1
|
Tuerhong N, Yang Y, Wang C, Huang P, Li Q. Interactions between platelets and the cancer immune microenvironment. Crit Rev Oncol Hematol 2024; 199:104380. [PMID: 38718939 DOI: 10.1016/j.critrevonc.2024.104380] [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: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 06/16/2024] Open
Abstract
Cancer is a leading cause of death in both China and developed countries due to its high incidence and low cure rate. Immune function is closely linked to the development and progression of tumors. Platelets, which are primarily known for their role in hemostasis, also play a crucial part in the spread and progression of tumors through their interaction with the immune microenvironment. The impact of platelets on tumor growth and metastasis depends on the type of cancer and treatment method used. This article provides an overview of the relationship between platelets and the immune microenvironment, highlighting how platelets can either protect or harm the immune response and cancer immune escape. We also explore the potential of available platelet-targeting strategies for tumor immunotherapy, as well as the promise of new platelet-targeted tumor therapy methods through further research.
Collapse
Affiliation(s)
- Nuerye Tuerhong
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China; West China Biomedical Big Data Center, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China
| | - Yang Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China; West China Biomedical Big Data Center, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China
| | - Chenyu Wang
- The Second Clinical Medical College, Lanzhou university, No. 222 South Tianshui Road, Gansu, China
| | - Peng Huang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China; West China Biomedical Big Data Center, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China; West China Biomedical Big Data Center, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Han X, Song X, Xiao Z, Zhu G, Gao R, Ni B, Li J. Study on the mechanism of MDSC-platelets and their role in the breast cancer microenvironment. Front Cell Dev Biol 2024; 12:1310442. [PMID: 38404689 PMCID: PMC10884319 DOI: 10.3389/fcell.2024.1310442] [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: 10/11/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are key immunosuppressive cells in the tumor microenvironment (TME) that play critical roles in promoting tumor growth and metastasis. Tumor-associated platelets (TAPs) help cancer cells evade the immune system and promote metastasis. In this paper, we describe the interaction between MDSCs and TAPs, including their generation, secretion, activation, and recruitment, as well as the effects of MDSCs and platelets on the generation and changes in the immune, metabolic, and angiogenic breast cancer (BC) microenvironments. In addition, we summarize preclinical and clinical studies, traditional Chinese medicine (TCM) therapeutic approaches, and new technologies related to targeting and preventing MDSCs from interacting with TAPs to modulate the BC TME, discuss the potential mechanisms, and provide perspectives for future development. The therapeutic strategies discussed in this review may have implications in promoting the normalization of the BC TME, reducing primary tumor growth and distant lung metastasis, and improving the efficiency of anti-tumor therapy, thereby improving the overall survival (OS) and progression-free survival (PFS) of patients. However, despite the significant advances in understanding these mechanisms and therapeutic strategies, the complexity and heterogeneity of MDSCs and side effects of antiplatelet agents remain challenging. This requires further investigation in future prospective cohort studies.
Collapse
Affiliation(s)
- Xinpu Han
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Hematology-Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaotong Song
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhigang Xiao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guanghui Zhu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruike Gao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baoyi Ni
- Department of Oncology, First Hospital of Heilongjiang University of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jie Li
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Shafqat A, Omer MH, Ahmed EN, Mushtaq A, Ijaz E, Ahmed Z, Alkattan K, Yaqinuddin A. Reprogramming the immunosuppressive tumor microenvironment: exploiting angiogenesis and thrombosis to enhance immunotherapy. Front Immunol 2023; 14:1200941. [PMID: 37520562 PMCID: PMC10374407 DOI: 10.3389/fimmu.2023.1200941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/15/2023] [Indexed: 08/01/2023] Open
Abstract
This review focuses on the immunosuppressive effects of tumor angiogenesis and coagulation on the tumor microenvironment (TME). We summarize previous research efforts leveraging these observations and targeting these processes to enhance immunotherapy outcomes. Clinical trials have documented improved outcomes when combining anti-angiogenic agents and immunotherapy. However, their overall survival benefit over conventional therapy remains limited and certain tumors exhibit poor response to anti-angiogenic therapy. Additionally, whilst preclinical studies have shown several components of the tumor coagulome to curb effective anti-tumor immune responses, the clinical studies reporting combinations of anticoagulants with immunotherapies have demonstrated variable treatment outcomes. By reviewing the current state of the literature on this topic, we address the key questions and future directions in the field, the answers of which are crucial for developing effective strategies to reprogram the TME in order to further the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Mohamed H. Omer
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | | | - Ali Mushtaq
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Eman Ijaz
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Zara Ahmed
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Khaled Alkattan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | |
Collapse
|
4
|
Pan Y, Wang Y, Wang Y, Xu S, Jiang F, Han Y, Hu M, Liu Z. Platelet-derived microvesicles (PMVs) in cancer progression and clinical applications. Clin Transl Oncol 2023; 25:873-881. [PMID: 36417084 DOI: 10.1007/s12094-022-03014-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/12/2022] [Indexed: 11/24/2022]
Abstract
Platelet-derived microvesicles (PMVs), the microvesicles with the highest concentration in the bloodstream, play a key role in the regulation of hemostasis, inflammation, and angiogenesis. PMVs have recently been identified as key factors in the link between platelets and cancer. PMVs bind to both cancer cells and nontransformed cells in the microenvironment of the tumor, and then transfer platelet-derived contents to the target cell. These contents have the potential to either stimulate or modulate the target cell's response. PMVs are encased in a lipid bilayer that contains surface proteins and lipids as well as components found inside the PMV. Each of these components participates in known and potential PMV roles in cancer. The complicated roles played by PMVs in the onset, development, and progression of cancer and cancer-related comorbidities are summarized in this study.
