1
|
An Y, Ji C, Zhang H, Jiang Q, Maitz MF, Pan J, Luo R, Wang Y. Engineered Cell Membrane Coating Technologies for Biomedical Applications: From Nanoscale to Macroscale. ACS NANO 2025; 19:11517-11546. [PMID: 40126356 DOI: 10.1021/acsnano.4c16280] [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: 03/25/2025]
Abstract
Cell membrane coating has emerged as a promising strategy for the surface modification of biomaterials with biological membranes, serving as a cloak that can carry more functions. The cloaked biomaterials inherit diverse intrinsic biofunctions derived from different cell sources, including enhanced biocompatibility, immunity evasion, specific targeting capacity, and immune regulation of the regenerative microenvironment. The intrinsic characteristics of biomimicry and biointerfacing have demonstrated the versatility of cell membrane coating technology on a variety of biomaterials, thus, furthering the research into a wide range of biomedical applications and clinical translation. Here, the preparation of cell membrane coatings is emphasized, and different sizes of coated biomaterials from nanoscale to macroscale as well as the engineering strategies to introduce additional biofunctions are summarized. Subsequently, the utilization of biomimetic membrane-cloaked biomaterials in biomedical applications is discussed, including drug delivery, imaging and phototherapy, cancer immunotherapy, anti-infection and detoxification, and implant modification. In conclusion, the latest advancements in clinical and preclinical studies, along with the multiple benefits of cell membrane-coated nanoparticles (NPs) in biomimetic systems, are elucidated.
Collapse
Affiliation(s)
- Yongqi An
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cheng Ji
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Hao Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Qing Jiang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Manfred F Maitz
- Max Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research Dresden, Dresden 01069, Germany
| | - Junqiang Pan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- Department of Cardiovascular Medicine, Xi'an Central Hospital, Xi'an 710003, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- Research Unit. of Minimally Invasive Treatment of Structural Heart-Disease, Chinese Academy of Medical Sciences (2021RU013), Chengdu, 610065, China
| |
Collapse
|
2
|
Lee CH, Lin CL, Yen TH, Hsieh SY. Daily Antiplatelets Other Than Aspirin Reduce Liver Cancer Risk but Increase Intracranial Hemorrhage Risk in Cirrhotic Patients. Int J Gen Med 2024; 17:4755-4767. [PMID: 39435412 PMCID: PMC11492919 DOI: 10.2147/ijgm.s472260] [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: 05/30/2024] [Accepted: 10/05/2024] [Indexed: 10/23/2024] Open
Abstract
Purpose Aspirin, known to reduce the risk of liver cancer, has been proposed as a preventive measure for patients with chronic hepatitis and cirrhosis. However, concerns regarding aspirin's potential to cause gastrointestinal (GI) mucosal injury and bleeding have emerged. Several antiplatelets other than aspirin (APOA) that pose a smaller risk of GI bleeding than aspirin have been proposed as potential aspirin substitutes. This study investigated whether APOAs were effective at reducing the risk of hepatocellular carcinoma (HCC). Additionally, we evaluated the safety of APOAs, specifically regarding their potential to increase the risk of GI bleeding, in a nationwide cirrhosis cohort. Patients and Methods For the period January 1, 2000, to December 31, 2017, we identified 686 993 patients with cirrhosis from a national database. A control group was established using 1:2 propensity score matching on the basis of sex, age, comorbidities, and medication use. Results Daily use of APOAs was significantly associated with lower incidences of HCC (aHR 0.67; 95% CI, 0.60-0.73; P < 0.001) and showed no significant increase in GI bleeding risk (aHR 1.04; 95% CI, 0.93-1.15; P = 0.533) compared to nonuse of APOAs. However, the risks of intracranial hemorrhage (aHR, 1.41; 95% CI, 1.18 to 1.69; P < 0.001) and overall mortality (aHR, 2.03; 95% CI, 1.95 to 2.10; P < 0.001) were higher in the APOA user group. Conclusion Our results suggest that although daily use of APOAs other than aspirin may decrease the HCC risk of patients with cirrhosis, it may also increase their risks of intracranial hemorrhage and overall mortality. Therefore, the use of APOAs as an alternative to aspirin for HCC prevention in patients with cirrhosis requires careful consideration.
