51
|
Randolph K, Hyder U, D’Orso I. KAP1/TRIM28: Transcriptional Activator and/or Repressor of Viral and Cellular Programs? Front Cell Infect Microbiol 2022; 12:834636. [PMID: 35281453 PMCID: PMC8904932 DOI: 10.3389/fcimb.2022.834636] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/03/2022] [Indexed: 01/01/2023] Open
Abstract
Several transcriptional and epigenetic regulators have been functionally linked to the control of viral and cellular gene expression programs. One such regulator is Krüppel-associated box (KRAB)-associated protein 1 (KAP1: also named TRIM28 or TIF1β), which has been extensively studied in the past three decades. Here we offer an up-to date review of its various functions in a diversity of contexts. We first summarize the discovery of KAP1 repression of endogenous retroviruses during development. We then deliberate evidence in the literature suggesting KAP1 is both an activator and repressor of HIV-1 transcription and discuss experimental differences and limitations of previous studies. Finally, we discuss KAP1 regulation of DNA and RNA viruses, and then expand on KAP1 control of cellular responses and immune functions. While KAP1 positive and negative regulation of viral and cellular transcriptional programs is vastly documented, our mechanistic understanding remains narrow. We thus propose that precision genetic tools to reveal direct KAP1 functions in gene regulation will be required to not only illuminate new biology but also provide the foundation to translate the basic discoveries from the bench to the clinics.
Collapse
|
52
|
An Y, Li X, Yao F, Duan J, Yang XD. Novel Complex of PD-L1 Aptamer and Albumin Enhances Antitumor Efficacy In Vivo. Molecules 2022; 27:1482. [PMID: 35268583 PMCID: PMC8911819 DOI: 10.3390/molecules27051482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 11/17/2022] Open
Abstract
The PD-1/PD-L1 pathway blockade can generate a good clinical response by reducing immunosuppression and provoking durable antitumor immunity. In addition to antibodies, aptamers can also block the interaction between PD-1 and PD-L1. For the in vivo application, however, free aptamers are usually too small in size and quickly removed from blood via glomerular filtration. To avoid renal clearance of aptamer, we conjugated the PD-L1 aptamer to albumin to form a larger complex (BSA-Apt) and evaluated whether BSA-Apt would enhance the in vivo antitumor efficacy. The PD-L1 aptamer was thiol-modified and conjugated to the amino group of BSA via a SMCC linker. The average size of BSA-Apt was 11.65 nm, which was above the threshold for renal clearance. Functionally, BSA-Apt retained the capability of the PD-L1 aptamer to bind with PDL1-expressing tumor cells. Moreover, both the free aptamer and BSA-Apt augmented the PBMC-induced antitumor cytotoxicity in vitro. Furthermore, BSA-Apt generated a significantly stronger antitumor efficacy than the free PD-L1 aptamer in vivo without raising systemic toxicity. The results indicate that conjugating the PD-L1 aptamer to albumin may serve as a promising strategy to improve the in vivo functionality of the aptamer and that BSA-Apt may have application potential in cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Xian-Da Yang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; (Y.A.); (X.L.); (F.Y.); (J.D.)
| |
Collapse
|
53
|
Sirasanagandla SR, Sofin RS, Al-Huseini I, Das S. Role of Bisphenol A in Autophagy Modulation: Understanding the Molecular Concepts and Therapeutic Options. Mini Rev Med Chem 2022; 22:2213-2223. [DOI: 10.2174/1389557522666220214094055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/05/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022]
Abstract
Abstract:
Bisphenol A (4,4′-isopropylidenediphenol) is an organic compound, commonly used in the plastic bottles, packaging containers, beverages and resin industry. The adverse effects of bisphenol A were studied in various systems of the body. Autophagy is a lysosomal degradation process meant for the regeneration of new cells. The role of bisphenol A on autophagy modulation in the pathogenesis of diseases is still debatable. Few research studies showed that bisphenol A-induced adverse effects were associated with autophagy dysregulation, while few showed the activation of autophagy by bisphenol A. Such contrasting views make the subject more interesting and debatable. In the present review, we discuss the different steps of autophagy, genes involved, and the effect of bisphenol A in autophagy modulation on different systems of the body. We also discuss the methods for monitoring autophagy and the roles of drugs such as chloroquine, verteporfin, and rapamycin in autophagy. Proper understanding of the role of bisphenol A in the modulation of autophagy may be important for future treatment and drug discovery.