Collapse
Affiliation(s)
- Yan Pan
- Department of Blood Transfusion, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, 100 Minjiang Road, Quzhou, 324000, Zhejiang, China
| | - Yingjian Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Yanzhong Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Shoufang Xu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Feiyu Jiang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Yetao Han
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Mengsi Hu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Zhiwei Liu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
5
|
Bian X, Yin S, Yang S, Jiang X, Wang J, Zhang M, Zhang L. Roles of platelets in tumor invasion and metastasis: A review. Heliyon 2022; 8:e12072. [PMID: 36506354 PMCID: PMC9730139 DOI: 10.1016/j.heliyon.2022.e12072] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/10/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
The invasion and metastasis of malignant tumors are major causes of death. The most common metastases of cancer are lymphatic metastasis and hematogenous metastasis. Hematogenous metastasis often leads to rapid tumor dissemination. The mechanism of hematogenous metastasis of malignant tumors is very complex. Some experts have found that platelets play an important role in promoting tumor hematogenous metastasis. Platelets may be involved in many processes, such as promoting tumor cell survival, helping tumor cells escape immune surveillance, helping tumors attach to endothelial cells and penetrating capillaries for distant metastasis. However, recent studies have shown that platelets can also inhibit tumor metastasis. At present, the function of platelets in tumor progression has been widely studied, and they not only promote tumor cell metastasis, but also have an inhibitory effect. Therefore, in-depth and summary research of the molecular mechanism of platelets in tumor cell metastasis is of great significance for the screening and treatment of cancer patients. The following is a brief review of the role of platelets in the process of malignant tumor metastasis.
Collapse
Affiliation(s)
- Xiulan Bian
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shengjie Yin
- Department of Oncology, Chifeng City Hospital, Chifeng, Inner Mongolia, China
| | - Shuo Yang
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinju Jiang
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiaqi Wang
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Minghui Zhang
- Department of Oncology, Chifeng City Hospital, Chifeng, Inner Mongolia, China
| | - Lei Zhang
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
6
|
Urbiola-Salvador V, Miroszewska D, Jabłońska A, Qureshi T, Chen Z. Proteomics approaches to characterize the immune responses in cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119266. [PMID: 35390423 DOI: 10.1016/j.bbamcr.2022.119266] [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: 10/14/2021] [Revised: 03/01/2022] [Accepted: 03/28/2022] [Indexed: 06/14/2023]
Abstract
Despite the dynamic development of cancer research, annually millions of people die of cancer. The human immune system is the major 'guard' against tumor development. Unfortunately, cancer cells have the ability to evade the immune system and continue to grow. The proper understanding of the intricate immune response in tumorigenesis remains the holy grail of cancer immunology and designing effective immunotherapy. To decode the immune responses in cancer, in recent years, proteomics studies have received considerable attention. Proteomics studies focus on the detection and quantification of proteins, which are the effectors of biological functions, and as such, are proven to reflect the cell state more accurately, in comparison to genomic or transcriptomic studies. In this review, we discuss the proteomics studies applied to characterize the immune responses in cancer and tumor immune microenvironment heterogeneity. Further, we describe emerging single-cell proteomics approaches that have the potential to be applied in cancer immunity studies.
Collapse
Affiliation(s)
- Víctor Urbiola-Salvador
- Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, University of Gdańsk, Poland.
| | - Dominika Miroszewska
- Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, University of Gdańsk, Poland.
| | - Agnieszka Jabłońska
- Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, University of Gdańsk, Poland.
| | - Talha Qureshi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| | - Zhi Chen
- Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, University of Gdańsk, Poland; Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
7
|
Cantrell R, Palumbo JS. Hemostasis and tumor immunity. Res Pract Thromb Haemost 2022; 6:e12728. [PMID: 35647476 PMCID: PMC9130907 DOI: 10.1002/rth2.12728] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/19/2022] [Accepted: 05/01/2022] [Indexed: 12/13/2022] Open
Abstract
Significant data have accumulated demonstrating a reciprocal relationship between cancer and the hemostatic system whereby cancer promotes life‐threatening hemostatic system dysregulation (e.g., thromboembolism, consumptive coagulopathy), and hemostatic system components directly contribute to cancer pathogenesis. The mechanistic underpinnings of this relationship continue to be defined, but it is becoming increasingly clear that many of these mechanisms involve crosstalk between the hemostatic and immune systems. This is perhaps not surprising given that there is ample evidence for bidirectional crosstalk between the hemostatic and immune systems at multiple levels that likely evolved to coordinate the response to injury, host defense, and tissue repair. Much of the data linking hemostasis and immunity in cancer biology focus on innate immune system components. However, the advent of adaptive immunity‐based cancer therapies such as immune checkpoint inhibitors has revealed that the relationship of hemostasis and immunity in cancer extends to the adaptive immune system. Adaptive immunity‐based cancer therapies appear to be associated with an increased risk of thromboembolic complications, and hemostatic system components appear to regulate adaptive immune functions through diverse mechanisms to affect tumor progression. In this review, the evidence for crosstalk between hemostatic and adaptive immune system components is discussed, and the implications of this relationship in the context of cancer therapy are reviewed. A better understanding of these relationships will likely lead to strategies to make existing adaptive immune based therapies safer by decreasing thromboembolic risk and may also lead to novel targets to improve adaptive immune‐based cancer treatments.
