1
|
Zhao Y, Zhang H, Wang W, Shen G, Wang M, Liu Z, Zhao J, Li J. The immune-related gene CD5 is a prognostic biomarker associated with the tumor microenvironment of breast cancer. Discov Oncol 2025; 16:39. [PMID: 39804513 PMCID: PMC11729608 DOI: 10.1007/s12672-024-01616-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Abstract
The occurrence and progression of breast cancer (BCa) are complex processes involving multiple factors and multiple steps. The tumor microenvironment (TME) plays an important role in this process, but the functions of immune components and stromal components in the TME require further elucidation. In this study, we obtained the RNA-seq data of 1086 patients from The Cancer Genome Atlas (TCGA) database. We calculated the proportions of tumor-infiltrating immune cells (TICs) and immune and stromal components using the CIBERSORT and ESTIMATE methods, and we screened differentially expressed genes (DEGs). Univariate Cox regression analysis of overall survival was performed on the DEGs, and a protein-protein interaction network of their protein products was generated. Finally, the hub gene CD5 was obtained. High CD5 expression was found to be associated with longer survival than low expression. Gene set enrichment analysis showed that DEGs upregulated in the high-CD5 expression group were mainly enriched in tumor- and immune-related pathways, while those upregulated in the low-expression group were enriched in protein export and lipid synthesis. TIC analysis showed that CD5 expression was positively correlated with the infiltration of CD8+ T cells, activated memory CD4+ T cells, gamma delta T cells, and M1 macrophages and negatively correlated with the infiltration of M2 macrophages. CD5 can increase anticancer immune cell infiltration and reduce M2 macrophage infiltration. These results suggest that CD5 is likely a potential prognostic biomarker and therapeutic target, providing novel insights into the treatment and prognostic assessment of BCa.
Collapse
Affiliation(s)
- Yi Zhao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, Qinghai, China
| | - Hengheng Zhang
- Graduate School of Qinghai University, Xining, 810000, Qinghai Province, People's Republic of China
| | - Wenwen Wang
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Guoshuang Shen
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, Qinghai, China
| | - Miaozhou Wang
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, Qinghai, China
| | - Zhen Liu
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, Qinghai, China
| | - Jiuda Zhao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, Qinghai, China
| | - Jinming Li
- Graduate School of Qinghai University, Xining, 810000, Qinghai Province, People's Republic of China.
| |
Collapse
|
2
|
Tian S, Xu M, Geng X, Fang J, Xu H, Xue X, Hu H, Zhang Q, Yu D, Guo M, Zhang H, Lu J, Guo C, Wang Q, Liu S, Zhang W. Network Medicine-Based Strategy Identifies Maprotiline as a Repurposable Drug by Inhibiting PD-L1 Expression via Targeting SPOP in Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410285. [PMID: 39499771 PMCID: PMC11714211 DOI: 10.1002/advs.202410285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/21/2024] [Indexed: 11/07/2024]
Abstract
Immune checkpoint inhibitors (ICIs) are drugs that inhibit immune checkpoint (ICP) molecules to restore the antitumor activity of immune cells and eliminate tumor cells. Due to the limitations and certain side effects of current ICIs, such as programmed death protein-1, programmed cell death-ligand 1, and cytotoxic T lymphocyte-associated antigen 4 (CTLA4) antibodies, there is an urgent need to find new drugs with ICP inhibitory effects. In this study, a network-based computational framework called multi-network algorithm-driven drug repositioning targeting ICP (Mnet-DRI) is developed to accurately repurpose novel ICIs from ≈3000 Food and Drug Administration-approved or investigational drugs. By applying Mnet-DRI to PD-L1, maprotiline (MAP), an antidepressant drug is repurposed, as a potential PD-L1 modifier for colorectal and lung cancers. Experimental validation revealed that MAP reduced PD-L1 expression by targeting E3 ubiquitin ligase speckle-type zinc finger structural protein (SPOP), and the combination of MAP and anti-CTLA4 in vivo significantly enhanced the antitumor effect, providing a new alternative for the clinical treatment of colorectal and lung cancer.
Collapse
Affiliation(s)
- Saisai Tian
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
| | - Mengting Xu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Xiangxin Geng
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Jiansong Fang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Hanchen Xu
- Institute of Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineShanghai200032China
| | - Xinying Xue
- Department of Respiratory and Critical CareEmergency and Critical Care Medical CenterBeijing Shijitan HospitalCapital Medical UniversityBeijing100038China
| | - Hongmei Hu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Qing Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Dianping Yu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Mengmeng Guo
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Hongwei Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Jinyuan Lu
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
| | - Chengyang Guo
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
| | - Qun Wang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Weidong Zhang
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di HerbsInstitute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100193China
- The Research Center for Traditional Chinese MedicineShanghai Institute of Infectious Diseases and BiosafetyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| |
Collapse
|
3
|
Elmetwalli A, El-Sewedy T, Hassan MG, Abdel-Monem MO, Hassan J, Ismail NF, Salama AF, Fu J, Mousa N, Sabir DK, El-Emam O, Hamdy G, El-Far AH. Gold nanoparticles mediate suppression of angiogenesis and breast cancer growth via MMP-9/NF-κB/mTOR and PD-L1/PD-1 signaling: integrative in vitro validation and network pharmacology insights. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03682-8. [PMID: 39718609 DOI: 10.1007/s00210-024-03682-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024]
Abstract
Gold nanoparticles (AuNPs) have emerged as promising candidates for cancer therapy due to their unique physicochemical properties and biocompatibility. In this study, we investigate the synthesis, characterization, and therapeutic potential of AuNPs in breast cancer treatment. Further, it establishes a comprehensive understanding of the mechanisms by which AuNPs suppress angiogenesis and breast cancer growth, identifying novel targets and signaling nodes contributing to the anti-tumor effects of AuNPs. AuNPs were synthesized and characterized using UV-Vis, crystallography, transmission electron microscopy (TEM), and energy-dispersive X-ray spectroscopy (EDX). The cytotoxicity of AuNPs was evaluated in WI-38 normal cells and MCF-7 breast cancer cells using the MTT assay. Additionally, the antioxidant activity of AuNPs was assessed through free radical scavenging and lipid peroxidation inhibition assays. Gene expression and pathway enrichment analyses were performed to elucidate the molecular mechanisms underlying the therapeutic effects of AuNPs in breast cancer. UV-Vis spectroscopy confirmed the successful synthesis of AuNPs, with a strong peak observed at 488.9 nm. Crystallography and TEM analysis revealed the crystalline nature and uniform size distribution of AuNPs, respectively. AuNPs exhibited concentration-dependent cytotoxic effects on MCF-7 cells, significantly inhibiting cancer cell proliferation at lower concentrations. Moreover, AuNPs demonstrated potent antioxidant activity, surpassing the effectiveness of vitamin C in scavenging free radicals and inhibiting lipid peroxidation. Gene expression analysis revealed modulation of crucial cancer-related genes and signaling pathways, including MMP-9/NF-κB/mTOR, PD-L1 expression and PD-1 checkpoint pathway, TNF signaling pathway, and adipocytokine signaling pathway, suggesting their potential as novel therapeutics for breast cancer treatment. Our findings support the promising role of AuNPs as effective and targeted therapeutics for breast cancer treatment. Further research is warranted to elucidate the precise mechanisms of action and evaluate the clinical efficacy and safety of AuNP-based therapies in breast cancer patients.
Collapse
Affiliation(s)
- Alaa Elmetwalli
- Department of Clinical Trial Research Unit and Drug Discovery, Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt.
- Higher Technological Institute of Applied Health Sciences, Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt.
| | - Tarek El-Sewedy
- Department of Applied Medical Chemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Mervat G Hassan
- Department of Botany and Microbiology, Faculty of Science, Benha University, Benha, 33516, Egypt
| | - Mohamed O Abdel-Monem
- Department of Botany and Microbiology, Faculty of Science, Benha University, Benha, 33516, Egypt
| | - Jihan Hassan
- Department of Applied Medical Chemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Nadia F Ismail
- Health Information Management Program, Biochemistry, Faculty of Health Science Technology, Borg El Arab Technological University, Alexandria, Egypt
| | - Afrah Fatthi Salama
- Biochemistry Section, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Nasser Mousa
- Tropical Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Deema Kamal Sabir
- Department of Medical Surgical Nursing, College of Nursing, Princess Nourah bint Abdulrahman University, P.O.Box 84428, 11671, Riyadh, Saudi Arabia
| | - Ola El-Emam
- Clinical Pathology Department, Mansoura University, Mansoura, Egypt
| | - Ghada Hamdy
- Higher Technological Institute of Applied Health Sciences, Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt
| | - Ali H El-Far
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, China
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt
| |
Collapse
|
4
|
Montoyo-Pujol YG, Ponce JJ, Delgado-García S, Martín TA, Ballester H, Castellón-Molla E, Ramos-Montoya A, Lozano-Cubo I, Sempere-Ortells JM, Peiró G. High CTLA-4 gene expression is an independent good prognosis factor in breast cancer patients, especially in the HER2-enriched subtype. Cancer Cell Int 2024; 24:371. [PMID: 39523362 PMCID: PMC11552348 DOI: 10.1186/s12935-024-03554-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is the most common cancer in women and the leading cause of cancer-related death worldwide. This heterogeneous disease has been historically considered a non-immunogenic type of cancer. However, recent advances in immunotherapy have increased the interest in knowing the role of the immune checkpoints (IC) and other immune regulation pathways in this neoplasia. METHODS In this retrospective study, we evaluated the correlation of mRNA expression of CTLA-4, PDCD1 (PD1), CD274 (PD-L1), PDCD1LG2 (PD-L2), CD276 (B7-H3), JAK2, and FOXO1 with clinicopathological factors and BC patient's outcome by real-time quantitative polymerase chain reaction (qPCR). RESULTS Our results showed that immunoregulatory gene expression depends on BC immunophenotype being CTLA-4 and PDCD1 (PD1) overexpressed on triple-negative/basal-like (TN/BL) and luminal B/HER2-positive phenotypes, respectively, and CD276 (B7-H3), JAK2 and FOXO1 associated with both luminal A and luminal B/HER2-negative tumors. In addition, we found that these genes can also be related to aggressive and non-aggressive clinicopathological characteristics in BC. Finally, survival analysis showed that CTLA-4 expression levels emerge as a significant independent factor of good prognosis in BC patients, especially in the HER2-enriched subtype. CONCLUSION Considering all these data, we can conclude that the expression of immunoregulatory genes depends on tumor phenotype and has potential clinical implications in BC patients.
Collapse
Affiliation(s)
- Yoel G Montoyo-Pujol
- Research Unit, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain.
- Medical Oncology Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain.
| | - José J Ponce
- Medical Oncology Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - Silvia Delgado-García
- Gynecology and Obstetrics Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - Tina A Martín
- Gynecology and Obstetrics Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - Hortensia Ballester
- Gynecology and Obstetrics Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - Elena Castellón-Molla
- Pathology Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - Angela Ramos-Montoya
- Gynecology and Obstetrics Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - Inmaculada Lozano-Cubo
- Medical Oncology Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - J Miguel Sempere-Ortells
- Research Unit, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
- Biotechnology Department, Immunology Division, University of Alicante, Ctra San Vicente s/n. 03080-San Vicente del Raspeig, Alicante, 03010, Spain
| | - Gloria Peiró
- Research Unit, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain.
- Pathology Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain.