Collapse
Affiliation(s)
- Srinivasa Rao Sirasanagandla
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Al-Khodh 123, Oman
| | - R.G. Sumesh Sofin
- Department of Physics, College of Science, Sultan Qaboos University, Muscat, Al-Khodh 123, Oman
| | - Isehaq Al-Huseini
- Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Al-Khodh 123, Oman
| | - Srijit Das
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Al-Khodh 123, Oman
| |
Collapse
|
54
|
Yamaguchi H, Hsu JM, Yang WH, Hung MC. Mechanisms regulating PD-L1 expression in cancers and associated opportunities for novel small-molecule therapeutics. Nat Rev Clin Oncol 2022; 19:287-305. [DOI: 10.1038/s41571-022-00601-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 02/06/2023]
|
55
|
Read RD. Repurposing the drug verteporfin as anti-neoplastic therapy for glioblastoma. Neuro Oncol 2022; 24:708-710. [PMID: 35100422 PMCID: PMC9071275 DOI: 10.1093/neuonc/noac019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Renee D Read
- Corresponding Author: Renee D. Read, PhD, Department of Pharmacology and Chemical Biology, Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA ()
| |
Collapse
|
56
|
Wang J, Xu Y, Rao X, Zhang R, Tang J, Zhang D, Jie X, Zhu K, Wang X, Xu Y, Zhang S, Dong X, Zhang T, Yang K, Xu S, Meng R, Wu G. BRD4-IRF1 axis regulates chemoradiotherapy-induced PD-L1 expression and immune evasion in non-small cell lung cancer. Clin Transl Med 2022; 12:e718. [PMID: 35083874 PMCID: PMC8792480 DOI: 10.1002/ctm2.718] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 01/07/2022] [Accepted: 01/13/2022] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Chemoradiotherapy-induced PD-L1 upregulation leads to therapeutic resistance and treatment failure. The PD-1/PD-L1 blocking antibodies sensitize cancers to chemoradiotherapy by blocking extracellular PD-1 and PD-L1 binding without affecting the oncogenic function of intracellular PD-L1. Reversing the chemoradiation-induced PD-L1 expression could provide a new strategy to achieve a greater anti-tumour effect of chemoradiotherapy. Here, we aimed to identify candidate small molecular inhibitors that might boost the anti-tumour immunity of chemoradiotherapy by decreasing treatment-induced PD-L1 expression in non-small cell lung cancer (NSCLC). METHODS A drug array was used to recognize compounds that can suppress the cisplatin-induced and radiation-induced PD-L1 expression in NSCLC via the flow cytometry-based assay. We examined whether and how targeting bromodomain containing 4 (BRD4) inhibits chemoradiation-induced PD-L1 expression and evaluated the effect of BRD4 inhibition and chemoradiation combination in vivo. RESULTS BRD4 inhibitors JQ1 and ARV-771 were identified as the most promising drugs both in the cisplatin and radiation screening projects in two NSCLC cell lines. Targeting BRD4 was supposed to block chemoradiotherapy inducible PD-L1 expression by disrupting the recruitment of BRD4-IRF1 complex to PD-L1 promoter. A positive correlation between BRD4 and PD-L1 expression was observed in human NSCLC tissues. Moreover, BRD4 inhibition synergized with chemoradiotherapy and PD-1 blockade to show a robust anti-tumour immunity dependent on CD8+ T cell through limiting chemoradiation-induced tumour cell surface PD-L1 upregulation in vivo. Notably, the BRD4-targeted combinatory treatments did not show increased toxicities. CONCLUSION The data showed that BRD4-targeted therapy synergized with chemoradiotherapy and anti-PD-1 antibody by boosting anti-tumour immunity in NSCLC.
