1
|
Quan L, Dai J, Luo Y, Wang L, Liu Y, Meng J, Yang F, You X. The 100 top-cited studies in systemic lupus erythematosus: A bibliometric analysis. Hum Vaccin Immunother 2024; 20:2387461. [PMID: 39149877 PMCID: PMC11328883 DOI: 10.1080/21645515.2024.2387461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/12/2024] [Accepted: 07/30/2024] [Indexed: 08/17/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune inflammatory tissue disease. In view of the explosive growth in research on SLE, bibliometrics was performed to evaluate the 100 top-cited papers in this realm. We performed the search with terms "systemic lupus erythematosus" the Web of Science Core Collection database on May 3, 2023. Relevant literatures were screened. Data were extracted and analyzed by SPSS. The citations of 100 top-cited SLE studies spanned from 472 to 13,557. Most studies (60 out of 100) were conducted in the United States. Total citation times were positively associated with ACY, which was negatively correlated with the length of time since publication. Approximately half of the studies focused on the underlying mechanisms of SLE. New biologic therapies garnered attention and development. Our findings provide valuable insights into the developments in crucial areas of SLE and shed contributions to future studies.
Collapse
Affiliation(s)
- Liuliu Quan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiawen Dai
- Tianjin Institutes of Health Science, Tianjin, China
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yuan Luo
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Lin Wang
- School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Yue Liu
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Jiaqi Meng
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Fan Yang
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xin You
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China
| |
Collapse
|
2
|
Cho LK, Jamal S. De novo Connective Tissue Disorders as Immune-related Adverse Events. Rheum Dis Clin North Am 2024; 50:301-312. [PMID: 38670728 DOI: 10.1016/j.rdc.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment through blocking immunoregulatory pathways, resulting in augmented antitumor responses. However, ICIs can cause inflammatory autoimmune toxicities, known as immune-related adverse events (irAEs). Common rheumatic irAEs include inflammatory arthritis, polymyalgia rheumatica-like symptoms, and myositis. Fewer cases of de novo connective tissue disease as irAEs have been described and have mainly presented with cutaneous manifestations of systemic lupus erythematosus (SLE) and systemic sclerosis (SSc). Treatments include glucocorticoids and steroid-sparing agents such as hydroxychloroquine, mycophenolate mofetil, and methotrexate with improvement of symptoms. In this review, the authors discuss immune-related SLE and SSc and their management.
Collapse
Affiliation(s)
- Lindsay Kristin Cho
- Department of Medicine, Division of Rheumatology, Gordon & Leslie Diamond Health Care Centre, University of British Columbia, 2775 Laurel Street, Suite 8205B, Vancouver, British Columbia V5Z 1M9, Canada
| | - Shahin Jamal
- Department of Medicine, Division of Rheumatology, Gordon & Leslie Diamond Health Care Centre, University of British Columbia, 2775 Laurel Street, Suite 8205B, Vancouver, British Columbia V5Z 1M9, Canada.
| |
Collapse
|
3
|
Gao M, Shi J, Xiao X, Yao Y, Chen X, Wang B, Zhang J. PD-1 regulation in immune homeostasis and immunotherapy. Cancer Lett 2024; 588:216726. [PMID: 38401888 DOI: 10.1016/j.canlet.2024.216726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/31/2024] [Accepted: 02/10/2024] [Indexed: 02/26/2024]
Abstract
Harnessing the programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) axis is pivotal in autoimmunity and cancer immunotherapy. PD-1 receptors on immune cells engage with one of its ligands, PD-L1 or PD-L2, expressed on antigen-presenting cells or tumor cells, driving T-cell dysfunction and tumor immune escape. Thus, targeting PD-1/PD-L1 revitalizes cytotoxic T cells for cancer elimination. However, a majority of cancer patients don't respond to PD-1/PD-L1 blockade, and the underlying mechanisms remain partially understood. Recent studies have revealed that PD-1 expression levels or modifications impact the effectiveness of anti-PD-1/PD-L1 treatments. Therefore, understanding the molecular mechanisms governing PD-1 expression and modifications is crucial for innovating therapeutic strategies to enhance the efficacy of PD-1/PD-L1 inhibition. This article presents a comprehensive overview of advancements in PD-1 regulation and highlights their potential in modulating immune homeostasis and cancer immunotherapy, aiming to refine clinical outcomes.
Collapse
Affiliation(s)
- Minling Gao
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jie Shi
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiangling Xiao
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yingmeng Yao
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xu Chen
- Chongqing University Medical School, Chongqing, 400044, China
| | - Bin Wang
- Department of Gastroenterology & Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Jinfang Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
4
|
Yang M, Liu Y, Zheng S, Geng P, He T, Lu L, Feng Y, Jiang Q. Associations of PD-1 and PD-L1 gene polymorphisms with cancer risk: a meta-analysis based on 50 studies. Aging (Albany NY) 2024; 16:6068-6097. [PMID: 38546391 PMCID: PMC11042937 DOI: 10.18632/aging.205689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 02/27/2024] [Indexed: 04/23/2024]
Abstract
Programmed death-1 and its ligand-1 (PD-1/PD-L1), immune checkpoints proteins, play a crucial role in anti-tumor responses. A large number of studies have evaluated the relationships of PD-1/PD-L1 polymorphisms with risk of cancer, but evidence for the associations remains inconsistent. Therefore, we performed a meta-analysis to examine the associations between PD-1/PD-L1 single nucleotide polymorphisms (SNPs) and cancer predisposition. Results showed that PD-1.3 and PD-L1 rs17718883 were significantly correlated with overall cancer risk. PD-1.5 was prominently linked with cervical cancer (CC), non-small cell lung cancer (NSCLC), TC (thyroid cancer), brain tumor, AML (acute myelocytic leukemia) and UCC (urothelial cell carcinoma) risk, PD-1.9 with breast cancer (BC), AML, esophageal cancer (EC) and ovarian cancer (OC) risk, and PD-1.3 with colorectal cancer (CRC) and BCC (basal cell carcinoma) risk. PD-1.1 polymorphism slightly elevated BC and OC susceptibility, whereas the rs4143815 variant notably decreased the risk of gastric cancer (GC), hepatocellular carcinoma (HCC) and OC, but increased the risk of BC. PD-1.6 was closely linked with AML risk, PD-L1 rs2890658 with NSCLC, HCC and BC risk, rs17718883 with HCC and GC risk, rs10815225 with GC risk, and rs2297136 with NSCLC and HCC risk. Interestingly, the rs7421861, rs10815225, and rs10815225 markedly reduced cancer susceptibility among Asians. The rs7421861 polymrophism decreased cancer risk among Caucasians, rather than the rs10815225 elevated cancer risk. Our results supported that PD-1 and PD-L1 SNPs were dramatically correlated with cancer risk.
Collapse
Affiliation(s)
- Maoquan Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261042, Shandong, China
| | - Yan Liu
- Department of Gastroenterology, Weifang People’s Hospital, The First Affiliated Hospital of Shandong Second Medical University, Kuiwen, Weifang 261000, Shandong, China
| | - Shuangshuang Zheng
- Department of Health, Weifang People’s Hospital, The First Affiliated Hospital of Shandong Second Medical University, Kuiwen, Weifang 261000, Shandong, China
| | - Peizhen Geng
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261042, Shandong, China
| | - Tianhao He
- Department of Gastroenterology, Weifang People’s Hospital, The First Affiliated Hospital of Shandong Second Medical University, Kuiwen, Weifang 261000, Shandong, China
| | - Linan Lu
- Department of Gastroenterology, Weifang People’s Hospital, The First Affiliated Hospital of Shandong Second Medical University, Kuiwen, Weifang 261000, Shandong, China
| | - Yikuan Feng
- Department of Gastroenterology, Weifang People’s Hospital, The First Affiliated Hospital of Shandong Second Medical University, Kuiwen, Weifang 261000, Shandong, China
| | - Qiqi Jiang
- Department of Gastroenterology, Weifang People’s Hospital, The First Affiliated Hospital of Shandong Second Medical University, Kuiwen, Weifang 261000, Shandong, China
| |
Collapse
|
5
|
Garcia JPT, Tayo LL. Theoretical Studies of DNA Microarray Present Potential Molecular and Cellular Interconnectivity of Signaling Pathways in Immune System Dysregulation. Genes (Basel) 2024; 15:393. [PMID: 38674328 PMCID: PMC11049615 DOI: 10.3390/genes15040393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/03/2024] [Accepted: 03/07/2024] [Indexed: 04/28/2024] Open
Abstract
Autoimmunity is defined as the inability to regulate immunological activities in the body, especially in response to external triggers, leading to the attack of the tissues and organs of the host. Outcomes include the onset of autoimmune diseases whose effects are primarily due to dysregulated immune responses. In past years, there have been cases that show an increased susceptibility to other autoimmune disorders in patients who are already experiencing the same type of disease. Research in this field has started analyzing the potential molecular and cellular causes of this interconnectedness, bearing in mind the possibility of advancing drugs and therapies for the treatment of autoimmunity. With that, this study aimed to determine the correlation of four autoimmune diseases, which are type 1 diabetes (T1D), psoriasis (PSR), systemic sclerosis (SSc), and systemic lupus erythematosus (SLE), by identifying highly preserved co-expressed genes among datasets using WGCNA. Functional annotation was then employed to characterize these sets of genes based on their systemic relationship as a whole to elucidate the biological processes, cellular components, and molecular functions of the pathways they are involved in. Lastly, drug repurposing analysis was performed to screen candidate drugs for repositioning that could regulate the abnormal expression of genes among the diseases. A total of thirteen modules were obtained from the analysis, the majority of which were associated with transcriptional, post-transcriptional, and post-translational modification processes. Also, the evaluation based on KEGG suggested the possible role of TH17 differentiation in the simultaneous onset of the four diseases. Furthermore, clomiphene was the top drug candidate for regulating overexpressed hub genes; meanwhile, prilocaine was the top drug for regulating under-expressed hub genes. This study was geared towards utilizing transcriptomics approaches for the assessment of microarray data, which is different from the use of traditional genomic analyses. Such a research design for investigating correlations among autoimmune diseases may be the first of its kind.
Collapse
Affiliation(s)
- Jon Patrick T. Garcia
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila 1002, Philippines;
- School of Graduate Studies, Mapúa University, Manila 1002, Philippines
| | - Lemmuel L. Tayo
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila 1002, Philippines;
- Department of Biology, School of Medicine and Health Sciences, Mapúa University, Makati 1200, Philippines
| |
Collapse
|
6
|
Ghoneim ME, Sheashaa H, Wafa E, Awadalla A, Ahmed AE, Sobh M, Shokeir AA. Impact of CD 28, CD86, CTLA-4 and PD-1 genes polymorphisms on acute renal allograft rejection and graft survival among Egyptian recipients. Sci Rep 2024; 14:2047. [PMID: 38267522 PMCID: PMC10808544 DOI: 10.1038/s41598-024-52195-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 01/15/2024] [Indexed: 01/26/2024] Open
Abstract
To study the impact of four gene polymorphisms on acute renal allograft rejection (AR) and graft survival among Egyptian population. These 4 gene polymorphisms include: (1) CD 28 (rs3116496), (2) CD86 (rs1129055), (3) CTLA-4 (rs3087243), (4) PD-1 (rs2227982). This is a non-concurrent cohort study including 50 kidney transplant recipients diagnosed histopathologically as (AR) [study group] and another 50 matched allograft recipients without AR [control group]. Blood samples were taken from both groups and subjected to genotyping for the selected four genetic polymorphisms by TaqMan genotyping assay. The difference in genotypic distribution of CD 28: rs3116496 and CD86: rs1129055 wasn't statistically significant between the study and control groups (P = 0.22 and 0.33 respectively) and also both polymorphisms had no effect on graft survival (P = 0.36 and 0.74 respectively) while the addition of C allele to IVS3 +17T/C polymorphism in CD28 gene showed a protective effect against AR (P = 0.03). CTLA-4: rs3087243 AG genotype showed a protective effect against AR as it was more frequent in no rejection group compared to those with AR (P = 0.001) with a statistically significant impact on graft survival (P < 0.001), while PD-1: rs2227982 AG genotype was equally distributed between both groups (variant of unknown significance). There was no detected association between CD86 polymorphism: rs1129055 and CD 28 polymorphism: rs3116496 with the development of AR. However, C allele of CD 28 IVS3 +17T/C polymorphism and CTLA-4 polymorphism: rs3087243AG genotype both demonstrated a protective effect against AR.