Collapse
Affiliation(s)
- Chern-Horng Lee
- Department of Geriatric Medicine and General Internal Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Cheng-Li Lin
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Tzung-Hai Yen
- Department of Nephrology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Sen-Yung Hsieh
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
3
|
Luo Z, Jiao B, Huang T, Zhao H, He W, Bo Y, Ding Z, Zhang G. Development and external validation of a novel nomogram to predict intravesical recurrence after radical nephroureterectomy: a multicenter study. J Cancer Res Clin Oncol 2023; 149:11223-11231. [PMID: 37355502 DOI: 10.1007/s00432-023-05016-2] [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: 05/23/2023] [Accepted: 06/19/2023] [Indexed: 06/26/2023]
Abstract
OBJECTIVE This study aimed to establish and validate nomograms to predict the probability of intravesical recurrence (IVR) after radical nephroureterectomy (RNU) for upper urinary tract epithelial carcinoma (UTUC). METHODS Clinical data of 528 patients with UTUC after RNU were collected from two medical centers between 2009 and 2020. We used the least absolute shrinkage and selection operator (LASSO) regression to select variables for multivariable Cox regression analysis in the training cohort and included independent risk factors into nomogram models predicting IVR-free survival (IVRFS). Another center was applied as the external cohort to validate the predictive accuracy and discriminative ability of the nomogram by performing area under the receiver operating curve (AUC), consistency index (C-index), and calibration curve. RESULTS History of bladder cancer, tumor size, preoperative urine cytology, postoperative instillation, Ki-67, and platelet-to-lymphocyte ratio (PLR) were identified as independent risk factors for IVR. The prognosis model including these predictors demonstrated excellent discriminatory performance in both the training cohort (C-index, 0.814) and external validation cohort (C-index, 0.748). The calibration plots of the nomogram revealed good consistency in both cohorts. Finally, patients could be classified into two risk groups based on scores obtained from the nomogram, with significant differences in IVRFS. CONCLUSION Our study provided a reliable nomogram for predicting the probability of IVR in patients with UTUC after RNU. Risk stratification based on this model may assist urologists make optimal clinical decisions on the management of UTUC.
Collapse
Affiliation(s)
- Zhenkai Luo
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, No. 9 Dongdan Santiao, Dongcheng District, Beijing, 100730, China
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Binbin Jiao
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China
| | - Tao Huang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, No. 300. Guangzhou Road, Nanjing, 210029, China
| | - Hang Zhao
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, 270 Dong an Road, Xuhui District, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong an Road, Xuhui District, Shanghai, 200032, China
| | - Weifeng He
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China
| | - Yuxuan Bo
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China
| | - Zhenshan Ding
- Department of Urology, China-Japan Friendship Hospital, Yinghuadong Road, Chaoyang District, Beijing, 100029, China.