Collapse
Affiliation(s)
- Jian Wang
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Yingzhuo Xu
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Xinrui Rao
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Ruiguang Zhang
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Jing Tang
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Dan Zhang
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Xiaohua Jie
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Kuikui Zhu
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Xu Wang
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Yunhong Xu
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Sheng Zhang
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Xiaorong Dong
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Tao Zhang
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Kunyu Yang
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Shuangbing Xu
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Rui Meng
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| | - Gang Wu
- Cancer Center, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhan430022China
| |
Collapse
|
57
|
Gultekin O, Gonzalez-Molina J, Hardell E, Moyano-Galceran L, Mitsios N, Mulder J, Kokaraki G, Isaksson A, Sarhan D, Lehti K, Carlson JW. FOXP3+ T cells in uterine sarcomas are associated with favorable prognosis, low extracellular matrix expression and reduced YAP activation. NPJ Precis Oncol 2021; 5:97. [PMID: 34799669 PMCID: PMC8604926 DOI: 10.1038/s41698-021-00236-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 09/22/2021] [Indexed: 02/03/2023] Open
Abstract
Uterine sarcomas are rare but deadly malignancies without effective treatment. Immunotherapy is a promising new approach to treat these tumors but has shown heterogeneous effects in sarcoma patients. With the goal of identifying key factors for improved patient treatment, we characterized the tumor immune landscape in 58 uterine sarcoma cases with full clinicopathological annotation. Immune cell characterization revealed the overall prevalence of FOXP3+ cells and pro-tumor M2-like macrophages. Hierarchical clustering of patients showed four tumor type-independent immune signatures, where infiltration of FOXP3+ cells and M1-like macrophages associated with favorable prognosis. High CD8+/FOXP3+ ratio in UUS and ESS correlated with poor survival, upregulation of immunosuppressive markers, extracellular matrix (ECM)-related genes and proteins, and YAP activation. This study shows that uterine sarcomas present distinct immune signatures with prognostic value, independent of tumor type, and suggests that targeting the ECM could be beneficial for future treatments.
Collapse
Affiliation(s)
- Okan Gultekin
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jordi Gonzalez-Molina
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Elin Hardell
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Lidia Moyano-Galceran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Nicholas Mitsios
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jan Mulder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Georgia Kokaraki
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Isaksson
- Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Dhifaf Sarhan
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Joseph W Carlson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden. .,Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden. .,Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
58
|
Gao L, Chen Y. Autophagy controls programmed death-ligand 1 expression on cancer cells (Review). Biomed Rep 2021; 15:84. [PMID: 34512972 DOI: 10.3892/br.2021.1460] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/29/2021] [Indexed: 12/17/2022] Open
Abstract
Programmed death-ligand 1 (PD-L1) is a transmembrane protein mainly located on cancer cells, including renal cell carcinoma, breast, colorectal, gastric and non-small cell lung cancer. PD-L1 binds to the PD-1 receptor expressed on T lymphocytes to inhibit the activation of T lymphocytes, thus allowing tumour cells to escape immune surveillance, leading to tumour growth and the poor prognosis of patients with cancer. Inhibitors targeting the programmed death-1/PD-L1 axis have been widely used in the clinical treatment of a variety of solid tumours in recent years. However, the clinical efficacy of these inhibitors varies. Studies have demonstrated that the effect of the targeted drug is positively associated with the expression of PD-L1 on the tumour membrane. Hence, exploring the mechanism of PD-L1 expression is very important for the treatment of tumours. Autophagy is a physiological process that maintains the stability of the internal environment. Autophagy degrades aging organelles and long-lived proteins and produces nutrients for cell recycling. To the best of our knowledge, the present review is the first to summarize the research that has been conducted on autophagy-regulated PD-L1 expression, which may provide new avenues for tumour immunotherapy.