Collapse
Affiliation(s)
- Rachel Cantrell
- Cancer and Blood Diseases Institute Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine Cincinnati Ohio USA
| | - Joseph S. Palumbo
- Cancer and Blood Diseases Institute Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine Cincinnati Ohio USA
| |
Collapse
|
8
|
Chemically Induced Colitis-Associated Cancer Models in Rodents for Pharmacological Modulation: A Systematic Review. J Clin Med 2022; 11:jcm11102739. [PMID: 35628865 PMCID: PMC9146029 DOI: 10.3390/jcm11102739] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 02/01/2023] Open
Abstract
Animal models for colitis-associated colorectal cancer (CACC) represent an important tool to explore the mechanistic basis of cancer-related inflammation, providing important evidence that several inflammatory mediators play specific roles in the initiation and perpetuation of colitis and CACC. Although several original articles have been published describing the CACC model in rodents, there is no consensus about the induction method. This review aims to identify, summarize, compare, and discuss the chemical methods for the induction of CACC through the PRISMA methodology. METHODS We searched MEDLINE via the Pubmed platform for studies published through March 2021, using a highly sensitive search expression. The inclusion criteria were only original articles, articles where a chemically-induced animal model of CACC is described, preclinical studies in vivo with rodents, and articles published in English. RESULTS Chemically inducible models typically begin with the administration of a carcinogenic compound (as azoxymethane (AOM) or 1,2-dimethylhydrazine (DMH)), and inflammation is caused by repeated cycles of colitis-inducing agents (such as 2,4,6-trinitrobenzenesulfonic acid (TNBS) or dextran sulfate sodium (DSS)). The strains mostly used are C57BL/6 and Balb/c with 5-6 weeks. To characterize the preclinical model, the parameters more used include body weight, stool consistency and morbidity, inflammatory biomarkers such as tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β, angiogenesis markers such as proliferating cell nuclear antigen (PCNA), marker of proliferation Ki-67, and caspase 3, the presence of ulcers, thickness or hyperemia in the colon, and histological evaluation of inflammation. CONCLUSION The AOM administration seems to be important to the CACC induction method, since the carcinogenic effect is achieved with just one administration. DSS has been the more used inflammatory agent; however, the TNBS contribution should be more studied, since it allows a reliable, robust, and a highly reproducible animal model of intestinal inflammation.
Collapse
|
9
|
Crosstalk between hemostasis and immunity in cancer pathogenesis. Thromb Res 2022; 213 Suppl 1:S3-S7. [DOI: 10.1016/j.thromres.2021.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 11/21/2022]
|
10
|
Yang WY, Izzi B, Bress AP, Thijs L, Citterio L, Wei FF, Salvi E, Delli Carpini S, Manunta P, Cusi D, Hoylaerts MF, Luttun A, Verhamme P, Hardikar S, Nawrot TS, Staessen JA, Zhang ZY. Association of colorectal cancer with genetic and epigenetic variation in PEAR1-A population-based cohort study. PLoS One 2022; 17:e0266481. [PMID: 35390065 PMCID: PMC8989234 DOI: 10.1371/journal.pone.0266481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 03/22/2022] [Indexed: 11/19/2022] Open
Abstract
Platelet Endothelial Aggregation Receptor 1 (PEAR1) modulates angiogenesis and platelet contact-induced activation, which play a role in the pathogenesis of colorectal cancer. We therefore tested the association of incident colorectal cancer and genetic and epigenetic variability in PEAR1 among 2532 randomly recruited participants enrolled in the family-based Flemish Study on Environment, Genes and Health Outcomes (51.2% women; mean age 44.8 years). All underwent genotyping of rs12566888 located in intron 1 of the PEAR1 gene; in 926 participants, methylation at 16 CpG sites in the PEAR1 promoter was also assessed. Over 18.1 years (median), 49 colorectal cancers occurred, all in different pedigrees. While accounting for clustering of risk factors within families and adjusting for sex, age, body mass index, the total-to-HDL cholesterol ratio, serum creatinine, plasma glucose, smoking and drinking, use of antiplatelet and nonsteroidal anti-inflammatory drug, the hazard ratio of colorectal cancer contrasting minor-allele (T) carriers vs. major-allele (GG) homozygotes was 2.17 (95% confidence interval, 1.18-3.99; P = 0.013). Bootstrapped analyses, from which we randomly excluded from two to nine cancer cases, provided confirmatory results. In participants with methylation data, we applied partial least square discriminant analysis (PLS-DA) and identified two methylation sites associated with higher colorectal cancer risk and two with lower risk. In-silico analysis suggested that methylation of the PEAR1 promoter at these four sites might affect binding of transcription factors p53, PAX5, and E2F-1, thereby modulating gene expression. In conclusion, our findings suggest that genetic and epigenetic variation in PEAR1 modulates the risk of colorectal cancer in white Flemish. To what extent, environmental factors as exemplified by our methylation data, interact with genetic predisposition and modulate penetrance of colorectal cancer risk is unknown.
