
| |
Collapse
|
138
|
Liu Q, An L, Qi Z, Zhao Y, Li C. Increased Expression of Programmed Cell Death-1 in Regulatory T Cells of Patients with Severe Sepsis and Septic Shock: An Observational Clinical Study. Scand J Immunol 2017; 86:408-417. [PMID: 28888058 DOI: 10.1111/sji.12612] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/04/2017] [Indexed: 12/31/2022]
Abstract
While regulatory T cells (Tregs) constitutively express programmed cell death-1 (PD-1), the role of PD-1 expression in Tregs of patients with sepsis remains unclear. Thus, we determined PD-1 expression in Tregs from the peripheral blood of patients with severe sepsis and septic shock. Seventy-eight patients with severe sepsis and 40 with septic shock, as well as 21 healthy subjects, were enrolled in this study. The percentage of peripheral blood PD-1+ Tregs, as well as absolute Treg counts, were compared between these three groups. PD-1 expression in Tregs and absolute Treg counts were also compared between survivors and non-survivors in patients with sepsis. PD-1 expression in Tregs increased in patients with sepsis compared to healthy controls. Conversely, absolute Treg counts were significantly decreased in patients with sepsis compared to healthy controls; patients with septic shock had the lowest absolute Treg counts. Among patients with sepsis, survivors had lower levels of PD-1 expression in Tregs, as well as higher absolute Treg counts, than non-survivors. Additionally, the percentage of PD-1+ Tregs correlated positively with lactate levels as well as the Acute Physiology and Chronic Health Evaluation II and Sequential Organ Failure Assessment scores in patients with sepsis. PD-1 was upregulated in Tregs of patients with sepsis and may indicate a state of immune dysfunction. Overexpression of PD-1 in Tregs was associated with more severe sepsis as well as poor outcomes.
Collapse
Affiliation(s)
- Q Liu
- Beijing Chao-yang Hospital Affiliated to Capital Medical University, Beijing, Chao-yang District, China.,Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - L An
- Beijing Chao-yang Hospital Affiliated to Capital Medical University, Beijing, Chao-yang District, China.,Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Z Qi
- Beijing Chao-yang Hospital Affiliated to Capital Medical University, Beijing, Chao-yang District, China.,Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Y Zhao
- Beijing Chao-yang Hospital Affiliated to Capital Medical University, Beijing, Chao-yang District, China.,Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - C Li
- Beijing Chao-yang Hospital Affiliated to Capital Medical University, Beijing, Chao-yang District, China.,Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| |
Collapse
|
139
|
Lou J, Zhou Y, Huang J, Qian X. Relationship Between PD-L1 Expression and Clinical Characteristics in Patients with Breast Invasive Ductal Carcinoma. Open Med (Wars) 2017; 12:288-292. [PMID: 28894845 PMCID: PMC5588755 DOI: 10.1515/med-2017-0042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 04/06/2017] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE To evaluate the expression of PD-L1 (programmed death 1 ligand 1, PD-L1) and its clinical significance in breast invasive ductal carcinoma. METHODS Tumor samples were collected from 64 cases of breast invasive ductal carcinoma patients, and tumor adjacent normal breast tissue were obtained as normal control. The expression of PD-L1 were examined by immunohistochemical staining and real time PCR assay, its correlations with patients' clinical pathological characteristics were analyzed. RESULTS PD-L1 was found to be over-expressed in 24 of 64 (37.5%) breast invasive ductal carcinoma samples, while in 1 of 22 (4.5%) tumor adjacent normal breast tissue which indicated PD-L1 was higher expressed in breast invasive ductal carcinoma samples than the tumor adjacent normal breast tissue (P < 0.05). PD-L1 positive expression was associated with clinical pathological characteristics of TNM stage and pathology grading (P < 0.05). However, PD-L1 positive expression was not correlated with age (P > 0.05), menstruation status (P >0.05), family history of breast cancer (P > 0.05), tumor diameter (P > 0.05), lymph node metastasis (P > 0.05) and tumor location (P > 0.05). CONCLUSION PD-L1 may play an important role in invasive ductal carcinoma, which could be a potential indicator for advanced clinical stage and poor prognosis.