Collapse
Affiliation(s)
- Lijuan Gao
- Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,The First Clinical College of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yongshun Chen
- Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,The First Clinical College of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
59
|
Duan Y, Tian X, Liu Q, Jin J, Shi J, Hou Y. Role of autophagy on cancer immune escape. Cell Commun Signal 2021; 19:91. [PMID: 34493296 PMCID: PMC8424925 DOI: 10.1186/s12964-021-00769-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/24/2021] [Indexed: 01/15/2023] Open
Abstract
Autophagy is catabolic process by degradation of intracellular components in lysosome including proteins, lipids, and mitochondria in response to nutrient deficiency or stress such as hypoxia or chemotherapy. Increasing evidence suggests that autophagy could induce immune checkpoint proteins (PD-L1, MHC-I/II) degradation of cancer cells, which play an important role in regulating cancer cell immune escape. In addition to autophagic degradation of immune checkpoint proteins, autophagy induction in immune cells (macrophages, dendritic cells) manipulates antigen presentation and T cell activity. These reports suggest that autophagy could negatively or positively regulate cancer cell immune escape by immune checkpoint protein and antigens degradation, cytokines release, antigens generation. These controversial phenomenon of autophagy on cancer cell immune evasion may be derived from different experimental context or models. In addition, autophagy maybe exhibit a role in regulating host excessive immune response. So rational combination with autophagy could enhance the efficacy of cancer immunotherapy. In this review, the current progress of autophagy on cancer immune escape is discussed. Video Abstract
Collapse
Affiliation(s)
- Yalan Duan
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University, Changzhou, 213017, Jiangsu Province, China.,School of Life Sciences, Jiangsu University, Zhenjiang, 213017, Jiangsu Province, China
| | - Xiaoqing Tian
- School of Life Sciences, Jiangsu University, Zhenjiang, 213017, Jiangsu Province, China
| | - Qian Liu
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University, Changzhou, 213017, Jiangsu Province, China.,Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, 213017, Jiangsu Province, China
| | - Jianhua Jin
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University, Changzhou, 213017, Jiangsu Province, China.,Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, 213017, Jiangsu Province, China
| | - Juanjuan Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, 213017, Jiangsu Province, China
| | - Yongzhong Hou
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University, Changzhou, 213017, Jiangsu Province, China. .,School of Life Sciences, Jiangsu University, Zhenjiang, 213017, Jiangsu Province, China.
| |
Collapse
|
60
|
Yuan L, Zhang X, Cheng K, Li L, Guo Z, Zeng L. IRF1 Inhibits Autophagy-Mediated Proliferation of Colorectal Cancer via Targeting ATG13. Cancer Invest 2021; 40:35-45. [PMID: 34313498 DOI: 10.1080/07357907.2021.1961265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
IRF1 is a nuclear transcription factor that mediates interferon effects and appears to have anti-tumor activity. To determine the roles of IRF1 in colorectal cancer (CRC), we investigated the effects of IRF1 in CRC cells. We found that IRF1 inhibit cell proliferation and tumor growth. Under starvation conditions, IRF1 enhanced apoptosis and reduced autophagic flux. ATG13, an important factor of autophagy complex, was confirmed as a target of IRF1. These findings indicated that IRF1 function as a tumor suppressor in CRC and inhibit autophagy through ATG13, targeting this pathway may provide new insights into the molecular mechanisms of CRC progression.