Collapse
Affiliation(s)
- Moatasem Elsayed Ghoneim
- Nephrology and Transplant Unit, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt.
| | - Hussein Sheashaa
- Nephrology and Transplant Unit, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Ehab Wafa
- Nephrology and Transplant Unit, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Amira Awadalla
- Center of Excellence for Genome and Cancer Research, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Asmaa E Ahmed
- Genetics Research Unit, Faculty of Medicine, Delta University for Science and Technology, Gamasa, Egypt
| | - Mohamed Sobh
- Nephrology and Transplant Unit, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Ahmed Abdulrahman Shokeir
- Department of Urology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
- Center of Excellence for Genome and Cancer Research, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| |
Collapse
|
7
|
May JF, Kelly RG, Suen AYW, Kim J, Kim J, Anderson CC, Rayat GR, Baldwin TA. Establishment of CD8+ T Cell Thymic Central Tolerance to Tissue-Restricted Antigen Requires PD-1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:271-283. [PMID: 37982696 DOI: 10.4049/jimmunol.2200775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 11/02/2023] [Indexed: 11/21/2023]
Abstract
Highly self-reactive T cells are censored from the repertoire by both central and peripheral tolerance mechanisms upon receipt of high-affinity TCR signals. Clonal deletion is considered a major driver of central tolerance; however, other mechanisms such as induction of regulatory T cells and functional impairment have been described. An understanding of the interplay between these different central tolerance mechanisms is still lacking. We previously showed that impaired clonal deletion to a model tissue-restricted Ag did not compromise tolerance. In this study, we determined that murine T cells that failed clonal deletion were rendered functionally impaired in the thymus. Programmed cell death protein 1 (PD-1) was induced in the thymus and was required to establish cell-intrinsic tolerance to tissue-restricted Ag in CD8+ thymocytes independently of clonal deletion. In bone marrow chimeras, tolerance was not observed in PD-L1-deficient recipients, but tolerance was largely maintained following adoptive transfer of tolerant thymocytes or T cells to PD-L1-deficient recipients. However, CRISPR-mediated ablation of PD-1 in tolerant T cells resulted in broken tolerance, suggesting different PD-1 signaling requirements for establishing versus maintaining tolerance. Finally, we showed that chronic exposure to high-affinity Ag supported the long-term maintenance of tolerance. Taken together, our study identifies a critical role for PD-1 in establishing central tolerance in autoreactive T cells that escape clonal deletion. It also sheds light on potential mechanisms of action of anti-PD-1 pathway immune checkpoint blockade and the development of immune-related adverse events.
Collapse
Affiliation(s)
- Julia F May
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Rees G Kelly
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Alexander Y W Suen
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Jeongbee Kim
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Jeongwoo Kim
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Colin C Anderson
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Gina R Rayat
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
- Ray Rajotte Surgical-Medical Research Institute, AB Diabetes and Transplant Institutes, University of Alberta, Edmonton, Alberta, Canada
| | - Troy A Baldwin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
8
|
Altan M, Li QZ, Wang Q, Vokes NI, Sheshadri A, Gao J, Zhu C, Tran HT, Gandhi S, Antonoff MB, Swisher S, Wang J, Byers LA, Abdel-Wahab N, Franco-Vega MC, Wang Y, Lee JJ, Zhang J, Heymach JV. Distinct patterns of auto-reactive antibodies associated with organ-specific immune-related adverse events. Front Immunol 2023; 14:1322818. [PMID: 38152395 PMCID: PMC10751952 DOI: 10.3389/fimmu.2023.1322818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/29/2023] [Indexed: 12/29/2023] Open
Abstract
The roles of preexisting auto-reactive antibodies in immune-related adverse events (irAEs) associated with immune checkpoint inhibitor therapy are not well defined. Here, we analyzed plasma samples longitudinally collected at predefined time points and at the time of irAEs from 58 patients with immunotherapy naïve metastatic non-small cell lung cancer treated on clinical protocol with ipilimumab and nivolumab. We used a proteomic microarray system capable of assaying antibody reactivity for IgG and IgM fractions against 120 antigens for systemically evaluating the correlations between auto-reactive antibodies and certain organ-specific irAEs. We found that distinct patterns of auto-reactive antibodies at baseline were associated with the subsequent development of organ-specific irAEs. Notably, ACHRG IgM was associated with pneumonitis, anti-cytokeratin 19 IgM with dermatitis, and anti-thyroglobulin IgG with hepatitis. These antibodies merit further investigation as potential biomarkers for identifying high-risk populations for irAEs and/or monitoring irAEs during immunotherapy treatment. Trial registration ClinicalTrials.gov identifier: NCT03391869.
Collapse
Affiliation(s)
- Mehmet Altan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Quan-Zhen Li
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Qi Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Natalie I. Vokes
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jianjun Gao
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Chengsong Zhu
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Hai T. Tran
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Saumil Gandhi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mara B. Antonoff
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Stephen Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jing Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lauren A. Byers
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Noha Abdel-Wahab
- Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria C. Franco-Vega
- Department of Hospital Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yinghong Wang
- Department of Gastroenterology Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - J. Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John V. Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
9
|
Zhou S, Zhao T, Chen X, Zhang W, Zou X, Yang Y, Wang Q, Zhang P, Zhou T, Feng T. Runx1 Deficiency Promotes M2 Macrophage Polarization Through Enhancing STAT6 Phosphorylation. Inflammation 2023; 46:2241-2253. [PMID: 37530929 DOI: 10.1007/s10753-023-01874-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 08/03/2023]
Abstract
Our previous study had demonstrated that Runx1 promoted LPS-induced macrophage inflammatory response, however, the role of Runx1 in M2 macrophage polarization still remains largely unknown. This study was conducted to investigate the role of Runx1 in IL-4/IL-13-induced M2 macrophage polarization and its potential regulatory mechanism. We found that exposure of macrophages to IL-4/IL-13 induced a remarkable increasement in Runx1 expression level. Specifically, we established genetically modified mice lacking Runx1 in myeloid cells, including macrophages. RNA-Seq was performed to identify differentially expressed genes (DEGs) between Runx1 knockout and WT control bone marrow-derived macrophages (BMDMs). We identified 686 DEGs, including many genes which were highly expressed in M2 macrophage. In addition, bioinformatics analysis indicated that these DEGs were significantly enriched in extracellular matrix-related processes. Moreover, RT-qPCR analysis showed that there was an obvious upregulation in the relative expression levels of M2 marker genes, including Arg1, Ym1, Fizz1, CD71, Mmp9, and Tgm2, in Runx1 knockout macrophages, as compared to WT controls. Consistently, similar results were obtained in the protein and enzymatic activity levels of Arg1. Finally, we found that the STAT6 phosphorylation level was significantly enhanced in Runx1 knockout macrophages, and the STAT6 inhibitor AS1517499 partly reduced the upregulated effect of Runx1 deficiency on the M2 macrophage polarization. Taken together, Runx1 deficiency facilitates IL-4/IL-13-induced M2 macrophage polarization through enhancing STAT6 phosphorylation.
Collapse
Affiliation(s)
- Siyuan Zhou
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Ting Zhao
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Xuqiong Chen
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Wuwen Zhang
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Xiaoyi Zou
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Yi Yang
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Qinshi Wang
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Ping Zhang
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Tong Zhou
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Tongbao Feng
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China.
| |
Collapse
|
10
|
Del Bello A, Treiner E. Immune Checkpoints in Solid Organ Transplantation. BIOLOGY 2023; 12:1358. [PMID: 37887068 PMCID: PMC10604300 DOI: 10.3390/biology12101358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
Allogenic graft acceptance is only achieved by life-long immunosuppression, which comes at the cost of significant toxicity. Clinicians face the challenge of adapting the patients' treatments over long periods to lower the risks associated with these toxicities, permanently leveraging the risk of excessive versus insufficient immunosuppression. A major goal and challenge in the field of solid organ transplantation (SOT) is to attain a state of stable immune tolerance specifically towards the grafted organ. The immune system is equipped with a set of inhibitory co-receptors known as immune checkpoints (ICs), which physiologically regulate numerous effector functions. Insufficient regulation through these ICs can lead to autoimmunity and/or immune-mediated toxicity, while excessive expression of ICs induces stable hypo-responsiveness, especially in T cells, a state sometimes referred to as exhaustion. IC blockade has emerged in the last decade as a powerful therapeutic tool against cancer. The opposite action, i.e., subverting IC for the benefit of establishing a state of specific hypo-responsiveness against auto- or allo-antigens, is still in its infancy. In this review, we will summarize the available literature on the role of ICs in SOT and the relevance of ICs with graft acceptance. We will also discuss the possible influence of current immunosuppressive medications on IC functions.
Collapse
Affiliation(s)
- Arnaud Del Bello
- Department of Nephrology, University Hospital of Toulouse, 31400 Toulouse, France
- Metabolic and Cardiovascular Research Institute (I2MC), Inserm UMR1297, CEDEX 4, 31432 Toulouse, France
- Faculty of Medicine, University Toulouse III Paul Sabatier, 31062 Toulouse, France
| | - Emmanuel Treiner
- Faculty of Medicine, University Toulouse III Paul Sabatier, 31062 Toulouse, France
- Laboratory of Immunology, University Hospital of Toulouse, 31300 Toulouse, France
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Inserm UMR1291, 31024 Toulouse, France
| |
Collapse
|
11
|
Ghorani E, Swanton C, Quezada SA. Cancer cell-intrinsic mechanisms driving acquired immune tolerance. Immunity 2023; 56:2270-2295. [PMID: 37820584 DOI: 10.1016/j.immuni.2023.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023]
Abstract
Immune evasion is a hallmark of cancer, enabling tumors to survive contact with the host immune system and evade the cycle of immune recognition and destruction. Here, we review the current understanding of the cancer cell-intrinsic factors driving immune evasion. We focus on T cells as key effectors of anti-cancer immunity and argue that cancer cells evade immune destruction by gaining control over pathways that usually serve to maintain physiological tolerance to self. Using this framework, we place recent mechanistic advances in the understanding of cancer immune evasion into broad categories of control over T cell localization, antigen recognition, and acquisition of optimal effector function. We discuss the redundancy in the pathways involved and identify knowledge gaps that must be overcome to better target immune evasion, including the need for better, routinely available tools that incorporate the growing understanding of evasion mechanisms to stratify patients for therapy and trials.
Collapse
Affiliation(s)
- Ehsan Ghorani
- Cancer Immunology and Immunotherapy Unit, Department of Surgery and Cancer, Imperial College London, London, UK; Department of Medical Oncology, Imperial College London Hospitals, London, UK.
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Department of Oncology, University College London Hospitals, London, UK
| | - Sergio A Quezada
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Cancer Immunology Unit, Research Department of Hematology, University College London Cancer Institute, London, UK.
| |
Collapse
|
12
|
Boguszewska-Byczkiewicz K, Wow T, Szymańska B, Kosny M, Kolacinska-Wow A. The PD-1 single-nucleotide polymorphism rs11568821 and rs2227981 as a novel prognosis model in a triple-negative breast cancer patient. Mol Biol Rep 2023; 50:6279-6285. [PMID: 37212960 DOI: 10.1007/s11033-023-08423-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 04/04/2023] [Indexed: 05/23/2023]
Abstract
INTRODUCTION The aim of the study is to determine the relationship between polymorphisms rs11568821 C/T and at rs2227981 G/A in the programmed cell death 1 gene (PDCD1) and the clinicopathologic characteristics of triple negative breast cancer patient (TNBC). MATERIAL AND METHODS The study included 30 TNBC patients and 30 healthy controls. Genotyping was performed with allelic discrimination using PCR with TaqMan SNP Genotyping Assays. RESULTS The presence of CC/CT in rs11568821and GG/AG in rs2227981 were not associated with the risk of progression of TNBC. The correlation between rs11568821 minor allele distribution and risk of TNBC has borderline significance (p = 0.0619). The rs2227981 polymorphism has a significant association with grade G (G3, p = 0.0229). There was a trend toward significance (p = 0.063448) in the minor allele presentation and Ki67 > 20% for rs2227981. Other clinical features (e.g. age, TNM stage) did not significantly correlate with the rs11568821 or the rs2227981 polymorphism. CONCLUSION rs2227981 is associated with grading; hence PDCD1 can be used as a prognostic marker in TNBC.