Collapse
Affiliation(s)
- Wen-Yi Yang
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Benedetta Izzi
- Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, Italy
| | - Adam P. Bress
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah, United States of America
| | - Lutgarde Thijs
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Lorena Citterio
- Division of Nephrology and Dialysis, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fang-Fei Wei
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Erika Salvi
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Simona Delli Carpini
- Division of Nephrology and Dialysis, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Manunta
- School of Nephrology, University Vita-Salute San Raffaele, Milan, Italy
| | | | | | - Aernout Luttun
- Center for Molecular and Vascular Biology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Peter Verhamme
- Center for Molecular and Vascular Biology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Sheetal Hardikar
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah, United States of America
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Tim S. Nawrot
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Jan A. Staessen
- Biomedical Science Group, University of Leuven, Leuven, Belgium
- Research Institute Association for the Promotion of Preventive Medicine, Mechelen, Belgium
| | - Zhen-Yu Zhang
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
11
|
The development of a nomogram and the prognostic prediction value of patients with esophageal squamous cell carcinoma undergoing radical radiotherapy. Future Sci OA 2022; 8:FSO781. [PMID: 35251695 PMCID: PMC8890129 DOI: 10.2144/fsoa-2021-0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 01/06/2022] [Indexed: 12/24/2022] Open
Abstract
Aim: This study aimed to explore the role of the developed nomogram in the prognosis of esophageal squamous cell carcinoma (ESCC). Methods: A total of 181 ESCC patients were randomly divided into a training cohort (n = 141) and a validation cohort (n = 40). Significant factors impacting overall survival (OS) were identified in the training set and integrated into the nomogram based on Cox proportional hazards regression. Results: In the training cohort, the median OS in the high group (≥222) was 49.9 months and the median OS in the low group (<222) was 14.4 months. Multivariate analysis revealed that age, Karnofsky performance status score, tumor stage, chemotherapy, BMI, cervical esophageal carcinoma index and neutrophil to lymphocyte ratio were predictors of OS. Conclusion: The developed nomogram can effectively predict the survival prognosis of ESCC patients.
Collapse
|
12
|
Zhu D, Li Y, Zhang Z, Xue Z, Hua Z, Luo X, Zhao T, Lu C, Liu Y. Recent advances of nanotechnology-based tumor vessel-targeting strategies. J Nanobiotechnology 2021; 19:435. [PMID: 34930293 PMCID: PMC8686559 DOI: 10.1186/s12951-021-01190-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor vessels can provide oxygen and nutrition for solid tumor tissue, create abnormal tumor microenvironment (TME), and play a vital role in the development, immune escape, metastasis and drug resistance of tumor. Tumor vessel-targeting therapy has become an important and promising direction in anti-tumor therapy, with the development of five anti-tumor therapeutic strategies, including vascular disruption, anti-angiogenesis, vascular blockade, vascular normalization and breaking immunosuppressive TME. However, the insufficient drug accumulation and severe side effects of vessel-targeting drugs limit their development in clinical application. Nanotechnology offers an excellent platform with flexible modified surface that can precisely deliver diverse cargoes, optimize efficacy, reduce side effects, and realize the combined therapy. Various nanomedicines (NMs) have been developed to target abnormal tumor vessels and specific TME to achieve more efficient vessel-targeting therapy. The article reviews tumor vascular abnormalities and the resulting abnormal microenvironment, the application of NMs in the tumor vessel-targeting strategies, and how NMs can improve these strategies and achieve multi-strategies combination to maximize anti-tumor effects. ![]()
Collapse
Affiliation(s)
- Dongjie Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhengjia Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zeyu Xue
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhenglai Hua
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xinyi Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ting Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
13
|
Du XZ, Wen B, Liu L, Wei YT, Zhao K. Role of immune escape in different digestive tumours. World J Clin Cases 2021; 9:10438-10450. [PMID: 35004976 PMCID: PMC8686128 DOI: 10.12998/wjcc.v9.i34.10438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/15/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023] Open
Abstract
A counterbalance between immune cells and tumour cells is key to fighting tumours, and immune escape is an important mechanism for the survival of tumour cells in the body. Tumor cells and their cytokines impair the activity of T cells, NK cells, macrophages and other immune cells through various ways, and change the expression of their own surface antigens so as to avoid the clearance of the immune system. Changes in major histocompatibility complex molecules, high expression of programmed death-ligand 1, and the presence of immunosuppressive cells in the tumor microenvironment (TME) are main means by which tumors impair the function of immune cells. During the development of tumours of the digestive system, different mechanisms acting on tumour cells, the TME, and immune cells lead to immune escape and promote tumour progression. In this paper, the mechanisms of immune escape in tumour cells of the digestive system are reviewed to provide a theoretical basis for the immunotherapy of gastrointestinal tumours.
