1
|
Xie H, Wei L, Tang S, Gan J. Serum albumin-carcinoembryonic antigen ratio as an effective clinical tool for predicting recurrence and overall survival in patients with rectal cancer. Front Nutr 2025; 11:1521691. [PMID: 39897534 PMCID: PMC11782030 DOI: 10.3389/fnut.2024.1521691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025] Open
Abstract
Background The albumin-carcinoembryonic antigen ratio (ACR), leveraging the strengths of albumin and CEA, has emerged as a promising serum prognostic marker. However, no studies to date have explored the association between ACR and the prognosis of patients with rectal cancer. This study aimed to determine the value of albumin-carcinoembryonic antigen ratio (ACR) in predicting the progression-free survival (PFS) and overall survival (OS) of patients with rectal cancer. Methods Survival analysis was conducted using the Kaplan-Meier method, and hazard ratios (HR) were calculated using Cox regression analyses. Nomograms were created based on variables with p < 0.05 in the multivariate Cox regression analysis. The predictive ability of the model was evaluated using the C-index and calibration curve, and its prognostic predictive abilities were compared to those of traditional Tumor Node Metastasis (TNM) stage using discriminant indices. Results A total of 736 patients with rectal cancer were included in the study. ACR was significantly higher in patients with poor survival or cancer recurrence. A low ACR was associated with increased tumor invasiveness, longer hospital stays, and higher hospitalization costs. Patients with a high ACR had significantly better PFS (62.9% vs. 35.2%, p < 0.001) and OS (67.0% vs. 37.2%, p < 0.001) than those with a low ACR. ACR can serve as an effective auxiliary tool for pathological staging, especially in patients with stage III-IV disease. The relationship between ACR and mortality risk was L-shaped. ACR is an independent prognostic factor for PFS [HR = 0.581, 95% confidence interval (CI): 0.458-0.738, p < 0.001] and OS (HR = 0.560, 95% CI: 0.435-0.720, p < 0.001) in rectal cancer patients. ACR-based nomograms have good predictive accuracy and outperform traditional TNM stage in predicting prognosis. Conclusion Albumin-carcinoembryonic antigen ratio is a simple and effective clinical tool for predicting the recurrence and survival of patients with rectal cancer and is a useful supplement to the TNM stage.
Collapse
Affiliation(s)
- Hailun Xie
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, Guangxi, China
| | - Lishuang Wei
- Department of Geriatric Respiratory Disease Ward, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Shuangyi Tang
- Department of Pharmacy, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Jialiang Gan
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, Guangxi, China
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
2
|
Bahrami A, Khalaji A, Bahri Najafi M, Sadati S, Raisi A, Abolhassani A, Eshraghi R, Khaksary Mahabady M, Rahimian N, Mirzaei H. NF-κB pathway and angiogenesis: insights into colorectal cancer development and therapeutic targets. Eur J Med Res 2024; 29:610. [PMID: 39702532 DOI: 10.1186/s40001-024-02168-w] [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: 06/11/2024] [Accepted: 11/21/2024] [Indexed: 12/21/2024] Open
Abstract
Colorectal cancer (CRC) is currently ranked as the third most common type of cancer, contributing significantly to mortality and morbidity worldwide. Epigenetic and genetic changes occurred during CRC progression resulted in the cell proliferation, cancer progression, angiogenesis, and invasion. Angiogenesis is one of the crucial steps during cancer progression required for the delivery of essential nutrients to cancer cells and removes metabolic waste. During angiogenesis, different molecules are secreted from tumoral cells to trigger vascular formation including epidermal growth factor and the vascular endothelial growth factor (VEGF). The production and regulation of the secretion of these molecules are modulated by different subcellular pathways such as NF-κB. NF-κB is involved in regulation of different homeostatic pathways including apoptosis, cell proliferation, inflammation, differentiation, tumor migration, and angiogenesis. Investigation of different aspects of this pathway and its role in angiogenesis could provide a comprehensive overview about the underlying mechanisms and could be used for development of further therapeutic targets. In this review of literature, we comprehensively reviewed the current understanding and potential of NF-κB-related angiogenesis in CRC. Moreover, we explored the treatments that are based on the NF-κB pathway.
Collapse
Affiliation(s)
- Ashkan Bahrami
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirreza Khalaji
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majed Bahri Najafi
- Applied Physiology Research Center, Cardiovascular Research Institute, Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sina Sadati
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Arash Raisi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Reza Eshraghi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Neda Rahimian
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
3
|
Zhu Y, Ke L, Wei Y, Zheng X. A High-Precision Real-Time Temperature Acquisition Method Based on Magnetic Nanoparticles. SENSORS (BASEL, SWITZERLAND) 2024; 24:7716. [PMID: 39686253 DOI: 10.3390/s24237716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024]
Abstract
The unique magnetothermal properties of magnetic nanoparticles enable the development of a high-precision, real-time, noninvasive temperature measurement method with significant potential in the biomedical field. Based on a low-frequency alternating magnetic field excitation model, we construct two additional magnetic field excitation models-alternating current-direct current superposition and dual-frequency superposition-to extract harmonic amplitude components from the magnetization response. To increase the accuracy of harmonic information acquisition, the effects of the truncation error, excitation magnetic field frequency, and amplitude are thoroughly analyzed, and optimal parameter values are selected to minimize the error. A single algorithm is designed for temperature inversion, and a joint algorithm is proposed to optimize the performance of the single algorithm. Under low-frequency alternating-current magnetic field excitation, the autonomous group particle swarm optimization method achieves superior real-time performance in terms of temperature inversion and running time. Compared with the opposition learning gray wolf optimizer and particle swarm optimization-gray wolf optimization, the proposed method achieves reductions of 52% and 68%, respectively. Additionally, under dual-frequency superimposed magnetic field excitation, a higher temperature inversion accuracy is achieved compared with that of the particle swarm optimization-gray wolf optimization algorithm, reducing the error from 0.237 K to 0.094 K.
Collapse
Affiliation(s)
- Yuchang Zhu
- School of Electrical Engineering, Shenyang University of Technology, Shenyang 110870, China
| | - Li Ke
- School of Electrical Engineering, Shenyang University of Technology, Shenyang 110870, China
| | - Yijing Wei
- School of Electrical Engineering, Shenyang University of Technology, Shenyang 110870, China
| | - Xiao Zheng
- CAS State Key Laboratory of Forest and Management, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China
| |
Collapse
|
4
|
Hameedat F, Mendes BB, Conniot J, Di Filippo LD, Chorilli M, Schroeder A, Conde J, Sousa F. Engineering nanomaterials for glioblastoma nanovaccination. NATURE REVIEWS MATERIALS 2024; 9:628-642. [DOI: 10.1038/s41578-024-00684-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 01/03/2025]
|
5
|
Xie H, Jia P, Wei L, Ruan G, Zhang H, Ge Y, Lin S, Song M, Wang Z, Liu C, Shi J, Liu X, Yang M, Zheng X, Chen Y, Zhang X, Shi H. Evaluation and validation of neutrophil to albumin ratio as a promising prognostic marker for all-cause mortality in patients with cancer: a multicenter cohort study. Nutrition 2024; 121:112365. [PMID: 38377700 DOI: 10.1016/j.nut.2024.112365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/31/2023] [Accepted: 01/21/2024] [Indexed: 02/22/2024]
Abstract
OBJECTIVES The practicality and effectiveness of using the prognostic value of the neutrophil-to-albumin ratio (NAR) in evaluating patients with cancer remain unclear, and research is needed to fully understand its potential application in the cancer population. METHODS The Kaplan-Meier method was used for survival analysis, and the log-rank test was employed for comparison. Univariate and multivariate Cox proportional hazards models were used to determine the prognostic biomarkers, and Logistic regression analysis was conducted to investigate the relationship between NAR and 90-day outcomes and cachexia. RESULTS The study included 14 682 patients with cancer, divided into discovery (6592 patients), internal validation (2820 patients), and external validation groups (5270 patients). Patients with high NAR had higher all-cause mortality than those with low NAR in the discovery (50.15% versus 69.29%, P < 0.001), internal validation (54.18% versus 70.91%, P < 0.001), and external validation cohorts (40.60% versus 66.68%, P < 0.001). In the discovery cohort, high NAR was observed to be independently associated with all-cause mortality in patients (HR 1.16, 95% CI 1.12-1.19; P < 0.001). Moreover, we validated the promising prognostic value of NAR as a predictor of survival in patients with cancer through internal validation (HR 1.21, 95% CI 1.16-1.27, P < 0.001) and external validation cohorts (HR 1.27, 95% CI 1.21-1.34, P < 0.001). Additionally, in the subgroup analysis by tumor type, high NAR was identified as a risk factor for most cancers, except for breast cancer. CONCLUSIONS This study showed that NAR is a feasible and promising biomarker for predicting prognosis and cancer cachexia in cancer patients.
Collapse
Affiliation(s)
- Hailun Xie
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Pingping Jia
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Lishuang Wei
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Guotian Ruan
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Heyang Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Yizhong Ge
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Shiqi Lin
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Mengmeng Song
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Ziwen Wang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Chenan Liu
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Jinyu Shi
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Xiaoyue Liu
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Ming Yang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Xin Zheng
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Yue Chen
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Xiaowei Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Hanping Shi
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China.
| |
Collapse
|
6
|
Krishnamohan M, Kaplanov I, Maudi-Boker S, Yousef M, Machluf-Katz N, Cohen I, Elkabets M, Titus J, Bersudsky M, Apte RN, Voronov E, Braiman A. Tumor Cell-Associated IL-1α Affects Breast Cancer Progression and Metastasis in Mice through Manipulation of the Tumor Immune Microenvironment. Int J Mol Sci 2024; 25:3950. [PMID: 38612760 PMCID: PMC11011794 DOI: 10.3390/ijms25073950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
IL-1α is a dual function cytokine that affects inflammatory and immune responses and plays a pivotal role in cancer. The effects of intracellular IL-1α on the development of triple negative breast cancer (TNBC) in mice were assessed using the CRISPR/Cas9 system to suppress IL-1α expression in 4T1 breast cancer cells. Knockout of IL-1α in 4T1 cells modified expression of multiple genes, including downregulation of cytokines and chemokines involved in the recruitment of tumor-associated pro-inflammatory cells. Orthotopical injection of IL-1α knockout (KO) 4T1 cells into BALB/c mice led to a significant decrease in local tumor growth and lung metastases, compared to injection of wild-type 4T1 (4T1/WT) cells. Neutrophils and myeloid-derived suppressor cells were abundant in tumors developing after injection of 4T1/WT cells, whereas more antigen-presenting cells were observed in the tumor microenvironment after injection of IL-1α KO 4T1 cells. This switch correlated with increased infiltration of CD3+CD8+ and NKp46+cells. Engraftment of IL-1α knockout 4T1 cells into immunodeficient NOD.SCID mice resulted in more rapid tumor growth, with increased lung metastasis in comparison to engraftment of 4T1/WT cells. Our results suggest that tumor-associated IL-1α is involved in TNBC progression in mice by modulating the interplay between immunosuppressive pro-inflammatory cells vs. antigen-presenting and cytotoxic cells.
Collapse
Affiliation(s)
- Mathumathi Krishnamohan
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Irena Kaplanov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Sapir Maudi-Boker
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Muhammad Yousef
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Noy Machluf-Katz
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Idan Cohen
- Cancer Center, Emek Medical Center, Afula 18101, Israel;
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Jaison Titus
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Marina Bersudsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Ron N. Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| |
Collapse
|
7
|
Iyer M, Ravichandran N, Karuppusamy PA, Gnanarajan R, Yadav MK, Narayanasamy A, Vellingiri B. Molecular insights and promise of oncolytic virus based immunotherapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:419-492. [PMID: 38762277 DOI: 10.1016/bs.apcsb.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Discovering a therapeutic that can counteract the aggressiveness of this disease's mechanism is crucial for improving survival rates for cancer patients and for better understanding the most different types of cancer. In recent years, using these viruses as an anticancer therapy has been thought to be successful. They mostly work by directly destroying cancer cells, activating the immune system to fight cancer, and expressing exogenous effector genes. For the treatment of tumors, oncolytic viruses (OVs), which can be modified to reproduce only in tumor tissues and lyse them while preserving the healthy non-neoplastic host cells and reinstating antitumor immunity which present a novel immunotherapeutic strategy. OVs can exist naturally or be created in a lab by altering existing viruses. These changes heralded the beginning of a new era of less harmful virus-based cancer therapy. We discuss three different types of oncolytic viruses that have already received regulatory approval to treat cancer as well as clinical research using oncolytic adenoviruses. The primary therapeutic applications, mechanism of action of oncolytic virus updates, future views of this therapy will be covered in this chapter.
Collapse
Affiliation(s)
- Mahalaxmi Iyer
- Department of Microbiology, Central University of Punjab, Bathinda, India
| | - Nandita Ravichandran
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | | | - Roselin Gnanarajan
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Mukesh Kumar Yadav
- Department of Microbiology, Central University of Punjab, Bathinda, India
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India.
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
8
|
Heshmat-Ghahdarijani K, Sarmadi V, Heidari A, Falahati Marvasti A, Neshat S, Raeisi S. The neutrophil-to-lymphocyte ratio as a new prognostic factor in cancers: a narrative review. Front Oncol 2023; 13:1228076. [PMID: 37860198 PMCID: PMC10583548 DOI: 10.3389/fonc.2023.1228076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
The increasing incidence of cancer globally has highlighted the significance of early diagnosis and improvement of treatment strategies. In the 19th century, a connection was made between inflammation and cancer, with inflammation recognized as a malignancy hallmark. The neutrophil-to-lymphocyte ratio (NLR), calculated from a complete blood count, is a simple and accessible biomarker of inflammation status. NLR has also been proven to be a prognostic factor for various medical conditions, including mortality classification in cardiac patients, infectious diseases, postoperative complications, and inflammatory states. In this narrative review, we aim to assess the prognostic potential of NLR in cancer. We will review recent studies that have evaluated the association between NLR and various malignancies. The results of this review will help to further understand the role of NLR in cancer prognosis and inform future research directions. With the increasing incidence of cancer, it is important to identify reliable and accessible prognostic markers to improve patient outcomes. The study of NLR in cancer may provide valuable insights into the development and progression of cancer and inform clinical decision-making.