| | - Guan Zhang
- Department of Urology, China-Japan Friendship Hospital, Yinghuadong Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
4
|
Liu G, Zhang B. Age-specific cardiovascular disease-related mortality among patients with major gastrointestinal cancers: A SEER population-based study. Cancer Med 2023; 12:17253-17265. [PMID: 37387603 PMCID: PMC10501270 DOI: 10.1002/cam4.6305] [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: 01/30/2023] [Revised: 05/29/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Studies have reported age as a risk factor for cardiovascular disease (CVD)-related mortality; however, only a few studies have focused on the relationship between age and CVD-related mortality, especially among major gastrointestinal cancers. METHOD The present retrospective cohort enrolled patients with colorectal, pancreatic, hepatocellular, gastric, and esophageal cancer between 2000 to 2015 from the Surveillance, Epidemiology and End Results Registry (SEER). Standardized mortality ratio (SMR), competing risk regression, and restricted cubic spline (RCS) analyses were used in our study. RESULTS We analyzed 576,713 patients with major gastrointestinal cancers (327,800 patients with colorectal cancer, 93,310 with pancreatic cancer, 69,757 with hepatocellular cancer, 52,024 with gastric cancer, and 33,822 with esophageal cancer). Overall, CVD-related mortality gradually decreased every year, and the majority were older patients. All cancer patients had a higher CVD-related mortality rate than the general U.S. POPULATION The adjusted sub-hazard ratios for middle-aged with colorectal cancer, pancreatic cancer, hepatocellular cancer, gastric cancer, and esophageal cancer were 2.55 (95% CI: 2.15-3.03), 1.77 (95% CI: 1.06-2.97), 2.64 (95% CI: 1.60-4.36), 2.15 (95% CI: 1.32-3.51), and 2.28 (95% CI: 1.17-4.44), respectively. The adjusted sub-hazard ratios for older patients with colorectal cancer, pancreatic cancer, hepatocellular cancer, gastric cancer, and esophageal cancer were 11.23 (95% CI: 9.50-13.27), 4.05 (95% CI: 2.46-6.66), 4.47 (95% CI: 2.72-7.35), 7.16 (95% CI: 4.49-11.41), and 4.40 (95% CI: 2.28-8.48), respectively. A non-linear relationship between age at diagnosis and CVD-related mortality was found in colorectal cancer, pancreatic cancer, and esophageal cancer; their reference ages were 67, 69, and 66 years old, respectively. CONCLUSION This study demonstrated that age was a risk factor for CVD-related mortality among major gastrointestinal cancers.
Collapse
Affiliation(s)
- Gen Liu
- Department of CardiologyRenmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of CardiologyWuhanChina
| | - Bo‐fang Zhang
- Department of CardiologyRenmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of CardiologyWuhanChina
| |
Collapse
|
5
|
Huang S, Zhang NQ, Xu CJ, Huang WQ, Li DX, Li J, Yao LL, Sundquist K, Sundquist J, Jiang SH, Xing X, Hu LP, Zhang ZG, Ji J, Zhang XL. Dipyridamole enhances the anti-cancer ability of aspirin against colorectal cancer by inducing apoptosis in an unfolded protein response-dependent manner. Cell Oncol (Dordr) 2023; 46:953-967. [PMID: 36939950 DOI: 10.1007/s13402-023-00789-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/21/2023] Open
Abstract
PURPOSE Available evidence indicates that dipyridamole enhances the anti-thrombotic effects of aspirin for the prevention of secondary strokes. Aspirin is a well-known non-steroid anti-inflammatory drug. This anti-inflammatory property has turned aspirin into a potential drug for inflammation-related cancers such as colorectal cancer (CRC). Here, we aimed to explore whether the anti-cancer effect of aspirin against CRC could be improved by combined administration with dipyridamole. METHODS Population-based clinical data analysis was conducted to assess a possible therapeutic effect of combined dipyridamole and aspirin treatment in inhibiting CRC compared with either monotherapy. This therapeutic effect was further verified in different CRC mouse models, i.e. an orthotopic xenograft mouse model, an AOM/DSS mouse model, an Apcmin/+ mouse model and a patient derived xenograft (PDX) mouse model. The in vitro effects of the drugs on CRC cells were tested using CCK8 and flow cytometry assays. RNA-Seq, Western blotting, qRT-PCR and flow cytometry were used to identify the underlying molecular mechanisms. RESULTS We found that dipyridamole combined with aspirin had a better inhibitory effect on CRC than either monotherapy alone. The enhanced anti-cancer effect of the combined use of dipyridamole with aspirin was found to rely on the induction of an overwhelmed endoplasmic reticulum (ER) stress and subsequent pro-apoptotic unfolded protein response (UPR), which was different from the anti-platelet effect. CONCLUSIONS Our data indicate that the anti-cancer effect of aspirin against CRC may be enhanced by combined administration with dipyridamole. In case further clinical studies confirm our findings, these may be repurposed as adjuvant agents.