Collapse
Affiliation(s)
- Jian Lou
- Department of Medical Oncology, Lishui Central Hospital, Lishui323000PR China
| | - Yuefen Zhou
- Department of Medical Oncology, Lishui Central Hospital, Lishui323000PR China
| | - Jianhui Huang
- Department of Medical Oncology, Lishui Central Hospital, Lishui323000PR China
| | - Xiaojun Qian
- Department of Breast and Thyroid Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Zhejiang, Shaoxing312000, China
| |
Collapse
|
140
|
Liu Q, Li CS. Programmed Cell Death-1/Programmed Death-ligand 1 Pathway: A New Target for Sepsis. Chin Med J (Engl) 2017; 130:986-992. [PMID: 28397730 PMCID: PMC5407047 DOI: 10.4103/0366-6999.204113] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Sepsis remains a leading cause of death in many Intensive Care Units worldwide. Immunosuppression has been a primary focus of sepsis research as a key pathophysiological mechanism. Given the important role of the negative costimulatory molecules programmed cell death-1 (PD-1) and programmed death-ligand 1 (PD-L1) in the occurrence of immunosuppression during sepsis, we reviewed literatures related to the PD-1/PD-L1 pathway to examine its potential as a new target for sepsis treatment. DATA SOURCES Studies of the association between PD-1/PD-L1 and sepsis published up to January 31, 2017, were obtained by searching the PubMed database. STUDY SELECTION English language studies, including those based on animal models, clinical research, and reviews, with data related to PD-1/PD-L1 and sepsis, were evaluated. RESULTS Immunomodulatory therapeutics could reverse the deactivation of immune cells caused by sepsis and restore immune cell activation and function. Blockade of the PD-1/PD-L1 pathway could reduce the exhaustion of T-cells and enhance the proliferation and activation of T-cells. CONCLUSIONS The anti-PD-1/PD-L1 pathway shows promise as a new target for sepsis treatment. This review provides a basis for clinical trials and future studies aimed at revaluating the efficacy and safety of this targeted approach.
Collapse
Affiliation(s)
- Qiang Liu
- Intensive Care Unit, Central Hospital of Dandong City, Dandong, Liaoning 118002; Department of Emergency, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Chun-Sheng Li
- Department of Emergency, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing 100020, China
| |
Collapse
|
141
|
Fang C, Huang Y, Pei Y, Zhang HH, Chen X, Guo H, Li S, Ji X, Hu J. Genome-wide gene expression profiling reveals that CD274 is up-regulated new-onset type 1 diabetes mellitus. Acta Diabetol 2017; 54:757-767. [PMID: 28577136 DOI: 10.1007/s00592-017-1005-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/19/2017] [Indexed: 11/25/2022]
Abstract
AIMS Early studies have identified type 1 diabetes mellitus (T1DM) as a disease that is caused by the autoimmune destruction of the insulin-producing pancreatic β-cells. Genetics, environment and the immune pathogenesis of T1DM are three major pillars of T1DM research. We try to understand the changes in the gene expression profile during the pathogenesis of T1DM. METHODS We performed a systematic search in the Gene Expression Omnibus (GEO) database for microarray studies of T1DM with samples taken at or before the T1DM onset. RESULTS The results of an integrated analysis of different GEO datasets and a comparison of the gene expression level in T1DM samples taken at the time of appearance of the islet autoantibodies, 1 year before T1DM onset, and at the time of T1DM onset showed that CD274, which encodes PD-L1, was up-regulated in the newly onset T1DM samples. CD274 had a stable expression level in the control samples but showed a gradual up-regulation from the appearance of autoantibodies to the onset of T1DM. CONCLUSIONS These results indicate that CD274 up-regulation in T1DM is correlated with disease pathogenesis. PD-L1 might play a protective role in preventing the pancreatic islets from autoimmune destruction, which may help researchers find strategies for preventing the destruction process of pancreas β-cells in T1DM.
Collapse
Affiliation(s)
- Chen Fang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Yun Huang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Yufang Pei
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College, Soochow University, Suzhou, 215003, Jiangsu, China
| | - Hong-Hong Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Xiaohong Chen
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Heming Guo
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Sicheng Li
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Xiaoyan Ji
- Department of Ophthalmology, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, China.
- Department of Ophthalmology, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215123, Jiangsu, China.