Collapse
Affiliation(s)
- Katarzyna Boguszewska-Byczkiewicz
- Department of Surgical Oncology, Copernicus Provincial Multidisciplinary Centre of Oncology and Traumatology, Paderewskiego 4, 93-513, Lodz, Poland.
| | - Thomas Wow
- Department of General Surgery and Surgical Oncology, Faculty of Medicine and Health Sciences, University of Zielona Gora, Zyty 26, 65-046, Zielona Gora, Poland
| | - Bożena Szymańska
- Research Labolatory CoreLab, Medical University of Lodz, Mazowiecka6/8 St., 92-215, Lodz, Poland
| | - Michał Kosny
- Copernicus Provincial Multidisciplinary Centre of Oncology and Traumatology, Paderewskiego 4, 93-513, Lodz, Poland
| | - Agnieszka Kolacinska-Wow
- Department of Oncological Physiotherapy, Medical University of Lodz, Paderewskiego 4, 93-509, Lodz, Poland
| |
Collapse
|
13
|
Zhu Y, Yu Q, Su G, Shao N, Feng J, Xiang L, Zhou C, Yang P. Interferon-α2a induces CD4+ T cell apoptosis and suppresses Th1/Th17 responses via upregulating IRF1-mediated PDL1 expression in dendritic cells from Behcet's uveitis. Clin Immunol 2023; 250:109303. [PMID: 36997038 DOI: 10.1016/j.clim.2023.109303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
Recombinant interferon-α2a (IFNα2a) has been widely used in the treatment of Behcet's uveitis (BU). However, the mechanism underlying its effects remains poorly understood. In this study, we investigated its effect on dendritic cells (DCs) and CD4+ T cells, which are essential for the development of BU. Our results showed that the expression of PDL1 and IRF1 was significantly decreased in DCs from active BU patients, and IFNα2a could significantly upregulate PDL1 expression in an IRF1-dependent manner. IFNα2a-treated DCs induced CD4+ T cells apoptosis and inhibited the Th1/Th17 immune response in association with reduced secretion of IFN-γ and IL-17. We also found that IFNα2a promoted Th1 cell differentiation and IL-10 secretion by CD4+ T cells. Finally, a comparison of patients before and after IFNα2a therapy revealed that the frequencies of Th1/Th17 cells significantly decreased in association with remission of uveitis after IFNα2a therapy. Collectively, these results show that IFNα2a could exert its effects by modulating the function of DCs and CD4+ T cells in BU.
Collapse
|
14
|
Cruz D, Rodríguez-Romanos R, González-Bartulos M, García-Cadenas I, de la Cámara R, Heras I, Buño I, Santos N, Lloveras N, Velarde P, Tuset E, Martínez C, González M, Sanz GF, Ferrá C, Sampol A, Coll R, Pérez-Simón JA, López-Jiménez J, Jurado M, Gallardo D. LAG3 genotype of the donor and clinical outcome after allogeneic transplantation from HLA-identical sibling donors. Front Immunol 2023; 14:1066393. [PMID: 36742309 PMCID: PMC9897054 DOI: 10.3389/fimmu.2023.1066393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023] Open
Abstract
Introduction The association of polymorphisms in molecules involved in the immune response (checkpoint inhibitors) with the clinical outcome after allogeneic transplantation (alloHSCT) has been described. Lymphocyte Activation 3 (LAG3) is a surface protein that plays a regulatory role in immunity as an inhibitory immune checkpoint molecule. Methods To determine its role in the alloHSCT setting, we analyzed 797 patients transplanted from HLA-identical sibling donors. The LAG3 rs870849 C>T polymorphism was genotyped in donors. Results We detected a higher incidence of severe acute GVHD in patients transplanted from donors with TT genotype (p: 0.047, HR 1.64; 95% CI 1.01 - 2.67). Overall survival (OS) was worse for patients transplanted from donors with the rs870849 CT/TT genotype (0.020; HR, 1.44; 95% CI 1.06 - 1.96), as well as disease-free survival (DFS) (p: 0.002; HR 1.58, 95%CI: 1.18 - 2.14) and transplant-related mortality (TRM) (p< 0.001; HR: 1.88, 95% CI 1.29 - 2.74). When combining the LAG3 rs870849 and the PDCD1 rs36084323 genotypes of the donor, three genetic groups were well defined, allowing a good stratification of the risk of acute GVHD, TRM, OS and DFS. Discussion We conclude that the LAG3 genotype of the donor may be considered in donors' selection. As this selection may be limited in the HLA-identical sibling donor scenario, further studies exploring the impact of LAG3 genotype of the donor in unrelated transplantation are warranted.
Collapse
Affiliation(s)
- David Cruz
- Hematology Department, Institut Català d’Oncologia - Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Girona, Spain
| | - Rocío Rodríguez-Romanos
- Hematology Department, Institut Català d’Oncologia - Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Girona, Spain
| | - Marta González-Bartulos
- Hematology Department, Institut Català d’Oncologia - Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Girona, Spain
| | - Irene García-Cadenas
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Institut d’Investigació Biomèdica Sant Pau, Universitat Autónoma de Barcelona, Barcelona, Spain
| | | | - Inmaculada Heras
- Hematology Department, Hospital General Universitario Morales Meseguer, Murcia, Spain
| | - Ismael Buño
- Hematology Department and Genomics Unit, Hospital General Universitario Gregorio Marañón, Gregorio Marañón Health Research Institute (IiSGM), Complutense University, Madrid, Spain
| | - Nazly Santos
- Hematology Department, Institut Català d’Oncologia - Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Girona, Spain
| | - Natàlia Lloveras
- Hematology Department, Institut Català d’Oncologia - Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Girona, Spain
- Department of Medicine, Universitat de Girona, Girona, Spain
| | - Pilar Velarde
- Hematology Department, Institut Català d’Oncologia - Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Girona, Spain
| | - Esperanza Tuset
- Hematology Department, Institut Català d’Oncologia - Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Girona, Spain
| | - Carmen Martínez
- Hematology Department, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marcos González
- Hematology Department, Hospital Clínico Universitario, Salamanca, Spain
| | - Guillermo F. Sanz
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Christelle Ferrá
- Hematology Department, Institut Català d’Oncologia – Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Badalona, Spain
| | - Antonia Sampol
- Hematology Department, Hospital Universitari Son Espases, IdISBa, Palma de Mallorca, Spain
| | - Rosa Coll
- Hematology Department, Institut Català d’Oncologia - Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Girona, Spain
- Department of Medicine, Universitat de Girona, Girona, Spain
| | - Jose A. Pérez-Simón
- Hematology Department, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Universidad de Sevilla, Sevilla, Spain
| | | | - Manuel Jurado
- Hematology Department, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - David Gallardo
- Hematology Department, Institut Català d’Oncologia - Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Girona, Spain
- Department of Medicine, Universitat de Girona, Girona, Spain
| |
Collapse
|
15
|
Wang B, Chen C, Liu X, Zhou S, Xu T, Wu M. The effect of combining PD-1 agonist and low-dose Interleukin-2 on treating systemic lupus erythematosus. Front Immunol 2023; 14:1111005. [PMID: 36969198 PMCID: PMC10030866 DOI: 10.3389/fimmu.2023.1111005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease involving multiple organs. It is often called "immortal cancer" due to the difficulties in disease treatment. As the cornerstone of immune regulation, the programmed cell death protein 1 (PD-1) has been extensively studied in the context of chronic inflammation due to its ability of regulating immune response and immunosuppression. Recently, more and more studies on rheumatic immune related complications have also focused on PD-1 and proposed that the use of PD-1 agonist could inhibit the activation of lymphocytes and alleviate SLE disease activity. In this review, we summarized the role of PD-1 in SLE, implicating its potential application as a biomarker to predict SLE disease activity; we also proposed that the combination of PD-1 agonist and low-dose IL-2 may have better therapeutic efficacy, shining light on a new direction for developing specific treatment approaches.
Collapse
Affiliation(s)
- Bing Wang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Can Chen
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xia Liu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Shuang Zhou
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Ting Xu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- *Correspondence: Ting Xu, ; Min Wu,
| | - Min Wu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- *Correspondence: Ting Xu, ; Min Wu,
| |
Collapse
|
16
|
Hassani N, Salmaninejad A, Aslani S, Kamali-sarvestani E, Vessal M. The association between PD-1 gene polymorphisms and susceptibility to multiple sclerosis. Immunol Med 2022; 46:69-76. [PMID: 36308011 DOI: 10.1080/25785826.2022.2137967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Programmed cell death 1 (PD-1) is an immune checkpoint and has been reported to be associated with several autoimmune diseases. We aimed to investigate the association between human PD-1 gene (PDCD1) polymorphisms and multiple sclerosis (MS). This case-control study was conducted on 229 MS patients and 246 healthy controls. Genotyping of rs36084323 (PD-1.1 G/A), rs11568821 (PD-1.3 G/A) and rs2227981 (PD-1.5 C/T) polymorphisms was performed by PCR-RFLP technique. The frequency difference of PD-1.1 genotypes and alleles (-536 G/A) between patients and healthy controls was not significant. Regarding PD-1.3, the AA + AG genotype was found to be relatively higher in the control group. Concerning PD-1.5 (+7785 C/T), the frequency of T allele carriers (TT + CT) was relatively higher in MS patients, which was marginally insignificant (p = .07). PD-1 gene polymorphisms may be associated with MS; however, accurate conclusions require further studies with a larger number of samples.
Collapse
Affiliation(s)
- Nasrin Hassani
- Department of Molecular Biology, Faculty of Medicine, Islamic Azad University, Shiraz, Iran
| | - Arash Salmaninejad
- Regenerative Medicine, Organ Procurement and Transplantation Multi Disciplinary Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Aslani
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Eskandar Kamali-sarvestani
- Department of Immunology and Autoimmune Diseases Research Center, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahmood Vessal
- Department of Molecular Biology, Faculty of Medicine, Islamic Azad University, Shiraz, Iran
| |
Collapse
|
17
|
The PD-1/PD-L1 Pathway: A Perspective on Comparative Immuno-Oncology. Animals (Basel) 2022; 12:ani12192661. [PMID: 36230402 PMCID: PMC9558501 DOI: 10.3390/ani12192661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/25/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022] Open
Abstract
Simple Summary The programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) pathway inhibits the function of activated immune cells. This mediates immune tolerance and prevents immune-mediated tissue destruction. The malfunction of this pathway is involved in the pathogenesis of chronic infections, autoimmunity, and cancer. The PD-1/PD-L1 pathway is an excellent example of the research benefits of comparative pathology and attests to the importance of the “one health one medicine” concept. Pioneering research was mainly focused on the examination of cells and tissues of human and mouse origin. It mainly revealed that PD-L1-positive tumor cells can paralyze PD-1-bearing immune cells, which prevents immunological destruction of cancer cells. This led to a major breakthrough in cancer treatment, i.e., the use of antibodies that block the interaction of these molecules and restore anti-cancer immune defense (immune checkpoint therapy). Further studies provided more detailed information on the tissue-specific context and fine-tuning of this pathway. The most recent research has extended the investigations to the examination of several animal species with the aim of improving disease diagnostics and treatment for certain animal diseases, in particular cancer, which is a major cause of disease and death in companion animals. Abstract The programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) pathway mainly attracted attention in immuno-oncology, leading to the development of immune checkpoint therapy. It has, however, much broader importance for tissue physiology and pathology. It mediates basic processes of immune tolerance and tissue homeostasis. In addition, it is involved in the pathogenesis of chronic infectious diseases, autoimmunity, and cancer. It is also an important paradigm for comparative pathology as well as the “one health one medicine” concept. The aim of this review is to provide an overview of novel research into the diverse facets of the PD-1/PD-L1 pathway and to give insights into its fine-tuning homeostatic role in a tissue-specific context. This review details early translational research from the discovery phase based on mice as animal models for understanding pathophysiological aspects in human tissues to more recent research extending the investigations to several animal species. The latter has the twofold goal of comparing this pathway between humans and different animal species and translating diagnostic tools and treatment options established for the use in human beings to animals and vice versa.
Collapse
|
18
|
Sprangers B, Leaf DE, Porta C, Soler MJ, Perazella MA. Diagnosis and management of immune checkpoint inhibitor-associated acute kidney injury. Nat Rev Nephrol 2022; 18:794-805. [PMID: 36168055 DOI: 10.1038/s41581-022-00630-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 11/10/2022]
Abstract
Since their introduction into clinical practice a decade ago, immune checkpoint inhibitors (ICIs) have had an overwhelming impact on cancer treatment. Use of these agents in oncology continues to grow; however, the increased use of these agents has been associated with a parallel increase in ICI-associated immune-related adverse events, which can affect virtually any organ, including the kidneys. ICI-associated acute kidney injury (ICI-AKI) occurs in 2-5% of patients treated with ICIs. Its occurrence can have important consequences, including the temporary or permanent discontinuation of ICIs or other concomitant anticancer therapies and the need for prolonged treatment with corticosteroids. Various mechanisms have been proposed to underlie the development of ICI-AKI, including loss of tolerance to self-antigens, reactivation of drug-specific effector T cells, and the production of kidney-specific autoantibodies. ICI-AKI most commonly manifests as acute tubulo-interstitial nephritis on kidney biopsy and generally shows a favourable response to early initiation of corticosteroids, with complete or partial remission achieved in most patients. The evaluation of patients with suspected ICI-AKI requires careful diagnostic work-up and kidney biopsy for patients with moderate-to-severe ICI-AKI to ensure accurate diagnosis and inform appropriate treatment.
Collapse
Affiliation(s)
- Ben Sprangers
- Division of Nephrology, Ziekenhuis Oost-Limburg, Genk, Belgium. .,Biomedical Research Institute, Department of Immunology and Infection, UHasselt, Diepenbeek, Belgium.