Collapse
Affiliation(s)
- Shan Huang
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, P.R. China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Nai-Qi Zhang
- Center for Primary Health Care Research, Lund University/Region Skåne, Lund, Sweden
| | - Chun-Jie Xu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Wu-Qing Huang
- School of Public Health, Fujian Medical University, Fuzhou City, P.R. China
| | - Dong-Xue Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Lin-Li Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Lund, Sweden
- Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Lund, Sweden
- Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Xin Xing
- Shanghai Fengxian District Central Hospital, No. 6600, Nanfeng Road, Shanghai, 201499, China
| | - Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China.
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China.
| | - Jianguang Ji
- Center for Primary Health Care Research, Lund University/Region Skåne, Lund, Sweden.
| | - Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China.
| |
Collapse
|
6
|
Shi Q, Ji T, Tang X, Guo W. The role of tumor-platelet interplay and micro tumor thrombi during hematogenous tumor metastasis. Cell Oncol (Dordr) 2023; 46:521-532. [PMID: 36652166 DOI: 10.1007/s13402-023-00773-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND In addition to their pivotal roles in coagulation and thrombosis, platelets are crucial in tumor progression, with plenty of clinical and experimental data demonstrating that the interplay of platelets and tumor cells is essential for hematogenous tumor metastasis. After detach from primary sites, tumor cells intravasate into the blood circulation becoming circulating tumor cells and induce platelet activation, aggregation and encasement around tumor cells to form micro tumor thrombi, which create a permissive tumor microenvironment for metastasis. Platelets in micro tumor thrombi protect tumor cells from immune surveillance and anoikis (detachment-triggered apoptosis) through various pathways, which are significant for tumor cell survival in the bloodstream. Moreover, platelets can facilitate tumor metastasis by expediting epithelial-mesenchymal transition (EMT), adhesion to the endothelium, angiogenesis, tumor proliferation processes and platelet-derived microvesicle (PMV) formation. CONCLUSIONS Here, we provide a synopsis of the current understanding of the formation of micro tumor thrombi and the role of micro tumor thrombi in tumor hematogenous metastasis based on the tumor-platelet interplay. We also highlight potential therapeutic strategies targeting platelets for tumor treatment, including cancer-associated platelet-targeted nanomedicines.
Collapse
Affiliation(s)
- Qianyu Shi
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, 100044, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Tao Ji
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, 100044, Beijing, China.
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China.
| | - Xiaodong Tang
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, 100044, Beijing, China
| | - Wei Guo
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, 100044, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| |
Collapse
|
7
|
Gao J, Zhou C, Zhong Y, Shi L, Luo X, Su H, Li M, Xu Y, Zhang N, Zhou H. Dipyridamole interacts with the N-terminal domain of HSP90 and antagonizes the function of the chaperone in multiple cancer cell lines. Biochem Pharmacol 2023; 207:115376. [PMID: 36513142 DOI: 10.1016/j.bcp.2022.115376] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/06/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Molecular chaperone HSP90 has been considered as a promising target for anti-cancer drug development for years. However, due to the heat shock response induced by the ATP competitive inhibitors against HSP90, the therapeutic efficacies of the compounds are compromised, which consequently restricts the clinical use of HSP90-targeted inhibitors. Therefore, there is a need to discover novel HSP90-targeted modulators which exhibit acceptable inhibition activity against the chaperone and do not induce significant heat shock response in the meantime. Here in this study, we firstly developed a tip-based affinity selection-mass spectrometry platform with optimized experimental conditions/parameters for HSP90-targeted active compound screening, and then applied it to fish out inhibitors against HSP90 from a collection of 2,395 compounds composed of FDA-approved drugs and drug candidates. Dipyridamole, which acts as an anti-thrombotic agent by modulating multiple targets and has a long history of safe use, was identified to interact with HSP90's N-terminal domain. The following conducted biophysical and biochemical experiments demonstrated that Dipyridamole could bind to HSP90's ATP binding pocket and function as an ATP competitive inhibitor of the chaperone. Finally, cellular-based assays including CESTA, cell viability assessment and proteomic analysis etc. were performed to evaluate whether the interaction between HSP90 and Dipyridamole contributes to the anti-tumor effects of the compound. We then found that Dipyridamole inhibits the growth and proliferation of human cancer cells by downregulating cell cycle regulators and upregulating apoptotic cell signaling, which are potentially mediated by the binding of Dipyridamole to HSP90 and to PDEs (phosphodiesterases), respectively.