Collapse
Affiliation(s)
- Kian Heshmat-Ghahdarijani
- Cardiac Rehabilitation, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vida Sarmadi
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Afshin Heidari
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Sina Neshat
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Biostatistics and Epidemiology, University of California, San Francisco, San Francisco, CA, United States
| | - Sina Raeisi
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
9
|
Dhanusu Sivakalai S, Sowndhar Rajan B, Vellaichamy E. C-type natriuretic peptide (CNP) inhibits 7,12-Dimethylbenz[a]anthracene (DMBA)/Croton oil-induced skin tumor growth by modulating inflammation in Swiss albino mice. J Biochem Mol Toxicol 2023; 37:e23423. [PMID: 37352108 DOI: 10.1002/jbt.23423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 04/14/2023] [Accepted: 06/12/2023] [Indexed: 06/25/2023]
Abstract
C-type natriuretic peptide (CNP) exhibits anti-inflammatory activity besides its natriuretic and diuretic functions. The present study aimed to determine the anticancer and synergistic therapeutic activity of CNP against a 7,12-Dimethylbenz[a]anthracene (DMBA)/Croton oil-induced skin tumor mouse model. CNP (2.5 µg/kg body weight) was injected either alone and/or in combination with Cisplatin (CDDP) (2 mg/kg body weight) for 4 weeks. The dorsal skin tumor incidences/growth and mortality rate were recorded during the experimental period of 16 weeks. The serum C-reactive protein (CRP), and lactate dehydrogenase (LDH) levels, infiltrating mast cells, and AgNORs proliferating cells count were analyzed in control and experimental mice. Further, the expression profile of marker genes of proliferation, inflammation, and progression molecules were analyzed using Reverse transcriptase-polymerase chain reaction (RT-PCR)/quantitative PCR (qPCR), western blot, and immunohistochemistry. The DMBA/Croton oil-induced mice exhibited 100% tumor incidence. Whereas, CNP alone, CDDP alone, and CNP+CDDP combination-treated mice exhibited 58%, 46%, and 24% tumor incidence, respectively. Also, a marked reduction in the levels of serum CRP and LDH, the number of infiltrating mast cells count and AgNORs proliferating cells count were noticed in the mice skin sections. Further, a significant reduction in both mRNA and protein expression levels of proliferation, inflammation, and progression markers were noticed in CNP (p < 0.01), CDDP (p < 0.01), and CNP+CDDP combination (p < 0.001) treated mice, respectively. The results of the present study suggest that CNP has anticancer activity. Further, the CNP+CDDP treatment has more promising anticancer activity as compared with CNP or CDDP alone treatment, probably due to the synergistic antiproliferative and anti-inflammatory activities of CNP and CDDP.
Collapse
Affiliation(s)
- Suresh Dhanusu Sivakalai
- Peptide Research and Molecular Cardiology Laboratory, Department of Biochemistry, Guindy Campus, University of Madras, Chennai, Tamil Nadu, India
| | - Boopathi Sowndhar Rajan
- Peptide Research and Molecular Cardiology Laboratory, Department of Biochemistry, Guindy Campus, University of Madras, Chennai, Tamil Nadu, India
| | - Elangovan Vellaichamy
- Peptide Research and Molecular Cardiology Laboratory, Department of Biochemistry, Guindy Campus, University of Madras, Chennai, Tamil Nadu, India
| |
Collapse
|
10
|
Santos AR, Koike TE, Santana AM, Miranda NC, Dell Aquila RA, Silva TC, Aoki MS, Miyabara EH. Glutamine supplementation accelerates functional recovery of EDL muscles after injury by modulating the expression of S100 calcium-binding proteins. Histochem Cell Biol 2023:10.1007/s00418-023-02194-5. [PMID: 37179509 DOI: 10.1007/s00418-023-02194-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2023] [Indexed: 05/15/2023]
Abstract
The aim of the current study was to investigate the effect of glutamine supplementation on the expression of HSP70 and the calcium-binding proteins from the S100 superfamily in the recovering extensor digitorum longus (EDL) muscle after injury. Two-month-old Wistar rats were subjected to cryolesion of the EDL muscle and then randomly divided into two groups (with or without glutamine supplementation). Starting immediately after the injury, the supplemented group received daily doses of glutamine (1 g/kg/day, via gavage) for 3 and 10 days orally. Then, muscles were subjected to histological, molecular, and functional analysis. Glutamine supplementation induced an increase in myofiber size of regenerating EDL muscles and prevented the decline in maximum tetanic strength of these muscles evaluated 10 days after injury. An accelerated upregulation of myogenin mRNA levels was detected in glutamine-supplemented injured muscles on day 3 post-cryolesion. The HSP70 expression increased only in the injured group supplemented with glutamine for 3 days. The increase in mRNA levels of NF-κB, the pro-inflammatory cytokines IL-1β and TNF-α, and the calcium-binding proteins S100A8 and S100A9 on day 3 post-cryolesion in EDL muscles was attenuated by glutamine supplementation. In contrast, the decrease in S100A1 mRNA levels in the 3-day-injured EDL muscles was minimized by glutamine supplementation. Overall, our results suggest that glutamine supplementation accelerates the recovery of myofiber size and contractile function after injury by modulating the expression of myogenin, HSP70, NF-κB, pro-inflammatory cytokines, and S100 calcium-binding proteins.
Collapse
Affiliation(s)
- Audrei R Santos
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av Prof. Lineu Prestes, 2415, Sao Paulo, SP, 05508-000, Brazil
| | - Tatiana E Koike
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av Prof. Lineu Prestes, 2415, Sao Paulo, SP, 05508-000, Brazil
| | - Alana M Santana
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av Prof. Lineu Prestes, 2415, Sao Paulo, SP, 05508-000, Brazil
| | - Natalya C Miranda
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av Prof. Lineu Prestes, 2415, Sao Paulo, SP, 05508-000, Brazil
| | - Rodrigo A Dell Aquila
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av Prof. Lineu Prestes, 2415, Sao Paulo, SP, 05508-000, Brazil
| | - Thiago C Silva
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av Prof. Lineu Prestes, 2415, Sao Paulo, SP, 05508-000, Brazil
| | - Marcelo S Aoki
- School of Arts, Sciences and Humanities, University of Sao Paulo, Sao Paulo, SP, 03828-000, Brazil
| | - Elen H Miyabara
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av Prof. Lineu Prestes, 2415, Sao Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
11
|
Carvalho R, Bonfá IS, de Araújo Isaías Muller J, Pando SC, Toffoli-Kadri MC. Protease inhibitor from Libidibia ferrea seeds attenuates inflammatory and nociceptive responses in mice. JOURNAL OF ETHNOPHARMACOLOGY 2023; 300:115694. [PMID: 36096346 DOI: 10.1016/j.jep.2022.115694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/15/2022] [Accepted: 08/31/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Libidibia ferrea (Mart. ex. Tul.) L.P. Queiroz is a Brazilian native tree locally known as jucá and pau-ferro, and it has been used in folk medicine for relieving, asthma, bronchitis, sore throat, rheumatism, enterocolitis and fever. The anti-inflammatory properties of L. ferrea were confirmed for its stem, fruit, leaves, bark and seeds extracts, however little is known about the natural compounds that may be associated with that response. AIM OF THIS STUDY In a normal physiological condition, many enzymes play an important role in catalyzing biological functions. Among them, proteases are of great interest. Although they take part of many biological systems, as the inflammatory process, when deregulated, proteases may cause system malfunctions, such as under- or overproduction of cytokines, or immune cells activation. Thus, protease inhibitors prevent these immune responses by regulating proteases. The objective of this study was to evaluate the anti-inflammatory and anti-nociceptive response of a protease inhibitor purified from L. ferrea seeds (LfTI). MATERIALS AND METHODS In vitro (5, 50 and 250 μg/mL of LfTI) and in vivo (0.6, 3 e 15 mg/kg of LfTI) assays were performed. Male Swiss mice weighing 18-25 g were used for cell harvesting and for the in vivo assays. The anti-inflammatory activity was analyzed in vitro by macrophage cytotoxicity, hydrogen peroxide (H2O2) production, and cell adhesion assays; and in vivo by leukocyte recruitment, nitric oxide (NO) production, vascular permeability, paw edema and mast cell degranulation assays. The anti-nociceptive activity was evaluated through abdominal writhing test induced by acetic acid and formalin sensitization. RESULTS Our results showed that, in vitro, LfTI is not cytotoxic. Also, LfTI (50 μg/mL) inhibited macrophage H2O2 production (48.2%), and adhesion (48.4%). LfTI (0.6, 3 e 15 mg/kg) decreased polymorphonuclear cell recruitment dose-dependently, and it inhibited NO production (53%), vascular permeability (40.7%) and paw edema at 3 mg/kg at different time, but it did not inhibit mast cell degranulation. Besides, LfTI did not inhibit either the number of writhing or the licking time in the formalin test in the second phase (inflammatory). However, LfTI (3 mg/kg) inhibited licking time at the first phase (neurogenic) in the formalin sensitization (46.1%). CONCLUSIONS Our results show that LfTI has anti-inflammatory and antinociceptive (neurogenic pain) effects, and these effects might be associated with the inhibition of inflammatory proteases and/or protease-activated receptors activation hindering.
Collapse
Affiliation(s)
- Raquel Carvalho
- Graduate Program in Biotechnology, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso do Sul (UFMS), Campo Grande, MS, Brazil; Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, UFMS, Campo Grande, MS, Brazil.
| | - Iluska Senna Bonfá
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, UFMS, Campo Grande, MS, Brazil.
| | - Jéssica de Araújo Isaías Muller
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, UFMS, Campo Grande, MS, Brazil; Multicenter Graduate Program in Biochemistry and Molecular Biology, Institute of Biosciences, UFMS, Campo Grande, MS, Brazil.
| | | | - Mônica Cristina Toffoli-Kadri
- Graduate Program in Biotechnology, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso do Sul (UFMS), Campo Grande, MS, Brazil; Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, UFMS, Campo Grande, MS, Brazil; Multicenter Graduate Program in Biochemistry and Molecular Biology, Institute of Biosciences, UFMS, Campo Grande, MS, Brazil.
| |
Collapse
|
12
|
Saha B, Vannucci L, Saha B, Tenti P, Baral R. Evolvability and emergence of tumor heterogeneity as a space-time function. Cytokine 2023; 161:156061. [PMID: 36252436 DOI: 10.1016/j.cyto.2022.156061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/20/2022] [Accepted: 09/30/2022] [Indexed: 11/22/2022]
Abstract
The loss of control of cell proliferation, apoptosis regulation and contact inhibition leads to tumor development. While benign tumors are restricted to their primary space, i.e. where these tumors first originate, the metastatic tumors not only disseminate- facilitated by hypoxia-driven neovascularization- to distant secondary sites but also show substantial changes in metabolism, tissue architectures, gene expression profiles and immune phenotypes. All these alterations result in radio-, chemo- and immune-resistance rendering these metastatic tumor cells refractory to therapy. Since the beginning of the transformation, these factors- which influence each other- are incorporated to the developing and metastasizing tumor. As a result, the complexities in the heterogeneity of tumor progressively increase. This space-time function in the heterogeneity of tumors is generated by various conditions and factors at the genetic as well as microenvironmental levels, for example, endogenous retroviruses, methylation and epigenetic dysregulation that may be etiology-specific, cancer associated inflammation, remodeling of the extracellular matrix and mesenchymal cell shifted functions. On the one hand, these factors may cause de-differentiation of the tumor cells leading to cancer stem cells that contribute to radio-, chemo- and immune-resistance and recurrence of tumors. On the other hand, they may also enhance the heterogeneity under specific microenvironment-driven proliferation. In this editorial, we intend to underline the importance of heterogeneity in cancer progress, its evaluation and its use in correlation with the tumor evolution in a specific patient as a field of research for achieving precise patient-tailored treatments and amelioration of diagnostic (monitoring) tools and prognostic capacity.
Collapse
Affiliation(s)
- Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| | - Luca Vannucci
- Institute of Microbiology, Czech Academy of Sciences, Videnska 1083, Praha, Czech Republic.
| | - Baibaswata Saha
- Institute of Microbiology, Czech Academy of Sciences, Videnska 1083, Praha, Czech Republic
| | - Paolo Tenti
- Institute of Microbiology, Czech Academy of Sciences, Videnska 1083, Praha, Czech Republic
| | - Rathindranath Baral
- Chittaranjan National Cancer Institute, Shyamaprasad Mukherjee Road, Calcutta 700026, India.