Collapse
Affiliation(s)
- Li Yuan
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiao Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kai Cheng
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Liping Li
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Zhongming Guo
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Liang Zeng
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
61
|
New Insights into the Clinical Implications of Yes-Associated Protein in Lung Cancer: Roles in Drug Resistance, Tumor Immunity, Autophagy, and Organoid Development. Cancers (Basel) 2021; 13:cancers13123069. [PMID: 34202980 PMCID: PMC8234989 DOI: 10.3390/cancers13123069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/09/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Innovative advancements in lung cancer treatment have developed over the past decade with the advent of targeted and immune therapies. Yes-associated protein (YAP), an effector of the Hippo pathway, promotes the resistance of these targeted drugs and modulates tumor immunity in lung cancer. YAP is involved in autophagy in lung cancer and plays a prominent role in forming the tubular structure in lung organoids and alveolar differentiation. In this review, we discuss the central roles of YAP in lung cancer and present YAP as a novel target for treating resistance to targeted therapies and immunotherapies in lung cancer. Abstract Despite significant innovations in lung cancer treatment, such as targeted therapy and immunotherapy, lung cancer is still the principal cause of cancer-associated death. Novel strategies to overcome drug resistance and inhibit metastasis in cancer are urgently needed. The Hippo pathway and its effector, Yes-associated protein (YAP), play crucial roles in lung development and alveolar differentiation. YAP is known to mediate mechanotransduction, an important process in lung homeostasis and fibrosis. In lung cancer, YAP promotes metastasis and confers resistance against chemotherapeutic drugs and targeted agents. Recent studies revealed that YAP directly controls the expression of programmed death-ligand 1 (PD-L1) and modulates the tumor microenvironment (TME). YAP not only has a profound relationship with autophagy in lung cancer but also controls alveolar differentiation, and is responsible for tubular structure formation in lung organoids. In this review, we discuss the various roles and clinical implications of YAP in lung cancer and propose that targeting YAP can be a promising strategy for treating lung cancer.
Collapse
|
62
|
Zhang H, Dai Z, Wu W, Wang Z, Zhang N, Zhang L, Zeng WJ, Liu Z, Cheng Q. Regulatory mechanisms of immune checkpoints PD-L1 and CTLA-4 in cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:184. [PMID: 34088360 PMCID: PMC8178863 DOI: 10.1186/s13046-021-01987-7] [Citation(s) in RCA: 235] [Impact Index Per Article: 78.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/17/2021] [Indexed: 02/01/2023]
Abstract
The cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4)/B7 and programmed death 1 (PD-1)/ programmed cell death-ligand 1 (PD-L1) are two most representative immune checkpoint pathways, which negatively regulate T cell immune function during different phases of T-cell activation. Inhibitors targeting CTLA-4/B7 and PD1/PD-L1 pathways have revolutionized immunotherapies for numerous cancer types. Although the combined anti-CTLA-4/B7 and anti-PD1/PD-L1 therapy has demonstrated promising clinical efficacy, only a small percentage of patients receiving anti-CTLA-4/B7 or anti-PD1/PD-L1 therapy experienced prolonged survival. Regulation of the expression of PD-L1 and CTLA-4 significantly impacts the treatment effect. Understanding the in-depth mechanisms and interplays of PD-L1 and CTLA-4 could help identify patients with better immunotherapy responses and promote their clinical care. In this review, regulation of PD-L1 and CTLA-4 is discussed at the levels of DNA, RNA, and proteins, as well as indirect regulation of biomarkers, localization within the cell, and drugs. Specifically, some potential drugs have been developed to regulate PD-L1 and CTLA-4 expressions with high efficiency.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Nan Zhang
- One-third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Jing Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
63
|
Blanchard L, Girard JP. High endothelial venules (HEVs) in immunity, inflammation and cancer. Angiogenesis 2021; 24:719-753. [PMID: 33956259 PMCID: PMC8487881 DOI: 10.1007/s10456-021-09792-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022]
Abstract
High endothelial venules (HEVs) are specialized blood vessels mediating lymphocyte trafficking to lymph nodes (LNs) and other secondary lymphoid organs. By supporting high levels of lymphocyte extravasation from the blood, HEVs play an essential role in lymphocyte recirculation and immune surveillance for foreign invaders (bacterial and viral infections) and alterations in the body’s own cells (neoantigens in cancer). The HEV network expands during inflammation in immune-stimulated LNs and is profoundly remodeled in metastatic and tumor-draining LNs. HEV-like blood vessels expressing high levels of the HEV-specific sulfated MECA-79 antigens are induced in non-lymphoid tissues at sites of chronic inflammation in many human inflammatory and allergic diseases, including rheumatoid arthritis, Crohn’s disease, allergic rhinitis and asthma. Such vessels are believed to contribute to the amplification and maintenance of chronic inflammation. MECA-79+ tumor-associated HEVs (TA-HEVs) are frequently found in human tumors in CD3+ T cell-rich areas or CD20+ B-cell rich tertiary lymphoid structures (TLSs). TA-HEVs have been proposed to play important roles in lymphocyte entry into tumors, a process essential for successful antitumor immunity and lymphocyte-mediated cancer immunotherapy with immune checkpoint inhibitors, vaccines or adoptive T cell therapy. In this review, we highlight the phenotype and function of HEVs in homeostatic, inflamed and tumor-draining lymph nodes, and those of HEV-like blood vessels in chronic inflammatory diseases. Furthermore, we discuss the role and regulation of TA-HEVs in human cancer and mouse tumor models.