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Camillo Porta
- Division of Medical Oncology, Azienda Ospedaliero-Universitaria Corsorziale Policlinico di Bari, Bari, Italy.,Oncology, Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Maria José Soler
- Nephrology Research Group, Vall d'hebrón Institut de Recerca (VHIR), Barcelona, Spain.,Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Mark A Perazella
- Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut, USA.,Veterans Affairs Medical Center, West Haven, Connecticut, USA
| |
Collapse
|
19
|
Prokunina-Olsson L. A Conversation with Dr. Ludmila Prokunina-Olsson. J Interferon Cytokine Res 2022; 42:462-466. [PMID: 36121375 PMCID: PMC9641989 DOI: 10.1089/jir.2022.29043.lup] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ludmila Prokunina-Olsson
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, USA
| |
Collapse
|
20
|
Zhou ZR, Chen X, Lv J, Li DW, Yang CD, Liu HL, Qian RC. A plasmonic nanoparticle-embedded polydopamine substrate for fluorescence detection of extracellular vesicle biomarkers in serum and urine from patients with systemic lupus erythematosus. Talanta 2022; 247:123620. [DOI: 10.1016/j.talanta.2022.123620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/16/2022] [Accepted: 05/25/2022] [Indexed: 12/21/2022]
|
21
|
Chye A, Allen I, Barnet M, Burnett DL. Insights Into the Host Contribution of Endocrine Associated Immune-Related Adverse Events to Immune Checkpoint Inhibition Therapy. Front Oncol 2022; 12:894015. [PMID: 35912205 PMCID: PMC9329613 DOI: 10.3389/fonc.2022.894015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
Blockade of immune checkpoints transformed the paradigm of systemic cancer therapy, enabling substitution of a cytotoxic chemotherapy backbone to one of immunostimulation in many settings. Invigorating host immune cells against tumor neo-antigens, however, can induce severe autoimmune toxicity which in many cases requires ongoing management. Many immune-related adverse events (irAEs) are clinically and pathologically indistinguishable from inborn errors of immunity arising from genetic polymorphisms of immune checkpoint genes, suggesting a possible shared driver for both conditions. Many endocrine irAEs, for example, have analogous primary genetic conditions with varied penetrance and severity despite consistent genetic change. This is akin to onset of irAEs in response to immune checkpoint inhibitors (ICIs), which vary in timing, severity and nature despite a consistent drug target. Host contribution to ICI response and irAEs, particularly those of endocrine origin, such as thyroiditis, hypophysitis, adrenalitis and diabetes mellitus, remains poorly defined. Improved understanding of host factors contributing to ICI outcomes is essential for tailoring care to an individual’s unique genetic predisposition to response and toxicity, and are discussed in detail in this review.
Collapse
Affiliation(s)
- Adrian Chye
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, Darlinghurst, NSW, Australia
| | - India Allen
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
| | - Megan Barnet
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, Darlinghurst, NSW, Australia
- *Correspondence: Megan Barnet, ; Deborah L. Burnett,
| | - Deborah L. Burnett
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
- *Correspondence: Megan Barnet, ; Deborah L. Burnett,
| |
Collapse
|
22
|
Kobayashi M, Numakura K, Hatakeyama S, Muto Y, Sekine Y, Sasagawa H, Kashima S, Yamamoto R, Koizumi A, Nara T, Saito M, Narita S, Ohyama C, Habuchi T. Severe Immune-Related Adverse Events in Patients Treated with Nivolumab for Metastatic Renal Cell Carcinoma Are Associated with PDCD1 Polymorphism. Genes (Basel) 2022; 13:genes13071204. [PMID: 35885987 PMCID: PMC9324515 DOI: 10.3390/genes13071204] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023] Open
Abstract
Single nucleotide polymorphisms (SNPs) reportedly influence the effect of nivolumab in metastatic renal cell carcinoma (mRCC). This study aimed to evaluate the relationship between the clinical outcomes of patients with mRCC and SNPs in programmed cell death protein 1 (PD-1) protein-coding gene (PDCD1) and explore any potential correlation with patient prognosis and incidence of immune-related adverse events (irAEs). In total, 106 patients with mRCC, who were treated with nivolumab alone (n = 59) or nivolumab and ipilimumab (n = 47), were enrolled in the study. Three SNPs in the PDCD1 gene, namely PD-1.3, PD-1.5, and PD-1.6, were assessed. Patients harboring the PD-1.6 G allele experienced more severe (odds ratio, 3.390; 95% confidence interval 1.517–7.756; p = 0.003) and multiple (OR, 2.778; 95% CI, 1.020–6.993 p = 0.031) irAEs than those harboring the AA genotype. Thus, the existence of the PDCD1 PD-1.6 polymorphism (G allele) was associated with the occurrence of severe and multiple irAEs in patients with mRCC. Further evaluation of PDCD1 polymorphisms might help identify patients experiencing irAE by nivolumab treatment.
Collapse
Affiliation(s)
- Mizuki Kobayashi
- Department of Urology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (M.K.); (Y.M.); (Y.S.); (H.S.); (S.K.); (R.Y.); (A.K.); (T.N.); (M.S.); (S.N.); (T.H.)
| | - Kazuyuki Numakura
- Department of Urology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (M.K.); (Y.M.); (Y.S.); (H.S.); (S.K.); (R.Y.); (A.K.); (T.N.); (M.S.); (S.N.); (T.H.)
- Correspondence: ; Tel.: +81-18-884-6460
| | - Shingo Hatakeyama
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8203, Japan; (S.H.); (C.O.)
| | - Yumina Muto
- Department of Urology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (M.K.); (Y.M.); (Y.S.); (H.S.); (S.K.); (R.Y.); (A.K.); (T.N.); (M.S.); (S.N.); (T.H.)
| | - Yuya Sekine
- Department of Urology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (M.K.); (Y.M.); (Y.S.); (H.S.); (S.K.); (R.Y.); (A.K.); (T.N.); (M.S.); (S.N.); (T.H.)
| | - Hajime Sasagawa
- Department of Urology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (M.K.); (Y.M.); (Y.S.); (H.S.); (S.K.); (R.Y.); (A.K.); (T.N.); (M.S.); (S.N.); (T.H.)
| | - Soki Kashima
- Department of Urology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (M.K.); (Y.M.); (Y.S.); (H.S.); (S.K.); (R.Y.); (A.K.); (T.N.); (M.S.); (S.N.); (T.H.)
| | - Ryohei Yamamoto
- Department of Urology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (M.K.); (Y.M.); (Y.S.); (H.S.); (S.K.); (R.Y.); (A.K.); (T.N.); (M.S.); (S.N.); (T.H.)
| | - Atsushi Koizumi
- Department of Urology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (M.K.); (Y.M.); (Y.S.); (H.S.); (S.K.); (R.Y.); (A.K.); (T.N.); (M.S.); (S.N.); (T.H.)
| | - Taketoshi Nara
- Department of Urology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (M.K.); (Y.M.); (Y.S.); (H.S.); (S.K.); (R.Y.); (A.K.); (T.N.); (M.S.); (S.N.); (T.H.)
| | - Mitsuru Saito
- Department of Urology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (M.K.); (Y.M.); (Y.S.); (H.S.); (S.K.); (R.Y.); (A.K.); (T.N.); (M.S.); (S.N.); (T.H.)
| | - Shintaro Narita
- Department of Urology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (M.K.); (Y.M.); (Y.S.); (H.S.); (S.K.); (R.Y.); (A.K.); (T.N.); (M.S.); (S.N.); (T.H.)
| | - Chikara Ohyama
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8203, Japan; (S.H.); (C.O.)
| | - Tomonori Habuchi
- Department of Urology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (M.K.); (Y.M.); (Y.S.); (H.S.); (S.K.); (R.Y.); (A.K.); (T.N.); (M.S.); (S.N.); (T.H.)
| |
Collapse
|
23
|
Li F, Liao B, Wang T, Qi T, Wang Y. Programmed Cell Death Protein 1/Programmed Cell Death Protein Ligand 1 Immunosuppressants in Advanced Non-Small Cell Lung Cancer Research Progress in Treatment. Front Pharmacol 2022; 13:918709. [PMID: 35784705 PMCID: PMC9243588 DOI: 10.3389/fphar.2022.918709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/25/2022] [Indexed: 12/03/2022] Open
Abstract
PD-1/PD-L1 play key roles in tumor immune escape and the formation of the tumor microenvironment, and are closely related to the generation and development of tumors. Blocking the PD-1/PD-L1 pathway can reshape the tumor microenvironment or block the formation of the tumor microenvironment and enhance endogenous antitumor immune response. Clinical trials show that the treatment of non-small cell lung cancer (NSCLC) with PD-1/PD-L1 inhibitors has significant advantages. The review briefly describes these basic principles of the PD-1/PD-L1 pathway and action mechanism in the treatment of NSCLC. A summary of global PD-1/PD-L1 clinical trials and five PD-1/PD-L1 inhibitors approved by FDA, EMA and NMPA for advanced NSCLC were analyzed.
Collapse
Affiliation(s)
- Feng Li
- Sichuan Cancer Hospital, Cancer Hospital Affiliate to School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Binchi Liao
- National Medical Products Administration Institute of Executive Development, Beijing, China
| | - Ting Wang
- Sichuan Cancer Hospital, Cancer Hospital Affiliate to School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tingting Qi
- Sichuan Cancer Hospital, Cancer Hospital Affiliate to School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yixin Wang
- Sichuan Cancer Hospital, Cancer Hospital Affiliate to School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Yixin Wang,
| |
Collapse
|
24
|
Mohammadi P, Hesari M, Chalabi M, Salari F, Khademi F. An overview of immune checkpoint therapy in autoimmune diseases. Int Immunopharmacol 2022; 107:108647. [DOI: 10.1016/j.intimp.2022.108647] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 02/17/2022] [Accepted: 02/20/2022] [Indexed: 02/06/2023]
|
25
|
Ni P, Wang G, Wang Y, Liu K, Chen W, Xiao J, Fan H, Ma X, Li Z, Shen K, Xu Z, Yang L. Correlation of MIF-AS1 polymorphisms with the risk and prognosis of gastric cancer. Pathol Res Pract 2022; 233:153850. [DOI: 10.1016/j.prp.2022.153850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/05/2022] [Accepted: 03/17/2022] [Indexed: 12/24/2022]
|
26
|
David S, Tan J, Siva S, Karroum L, Savas P, Loi S. Combining Radiotherapy and Immunotherapy in Metastatic Breast Cancer: Current Status and Future Directions. Biomedicines 2022; 10:821. [PMID: 35453571 PMCID: PMC9024725 DOI: 10.3390/biomedicines10040821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
The role of radiotherapy and immunotherapy with immune checkpoint inhibitors (ICI) is of emerging interest in many solid tumours, including breast cancer. There is increasing evidence that the host's immune system plays an important role in influencing the response to treatment and prognosis in breast cancer. Several pre-clinical studies and clinical trials have reported on the 'abscopal effect-regression of distant untreated tumour sites, mediated by an immunological response following ionizing radiation to a targeted tumour site. Stereotactic Ablative Body Radiotherapy (SABR) is a non-invasive technique used to augment various immune responses with an ablative tumoricidal dose when compared to conventional radiotherapy. SABR is characterized by typically 1-5 precision radiotherapy treatments that simultaneously deliver a high dose, whilst sparing normal tissues. Following SABR, there is evidence of systemic immune activation in patients with increased PD1 expression on CD8+ and CD4+ T cells. Studies continue to focus on metastatic triple-negative disease, a highly immunogenic subtype of breast cancer with poor prognosis. In this review, we discuss the immunological effect of SABR, alone and in combination with immunotherapy, and the importance of dose and fractionation. We also propose future strategies for treating oligometastatic disease, where this approach may be most useful for producing durable responses.
Collapse
Affiliation(s)
- Steven David
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (J.T.); (S.S.); (L.K.)
| | - Jennifer Tan
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (J.T.); (S.S.); (L.K.)
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3000, Australia; (P.S.); (S.L.)
| | - Shankar Siva
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (J.T.); (S.S.); (L.K.)
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3000, Australia; (P.S.); (S.L.)
| | - Lama Karroum
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (J.T.); (S.S.); (L.K.)
| | - Peter Savas
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3000, Australia; (P.S.); (S.L.)
- Department of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Sherene Loi
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3000, Australia; (P.S.); (S.L.)