| |
Collapse
|
13
|
Tuli HS, Kaur J, Vashishth K, Sak K, Sharma U, Choudhary R, Behl T, Singh T, Sharma S, Saini AK, Dhama K, Varol M, Sethi G. Molecular mechanisms behind ROS regulation in cancer: A balancing act between augmented tumorigenesis and cell apoptosis. Arch Toxicol 2023; 97:103-120. [PMID: 36443493 DOI: 10.1007/s00204-022-03421-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/14/2022] [Indexed: 11/29/2022]
Abstract
ROS include hydroxyl radicals (HO.), superoxide (O2..), and hydrogen peroxide (H2O2). ROS are typically produced under physiological conditions and play crucial roles in living organisms. It is known that ROS, which are created spontaneously by cells through aerobic metabolism in mitochondria, can have either a beneficial or detrimental influence on biological systems. Moderate levels of ROS can cause oxidative damage to proteins, DNA and lipids, which can aid in the pathogenesis of many disorders, including cancer. However, excessive concentrations of ROS can initiate programmed cell death in cancer. Presently, a variety of chemotherapeutic drugs and herbal agents are being investigated to induce ROS-mediated cell death in cancer. Therefore, preserving ROS homeostasis is essential for ensuring normal cell development and survival. On account of a significant association of ROS levels at various concentrations with carcinogenesis in a number of malignancies, further studies are needed to determine the underlying molecular mechanisms and develop the possibilities for intervening in these processes.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133207, India
| | - Jagjit Kaur
- Graduate School of Biomedical Engineering, Faculty of Engineering, The University of New South Wales, Sydney, 2052, Australia
| | - Kanupriya Vashishth
- Advance Cardiac Centre Department of Cardiology, PGIMER, Chandigarh, 160012, India
| | | | - Ujjawal Sharma
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133207, India
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Renuka Choudhary
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133207, India
| | - Tapan Behl
- Department of Pharmacology, School of Health Sciences & Technology (SoHST), University of Petroleum and Energy Studies, Bidholi, Dehradun, Uttarakhand, 248007, India
| | - Tejveer Singh
- Translanatal Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
| | - Sheetu Sharma
- Department of Pharmacovigilace and Clinical Research, Chitkara University, Rajpura, 140401, India
| | - Adesh K Saini
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133207, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Mehmet Varol
- Department of Molecular Biology and Genetics, Faculty of Science, Mugla Sitki Kocman University, Mugla, 48000, Turkey
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| |
Collapse
|
14
|
Uchendu I, Zhilenkova A, Pirogova Y, Basova M, Bagmet L, Kohanovskaia I, Ngaha Y, Ikebunwa O, Sekacheva M. Cytokines as Potential Therapeutic Targets and their Role in the Diagnosis and Prediction of Cancers. Curr Pharm Des 2023; 29:2552-2567. [PMID: 37916493 DOI: 10.2174/0113816128268111231024054240] [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: 06/19/2023] [Accepted: 09/26/2023] [Indexed: 11/03/2023]
Abstract
The death rate from cancer is declining as a result of earlier identification and more advanced treatments. Nevertheless, a number of unfavourable adverse effects, including prolonged, long-lasting inflammation and reduced immune function, usually coexist with anti-cancer therapies and lead to a general decline in quality of life. Improvements in standardized comprehensive therapy and early identification of a variety of aggressive tumors remain the main objectives of cancer research. Tumor markers in those with cancer are tumor- associated proteins that are clinically significant. Even while several tumor markers are routinely used, they don't always provide reliable diagnostic information. Serum cytokines are promising markers of tumor stage, prognosis, and responsiveness to therapy. In fact, several cytokines are currently proposed as potential biomarkers in a variety of cancers. It has actually been proposed that the study of circulatory cytokines together with biomarkers that are particular to cancer can enhance and accelerate cancer diagnosis and prediction, particularly via blood samples that require minimal to the absence of invasion. The purpose of this review was to critically examine relevant primary research literature in order to elucidate the role and importance of a few identified serum cytokines as prospective therapeutic targets in oncological diseases.
Collapse
Affiliation(s)
- Ikenna Uchendu
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
- Department of Medical Laboratory Science, Faculty of Health Science and Technology, University of Nigeria, Enugu Campus, Enugu, Nigeria
| | - Angelina Zhilenkova
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Yuliya Pirogova
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Maria Basova
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Leonid Bagmet
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Iana Kohanovskaia
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Yvan Ngaha
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Obinna Ikebunwa
- Department of Medical Laboratory Science, Faculty of Health Science and Technology, University of Nigeria, Enugu Campus, Enugu, Nigeria
- Department of Biotechnology, First Moscow State Medical University of The Ministry of Health of Russia (Sechenov University), Moscow, Russia
| | - Marina Sekacheva
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), Moscow, Russia
| |
Collapse
|
15
|
Jiang R, Cao M, Mei S, Guo S, Zhang W, Ji N, Zhao Z. Trends in metabolic signaling pathways of tumor drug resistance: A scientometric analysis. Front Oncol 2022; 12:981406. [DOI: 10.3389/fonc.2022.981406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/12/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundCancer chemotherapy resistance is one of the most critical obstacles in cancer therapy. Since Warburg O first observed alterations in cancer metabolism in the 1950s, people gradually found tumor metabolism pathways play a fundamental role in regulating the response to chemotherapeutic drugs, and the attempts of targeting tumor energetics have shown promising preclinical outcomes in recent years. This study aimed to summarize the knowledge structure and identify emerging trends and potential hotspots in metabolic signaling pathways of tumor drug resistance research.MethodsPublications related to metabolic signaling pathways of tumor drug resistance published from 1992 to 2022 were retrieved from the Web of Science Core Collection database. The document type was set to articles or reviews with language restriction to English. Two different scientometric software including Citespace and VOS viewer were used to conduct this scientometric analysis.ResultsA total of 2,537 publications including 1,704 articles and 833 reviews were retrieved in the final analysis. The USA made the most contributions to this field. The leading institution was the University of Texas MD Anderson Cancer Center. Avan A was the most productive author, and Hanahan D was the key researcher with the most co-citations, but there is no leader in this field yet. Cancers was the most influential academic journal, and Oncology was the most popular research field. Based on keywords occurrence analysis, these selected keywords could be roughly divided into five main topics: cluster 1 (study of cancer cell apoptosis pathway); cluster 2 (study of resistance mechanisms of different cancer types); cluster 3 (study of cancer stem cells); cluster 4 (study of tumor oxidative stress and inflammation signaling pathways); and cluster 5 (study of autophagy). The keywords burst detection identified several keywords as new research hotspots, including “tumor microenvironment,” “invasion,” and “target”.ConclusionTumor metabolic reprogramming of drug resistance research is advancing rapidly. This study serves as a starting point, providing a thorough overview, the development landscape, and future opportunities in this field.
Collapse
|
16
|
Triyasa KS, Diantini A, Barliana MI. A Review of Herbal Medicine-Based Phytochemical of Garcinia as Molecular Therapy for Breast Cancer. Drug Des Devel Ther 2022; 16:3573-3588. [PMID: 36248245 PMCID: PMC9554952 DOI: 10.2147/dddt.s358229] [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: 01/31/2022] [Accepted: 09/07/2022] [Indexed: 11/07/2022] Open
Abstract
Data from globocan statistic in 2020 indicate that breast cancer has become highest incidence rate of cancer. Estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) are known immunohistochemistry (IHC) markers that mediate cell growth and survival signaling. Furthermore, regulator proteins, receptors, and their downstream signaling pathways have emerged as critical components in breast cancer formation and proliferation, and have become well-established therapeutic targets and the core focus of breast cancer therapy research. Garcinia is a big genus in the Clusiaceae family that contains a wide spectrum of biologically active metabolites for the chemical composition of their isolated fruits, stem barks, seeds, leaves, and roots, have resulted including polyisoprenylated benzophenones, polyphenols, bioflavonoids, xanthones, lactones, and triterpenes. This review article aimed to analyze the potential of Garcinia phytochemicals as a molecular therapy of breast cancer. The results showed that phytochemicals of Garcinia (i.e., α-mangostin, Cambogin, Gambogic Acid [GA], Garcinol, Griffipavixanthone, Friedolanostane triterpenoid, Hexane, Neobractatin, 7-Epiclusianone, xanthochymol - guttiferone E, and isoxanthochymol - cycloxanthochymol) have anticancer properties, including apoptosis, inhibition of proliferation, and metastasis. This review is important to provide information regarding phytochemicals of Garcinia as an alternative treatment for breast cancer patients. This article selected 28 article researches based on inclusion criteria with the keyword “Garcinia” and “Breast cancer”, in English, and available in full text and abstract searching on PubMed.
Collapse
Affiliation(s)
- Komang Suma Triyasa
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Ajeng Diantini
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia,Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
| | - Melisa Intan Barliana
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia,Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia,Correspondence: Melisa Intan Barliana, Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Ir. Soekarno KM. 21, Jatinangor, Bandung, 45363, Indonesia, Email
| |
Collapse
|
17
|
Chang Z, Zhang Y, Fan J, Zhang L, Liu S, Liu G, Tu J. The potential effects and mechanisms of breast inflammatory lesions on the occurrence and development of breast cancer. Front Oncol 2022; 12:932743. [PMID: 35992864 PMCID: PMC9389363 DOI: 10.3389/fonc.2022.932743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/30/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer as the most common cancer in women has become the leading cause of cancer death for women. Although many inflammatory factors increase the risk of breast cancer, there are very few studies on the mechanisms by which inflammation affects the initiation and progression of breast cancer. Here, we profiled and compared the transcriptome of normal tissues, inflammatory breast tissues, benign breast tumors, and malignant breast tumors. To find key regulatory factors, a protein interaction network between characteristic modules in inflammatory lesions and ER-negative (ER−) breast cancer was constructed and inflammation-cancer interface genes were identified. We found that the transcriptional profile of inflammatory breast tissues was similar with ER− malignant tumors, featured with low ER expression levels and similar immune signaling pathway activation. Through comprehensive protein network analysis, we identified the interface genes and chemokine signaling pathway that have the potential to promote inflammatory cancer transformation. These interface genes could be used as a risk factor to provide a certain basis for the clinical early detection and treatment of breast cancer. This is the first study to explore the association between breast inflammatory lesions and breast cancer at the transcriptome level. Our inflammation data and research results provide a basis for future inflammation-cancer transformation analysis.
Collapse
Affiliation(s)
- Zhaoxia Chang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Zhang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jue Fan
- Singleron Biotechnologies, Nanjing, China
| | - Lixing Zhang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Juchuanli Tu, ; Guangyu Liu, ; Suling Liu,
| | - Guangyu Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Juchuanli Tu, ; Guangyu Liu, ; Suling Liu,
| | - Juchuanli Tu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Juchuanli Tu, ; Guangyu Liu, ; Suling Liu,
| |
Collapse
|
18
|
Pardo-Sánchez I, García-Moreno D, Mulero V. Zebrafish Models to Study the Crosstalk between Inflammation and NADPH Oxidase-Derived Oxidative Stress in Melanoma. Antioxidants (Basel) 2022; 11:1277. [PMID: 35883768 PMCID: PMC9311651 DOI: 10.3390/antiox11071277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 02/01/2023] Open
Abstract
Melanoma is the deadliest form of skin cancer, and its incidence continues to increase. In the early stages of melanoma, when the malignant cells have not spread to lymph nodes, they can be removed by simple surgery and there is usually low recurrence. Melanoma has a high mortality rate due to its ability to metastasize; once melanoma has spread, it becomes a major health complication. For these reasons, it is important to study how healthy melanocytes transform into melanoma cells, how they interact with the immune system, which mechanisms they use to escape immunosurveillance, and, finally, how they spread and colonize other tissues, metastasizing. Inflammation and oxidative stress play important roles in the development of several types of cancer, including melanoma, but it is not yet clear under which conditions they are beneficial or detrimental. Models capable of studying the relevance of inflammation and oxidative stress in the early steps of melanocyte transformation are urgently needed, as they are expected to help recognize premetastatic lesions in patients by improving both early detection and the development of new therapies.
Collapse
Affiliation(s)
- Irene Pardo-Sánchez
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain;
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Diana García-Moreno
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain;
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Victoriano Mulero
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain;
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
19
|
Larsen O, van der Velden WJC, Mavri M, Schuermans S, Rummel PC, Karlshøj S, Gustavsson M, Proost P, Våbenø J, Rosenkilde MM. Identification of a conserved chemokine receptor motif that enables ligand discrimination. Sci Signal 2022; 15:eabg7042. [PMID: 35258997 DOI: 10.1126/scisignal.abg7042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Extensive ligand-receptor promiscuity in the chemokine signaling system balances beneficial redundancy and specificity. However, this feature poses a major challenge to selectively modulate the system pharmacologically. Here, we identified a conserved cluster of three aromatic receptor residues that anchors the second extracellular loop (ECL2) to the top of receptor transmembrane helices (TM) 4 and 5 and enables recognition of both shared and specific characteristics of interacting chemokines. This cluster was essential for the activation of several chemokine receptors. Furthermore, characteristic motifs of the ß1 strand and 30s loop make the two main CC-chemokine subgroups-the macrophage inflammatory proteins (MIPs) and monocyte chemoattractant proteins (MCPs)-differentially dependent on this cluster in the promiscuous receptors CCR1, CCR2, and CCR5. The cluster additionally enabled CCR1 and CCR5 to discriminate between closely related MIPs based on the N terminus of the chemokine. G protein signaling and β-arrestin2 recruitment assays confirmed the importance of the conserved cluster in receptor discrimination of chemokine ligands. This extracellular site may facilitate the development of chemokine-related therapeutics.
Collapse
Affiliation(s)
- Olav Larsen
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Wijnand J C van der Velden
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Maša Mavri
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia
| | - Sara Schuermans
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Pia C Rummel
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Stefanie Karlshøj
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Martin Gustavsson
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Jon Våbenø
- Helgeland Hospital Trust, Prestmarkveien 1, 8800 Sandnessjøen, Norway
| | - Mette M Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
20
|
Wetzel A, Bonnefoy F, Chagué C, Vetter M, Couturier M, Baffert B, Adotévi O, Saas P, Perruche S. Pro-Resolving Factor Administration Limits Cancer Progression by Enhancing Immune Response Against Cancer Cells. Front Immunol 2022; 12:812171. [PMID: 35116038 PMCID: PMC8804172 DOI: 10.3389/fimmu.2021.812171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/21/2021] [Indexed: 12/27/2022] Open
Abstract
Cancers are consequences of cellular dysfunction leading to an aberrant cellular multiplication and proliferation, subsequently yielding metastasis formation. Inflammatory reaction, with immune cell recruitment, is the main defense against precancerous lesions. However, an inflammatory environment also favors cancer cell progression, with cancer cell evasion from immune surveillance, leading to cancer development. Current therapeutic strategies enhance this natural immune response in order to restore immunosurveillance. The variety of these strategies is a predominant source of inflammatory mediators used by cancer cells to grow, differentiate, and migrate, therefore encouraging metastasis formation. For this reason, during cancer progression, limiting inflammation appears to be an innovative strategy to avoid the escape of cancer cells and potentially enhance the efficacy of antitumor therapies. Thus, this study aims to investigate the impact of administering pro-resolving factors (SuperMApo® drug candidate), which are inducers of inflammation resolution, in the framework of cancer treatment. We have observed that administering pro-resolving mediators issued from apoptotic cell efferocytosis by macrophages controlled peritoneal cancer progression by limiting cancer cell dissemination to the blood and mesenteric lymph nodes. This observation has been linked to an increase of macrophage mobilization in both peritoneal cavity and mesenteric lymph nodes. This control is associated to a restricted immunosuppressive myeloid cell circulation and to an IFN-γ-specific anti-tumor T-cell response. Altogether, these results suggest that administering proresolving factors could provide a new additional therapeutic alternative to control cancer progression.