Collapse
Affiliation(s)
- Jing Gao
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Chen Zhou
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Yan Zhong
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Li Shi
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Xuanyang Luo
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Haixia Su
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Minjun Li
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yechun Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Naixia Zhang
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China.
| | - Hu Zhou
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China.
| |
Collapse
|
8
|
van den Kerkhof DL, Nagy M, Wichapong K, Brouns SL, Suylen DP, Hackeng TM, Dijkgraaf I. Unraveling the role of the homoarginine residue in antiplatelet drug eptifibatide in binding to the αIIbβ3 integrin receptor. Thromb Res 2022; 217:96-103. [DOI: 10.1016/j.thromres.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/19/2022] [Accepted: 07/21/2022] [Indexed: 10/16/2022]
|
9
|
Secretory SERPINE1 Expression Is Increased by Antiplatelet Therapy, Inducing MMP1 Expression and Increasing Colon Cancer Metastasis. Int J Mol Sci 2022; 23:ijms23179596. [PMID: 36076991 PMCID: PMC9455756 DOI: 10.3390/ijms23179596] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/21/2022] Open
Abstract
Contrary to many reports that antiplatelet agents inhibit cancer growth and metastasis, new solid tumors have been reported in patients receiving long-term antiplatelet therapy. We investigated the effects of these agents directly on cancer cells in the absence of platelets to mimic the effects of long-term therapy. When four antiplatelet agents (aspirin, clopidogrel, prasugrel, and ticagrelor) were administered to colon cancer cells, cancer cell proliferation was inhibited similarly to a previous study. However, surprisingly, when cells were treated with a purinergic P2Y12 inhibitor (purinergic antiplatelet agent), the motility of the cancer cells was significantly increased. Therefore, gene expression profiles were identified to investigate the effect of P2Y12 inhibitors on cell mobility, and Serpin family 1 (SERPINE1) was identified as a common gene associated with cell migration and cell death in three groups. Antiplatelet treatment increased the level of SERPINE1 in cancer cells and also promoted the secretion of SERPINE1 into the medium. Increased SERPINE1 was found to induce MMP1 and, thus, increase cell motility. In addition, an increase in SERPINE1 was confirmed using the serum of patients who received these antiplatelet drugs. With these results, we propose that SERPINE1 could be used as a new target gene to prevent the onset and metastasis of cancer in patients with long-term antiplatelet therapy.
Collapse
|
10
|
Huang WJ, Wang GY, Liu ZY, Zhang ML, Wang W, Zhang X, Wang RT. Preoperative PDW levels predict pulmonary metastasis in patients with hepatocellular carcinoma. BMC Cancer 2022; 22:683. [PMID: 35729523 PMCID: PMC9215007 DOI: 10.1186/s12885-022-09754-3] [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: 05/17/2021] [Accepted: 06/10/2022] [Indexed: 12/03/2022] Open
Abstract
Background In hepatocellular carcinoma (HCC), pulmonary metastasis (PM) after hepatectomy is associated with poor clinical outcomes. The crucial phases of tumour cell proliferation, angiogenesis, and metastasis all entail platelet activation. In HCC, platelet distribution width (PDW) suggests platelet size changes and predicts a worse prognosis. The aim of this study was to assess the association between PDW and PMs in HCC patients receiving hepatectomy. Material/methods From January 2013 to December 2015, a cohort of patients who underwent hepatectomy for HCC at the Harbin Medical University Cancer Hospital in China were retrospectively evaluated. The relationship between PDW levels and clinical and demographic parameters was examined. To investigate the relationships between predicted factors and PM, a competing risk model was used. From January 2016 to December 2018, a validation cohort of 109 patients from the First Affiliated Hospital of Harbin Medical University was studied independently. Results In the primary cohort, 19 out of 214 patients had postoperative PMs. In HCC patients with PM, PDW levels were lower than in those without PM. There was a significant difference in the cumulative incidence of 2-year PM between the high-PDW and low-PDW groups after controlling for competing risk events (death prior to the development of PM) (p < 0.001). In addition, PDW was also found to be an independent predictor for PM in a multivariable competing risk analysis. The results were externally validated in another cohort. Conclusions In HCC, preoperative PDW is significantly associated with PM. PDW could be a biomarker for post-operative PM in HCC patients.