Collapse
Affiliation(s)
- Lucas Blanchard
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
64
|
Zhao G, Liu C, Wen X, Luan G, Xie L, Guo X. The translational values of TRIM family in pan-cancers: From functions and mechanisms to clinics. Pharmacol Ther 2021; 227:107881. [PMID: 33930453 DOI: 10.1016/j.pharmthera.2021.107881] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/08/2023]
Abstract
Cancer is the second leading cause of human death across the world. Tripartite motif (TRIM) family, with E3 ubiquitin ligase activities in majority of its members, is reported to be involved in multiple cellular processes and signaling pathways. TRIM proteins have critical effects in the regulation of biological behaviors of cancer cells. Here, we discussed the current understanding of the molecular mechanism of TRIM proteins regulation of cancer cells. We also comprehensively reviewed published studies on TRIM family members as oncogenes or tumor suppressors in the oncogenesis, development, and progression of a variety of types of human cancers. Finally, we highlighted that certain TRIM family members are potential molecular biomarkers for cancer diagnosis and prognosis, and potential therapeutic targets.
Collapse
Affiliation(s)
- Guo Zhao
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Chuan Liu
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Xin Wen
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Gan Luan
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Longxiang Xie
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Xiangqian Guo
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| |
Collapse
|
65
|
Jiang T, Chen X, Ren X, Yang JM, Cheng Y. Emerging role of autophagy in anti-tumor immunity: Implications for the modulation of immunotherapy resistance. Drug Resist Updat 2021; 56:100752. [PMID: 33765484 DOI: 10.1016/j.drup.2021.100752] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023]
Abstract
Immunotherapies such as CAR-T cell transfer and antibody-targeted therapy have produced promising clinical outcomes in patients with advanced and metastatic cancer that are resistant to conventional therapies. However, with increasing use of cancer immunotherapy in clinical treatment, multiple therapy-resistance mechanisms have gradually emerged. The tumor microenvironment (TME), an integral component of cancer, can significantly influence the therapeutic response. Thus, it is worth exploring the potential of TME in modulating therapy resistance, in the hope to devise novel strategies to reinforcing anti-cancer treatments such as immunotherapy. As a crucial recycling process in the complex TME, the role of autophagy in tumor immunity has been increasingly appreciated. Firstly, autophagy in tumor cells can affect their immune response through modulating MHC-I-antigen complexes, thus modulating immunogenic tumor cell death, changing functions of immune cells via secretory autophagy, reducing the NK- and CTL-mediated cell lysis and degradation of immune checkpoint proteins. Secondly, autophagy is critical for the differentiation, maturation and survival of immune cells in the TME and can significantly affect the immune function of these cells, thereby regulating the anti-tumor immune response. Thirdly, alteration of autophagic activity in stromal cells, especially in fibroblasts, can reconstruct the three-dimensional stromal environment and metabolic reprogramming in the TME. A number of studies have demonstrated that optimal induction or inhibition of autophagy may lead to effective therapeutic regimens when combined with immunotherapy. This review discusses the important roles of autophagy in tumor cells, immune cells and stromal cells in the context of tumor immunity, and the potential of combining the autophagy-based therapy with immunotherapy as novel therapeutic approaches against cancer.