Collapse
Affiliation(s)
- Audrey Wetzel
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, LabEx LipSTIC, Besançon, France
- MED’INN’Pharma, Besançon, France
| | - Francis Bonnefoy
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, LabEx LipSTIC, Besançon, France
- MED’INN’Pharma, Besançon, France
| | - Cécile Chagué
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, LabEx LipSTIC, Besançon, France
| | - Mathieu Vetter
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, LabEx LipSTIC, Besançon, France
| | | | - Blandine Baffert
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, LabEx LipSTIC, Besançon, France
| | - Olivier Adotévi
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, LabEx LipSTIC, Besançon, France
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Philippe Saas
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, LabEx LipSTIC, Besançon, France
| | - Sylvain Perruche
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, LabEx LipSTIC, Besançon, France
- MED’INN’Pharma, Besançon, France
- *Correspondence: Sylvain Perruche,
| |
Collapse
|
21
|
Preoperative Neutrophil-to-Lymphocyte Ratio, Platelet-to-Lymphocyte Ratio, and CEA as the Potential Prognostic Biomarkers for Colorectal Cancer. Can J Gastroenterol Hepatol 2022; 2022:3109165. [PMID: 35028301 PMCID: PMC8752269 DOI: 10.1155/2022/3109165] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 11/27/2021] [Accepted: 12/04/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND This study was to evaluate the prognostic value of the preoperative neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and carcinoembryonic antigen (CEA) in colorectal cancer (CRC) patients and to identify the potential and easily accessible prognostic biomarkers for CRC. METHODS We retrospectively reviewed altogether the records of 330 CRC patients according to inclusion criteria. The clinical characteristics include age at diagnosis, body mass index (BMI), preoperative CEA level, neutrophil , lymphocyte, and platelet count, tumor primary site and size, clinical pathological TNM stage, and survival status were recorded through the review of medical records. The overall survival (OS) was calculated using the Kaplan-Meier method. The Cox proportional hazards model was used for the univariate and multivariate analysis to evaluate the prognostic factors of CRC. RESULTS A total of 330 patients were finally included in the current study. The mean follow-up duration was 32.8 ± 19.1 months (range, 0.1-67.7). Compared with the median OS, preoperative high NLR, PLR, and CEA, and low BMI had lower median OS. The NLR and PLR value rise indicates lower median OS in stage I-II CRC; however, the NLR value and CEA level rise indicates lower median OS in stage III-IV CRC. Preoperative high NLR, PLR, and CEA level and low BMI have poorer OS by univariate analysis. By multivariate analysis, the age, sex, N, M stage, and BMI demonstrated independently influence the OS of CRC. NLR was an independent predictor of stage I-II CRC, and the CEA level was an independent predictor of stage III-IV CRC. CONCLUSIONS Our results show that preoperative high NLR, PLR, CEA, and low BMI had poorer OS, NLR was an independent predictor of stage I-II CRC, and the CEA level was an independent predictor of stage III-IV CRC.
Collapse
|
22
|
Wang S, Liu Q, Zeng T, Zhan J, Zhao H, Ho CT, Xiao Y, Li S. Immunomodulatory effects and associated mechanisms of Momordica charantia and its phytochemicals. Food Funct 2022; 13:11986-11998. [DOI: 10.1039/d2fo02096c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Momordica charantia L. (M. charantia), which is a member of the Cucurbitaceae family and widely distributed in tropical and subtropical regions, has been consumed as a vegetable and also used as herbal medicine for thousands of years worldwide.
Collapse
Affiliation(s)
- Shuzhen Wang
- College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang, 438000, Hubei Province, P.R. China
| | - Qian Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, 250355, Shandong Province, P.R. China
| | - Ting Zeng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, 250355, Shandong Province, P.R. China
| | - Jianfeng Zhan
- College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang, 438000, Hubei Province, P.R. China
| | - Hui Zhao
- Tianjin Key Laboratory of Food and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA
| | - Yunli Xiao
- College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang, 438000, Hubei Province, P.R. China
| | - Shiming Li
- College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang, 438000, Hubei Province, P.R. China
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA
| |
Collapse
|
23
|
Neganova M, Liu J, Aleksandrova Y, Klochkov S, Fan R. Therapeutic Influence on Important Targets Associated with Chronic Inflammation and Oxidative Stress in Cancer Treatment. Cancers (Basel) 2021; 13:6062. [PMID: 34885171 PMCID: PMC8657135 DOI: 10.3390/cancers13236062] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/28/2021] [Accepted: 11/28/2021] [Indexed: 01/17/2023] Open
Abstract
Chronic inflammation and oxidative stress are the interconnected pathological processes, which lead to cancer initiation and progression. The growing level of oxidative and inflammatory damage was shown to increase cancer severity and contribute to tumor spread. The overproduction of reactive oxygen species (ROS), which is associated with the reduced capacity of the endogenous cell defense mechanisms and/or metabolic imbalance, is the main contributor to oxidative stress. An abnormal level of ROS was defined as a predisposing factor for the cell transformation that could trigger pro-oncogenic signaling pathways, induce changes in gene expression, and facilitate accumulation of mutations, DNA damage, and genomic instability. Additionally, the activation of transcription factors caused by a prolonged oxidative stress, including NF-κB, p53, HIF1α, etc., leads to the expression of several genes responsible for inflammation. The resulting hyperactivation of inflammatory mediators, including TNFα, TGF-β, interleukins, and prostaglandins can contribute to the development of neoplasia. Pro-inflammatory cytokines were shown to trigger adaptive reactions and the acquisition of resistance by tumor cells to apoptosis, while promoting proliferation, invasion, and angiogenesis. Moreover, the chronic inflammatory response leads to the excessive production of free radicals, which further aggravate the initiated reactions. This review summarizes the recent data and progress in the discovery of mechanisms that associate oxidative stress and chronic inflammation with cancer onset and metastasis. In addition, the review provides insights for the development of therapeutic approaches and the discovery of natural substances that will be able to simultaneously inhibit several key oncological and inflammation-related targets.
Collapse
Affiliation(s)
- Margarita Neganova
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China; (M.N.); (J.L.)
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China; (M.N.); (J.L.)
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yulia Aleksandrova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Sergey Klochkov
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China; (M.N.); (J.L.)
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
24
|
de Villiers EM, zur Hausen H. Bovine Meat and Milk Factors (BMMFs): Their Proposed Role in Common Human Cancers and Type 2 Diabetes Mellitus. Cancers (Basel) 2021; 13:cancers13215407. [PMID: 34771570 PMCID: PMC8582480 DOI: 10.3390/cancers13215407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary This manuscript emphasizes the mechanistic differences of infectious agents contributing to human cancers either by “direct” or “indirect” interactions. The epidemiology of cancers linked to direct carcinogens differs (e.g., response to immunosuppression) from those cancers linked with indirect infectious interactions. We discuss their role in colon, breast, and prostate cancers and type II diabetes mellitus. A brief discussion covers the potential role of BMMF (bovine meat and milk factor) infections in acute myeloid leukemia. Abstract Exemplified by infections with bovine meat and milk factors (BMMFs), this manuscript emphasizes the different mechanistic aspects of infectious agents contributing to human cancers by “direct” or “indirect” interactions. The epidemiology of cancers linked to direct carcinogens (e.g., response to immunosuppression) differs from those cancers linked with indirect infectious interactions. Cancers induced by direct infectious carcinogens commonly increase under immunosuppression, whereas the cancer risk by indirect carcinogens is reduced. This influences their responses to preventive and therapeutic interferences. In addition, we discuss their role in colon, breast and prostate cancers and type II diabetes mellitus. A brief discussion covers the potential role of BMMF infections in acute myeloid leukemia.
Collapse
Affiliation(s)
- Ethel-Michele de Villiers
- Correspondence: (E.-M.d.V.); (H.z.H.); Tel.: +49-151-4312-3085 (E.-M.d.V.); +49-6221-423850 (H.z.H.)
| | - Harald zur Hausen
- Correspondence: (E.-M.d.V.); (H.z.H.); Tel.: +49-151-4312-3085 (E.-M.d.V.); +49-6221-423850 (H.z.H.)
| |
Collapse
|
25
|
Takenaka M, Yabuta A, Takahashi Y, Takakura Y. Interleukin-4-carrying small extracellular vesicles with a high potential as anti-inflammatory therapeutics based on modulation of macrophage function. Biomaterials 2021; 278:121160. [PMID: 34653934 DOI: 10.1016/j.biomaterials.2021.121160] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 12/18/2022]
Abstract
Interleukin-4 (IL4), a Th2-type cytokine that can drive M2 macrophage polarization, is expected to be used as an anti-inflammatory therapy agent as M2 polarization of macrophages can ameliorate chronic inflammation. However, several problems, such as the low effectiveness and side effects, have hampered the clinical application. To safely and effectively use IL4, an efficient delivery of IL4 to target cells, macrophages, is necessary. Small extracellular vesicles (sEVs) are promising candidates as macrophage delivery carriers because they are efficiently recognized by macrophages. In addition, considering the property of IL4 signaling, for which the internalization of IL4 receptor into the cellular compartment is important, and sEV uptake mechanism by macrophages, sEVs are expected to amplify IL4 signaling. In this study, we developed IL4-carrying sEVs (IL4-sEVs) by genetically engineering sEV-producing cells. We investigated the bioactivity of IL4-sEVs using RAW264.7 macrophages and their potential for therapeutic application to the treatment of an inflammatory disease using collagen-induced arthritis model mice. IL4-sEVs exhibited stronger anti-inflammatory effects on M1-polarized macrophages through M2 polarization of macrophages than those of soluble IL4 proteins. Moreover, IL4-sEVs exhibited more effective therapeutic effects on rheumatoid arthritis than those of IL4. These results indicate that IL4-carrying sEVs are promising anti-inflammatory therapeutics.
Collapse
Affiliation(s)
- Misako Takenaka
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Ayane Yabuta
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Yuki Takahashi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| |
Collapse
|
26
|
Pinto C, Zucali PA, Pagano M, Grosso F, Pasello G, Garassino MC, Tiseo M, Soto Parra H, Grossi F, Cappuzzo F, de Marinis F, Pedrazzoli P, Bonomi M, Gianoncelli L, Perrino M, Santoro A, Zanelli F, Bonelli C, Maconi A, Frega S, Gervasi E, Boni L, Ceresoli GL. Gemcitabine with or without ramucirumab as second-line treatment for malignant pleural mesothelioma (RAMES): a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol 2021; 22:1438-1447. [PMID: 34499874 DOI: 10.1016/s1470-2045(21)00404-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND There is a preclinical rationale for inhibiting angiogenesis in mesothelioma. We aimed to assess the efficacy and safety of the anti-VEGFR-2 antibody ramucirumab combined with gemcitabine in patients with pretreated malignant pleural mesothelioma. METHODS RAMES was a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial done at 26 hospitals in Italy. Eligible patients were aged 18 years or older, had Eastern Cooperative Oncology Group performance status 0-2, and histologically proven malignant pleural mesothelioma progressing during or after first-line treatment with pemetrexed plus platinum. Patients were randomly assigned (1:1) to receive intravenous gemcitabine 1000 mg/m2 on days 1 and 8 every 3 weeks plus either intravenous placebo (gemcitabine plus placebo group) or ramucirumab 10 mg/kg (gemcitabine plus ramucirumab group) on day 1 every 3 weeks, until tumour progression or unacceptable toxicity. Central randomisation was done according to a minimisation algorithm method, associated with a random element using the following stratification factors: ECOG performance status, age, histology, and first-line time-to-progression. The primary endpoint was overall survival, measured from the date of randomisation to the date of death from any cause. Efficacy analyses were assessed in all patients who had been correctly randomised and received their allocated treatment, and safety analyses were assessed in all patients who received at least one dose of their assigned treatment. This trial is registered with ClinicalTrials.gov, NCT03560973, and with EudraCT, 2016-001132-36. FINDINGS Between Dec 22, 2016, and July 30, 2018, of 165 patients enrolled 161 were correctly assigned and received either gemcitabine plus placebo (n=81) or gemcitabine plus ramucirumab (n=80). At database lock (March 8, 2020), with a median follow-up of 21·9 months (IQR 17·7-28·5), overall survival was longer in the ramucirumab group (HR 0·71, 70% CI 0·59-0·85; p=0·028). Median overall survival was 13·8 months (70% CI 12·7-14·4) in the gemcitabine plus ramucirumab group and 7·5 months (6·9-8·9) in the gemcitabine plus placebo group. Grade 3-4 treatment-related adverse events were reported in 35 (44%) of 80 patients in the gemcitabine plus ramucirumab group and 24 (30%) of 81 in the gemcitabine plus placebo group. The most common treatment-related grade 3-4 adverse events were neutropenia (16 [20%] for gemcitabine plus ramucirumab vs ten [12%] for gemcitabine plus placebo) and hypertension (five [6%] vs none). Treatment-related serious adverse events were reported in five (6%) in the gemcitabine plus ramucirumab group and in four (5%) patients in the gemcitabine plus placebo group; the most common was thromboembolism (three [4%] for gemcitabine plus ramucirumab vs two [2%] for gemcitabine plus placebo). There were no treatment-related deaths. INTERPRETATION Ramucirumab plus gemcitabine significantly improved overall survival after first-line standard chemotherapy, with a favourable safety profile. This combination could be a new option in this setting. FUNDING Eli Lilly Italy. TRANSLATION For the Italian translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Carmine Pinto
- Medical Oncology Unit, Clinical Cancer Centre, AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Paolo Andrea Zucali
- Department of Biomedical Sciences, Humanitas University, Milan, Italy; Department of Oncology, IRCCS Humanitas Research Hospital, Milan, Italy.