Collapse
Affiliation(s)
- Wen-Juan Huang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Guang-Yu Wang
- Department of Digestive Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Zeng-Yao Liu
- Department of Interventional Medicine, The First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - Meng-Lin Zhang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Wen Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Xin Zhang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China.
| | - Rui-Tao Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China.
| |
Collapse
|
11
|
Li QR, Xu HZ, Xiao RC, Liu Y, Tang JM, Li J, Yu TT, Liu B, Li LG, Wang MF, Han N, Xu YH, Wang C, Komatsu N, Zhao L, Peng XC, Li TF, Chen X. Platelets are highly efficient and efficacious carriers for tumor-targeted nano-drug delivery. Drug Deliv 2022; 29:937-949. [PMID: 35319321 PMCID: PMC8956315 DOI: 10.1080/10717544.2022.2053762] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The present work aims to prove the concept of tumor-targeted drug delivery mediated by platelets. Doxorubicin (DOX) attached to nanodiamonds (ND-DOX) was investigated as the model payload drug of platelets. In vitro experiments first showed that ND-DOX could be loaded in mouse platelets in a dose-dependent manner with a markedly higher efficiency and capacity than free DOX. ND-DOX-loaded platelets (Plt@ND-DOX) maintained viability and ND-DOX could be stably held in the platelets for at least 4 hr. Next, mouse Lewis lung cancer cells were found to activate Plt@ND-DOX and thereby stimulate cargo unloading of Plt@ND-DOX. The unloaded ND-DOX was taken up by co-cultured cancer cells which consequently exhibited loss of viability, proliferation suppression and apoptosis. In vivo, Plt@ND-DOX displayed significantly prolonged blood circulation time over ND-DOX and DOX in mice, and Lewis tumor grafts demonstrated infiltration, activation and cargo unloading of Plt@ND-DOX in the tumor tissue. Consequently, Plt@ND-DOX effectively reversed the growth of Lewis tumor grafts which exhibited significant inhibition of cell proliferation and apoptosis. Importantly, Plt@ND-DOX displayed a markedly higher therapeutic potency than free DOX but without the severe systemic toxicity associated with DOX. Our findings are concrete proof of platelets as efficient and efficacious carriers for tumor-targeted nano-drug delivery with the following features: 1) large loading capacity and high loading efficiency, 2) good tolerance of cargo drug, 3) stable cargo retention and no cargo unloading in the absence of stimulation, 4) prolonged blood circulation time, and 5) excellent tumor distribution and tumor-activated drug unloading leading to high therapeutic potency and few adverse effects. Platelets hold great potential as efficient and efficacious carriers for tumor-targeted nano-drug delivery.