Collapse
Affiliation(s)
- Xin-Zhu Du
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Bin Wen
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Lin Liu
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Ying-Ting Wei
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Kui Zhao
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| |
Collapse
|
14
|
Cariello M, Piccinin E, Zerlotin R, Piglionica M, Peres C, Divella C, Signorile A, Villani G, Ingravallo G, Sabbà C, Moschetta A. Adhesion of Platelets to Colon Cancer Cells Is Necessary to Promote Tumor Development in Xenograft, Genetic and Inflammation Models. Cancers (Basel) 2021; 13:cancers13164243. [PMID: 34439397 PMCID: PMC8394609 DOI: 10.3390/cancers13164243] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Platelets are small, anucleate, metabolically active cells and they represent an important linkage between tissue damage and inflammatory response. Several studies focused on the central role of platelets in inflammation and tumor development through their direct interaction with other cell types. Mice lacking the vascular adhesion molecules P-selectin exhibited a reduction in tumor metastases. We demonstrated that P-selectin-ablated platelets reduced tumor growth in a xenograft adenocarcinoma model. Furthermore, the lack of P-selectin decreased colon cancer progression in genetic mouse models and in chemically-induced colitis colorectal carcinogenesis. Our results suggest that platelets-cancer cells crosstalk via P-selectin is fundamental for tumor development. Abstract Platelets represent the linkage between tissue damage and inflammatory response with a putative role in tumorigenesis. Given the importance of the microenvironment in colon cancer development, we elucidated the eventual role of platelets-cancer cells crosstalk in in vivo colon cancer models. To evaluate the involvement of platelets in intestinal tumorigenesis, we first analyzed if the ablation of β-integrin P-selectin that drives platelets-cell adhesion, would contribute to platelets-colon cancer cell interaction and drive cancer progression. In a xenograft tumor model, we observed that when tumors are inoculated with platelets, the ablation of P-selectin significantly reduced tumor growth compared to control platelets. Furthermore, in genetic models, as well as in chronic colitis-associated colorectal carcinogenesis, P-selectin ablated mice displayed a significant reduction in tumor number and size compared to control mice. Taken together, our data highlights the importance of platelets in the tumor microenvironment for intestinal tumorigenesis. These results support the hypothesis that a strategy aimed to inhibit platelets adhesion to tumor cells are able to block tumor growth and could represent a novel therapeutic approach to colon cancer treatment.
Collapse
Affiliation(s)
- Marica Cariello
- Department of Interdisciplinary Medicine, “Aldo Moro” University of Bari, 70124 Bari, Italy; (M.C.); (E.P.); (R.Z.); (M.P.); (C.S.)
| | - Elena Piccinin
- Department of Interdisciplinary Medicine, “Aldo Moro” University of Bari, 70124 Bari, Italy; (M.C.); (E.P.); (R.Z.); (M.P.); (C.S.)
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, “Aldo Moro” University of Bari, 70124 Bari, Italy; (A.S.); (G.V.)
| | - Roberta Zerlotin
- Department of Interdisciplinary Medicine, “Aldo Moro” University of Bari, 70124 Bari, Italy; (M.C.); (E.P.); (R.Z.); (M.P.); (C.S.)
| | - Marilidia Piglionica
- Department of Interdisciplinary Medicine, “Aldo Moro” University of Bari, 70124 Bari, Italy; (M.C.); (E.P.); (R.Z.); (M.P.); (C.S.)
| | - Claudia Peres
- INBB, National Institute for Biostructures and Biosystems, 00136 Rome, Italy;
| | - Chiara Divella
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, 71024 Bari, Italy;
| | - Anna Signorile
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, “Aldo Moro” University of Bari, 70124 Bari, Italy; (A.S.); (G.V.)
| | - Gaetano Villani
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, “Aldo Moro” University of Bari, 70124 Bari, Italy; (A.S.); (G.V.)
| | - Giuseppe Ingravallo
- Pathology Section, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, 70124 Bari, Italy;
| | - Carlo Sabbà
- Department of Interdisciplinary Medicine, “Aldo Moro” University of Bari, 70124 Bari, Italy; (M.C.); (E.P.); (R.Z.); (M.P.); (C.S.)
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, “Aldo Moro” University of Bari, 70124 Bari, Italy; (M.C.); (E.P.); (R.Z.); (M.P.); (C.S.)