| | - Maria Pagano
- Medical Oncology Unit, Clinical Cancer Centre, AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Federica Grosso
- Mesothelioma Unit, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy; Infrastruttura Ricerca Formazione e Innovazione, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Giulia Pasello
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padua, Italy
| | - Marina Chiara Garassino
- Thoracic Oncology Unit, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Hector Soto Parra
- Medical Oncology Unit, AOU Policlinico Vittorio Emanuele, Catania, Italy
| | - Francesco Grossi
- Medical Oncology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore, Milan, Italy; Medical Oncology Unit, University of Insubria, Varese, Italy
| | - Federico Cappuzzo
- Medical Oncology Unit, IRCCS Istituto Nazionale Tumori Regina Elena, Rome, Italy
| | - Filippo de Marinis
- Thoracic Oncology Division, Istituto Europeo di Oncologia IRCCS, Milan, Italy
| | - Paolo Pedrazzoli
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy
| | - Maria Bonomi
- Department of Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy; Department of Oncology, ASST Cremona, Cremona, Italy
| | - Letizia Gianoncelli
- Department of Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy; Department of Oncology, Ospedale San Paolo, Milan, Italy
| | - Matteo Perrino
- Department of Oncology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Milan, Italy; Department of Oncology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Francesca Zanelli
- Medical Oncology Unit, Clinical Cancer Centre, AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Candida Bonelli
- Medical Oncology Unit, Clinical Cancer Centre, AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Antonio Maconi
- Infrastruttura Ricerca Formazione e Innovazione, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Stefano Frega
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padua, Italy
| | - Erika Gervasi
- Medical Oncology Unit, Clinical Cancer Centre, AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Luca Boni
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | |
Collapse
|
27
|
Zhong H, Liu S, Cao F, Zhao Y, Zhou J, Tang F, Peng Z, Li Y, Xu S, Wang C, Yang G, Li ZQ. Dissecting Tumor Antigens and Immune Subtypes of Glioma to Develop mRNA Vaccine. Front Immunol 2021; 12:709986. [PMID: 34512630 PMCID: PMC8429949 DOI: 10.3389/fimmu.2021.709986] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/28/2021] [Indexed: 01/02/2023] Open
Abstract
Background Nowadays, researchers are leveraging the mRNA-based vaccine technology used to develop personalized immunotherapy for cancer. However, its application against glioma is still in its infancy. In this study, the applicable candidates were excavated for mRNA vaccine treatment in the perspective of immune regulation, and suitable glioma recipients with corresponding immune subtypes were further investigated. Methods The RNA-seq data and clinical information of 702 and 325 patients were recruited from TCGA and CGGA, separately. The genetic alteration profile was visualized and compared by cBioPortal. Then, we explored prognostic outcomes and immune correlations of the selected antigens to validate their clinical relevance. The prognostic index was measured via GEPIA2, and infiltration of antigen-presenting cells (APCs) was calculated and visualized by TIMER. Based on immune-related gene expression, immune subtypes of glioma were identified using consensus clustering analysis. Moreover, the immune landscape was visualized by graph learning-based dimensionality reduction analysis. Results Four glioma antigens, namely ANXA5, FKBP10, MSN, and PYGL, associated with superior prognoses and infiltration of APCs were selected. Three immune subtypes IS1-IS3 were identified, which fundamentally differed in molecular, cellular, and clinical signatures. Patients in subtypes IS2 and IS3 carried immunologically cold phenotypes, whereas those in IS1 carried immunologically hot phenotype. Particularly, patients in subtypes IS3 and IS2 demonstrated better outcomes than that in IS1. Expression profiles of immune checkpoints and immunogenic cell death (ICD) modulators showed a difference among IS1-IS3 tumors. Ultimately, the immune landscape of glioma elucidated considerable heterogeneity not only between individual patients but also within the same immune subtype. Conclusions ANXA5, FKBP10, MSN, and PYGL are identified as potential antigens for anti-glioma mRNA vaccine production, specifically for patients in immune subtypes 2 and 3. In summary, this study may shed new light on the promising approaches of immunotherapy, such as devising mRNA vaccination tailored to applicable glioma recipients.
Collapse
Affiliation(s)
- Hua Zhong
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Shuai Liu
- Department of Biochemistry, Molecular Biology, Entomology and Plant Pathology, Mississippi State University, Starkville, MS, United States
| | - Fang Cao
- Department of Cerebrovascular Disease, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yi Zhao
- Department of Medical Genetics, School of Basic Medical Science, Demonstration Center for Experimental Basic Medicine Education, Wuhan University, Wuhan, China
| | - Jianguo Zhou
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Feng Tang
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Zhaohua Peng
- Department of Biochemistry, Molecular Biology, Entomology and Plant Pathology, Mississippi State University, Starkville, MS, United States
| | - Yangsheng Li
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Shen Xu
- Department of Neurosurgery, No. 901 Hospital of the Chinese People’s Liberation Army Logistic Support Force, Hefei, China
| | - Chunlin Wang
- Department of Neurosurgery, No. 901 Hospital of the Chinese People’s Liberation Army Logistic Support Force, Hefei, China
| | - Guohua Yang
- Department of Medical Genetics, School of Basic Medical Science, Demonstration Center for Experimental Basic Medicine Education, Wuhan University, Wuhan, China
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
28
|
Ha MW, Paek SM. Recent Advances in the Synthesis of Ibuprofen and Naproxen. Molecules 2021; 26:4792. [PMID: 34443379 PMCID: PMC8399189 DOI: 10.3390/molecules26164792] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 11/16/2022] Open
Abstract
Herein, we review the recent progress in the synthesis of representative nonsteroidal anti-inflammatory drugs (NSAIDs), ibuprofen and naproxen. Although these drugs were discovered over 50 years ago, novel practical and asymmetric approaches are still being developed for their synthesis. In addition, this endeavor has enabled access to more potent and selective derivatives from the key frameworks of ibuprofen and naproxen. The development of a synthetic route to ibuprofen and naproxen over the last 10 years is summarized, including developing methodologies, finding novel synthetic routes, and applying continuous-flow chemistry.
Collapse
Affiliation(s)
- Min-Woo Ha
- Jeju Research Institute of Pharmaceutical Sciences, College of Pharmacy, Jeju National University, 102 Jejudaehak-ro, Jeju 63243, Jeju-do, Korea;
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, 102 Jejudaehak-ro, Jeju 63243, Jeju-do, Korea
| | - Seung-Mann Paek
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Gyeongnam-do, Korea
| |
Collapse
|
29
|
Spyrou N, Vallianou N, Kadillari J, Dalamaga M. The interplay of obesity, gut microbiome and diet in the immune check point inhibitors therapy era. Semin Cancer Biol 2021; 73:356-376. [PMID: 33989733 DOI: 10.1016/j.semcancer.2021.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 03/22/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022]
|
30
|
Martin M, Sun M, Motolani A, Lu T. The Pivotal Player: Components of NF-κB Pathway as Promising Biomarkers in Colorectal Cancer. Int J Mol Sci 2021; 22:7429. [PMID: 34299049 PMCID: PMC8303169 DOI: 10.3390/ijms22147429] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/04/2021] [Accepted: 07/07/2021] [Indexed: 12/22/2022] Open
Abstract
Over the last several decades, colorectal cancer (CRC) has been one of the most prevalent cancers. While significant progress has been made in both diagnostic screening and therapeutic approaches, a large knowledge gap still remains regarding the early identification and treatment of CRC. Specifically, identification of CRC biomarkers that can help with the creation of targeted therapies as well as increasing the ability for clinicians to predict the biological response of a patient to therapeutics, is of particular importance. This review provides an overview of CRC and its progression stages, as well as the basic types of CRC biomarkers. We then lay out the synopsis of signaling pathways related to CRC, and further highlight the pivotal and multifaceted role of nuclear factor (NF) κB signaling in CRC. Particularly, we bring forth knowledge regarding the tumor microenvironment (TME) in CRC, and its complex interaction with cancer cells. We also provide examples of NF-κB signaling-related CRC biomarkers, and ongoing efforts made at targeting NF-κB signaling in CRC treatment. We conclude and anticipate that with more emerging novel regulators of the NF-κB pathway being discovered, together with their in-depth characterization and the integration of large groups of genomic, transcriptomic and proteomic data, the day of successful development of more ideal NF-κB inhibitors is fast approaching.
Collapse
Affiliation(s)
- Matthew Martin
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, USA; (M.M.); (M.S.); (A.M.)
| | - Mengyao Sun
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, USA; (M.M.); (M.S.); (A.M.)
| | - Aishat Motolani
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, USA; (M.M.); (M.S.); (A.M.)
| | - Tao Lu
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, USA; (M.M.); (M.S.); (A.M.)
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 975 West Walnut Street, Indianapolis, IN 46202, USA
| |
Collapse
|
31
|
Zhao Q, Jiang D, Sun X, Mo Q, Chen S, Chen W, Gui R, Ma X. Biomimetic nanotherapy: core-shell structured nanocomplexes based on the neutrophil membrane for targeted therapy of lymphoma. J Nanobiotechnology 2021; 19:179. [PMID: 34120620 PMCID: PMC8201715 DOI: 10.1186/s12951-021-00922-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/01/2021] [Indexed: 12/24/2022] Open
Abstract
Background Non-Hodgkin’s lymphoma (NHL) is a malignant disease of lymphoid tissue. At present, chemotherapy is still the main method for the treatment of NHL. R-CHOP can significantly improve the survival rate of patients. Unfortunately, DOX is the main cytotoxic drug in R-CHOP and it can lead to adverse reactions. Therefore, it is particularly important to uncover new treatment options for NHL. Results In this study, a novel anti-tumor nanoparticle complex Nm@MSNs-DOX/SM was designed and constructed in this study. Mesoporous silica nanoparticles (MSNs) loaded with Doxorubicin (DOX) and anti-inflammatory drugs Shanzhiside methylester (SM) were used as the core of nanoparticles. Neutrophil membrane (Nm) can be coated with multiple nanonuclei as a shell. DOX combined with SM can enhance the anti-tumor effect, and induce apoptosis of lymphoma cells and inhibit the expression of inflammatory factors related to tumorigenesis depending on the regulation of Bcl-2 family-mediated mitochondrial pathways, such as TNF-α and IL-1β. Consequently, the tumor microenvironment (TME) was reshaped, and the anti-tumor effect of DOX was amplified. Besides, Nm has good biocompatibility and can enhance the EPR effect of Nm@MSNs-DOX/SM and increase the effect of active targeting tumors. Conclusions This suggests that the Nm-modified drug delivery system Nm@MSNs-DOX/SM is a promising targeted chemotherapy and anti-inflammatory therapy nanocomplex, and may be employed as a specific and efficient anti-Lymphoma therapy. ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-00922-4.
Collapse
Affiliation(s)
- Qiangqiang Zhao
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, 410013, People's Republic of China.,Department of Hematology, The Qinghai Provincial People's Hospital, Xining, 810007, People's Republic of China
| | - Duanfeng Jiang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, 410013, People's Republic of China
| | - Xiaoying Sun
- Nursing School, Soochow University, Suzhou, 215000, People's Republic of China.,Department of Emergency, The Qinghai Provincial People's Hospital, Xining, 810007, People's Republic of China
| | - Qiuyu Mo
- Department of Hematology, Affiliated Hospital of Guilin Medical University, Guilin, 541002, People's Republic of China
| | - Shaobin Chen
- Department of Hematology, The Qinghai Provincial People's Hospital, Xining, 810007, People's Republic of China
| | - Wansong Chen
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, People's Republic of China
| | - Rong Gui
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, 410013, People's Republic of China.
| | - Xianjun Ma
- Department of Blood Transfusion, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China.
| |
Collapse
|
32
|
Local Immune Changes in Early Stages of Inflammation and Carcinogenesis Correlate with the Collagen Scaffold Changes of the Colon Mucosa. Cancers (Basel) 2021; 13:cancers13102463. [PMID: 34070183 PMCID: PMC8158480 DOI: 10.3390/cancers13102463] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/09/2021] [Accepted: 05/13/2021] [Indexed: 11/22/2022] Open
Abstract
Simple Summary Chronic colitis and colon cancer develop for alteration of the mucosa homeostatic regulation, also involving TGF-β1. Dextran sulphate sodium (DSS)-induced colitis and azoxymethane (AOM)-induced colorectal carcinogenesis animal models allow for the investigation of the pathological evolution steps. Since chronic inflammation is a common factor, we aimed to explore in rat models the colon mucosa immunological and structural conditions at one month after the end of the inductions, a transition period between acute effects and established lesions. We found, in comparison to healthy controls, downregulation of inflammatory cytokines (except IL-6) and of TGF-β1. At the same time, the collagen scaffold was significantly remodelled in both groups. We conclude that the pro-inflammatory cytokines, in front of a downregulated TGF-β1, sustained a smouldering inflammation with structural changes preparing the niche of both pathologies (ulcerative colitis with fibrosis; tumour). The collagen scaffold changes pointing to an unnoticed inflammation may be suggested as a possible pre-neoplastic condition marker. Abstract Continuous activation of the immune system inside a tissue can lead to remodelling of the tissue structure and creation of a specific microenvironment, such as during the tumour development. Chronic inflammation is a central player in stimulating changes that alter the tissue stroma and can lead to fibrotic evolution. In the colon mucosa, regulatory mechanisms, including TGF-β1, avoid damaging inflammation in front of the continuous challenge by the intestinal microbiome. Inducing either DSS colitis or AOM colorectal carcinogenesis in AVN-Wistar rats, we evaluated at one month after the end of each treatment whether immunological changes and remodelling of the collagen scaffold were already in development. At this time point, we found in both models a general downregulation of pro-inflammatory cytokines and even of TGF-β1, but not of IL-6. Moreover, we demonstrated by multi-photon microscopy the simultaneously presence of pro-fibrotic remodelling of the collagen scaffold, with measurable changes in comparison to the control mucosa. The scaffold was significantly modified depending on the type of induced stimulation. These results suggest that at one month after the end of the DSS or AOM inductions, a smouldering inflammation is present in both induced conditions, since the pro-inflammatory cytokines still exceed, in proportion, the local homeostatic regulation of which TGF-β1 is a part (inflammatory threshold). Such an inflammation appears sufficient to sustain remodelling of the collagen scaffold that may be taken as a possible pathological marker for revealing pre-neoplastic inflammation.