- Biotechnology Department, Immunology Division, University of Alicante, Ctra San Vicente s/n. 03080-San Vicente del Raspeig, Alicante, 03010, Spain.
| |
Collapse
|
5
|
Mao W, Yoo HS. Inorganic Nanoparticle Functionalization Strategies in Immunotherapeutic Applications. Biomater Res 2024; 28:0086. [PMID: 39323561 PMCID: PMC11423863 DOI: 10.34133/bmr.0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/20/2024] [Accepted: 09/05/2024] [Indexed: 09/27/2024] Open
Abstract
Nanotechnology has been increasingly utilized in anticancer treatment owing to its ability of engineering functional nanocarriers that enhance therapeutic effectiveness while minimizing adverse effects. Inorganic nanoparticles (INPs) are prevalent nanocarriers to be customized for a wide range of anticancer applications, including theranostics, imaging, targeted drug delivery, and therapeutics, because they are advantageous for their superior biocompatibility, unique optical properties, and capacity of being modified via versatile surface functionalization strategies. In the past decades, the high adaptation of INPs in this emerging immunotherapeutic field makes them good carrier options for tumor immunotherapy and combination immunotherapy. Tumor immunotherapy requires targeted delivery of immunomodulating therapeutics to tumor locations or immunological organs to provoke immune cells and induce tumor-specific immune response while regulating immune homeostasis, particularly switching the tumor immunosuppressive microenvironment. This review explores various INP designs and formulations, and their employment in tumor immunotherapy and combination immunotherapy. We also introduce detailed demonstrations of utilizing surface engineering tactics to create multifunctional INPs. The generated INPs demonstrate the abilities of stimulating and enhancing the immune response, specific targeting, and regulating cancer cells, immune cells, and their resident microenvironment, sometimes along with imaging and tracking capabilities, implying their potential in multitasking immunotherapy. Furthermore, we discuss the promises of INP-based combination immunotherapy in tumor treatments.
Collapse
Affiliation(s)
- Wei Mao
- Department of Biomedical Materials Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hyuk Sang Yoo
- Department of Biomedical Materials Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea
- Kangwon Radiation Convergence Research Center, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
6
|
Yang X, Li X, Xu H, Du S, Wang C, He H. Predicting CTLA4 expression and prognosis in clear cell renal cell carcinoma using a pathomics signature of histopathological images and machine learning. Heliyon 2024; 10:e34877. [PMID: 39145002 PMCID: PMC11320204 DOI: 10.1016/j.heliyon.2024.e34877] [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: 08/08/2023] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
Background CTLA4, an immune checkpoint, plays an important role in tumor immunotherapy. The purpose of this study was to develop a pathomics signature to evaluate CTLA4 expression and predict clinical outcomes in clear cell renal cell carcinoma (ccRCC) patients. Methods A total of 354 patients from the TCGA-KIRC dataset were enrolled in this study. The patients were stratified into two groups based on the level of CTLA4 expression, and overall survival rates were analyzed between groups. Pathological features were identified using machine learning algorithms, and a gradient boosting machine (GBM) was employed to construct the pathomics signatures for predicting prognosis and CTLA4 expression. The predictive performance of the model was subsequently assessed. Enrichment analysis was performed on diferentially expressed genes related to the pathomics score (PS). Additionally, correlations between PS and TMB, as well as immune infiltration profiles associated with different PS values, were explored. In vitro experiments, CTLA4 knockdown was performed to investigate its impact on cell proliferation, migration, invasion, TGF-β signaling pathway, and macrophage polarization. Results High expression of CTLA4 was associated with an unfavorable prognosis in ccRCC patients. The pathomics signature displayed good performance in the validation set (AUC = 0.737; P < 0.001 in the log-rank test). The PS was positively correlated with CTLA4 expression. We next explored the underlying mechanism and found the associations between the pathomics signature and TGF-β signaling pathways, TMB, and Tregs. Further in vitro experiments demonstrated that CTLA4 knockdown inhibited cell proliferation, migration, invasion, TGF-β expression, and macrophage M2 polarization. Conclusion High expression of CTLA4 was found to correlate with poor prognosis in ccRCC patients. The pathomics signature established by our group using machine learning effectively predicted both patient prognosis and CTLA4 expression levels in ccRCC cases.
Collapse
Affiliation(s)
- Xiaoqun Yang
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiangyun Li
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Haimin Xu
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Silin Du
- University Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Chaofu Wang
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongchao He
- Department of Urology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
7
|
Seki Y, Yamana K, Yoshida R, Inoue J, Shinohara K, Oyama T, Kubo R, Nagata M, Kawahara K, Hirayama M, Takahashi N, Nakamoto M, Hirosue A, Kariya R, Okada S, Nakayama H. Programmed death-ligand 1-expressing extracellular vesicles are a prognostic factor in patients with oral squamous cell carcinoma treated with immune checkpoint inhibitors. JOURNAL OF ORAL AND MAXILLOFACIAL SURGERY, MEDICINE, AND PATHOLOGY 2024; 36:518-525. [DOI: 10.1016/j.ajoms.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
|
8
|
Xu C, Sun Z, Zhang X, Zang Q, Yang Z, Li L, Yang X, He Y, Ma Z, Chen J. Discovery of 4-phenyl-1H-indazole derivatives as novel small-molecule inhibitors targeting the PD-1/PD-L1 interaction. Bioorg Chem 2024; 147:107376. [PMID: 38640722 DOI: 10.1016/j.bioorg.2024.107376] [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: 01/29/2024] [Revised: 03/30/2024] [Accepted: 04/14/2024] [Indexed: 04/21/2024]
Abstract
The inhibition of the programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) pathway with small molecules is a promising approach for cancer immunotherapy. Herein, novel small molecules compounds bearing various scaffolds including thiophene, thiazole, tetrahydroquinoline, benzimidazole and indazole were designed, synthesized and evaluated for their inhibitory activity against the PD-1/PD-L1 interaction. Among them, compound Z13 exhibited the most potent activity with IC50 of 189.6 nM in the homogeneous time-resolved fluorescence (HTRF) binding assay. Surface plasmon resonance (SPR) assay demonstrated that Z13 bound to PD-L1 with high affinity (KD values of 231 nM and 311 nM for hPD-L1 and mPD-L1, respectively). In the HepG2/Jurkat T co-culture cell model, Z13 decreased the viability rate of HepG2 cells in a concentration-dependent manner. In addition, Z13 showed significant in vivo antitumor efficacy (TGI = 52.6 % at 40 mg/kg) without obvious toxicity in the B16-F10 melanoma model. Furthermore, flow cytometry analysis demonstrated that Z13 inhibited tumor growth in vivo by activating the tumor immune microenvironment. These findings indicate that Z13 is a promising PD-1/PD-L1 inhibitor deserving further investigation.
Collapse
Affiliation(s)
- Chenglong Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhiqiang Sun
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xuewen Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qinru Zang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zichao Yang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ling Li
- The Eighth Affiliated Hospital, Sun Yat sen University, Shenzhen 518033, China
| | - Xixiang Yang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yueyu He
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zeli Ma
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
9
|
Kheraldine H, Gupta I, Cyprian FS, Vranic S, Al-Farsi HF, Merhi M, Dermime S, Al Moustafa AE. Targeting HER2-positive breast cancer cells by a combination of dasatinib and BMS-202: Insight into the molecular pathways. Cancer Cell Int 2024; 24:94. [PMID: 38431613 PMCID: PMC10909263 DOI: 10.1186/s12935-023-03195-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 12/26/2023] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Recent investigations have reported the benefits of using a tyrosine kinase inhibitor, dasatinib (DA), as well as programmed death-ligand 1 (PD-L1) inhibitors in the management of several solid tumors, including breast cancer. Nevertheless, the outcome of the combination of these inhibitors on HER2-positive breast cancer is not explored yet. METHODS Herein, we investigated the impact of DA and PD-L1 inhibitor (BMS-202) combination on HER2-positive breast cancer cell lines, SKBR3 and ZR75. RESULTS Our data reveal that the combination significantly inhibits cell viability of both cancer cell lines as compared to monotreatment. Moreover, the combination inhibits epithelial-mesenchymal transition (EMT) progression and reduces cancer cell invasion by restoring E-cadherin and β-catenin expressions and loss of vimentin, major biomarkers of EMT. Additionally, the combination reduces the colony formation of both cell lines in comparison with their matched control. Also, the combination considerably inhibits the angiogenesis of the chorioallantoic membrane model compared with monotreatment. Molecular pathway analysis of treated cells shows that this combination blocks HER2, AKT, β-catenin, and JNK1/2/3 activities. CONCLUSION Our findings implicate that a combination of DA and BMS-202 could have a significant impact on the management of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Hadeel Kheraldine
- College of Medicine, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
- Biomedical Research Centre, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Ishita Gupta
- College of Medicine, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
- Sidra Medicine, Doha, Qatar
| | - Farhan Sachal Cyprian
- College of Medicine, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
- Biomedical Research Centre, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Halema F Al-Farsi
- College of Medicine, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Maysaloun Merhi
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
- Translational Cancer Research Facility, Interim Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Said Dermime
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
- Translational Cancer Research Facility, Interim Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar.
- Biomedical Research Centre, Qatar University, P. O. Box 2713, Doha, Qatar.
- Oncology Department, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
10
|
Croft D, Lodhia P, Lourenco S, MacKay C. Effectively utilizing publicly available databases for cancer target evaluation. NAR Cancer 2023; 5:zcad035. [PMID: 37457379 PMCID: PMC10346432 DOI: 10.1093/narcan/zcad035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/12/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
The majority of compounds designed against cancer drug targets do not progress to become approved drugs, mainly due to lack of efficacy and/or unmanageable toxicity. Robust target evaluation is therefore required before progressing through the drug discovery process to reduce the high attrition rate. There are a wealth of publicly available databases that can be mined to generate data as part of a target evaluation. It can, however, be challenging to learn what databases are available, how and when they should be used, and to understand the associated limitations. Here, we have compiled and present key, freely accessible and easy-to-use databases that house informative datasets from in vitro, in vivo and clinical studies. We also highlight comprehensive target review databases that aim to bring together information from multiple sources into one-stop portals. In the post-genomics era, a key objective is to exploit the extensive cell, animal and patient characterization datasets in order to deliver precision medicine on a patient-specific basis. Effective utilization of the highlighted databases will go some way towards supporting the cancer research community achieve these aims.
Collapse
Affiliation(s)
- Daniel Croft
- Cancer Research Horizons, The Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
| | - Puja Lodhia
- Cancer Research Horizons, The Francis Crick Institute, London, NW1 1AT, UK
| | - Sofia Lourenco
- Cancer Research Horizons, The Francis Crick Institute, London, NW1 1AT, UK
| | - Craig MacKay
- Cancer Research Horizons, The Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
| |
Collapse
|
11
|
Huang PL, Kan HT, Hsu CH, Hsieh HT, Cheng WC, Huang RY, You JJ. A bispecific antibody AP203 targeting PD-L1 and CD137 exerts potent antitumor activity without toxicity. J Transl Med 2023; 21:346. [PMID: 37226226 PMCID: PMC10210478 DOI: 10.1186/s12967-023-04193-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Bispecific antibody has garnered considerable attention in the recent years due to its impressive preliminary efficacy in hematological malignancies. For solid tumors, however, the main hindrance is the suppressive tumor microenvironment, which effectively impedes the activation of infiltrating T cells. Herein, we designed a bispecific antibody AP203 with high binding affinity to PD-L1 and CD137 and assessed its safety and anti-tumor efficacy, as well as explored the mechanism of action. METHODS The optimal antibody binders against PD-L1 and CD137 were screened from the OmniMab phagemid library. The binding affinity of the constructed AP203 were evaluated using enzyme-linked immunosorbent assay (ELISA) and biolayer interferometry (BLI). T-cell stimulatory capacity was assessed using the allogeneic mixed lymphocyte reaction (MLR), antigen-specific recall response, and coculture with PD-L1-expressing cells. In vivo antitumor efficacy was evaluated using two models of tumor-xenografted humanized mice with profiling of tumor infiltrating lymphocytes (TILs). The possible toxicity of AP203 was examined using in vitro cytokine release assay by human PBMCs. RESULTS AP203, which simultaneously targeted PD-L1 and costimulatory CD137, elicit superior agonistic effects over parental antibodies alone or in combination in terms of T cell activation, enhanced memory recall responses, and overcoming Treg-mediated immunosuppression (P < 0.05). The agonistic activity of AP203 was further demonstrated PD-L1-dependent by coculturing T cells with PD-L1-expressing cells. In vivo animal studies using immunodeficient or immunocompetent mice both showed a dose-related antitumor efficacy superior to parental antibodies in combination (P < 0.05). Correspondingly, AP203 significantly increased tumor infiltrating CD8 + T cells, while decreased CD4 + T cells, as well as Treg cells (P < 0.05), resulting in a dose-dependent increase in the CD8 + /CD4 + ratio. Moreover, either soluble or immobilized AP203 did not induce the production of inflammatory cytokines by human PBMCs. CONCLUSIONS AP203 exerts potent antitumor activity not only by blocking PD-1/PD-L1 inhibitory signaling, but also by activating CD137 costimulatory signaling in effector T cells that consequently counteracts Treg-mediated immunosuppression. Based on promising preclinical results, AP203 should be a suitable candidate for clinical treatment of solid tumors.