| | - Ji Hu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
142
|
|
143
|
Richardson DR, Ellis B, Mehmi I, Leys M. Bilateral uveitis associated with nivolumab therapy for metastatic melanoma: a case report. Int J Ophthalmol 2017; 10:1183-1186. [PMID: 28730129 DOI: 10.18240/ijo.2017.07.28] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/23/2017] [Indexed: 11/23/2022] Open
Affiliation(s)
| | - Brian Ellis
- West Virginia University Eye Institute, Morgantown, WV 26506, USA
| | - Inderjit Mehmi
- West Virginia University Cancer Institute, Morgantown, WV 26506, USA
| | - Monique Leys
- West Virginia University Eye Institute, Morgantown, WV 26506, USA
| |
Collapse
|
144
|
|
145
|
Kuo R, Saito E, Miller SD, Shea LD. Peptide-Conjugated Nanoparticles Reduce Positive Co-stimulatory Expression and T Cell Activity to Induce Tolerance. Mol Ther 2017; 25:1676-1685. [PMID: 28408181 PMCID: PMC5498812 DOI: 10.1016/j.ymthe.2017.03.032] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 12/11/2022] Open
Abstract
Targeted approaches to treat autoimmune diseases would improve upon current therapies that broadly suppress the immune system and lead to detrimental side effects. Antigen-specific tolerance was induced using poly(lactide-co-glycolide) nanoparticles conjugated with disease-relevant antigen to treat a model of multiple sclerosis. Increasing the nanoparticle dose and amount of conjugated antigen both resulted in more durable immune tolerance. To identify active tolerance mechanisms, we investigated downstream cellular and molecular events following nanoparticle internalization by antigen-presenting cells. The initial cell response to nanoparticles indicated suppression of inflammatory signaling pathways. Direct and functional measurement of surface MHC-restricted antigen showed positive correlation with both increasing particle dose from 1 to 100 μg/mL and increasing peptide conjugation by 2-fold. Co-stimulatory analysis of cells expressing MHC-restricted antigen revealed most significant decreases in positive co-stimulatory molecules (CD86, CD80, and CD40) following high doses of nanoparticles with higher peptide conjugation, whereas expression of a negative co-stimulatory molecule (PD-L1) remained high. T cells isolated from mice immunized against myelin proteolipid protein (PLP139-151) were co-cultured with antigen-presenting cells administered PLP139-151-conjugated nanoparticles, which resulted in reduced T cell proliferation, increased T cell apoptosis, and a stronger anti-inflammatory response. These findings indicate several potential mechanisms used by peptide-conjugated nanoparticles to induce antigen-specific tolerance.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/drug effects
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/pathology
- Antigens/chemistry
- Antigens/immunology
- Antigens/pharmacology
- B7-1 Antigen/genetics
- B7-1 Antigen/immunology
- B7-2 Antigen/genetics
- B7-2 Antigen/immunology
- CD40 Antigens/genetics
- CD40 Antigens/immunology
- Delayed-Action Preparations/administration & dosage
- Delayed-Action Preparations/chemistry
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Female
- Gene Expression
- Immune Tolerance/drug effects
- Immunoconjugates/chemistry
- Immunoconjugates/metabolism
- Immunoconjugates/pharmacology
- Mice
- Mice, Inbred C57BL
- Myelin Proteolipid Protein/chemistry
- Myelin Proteolipid Protein/immunology
- Myelin Proteolipid Protein/pharmacology
- Nanoparticles/administration & dosage
- Nanoparticles/chemistry
- Ovalbumin/chemistry
- Ovalbumin/immunology
- Ovalbumin/pharmacology
- Particle Size
- Polyglactin 910/chemistry
- Polyglactin 910/metabolism
- Primary Cell Culture
- Spleen/drug effects
- Spleen/immunology
- Spleen/pathology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- Robert Kuo
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eiji Saito
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Northwestern University, Chicago, IL 60611, USA
| | - Lonnie D Shea
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
146
|
Zhao Y, Jia Y, Li C, Fang Y, Shao R. The risk stratification and prognostic evaluation of soluble programmed death-1 on patients with sepsis in emergency department. Am J Emerg Med 2017; 36:43-48. [PMID: 28716595 DOI: 10.1016/j.ajem.2017.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 07/02/2017] [Accepted: 07/02/2017] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE To evaluate the efficacy of soluble programmed death-1 (sPD-1) for risk stratification and prediction of 28-day mortality in patients with sepsis, we compared serum sPD-1 with procalcitonin (PCT), C-reactive protein (CRP), and the Mortality in Emergency Department Sepsis (MEDS) score. METHODS A total of 60 healthy volunteers and 595 emergency department (ED) patients were recruited for this prospective cohort study. According to the severity of their condition on ED arrival, the patients were allocated to the systemic inflammatory response syndrome group (130 cases), sepsis group (276 cases), severe sepsis group (121 cases), and septic shock group (68 cases). In addition, all patients with sepsis were also divided into the survivor group (349 cases) and nonsurvivor group (116 cases) according to the 28-day outcomes. RESULTS When the severity of sepsis increased, the levels of sPD-1 gradually increased. The levels of sPD-1, PCT, CRP and the MEDS score were also higher in the nonsurvivor group compared to the survivor group. Logistic regression suggested that sPD-1, PCT, and the MEDS score were independent risk factors for 28-day mortality of patients with sepsis. Area under the curve (AUC) of sPD-1, PCT and the MEDS score for 28-day mortality was 0.725, 0.693, and 0.767, respectively, and the AUC was improved when all 3 factors were combined (0.843). CONCLUSION Serum sPD-1 is positively correlated with the severity of sepsis, and it is valuable for risk stratification of patients and prediction of 28-day mortality. Combining sPD-1 with PCT and the MEDS score improves the prognostic evaluation.