Collapse
|
33
|
Mishra R, Rajsiglová L, Lukáč P, Tenti P, Šima P, Čaja F, Vannucci L. Spontaneous and Induced Tumors in Germ-Free Animals: A General Review. ACTA ACUST UNITED AC 2021; 57:medicina57030260. [PMID: 33799911 PMCID: PMC8002107 DOI: 10.3390/medicina57030260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/27/2022]
Abstract
Cancer, bacteria, and immunity relationships are much-debated topics in the last decade. Microbiome’s importance for metabolic and immunologic modulation of the organism adaptation and responses has become progressively evident, and models to study these relationships, especially about carcinogenesis, have acquired primary importance. The availability of germ-free (GF) animals, i.e., animals born and maintained under completely sterile conditions avoiding the microbiome development offers a unique tool to investigate the role that bacteria can have in carcinogenesis and tumor development. The comparison between GF animals with the conventional (CV) counterpart with microbiome can help to evidence conditions and mechanisms directly involving bacterial activities in the modulation of carcinogenesis processes. Here, we review the literature about spontaneous cancer and cancer modeling in GF animals since the early studies, trying to offer a practical overview on the argument.
Collapse
Affiliation(s)
- Rajbardhan Mishra
- Laboratory of Immunotherapy, Institute of Microbiology v.v.i., Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (R.M.); (L.R.); (P.L.); (P.T.); (P.Š.); (F.Č.)
| | - Lenka Rajsiglová
- Laboratory of Immunotherapy, Institute of Microbiology v.v.i., Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (R.M.); (L.R.); (P.L.); (P.T.); (P.Š.); (F.Č.)
- Faculty of Science, Charles University, Albertov 6, 12800 Prague, Czech Republic
| | - Pavol Lukáč
- Laboratory of Immunotherapy, Institute of Microbiology v.v.i., Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (R.M.); (L.R.); (P.L.); (P.T.); (P.Š.); (F.Č.)
- Faculty of Science, Charles University, Albertov 6, 12800 Prague, Czech Republic
| | - Paolo Tenti
- Laboratory of Immunotherapy, Institute of Microbiology v.v.i., Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (R.M.); (L.R.); (P.L.); (P.T.); (P.Š.); (F.Č.)
- Faculty of Science, Charles University, Albertov 6, 12800 Prague, Czech Republic
| | - Peter Šima
- Laboratory of Immunotherapy, Institute of Microbiology v.v.i., Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (R.M.); (L.R.); (P.L.); (P.T.); (P.Š.); (F.Č.)
| | - Fabián Čaja
- Laboratory of Immunotherapy, Institute of Microbiology v.v.i., Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (R.M.); (L.R.); (P.L.); (P.T.); (P.Š.); (F.Č.)
- Faculty of Science, Charles University, Albertov 6, 12800 Prague, Czech Republic
| | - Luca Vannucci
- Laboratory of Immunotherapy, Institute of Microbiology v.v.i., Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (R.M.); (L.R.); (P.L.); (P.T.); (P.Š.); (F.Č.)
- Correspondence: ; Tel.: +42-024-106-2394
| |
Collapse
|
34
|
Wang RX, Zhou M, Ma HL, Qiao YB, Li QS. The Role of Chronic Inflammation in Various Diseases and Anti-inflammatory Therapies Containing Natural Products. ChemMedChem 2021; 16:1576-1592. [PMID: 33528076 DOI: 10.1002/cmdc.202000996] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Indexed: 12/13/2022]
Abstract
Chronic inflammation represents a long-term reaction of the body's immune system to noxious stimuli. Such a sustained inflammatory response sometimes results in lasting damage to healthy tissues and organs. In fact, chronic inflammation is implicated in the development and progression of various diseases, including cardiovascular diseases, respiratory diseases, metabolic diseases, neurodegenerative diseases, and even cancers. Targeting nonresolving inflammation thus provides new opportunities for treating relevant diseases. In this review, we will go over several chronic inflammation-associated diseases first with emphasis on the role of inflammation in their pathogenesis. Then, we will summarize a number of natural products that exhibit therapeutic effects against those diseases by acting on different markers in the inflammatory response. We envision that natural products will remain a rich resource for the discovery of new drugs treating diseases associated with chronic inflammation.
Collapse
Affiliation(s)
- Ren-Xiao Wang
- Shanxi Key Laboratory of Innovative Drugs for the, Treatment of Serious Diseases Based on Chronic Inflammation, College of Traditional Chinese Medicines, Shanxi University of Chinese Medicine, Taiyuan, Shanxi, 030619, P. R. China.,Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Mi Zhou
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Hui-Lai Ma
- Shanxi Key Laboratory of Innovative Drugs for the, Treatment of Serious Diseases Based on Chronic Inflammation, College of Traditional Chinese Medicines, Shanxi University of Chinese Medicine, Taiyuan, Shanxi, 030619, P. R. China
| | - Yuan-Biao Qiao
- Shanxi Key Laboratory of Innovative Drugs for the, Treatment of Serious Diseases Based on Chronic Inflammation, College of Traditional Chinese Medicines, Shanxi University of Chinese Medicine, Taiyuan, Shanxi, 030619, P. R. China
| | - Qing-Shan Li
- Shanxi Key Laboratory of Innovative Drugs for the, Treatment of Serious Diseases Based on Chronic Inflammation, College of Traditional Chinese Medicines, Shanxi University of Chinese Medicine, Taiyuan, Shanxi, 030619, P. R. China
| |
Collapse
|
35
|
Kotsiou OS, Papagiannis D, Papadopoulou R, Gourgoulianis KI. Calprotectin in Lung Diseases. Int J Mol Sci 2021; 22:1706. [PMID: 33567747 PMCID: PMC7915440 DOI: 10.3390/ijms22041706] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 12/14/2022] Open
Abstract
Calprotectin (CLP) is a heterodimer formed by two S-100 calcium-binding cytosolic proteins, S100A8 and S100A9. It is a multifunctional protein expressed mainly by neutrophils and released extracellularly by activated or damaged cells mediating a broad range of physiological and pathological responses. It has been more than 20 years since the implication of S100A8/A9 in the inflammatory process was shown; however, the evaluation of its role in the pathogenesis of respiratory diseases or its usefulness as a biomarker for the appropriate diagnosis and prognosis of lung diseases have only gained attention in recent years. This review aimed to provide current knowledge regarding the potential role of CLP in the pathophysiology of lung diseases and describe how this knowledge is, up until now, translated into daily clinical practice. CLP is involved in numerous cellular processes in lung health and disease. In addition to its anti-microbial functions, CLP also serves as a molecule with pro- and anti-tumor properties related to cell survival and growth, angiogenesis, DNA damage response, and the remodeling of the extracellular matrix. The findings of this review potentially introduce CLP in daily clinical practice within the spectrum of respiratory diseases.
Collapse
Affiliation(s)
- Ourania S. Kotsiou
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, 41110 Larissa, Greece;
- Department of Nursing, Faculty of Medicine, University of Thessaly, 41110 Larissa, Greece;
| | - Dimitrios Papagiannis
- Department of Nursing, Faculty of Medicine, University of Thessaly, 41110 Larissa, Greece;
| | - Rodanthi Papadopoulou
- Human Nutrition, School of Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow Royal Infirmary, Glasgow G31 2ER, UK;
| | | |
Collapse
|
36
|
Zhu Z, Liu JB, Liu X, Qian L. Association of interleukin 10 rs1800896 polymorphism with susceptibility to breast cancer: a meta-analysis. J Int Med Res 2021; 48:300060520904863. [PMID: 32349574 PMCID: PMC7218478 DOI: 10.1177/0300060520904863] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective To evaluate the correlation between interleukin 10 (IL-10) −1082A/G polymorphism (rs1800896) and breast cancers by performing a meta-analysis. Methods The Embase and Medline databases were searched through 1 September 2018 to identify qualified articles. Odds ratios (OR) and corresponding 95% confidence intervals (CIs) were applied to evaluate associations. Results In total, 14 case-control studies, including 5320 cases and 5727 controls, were analyzed. We detected significant associations between the IL10 −1082 G/G genotype and risk of breast cancer (AA + AG vs. GG: OR = 0.88, 95% CI = 0.80–0.97). Subgroup analyses confirmed a significant association in Caucasian populations (OR = 0.89, 95% CI = 0.80–0.99), in population-based case-control studies (OR = 0.87, 95% CI = 0.78–0.96), and in studies with ≥500 subjects (OR = 0.88, 95% CI = 0.79–0.99) under the recessive model (AA + AG vs. GG). No associations were found in Asian populations. Conclusions The IL10 −1082A/G polymorphism is associated with an increased risk of breast cancer. The association between IL10 −1082 G/G genotype and increased risk of breast cancer is more significant in Caucasians, in population-based studies, and in larger studies.
Collapse
Affiliation(s)
- ZiYin Zhu
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ji-Bin Liu
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Xi Liu
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - LinXue Qian
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
37
|
Jarosz-Biej M, Smolarczyk R, Cichoń T, Szala S. Reprogramming of Tumor Microenvironment in Therapy. CANCER IMMUNOLOGY 2021:403-412. [DOI: 10.1007/978-3-030-50287-4_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
38
|
Wen B, Zhang C, Zhou J, Zhang Z, Che Q, Cao H, Bai Y, Guo J, Su Z. Targeted treatment of alcoholic liver disease based on inflammatory signalling pathways. Pharmacol Ther 2020; 222:107752. [PMID: 33253739 DOI: 10.1016/j.pharmthera.2020.107752] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023]
Abstract
Targeted therapy is an emerging treatment strategy for alcoholic liver disease (ALD). Inflammation plays an important role in the occurrence and development of ALD, and is a key choice for its targeted treatment, and anti-inflammatory treatment has been considered beneficial for liver disease. Surprisingly, immune checkpoint inhibitors have become important therapeutic agents for hepatocellular carcinoma (HCC). Moreover, studies have shown that the combination of inflammatory molecule inhibitors and immune checkpoint inhibitors can exert better effects than either alone in mouse models of HCC. This review discusses the mechanism of hepatic ethanol metabolism and the conditions under which inflammation occurs. In addition, we focus on the potential molecular targets in inflammatory signalling pathways and summarize the potential targeted inhibitors and immune checkpoint inhibitors, providing a theoretical basis for the targeted treatment of ALD and the development of new combination therapy strategies for HCC.
Collapse
Affiliation(s)
- Bingjian Wen
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chengcheng Zhang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jingwen Zhou
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhengyan Zhang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd., Guangzhou 510663, China
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
39
|
Shi J, Wang K, Xiong Z, Yuan C, Wang C, Cao Q, Yu H, Meng X, Xie K, Cheng Z, Yang H, Chen K, Zhang X. Impact of inflammation and immunotherapy in renal cell carcinoma. Oncol Lett 2020; 20:272. [PMID: 33014151 PMCID: PMC7520756 DOI: 10.3892/ol.2020.12135] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 07/23/2020] [Indexed: 12/13/2022] Open
Abstract
Substantial research attention has been directed at exploring the mechanisms and treatment of renal cell carcinoma (RCC). Indeed, the association between inflammation and tumor phenotypes has been at the center of cancer research. Concomitant with research on the inflammation response and inflammatory molecules involved in RCC, new breakthroughs have emerged. A large body of knowledge now shows that treatments targeting inflammation and immunity in RCC provide substantial clinical benefits. Adequate analysis and a better understanding of the mechanisms of inflammatory factors in the occurrence and progression of RCC are highly desirable. Currently, numerous RCC treatments targeted at inflammation and immunotherapy are available. The current review describes in detail the link between inflammation and RCC.
Collapse
Affiliation(s)
- Jian Shi
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Keshan Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhiyong Xiong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Changfei Yuan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Cheng Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qi Cao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Huang Yu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiangui Meng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Kairu Xie
- Department of Pathogenic Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhixian Cheng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ke Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
40
|
Nouri Z, Fakhri S, Nouri K, Wallace CE, Farzaei MH, Bishayee A. Targeting Multiple Signaling Pathways in Cancer: The Rutin Therapeutic Approach. Cancers (Basel) 2020; 12:E2276. [PMID: 32823876 PMCID: PMC7463935 DOI: 10.3390/cancers12082276] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022] Open
Abstract
Multiple dysregulated signaling pathways are implicated in the pathogenesis of cancer. The conventional therapies used in cancer prevention/treatment suffer from low efficacy, considerable toxicity, and high cost. Hence, the discovery and development of novel multi-targeted agents to attenuate the dysregulated signaling in cancer is of great importance. In recent decades, phytochemicals from dietary and medicinal plants have been successfully introduced as alternative anticancer agents due to their ability to modulate numerous oncogenic and oncosuppressive signaling pathways. Rutin (also known as rutoside, quercetin-3-O-rutinoside and sophorin) is an active plant-derived flavonoid that is widely distributed in various vegetables, fruits, and medicinal plants, including asparagus, buckwheat, apricots, apples, cherries, grapes, grapefruit, plums, oranges, and tea. Rutin has been shown to target various inflammatory, apoptotic, autophagic, and angiogenic signaling mediators, including nuclear factor-κB, tumor necrosis factor-α, interleukins, light chain 3/Beclin, B cell lymphoma 2 (Bcl-2), Bcl-2 associated X protein, caspases, and vascular endothelial growth factor. A comprehensive and critical analysis of the anticancer potential of rutin and associated molecular targets amongst various cancer types has not been performed previously. Accordingly, the purpose of this review is to present an up-to-date and critical evaluation of multiple cellular and molecular mechanisms through which the anticancer effects of rutin are known to be exerted. The current challenges and limitations as well as future directions of research are also discussed.