Collapse
Affiliation(s)
- Po-Lin Huang
- AP Biosciences, Inc., 17F., No. 3, Yuanqu St., Nangang Dist., Taipei, 115603, Taiwan.
| | - Hung-Tsai Kan
- AP Biosciences, Inc., 17F., No. 3, Yuanqu St., Nangang Dist., Taipei, 115603, Taiwan
| | - Ching-Hsuan Hsu
- AP Biosciences, Inc., 17F., No. 3, Yuanqu St., Nangang Dist., Taipei, 115603, Taiwan
| | - Hsin-Ta Hsieh
- AP Biosciences, Inc., 17F., No. 3, Yuanqu St., Nangang Dist., Taipei, 115603, Taiwan
| | - Wan-Chien Cheng
- Department of Periodontology, School of Dentistry, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Ren-Yeong Huang
- Department of Periodontology, School of Dentistry, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Jhong-Jhe You
- AP Biosciences, Inc., 17F., No. 3, Yuanqu St., Nangang Dist., Taipei, 115603, Taiwan.
| |
Collapse
|
12
|
Ramírez-González A, Ávila-López P, Bahena-Román M, Contreras-Ochoa CO, Lagunas-Martínez A, Langley E, Manzo-Merino J, Madrid-Marina V, Torres-Poveda K. Critical Role of the Transcription Factor AKNA in T-Cell Activation: An Integrative Bioinformatics Approach. Int J Mol Sci 2023; 24:ijms24044212. [PMID: 36835622 PMCID: PMC9965657 DOI: 10.3390/ijms24044212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
The human akna gene encodes an AT-hook transcription factor, the expression of which is involved in various cellular processes. The goal of this study was to identify potential AKNA binding sites in genes that participate in T-cell activation and validate selected genes. Here we analyzed ChIP-seq and microarray assays to determine AKNA-binding motifs and the cellular process altered by AKNA in T-cell lymphocytes. In addition, we performed a validation analysis by RT-qPCR to assess AKNA's role in promoting IL-2 and CD80 expression. We found five AT-rich motifs that are potential candidates as AKNA response elements. We identified these AT-rich motifs in promoter regions of more than a thousand genes in activated T-cells, and demonstrated that AKNA induces the expression of genes involved in helper T-cell activation, such as IL-2. The genomic enrichment and prediction of AT-rich motif analyses demonstrated that AKNA is a transcription factor that can potentially modulate gene expression by recognizing AT-rich motifs in a plethora of genes that are involved in different molecular pathways and processes. Among the cellular processes activated by AT-rich genes, we found inflammatory pathways potentially regulated by AKNA, suggesting AKNA is acting as a master regulator during T-cell activation.
Collapse
Affiliation(s)
- Abrahan Ramírez-González
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico
| | - Pedro Ávila-López
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Margarita Bahena-Román
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico
| | - Carla O. Contreras-Ochoa
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico
| | - Alfredo Lagunas-Martínez
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico
| | - Elizabeth Langley
- Department of Basic Research, Instituto Nacional de Cancerología, Mexico City 14080, Mexico
| | - Joaquín Manzo-Merino
- Department of Basic Research, Instituto Nacional de Cancerología, Mexico City 14080, Mexico
- Consejo Nacional de Ciencia y Tecnología (CONACyT), Instituto Nacional de Cancerología, Mexico City 03940, Mexico
| | - Vicente Madrid-Marina
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico
| | - Kirvis Torres-Poveda
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico
- Consejo Nacional de Ciencia y Tecnología (CONACyT), Instituto Nacional de Salud Pública, Cuernavaca 03940, Mexico
- Correspondence: ; Tel.:+52-777-3293000 (ext. 2204)
| |
Collapse
|
13
|
Schafer JB, Lucas ED, Dzieciatkowska M, Forward T, Tamburini BAJ. Programmed death ligand 1 intracellular interactions with STAT3 and focal adhesion protein Paxillin facilitate lymphatic endothelial cell remodeling. J Biol Chem 2022; 298:102694. [PMID: 36375639 PMCID: PMC9761386 DOI: 10.1016/j.jbc.2022.102694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/20/2022] [Accepted: 10/22/2022] [Indexed: 11/13/2022] Open
Abstract
Lymphatic endothelial cells (LECs) comprise lymphatic capillaries and vessels that guide immune cells to lymph nodes (LNs) and form the subcapsular sinus and cortical and medullary lymphatic structures of the LN. During an active immune response, the lymphatics remodel to accommodate the influx of immune cells from the tissue, but factors involved in remodeling are unclear. Here, we determined that a TSS motif within the cytoplasmic domain of programmed death ligand 1 (PD-L1), expressed by LECs in the LN, participates in lymphatic remodeling. Mutation of the TSS motif to AAA does not affect surface expression of PD-L1, but instead causes defects in LN cortical and medullary lymphatic organization following immunostimulant, Poly I:C, administration in vivo. Supporting this observation, in vitro treatment of the LEC cell line, SVEC4-10, with cytokines TNFα and IFNα significantly impeded SVEC4-10 movement in the presence of the TSS-AAA cytoplasmic mutation. The cellular movement defects coincided with reduced F-actin polymerization, consistent with differences previously found in dendritic cells. Here, in addition to loss of actin polymerization, we define STAT3 and Paxillin as important PD-L1 binding partners. STAT3 and Paxillin were previously demonstrated to be important at focal adhesions for cellular motility. We further demonstrate the PD-L1 TSS-AAA motif mutation reduced the amount of pSTAT3 and Paxillin bound to PD-L1 both before and after exposure to TNFα and IFNα. Together, these findings highlight PD-L1 as an important component of a membrane complex that is involved in cellular motility, which leads to defects in lymphatic organization.
Collapse
Affiliation(s)
- Johnathon B Schafer
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, Colorado, USA; Molecular Biology Graduate Program, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Erin D Lucas
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, Colorado, USA; Immunology Graduate Program, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Tadg Forward
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Beth A Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, Colorado, USA; Molecular Biology Graduate Program, University of Colorado School of Medicine, Aurora, Colorado, USA; Immunology Graduate Program, University of Colorado School of Medicine, Aurora, Colorado, USA; Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA.
| |
Collapse
|
14
|
Warren SI, Charville GW, Manasherob R, Amanatullah DF. Immune checkpoint upregulation in periprosthetic joint infection. J Orthop Res 2022; 40:2663-2669. [PMID: 35124851 PMCID: PMC9352818 DOI: 10.1002/jor.25276] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/06/2022] [Accepted: 01/16/2022] [Indexed: 02/04/2023]
Abstract
Periprosthetic joint infections (PJI) induce an immunosuppressive cytokine profile through an unknown mechanism. Immune checkpoints, like programmed cell death 1 (PD-1) and its ligand (PD-L1), initiate innate immunosuppressive pathways essential for self-tolerance. Several malignancies and chronic infections co-opt these pathways to derive a survival advantage. This study evaluates PD-1/PD-L1 expression in periprosthetic tissue from patients undergoing revision hip or knee arthroplasty for a PJI versus an aseptic failure. PD-1/PD-L1 in the global tissue sample and the high-power microscopic field of maximum expression was analyzed prospectively using immunohistochemistry. Fifteen patients with a PJI (45%) and 16 patients with an aseptic failure (52%) were included. PD-1 expression was uniformly low. Maximum PD-L1 expression was upregulated in patients with a PJI (25%, interquartile range [IQR]: 5%-75%) versus an aseptic failure, (8%, IQR: 1%-48%, p = 0.039). In the PJI cohort, maximum PD-L1 expression was higher among patients who developed a recurrent PJI (68%, IQR: 53%-86% vs. 15%, IQR: 5%-70%, p = 0.039). Patients with global PD-L1 over 5% trended toward a near 22-fold increase in the odds of reinfection (odds ratio [OR]: 21.9, 95% confidence interval [CI]: 0.9-523.5, p = 0.057) and patients with maximum PD-L1 over 20% trended toward a 15-fold increase in the odds of reinfection (OR: 15.0, 95% CI: 0.6-348.9, p = 0.092). These results support immune checkpoint upregulation as a mechanism of PJI-induced local immune dysfunction. Future studies should confirm PD-L1 as a risk factor for reinfection in larger cohorts.
Collapse
Affiliation(s)
- Shay I. Warren
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | | | - Robert Manasherob
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Derek F. Amanatullah
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| |
Collapse
|
15
|
Hu C, Jiang W, Lv M, Fan S, Lu Y, Wu Q, Pi J. Potentiality of Exosomal Proteins as Novel Cancer Biomarkers for Liquid Biopsy. Front Immunol 2022; 13:792046. [PMID: 35757760 PMCID: PMC9218252 DOI: 10.3389/fimmu.2022.792046] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
Liquid biopsy has been rapidly developed in recent years due to its advantages of non-invasiveness and real-time sampling in cancer prognosis and diagnosis. Exosomes are nanosized extracellular vesicles secreted by all types of cells and abundantly distributed in all types of body fluid, carrying diverse cargos including proteins, DNA, and RNA, which transmit regulatory signals to recipient cells. Among the cargos, exosomal proteins have always been used as immunoaffinity binding targets for exosome isolation. Increasing evidence about the function of tumor-derived exosomes and their proteins is found to be massively associated with tumor initiation, progression, and metastasis in recent years. Therefore, exosomal proteins and some nucleic acids, such as miRNA, can be used not only as targets for exosome isolation but also as potential diagnostic markers in cancer research, especially for liquid biopsy. This review will discuss the existing protein-based methods for exosome isolation and characterization that are more appropriate for clinical use based on current knowledge of the exosomal biogenesis and function. Additionally, the recent studies for the use of exosomal proteins as cancer biomarkers are also discussed and summarized, which might contribute to the development of exosomal proteins as novel diagnostic tools for liquid biopsy.