- INBB, National Institute for Biostructures and Biosystems, 00136 Rome, Italy;
- Correspondence: ; Tel.: +39-0805593262; Fax: +39-0805478126
| |
Collapse
|
15
|
Incidence, risk factors and clinical outcome of venous thromboembolism in non-small cell lung cancer patients receiving immune checkpoint inhibition. THROMBOSIS UPDATE 2021. [DOI: 10.1016/j.tru.2021.100056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
16
|
Miao S, Zhang Q, Chang W, Wang J. New Insights Into Platelet-enriched miRNAs: Production, Functions, Roles in Tumors, and Potential Targets for Tumor Diagnosis and Treatment. Mol Cancer Ther 2021; 20:1359-1366. [PMID: 34045229 DOI: 10.1158/1535-7163.mct-21-0050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/12/2021] [Accepted: 05/03/2021] [Indexed: 11/16/2022]
Abstract
In view of the increasing number of malignant tumors worldwide and their high mortality, efforts are being made to find effective biomarkers for early detection and effective treatment measures of cancer. In recent years, the roles of platelets in tumors have attracted considerable attention. Although platelets do not have nuclei, they are rich in miRNAs, which are important molecules in platelet regulation of tumors. Platelet miRNA expression in tumor patients is abnormal and tumor-specific. Platelet miRNAs have higher accuracy and specificity than conventional tumor detection markers and circulating miRNAs in tumor diagnosis. Platelets enriched miRNAs are involved in the regulation of tumor proliferation, metastasis, tumor-related immunity, tumor-related thrombosis, and antitumor therapy. To understand the role of platelet miRNAs in tumors, this article reviews the biological functions of miRNAs in platelets and summarizes the regulatory roles of platelet miRNAs in tumors and the potential roles of platelet miRNAs in tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Shuo Miao
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Qingsong Zhang
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenguang Chang
- Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
17
|
Spinelli C, Tawil N, Adnani L, Rak J, Choi D. Extracellular Vesicle Mediated Vascular Pathology in Glioblastoma. Subcell Biochem 2021; 97:247-273. [PMID: 33779920 DOI: 10.1007/978-3-030-67171-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Glioblastoma (GBM) is an incurable, infiltrative high-grade brain tumour associated with dramatic vascular responses observed both locally (angiogenesis, vascular cooption, angiocrine effects, microthrombosis) and systemically (venous thromboembolism). GBM-associated vascular pathology is diagnostically relevant and constitutes a source of morbidity, mortality and progressive changes in tumour biology. Extracellular vesicles (EVs) have emerged as unique mediators of vascular effects in brain tumours acting as vehicles for intercellular transfer of oncoproteins (e.g. EGFRvIII), RNA, DNA and molecular effectors of angiogenesis and thrombosis. Vascular effects of GBM EVs are regulated by cancer cell genome, epigenome and microenvironment and differ between subtypes of cancer cells and stem cells. Understanding and targeting EV-driven vascular processes in GBM may offer new approaches to diagnose and treat these intractable tumours.
Collapse
Affiliation(s)
- Cristiana Spinelli
- McGill University and the Research Institute of the McGill University Health Centre, QC, Canada
| | - Nadim Tawil
- McGill University and the Research Institute of the McGill University Health Centre, QC, Canada
| | - Lata Adnani
- McGill University and the Research Institute of the McGill University Health Centre, QC, Canada
| | - Janusz Rak
- McGill University and the Research Institute of the McGill University Health Centre, QC, Canada.
| | - Dongsic Choi
- McGill University and the Research Institute of the McGill University Health Centre, QC, Canada.
| |
Collapse
|
18
|
Shi T, Kobara H, Oura K, Masaki T. Mechanisms Underlying Hepatocellular Carcinoma Progression in Patients with Type 2 Diabetes. J Hepatocell Carcinoma 2021; 8:45-55. [PMID: 33604315 PMCID: PMC7886236 DOI: 10.2147/jhc.s274933] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/25/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) ranks third in cancer-related deaths from solid tumors worldwide. The incidence of type 2 diabetes mellitus (T2DM) has increased worldwide in conjunction with the expansion of the Western lifestyle. Furthermore, patients with T2DM have been documented to have an increased risk of HCC, as well as bile tract cancer. Growing evidence shows that T2DM is a strong additive metabolic risk factor for HCC, but how diabetes affects the incidence of HCC requires additional investigation. In this review, we discuss the underlying mechanisms of HCC in patients with T2DM. Topics covered include abnormal glucose and lipid metabolism, hyperinsulinemia, and insulin resistance; the effect of activated platelets; hub gene expression associated with HCC; inflammation and signaling pathways; miRNAs; altered gut microbiota and immunomodulation. The evidence suggests that reducing obesity, diabetes, and nonalcoholic fatty liver disease/nonalcoholic steatohepatitis through efficient measures of prevention may lead to decreased rates of T2DM-related HCC.