Collapse
Affiliation(s)
- Zeinab Nouri
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran;
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
| | - Keyvan Nouri
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran;
| | - Carly E. Wallace
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
| |
Collapse
|
41
|
Intercellular Bioimaging and Biodistribution of Gold Nanoparticle-Loaded Macrophages for Targeted Drug Delivery. ELECTRONICS 2020. [DOI: 10.3390/electronics9071105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In order to effectively apply nanoparticles to clinical use, macrophages have been used as vehicles to deliver genes, drugs or nanomaterials into tumors. In this study, the effectiveness of macrophage as a drug delivery system was validated by biodistribution imaging modalities at intercellular and ex vivo levels. We focused on biodistribution imaging, namely, the characterization of the gold nanoparticle-loaded macrophages using intracellular holotomography and target delivery efficiency analysis using ex vivo fluorescence imaging techniques. In more detail, gold nanoparticles (AuNPs) were prepared with trisodium citrate method and loaded into macrophage cells (RAW 264.7). First, AuNPs loading into macrophages was confirmed using the conventional ultraviolet-visible (UV-VIS) spectroscopy and inductively coupled plasma-mass spectrometry (ICP-MS). Then, the holotomographic imaging was employed to characterize the intracellular biodistribution of the AuNPs-loaded macrophages. The efficacy of target delivery of the well AuNPs uptake macrophages was studied in a mouse model, established via lipopolysaccharide (LPS)-induced inflammation. The fluorescent images and the ex vivo ICP-MS evaluated the delivery efficiency of the AuNPs-loaded macrophages. Results revealed that the holotomographic imaging techniques can be promising modalities to understand intracellular biodistribution and ex vivo fluorescence imaging can be useful to validate the target delivery efficacy of the AuNPs-loaded macrophages.
Collapse
|
42
|
Amplification of transglutaminase 2 enhances tumor-promoting inflammation in gastric cancers. Exp Mol Med 2020; 52:854-864. [PMID: 32467608 PMCID: PMC7272405 DOI: 10.1038/s12276-020-0444-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor-promoting inflammation is a hallmark of cancer and is highly associated with tumor progression, angiogenesis, and metastasis. Tumor-associated macrophages (TAMs) are major drivers of tumor-promoting inflammation, but due to the complexity of the tumor microenvironment, the detailed regulatory mechanisms are still under investigation. Here, we investigated a novel role for transglutaminase 2 (TGM2) in the development of tumor-promoting inflammation and recruitment of TAMs to gastric cancer (GC) tissues. When estimated by array comparative genomic hybridization and droplet digital PCR, the copy numbers of the TGM2 gene were amplified in 13.6% (14/103) of GC patients and positively associated with TGM2 expression. Gene set enrichment analysis of expression microarray data for GC samples with high or low TGM2 expression showed that increased TGM2 expression was associated with tumor-promoting inflammation in GC. In addition, the expression of TGM2 was correlated with the expression of markers for macrophages, neutrophils, blood vessels, and lymphatic vessels. Overexpression of TGM2 in GC cells augmented the IL-1β-induced secretion of macrophage-recruiting chemokines and NF-κB activation. TGM2 protein levels were associated with the expression levels of the macrophage marker CD163 in human GC tissue samples. Moreover, GC patients with high expression of TGM2 had a worse prognosis than those with low expression of TGM2. These results suggest TGM2 as a novel regulator of the tumor microenvironment of GC and provide a promising target for constraining tumor-promoting inflammation. An enzyme linked to harmful inflammation that promotes tumor growth could be a target for stomach cancer treatment. The expression of the enzyme transglutaminase 2 (TGM2) has been shown to increase in several types of cancer, but its precise role has been unclear. Sung-Yup Cho at Seoul National University College of Medicine, Korea and co-workers measured copy numbers and expression levels of TGM2 in gastric cancer cell lines and in tissue samples from patients undergoing gastrectomy. They found that higher levels of TGM2 were closely associated with increases in various genes that promote the recruitment and activity of tumor-associated macrophages, malfunctioning white blood cells that drive tumor-promoting inflammation. In addition, stomach cancer patients with higher expression of TGM2 had poorer prognoses, indicating the potential therapeutic value of targeting this enzyme.
Collapse
|
43
|
Zhao X, Qi X, Lian W, Tong X, Wang H, Su L, Wei P, Zhuang Z, Gong J, Bai L. Trichomicin Suppresses Colorectal Cancer via Comprehensive Regulation of IL-6 and TNFα in Tumor Cells, TAMs, and CAFs. Front Pharmacol 2020; 11:386. [PMID: 32317968 PMCID: PMC7146085 DOI: 10.3389/fphar.2020.00386] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 03/13/2020] [Indexed: 12/19/2022] Open
Abstract
Trichomicin, a small-molecule compound isolated from fungi, has been identified with bioactivity of antitumor. In this study, a colon cancer subcutaneous mice model was used to evaluate the antitumor effects of Trichomicin in vivo. Treatment with Trichomicin significantly inhibited tumor growth in a xenograft mouse colon cancer model. The underlying molecular mechanism has also been investigated through the quantification of relevant proteins. The expression levels of IL-6 and TNFα were reduced in tumor tissues of mice treated with Trichomicin, which was consistent with results of in vitro experiments in which Trichomicin suppressed the expression of IL-6 and TNFα in tumor and stromal cells. In addition, Trichomicin inhibited TNFα-induced activation of NF-κB and basal Stat3 signaling in vitro, which resulted in reduced expression of the immune checkpoint protein PD-L1 in tumor and stromal cells. Conclusively, Trichomicin, a promising new drug candidate with antitumor activity, exerted antitumor effects against colon cancer through inhibition of the IL-6 and TNFα signaling pathways.
Collapse
Affiliation(s)
- Xi Zhao
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoqiang Qi
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Surgery and Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO, United States
| | - Wenrui Lian
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Gastrointestinal Surgery & Clinical Medicine Research Center, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xin Tong
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Gastrointestinal Surgery & Clinical Medicine Research Center, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hua Wang
- Department of Gastrointestinal Surgery & Clinical Medicine Research Center, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Liya Su
- Department of Gastrointestinal Surgery & Clinical Medicine Research Center, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Ping Wei
- Department of Medical Immunology, Basic Medical College, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Zhuochen Zhuang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianhua Gong
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liping Bai
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Chen X, Xu C, Hong S, Xia X, Cao Y, McDermott J, Mu Y, Han JDJ. Immune Cell Types and Secreted Factors Contributing to Inflammation-to-Cancer Transition and Immune Therapy Response. Cell Rep 2020; 26:1965-1977.e4. [PMID: 30759403 DOI: 10.1016/j.celrep.2019.01.080] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 09/17/2018] [Accepted: 01/22/2019] [Indexed: 12/23/2022] Open
Abstract
Although chronic inflammation increases many cancers' risk, how inflammation facilitates cancer development is still not well studied. Recognizing whether and when inflamed tissues transition to cancerous tissues is of utmost importance. To unbiasedly infer molecular events, immune cell types, and secreted factors contributing to the inflammation-to-cancer (I2C) transition, we develop a computational package called "SwitchDetector" based on liver, gastric, and colon cancer I2C data. Using it, we identify angiogenesis associated with a common critical transition stage for multiple I2C events. Furthermore, we infer infiltrated immune cell type composition and their secreted or suppressed extracellular proteins to predict expression of important transition stage genes. This identifies extracellular proteins that may serve as early-detection biomarkers for pre-cancer and early-cancer stages. They alone or together with I2C hallmark angiogenesis genes are significantly related to cancer prognosis and can predict immune therapy response. The SwitchDetector and I2C database are publicly available at www.inflammation2cancer.org.
Collapse
Affiliation(s)
- Xingwei Chen
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chi Xu
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shengjun Hong
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xian Xia
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaqiang Cao
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Joseph McDermott
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Yonglin Mu
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing-Dong J Han
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
45
|
Dadiani M, Necula D, Kahana-Edwin S, Oren N, Baram T, Marin I, Morzaev-Sulzbach D, Pavlovski A, Balint-Lahat N, Anafi L, Wiemann S, Korner C, Gal-Yam EN, Avivi C, Kaufman B, Barshack I, Ben-Baruch A. TNFR2+ TILs are significantly associated with improved survival in triple-negative breast cancer patients. Cancer Immunol Immunother 2020; 69:1315-1326. [PMID: 32198536 DOI: 10.1007/s00262-020-02549-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/10/2020] [Indexed: 12/12/2022]
Abstract
In view of the relatively limited efficacy of immunotherapies targeting the PD-1-PD-L1 axis in triple-negative breast cancer (TNBC) and of published reports on tumor-promoting roles of TNFR2+ tumor-infiltrating lymphocytes (TNFR2+ TILs), we determined the incidence of TNFR2+ TILs in TNBC patient tumors, their association with disease outcome and relations with PD-1+ TILs. Using a cohort of treatment-naïve TNBC patients with long follow-up (n = 70), we determined the presence of TNFR2+ TILs and PD-1+ TILs by immunohistochemistry. TILs (≥ 1% of cellular mass) and TNFR2+ TILs (≥ 1% of total TILs) were detected in 96% and 74% of tumors, respectively. The presence of TILs at > 5% of tumor cell mass ("Positive TILs"), as well as of positive TNFR2+ TILs (> 5%), was independently associated with good prognosis, and combination of both parameters demonstrated superior outcome relative to their lower levels. PD1+ TILs (> 5/hot spot) were detected in 63% of patients. High levels of PD-1+ TILs (> 20/hot spot) showed an unfavorable disease outcome, and in their presence, the favorable outcome of positive TNFR2+ TILs was ablated. Thus, TNFR2+ TILs are strongly connected to improved prognosis in TNBC; these findings suggest that TNFR2+ TILs have favorable effects in TNBC patients, unlike the tumor-promoting roles attributed to them in other cancer systems. Overall, our observations propose that the TNFR2+ TIL subset should not be targeted in the course of TNBC therapy; rather, its beneficial impacts may become into power when anti-PD-1 regimens-that may potentiate immune activities-are administered to TNBC patients.
Collapse
Affiliation(s)
- Maya Dadiani
- Cancer Research Center, Sheba Medical Center, Ramat Gan, Israel
| | - Daniela Necula
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel
| | | | - Nino Oren
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Tamir Baram
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Irina Marin
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel
| | | | - Anya Pavlovski
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel
| | | | - Liat Anafi
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cindy Korner
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Camila Avivi
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel
| | - Bella Kaufman
- Breast Oncology Institute, Sheba Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Iris Barshack
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adit Ben-Baruch
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel.
| |
Collapse
|
46
|
Carbotti G, Petretto A, Naschberger E, Stürzl M, Martini S, Mingari MC, Filaci G, Ferrini S, Fabbi M. Cytokine-Induced Guanylate Binding Protein 1 (GBP1) Release from Human Ovarian Cancer Cells. Cancers (Basel) 2020; 12:E488. [PMID: 32093058 PMCID: PMC7072386 DOI: 10.3390/cancers12020488] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/08/2020] [Accepted: 02/17/2020] [Indexed: 01/01/2023] Open
Abstract
We showed that IL-27 shares several effects with IFN-γ in human cancer cells. To identify novel extracellular mediators, potentially involved in epithelial ovarian cancer (EOC) biology, we analyzed the effect of IL-27 or IFN-γ on the secretome of cultured EOC cells by mass-spectrometry (nano-UHPLC-MS/MS). IL-27 and IFN-γ modulate the release of a limited fraction of proteins among those induced in the whole cell. We focused our attention on GBP1, a guanylate-binding protein and GTPase, which mediates several biological activities of IFNs. Cytokine treatment induced GBP1, 2, and 5 expressions in EOC cells, but only GBP1 was secreted. ELISA and immunoblotting showed that cytokine-stimulated EOC cells release full-length GBP1 in vitro, through non-classical pathways, not involving microvesicles. Importantly, full-length GBP1 accumulates in the ascites of most EOC patients and ex-vivo EOC cells show constitutive tyrosine-phosphorylated STAT1/3 proteins and GBP1 expression, supporting a role for Signal Transducer And Activator Of Transcription (STAT)-activating cytokines in vivo. High GBP1 gene expression correlates with better overall survival in the TCGA (The Cancer Genome Atlas) dataset of EOC. In addition, GBP1 transfection partially reduced EOC cell viability in an MTT assay. Our data show for the first time that cytokine-stimulated tumor cells release soluble GBP1 in vitro and in vivo and suggest that GBP1 may have anti-tumor effects in EOC.
Collapse
Affiliation(s)
- Grazia Carbotti
- IRCCS Ospedale Policlinico San Martino, Biotherapies Unit, Largo R. Benzi 10, 16132 Genoa, Italy; (G.C.); (G.F.)
| | - Andrea Petretto
- Core Facilities—Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genoa, Italy;
| | - Elisabeth Naschberger
- Division of Molecular and Experimental Surgery, University Medical Center Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Schwabachanlage 12, 91054 Erlangen, Germany; (E.N.); (M.S.)
| | - Michael Stürzl
- Division of Molecular and Experimental Surgery, University Medical Center Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Schwabachanlage 12, 91054 Erlangen, Germany; (E.N.); (M.S.)
| | - Stefania Martini
- IRCCS Ospedale Policlinico San Martino, Immunology Unit, Largo R. Benzi 10, 16132 Genoa, Italy; (S.M.); (M.C.M.)
| | - Maria Cristina Mingari
- IRCCS Ospedale Policlinico San Martino, Immunology Unit, Largo R. Benzi 10, 16132 Genoa, Italy; (S.M.); (M.C.M.)
- Department of Experimental Medicine and Centre of Excellence for Biomedical Research, University of Genoa, Via L.B. Alberti 2, 16132 Genoa, Italy
| | - Gilberto Filaci
- IRCCS Ospedale Policlinico San Martino, Biotherapies Unit, Largo R. Benzi 10, 16132 Genoa, Italy; (G.C.); (G.F.)