Collapse
Affiliation(s)
- Chunmiao Hu
- Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.,Department of Cellular Biology, Dakewe, Shenzhen, China
| | - Wei Jiang
- Department of Cellular Biology, Dakewe, Shenzhen, China
| | - Mingjin Lv
- Department of Cellular Biology, Dakewe, Shenzhen, China
| | - Shuhao Fan
- Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Yujia Lu
- Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Qingjun Wu
- Department of Cellular Biology, Dakewe, Shenzhen, China
| | - Jiang Pi
- Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| |
Collapse
|
16
|
Dum D, Henke TLC, Mandelkow T, Yang C, Bady E, Raedler JB, Simon R, Sauter G, Lennartz M, Büscheck F, Luebke AM, Menz A, Hinsch A, Höflmayer D, Weidemann S, Fraune C, Möller K, Lebok P, Uhlig R, Bernreuther C, Jacobsen F, Clauditz TS, Wilczak W, Minner S, Burandt E, Steurer S, Blessin NC. Semi-automated validation and quantification of CTLA-4 in 90 different tumor entities using multiple antibodies and artificial intelligence. J Transl Med 2022; 102:650-657. [PMID: 35091676 PMCID: PMC9162915 DOI: 10.1038/s41374-022-00728-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/17/2021] [Accepted: 12/17/2021] [Indexed: 11/26/2022] Open
Abstract
CTLA-4 is an inhibitory immune checkpoint receptor and a negative regulator of anti-tumor T-cell function. This study is aimed for a comparative analysis of CTLA-4+ cells between different tumor entities. To quantify CTLA-4+ cells, 4582 tumor samples from 90 different tumor entities as well as 608 samples of 76 different normal tissue types were analyzed by immunohistochemistry in a tissue microarray format. Two different antibody clones (MSVA-152R and CAL49) were validated and quantified using a deep learning framework for automated exclusion of unspecific immunostaining. Comparing both CTLA-4 antibodies revealed a clone dependent unspecific staining pattern in adrenal cortical adenoma (63%) for MSVA-152R and in pheochromocytoma (67%) as well as hepatocellular carcinoma (36%) for CAL49. After automated exclusion of non-specific staining reaction (3.6%), a strong correlation was observed for the densities of CTLA-4+ lymphocytes obtained by both antibodies (r = 0.87; p < 0.0001). A high CTLA-4+ cell density was linked to low pT category (p < 0.0001), absent lymph node metastases (p = 0.0354), and PD-L1 expression in tumor cells or inflammatory cells (p < 0.0001 each). A high CTLA-4/CD3-ratio was linked to absent lymph node metastases (p = 0.0295) and to PD-L1 positivity on immune cells (p = 0.0026). Marked differences exist in the number of CTLA-4+ lymphocytes between tumors. Analyzing two independent antibodies by a deep learning framework can facilitate automated quantification of immunohistochemically analyzed target proteins such as CTLA-4.
Collapse
Affiliation(s)
- David Dum
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tjark L C Henke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Mandelkow
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cheng Yang
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elena Bady
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonas B Raedler
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- College of Arts and Sciences, Boston University, Boston, MA, USA
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franziska Büscheck
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas M Luebke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Menz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Doris Höflmayer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sören Weidemann
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Möller
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ria Uhlig
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till S Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Waldemar Wilczak
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Niclas C Blessin
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
17
|
Chen L, Huang S, Liu Q, Kong X, Su Z, Zhu M, Fang Y, Zhang L, Li X, Wang J. PD-L1 Protein Expression Is Associated With Good Clinical Outcomes and Nomogram for Prediction of Disease Free Survival and Overall Survival in Breast Cancer Patients Received Neoadjuvant Chemotherapy. Front Immunol 2022; 13:849468. [PMID: 35669769 PMCID: PMC9163312 DOI: 10.3389/fimmu.2022.849468] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveThis study aims to investigate the potential prognostic significance of programmed death ligand-1 (PD-L1) protein expression in tumor cells of breast cancer patients received neoadjuvant chemotherapy (NACT).MethodsUsing semiquantitative immunohistochemistry, the PD-L1 protein expression in breast cancer tissues was analyzed. The correlations between PD-L1 protein expression and clinicopathologic characteristics were analyzed using Chi-square test or Fisher’s exact test. The survival curve was stemmed from Kaplan-Meier assay, and the log-rank test was used to compare survival distributions against individual index levels. Univariate and multivariate Cox proportional hazards regression models were accessed to analyze the associations between PD-L1 protein expression and survival outcomes. A predictive nomogram model was constructed in accordance with the results of multivariate Cox model. Calibration analyses and decision curve analyses (DCA) were performed for the calibration of the nomogram model, and subsequently adopted to assess the accuracy and benefits of the nomogram model.ResultsA total of 104 breast cancer patients received NACT were enrolled into this study. According to semiquantitative scoring for IHC, patients were divided into: low PD-L1 group (61 cases) and high PD-L1 group (43 cases). Patients with high PD-L1 protein expression were associated with longer disease free survival (DFS) (mean: 48.21 months vs. 31.16 months; P=0.011) and overall survival (OS) (mean: 83.18 months vs. 63.31 months; P=0.019) than those with low PD-L1 protein expression. Univariate and multivariate analyses indicated that PD-L1, duration of neoadjuvant therapy, E-Cadherin, targeted therapy were the independent prognostic factors for patients’ DFS and OS. Nomogram based on these independent prognostic factors was used to evaluate the DFS and OS time. The calibration plots shown PD-L1 based nomogram predictions were basically consistent with actual observations for assessments of 1-, 3-, and 5-year DFS and OS time. The DCA curves indicated the PD-L1 based nomogram had better predictive clinical applications regarding prognostic assessments of 3- and 5-year DFS and OS, respectively.ConclusionHigh PD-L1 protein expression was associated with significantly better prognoses and longer DFS and OS in breast cancer patients. Furthermore, PD-L1 protein expression was found to be a significant prognostic factor for patients who received NACT.
Collapse
Affiliation(s)
- Li Chen
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shaolong Huang
- Department of Thyroid and Breast, Burn and Plastic Surgery, Tongren City People’s Hospital, Tongren, China
| | - Qiang Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhaohui Su
- School of Public Health, Southeast University, Nanjing, China
| | - Mengliu Zhu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Zhang
- Melbourne School of Population and Global Health, The University of Melbourne, VIC, Australia
- Centre of Cancer Research, Victorian Comprehensive Cancer Centre, Melbourne, VIC, Australia
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Lin Zhang, ; Xingrui Li, ; Jing Wang,
| | - Xingrui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Lin Zhang, ; Xingrui Li, ; Jing Wang,
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Lin Zhang, ; Xingrui Li, ; Jing Wang,
| |
Collapse
|
18
|
Hamed MM, Gouida MS, Abd EL-Aziz SR, EL-Sokkary AM. Evaluation PD-L1, CD8 and CD20 as early predictor and tracking markers for breast cancer (BC) in Egypt. Heliyon 2022; 8:e09474. [PMID: 35647336 PMCID: PMC9136277 DOI: 10.1016/j.heliyon.2022.e09474] [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: 01/16/2022] [Revised: 02/20/2022] [Accepted: 05/13/2022] [Indexed: 11/15/2022] Open
Abstract
Background Breast cancer (BC) is considered as a common type of cancer threatening women throughout the world. Therefore, development of early predication biomarkers for BC got more concern especially for Egyptian females. This study was aimed to evaluate PD-L1, CD8, and CD20 as early prediction breast cancer biomarkers. Methods Flow cytometry (FC), immunohistochemistry (IHC), Western Blot, and q-PCR were used to compare PD-L1, CD20, and CD8 levels in tissues and blood samples of Breast Cancer and controls. Results Blood samples showed a significant increase in PD-L1, CD20, and CD8 compared to controls (p˂0.005). A Significant correlation was shown between PD-L1, CD8, and CD20 in tissue and breast cancer subtypes. Whereas, invasive lobular carcinoma (ILC) was characterized by superior PD-L1 and CD20 levels compared to invasive ductal carcinoma (IDC). FC studies on Blood showed 83% and 45.7% PD-L1 expressions for IDC and ILC, respectively. CD20 in ILC and IDC were 78.2% and 62.5%, respectively. Nevertheless, CD8 was 74.2% for IDC and 67.7% for ILC. Whereas, FC studies for PD-L1, CD20, and CD8 in ILC in tissues gave 34.4%, 30.2% and 35.1%, respectively. In addition, IDC tissue samples showed 16%, 12.5, and 13.5% for PD-L1, CD20, and CD8. The moderate stage of adenocarcinoma caused expression of PD-L1 within inflammatory cells, while expression was within neoplastic glandular cells in late stage. Conclusion PD-L1, CD8, and CD20 are considered as early predictor and tracking markers for breast cancer.
Collapse
Affiliation(s)
- Manar M. Hamed
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Egypt
| | - Mona S. Gouida
- Genetic Unit, Children Hospital, Mansoura University, Egypt
| | | | - Ahmed M.A. EL-Sokkary
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Egypt
| |
Collapse
|
19
|
Junjun S, Yangyanqiu W, Jing Z, Jie P, Jian C, Yuefen P, Shuwen H. Prognostic model based on six PD-1 expression and immune infiltration-associated genes predicts survival in breast cancer. Breast Cancer 2022; 29:666-676. [PMID: 35233733 PMCID: PMC9226094 DOI: 10.1007/s12282-022-01344-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 02/13/2022] [Indexed: 11/23/2022]
Abstract
Background The prognosis of breast cancer (BC) was associated with the expression of programmed cell death-1 (PD-1). Methods BC-related expression and clinical data were downloaded from TCGA database. PD-1 expression with overall survival and clinical factors were investigated. Gene set variation analysis (GSVA) and weighted gene correlation network analysis were performed to investigate the PD-1 expression-associated KEGG pathways and genes, respectively. Immune infiltration was analyzed using the ssGSEA algorithm and DAVID, respectively. Univariate and multivariable Cox and LASSO regression analyses were performed to select prognostic genes for modeling. Results High PD-1 expression was related to prolonged survival time (P = 0.014). PD-1 expression status showed correlations with age, race, and pathological subtype. ER- and PR-negative patients exhibited high PD-1 expression. The GSVA revealed that high PD-1 expression was associated with various immune-associated pathways, such as T cell/B cell receptor signaling pathway or natural killer cell-mediated cytotoxicity. The patients in the high-immune infiltration group exhibited significantly higher PD-1 expression levels. In summary, 397 genes associated with both immune infiltration and PD-1 expression were screened. Univariate analysis and LASSO regression model identified the six most valuable prognostic genes, namely IRC3, GBP2, IGJ, KLHDC7B, KLRB1, and RAC2. The prognostic model could predict survival for BC patients. Conclusion High PD-1 expression was associated with high-immune infiltration in BC patients. Genes closely associated with PD-1, immune infiltration and survival prognosis were screened to predict prognosis. Supplementary Information The online version contains supplementary material available at 10.1007/s12282-022-01344-2.
Collapse
Affiliation(s)
- Shen Junjun
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No. 1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang, China
| | - Wang Yangyanqiu
- Graduate School of Medical College of Zhejiang University, No. 268 Kaixuan Road, Jianggan District, Hangzhou, 310029, Zhejiang, China
| | - Zhuang Jing
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No. 1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang, China
| | - Pu Jie
- Graduate School of Nursing, Huzhou University, No. 1 Bachelor Road, Huzhou, 313000, Zhejiang, China
| | - Chu Jian
- Graduate School of Second Clinical Medicine Faculty, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, Zhejiang, China
| | - Pan Yuefen
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No. 1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang, China.
| | - Han Shuwen
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No. 1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang, China.
| |
Collapse
|
20
|
da Silva MC, Medeiros FS, da Silva NCH, Paiva LA, Gomes FODS, Costa E Silva M, Gomes TT, Peixoto CA, Rygaard MCV, Menezes MLB, Welkovic S, Donadi EA, Lucena-Silva N. Increased PD-1 Level in Severe Cervical Injury Is Associated With the Rare Programmed Cell Death 1 ( PDCD1) rs36084323 A Allele in a Dominant Model. Front Cell Infect Microbiol 2021; 11:587932. [PMID: 34290992 PMCID: PMC8288189 DOI: 10.3389/fcimb.2021.587932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 04/21/2021] [Indexed: 11/16/2022] Open
Abstract
The high-risk oncogenic human papillomavirus (HPV) has developed mechanisms for evasion of the immune system, favoring the persistence of the infection. The chronic inflammation further contributes to the progression of tissue injury to cervical cancer. The programmed cell death protein (PD-1) after contacting with its ligands (PD-L1 and PD-L2) exerts an inhibitory effect on the cellular immune response, maintaining the balance between activation, tolerance, and immune cell-dependent lesion. We evaluated 295 patients exhibiting or not HPV infection, stratified according to the location (injured and adjacent non-injured areas) and severity of the lesion (benign, pre-malignant lesions). Additionally, we investigated the role of the promoter region PDCD1 -606G>A polymorphism (rs36084323) on the studied variables. PD-1 and PDCD1 expression were evaluated by immunohistochemistry and qPCR, respectively, and the PDCD1 polymorphism was evaluated by nucleotide sequencing. Irrespective of the severity of the lesion, PD-1 levels were increased compared to adjacent uninjured areas. Additionally, in cervical intraepithelial neoplasia (CIN) I, the presence of HPV was associated with increased (P = 0.0649), whereas in CIN III was associated with decreased (P = 0.0148) PD-1 levels, compared to the uninjured area in absence of HPV infection. The PDCD1 -606A allele was rare in our population (8.7%) and was not associated with the risk for development of HPV infection, cytological and histological features, and aneuploidy. In contrast, irrespective of the severity of the lesion, patients exhibiting the mutant PDCD1 -606A allele at single or double doses exhibited increased protein and gene expression when compared to the PDCD1 -606GG wild type genotype. Besides, the presence of HPV was associated with the decrease in PDCD1 expression and PD-1 levels in carriers of the -606 A allele presenting severe lesions, suggesting that other mediators induced during the HPV infection progression may play an additional role. This study showed that increased PD-1 levels are influenced by the -606G>A nucleotide variation, particularly in low-grade lesions, in which the A allele favors increased PDCD1 expression, contributing to HPV immune system evasion, and in the high-grade lesion, by decreasing tissue PD-1 levels.