- Department of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| |
Collapse
|
27
|
Yaseen MM, Abuharfeil NM, Darmani H. CMTM6 as a master regulator of PD-L1. Cancer Immunol Immunother 2022; 71:2325-2340. [PMID: 35294592 DOI: 10.1007/s00262-022-03171-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/08/2022] [Indexed: 12/15/2022]
Abstract
Immune checkpoint proteins, such as programmed cell death receptor 1 (PD-1) and its ligand (PD-L1), play critical roles in the pathology of chronic inflammatory pathological conditions, particularly cancer. In addition, the activation of PD-1/PD-L1 pathway is involved in mediating resistance to certain anti-cancer chemo- and immuno-therapeutics. Unfortunately, targeting the PD-1/PD-L1 pathway by the available anti-PD-1/PD-L1 drugs can benefit only a small proportion of cancer patients. Thus, studying the factors that regulate the expression of these immune checkpoint proteins is of central importance in this context. Recent investigations have identified CMTM6 and, to a lesser extent, CMTM4, as master regulators of PD-L1 expression in various cancer cells. Understanding the mechanisms by which such proteins upregulate the expression of PD-L1 in tumor cells, and determining the potential regulators of CMTM6 expression in different types of cancers will accelerate the development of new therapeutic targets and/or lead to the enhancement of the currently available PD-1/PD-L1 blockade therapies.
Collapse
Affiliation(s)
- Mahmoud Mohammad Yaseen
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Nizar Mohammad Abuharfeil
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Homa Darmani
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
28
|
Expression of PD-1, PD-L1 and PD-L2 in Lymphomas in Patients with Pre-Existing Rheumatic Diseases-A Possible Association with High Rheumatoid Arthritis Disease Activity. Cancers (Basel) 2022; 14:cancers14061509. [PMID: 35326658 PMCID: PMC8946311 DOI: 10.3390/cancers14061509] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
Current research seeks to identify subgroups of non-Hodgkin lymphoma (NHL) patients responsive to PD-1 blocking agents. Whether patients with pre-existing rheumatic diseases might constitute such a subgroup is unknown. We determined intratumoral expression of PD-1 and its ligands in lymphoma patients with pre-existing rheumatic diseases. We included 215 patients with rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) or Sjögren's syndrome with subsequent lymphoma and 74 diffuse large B-cell lymphoma (DLBCL) controls without rheumatic disease. PD-1 and PD-ligand immunohistochemical markers were applied on tumor tissue microarrays. The number of PD-1+ tumor infiltrating leukocytes (TILs) and proportions of PD-L1+ and PD-L2+ tumor cells and TILs were calculated and correlated with clinical data. Expression of PD-L1 in tumor cells and TILs was highest in classical Hodgkin lymphoma and DLBCL. In DLBCLs, expression of PD-1 in TILs and PD-L1 in tumor cells was similar in RA, SLE and controls. In RA-DLBCL, high expression of PD-L1 in tumor cells was significantly more common in patients with the most severe RA disease and was associated with inferior overall survival in multivariable analysis.
Collapse
|
29
|
Merlo LM, Peng W, DuHadaway JB, Montgomery JD, Prendergast GC, Muller AJ, Mandik-Nayak L. The Immunomodulatory Enzyme IDO2 Mediates Autoimmune Arthritis through a Nonenzymatic Mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:571-581. [PMID: 34965962 PMCID: PMC8770583 DOI: 10.4049/jimmunol.2100705] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/15/2021] [Indexed: 02/03/2023]
Abstract
IDO2 is one of two closely related tryptophan catabolizing enzymes induced under inflammatory conditions. In contrast to the immunoregulatory role defined for IDO1 in cancer models, IDO2 has a proinflammatory function in models of autoimmunity and contact hypersensitivity. In humans, two common single-nucleotide polymorphisms have been identified that severely impair IDO2 enzymatic function, such that <25% of individuals express IDO2 with full catalytic potential. This, together with IDO2's relatively weak enzymatic activity, suggests that IDO2 may have a role outside of its function in tryptophan catabolism. To determine whether the enzymatic activity of IDO2 is required for its proinflammatory function, we used newly generated catalytically inactive IDO2 knock-in mice together with established models of contact hypersensitivity and autoimmune arthritis. Contact hypersensitivity was attenuated in catalytically inactive IDO2 knock-in mice. In contrast, induction of autoimmune arthritis was unaffected by the absence of IDO2 enzymatic activity. In pursuing this nonenzymatic IDO2 function, we identified GAPDH, Runx1, RANbp10, and Mgea5 as IDO2-binding proteins that do not interact with IDO1, implicating them as potential mediators of IDO2-specific function. Taken together, our findings identify a novel function for IDO2, independent of its tryptophan catabolizing activity, and suggest that this nonenzymatic function could involve multiple signaling pathways. These data show that the enzymatic activity of IDO2 is required only for some inflammatory immune responses and provide, to our knowledge, the first evidence of a nonenzymatic role for IDO2 in mediating autoimmune disease.
Collapse
Affiliation(s)
| | - Weidan Peng
- Lankenau Institute for Medical Research, Wynnewood, PA
| | | | | | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA,Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | | | |
Collapse
|
30
|
Dolatkhah K, Alizadeh N, Mohajjel-Shoja H, Abdoli Shadbad M, Hajiasgharzadeh K, Aghebati-Maleki L, Baghbanzadeh A, Hosseinkhani N, Karim Ahangar N, Baradaran B. B7 immune checkpoint family members as putative therapeutics in autoimmune disease: An updated overview. Int J Rheum Dis 2022; 25:259-271. [PMID: 34994525 DOI: 10.1111/1756-185x.14273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/11/2021] [Accepted: 12/18/2021] [Indexed: 12/23/2022]
Abstract
Autoimmune diseases, especially among young people in the US, are one of the leading causes of morbidity and death. The immune responses are the fundamental pathogenicity of autoimmune disorders. The equilibrium between stimulatory and inhibitory signals is critical for the stimulation, migration, survival, and T cell-related immune responses. The B7 family can substantially regulate T cell-mediated immune responses. Nevertheless, recent breakthroughs in immune checkpoint blockade in cancer immunotherapy have facilitated autoimmune diseases, especially among the prone populations. In the current study, we tried to concisely review the role of the B7 family in regulating immune reactions and the influence of immune checkpoint inhibitors on autoimmunity development.
Collapse
Affiliation(s)
- Katayoun Dolatkhah
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hanieh Mohajjel-Shoja
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | | | | | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negar Hosseinkhani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Noora Karim Ahangar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
31
|
Mor A, Strazza M. Bridging the Gap: Connecting the Mechanisms of Immune-Related Adverse Events and Autoimmunity Through PD-1. Front Cell Dev Biol 2022; 9:790386. [PMID: 35047501 PMCID: PMC8762228 DOI: 10.3389/fcell.2021.790386] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/30/2021] [Indexed: 12/19/2022] Open
Abstract
The emergence of anti-cytotoxic T-lymphocyte antigen 4 (anti-CTLA-4), anti-programmed cell death 1 ligand (anti-PD-1), and anti-PD-L1 antibodies as immune checkpoint inhibitors (ICIs) revolutionized the treatment of numerous types of tumors. These antibodies, both alone and in combination, provide great clinical efficacy as evidenced by tumor regression and increased overall patients' survival. However, with this success comes multiple challenges. First, while patients who respond to ICIs have outstanding outcomes, there remains a large proportion of patients who do not respond at all. This all-or-none response has led to looking downstream of programmed cell death 1 (PD-1) for additional therapeutic targets and for new combination therapies. Second, a majority of patients who receive ICIs go on to develop immune-related adverse events (irAEs) characterized by end-organ inflammation with T-cell infiltrates. The hallmarks of these clinically observed irAEs share many similarities with primary autoimmune diseases. The contribution of PD-1 to peripheral tolerance is a major mechanism for protection against expansion of self-reactive T-cell clones and autoimmune disease. In this review, we aim to bridge the gaps between our cellular and molecular knowledge of PD-1 signaling in T cells, ICI-induced irAEs, and autoimmune diseases. We will highlight shared mechanisms and the potential for new therapeutic strategies.
Collapse
Affiliation(s)
- Adam Mor
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Marianne Strazza
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
32
|
Mi Y, Han J, Zhu J, Jin T. Role of the PD-1/PD-L1 Signaling in Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis: Recent Insights and Future Directions. Mol Neurobiol 2021; 58:6249-6271. [PMID: 34480337 PMCID: PMC8639577 DOI: 10.1007/s12035-021-02495-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is an autoimmunity-related chronic demyelination disease of the central nervous system (CNS), causing young disability. Currently, highly specific immunotherapies for MS are still lacking. Programmed cell death 1 (PD-1) is an immunosuppressive co-stimulatory molecule, which is expressed on activated T lymphocytes, B lymphocytes, natural killer cells, and other immune cells. PD-L1, the ligand of PD-1, is expressed on T lymphocytes, B lymphocytes, dendritic cells, and macrophages. PD-1/PD-L1 delivers negative regulatory signals to immune cells, maintaining immune tolerance and inhibiting autoimmunity. This review comprehensively summarizes current insights into the role of PD-1/PD-L1 signaling in MS and its animal model experimental autoimmune encephalomyelitis (EAE). The potentiality of PD-1/PD-L1 as biomarkers or therapeutic targets for MS will also be discussed.
Collapse
Affiliation(s)
- Yan Mi
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
| | - Jinming Han
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
- Present Address: Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
| |
Collapse
|
33
|
Prazak L, Iwasaki Y, Kim AR, Kozlov K, King K, Gergen JP. A dual role for DNA binding by Runt in activation and repression of sloppy paired transcription. Mol Biol Cell 2021; 32:ar26. [PMID: 34432496 PMCID: PMC8693977 DOI: 10.1091/mbc.e20-08-0509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
This work investigates the role of DNA binding by Runt in regulating the sloppy paired 1 (slp1) gene and in particular two distinct cis-regulatory elements that mediate regulation by Runt and other pair-rule transcription factors during Drosophila segmentation. We find that a DNA-binding-defective form of Runt is ineffective at repressing both the distal (DESE) and proximal (PESE) early stripe elements of slp1 and is also compromised for DESE-dependent activation. The function of Runt-binding sites in DESE is further investigated using site-specific transgenesis and quantitative imaging techniques. When DESE is tested as an autonomous enhancer, mutagenesis of the Runt sites results in a clear loss of Runt-dependent repression but has little to no effect on Runt-dependent activation. Notably, mutagenesis of these same sites in the context of a reporter gene construct that also contains the PESE enhancer results in a significant reduction of DESE-dependent activation as well as the loss of repression observed for the autonomous mutant DESE enhancer. These results provide strong evidence that DNA binding by Runt directly contributes to the regulatory interplay of interactions between these two enhancers in the early embryo.
Collapse
Affiliation(s)
- Lisa Prazak
- Department of Biology, Farmingdale State College, Farmingdale, NY 11735-1021.,Department of Biochemistry and Cell Biology and Center for Developmental Genetics.,Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794-5215
| | - Yasuno Iwasaki
- Department of Biochemistry and Cell Biology and Center for Developmental Genetics
| | - Ah-Ram Kim
- Graduate Program in Biochemistry and Structural Biology, and
| | - Konstantin Kozlov
- Department of Applied Mathematics, St. Petersburg State Polytechnical University, St. Petersburg, Russia 195251
| | - Kevin King
- Department of Biochemistry and Cell Biology and Center for Developmental Genetics.,Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794-5215
| | - J Peter Gergen
- Department of Biochemistry and Cell Biology and Center for Developmental Genetics
| |
Collapse
|
34
|
Curnock AP, Bossi G, Kumaran J, Bawden LJ, Figueiredo R, Tawar R, Wiseman K, Henderson E, Hoong SJ, Gonzalez V, Ghadbane H, Knight DE, O'Dwyer R, Overton DX, Lucato CM, Smith NM, Reis CR, Page K, Whaley LM, McCully ML, Hearty S, Mahon TM, Weber P. Cell-targeted PD-1 agonists that mimic PD-L1 are potent T cell inhibitors. JCI Insight 2021; 6:e152468. [PMID: 34491911 PMCID: PMC8564903 DOI: 10.1172/jci.insight.152468] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/03/2021] [Indexed: 12/21/2022] Open
Abstract
The PD-1/PD-L1 pathway is a key immune checkpoint that regulates T cell activation. There is strong rationale to develop PD-1 agonists as therapeutics against autoimmunity, but progress in this area has been limited. Here, we generated T cell receptor (TCR) targeting, PD-1 agonist bispecifics called ImmTAAI molecules that mimic the ability of PD-L1 to facilitate the colocalization of PD-1 with the TCR complex at the target cell-T cell interface. PD-1 agonist ImmTAAI molecules specifically bound to target cells and were highly effective in activating the PD-1 receptor on interacting T cells to achieve immune suppression. Potent PD-1 antibody ImmTAAI molecules closely mimicked the mechanism of action of endogenously expressed PD-L1 in their localization to the target cell-T cell interface, inhibition of proximal TCR signaling events, and suppression of T cell function. At picomolar concentrations, these bispecifics suppressed cytokine production and inhibited CD8+ T cell-mediated cytotoxicity in vitro. Crucially, in soluble form, the PD-1 ImmTAAI molecules were inactive and, hence, could avoid systemic immunosuppression. This study outlines a promising new route to generate more effective, potent, tissue-targeted PD-1 agonists that can inhibit T cell function locally with the potential to treat autoimmune and chronic inflammatory diseases of high unmet need.