Collapse
Affiliation(s)
- Ting Jiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xisha Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xingcong Ren
- Department of Toxicology and Cancer Biology, Department of Pharmacology, and Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Jin-Ming Yang
- Department of Toxicology and Cancer Biology, Department of Pharmacology, and Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA.
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
66
|
Yan Y, Zheng L, Du Q, Yazdani H, Dong K, Guo Y, Geller DA. Interferon regulatory factor 1(IRF-1) activates anti-tumor immunity via CXCL10/CXCR3 axis in hepatocellular carcinoma (HCC). Cancer Lett 2021; 506:95-106. [PMID: 33689775 DOI: 10.1016/j.canlet.2021.03.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023]
Abstract
Interferon regulatory factor 1 (IRF-1) is a tumor suppressor gene in cancer biology with anti-proliferative and pro-apoptotic effect on cancer cells, however mechanisms of IRF-1 regulating tumor microenvironment (TME) in hepatocellular carcinoma (HCC) remain only partially characterized. Here, we investigated that IRF-1 regulates C-X-C motif chemokine 10 (CXCL10) and chemokine receptor 3 (CXCR3) to activate anti-tumor immunity in HCC. We found that IRF-1 mRNA expression was positively correlated with CXCL10 and CXCR3 through qRT-PCR assay in HCC tumors and in analysis of the TCGA database. IRF-1 response elements were identified in the CXCL10 promoter region, and ChIP-qPCR confirmed IRF-1 binding to promote CXCL10 transcription. IRF-2 is a competitive antagonist for IRF-1 mediated transcriptional effects, and overexpression of IRF-2 decreased basal and IFN-γ induced CXCL10 expression. Although IRF-1 upregulated CXCR3 expression in HCC cells, it inhibited proliferation and exerted pro-apoptotic effects, which overcome proliferation partly mediated by activating the CXCL10/CXCR3 autocrine axis. In vitro and in vivo studies showed that IRF-1 increased CD8+ T cells, NK and NKT cells migration, and activated IFN-γ secretion in NK and NKT cells to induce tumor apoptosis through the CXCL10/CXCR3 paracrine axis. Conversely, this effect was markedly abrogated in HCC tumor bearing mice deficient in CXCR3. Therefore, the IRF-1/CXCL10/CXCR3 axis contributes to the anti-tumor microenvironment in HCC.
Collapse
Affiliation(s)
- Yihe Yan
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China; Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15260, USA.
| | - Leting Zheng
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15260, USA; Department of Rheumatology and Immunology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Qiang Du
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15260, USA
| | - Hamza Yazdani
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15260, USA
| | - Kun Dong
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15260, USA
| | - Yarong Guo
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15260, USA
| | - David A Geller
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
67
|
Cheng B, Xiao Y, Xue M, Cao H, Chen J. Recent Advances in the Development of PD-L1 Modulators: Degraders, Downregulators, and Covalent Inhibitors. J Med Chem 2020; 63:15389-15398. [PMID: 33272018 DOI: 10.1021/acs.jmedchem.0c01362] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Therapeutic interference of the programmed cell death protein 1(PD-1)/immunosuppressive programmed cell death ligand 1 (PD-L1) signaling pathway by monoclonal antibodies has achieved spectacular success for treating various tumors. However, the development of small molecule inhibitors of PD-1/PD-L1 has lagged far behind due to the challenge of targeting the highly hydrophobic and relatively flat binding interface, despite the benefits small molecule can bring over therapeutic antibodies. This technical challenge provokes the adoption of different strategies in searching for small, medium-sized, and large molecule modulators (e.g., degraders, downregulators, and covalent inhibitors) of the PD-1/PD-L1 protein-protein interaction. In this review article, we discuss latest advances in the development of PD-L1 modulators, with a focus on degraders, downregulators, and covalent inhibitors.