Collapse
Affiliation(s)
- Mauro César da Silva
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | - Fernanda Silva Medeiros
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | | | | | | | - Matheus Costa E Silva
- Clinical Immunology Division, Department of Medicine, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Thailany Thays Gomes
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | - Christina Alves Peixoto
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | | | | | - Stefan Welkovic
- Integrated Health Center Amaury de Medeiros (CISAM), University of Pernambuco, Recife, Brazil
| | - Eduardo Antônio Donadi
- Clinical Immunology Division, Department of Medicine, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Norma Lucena-Silva
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil.,Laboratory of Molecular Biology, IMIP Hospital, Pediatric Oncology Service, Recife, Brazil
| |
Collapse
|
21
|
Guo J, Wang S, Han Y, Jia Z, Wang R. Effects of transarterial chemoembolization on the immunological function of patients with hepatocellular carcinoma. Oncol Lett 2021; 22:554. [PMID: 34084221 PMCID: PMC8161415 DOI: 10.3892/ol.2021.12815] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 03/25/2021] [Indexed: 12/18/2022] Open
Abstract
The present study aimed to investigate the effects of transarterial chemoembolization (TACE) on the immune function of patients with hepatocellular carcinoma (HCC). A total of 114 patients with HCC were selected and their peripheral blood was collected before and 1 month after TACE treatment. Flow cytometry and reverse transcription-quantitative PCR were performed to analyze the changes in immune function in patients before and after treatment. Kaplan-Meier curves were plotted for survival analysis. The programmed cell death ligand 1 (PD-L1) and programmed cell death protein 1 (PD1) expression before TACE treatment were significantly higher in patients with poor TACE response compared with those patients with well response. Higher PD-L1 mRNA expression in the peripheral blood mononuclear cells after TACE predicted a superior prognosis. After TACE treatment, the proportion of CD4+/CD8+ cells were decreased while the expression levels of programmed cell death protein 1 (PD1) were significantly increased. To conclude, TACE could reduce the proportion of CD4+/CD8+ cells and improve the mRNA expression levels of PD1 in patients with HCC. The expression levels of PD1 and PD-L1 were closely related to the therapeutic effect of TACE and the prognosis of patients with HCC. TACE combined with immunotherapy may have potential clinical value for patients with HCC.
Collapse
Affiliation(s)
- Jingjing Guo
- Interventional Department of Liver Diseases, Qingdao Sixth People's Hospital, Qingdao, Shandong 266000, P.R. China
| | - Saixia Wang
- Integrated Outpatient Department, Qingdao Sixth People's Hospital, Qingdao, Shandong 266000, P.R. China
| | - Yujing Han
- Integrated Traditional Chinese Medicine and Western Medicine Department, Qingdao Sixth People's Hospital, Qingdao, Shandong 266000, P.R. China
| | - Zhongyuan Jia
- Department of General Practice, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan Maternity and Child Care Hospital, Jinan, Shandong 250000, P.R. China
| | - Runchao Wang
- Integrated Traditional Chinese Medicine and Western Medicine Department, Qingdao Sixth People's Hospital, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
22
|
Liu C, Seeram NP, Ma H. Small molecule inhibitors against PD-1/PD-L1 immune checkpoints and current methodologies for their development: a review. Cancer Cell Int 2021; 21:239. [PMID: 33906641 PMCID: PMC8077906 DOI: 10.1186/s12935-021-01946-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Programmed death-1/programmed death ligand-1 (PD-1/PD-L1) based immunotherapy is a revolutionary cancer therapy with great clinical success. The majority of clinically used PD-1/PD-L1 inhibitors are monoclonal antibodies but their applications are limited due to their poor oral bioavailability and immune-related adverse effects (irAEs). In contrast, several small molecule inhibitors against PD-1/PD-L1 immune checkpoints show promising blockage effects on PD-1/PD-L1 interactions without irAEs. However, proper analytical methods and bioassays are required to effectively screen small molecule derived PD-1/PD-L1 inhibitors. Herein, we summarize the biophysical and biochemical assays currently employed for the measurements of binding capacities, molecular interactions, and blocking effects of small molecule inhibitors on PD-1/PD-L1. In addition, the discovery of natural products based PD-1/PD-L1 antagonists utilizing these screening assays are reviewed. Potential pitfalls for obtaining false leading compounds as PD-1/PD-L1 inhibitors by using certain binding bioassays are also discussed in this review.
Collapse
Affiliation(s)
- Chang Liu
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Avedisian Hall Lab 440, 7 Greenhouse Road, Kingston, RI, 02881, USA.
| | - Navindra P Seeram
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Avedisian Hall Lab 440, 7 Greenhouse Road, Kingston, RI, 02881, USA
| | - Hang Ma
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Avedisian Hall Lab 440, 7 Greenhouse Road, Kingston, RI, 02881, USA.
| |
Collapse
|
23
|
Huang S, Liang C, Zhao Y, Deng T, Tan J, Lu Y, Liu S, Li Y, Chen S. Increased TOX expression concurrent with PD-1, Tim-3, and CD244 in T cells from patients with non-Hodgkin lymphoma. Asia Pac J Clin Oncol 2021; 18:143-149. [PMID: 33608984 DOI: 10.1111/ajco.13545] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 12/27/2022]
Abstract
AIM To characterize immune suppression in lymphoma, thymocyte selection-associated high mobility group box protein (TOX) expression and co-expression with programmed cell death receptor-1 (PD-1), T cell immunoglobulin mucin-domain-containing-3 (Tim-3), and CD244 in CD3+, CD4+, CD8+, and regulatory T (Treg) cells from patients with lymphomas were analyzed. METHODS TOX expression and co-expression with PD-1, Tim-3, and CD244 in CD3+, CD4+, Treg, and CD8+ T cells were analyzed by multi-color fluorescent flow cytometry using peripheral blood (PB) from 13 newly diagnosed, untreated lymphoma patients, and 11 healthy individuals. RESULTS An increased percentage of TOX+ CD3+, CD4+, and CD8+ T cells was found in PB from patients with B cell non-Hodgkin's lymphoma (B-NHL) in comparison with healthy controls. Moreover, TOX+PD-1+ and TOX+Tim-3+ double-positive T cells increased among the CD3+, CD4+, and CD8+T cell populations in the B-NHL group. There was apparent heterogeneity in TOX expression and co-expression with PD-1, Tim-3, and CD244 in CD3+, CD4+, and CD8+ T cells in different lymphoma patients. In addition, the percentage of CD4+CD25+FoxP3+ T cells (Treg) among the CD3+ and CD4+ T cells significantly increased, and the number of TOX+ and TOX+PD-1+ Treg cells also significantly increased in the B-NHL group. CONCLUSIONS Higher expression of TOX concurrent with PD-1, Tim-3, and CD244 in T cells from patients with B-NHL may contribute to T cell exhaustion and impair their special anti-tumor T cell activity. TOX may be considered a potential target for reversing T cell exhaustion and improving T cell function in hematological malignancies.
Collapse
Affiliation(s)
- Shuxin Huang
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Chaofeng Liang
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Yujie Zhao
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Tairan Deng
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Jiaxiong Tan
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yuhong Lu
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Sichu Liu
- Lymphoma Division, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yangqiu Li
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Shaohua Chen
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| |
Collapse
|
24
|
Clinical Implications of Exosomal PD-L1 in Cancer Immunotherapy. J Immunol Res 2021; 2021:8839978. [PMID: 33628854 PMCID: PMC7886511 DOI: 10.1155/2021/8839978] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/07/2021] [Accepted: 01/29/2021] [Indexed: 02/08/2023] Open
Abstract
Inhibiting the programmed cell death ligand-1 (PD-L1)/programmed cell death receptor-1 (PD-1) signaling axis reinvigorates the antitumor immune response with remarkable clinical efficacy. Yet, low response rates limit the benefits of immunotherapy to a minority of patients. Recent studies have explored the importance of PD-L1 as a transmembrane protein in exosomes and have revealed exosomal PD-L1 as a mechanism of tumor immune escape and immunotherapy resistance. Exosomal PD-L1 suppresses T cell effector function, induces systemic immunosuppression, and transfers functional PD-L1 across the tumor microenvironment (TME). Because of its significant contribution to immune escape, exosomal PD-L1 has been proposed as a biomarker to predict immunotherapy response and to assess therapeutic efficacy. In this review, we summarize the immunological mechanisms of exosomal PD-L1, focusing on the factors that lead to exosome biogenesis and release. Next, we review the effect of exosomal PD-L1 on T cell function and its role across the TME. In addition, we discuss the latest findings on the use of exosomal PD-L1 as a biomarker for cancer immunotherapy. Throughout this review, we propose exosomal PD-L1 as a critical mediator of tumor progression and highlight the clinical implications that follow for immuno-oncology, discussing the potential to target exosomes to advance cancer treatment.
Collapse
|
25
|
PD-L1 Protein Expression in Middle Eastern Breast Cancer Predicts Favorable Outcome in Triple-Negative Breast Cancer. Cells 2021; 10:cells10020229. [PMID: 33503961 PMCID: PMC7910988 DOI: 10.3390/cells10020229] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 12/14/2022] Open
Abstract
Programmed cell-death ligand 1 (PD-L1) has been shown to induce potent T-cell mediated anti-tumoral immunity. The significance of PD-L1 expression in the prognosis of breast cancer (BC) remains controversial and its prevalence and prognostic value in breast cancer from Middle Eastern ethnicity is lacking. A total of 1003 unselected Middle Eastern breast cancers were analyzed for PD-L1 expression using immunohistochemistry. PD-L1 expression, seen in 32.8% (329/1003) of cases, was significantly associated with poor prognostic indicators such as younger patients, high-grade tumors, estrogen-receptor (ER)-negative, progesterone-receptor (PR)-negative, and triple-negative breast cancers (TNBC) as well as high Ki-67 index. We also found a significant association between PD-L1 expression and deficient mismatch repair protein expression. No association was found between PD-L1 expression and clinical outcome. However, on further subgroup analysis, PD-L1 expression was found to be an independent marker for favorable overall survival and recurrence-free survival in TNBC. In conclusion, we demonstrated strong association between PD-L1 and mismatch repair deficiency in Middle Eastern BC patients and that PD-L1 overexpression in tumor cells was an independent prognostic marker in TNBCs from Middle Eastern ethnicity. Overall, these findings might help in the development of more appropriate treatment strategies for BC in Middle Eastern population.