Collapse
Affiliation(s)
- Tingting Shi
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa, 761-0793, Japan
| | - Hideki Kobara
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa, 761-0793, Japan
| | - Kyoko Oura
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa, 761-0793, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa, 761-0793, Japan
| |
Collapse
|
19
|
Cantrell R, Palumbo JS. The thrombin–inflammation axis in cancer progression. Thromb Res 2020; 191 Suppl 1:S117-S122. [DOI: 10.1016/s0049-3848(20)30408-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/18/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023]
|
20
|
Oury C, Côté N, Clavel MA. Biomarkers Associated with Aortic Stenosis and Structural Bioprosthesis Dysfunction. Cardiol Clin 2019; 38:47-54. [PMID: 31753176 DOI: 10.1016/j.ccl.2019.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Prediction of patients at risk of aortic valve stenosis (AS), AS progression rate, and aortic bioprosthesis dysfunction are of major importance for clinical management and/or prevention. Many imaging modalities may be used; however, they may not be conclusive or available for all patients. Circulating biomarkers are easily available and may be related to a disease or process such as aortic valve calcification or associated with a risk factor of the disease. This article reviews current blood biomarkers associated with aortic valve stenosis/calcification and bioprosthesis dysfunction.
Collapse
Affiliation(s)
- Cécile Oury
- Laboratory of Cardiology, Department of Cardiology, GIGA-Cardiovascular Sciences, University of Liège Hospital, University of Liège, CHU du Sart Tilman, Domaine Universitaire du Sart Tilman, Batiment B35, Liège 4000, Belgium.
| | - Nancy Côté
- Institut universitaire de cardiologie et de Pneumologie de Québec, 2725, Chemin Sainte-Foy, A-2047, Québec, Québec G1V 4G5, Canada
| | - Marie-Annick Clavel
- Institut universitaire de cardiologie et de Pneumologie de Québec, 2725, Chemin Sainte-Foy, A-2047, Québec, Québec G1V 4G5, Canada
| |
Collapse
|
21
|
Jovani M, Chan AT. Do Aspirin and Clopidogrel Follow the Same Road Toward Prevention of Colorectal Cancer? Clin Gastroenterol Hepatol 2019; 17:1945-1947. [PMID: 30807844 DOI: 10.1016/j.cgh.2019.02.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/20/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Manol Jovani
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
22
|
Panigrahy D, Gartung A, Yang J, Yang H, Gilligan MM, Sulciner ML, Bhasin SS, Bielenberg DR, Chang J, Schmidt BA, Piwowarski J, Fishbein A, Soler-Ferran D, Sparks MA, Staffa SJ, Sukhatme V, Hammock BD, Kieran MW, Huang S, Bhasin M, Serhan CN, Sukhatme VP. Preoperative stimulation of resolution and inflammation blockade eradicates micrometastases. J Clin Invest 2019; 129:2964-2979. [PMID: 31205032 DOI: 10.1172/jci127282] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022] Open
Abstract
Cancer therapy is a double-edged sword, as surgery and chemotherapy can induce an inflammatory/immunosuppressive injury response that promotes dormancy escape and tumor recurrence. We hypothesized that these events could be altered by early blockade of the inflammatory cascade and/or by accelerating the resolution of inflammation. Preoperative, but not postoperative, administration of the nonsteroidal antiinflammatory drug ketorolac and/or resolvins, a family of specialized proresolving autacoid mediators, eliminated micrometastases in multiple tumor-resection models, resulting in long-term survival. Ketorolac unleashed anticancer T cell immunity that was augmented by immune checkpoint blockade, negated by adjuvant chemotherapy, and dependent on inhibition of the COX-1/thromboxane A2 (TXA2) pathway. Preoperative stimulation of inflammation resolution via resolvins (RvD2, RvD3, and RvD4) inhibited metastases and induced T cell responses. Ketorolac and resolvins exhibited synergistic antitumor activity and prevented surgery- or chemotherapy-induced dormancy escape. Thus, simultaneously blocking the ensuing proinflammatory response and activating endogenous resolution programs before surgery may eliminate micrometastases and reduce tumor recurrence.