Collapse
Affiliation(s)
- Binbin Cheng
- Drug Design and Discovery Research Innovation Community, School of Pharmaceutical Sciences, Southern Medical University, Baiyun District, Guangzhou 510515, China
| | - Yao Xiao
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan Wuchang Hospital, Wuchang 430063, China
| | - Mingming Xue
- Tianjin Tiancheng Chemical Co., Ltd., Chemical Street, Binhai New District, Tianjin 300480, China
| | - Hao Cao
- Drug Design and Discovery Research Innovation Community, School of Pharmaceutical Sciences, Southern Medical University, Baiyun District, Guangzhou 510515, China
| | - Jianjun Chen
- Drug Design and Discovery Research Innovation Community, School of Pharmaceutical Sciences, Southern Medical University, Baiyun District, Guangzhou 510515, China
| |
Collapse
|
68
|
TRIM28 is a distinct prognostic biomarker that worsens the tumor immune microenvironment in lung adenocarcinoma. Aging (Albany NY) 2020; 12:20308-20331. [PMID: 33091876 PMCID: PMC7655206 DOI: 10.18632/aging.103804] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/09/2020] [Indexed: 12/26/2022]
Abstract
The tumor immune microenvironment (TIME) is an important determinant of cancer prognosis and treatment efficacy. To identify immune-related prognostic biomarkers of lung adenocarcinoma, we used the ESTIMATE algorithm to calculate the immune and stromal scores of 517 lung adenocarcinoma patients from The Cancer Genome Atlas (TCGA). We detected 985 differentially expressed genes (DEGs) between patients with high and low immune and stromal scores, and we analyzed their functions and protein-protein interactions. TRIM28 was upregulated in lung adenocarcinoma patients with low immune and stromal scores, and was associated with a poor prognosis. The TISIDB and TIMER databases indicated that TRIM28 expression correlated negatively with immune infiltration. We then explored genes that were co-expressed with TRIM28 in TCGA, and investigated DEGs based on TRIM28 expression in GSE43580 and GSE7670. The 429 common DEGs from these analyses were functionally analyzed. We also performed a Gene Set Enrichment Analysis using TCGA data, and predicted substrates of TRIM28 using UbiBrowser. The results indicated that TRIM28 may negatively regulate the TIME by increasing the SUMOylation of IRF5 and IRF8. Correlation analyses and validations in two lung adenocarcinoma cell lines (PC9 and H1299) confirmed these findings. Thus, TRIM28 may worsen the TIME and prognosis of lung adenocarcinoma.
Collapse
|
69
|
Spatial and Temporal Changes in PD-L1 Expression in Cancer: The Role of Genetic Drivers, Tumor Microenvironment and Resistance to Therapy. Int J Mol Sci 2020; 21:ijms21197139. [PMID: 32992658 PMCID: PMC7583014 DOI: 10.3390/ijms21197139] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
Immunotherapies blocking immune inhibitory receptors programmed cell death-1 (PD-1) and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) on T-cells have dramatically improved patient outcomes in a range of advanced cancers. However, the lack of response, and the development of resistance remain major obstacles to long-term improvements in patient outcomes. There is significant interest in the clinical use of biomarkers to improve patient selection, and the expression of PD-1 ligand 1 (PD-L1) is often reported as a potential biomarker of response. However, accumulating evidence suggests that the predictive value of PD-L1 expression in tumor biopsies is relatively low due, in part, to its complex biology. In this review, we discuss the biological consequences of PD-L1 expression by various cell types within the tumor microenvironment, and the complex mechanisms that regulate PD-L1 expression at the genomic, transcriptomic and proteomic levels.
Collapse
|