Collapse
|
35
|
Gatselis NK, Azariadis K, Lyberopoulou A, Dalekos GN. Programmed cell death-1 rs11568821 and interleukin-28B rs12979860 polymorphisms in autoimmune hepatitis. J Transl Autoimmun 2021; 4:100126. [PMID: 34632357 PMCID: PMC8488593 DOI: 10.1016/j.jtauto.2021.100126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Autoimmune hepatitis (AIH) is a relatively rare chronic liver disease of unknown etiology. The genetic background affects susceptibility, clinical phenotype, and prognosis. The programmed cell death-1 rs11568821 polymorphism (PD1.3) has been associated with susceptibility to autoimmune diseases. The interleukin-28B (IL28B) rs12979860 polymorphism has been associated with steatosis, inflammation, and fibrosis in liver diseases. AIM Our aim was to investigate for the first time the incidence and clinical significance of PD1.3 and IL28B rs12979860 in AIH. METHODS Two hundred patients with AIH were evaluated, while 100 healthy subjects were used as controls. Genotyping was performed with in-house allelic discrimination End-Point PCR. RESULTS The SNP PD1.3/A was present in 36/200 (18%) AIH patients compared to 28/100 (28%) healthy controls (p = 0.065). The AA/GA genotypes were not associated with the mode of presentation of AIH, the histological grade or stage, the presence of cirrhosis, risk of disease progression, response to treatment and survival. The IL28B rs12979860 genotype distribution was CC 79/200 (39.5%), TT 36/200 (18%) and CT 85/200 (42.5%), in similar rates with healthy controls (p = 0.878). Inflammatory activity and fibrosis stage did not differ between CC homozygotes and CT/TT carriers. LDL cholesterol was significantly higher in CC than CT/TT patients (P = 0.027), though no differences was found regarding the presence of steatosis or steatohepatitis. On-treatment response to immunosuppressive treatment was not affected by the IL28B rs12979860 polymorphism. However, CC homozygotes AIH patients achieved treatment withdrawal in significantly higher rates (OR 2.3, 95%CI: 1.1-4.7, P = 0.02) irrespective of the presence of steatosis or steatohepatitis. CONCLUSIONS The PD1.3 and IL28B rs12979860 variants are unlikely to contribute to AIH susceptibility, disease presentation and prognosis. The IL28B rs12979860 is not associated with the presence of concurrent steatosis or steatohepatitis. However, although on-treatment response rates to immunosuppression were not affected by the IL28B rs12979860 polymorphism, AIH patients with CC homozygosity were more likely to achieve complete treatment withdrawal. This novel finding needs validation and further clarification from larger multicenter studies.
Collapse
Key Words
- AIH, Autoimmune hepatitis.
- ANA, Antinuclear antibodies.
- Anti-LC1, Liver cytosol type-1 antibodies.
- Anti-LKM1, Liver kidney microsomal type-1 antibodies
- Anti-SLA/LP, Soluble liver antigen/liver pancreas antibodies.
- Autoimmune hepatitis
- CR, Complete response.
- HCC, Hepatocellular carcinoma.
- HCV, Hepatis C virus.
- HDL, High density lipoprotein.
- HLA, Human leukocyte antigen.
- HWE, Hardy-weinberg equilibrium.
- IL28B, Interleukin 28B.
- INR, International normalized ratio.
- IQR, Interquartile range.
- IgG, Immunoglobulin class G.
- Interleukin-28B
- LDL, Low density lipoprotein
- MetS, Metabolic syndrome.
- NAFLD, Non-alcoholic fatty liver disease.
- PCR, Polymerase chain reaction.
- PD1, Programmed cell death-1.
- Polymorphisms
- Programmed cell death-1
- SD, Standard deviation.
- SLE, Systemic lupus erythematosus.
- SMA, Smooth muscle antibodies.
- SNP, Single nucleotide polymorphism.
- ULN, Upper limit of normal.
Collapse
Affiliation(s)
- Nikolaos K. Gatselis
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, General University Hospital of Larissa, 41110, Larissa, Greece
| | - Kalliopi Azariadis
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, General University Hospital of Larissa, 41110, Larissa, Greece
| | - Aggeliki Lyberopoulou
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, General University Hospital of Larissa, 41110, Larissa, Greece
| | - George N. Dalekos
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, General University Hospital of Larissa, 41110, Larissa, Greece
| |
Collapse
|
36
|
CD8 + T effector and immune checkpoint signatures predict prognosis and responsiveness to immunotherapy in bladder cancer. Oncogene 2021; 40:6223-6234. [PMID: 34552192 DOI: 10.1038/s41388-021-02019-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/25/2021] [Accepted: 09/10/2021] [Indexed: 11/08/2022]
Abstract
Immune-checkpoint blockade (ICB) has been routinely implemented to treat bladder cancer; however, most patients have little or no clinical benefit. In this study, 348 pretreated metastatic urothelial cancer samples from the IMvigor210 cohort were used to identify important genes significantly associated with CD8+ T effector and immune checkpoint signatures. The immune checkpoint inhibitor score (IMS) scoring system was constructed to predict the immunotherapy responsiveness. Transcriptome analysis confirmed that the high IMS score group had significant immune activation with better prognosis and higher immunotherapy responsiveness, which was a powerful biomarker for predicting the prognosis and responsiveness of ICB. Tumor immune dysfunction and exclusion (TIDE) scores were calculated using 2031 external bladder cancer samples for further validation. We selected the important Hub genes as potential therapeutic targets, and validated the genes using genomic, transcriptomic, immunomic, and other multi-omics methods. In addition, we construct a risk prediction model which could stratify patients with bladder cancer and predict patient prognosis and ICB treatment responsiveness. In conclusion, this study identified effective biomarkers for the prediction of immune checkpoint inhibitor treatment responsiveness in bladder cancer patients and provided new immunotherapeutic targets.
Collapse
|
37
|
Foster CC, Couey MA, Kochanny SE, Khattri A, Acharya RK, Tan YHC, Brisson RJ, Leidner RS, Seiwert TY. Immune-related adverse events are associated with improved response, progression-free survival, and overall survival for patients with head and neck cancer receiving immune checkpoint inhibitors. Cancer 2021; 127:4565-4573. [PMID: 34547103 DOI: 10.1002/cncr.33780] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/31/2021] [Accepted: 06/05/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND The authors hypothesized that patients developing immune-related adverse events (irAEs) while receiving immune checkpoint inhibition (ICI) for recurrent/metastatic head and neck cancer (HNC) would have improved oncologic outcomes. METHODS Patients with recurrent/metastatic HNC received ICI at 2 centers. Univariate and multivariate logistic regression, Kaplan-Meier methods, and Cox proportional hazards regression were used to associate the irAE status with the overall response rate (ORR), progression-free survival (PFS), and overall survival (OS) in cohort 1 (n = 108). These outcomes were also analyzed in an independent cohort of patients receiving ICI (cohort 2; 47 evaluable for irAEs). RESULTS The median follow-up was 8.4 months for patients treated in cohort 1. Sixty irAEs occurred in 49 of 108 patients with 5 grade 3 or higher irAEs (10.2%). ORR was higher for irAE+ patients (30.6%) in comparison with irAE- patients (12.3%; P = .02). The median PFS was 6.9 months for irAE+ patients and 2.1 months for irAE- patients (P = .0004), and the median OS was 12.5 and 6.8 months, respectively (P = .007). Experiencing 1 or more irAEs remained associated with ORR (P = .03), PFS (P = .003), and OS (P = .004) in multivariate analyses. The association between development of irAEs and prolonged OS persisted in a 22-week landmark analysis (P = .049). The association between development of irAEs and favorable outcomes was verified in cohort 2. CONCLUSIONS The development of irAEs was strongly associated with an ICI benefit, including overall response, PFS, and OS, in 2 separate cohorts of patients with recurrent/metastatic HNC.
Collapse
Affiliation(s)
- Corey C Foster
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Marcus A Couey
- Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, Oregon
| | - Sara E Kochanny
- Section of Hematology/Oncology, Department of Medicine and Comprehensive Cancer Center, University of Chicago Medicine, Chicago, Illinois
| | - Arun Khattri
- Section of Hematology/Oncology, Department of Medicine and Comprehensive Cancer Center, University of Chicago Medicine, Chicago, Illinois
| | - Rajesh K Acharya
- Section of Hematology/Oncology, Department of Medicine and Comprehensive Cancer Center, University of Chicago Medicine, Chicago, Illinois
| | - Yi-Hung Carol Tan
- Section of Hematology/Oncology, Department of Medicine and Comprehensive Cancer Center, University of Chicago Medicine, Chicago, Illinois
| | - Ryan J Brisson
- Department of Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Rom S Leidner
- Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, Oregon
| | - Tanguy Y Seiwert
- Departments of Oncology and Otolaryngology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
38
|
Inherited PD-1 deficiency underlies tuberculosis and autoimmunity in a child. Nat Med 2021; 27:1646-1654. [PMID: 34183838 PMCID: PMC8446316 DOI: 10.1038/s41591-021-01388-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
The pathophysiology of adverse events following programmed cell death protein 1 (PD-1) blockade, including tuberculosis (TB) and autoimmunity, remains poorly characterized. We studied a patient with inherited PD-1 deficiency and TB who died of pulmonary autoimmunity. The patient's leukocytes did not express PD-1 or respond to PD-1-mediated suppression. The patient's lymphocytes produced only small amounts of interferon (IFN)-γ upon mycobacterial stimuli, similarly to patients with inborn errors of IFN-γ production who are vulnerable to TB. This phenotype resulted from a combined depletion of Vδ2+ γδ T, mucosal-associated invariant T and CD56bright natural killer lymphocytes and dysfunction of other T lymphocyte subsets. Moreover, the patient displayed hepatosplenomegaly and an expansion of total, activated and RORγT+ CD4-CD8- double-negative αβ T cells, similar to patients with STAT3 gain-of-function mutations who display lymphoproliferative autoimmunity. This phenotype resulted from excessive amounts of STAT3-activating cytokines interleukin (IL)-6 and IL-23 produced by activated T lymphocytes and monocytes, and the STAT3-dependent expression of RORγT by activated T lymphocytes. Our work highlights the indispensable role of human PD-1 in governing both antimycobacterial immunity and self-tolerance, while identifying potentially actionable molecular targets for the diagnostic and therapeutic management of TB and autoimmunity in patients on PD-1 blockade.
Collapse
|
39
|
Nuzzo PV, Pond GR, Abou Alaiwi S, Nassar AH, Flippot R, Curran C, Kilbridge KL, Wei XX, McGregor BA, Choueiri T, Harshman LC, Sonpavde G. Conditional immune toxicity rate in patients with metastatic renal and urothelial cancer treated with immune checkpoint inhibitors. J Immunother Cancer 2021; 8:jitc-2019-000371. [PMID: 32234849 PMCID: PMC7174062 DOI: 10.1136/jitc-2019-000371] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2020] [Indexed: 12/31/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) are associated with immune-related adverse events (irAEs). Although the incidence and prevalence of irAEs have been well characterized in the literature, less is known about the cumulative incidence rate of irAEs. We studied the cumulative incidence of irAEs, defined as the probability of irAE occurrence over time and the risk factors for irAE development in metastatic urothelial carcinoma (mUC) and renal cell carcinoma (mRCC) patients treated with ICIs. Methods We identified a cohort of patients who received ICIs for mUC and mRCC. irAEs were classified using Common Terminology Criteria for Adverse Event (CTCAE) V.5.0 guidelines. The monthly incidence of irAEs over time was reported after landmark duration of therapy. Cumulative incidence of irAEs was calculated to evaluate the time to the first occurrence of an irAE accounting for the competing risk of death. Prognostic factors for irAE were assessed using the Fine and Gray method. Results A total of 470 patients were treated with ICIs between July 2013 and October 2018 (mUC: 199 (42.3%); mRCC: 271 (57.7%)). 341 (72.6%) patients received monotherapy, 86 (18.3%) received ICIs in combination with targeted therapies, and 43 (9.2%) received dual ICI therapy. Overall, 186 patients (39.5%) experienced an irAE at any time point. Common irAEs included hypothyroidism (n=42, 22.6%), rush and pruritus (n=36, 19.4%), diarrhea/colitis (n=35, 18.8%), transaminitis (n=32, 17.2%), and pneumonitis (n=14, 7.5%). Monthly incidence rates decreased over time; however, 17 of 109 (15.6%, 95% CI: 9.4% to 23.8%) experienced their first irAE at least 1 year after treatment initiation. No differences in cumulative incidence were observed based on cancer type, agent, or irAE grade. On multivariable analysis, combined ICI therapy with another ICI or with targeted therapy (p<0.001), first-line ICI therapy (p=0.011), and PD-1 inhibitor therapy (p=0.007) were all significantly associated with irAE development. Conclusions This study quantitates the incidence of developing irAEs due to ICI conditioned on time elapsed without irAE development. Although the monthly incidence of irAEs decreased over time on therapy, patients can still develop delayed irAEs beyond ICI discontinuation, and thus, continuous vigilant monitoring is warranted.