Collapse
|
26
|
Li J, Yin L, Chen Y, An S, Xiong Y, Huang G, Liu J. PD-L1 correlated gene expression profiles and tumor infiltrating lymphocytes in pancreatic cancer. Int J Med Sci 2021; 18:3150-3157. [PMID: 34400885 PMCID: PMC8364450 DOI: 10.7150/ijms.61771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/16/2021] [Indexed: 11/16/2022] Open
Abstract
Objective: To study the expression and clinical value of PD-L1 gene in pancreatic cancer, and to predict the role of PD-L1 gene in the development of pancreatic cancer. Methods: The pancreatic cancer datasets were downloaded from the Cancer Genome Atlas (TCGA) and the Oncomine to obtain the PD-L1 gene expression profile and clinical information. Bioinformatics methods were used to analyze the correlation between the expression level of PD-L1 gene in pancreatic cancer and clinicopathological indicators, as well as its influence on prognosis. GSEA and WGCNA analysis was performed to predict the possible pathways of PD-L1 gene regulation in pancreatic cancer. TIMER and MCP-counter were used for PD-L1 with immune infiltration. The genes interact with PD-L1 were also investigated by STING and immunoco-precipitation combined with mass spectrometry analysis (IP-MS). Results: In TCGA database, the overall survival of patients with high expression of PD-L1 gene was significantly lower than that of patients with low expression of PD-L1 gene (χ2 = 12.52, P < 0.001). The samples with high expression of PD-L1 gene showed enrichment of 8 pathways including toll-like receptor signaling pathway and NOD receptor signaling pathway (P < 0.01, FDR < 0.05). Immune infiltration analysis suggested that PD-L1 were associated with monocytic lineage (r = 0.5). The proteins interacting with PD-L1 are mainly concentrated in RNA binding, ribosome, spliceosome and other biological processes or pathways. Conclusion: PD-L1 gene may play an important role in the development of pancreatic cancer and is expected to be a prognostic indicator of pancreatic cancer.
Collapse
Affiliation(s)
- Jiajin Li
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lu Yin
- Department of Cardiology, Gongli Hospital of Shanghai Pudong New Area, Shanghai 200135, China
| | - Yumei Chen
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shuxian An
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yi Xiong
- Department of Bioinformatics and Biostatistics, School of Life Science and Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
27
|
Rrapaj E, Giacometti L, Spina P, Salvo M, Baselli GA, Veggiani C, Rena O, Trisolini E, Boldorini RL. Programmed cell death 1 ligand 1 (PD-L1) expression is associated with poor prognosis of malignant pleural mesothelioma patients with good performance status. Pathology 2020; 53:462-469. [PMID: 33272690 DOI: 10.1016/j.pathol.2020.09.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/30/2020] [Accepted: 09/04/2020] [Indexed: 01/22/2023]
Abstract
Malignant pleural mesothelioma (MPM) is often associated with a poor prognosis and options for the treatment of this disease are few. To date, the important role of the immune microenvironment in modifying the disease natural history is well established. The programmed cell death pathway (PD-1/PD-L1) limits the T lymphocyte activation in peripheral tissues when an inflammatory response occurs, and controls the tumour immune escape. PD-L1 is broadly expressed in several malignant tumours and associated with poor clinical outcomes. Thus, the aim of our study is to investigate the potential role of PD-L1 expression in MPM prognosis. Biopsy samples from 198 patients diagnosed with MPM were examined by immunohistochemistry (IHC) and reverse transcription-polymerase chain reaction (RT-PCR) to evaluate PD-L1 protein and gene expression. For PD-L1 protein expression we consider at least 5% membranous staining as positive. Gene expression levels were calculated with ΔΔCt method. Positive expression of PD-L1 by IHC was correlated with worse overall survival (OS; p=0.0225) in MPM patients. PD-L1 positive status was correlated with worse OS in the subgroup of patients with ECOG score <2 (p=0.0004, n=129) and these data were confirmed by multivariate analysis. No significant correlation was found between PD-L1 gene expression and OS. Our results show that PD-L1 evaluated by IHC assay may be a prognostic biomarker for MPM patients with good performance status.
Collapse
Affiliation(s)
- Eltjona Rrapaj
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy.
| | - Lorenzo Giacometti
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Paolo Spina
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy; Cantonal Institute of Pathology, Locarno, Switzerland
| | - Michela Salvo
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy; Unit of Pathology, Maggiore Della Carita Hospital, Novara, Italy
| | - Guido Alessandro Baselli
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Claudia Veggiani
- Unit of Pathology, Maggiore Della Carita Hospital, Novara, Italy
| | - Ottavio Rena
- Unit of Thoracic Surgery, Maggiore Della Carita Hospital, Novara, Italy
| | - Elena Trisolini
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy; Unit of Pathology, Maggiore Della Carita Hospital, Novara, Italy
| | - Renzo Luciano Boldorini
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy; Unit of Pathology, Maggiore Della Carita Hospital, Novara, Italy
| |
Collapse
|
28
|
Rao J, Xia J, Yang W, Wu C, Sha B, Zheng Q, Cheng F, Lu L. Complete response to immunotherapy combined with an antiangiogenic agent in multiple hepatic metastases after radical surgery for advanced gallbladder cancer: a case report. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1609. [PMID: 33437808 PMCID: PMC7791255 DOI: 10.21037/atm-20-4420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Most advanced gallbladder cancers (GBCa) are unresectable or metastatic once diagnosed, and even patients who undergo surgery have a high risk of recurrence and metastasis. Immunotherapy, especially immune checkpoint inhibitors (ICIs), combined with an antiangiogenic agent, is an emerging prospective treatment for GBCa. However, the efficacy and safety of this combination therapy have not yet been investigated. We report the case of a 70-year-old female patient with recurrent metastatic GBCa (stage IVB) after radical surgery. Immunohistochemical examination revealed that 10% of the tumor cells expressed programmed cell death protein-1 (PD-1) and programmed cell death receptor ligand 1 (PD-L1). Whole-exome sequencing showed cancer tissues with a low tumor mutational burden (TMB) and microsatellite stability (MSS). The patient received Camrelizumab (200 mg, every three weeks) and Apatinib (40 mg/d). The clinical and immunological responses were observed, and the patient achieved a complete response after five cycles. This is the first case describing the efficacy and safety of Camrelizumab plus Apatinib in a GBCa patient with weak PD-1 and PD-L1 expression, and low TMB and MSS. The treatment had a tolerable safety profile and a complete response in the patient. Also, we found that the cluster of differentiation (CD)16+CD56+natural killer (NK) cell ratio in peripheral blood was increased after the combined treatment. Immunotherapy with antiangiogenic drugs may be a potential treatment option for patients with recurrent GBC or GBCa.
Collapse
Affiliation(s)
- Jianhua Rao
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Jinguo Xia
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Wenjie Yang
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Chen Wu
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Bowen Sha
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Qitong Zheng
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Feng Cheng
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Ling Lu
- Hepatobiliary Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| |
Collapse
|
29
|
Liu Q, Cheng R, Kong X, Wang Z, Fang Y, Wang J. Molecular and Clinical Characterization of PD-1 in Breast Cancer Using Large-Scale Transcriptome Data. Front Immunol 2020; 11:558757. [PMID: 33329517 PMCID: PMC7718028 DOI: 10.3389/fimmu.2020.558757] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 10/16/2020] [Indexed: 12/20/2022] Open
Abstract
Despite the impressive impact of PD-1 (programmed cell death protein 1)-targeted cancer immunotherapy, a great part of patients with cancer fail to respond. PD-1 impact on immune cells in addition to T cells, and the synergistic role of PD-1 with other immune modulators remain largely unknown. To fill this gap, we systematically investigated PD-1-related transcriptome data and relevant clinical information derived from TCGA (The Cancer Genome Atlas) and METABRIC (Molecular Taxonomy of Breast Cancer International Consortium), which involved a total of 2,994 breast cancer patients. Our results revealed the relationship among PD-1 and major molecular and clinical characteristics in breast cancer. More importantly, we depicted the association landscape between PD-1 and other immune cell populations. Gene ontology analyses and gene set variation analysis (GSVA) of genes correlated with PD-1 revealed that PD-1 was mainly involved in immune responses and inflammatory activities. We also elucidated the association of PD-1 with other immune modulators in pan-cancer level, especially the potential synergistic relationship between PD-1 and other immune checkpoints members in breast cancer. In short, the expression level of PD-1 was bound up with breast cancer malignancy, which could be used as a potential biomarker; PD-1 might manipulate the anti-tumor immune response by impacting not just T cells, and this might vary among different tumor types. Furthermore, PD-1 might synergize with other immune checkpoint members to modulate the immune microenvironment in breast cancer.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ran Cheng
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhongzhao Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
30
|
Matikas A, Zerdes I, Lövrot J, Sifakis E, Richard F, Sotiriou C, Rassidakis G, Bergh J, Valachis A, Foukakis T. PD-1 protein and gene expression as prognostic factors in early breast cancer. ESMO Open 2020; 5:e001032. [PMID: 33172959 PMCID: PMC7656908 DOI: 10.1136/esmoopen-2020-001032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/30/2020] [Accepted: 10/03/2020] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND There is a paucity of data on the prognostic value of programmed cell death protein 1 (PD-1) protein and gene expression in early breast cancer (BC) and the present study's aim was to comprehensively investigate it. METHODS The study consisted of three parts: a correlative analysis of PD-1 protein and gene expression from an original patient cohort of 564 patients with early BC; a systematic review and trial-level meta-analysis on the association between PD-1 protein expression and disease-free survival/overall survival (OS) in early BC; and a pooled gene expression analysis from publicly available transcriptomic datasets regarding PDCD1 expression. RESULTS In the study cohort, PD-1 protein, but not gene expression, was associated with improved OS (HRadj=0.73, 95% CI 0.55 to 0.97, p=0.027 and HRadj=0.88, 95% CI 0.68 to 1.13, p=0.312, respectively). In the trial-level meta-analysis, PD-1 protein expression was not found to be statistically significantly associated with outcomes in the overall population. Finally, in the pooled gene expression analysis, higher PDCD1 expression was associated with better OS in multivariable analysis in the entire population (HRadj=0.89, 95% CI 0.80 to 0.99, p=0.025) and in basal-like tumours. CONCLUSIONS PD-1 protein and gene expression seem to be promising prognostic factors in early BC. Standardisation of detection and assessment methods is of utmost importance.
Collapse
Affiliation(s)
- Alexios Matikas
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis Zerdes
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| | - John Lövrot
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Emmanouil Sifakis
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Christos Sotiriou
- Department of Medical Oncology, Institute Jules Bordet, Brussels, Belgium
| | - Georgios Rassidakis
- Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Bergh
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Antonis Valachis
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Theodoros Foukakis
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Analysis of Cross-Association between mRNA Expression and RNAi Efficacy for Predictive Target Discovery in Colon Cancers. Cancers (Basel) 2020; 12:cancers12113091. [PMID: 33114107 PMCID: PMC7690798 DOI: 10.3390/cancers12113091] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/20/2022] Open
Abstract
Simple Summary This study focused on finding genes for which mRNA expression was able to predict the anticancer efficacy of its RNAi treatment. Predictive target discovery is of critical importance in developing biomarker-based strategies of precision medicine. We demonstrated this carrying out cross-association analysis on collateral mRNA expression and RNAi treatment data of ~12,000 genes on a colon cell line panel. The analysis revealed several genes with significant association between mRNA expression level and the inhibitory efficacy of its RNAi treatment. The experimental validation confirm that this simple approach has general applications for studying gene association between omics data from diverse cancer lineages. Abstract The availability of large-scale, collateral mRNA expression and RNAi data from diverse cancer cell types provides useful resources for the discovery of anticancer targets for which inhibitory efficacy can be predicted from gene expression. Here, we calculated bidirectional cross-association scores (predictivity and descriptivity) for each of approximately 18,000 genes identified from mRNA and RNAi (i.e., shRNA and sgRNA) data from colon cancer cell lines. The predictivity score measures the difference in RNAi efficacy between cell lines with high vs. low expression of the target gene, while the descriptivity score measures the differential mRNA expression between groups of cell lines exhibiting high vs. low RNAi efficacy. The mRNA expression of 90 and 74 genes showed significant (p < 0.01) cross-association scores with the shRNA and sgRNA data, respectively. The genes were found to be from diverse molecular classes and have different functions. Cross-association scores for the mRNA expression of six genes (CHAF1B, HNF1B, HTATSF1, IRS2, POLR2B and SATB2) with both shRNA and sgRNA efficacy were significant. These genes were interconnected in cancer-related transcriptional networks. Additional experimental validation confirmed that siHNF1B efficacy is correlated with HNF1B mRNA expression levels in diverse colon cancer cell lines. Furthermore, KIF26A and ZIC2 gene expression, with which shRNA efficacy displayed significant scores, were found to correlate with the survival rate from colon cancer patient data. This study demonstrates that bidirectional predictivity and descriptivity calculations between mRNA and RNAi data serve as useful resources for the discovery of predictive anticancer targets.