Collapse
Affiliation(s)
- Yongzhen Zhao
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Cardiopulmonary-Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing,China
| | - Yumei Jia
- Department of Endocrinology, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Chunsheng Li
- Beijing Key Laboratory of Cardiopulmonary-Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing,China.
| | - Yingying Fang
- Beijing Key Laboratory of Cardiopulmonary-Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing,China
| | - Rui Shao
- Beijing Key Laboratory of Cardiopulmonary-Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing,China
| |
Collapse
|
147
|
Salmaninejad A, Khoramshahi V, Azani A, Soltaninejad E, Aslani S, Zamani MR, Zal M, Nesaei A, Hosseini SM. PD-1 and cancer: molecular mechanisms and polymorphisms. Immunogenetics 2017. [PMID: 28642997 DOI: 10.1007/s00251-017-1015-5] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The programmed cell death protein 1 (PD-1) is expressed by activated T cells that act as an immunoregulatory molecule, and are responsible for the negative regulation of T cell activation and peripheral tolerance. The PD-1 gene also encodes an inhibitory cell surface receptor involved in the regulation of T cell functions during immune responses/tolerance. Beyond potent inhibitory effects on T cells, PD-1 also has a role in regulating B cell and monocyte responses. An overexpression of PD-1 has been reported to contribute to immune system avoidance in different cancers. In particular, PD-1 over-expression influences tumor-specific T cell immunity in a cancer microenvironment. Blocking the PD-1/PD-1 ligand (PD-L1) pathway could potentially augment endogenous antitumor responses. Along these lines, the use of PD-1/PD-L1 inhibitors has been applied in clinical trials against diverse forms of cancer. It was believed that antibodies targeting PD-1/PD-L1 might synergize with other treatments that enhance endogenous antitumor immunity by blocking inhibitory receptor-ligand interactions. However, in all cases, the host genetic status (as well as that of the tumor) is likely to have an impact on the expected outcomes. Various investigations have evaluated the association between PD-1 polymorphisms and the risk of various types of cancer. Frequently studied PD-1 polymorphisms, PD-1.1 (rs36084323), PD-1.3 (rs11568821), PD-1.5 (rs2227981), PD-1.9 (rs2227982), and PD-1 rs7421861, and their associations in the risk of susceptibility to different types of cancer are mentioned in this review, as are studies highlighting the significance of conducting genetic association studies in different ethnic populations.
Collapse
Affiliation(s)
- Arash Salmaninejad
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Medical Genetics Research Center, Student Research Committee, Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Khoramshahi
- Department of Immunology, International Campus of Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alireza Azani
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ehsan Soltaninejad
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Aslani
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zamani
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Zal
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Nesaei
- Department of Basic Sciences, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Sayed Mostafa Hosseini
- Human Genetic Research Center, Baqiyatallah University of Medical Science, Tehran, Iran.
| |
Collapse
|
148
|
Läubli H, Hench J, Stanczak M, Heijnen I, Papachristofilou A, Frank S, Zippelius A, Stenner-Liewen F. Cerebral vasculitis mimicking intracranial metastatic progression of lung cancer during PD-1 blockade. J Immunother Cancer 2017; 5:46. [PMID: 28642817 PMCID: PMC5477093 DOI: 10.1186/s40425-017-0249-y,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Stimulation of the immune system by targeting the PD-1/PD-L1 pathway can result in activation of anti-tumor immunity. Besides its clinical benefit immune checkpoint therapy leads to significant immune-related adverse events (irAEs). Some rare irAEs are not well described yet but are critical in patient management. CASE PRESENTATION Here, we describe a case of autoimmune cerebral vasculitis/encephalitis after PD-1 inhibitor treatment for metastatic adenocarcinoma of the lung. Upon PD-1 blockade, the patient developed cerebral lesions, while having disease stabilization of extracranial metastases. Imaging suggested that the patient had new progressing brain metastases. Despite stereotactic irradiation the lesions progressed further. The largest lesion became symptomatic and had to be surgically resected. On examination, cerebral vasculitis was detected but not evidence of metastatic lung cancer. Analysis of the patient's serum revealed the presence of antinuclear antibodies that were already present before starting PD-1 blockade. In addition, we also found anti-vascular endothelial antibodies in the serum. CONCLUSION This finding suggests that the patient had preformed autoantibodies and the checkpoint inhibitor induced a clinically relevant autoimmune disease. Taken together, encephalitic lesions in patients under PD-1/PD-L1 blockade can mimic metastatic brain lesions and this rare irAE has to be considered as a differential diagnosis in patients treated with immunotherapy.