Collapse
Affiliation(s)
- Pier Vitale Nuzzo
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Gregory R Pond
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Sarah Abou Alaiwi
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Amin H Nassar
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ronan Flippot
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Catherine Curran
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kerry L Kilbridge
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Xiao X Wei
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Bradley A McGregor
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Toni Choueiri
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Lauren C Harshman
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Guru Sonpavde
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
40
|
Kelkar MG, Bargir UA, Malik-Yadav R, Gupta M, Dalvi A, Jodhawat N, Shinde S, Madkaikar MR. CD8 + T Cells Exhibit an Exhausted Phenotype in Hemophagocytic Lymphohistiocytosis. J Clin Immunol 2021; 41:1794-1803. [PMID: 34389889 DOI: 10.1007/s10875-021-01109-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/22/2021] [Indexed: 12/31/2022]
Abstract
PURPOSE Hemophagocytic lymphohistiocytosis (HLH) is a hyperinflammatory syndrome mainly caused by uncontrolled activation of antigen presenting cells and CD8 T cells. CD8 T cell exhaustion is a known phenomenon in chronic viral infections and cancer. However, the role of T cell exhaustion is not yet identified in HLH in the background of persistent inflammation. So, currently, we have characterized the CD8 T cells using flow cytometry to understand the phenomenon of exhaustion in these cells in HLH. METHODS We have comprehensively evaluated lymphocyte subsets and characterized CD8 T cells using immunophenotypic markers like PD1, TIM3, LAG3, Ki67, Granzyme B, etc. in a cohort of 21 HLH patients. Effector cytokine secretion and degranulation by CD8 T cells are also studied. RESULTS Our findings indicate skewed lymphocyte subsets and aberrantly activated CD8 T cells in HLH. CD8 T cells exhibit significantly increased expression of PD1, TIM3, and LAG3 prominently in primary HLH as compared to controls. PD1 + CD8 T cells express elevated levels of Granzyme B and Ki67. Moreover, CD8 T cells are hypofunctional as evidenced by significantly reduced cytokine secretion and compromised CD107a degranulation. CONCLUSION The study has revealed that CD8 + cytotoxic T lymphocytes from HLH patients exhibited high expression of exhaustion markers with overall impaired function. To the best of our understanding, this is the first report suggesting functional exhaustion of CD8 T cells in both primary and secondary HLH. Future studies to understand the association of exhaustion with disease outcome are needed for its probable therapeutic implementation.
Collapse
Affiliation(s)
- Madhura G Kelkar
- Department of Pediatric Immunology and Leukocyte Biology, ICMR-National Institute of Immunohaematology (NIIH), 13th Floor, NMS Building, KEM Hospital Campus, Parel, , Mumbai, 400012, India
| | - Umair Ahmad Bargir
- Department of Pediatric Immunology and Leukocyte Biology, ICMR-National Institute of Immunohaematology (NIIH), 13th Floor, NMS Building, KEM Hospital Campus, Parel, , Mumbai, 400012, India
| | - Reetika Malik-Yadav
- Department of Pediatric Immunology and Leukocyte Biology, ICMR-National Institute of Immunohaematology (NIIH), 13th Floor, NMS Building, KEM Hospital Campus, Parel, , Mumbai, 400012, India
| | - Maya Gupta
- Department of Pediatric Immunology and Leukocyte Biology, ICMR-National Institute of Immunohaematology (NIIH), 13th Floor, NMS Building, KEM Hospital Campus, Parel, , Mumbai, 400012, India
| | - Aparna Dalvi
- Department of Pediatric Immunology and Leukocyte Biology, ICMR-National Institute of Immunohaematology (NIIH), 13th Floor, NMS Building, KEM Hospital Campus, Parel, , Mumbai, 400012, India
| | - Neha Jodhawat
- Department of Pediatric Immunology and Leukocyte Biology, ICMR-National Institute of Immunohaematology (NIIH), 13th Floor, NMS Building, KEM Hospital Campus, Parel, , Mumbai, 400012, India
| | - Shweta Shinde
- Department of Pediatric Immunology and Leukocyte Biology, ICMR-National Institute of Immunohaematology (NIIH), 13th Floor, NMS Building, KEM Hospital Campus, Parel, , Mumbai, 400012, India
| | - Manisha R Madkaikar
- Department of Pediatric Immunology and Leukocyte Biology, ICMR-National Institute of Immunohaematology (NIIH), 13th Floor, NMS Building, KEM Hospital Campus, Parel, , Mumbai, 400012, India.
| |
Collapse
|
41
|
Li H, Zheng C, Han J, Zhu J, Liu S, Jin T. PD-1/PD-L1 Axis as a Potential Therapeutic Target for Multiple Sclerosis: A T Cell Perspective. Front Cell Neurosci 2021; 15:716747. [PMID: 34381337 PMCID: PMC8350166 DOI: 10.3389/fncel.2021.716747] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 06/22/2021] [Indexed: 12/19/2022] Open
Abstract
The programmed cell death protein-1/programmed death ligand-1 (PD-1/PD-L1) axis is a widely studied immune checkpoint that modulates signaling pathways related to T cell activation. The use of PD-1/PD-L1 inhibitors is a promising immune therapy strategy for cancer patients. However, individuals treated with PD-1/PD-L1 inhibitors may develop immune-related adverse events due to excessive immune reactions. Multiple sclerosis (MS) is a chronic demyelinating and neurodegenerative disease of the central nervous system. T cells and the PD-1/PD-L1 axis play vital roles in the pathogenesis of MS. A better understanding of the complex relationship between the PD-1/PD-L1 axis and T cells may extend our knowledge of the molecular mechanisms and therapeutic approaches for MS. In this review, we summarize the most recent findings regarding the role of the PD-1/PD-L1 axis in MS and discuss the potential therapeutic strategies to modulate the expression of PD-1/PD-L1 in MS.
Collapse
Affiliation(s)
- HaiXia Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Chao Zheng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jinming Han
- Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Shan Liu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
42
|
Abstract
Programmed cell death protein-1 (PD-1) inhibitory antibodies, often referred to as checkpoint inhibitors, have revolutionized the treatment of cancer. However, there is an unmet need for PD-1 agonists to treat autoimmune disorders. Herein, we describe the de novo design of a small, stable PD-1 binding protein and development of a synthetic PD-1 agonist. Programmed cell death protein-1 (PD-1) expressed on activated T cells inhibits T cell function and proliferation to prevent an excessive immune response, and disease can result if this delicate balance is shifted in either direction. Tumor cells often take advantage of this pathway by overexpressing the PD-1 ligand PD-L1 to evade destruction by the immune system. Alternatively, if there is a decrease in function of the PD-1 pathway, unchecked activation of the immune system and autoimmunity can result. Using a combination of computation and experiment, we designed a hyperstable 40-residue miniprotein, PD-MP1, that specifically binds murine and human PD-1 at the PD-L1 interface with a Kd of ∼100 nM. The apo crystal structure shows that the binder folds as designed with a backbone RMSD of 1.3 Å to the design model. Trimerization of PD-MP1 resulted in a PD-1 agonist that strongly inhibits murine T cell activation. This small, hyperstable PD-1 binding protein was computationally designed with an all-beta interface, and the trimeric agonist could contribute to treatments for autoimmune and inflammatory diseases.
Collapse
|
43
|
Ponce de León C, Lorite P, López-Casado MÁ, Barro F, Palomeque T, Torres MI. Significance of PD1 Alternative Splicing in Celiac Disease as a Novel Source for Diagnostic and Therapeutic Target. Front Immunol 2021; 12:678400. [PMID: 34220824 PMCID: PMC8242946 DOI: 10.3389/fimmu.2021.678400] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/26/2021] [Indexed: 02/05/2023] Open
Abstract
Background We have focused on the alteration of the PD-1/PD-L1 pathway in celiac disease and discussed the roles of the PD1 pathway in regulating the immune response. We explored the idea that the altered mRNA splicing process in key regulatory proteins could represent a novel source to identify diagnostic, prognostic, and therapeutic targets in celiac disease. Methods We characterized the PD1 mRNA variants' profile in CD patients and in response to gluten peptides' incubation after in vitro experiments. Total RNA from whole blood was isolated, and the coding region of the human PD-1 mRNA was amplified by cDNA PCR. Results PCR amplification of the human PD-1 coding sequence revealed an association between the over-expression of the sPD-1 protein and the PD-1Δex3 transcript in celiac disease. Thus, we have found three novel alternative spliced isoforms, two of which result in a truncated protein and the other isoform with a loss of 14 aa of exon 2 and complete exon 3 (Δ3) which could encode a new soluble form of PD1 (sPD-1). Conclusions Our study provides evidence that dietary gluten can modulate processes required for cell homeostasis through the splicing of pre-mRNAs encoding key regulatory proteins, which represents an adaptive mechanism in response to different nutritional conditions.
Collapse
Affiliation(s)
| | - Pedro Lorite
- Department of Experimental Biology, Faculty of Health Sciences, University of Jaén, Jaén, Spain
| | | | - Francisco Barro
- Department of Plant Genetic Improvement, Institute for Sustainable Agriculture, Spanish National Research Council (CSIC), Córdoba, Spain
| | - Teresa Palomeque
- Department of Experimental Biology, Faculty of Health Sciences, University of Jaén, Jaén, Spain
| | - María Isabel Torres
- Department of Experimental Biology, Faculty of Health Sciences, University of Jaén, Jaén, Spain
| |
Collapse
|
44
|
Parakh S, Musafer A, Paessler S, Witkowski T, Suen CSNLW, Tutuka CSA, Carlino MS, Menzies AM, Scolyer RA, Cebon J, Dobrovic A, Long GV, Klein O, Behren A. PDCD1 Polymorphisms May Predict Response to Anti-PD-1 Blockade in Patients With Metastatic Melanoma. Front Immunol 2021; 12:672521. [PMID: 34177913 PMCID: PMC8220213 DOI: 10.3389/fimmu.2021.672521] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/21/2021] [Indexed: 01/06/2023] Open
Abstract
A significant number of patients (pts) with metastatic melanoma do not respond to anti-programmed cell death 1 (PD1) therapies. Identifying predictive biomarkers therefore remains an urgent need. We retrospectively analyzed plasma DNA of pts with advanced melanoma treated with PD-1 antibodies, nivolumab or pembrolizumab, for five PD-1 genotype single nucleotide polymorphisms (SNPs): PD1.1 (rs36084323, G>A), PD1.3 (rs11568821, G>A), PD1.5 (rs2227981, C>T) PD1.6 (rs10204225, G>A) and PD1.9 (rs2227982, C>T). Clinico-pathological and treatment parameters were collected, and presence of SNPs correlated with response, progression free survival (PFS) and overall survival (OS). 115 patients were identified with a median follow up of 18.7 months (range 0.26 – 52.0 months). All were Caucasian; 27% BRAF V600 mutation positive. At PD-1 antibody commencement, 36% were treatment-naïve and 52% had prior ipilimumab. The overall response rate was 43%, 19% achieving a complete response. Overall median PFS was 11.0 months (95% CI 5.4 - 17.3) and median OS was 31.1 months (95% CI 23.2 - NA). Patients with the G/G genotype had more complete responses than with A/G genotype (16.5% vs. 2.6% respectively) and the G allele of PD1.3 rs11568821 was significantly associated with a longer median PFS than the AG allele, 14.1 vs. 7.0 months compared to the A allele (p=0.04; 95% CI 0.14 – 0.94). No significant association between the remaining SNPs and responses, PFS or OS were observed. Despite limitations in sample size, this is the first study to demonstrate an association of a germline PD-1 polymorphism and PFS in response to anti-PD-1 therapy in pts with metastatic melanoma. Extrinsic factors like host germline polymorphisms should be considered with tumor intrinsic factors as predictive biomarkers for immune checkpoint regulators.