Collapse
|
32
|
Yang DP, Lu HP, Chen G, Yang J, Gao L, Song JH, Chen SW, Mo JX, Kong JL, Tang ZQ, Li CB, Zhou HF, Yang LJ. Integrated expression analysis revealed RUNX2 upregulation in lung squamous cell carcinoma tissues. IET Syst Biol 2020; 14:252-260. [PMID: 33095746 PMCID: PMC8687175 DOI: 10.1049/iet-syb.2020.0063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/01/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
This study aimed to investigate the clinicopathological significance and prospective molecular mechanism of RUNX family transcription factor 2 (RUNX2) in lung squamous cell carcinoma (LUSC). The authors used immunohistochemistry (IHC), RNA-seq, and microarray data from multi-platforms to conduct a comprehensive analysis of the clinicopathological significance and molecular mechanism of RUNX2 in the occurrence and development of LUSC. RUNX2 expression was significantly higher in 16 LUSC tissues than in paired non-cancerous tissues detected by IHC (P < 0.05). RNA-seq data from the combination of TCGA and genotype-tissue expression (GTEx) revealed significantly higher expression of RUNX2 in 502 LUSC samples than in 476 non-cancer samples. The expression of RUNX2 protein was also significantly higher in pathologic T3-T4 than in T1-T2 samples (P = 0.031). The pooled standardised mean difference (SMD) for RUNX2 was 0.87 (95% CI, 0.58-1.16), including 29 microarrays from GEO and one from ArrayExpress. The co-expression network of RUNX2 revealed complicated connections between RUNX2 and 45 co-expressed genes, which were significantly clustered in pathways including ECM-receptor interaction, focal adhesion, protein digestion and absorption, human papillomavirus infection and PI3K-Akt signalling pathway. Overexpression of RUNX2 plays an essential role in the clinical progression of LUSC.
Collapse
Affiliation(s)
- Da-Ping Yang
- Department of Pathology, The Eighth Affiliated Hospital of Guangxi Medical University/Guigang People's Hospital, Guigang, Guangxi, People's Republic of China
| | - Hui-Ping Lu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Jie Yang
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Li Gao
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Jian-Hua Song
- Department of Pathology, The Eighth Affiliated Hospital of Guangxi Medical University/Guigang People's Hospital, Guigang, Guangxi, People's Republic of China
| | - Shang-Wei Chen
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Jun-Xian Mo
- Department of Cardio-Thoracic Surgery, The Seventh Affiliated Hospital of Guangxi Medical University/Wuzhou Gongren Hospital, Wuzhou, Guangxi, People's Republic of China
| | - Jin-Liang Kong
- Ward of Pulmonary and Critical Care Medicine, Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Zhong-Qing Tang
- Department of Pathology, The Seventh Affiliated Hospital of Guangxi Medical University/Wuzhou Gongren Hospital, Wuzhou, Guangxi, People's Republic of China
| | - Chang-Bo Li
- Department of Cardio-Thoracic Surgery, The Seventh Affiliated Hospital of Guangxi Medical University/Wuzhou Gongren Hospital, Wuzhou, Guangxi, People's Republic of China
| | - Hua-Fu Zhou
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China.
| | - Lin-Jie Yang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| |
Collapse
|
33
|
PD-1/PD-L1 axis regulation in cancer therapy: The role of long non-coding RNAs and microRNAs. Life Sci 2020; 256:117899. [DOI: 10.1016/j.lfs.2020.117899] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/31/2020] [Accepted: 05/31/2020] [Indexed: 02/07/2023]
|
34
|
Eddy JA, Thorsson V, Lamb AE, Gibbs DL, Heimann C, Yu JX, Chung V, Chae Y, Dang K, Vincent BG, Shmulevich I, Guinney J. CRI iAtlas: an interactive portal for immuno-oncology research. F1000Res 2020; 9:1028. [PMID: 33214875 PMCID: PMC7658727 DOI: 10.12688/f1000research.25141.1] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/22/2020] [Indexed: 12/18/2022] Open
Abstract
The Cancer Research Institute (CRI) iAtlas is an interactive web platform for data exploration and discovery in the context of tumors and their interactions with the immune microenvironment. iAtlas allows researchers to study immune response characterizations and patterns for individual tumor types, tumor subtypes, and immune subtypes. iAtlas supports computation and visualization of correlations and statistics among features related to the tumor microenvironment, cell composition, immune expression signatures, tumor mutation burden, cancer driver mutations, adaptive cell clonality, patient survival, expression of key immunomodulators, and tumor infiltrating lymphocyte (TIL) spatial maps. iAtlas was launched to accompany the release of the TCGA PanCancer Atlas and has since been expanded to include new capabilities such as (1) user-defined loading of sample cohorts, (2) a tool for classifying expression data into immune subtypes, and (3) integration of TIL mapping from digital pathology images. We expect that the CRI iAtlas will accelerate discovery and improve patient outcomes by providing researchers access to standardized immunogenomics data to better understand the tumor immune microenvironment and its impact on patient responses to immunotherapy.
Collapse
Affiliation(s)
| | | | | | - David L Gibbs
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | | | - Jia Xin Yu
- Anna-Maria Kellen Clinical Accelerator, Cancer Research Institute, New York, NY, 10006, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Huang Y, Shen A. The prediction potential of neutrophil-to-lymphocyte ratio for the therapeutic outcomes of programmed death receptor-1/programmed death ligand 1 inhibitors in non-small cell lung cancer patients: A meta-analysis. Medicine (Baltimore) 2020; 99:e21718. [PMID: 32846790 PMCID: PMC7447402 DOI: 10.1097/md.0000000000021718] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Programmed death receptor-1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitors have been demonstrated to improve the prognosis of patients with advanced non-small cell lung cancer (NSCLC) compared with chemotherapy. However, there were still some non-responders. Thus, how to effectively screen the responder may be an important issue. Recent studies revealed the immune-related indicator, neutrophil-lymphocyte ratio (NLR), may predict the therapeutic effects of anti-PD1/PD-L1 antibodies; however, the results were controversial. This study was to re-evaluate the prognostic potential of NLR for NSCLC patients receiving PD1/PD-L1 inhibitors by performing a meta-analysis. METHODS Eligible studies were identified by searching online databases of PubMed, EMBASE and Cochrane Library. The predictive values of NLR for overall survival, (OS), progression free survival (PFS) and overall response rate (ORR) were estimated by hazard ratio (HR) with 95% confidence interval (CI). RESULTS Twenty-four studies involving 2196 patients were included. The pooled analysis demonstrated that elevated NLR before PD-1/PD-L1 inhibitor treatment was a predictor of poor OS (HR = 2.17; 95% CI: 1.64 - 2.87, P < .001), PFS (HR = 1.54; 95% CI: 1.34 - 1.78, P < .001) and low ORR (HR = 0.64; 95% CI: 0.44 - 0.95, P = .027) in NSCLC patients. Subgroup analysis revealed the predictive ability of NLR for OS and PFS was not changed by ethnicity, sample size, cut-off, HR source, study design or inhibitor type (except the combined anti-PD-L1 group); while its association with ORR was only significant when the cut-off value was less than 5 and the studies were prospectively designed. CONCLUSION Our findings suggest patients with lower NLR may benefit from the use of PD-1/PD-L1 inhibitors to prolong their survival period.
Collapse
|
36
|
Prognostic Role of Immune Markers in Triple Negative Breast Carcinoma. Pathol Oncol Res 2020; 26:2733-2745. [PMID: 32681436 DOI: 10.1007/s12253-020-00874-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/09/2020] [Indexed: 12/21/2022]
Abstract
Tumor immune microenvironment (TIME) is a significant prognostic parameter for triple negative breast carcinomas (TNBC) due to being a target for immunotherapeutic agents and its essential role during the cancer immunoediting process. In this study, CD8, FOXP3, CD163, PD-L1/SP142 and PD-L1/SP263 antibodies were examined in a sample of 51 TNBC cases. Patients who received neoadjuvant therapy were excluded. CD8, FOXP3 and CD163 antibodies were evaluated separately in intratumoral area (ITA) and tumor stroma (TS). PD-L1 status was also examined in tumor cells (TC) and immune cells (IC) using both SP142 and SP263 antibodies. In multivariate Cox regressions, the only antibody that was found to be significantly associated with survival was SP142. SP142-positivity in TC and IC was related to increased overall survival. Higher CD163 expression in ITA and SP263-positivity in IC were associated with younger age. Lymphatic/angioinvasion was more frequent in cases with negative/low CD8 and FOXP3 expressions. Moreover, metastatic axillary lymph node(s) was associated with negative/low FOXP3 expression in TS. CD8, FOXP3, CD163, SP142 and SP263 expressions were positively correlated with each other, except a mild discordance caused by CD163 in ITA. Although PD-L1 status with both SP142 and SP263 antibodies were concordant in the majority of cases, 33.3% and 13.7% of the cases showed SP142-negative/SP263-positive pattern in TC and IC respectively. In conclusion, we suggest that composition, density and localization of the immune cells and the check point molecules are important prognostic parameters in TNBC. Immunohistochemistry can be used as an accessible and less expensive tool to demonstrate TIME.
Collapse
|
37
|
Su J, Zhang X, Zhao Q, Guo Z, Wu J, Chen G, Liang Q, Chen Z, He Z, Cai X, Xie M, Zheng L, Zhao K. PD-1 mRNA expression in peripheral blood mononuclear cells as a biomarker for different stages of primary gouty arthritis. J Cell Mol Med 2020; 24:9323-9331. [PMID: 32639111 PMCID: PMC7417685 DOI: 10.1111/jcmm.15582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 06/05/2020] [Indexed: 12/31/2022] Open
Abstract
There is currently a lack of biomarkers to assist the diagnosis and prediction of primary gouty arthritis (PG). Therefore, we evaluated the clinical value of programmed cell death protein 1 (PD-1) mRNA expression in peripheral blood mononuclear cells (PBMCs) of patients with PG. This study included 36 patients with acute phase PG (APPG), 48 with non-acute phase PG (NAPPG), 42 with asymptomatic hyperuricemia (AH) and 79 normal controls (NCs). PD-1 mRNA expression levels were detected by qRT-PCR. PD-1 mRNA expression was statistically analysed by ANOVA or t tests, while correlations between PD-1 mRNA and clinical variables were assessed using Pearson correlation tests. Receiver operator characteristic (ROC) curve analysis was used to evaluate the diagnostic value of PD-1 in different PG stages. PD-1 mRNA expression was significantly lower in patients with APPG than that in NAPPG, AH and NCs (P < 0.01). Correlation analysis revealed that PD-1 mRNA levels correlated negatively with T-score (r = -0.209, P < 0.01). ROC curve analysis showed that serum uric acid (SUA), PD-1 mRNA and both combined displayed higher diagnostic value in patients with PG, NAPPG and APPG compared to that in NCs and patients with non-PG arthritis (NPG). Moreover, ROC curve analysis showed that SUA and PD-1 mRNA had good diagnostic value in APPG, with the greatest diagnostic power when combined. PD-1 mRNA could be a clinical auxiliary diagnostic biomarker for APPG, and the combined use of PD-1 mRNA and SUA is better than that of SUA alone.