- Centre of Excellence for Biomedical Research and Department of Internal Medicine, University of Genoa, Via De Toni 14, 16132 Genoa, Italy
| | - Silvano Ferrini
- IRCCS Ospedale Policlinico San Martino, Biotherapies Unit, Largo R. Benzi 10, 16132 Genoa, Italy; (G.C.); (G.F.)
| | - Marina Fabbi
- IRCCS Ospedale Policlinico San Martino, Biotherapies Unit, Largo R. Benzi 10, 16132 Genoa, Italy; (G.C.); (G.F.)
| |
Collapse
|
47
|
Li J, Lin TY, Chen L, Liu Y, Dian MJ, Hao WC, Lin XL, Li XY, Li YL, Lian M, Chen HW, Jia JS, Zhang XL, Xiao SJ, Xiao D, Sun Y. miR-19 regulates the expression of interferon-induced genes and MHC class I genes in human cancer cells. Int J Med Sci 2020; 17:953-964. [PMID: 32308549 PMCID: PMC7163354 DOI: 10.7150/ijms.44377] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/19/2020] [Indexed: 01/13/2023] Open
Abstract
MicroRNA-19 (miR-19) is identified as the key oncogenic component of the miR-17-92 cluster. When we explored the functions of the dysregulated miR-19 in lung cancer, microarray-based data unexpectedly demonstrated that some immune and inflammatory response genes (i.e., IL32, IFI6 and IFIT1) were generally down-regulated by miR-19 overexpression in A549 cells, which prompted us to fully investigate whether the miR-19 family (i.e., miR-19a and miR-19b-1) was implicated in regulating the expression of immune and inflammatory response genes in cancer cells. In the present study, we observed that miR-19a or miR-19b-1 overexpression by miRNA mimics in the A549, HCC827 and CNE2 cells significantly downregulated the expression of interferon (IFN)-regulated genes (i.e., IRF7, IFI6, IFIT1, IFITM1, IFI27 and IFI44L). Furthermore, the ectopic miR-19a or miR-19b-1 expression in the A549, HCC827, CNE2 and HONE1 cells led to a general downward trend in the expression profile of major histocompatibility complex (MHC) class I genes (such as HLA-B, HLA-E, HLA-F or HLA-G); conversely, miR-19a or miR-19b-1 inhibition by the miRNA inhibitor upregulated the aforementioned MHC Class I gene expression, suggesting that miR-19a or miR-19b-1 negatively modulates MHC Class I gene expression. The miR-19a or miR-19b-1 mimics reduced the expression of interleukin (IL)-related genes (i.e., IL1B, IL11RA and IL6) in the A549, HCC827, CNE2 or HONE1 cells. The ectopic expression of miR-19a or miR-19b-1 downregulated IL32 expression in the A549 and HCC827 cells and upregulated IL32 expression in CNE2 and HONE1 cells. In addition, enforced miR-19a or miR-19b-1 expression suppressed IL-6 production by lung cancer and nasopharyngeal carcinoma (NPC) cells. Taken together, these findings demonstrate, for the first time, that miR-19 can modulate the expression of IFN-induced genes and MHC class I genes in human cancer cells, suggesting a novel role of miR-19 in linking inflammation and cancer, which remains to be fully characterized.
Collapse
Affiliation(s)
- Jing Li
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Radiotherapy Center, the First People's Hospital of Chenzhou, Chenzhou 423000, China
| | - Tao-Yan Lin
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lin Chen
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Yu Liu
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Mei-Juan Dian
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Wei-Chao Hao
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Lin Lin
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Yan Li
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Yong-Long Li
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Mei Lian
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Heng-Wei Chen
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Jun-Shuang Jia
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Ling Zhang
- Department of Physiology, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China
| | - Sheng-Jun Xiao
- Department of Pathology, the Second Affiliated Hospital, Guilin Medical University, Guilin 541199, China
| | - Dong Xiao
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Yan Sun
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
48
|
Tsai CL, Chang JS, Yu MC, Lee CH, Chen TC, Chuang WY, Kuo WL, Lin CC, Lin SM, Hsieh SY. Functional Genomics Identifies Hepatitis-Induced STAT3-TYRO3-STAT3 Signaling as a Potential Therapeutic Target of Hepatoma. Clin Cancer Res 2019; 26:1185-1197. [PMID: 31831556 DOI: 10.1158/1078-0432.ccr-18-3531] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 03/27/2019] [Accepted: 12/09/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Hepatitis promotes the development and recurrence of hepatocellular carcinoma (HCC). Receptor tyrosine kinases (RTK) play critical roles in the development of many cancers. We explored the potential roles of RTKs in hepatitis-related liver cancers. EXPERIMENTAL DESIGN We conducted loss-of-function screening to elucidate the roles of RTKs in the development of HCC in vitro and in vivo. RESULTS Many RTKs were coexpressed in HCC and were involved in tumor development and growth. Of these, TYRO3 promoted tumor growth and was clinically associated with hepatitis activity and poor prognosis. In mice, chemical-induced hepatitis transcriptionally activated Tyro3 expression via IL-6/IL6R-STAT3 signaling. Moreover, hepatitis-associated apoptotic cells facilitated the presentation of GAS6, a TYRO3 ligand, to further activate TYRO3-mediated signaling. Furthermore, TYRO3 activation elicited intracellular SRC- and STAT3 signaling. In mice, hepatitis and Tyro3 synergistically promoted HCC development. Silencing TYRO3 expression or inhibiting its kinase activity suppressed xenograft HCC growth in nude mice. CONCLUSIONS Many RTKs are simultaneously involved in HCC development. Hepatitis exerts dual effects on the activation of TYRO3-mediated signaling in HCC cells, which further elicits the "TYRO3-STAT3-TYRO3" signaling loop to facilitate tumor growth. Our findings unveil a previously unrecognized link between RTKs and hepatitis-associated HCC and suggest TYRO3 as a marker and therapeutic target for the HCCs with higher hepatitis activity.
Collapse
Affiliation(s)
- Chia-Liang Tsai
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jeng-Shou Chang
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ming-Chin Yu
- Department of General Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chern-Horng Lee
- Department of General Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Tse-Ching Chen
- Department of Pathology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Wen-Yu Chuang
- Department of Pathology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Wei-Liang Kuo
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chen-Chun Lin
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shi-Ming Lin
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Sen-Yung Hsieh
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan. .,Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
49
|
Setaro F, Wennink JWH, Mäkinen PI, Holappa L, Trohopoulos PN, Ylä-Herttuala S, van Nostrum CF, de la Escosura A, Torres T. Amphiphilic phthalocyanines in polymeric micelles: a supramolecular approach toward efficient third-generation photosensitizers. J Mater Chem B 2019; 8:282-289. [PMID: 31803886 DOI: 10.1039/c9tb02014d] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In this paper we describe a straightforward supramolecular strategy to encapsulate silicon phthalocyanine (SiPc) photosensitizers (PS) in polymeric micelles made of poly(ε-caprolactone)-b-methoxypoly(ethylene glycol) (PCL-PEG) block copolymers. While PCL-PEG micelles are promising nanocarriers based on their biocompatibility and biodegradability, the design of our new PS favors their encapsulation. In particular, they combine two axial benzoyl substituents, each of them carrying either three hydrophilic methoxy(triethylenoxy) chains (1), three hydrophobic dodecyloxy chains (3), or both kinds of chains (2). The SiPc derivatives 1 and 2 are therefore amphiphilic, with the SiPc unit contributing to the hydrophobic core, while lipophilicity increases along the series, making it possible to correlate the loading efficacy in PCL-PEG micelles with the hydrophobic/hydrophilic balance of the PS structure. This has led to a new kind of third-generation nano-PS that efficiently photogenerates 1O2, while preliminary in vitro experiments demonstrate an excellent cellular uptake and a promising PDT activity.
Collapse
Affiliation(s)
- Francesca Setaro
- Organic Chemistry Department, Universidad Autónoma de Madrid, 28049 Cantoblanco, Madrid, Spain.
| | - Jos W H Wennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands.
| | - Petri I Mäkinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland
| | - Lari Holappa
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland
| | | | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland
| | - Cornelus F van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands.
| | - Andres de la Escosura
- Organic Chemistry Department, Universidad Autónoma de Madrid, 28049 Cantoblanco, Madrid, Spain. and Institute for Advanced Research in Chemical Sciences (IAdChem), 28049 Cantoblanco, Madrid, Spain
| | - Tomas Torres
- Organic Chemistry Department, Universidad Autónoma de Madrid, 28049 Cantoblanco, Madrid, Spain. and Institute for Advanced Research in Chemical Sciences (IAdChem), 28049 Cantoblanco, Madrid, Spain and IMDEA Nanosience, Ciudad Universitaria de Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
50
|
Fagan V, Johansson C, Gileadi C, Monteiro O, Dunford JE, Nibhani R, Philpott M, Malzahn J, Wells G, Faram R, Cribbs AP, Halidi N, Li F, Chau I, Greschik H, Velupillai S, Allali-Hassani A, Bennett J, Christott T, Giroud C, Lewis AM, Huber KVM, Athanasou N, Bountra C, Jung M, Schüle R, Vedadi M, Arrowsmith C, Xiong Y, Jin J, Fedorov O, Farnie G, Brennan PE, Oppermann U. A Chemical Probe for Tudor Domain Protein Spindlin1 to Investigate Chromatin Function. J Med Chem 2019; 62:9008-9025. [PMID: 31550156 DOI: 10.1021/acs.jmedchem.9b00562] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Modifications of histone tails, including lysine/arginine methylation, provide the basis of a "chromatin or histone code". Proteins that contain "reader" domains can bind to these modifications and form specific effector complexes, which ultimately mediate chromatin function. The spindlin1 (SPIN1) protein contains three Tudor methyllysine/arginine reader domains and was identified as a putative oncogene and transcriptional coactivator. Here we report a SPIN1 chemical probe inhibitor with low nanomolar in vitro activity, exquisite selectivity on a panel of methyl reader and writer proteins, and with submicromolar cellular activity. X-ray crystallography showed that this Tudor domain chemical probe simultaneously engages Tudor domains 1 and 2 via a bidentate binding mode. Small molecule inhibition and siRNA knockdown of SPIN1, as well as chemoproteomic studies, identified genes which are transcriptionally regulated by SPIN1 in squamous cell carcinoma and suggest that SPIN1 may have a role in cancer related inflammation and/or cancer metastasis.
Collapse
Affiliation(s)
- Vincent Fagan
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Target Discovery Institute, Nuffield Department of Medicine , University of Oxford , OX3 7FZ Oxford , U.K
| | - Catrine Johansson
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, NIHR Bio-medical Research Centre , University of Oxford , Oxford OX3 7LD , U.K
| | - Carina Gileadi
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, NIHR Bio-medical Research Centre , University of Oxford , Oxford OX3 7LD , U.K
| | - Octovia Monteiro
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Target Discovery Institute, Nuffield Department of Medicine , University of Oxford , OX3 7FZ Oxford , U.K
| | - James E Dunford
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, NIHR Bio-medical Research Centre , University of Oxford , Oxford OX3 7LD , U.K
| | - Reshma Nibhani
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, NIHR Bio-medical Research Centre , University of Oxford , Oxford OX3 7LD , U.K
| | - Martin Philpott
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, NIHR Bio-medical Research Centre , University of Oxford , Oxford OX3 7LD , U.K
| | - Jessica Malzahn
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, NIHR Bio-medical Research Centre , University of Oxford , Oxford OX3 7LD , U.K
| | - Graham Wells
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, NIHR Bio-medical Research Centre , University of Oxford , Oxford OX3 7LD , U.K
| | - Ruth Faram
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, NIHR Bio-medical Research Centre , University of Oxford , Oxford OX3 7LD , U.K
| | - Adam P Cribbs
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, NIHR Bio-medical Research Centre , University of Oxford , Oxford OX3 7LD , U.K
| | - Nadia Halidi
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, NIHR Bio-medical Research Centre , University of Oxford , Oxford OX3 7LD , U.K
| | - Fengling Li
- Structural Genomics Consortium , University of Toronto , 101 College Street , Toronto , Ontario M5G 1L7 , Canada
| | - Irene Chau
- Structural Genomics Consortium , University of Toronto , 101 College Street , Toronto , Ontario M5G 1L7 , Canada
| | - Holger Greschik
- Department of Urology, Center for Clinical Research, Medical Center, Signalling Research Centres BIOSS and CIBSS , University of Freiburg , D-79106 Freiburg , Germany
| | - Srikannathasan Velupillai
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
| | - Abdellah Allali-Hassani
- Structural Genomics Consortium , University of Toronto , 101 College Street , Toronto , Ontario M5G 1L7 , Canada
| | - James Bennett
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Target Discovery Institute, Nuffield Department of Medicine , University of Oxford , OX3 7FZ Oxford , U.K
| | - Thomas Christott
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Target Discovery Institute, Nuffield Department of Medicine , University of Oxford , OX3 7FZ Oxford , U.K
| | - Charline Giroud
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Target Discovery Institute, Nuffield Department of Medicine , University of Oxford , OX3 7FZ Oxford , U.K
| | - Andrew M Lewis
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Target Discovery Institute, Nuffield Department of Medicine , University of Oxford , OX3 7FZ Oxford , U.K
| | - Kilian V M Huber
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Target Discovery Institute, Nuffield Department of Medicine , University of Oxford , OX3 7FZ Oxford , U.K
| | - Nick Athanasou
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, NIHR Bio-medical Research Centre , University of Oxford , Oxford OX3 7LD , U.K
| | - Chas Bountra
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
| | - Manfred Jung
- FRIAS-Freiburg Institute of Advanced Studies , University of Freiburg , 79104 Freiburg , Germany
- Institute of Pharmaceutical Sciences , University of Freiburg , Albertstraße 25 , 79104 Freiburg , Germany
| | - Roland Schüle
- Department of Urology, Center for Clinical Research, Medical Center, Signalling Research Centres BIOSS and CIBSS , University of Freiburg , D-79106 Freiburg , Germany
| | - Masoud Vedadi
- Structural Genomics Consortium , University of Toronto , 101 College Street , Toronto , Ontario M5G 1L7 , Canada
| | - Cheryl Arrowsmith
- Structural Genomics Consortium , University of Toronto , 101 College Street , Toronto , Ontario M5G 1L7 , Canada
| | - Yan Xiong
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences , Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences , Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Oleg Fedorov
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Target Discovery Institute, Nuffield Department of Medicine , University of Oxford , OX3 7FZ Oxford , U.K
| | - Gillian Farnie
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, NIHR Bio-medical Research Centre , University of Oxford , Oxford OX3 7LD , U.K
| | - Paul E Brennan
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Target Discovery Institute, Nuffield Department of Medicine , University of Oxford , OX3 7FZ Oxford , U.K
- Alzheimer's Research UK Oxford Drug Discovery Institute, Nuffield Department of Medicine , University of Oxford , OX3 7FZ Oxford , U.K
| | - Udo Oppermann
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , OX3 7DQ Oxford , U.K
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, NIHR Bio-medical Research Centre , University of Oxford , Oxford OX3 7LD , U.K
- FRIAS-Freiburg Institute of Advanced Studies , University of Freiburg , 79104 Freiburg , Germany
| |
Collapse
|