Collapse
Affiliation(s)
- Sagun Parakh
- Medical Oncology Unit, Austin Health, Melbourne, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia.,La Trobe University School of Cancer Medicine, Melbourne, VIC, Australia
| | - Ashan Musafer
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia.,La Trobe University School of Cancer Medicine, Melbourne, VIC, Australia
| | - Sabrina Paessler
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
| | - Tom Witkowski
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia.,La Trobe University School of Cancer Medicine, Melbourne, VIC, Australia
| | - Connie S N Li Wai Suen
- Department of Mathematics and Statistics, La Trobe University, Melbourne, VIC, Australia
| | | | - Matteo S Carlino
- Department of Medical Oncology, Westmead and Blacktown Hospitals, Sydney, NSW, Australia.,Melanoma Institute Australia, The University of Sydney, North Sydney, NSW, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, North Sydney, NSW, Australia.,Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, North Sydney, NSW, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Jonathan Cebon
- Medical Oncology Unit, Austin Health, Melbourne, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia.,La Trobe University School of Cancer Medicine, Melbourne, VIC, Australia
| | - Alexander Dobrovic
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia.,La Trobe University School of Cancer Medicine, Melbourne, VIC, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, North Sydney, NSW, Australia.,Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, NSW, Australia.,Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - Oliver Klein
- Medical Oncology Unit, Austin Health, Melbourne, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
| | - Andreas Behren
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia.,La Trobe University School of Cancer Medicine, Melbourne, VIC, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
45
|
Bolstad AI, Sehjpal P, Lie SA, Fevang BTS. Periodontitis in patients with systemic lupus erythematosus: A nation-wide study of 1990 patients. J Periodontol 2021; 93:364-372. [PMID: 34076274 DOI: 10.1002/jper.21-0181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/16/2021] [Accepted: 05/27/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND The aim of this study was to examine the association between systemic lupus erythematosus (SLE) and periodontitis in Norway during a 10-year period from 2008 through 2017. METHODS In this population-based study, 1990 patients were included in the SLE-cohort based on diagnostic codes registered in the Norwegian Patient Registry. The control group (n = 170,332) comprised patients registered with diagnostic codes for non-osteoporotic fractures or hip or knee replacement because of osteoarthritis. The outcome was periodontitis, defined by procedure codes registered in the Control and Payment of Health Refunds database. Logistic regression analyses were performed to estimate odds ratio for periodontitis in patients versus controls adjusted for potential covariates. RESULTS Periodontitis was significantly more common in SLE patients compared to controls (OR 1.78, 95% CI 1.47-2.14) and the difference was highest in SLE-patients 20 to 30 years of age (OR 3.24, 95% CI 1.23 - 8.52). The periodontitis rate in SLE patients was in the same range as for patients with diabetes mellitus type 2. CONCLUSIONS Patients with SLE had an almost doubled risk of periodontitis compared with the control population, and the difference was most accentuated in the young patients. These findings warrant an increased focus on dental health in SLE-patients.
Collapse
Affiliation(s)
- Anne Isine Bolstad
- Department of Clinical Dentistry, The Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Pria Sehjpal
- Department of Clinical Science, The Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Stein Atle Lie
- Department of Clinical Dentistry, The Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Bjørg-Tilde Svanes Fevang
- Department of Clinical Science, The Faculty of Medicine, University of Bergen, Bergen, Norway.,Department of Rheumatology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
46
|
Yoon JH, Shin MH, Kim HN, Choi W, Park JY, Hong AR, Kim HK, Kang HC. Programmed Cell Death-Ligand 1 (PD-L1) gene Single Nucleotide Polymorphism in Graves' Disease and Hashimoto's Thyroiditis in Korean Patients. Endocrinol Metab (Seoul) 2021; 36:599-606. [PMID: 34074094 PMCID: PMC8258335 DOI: 10.3803/enm.2021.965] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/01/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Programmed cell death-ligand 1 (PD-L1) has an important role in regulating immune reactions by binding to programmed death 1 (PD-1) on immune cells, which could prevent the exacerbation of autoimmune thyroid disease (AITD). The aim of this study was to evaluate the association of PD-L1 polymorphism with AITD, including Graves' disease (GD) and Hashimoto's thyroiditis (HT). METHODS A total of 189 GD patients, 234 HT patients, and 846 healthy age- and sex-matched controls were enrolled in this study. We analyzed PD-L1 single nucleotide polymorphism (SNP) (rs822339) and investigated the associations with clinical disease course and outcome. RESULTS Genotype frequency at the PD-L1 marker RS822339 in GD (P=0.219) and HT (P=0.764) patients did not differ from that among healthy controls. In patients with GD, the A/G or G/G genotype group demonstrated higher TBII titer (20.6±20.5 vs. 28.0± 25.8, P=0.044) and longer treatment duration (39.0±40.4 months vs. 62.4±65.0 months, P=0.003) compared to the A/A genotype group. Among patients in whom anti-thyroid peroxidase (TPO) antibody was measured after treatment of GD, post-treatment antiTPO positivity was higher in the A/G or G/G genotype group compared to the A/A genotype group (48.1% vs. 69.9%, P=0.045). Among patients with HT, there was no significant difference of anti-TPO antibody positivity (79.4% vs. 68.6%, P=0.121), anti-thyroglobulin antibody positivity (80.9% vs. 84.7%, P=0.661), or development to overt hypothyroidism (68.0% vs. 71.1%, P=0.632) between the A/A genotype group and the A/G or G/G genotype group. CONCLUSION The genotype frequency of PD-L1 (rs822339) is not different in patients with AITD compared with healthy controls. The intact PD-1/PD-L1 pathway in GD and HT might be important to maintain chronicity of AITD by protecting immune tolerance. However, the PD-L1 SNP could be associated with difficulty in achieving remission in patients with GD, which may be helpful to predict the possibility of longer treatment. Further studies are required to investigate the complex immune tolerance system in patients with AITD.
Collapse
Affiliation(s)
- Jee Hee Yoon
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun,
Korea
| | - Min-ho Shin
- Department of Preventive Medicine, Chonnam National University Medical School, Hwasun,
Korea
| | - Hee Nam Kim
- Department of Preventive Medicine, Chonnam National University Medical School, Hwasun,
Korea
| | - Wonsuk Choi
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun,
Korea
| | - Ji Yong Park
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun,
Korea
| | - A Ram Hong
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun,
Korea
| | - Hee Kyung Kim
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun,
Korea
| | - Ho-Cheol Kang
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun,
Korea
| |
Collapse
|
47
|
Single nucleotide polymorphisms in the PD1 gene with susceptibility to breast cancer in women. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
48
|
Hua S, Fan B, Mao W, Xu R, Wang Y, Kuai L, Ding X, Li B, Chen J, Miao X. Association between PDCD1 gene polymorphisms and psoriasis susceptibility in the Chinese population. Int J Dermatol 2021; 60:1411-1417. [PMID: 34057203 PMCID: PMC8596410 DOI: 10.1111/ijd.15665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/31/2021] [Accepted: 04/21/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND As an immune regulator expressed on the surface of activated T cells, programmed cell death 1 (PDCD1) plays an important role in psoriasis. However, whether PDCD1 genetic polymorphism is associated with psoriasis has yet to be explored. OBJECTIVE To study the association between polymorphisms of the immune-related gene PDCD1 and psoriasis susceptibility in the Chinese population, to illustrate the genetic mechanism of psoriasis and provide new research ideas for the diagnosis and treatment of psoriasis (PS). METHODS Overall, 128 psoriasis patients and 88 healthy controls were included in this study. Using polymerase chain reaction (PCR)-Sanger sequencing analysis, six PDCD1 single nucleotide polymorphisms (SNPs) were sequenced: PD1.1, PD1.3, PD1.4, PD1.5, PD1.6, and PD1.9. RESULTS Among the six tested SNPs, PD1.6 showed a significant association with psoriasis in genotype and allele frequency distribution. The G allele of PD1.6 increased the risk of psoriasis (P = 0.03). In contrast, the other five SNPs failed to show association with psoriasis. Further analysis within the patient group showed that the frequency of the PD1.6 G allele was relatively high in severe psoriasis, but the difference was nonsignificant. CONCLUSION PDCD1 gene polymorphism is associated with psoriasis. The population carrying PD1.6 allele G are at a higher risk of developing psoriasis, though the severity of psoriasis does not correlate with PD1.6 polymorphism.
Collapse
Affiliation(s)
- Shengyuan Hua
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bin Fan
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiwei Mao
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Xu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifei Wang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojie Ding
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Chen
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao Miao
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
49
|
Baba H, Kettani A, Bouqdayr M, Ouladlahsen A, Bensghir R, Marih L, Sodqi M, Benjelloun S, Ezzikouri S, Zaidane I, Jadid FZ, Filali KME, Wakrim L. Programmed cell death-1 single-nucleotide polymorphism rs10204525 is associated with human immunodeficiency virus type 1 RNA viral load in HIV-1-infected Moroccan subjects. Med Microbiol Immunol 2021; 210:187-196. [PMID: 34031715 DOI: 10.1007/s00430-021-00712-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 05/15/2021] [Indexed: 10/21/2022]
Abstract
Human Immunodeficiency Virus (HIV-1) infections are characterized by dysfunctional cellular and humoral antiviral immune responses. The progressive loss of effector functions in chronic viral infection has been associated with the up-regulation of programmed death-1 (PD-1), a negative regulator of activated T cells and Natural Killer cells. In HIV-1 infection, increased levels of PD-1 expression correlate with CD8 + T-cell exhaustion. In vitro, PD-1 blockade using PD-1 antibodies led to an increase in HIV-1 specific CD8 + T and memory B cell proliferation. We aimed to investigate the impact of PDCD1 rs10204525 polymorphism on HIV-1 susceptibility, AIDS development, and treatment response outcomes in HIV-1 infection in a Moroccan population. A total of 214 HIV-1 seropositive and 250 seronegative subjects were enrolled to investigate the association between the between the single-nucleotide polymorphism (SNP) rs10204525 of PDCD1 gene and HIV-1 pathogenesis using a predesigned TaqMan SNP genotyping assay. No significant association was found between rs10204525 and susceptibility to HIV-1 infection and AIDS development (p > 0.05). Genotype frequencies were significantly associated with the viral load before ART (p = 0.0105). HIV-1 viral load was significantly higher among subjects with the CC compared to TT genotype (p = 0.0043). In treated subjects, the median of viral load levels was significantly higher in CC and CT groups than TT subjects (p < 0.005). However, analysis of the correlation between CD4 + T-cell levels and PDCD1 polymorphism before and after ART showed no significant difference (p > 0.05). Our results demonstrated that rs10204525 polymorphism does not affect HIV-1 infection. However, this polymorphism may affect the response to treatment as measured by RNA viral load levels.
Collapse
Affiliation(s)
- Hanâ Baba
- Virology Unit, Immuno-Virology Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco.
- Laboratory of Biology and Health, URAC 34, Faculty of Sciences Ben M'Sik, Hassan II University-Casablanca, Casablanca, Morocco.
| | - Anass Kettani
- Laboratory of Biology and Health, URAC 34, Faculty of Sciences Ben M'Sik, Hassan II University-Casablanca, Casablanca, Morocco
| | - Meryem Bouqdayr
- Virology Unit, Immuno-Virology Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
- Laboratory of Biology and Health, URAC 34, Faculty of Sciences Ben M'Sik, Hassan II University-Casablanca, Casablanca, Morocco
| | - Ahd Ouladlahsen
- Service Des Maladies Infectieuses, CHU Ibn Rochd, Casablanca, Morocco
| | - Rajaa Bensghir
- Service Des Maladies Infectieuses, CHU Ibn Rochd, Casablanca, Morocco
| | - Latifa Marih
- Service Des Maladies Infectieuses, CHU Ibn Rochd, Casablanca, Morocco
| | - Mustapha Sodqi
- Service Des Maladies Infectieuses, CHU Ibn Rochd, Casablanca, Morocco
| | - Soumaya Benjelloun
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Sayeh Ezzikouri
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Imane Zaidane
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Fatima-Zahra Jadid
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | | | - Lahcen Wakrim
- Virology Unit, Immuno-Virology Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| |
Collapse
|
50
|
Yousefzadeh Y, Soltani-Zangbar MS, Hemmatzadeh M, Shomali N, Mahmoodpoor A, Ahmadian Heris J, Yousefi M. Fetomaternal Immune Tolerance: Crucial Mechanisms of Tolerance for Successful Pregnancy in Humans. Immunol Invest 2021; 51:1108-1125. [PMID: 33830854 DOI: 10.1080/08820139.2021.1909061] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
For many years, the question of how the maternal immune system tolerates the foreign fetus has remained unanswered, and numerous studies have considerably attempted to elucidate underlying mechanisms for fetomaternal tolerance. This review aimed at discussing various significant mechanisms in fetomaternal compatibility. At the fetomaternal interface, in addition to having efficient control against infections, innate and adaptive maternal immune systems selectively prevent fetal rejection. In general, understanding the complex mechanisms of fetomaternal tolerance is critical for immunologic tolerance induction and spontaneous abortion prevention in high-risk populations. Different cells and molecules, such as regulatory T-cells, dendritic cells, decidua cells, IDO, Class I HLA molecules, TGF-β, and IL-10, induce maternal immune tolerance in the fetus in numerous ways. The findings on fetomaternal immune tolerance have remained controversial and require further research.
Collapse
Affiliation(s)
- Yousef Yousefzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Committee Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Sadegh Soltani-Zangbar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hemmatzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Navid Shomali
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Ahmadian Heris
- Department of Allergy and Clinical Immunology, Pediatric Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|