Collapse
Affiliation(s)
- Jing Su
- Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuefang Zhang
- Hai Zhu Qu Jiang Hai Jie Community Health Service Center, Guangzhou, China
| | - Qing Zhao
- Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhaodi Guo
- Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianxiong Wu
- Hai Zhu Qu Jiang Hai Jie Community Health Service Center, Guangzhou, China
| | - Guoqiang Chen
- Hai Zhu Qu Jiang Hai Jie Community Health Service Center, Guangzhou, China
| | - Qianxin Liang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhixiang Chen
- Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhiliang He
- Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiuping Cai
- Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Manlin Xie
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Zheng
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kewei Zhao
- Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
38
|
Profiles of PD-1, PD-L1, PD-L2 in Gastric Cancer and Their Relation with Mutation, Immune Infiltration, and Survival. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2496582. [PMID: 32596285 PMCID: PMC7298268 DOI: 10.1155/2020/2496582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
Abstract
Background Although multiple types of cancers demonstrated favorable outcome after immunotherapy of PD-1/PD-L1 blockade, the specific regulatory mechanism of PD genes in gastric cancer (GC) remains largely unknown. Materials and Methods Expression of RNA, copy number variants, and clinical parameters of GC individuals from TCGA were analyzed. Coexpressed genes for PD-1, PD-L1, and PD-L2 were selected by correlation analysis and confirmed by STRING. Gene Ontology and KEGG pathway analyses were performed by clusterProfiler. The influence of PD-1/PD-L1/PD-L2 on immune cell infiltration was investigated by MCP-counter. Results PD-L2 demonstrated significant relation with clinical stage of GC (P = 0.043). Survival analysis showed that PD-1 expression was correlated with better prognosis of GC patients (HR = 0.70, P = 0.031), but PD-L2 expression was related with worse survival (HR = 1.42, P = 0.032). Mutation of PIK3CA could alter the level of PD-1, PD-L1, and PD-L2 (P < 0.001), and TP53 mutation demonstrated significant correlation with PD-L1 (P = 0.015) and PD-L2 (P = 0.014) expression. Enrichment analysis of PD-1/PD-L1/PD-L2 coexpressed genes indicated a biological process of mononuclear cell proliferation, leukocyte cell-cell adhesion, and lymphocyte activation as well as KEGG pathways including cell differentiation of Th1 and Th2, cell differentiation of Th17, and hematopoietic cell landscape. As for immune infiltration analysis, PD-1 was mainly related with cytotoxic lymphocytes and endothelial cells; PD-L1 were associated with monocytic lineage; PD-L2 showed significant correlation with myeloid dendritic cells. Conclusion PD-1 expression showed association with better prognosis of GC, and PD-L2 expression was related with worse survival. Mutations of PIK3CA and TP53 significantly correlated with PD-1/PD-L1/PD-L2 axis. PD-1/PD-L1/PD-L2 coexpressed genes demonstrated enrichment in mononuclear cell proliferation, leukocyte cell-cell adhesion, and lymphocyte activation as well as KEGG pathways including cell differentiation of Th1, Th2, and Th17.
Collapse
|
39
|
Zhou Y, Zong H, Han L, Xie Y, Jiang H, Gilly J, Zhang B, Lu H, Chen J, Sun R, Pan Z, Zhu J. A novel bispecific antibody targeting CD3 and prolactin receptor (PRLR) against PRLR-expression breast cancer. J Exp Clin Cancer Res 2020; 39:87. [PMID: 32398042 PMCID: PMC7216678 DOI: 10.1186/s13046-020-01564-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/24/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Prolactin receptor (PRLR) is highly expressed in a subset of human breast cancer and prostate cancer, which makes it a potential target for cancer treatment. In clinical trials, the blockade of PRLR was shown to be safe but with poor efficacy. It is therefore urgent to develop new therapies against PRLR target. Bispecific antibodies (BsAbs) could guide immune cells toward tumor cells, and produced remarkable effects in some cancers. METHODS In this study, a bispecific antibody targeting both tumor antigen PRLR and T cell surface CD3 antigen (PRLR-DbsAb) was constructed by split intein mediated protein transsplicing (BAPTS) system for the first time. Its binding activity was determined by Biacore and Flow cytometry, and target-dependent T cell mediated cytotoxicity was detected using LDH release assay. ELISA was utilized to study the secretion of cytokines by immune cells. Subcutaneous tumor mouse models were used to analyze the in vivo anti-tumor effects of PRLR-DbsAb. RESULTS PRLR-DbsAb in vitro could recruit and activate T cells to promote the release of Th1 cytokines IFN- γ and TNF- α, which could kill PRLR expressed breast cancer cells. In xenograft models with breast cancer cell line T47D, NOD/SCID mice intraperitoneally injected with PRLR-DbsAb exhibited significant inhibition of tumor growth and a longer survival compared to mice treated with PRLR monoclonal antibody (PRLR mAb). CONCLUSIONS Both in vitro and in vivo experiments showed PRLR-DbsAb had a potential therapy of cancer treatment potential therapy for cancer. Immunotherapy may be a promising treatment against the tumor target of PRLR.
Collapse
Affiliation(s)
- Yuexian Zhou
- Engineering Research Center of Cell & Therapeutic Antibody, MOE,Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
- School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
| | - Huifang Zong
- Engineering Research Center of Cell & Therapeutic Antibody, MOE,Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
- School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
| | - Lei Han
- Engineering Research Center of Cell & Therapeutic Antibody, MOE,Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
- School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
| | | | - Hua Jiang
- Jecho Laboratories, Inc, Frederick, MD, USA
| | - John Gilly
- Jecho Laboratories, Inc, Frederick, MD, USA
| | - Baohong Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, MOE,Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
- School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
| | - Huili Lu
- Engineering Research Center of Cell & Therapeutic Antibody, MOE,Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
- School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
| | - Jie Chen
- Engineering Research Center of Cell & Therapeutic Antibody, MOE,Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
- School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
| | - Rui Sun
- Engineering Research Center of Cell & Therapeutic Antibody, MOE,Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
- School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
| | - Zhidi Pan
- Engineering Research Center of Cell & Therapeutic Antibody, MOE,Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
- School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China
| | - Jianwei Zhu
- Engineering Research Center of Cell & Therapeutic Antibody, MOE,Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China.
- School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road, Shanghai, China.
- Jecho Laboratories, Inc, Frederick, MD, USA.
- Jecho Biopharmaceuticals Co., Ltd, Tianjin, China.
| |
Collapse
|
40
|
Tu C, Zheng Y, Zhang H, Wang J. Exploration of the personalized immune checkpoint atlas of plasma cell dyscrasias patients using high‑dimensional single‑cell analysis. Oncol Rep 2020; 44:224-240. [PMID: 32319658 PMCID: PMC7251663 DOI: 10.3892/or.2020.7587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 04/01/2020] [Indexed: 01/22/2023] Open
Abstract
Immune checkpoint blockade endows patients with unparalleled success in conquering cancer. Unfortunately, inter-individual heterogeneity causes failure in controlling tumors in many patients. Emerging mass cytometry technology is capable of revealing a multiscale onco-immune landscape that improves the efficacy of cancer immunotherapy. We introduced mass cytometry to determine the personalized immune checkpoint status in bone marrow and peripheral blood samples from 3 patients with multiple myeloma, amyloid light-chain amyloidosis, and solitary bone plasmacytoma and 1 non-hematologic malignancy patient. The expression of 18 immune regulatory receptors and ligands on 17 defined cell populations was simultaneously examined. By single-cell analyses, we identified the T cell clusters that serve as immunosuppressive signal source and revealed integrated immune checkpoint axes of individuals, thereby providing multiple potential immunotherapeutic targets, including programmed cell death protein 1 (PD-1), inducible co-stimulator (ICOS), and cluster of differentiation 28 (CD28), for each patient. Distinguishing the cell populations that function as providers and receivers of the immune checkpoint signals demonstrated a distinct cross-interaction network of immunomodulatory signals in individuals. These in-depth personalized data demonstrate mass cytometry as a powerful innovation to discover the systematical immune status in the primary and peripheral tumor microenvironment.
Collapse
Affiliation(s)
- Chenggong Tu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Yongjiang Zheng
- Department of Hematology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Hui Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Jinheng Wang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| |
Collapse
|
41
|
Padmanabhan R, Kheraldine HS, Meskin N, Vranic S, Al Moustafa AE. Crosstalk between HER2 and PD-1/PD-L1 in Breast Cancer: From Clinical Applications to Mathematical Models. Cancers (Basel) 2020; 12:E636. [PMID: 32164163 PMCID: PMC7139939 DOI: 10.3390/cancers12030636] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/12/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is one of the major causes of mortality in women worldwide. The most aggressive breast cancer subtypes are human epidermal growth factor receptor-positive (HER2+) and triple-negative breast cancers. Therapies targeting HER2 receptors have significantly improved HER2+ breast cancer patient outcomes. However, several recent studies have pointed out the deficiency of existing treatment protocols in combatting disease relapse and improving response rates to treatment. Overriding the inherent actions of the immune system to detect and annihilate cancer via the immune checkpoint pathways is one of the important hallmarks of cancer. Thus, restoration of these pathways by various means of immunomodulation has shown beneficial effects in the management of various types of cancers, including breast. We herein review the recent progress in the management of HER2+ breast cancer via HER2-targeted therapies, and its association with the programmed death receptor-1 (PD-1)/programmed death ligand-1 (PD-L1) axis. In order to link research in the areas of medicine and mathematics and point out specific opportunities for providing efficient theoretical analysis related to HER2+ breast cancer management, we also review mathematical models pertaining to the dynamics of HER2+ breast cancer and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Regina Padmanabhan
- Department of Electrical Engineering, Qatar University, 2713 Doha, Qatar;
- Biomedical Research Centre, Qatar University, 2713 Doha, Qatar;
| | - Hadeel Shafeeq Kheraldine
- Biomedical Research Centre, Qatar University, 2713 Doha, Qatar;
- College of Pharmacy, QU Health, Qatar University, 2713 Doha, Qatar
| | - Nader Meskin
- Department of Electrical Engineering, Qatar University, 2713 Doha, Qatar;
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, 2713 Doha, Qatar;
| | - Ala-Eddin Al Moustafa
- Biomedical Research Centre, Qatar University, 2713 Doha, Qatar;
- College of Medicine, QU Health, Qatar University, 2713 Doha, Qatar;
| |
Collapse
|
42
|
Xiao S, Zhu H, Luo J, Wu Z, Xie M. miR‑425‑5p is associated with poor prognosis in patients with breast cancer and promotes cancer cell progression by targeting PTEN. Oncol Rep 2019; 42:2550-2560. [PMID: 31638259 PMCID: PMC6826330 DOI: 10.3892/or.2019.7371] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BC) is the most common invasive cancer in women, and it imposes a heavy burden on patients. microRNAs (miRNAs/miRs) have been found to play an important role in the development of tumors, but their role in the malignant progression of BC is unclear. In the present study, the expression level of miR‑425‑5p was examined in patients with BC, and its association with prognosis was investigated. In vitro experiments were performed to examine role of miR‑425‑5p in the development of BC cells. A downstream target gene of miR‑425‑5p was predicted using a miRNA target prediction tool and validated with a luciferase reporter assay. It was found that miR‑425‑5p expression was increased in BC tissues and cell lines, and was associated with tumor size, clinical stage, lymph node metastasis, distant metastasis and poor overall survival in patients with BC. Knockdown of miR‑425‑5p in BC cell lines inhibited proliferation and migration. PTEN was identified as a downstream target gene of miR‑425‑5p. Overexpression of PTEN was demonstrated to partially inhibit the promotional effect of miR‑425‑5p on cell proliferation and migration. Taken together, miR‑425‑5p is associated with poor prognosis, and promotes cell proliferation and migration via PTEN. Thus, miR‑425‑5p may serve as a therapeutic and prognostic marker for BC.
Collapse
Affiliation(s)
- Sheng Xiao
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Hongjia Zhu
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jian Luo
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Zhenru Wu
- Laboratory of Pathology, West China Hospital, Chengdu, Sichuan 610041, P.R. China
| | - Mingjun Xie
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|