Collapse
Affiliation(s)
- Qi-Rui Li
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Hua-Zhen Xu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Rong-Cheng Xiao
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Yan Liu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Jun-Ming Tang
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Jian Li
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Ting-Ting Yu
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Bin Liu
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Liu-Gen Li
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Mei-Fang Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Ning Han
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Yong-Hong Xu
- Institute of Ophthalmological Research, Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chao Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Naoki Komatsu
- Graduate School of Human and Environmental Studies, Kyoto University, Kyoto, Japan
| | - Li Zhao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Xing-Chun Peng
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Tong-Fei Li
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Xiao Chen
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| |
Collapse
|
12
|
Fernández DI, Provenzale I, Cheung HY, van Groningen J, Tullemans BM, Veninga A, Dunster JL, Honarnejad S, van den Hurk H, Kuijpers MJ, Heemskerk JW. Ultra-high-throughput Ca 2+ assay in platelets to distinguish ITAM-linked and G-protein-coupled receptor activation. iScience 2022; 25:103718. [PMID: 35072010 PMCID: PMC8762394 DOI: 10.1016/j.isci.2021.103718] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/12/2021] [Accepted: 12/29/2021] [Indexed: 12/30/2022] Open
Abstract
Antiplatelet drugs targeting G-protein-coupled receptors (GPCRs), used for the secondary prevention of arterial thrombosis, coincide with an increased bleeding risk. Targeting ITAM-linked receptors, such as the collagen receptor glycoprotein VI (GPVI), is expected to provide a better antithrombotic-hemostatic profile. Here, we developed and characterized an ultra-high-throughput (UHT) method based on intracellular [Ca2+]i increases to differentiate GPVI and GPCR effects on platelets. In 96-, 384-, or 1,536-well formats, Calcium-6-loaded human platelets displayed a slow-prolonged or fast-transient [Ca2+]i increase when stimulated with the GPVI agonist collagen-related peptide or with thrombin and other GPCR agonists, respectively. Semi-automated curve fitting revealed five parameters describing the Ca2+ responses. Verification of the UHT assay was done with a robustness compound library and clinically relevant platelet inhibitors. Taken together, these results present proof of principle of distinct receptor-type-dependent Ca2+ signaling curves in platelets, which allow identification of new inhibitors in a UHT way.
Collapse
Affiliation(s)
- Delia I. Fernández
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Isabella Provenzale
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- Institute for Cardiovascular and Metabolic Research, University of Reading, RG6 6AX Reading, UK
| | - Hilaire Y.F. Cheung
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- ISASLeibniz-Institut fur Analytische Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany
- Institute of Cardiovascular Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | | | - Bibian M.E. Tullemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Alicia Veninga
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Joanne L. Dunster
- Institute for Cardiovascular and Metabolic Research, University of Reading, RG6 6AX Reading, UK
| | | | | | - Marijke J.E. Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- Thrombosis Expertise Centre, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Johan W.M. Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- Synapse Research Institute, Kon. Emmaplein 7, 6214 AC, Maastricht, the Netherlands
| |
Collapse
|
13
|
Ruksiriwanich W, Khantham C, Linsaenkart P, Chaitep T, Rachtanapun P, Jantanasakulwong K, Phimolsiripol Y, Režek Jambrak A, Nazir Y, Yooin W, Sommano SR, Jantrawut P, Sainakham M, Tocharus J, Mingmalairak S, Sringarm K. Anti‐inflammation of bioactive compounds from ethanolic extracts of edible bamboo mushroom (