Collapse
Affiliation(s)
- Heinz Läubli
- grid.410567.1Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland ,grid.410567.1Department of Biomedicine, Cancer Immunology Laboratory, University Hospital Basel, Basel, Switzerland ,grid.410567.1Medical Oncology and Cancer Immunology, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Jürgen Hench
- grid.410567.1Department of Pathology, Division of Neuropathology, University Hospital Basel, Basel, Switzerland
| | - Michal Stanczak
- grid.410567.1Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland ,grid.410567.1Department of Biomedicine, Cancer Immunology Laboratory, University Hospital Basel, Basel, Switzerland
| | - Ingmar Heijnen
- grid.410567.1Department of Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Alexandros Papachristofilou
- grid.410567.1Department of Radiology, Division of Radiotherapy, University Hospital Basel, Basel, Switzerland
| | - Stephan Frank
- grid.410567.1Department of Pathology, Division of Neuropathology, University Hospital Basel, Basel, Switzerland
| | - Alfred Zippelius
- grid.410567.1Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland ,grid.410567.1Department of Biomedicine, Cancer Immunology Laboratory, University Hospital Basel, Basel, Switzerland
| | - Frank Stenner-Liewen
- grid.410567.1Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland ,grid.410567.1Department of Biomedicine, Cancer Immunology Laboratory, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
149
|
Läubli H, Hench J, Stanczak M, Heijnen I, Papachristofilou A, Frank S, Zippelius A, Stenner-Liewen F. Cerebral vasculitis mimicking intracranial metastatic progression of lung cancer during PD-1 blockade. J Immunother Cancer 2017. [PMID: 28642817 PMCID: PMC5477093 DOI: 10.1186/s40425-017-0249-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Stimulation of the immune system by targeting the PD-1/PD-L1 pathway can result in activation of anti-tumor immunity. Besides its clinical benefit immune checkpoint therapy leads to significant immune-related adverse events (irAEs). Some rare irAEs are not well described yet but are critical in patient management. Case presentation Here, we describe a case of autoimmune cerebral vasculitis/encephalitis after PD-1 inhibitor treatment for metastatic adenocarcinoma of the lung. Upon PD-1 blockade, the patient developed cerebral lesions, while having disease stabilization of extracranial metastases. Imaging suggested that the patient had new progressing brain metastases. Despite stereotactic irradiation the lesions progressed further. The largest lesion became symptomatic and had to be surgically resected. On examination, cerebral vasculitis was detected but not evidence of metastatic lung cancer. Analysis of the patient’s serum revealed the presence of antinuclear antibodies that were already present before starting PD-1 blockade. In addition, we also found anti-vascular endothelial antibodies in the serum. Conclusion This finding suggests that the patient had preformed autoantibodies and the checkpoint inhibitor induced a clinically relevant autoimmune disease. Taken together, encephalitic lesions in patients under PD-1/PD-L1 blockade can mimic metastatic brain lesions and this rare irAE has to be considered as a differential diagnosis in patients treated with immunotherapy.
Collapse
Affiliation(s)
- Heinz Läubli
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, Cancer Immunology Laboratory, University Hospital Basel, Basel, Switzerland.,Medical Oncology and Cancer Immunology, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Jürgen Hench
- Department of Pathology, Division of Neuropathology, University Hospital Basel, Basel, Switzerland
| | - Michal Stanczak
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, Cancer Immunology Laboratory, University Hospital Basel, Basel, Switzerland
| | - Ingmar Heijnen
- Department of Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | | | - Stephan Frank
- Department of Pathology, Division of Neuropathology, University Hospital Basel, Basel, Switzerland
| | - Alfred Zippelius
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, Cancer Immunology Laboratory, University Hospital Basel, Basel, Switzerland
| | - Frank Stenner-Liewen
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, Cancer Immunology Laboratory, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
150
|
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: 240] [Impact Index Per Article: 30.0] [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
|