Collapse
Affiliation(s)
- Dipak Panigrahy
- Center for Vascular Biology Research.,Department of Pathology, and.,Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Allison Gartung
- Center for Vascular Biology Research.,Department of Pathology, and.,Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jun Yang
- Department of Entomology and Nematology, and UC Davis Comprehensive Cancer Center, University of California, Davis, California, USA
| | - Haixia Yang
- Center for Vascular Biology Research.,Department of Pathology, and.,Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Molly M Gilligan
- Center for Vascular Biology Research.,Department of Pathology, and.,Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Megan L Sulciner
- Center for Vascular Biology Research.,Department of Pathology, and.,Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Swati S Bhasin
- Division of Interdisciplinary Medicine and Biotechnology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Jaimie Chang
- Center for Vascular Biology Research.,Department of Pathology, and.,Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Birgitta A Schmidt
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julia Piwowarski
- Center for Vascular Biology Research.,Department of Pathology, and.,Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Anna Fishbein
- Center for Vascular Biology Research.,Department of Pathology, and.,Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Dulce Soler-Ferran
- Center for Vascular Biology Research.,Department of Pathology, and.,Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew A Sparks
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, USA
| | - Steven J Staffa
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Bruce D Hammock
- Department of Entomology and Nematology, and UC Davis Comprehensive Cancer Center, University of California, Davis, California, USA
| | - Mark W Kieran
- Division of Pediatric Oncology, Dana-Farber Cancer Institute, and.,Department of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sui Huang
- Institute for Systems Biology, Seattle, Washington, USA
| | - Manoj Bhasin
- Division of Interdisciplinary Medicine and Biotechnology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Vikas P Sukhatme
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Division of Interdisciplinary Medicine and Biotechnology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Medicine and Center for Affordable Medical Innovation, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
23
|
Perfluorocarbon nanoparticle-mediated platelet inhibition promotes intratumoral infiltration of T cells and boosts immunotherapy. Proc Natl Acad Sci U S A 2019; 116:11972-11977. [PMID: 31142648 DOI: 10.1073/pnas.1901987116] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cancer immunotherapy can stimulate and enhance the ability of the immune system to recognize, arrest, and eliminate tumor cells. Immune checkpoint therapies (e.g., PD-1/PD-L1) have shown an unprecedented and durable clinical response rate in patients among various cancer types. However, a large fraction of patients still does not respond to these checkpoint inhibitors. The main cause of this phenomenon is the limited T-cell infiltration in tumors. Therefore, additional strategies to enhance T-cell trafficking into tumors are urgently needed to improve patients' immune responses. In this study, we screened an array of perfluorocarbon compounds, reporting that albumin-based perfluorotributylamine nanoparticles (PFTBA@Alb) can effectively increase the permeability of tumor blood vessels, and no distinct side effects were found on normal blood vessels. After i.v. administration of PFTBA@Alb, the number of tumor-infiltrating CD8+ and CD4+ T cells showed an obvious rising trend. More important, a striking tumor inhibition rate, reaching nearly 90%, was observed when combining PFTBA@Alb with anti-PD-L1 antibody. These findings suggest that PFTBA@Alb can be regarded as an enhancer for anti-PD-L1 immunotherapy.
Collapse
|
24
|
Abstract
Several pieces of evidence support the role of activated platelets in the development of the chronic inflammation-related diseases, such as atherothrombosis and cancer, mainly via the release of soluble factors and microparticles (MPs). Platelets and MPs contain a repertoire of proteins and genetic material (i.e., mRNAs and microRNAs) which may be influenced by the clinical condition of the individuals. In fact, platelets are capable of up-taking proteins and genetic material during their lifespan. Moreover, the content of platelet-derived MPs can be delivered to other cells, including stromal, immune, epithelial, and cancer cells, to change their phenotype and functions, thus contributing to cancer promotion and its metastasization. Platelets and MPs can play an indirect role in the metastatic process by helping malignant cells to escape from immunological surveillance. Furthermore, platelets and their derived MPs represent a potential source for blood biomarker development in oncology. This review provides an updated overview of the roles played by platelets and MPs in cancer and metastasis formation. The possible analysis of platelet and MP molecular signatures for the detection of cancer and monitoring of anticancer treatments is discussed. Finally, the potential use of MPs as vectors for drug delivery systems to cancer cells is put forward.
Collapse
|
25
|
Luu S, Gardiner EE, Andrews RK. Bone Marrow Defects and Platelet Function: A Focus on MDS and CLL. Cancers (Basel) 2018; 10:E147. [PMID: 29783667 PMCID: PMC5977120 DOI: 10.3390/cancers10050147] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/11/2018] [Accepted: 05/16/2018] [Indexed: 12/16/2022] Open
Abstract
The bloodstream typically contains >500 billion anucleate circulating platelets, derived from megakaryocytes in the bone marrow. This review will focus on two interesting aspects of bone marrow dysfunction and how this impacts on the quality of circulating platelets. In this regard, although megakaryocytes are from the myeloid lineage leading to granulocytes (including neutrophils), erythrocytes, and megakaryocytes/platelets, recent evidence has shown that defects in the lymphoid lineage leading to B cells, T cells, and natural killer (NK) cells also result in abnormal circulating platelets. Current evidence is limited regarding whether this latter phenomenon might potentially arise from (a) some form of as-yet-undetected defect common to both lineages; (b) adverse interactions occurring between cells of different lineages within the bone marrow environment; and/or (c) unknown disease-related factor(s) affecting circulating platelet receptor expression/function after their release from megakaryocytes. Understanding the mechanisms underlying how both myeloid and lymphoid lineage bone marrow defects lead to dysfunction of circulating platelets is significant because of the potential diagnostic and predictive value of peripheral platelet analysis for bone marrow disease progression, the additional potential effects of new anti-cancer drugs on platelet function, and the critical role platelets play in regulation of bleeding risk, inflammation, and innate immunity.
Collapse
Affiliation(s)
- Sarah Luu
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3004, Australia.
| | - Elizabeth E Gardiner
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600, Australia.
| | - Robert K Andrews
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|
26
|
Flick MJ, Palumbo JS. Platelets couple inflammation to tumorigenesis, a bridge too far. J Thromb Haemost 2018; 16:759-761. [PMID: 29418061 DOI: 10.1111/jth.13967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Indexed: 12/16/2022]
Affiliation(s)
- M J Flick
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - J S Palumbo
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|