Dictyophora indusiata
) as functional health promoting food ingredients. Int J Food Sci Technol 2022. [DOI: 10.1111/ijfs.15338] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Warintorn Ruksiriwanich
- Department of Pharmaceutical Sciences Faculty of Pharmacy Chiang Mai University Chiang Mai 50200 Thailand
- Cluster of Research and Development of Pharmaceutical and Natural Products Innovation for Human or Animal Chiang Mai University Chiang Mai 50200 Thailand
- Cluster of Agro Bio‐Circular‐Green Industry Faculty of Agro‐Industry Chiang Mai University Chiang Mai 50100 Thailand
| | - Chiranan Khantham
- Department of Pharmaceutical Sciences Faculty of Pharmacy Chiang Mai University Chiang Mai 50200 Thailand
| | - Pichchapa Linsaenkart
- Department of Pharmaceutical Sciences Faculty of Pharmacy Chiang Mai University Chiang Mai 50200 Thailand
| | - Tanakarn Chaitep
- Department of Pharmaceutical Sciences Faculty of Pharmacy Chiang Mai University Chiang Mai 50200 Thailand
| | - Pornchai Rachtanapun
- Cluster of Agro Bio‐Circular‐Green Industry Faculty of Agro‐Industry Chiang Mai University Chiang Mai 50100 Thailand
- Faculty of Agro‐Industry Chiang Mai University Chiang Mai 50100 Thailand
| | - Kittisak Jantanasakulwong
- Cluster of Agro Bio‐Circular‐Green Industry Faculty of Agro‐Industry Chiang Mai University Chiang Mai 50100 Thailand
- Faculty of Agro‐Industry Chiang Mai University Chiang Mai 50100 Thailand
| | - Yuthana Phimolsiripol
- Cluster of Agro Bio‐Circular‐Green Industry Faculty of Agro‐Industry Chiang Mai University Chiang Mai 50100 Thailand
- Faculty of Agro‐Industry Chiang Mai University Chiang Mai 50100 Thailand
| | - Anet Režek Jambrak
- Faculty of Food Technology and Biotechnology University of Zagreb Pierottijeva 6 Zagreb 1000 Croatia
| | - Yasir Nazir
- Department of Pharmaceutical Sciences Faculty of Pharmacy Chiang Mai University Chiang Mai 50200 Thailand
| | - Wipawadee Yooin
- Department of Pharmaceutical Sciences Faculty of Pharmacy Chiang Mai University Chiang Mai 50200 Thailand
- Cluster of Research and Development of Pharmaceutical and Natural Products Innovation for Human or Animal Chiang Mai University Chiang Mai 50200 Thailand
| | - Sarana Rose Sommano
- Cluster of Research and Development of Pharmaceutical and Natural Products Innovation for Human or Animal Chiang Mai University Chiang Mai 50200 Thailand
- Cluster of Agro Bio‐Circular‐Green Industry Faculty of Agro‐Industry Chiang Mai University Chiang Mai 50100 Thailand
| | - Pensak Jantrawut
- Department of Pharmaceutical Sciences Faculty of Pharmacy Chiang Mai University Chiang Mai 50200 Thailand
- Cluster of Research and Development of Pharmaceutical and Natural Products Innovation for Human or Animal Chiang Mai University Chiang Mai 50200 Thailand
- Cluster of Agro Bio‐Circular‐Green Industry Faculty of Agro‐Industry Chiang Mai University Chiang Mai 50100 Thailand
| | - Mathukorn Sainakham
- Department of Pharmaceutical Sciences Faculty of Pharmacy Chiang Mai University Chiang Mai 50200 Thailand
- Cluster of Research and Development of Pharmaceutical and Natural Products Innovation for Human or Animal Chiang Mai University Chiang Mai 50200 Thailand
| | | | | | - Korawan Sringarm
- Cluster of Research and Development of Pharmaceutical and Natural Products Innovation for Human or Animal Chiang Mai University Chiang Mai 50200 Thailand
- Cluster of Agro Bio‐Circular‐Green Industry Faculty of Agro‐Industry Chiang Mai University Chiang Mai 50100 Thailand
- Department of Animal and Aquatic Sciences Faculty of Agriculture Chiang Mai University Chiang Mai 50200 Thailand
| |
Collapse
|
14
|
Yu L, Guo Y, Chang Z, Zhang D, Zhang S, Pei H, Pang J, Zhao ZJ, Chen Y. Bidirectional Interaction Between Cancer Cells and Platelets Provides Potential Strategies for Cancer Therapies. Front Oncol 2021; 11:764119. [PMID: 34722319 PMCID: PMC8551800 DOI: 10.3389/fonc.2021.764119] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
Platelets are essential components in the tumor microenvironment. For decades, clinical data have demonstrated that cancer patients have a high risk of thrombosis that is associated with adverse prognosis and decreased survival, indicating the involvement of platelets in cancer progression. Increasing evidence confirms that cancer cells are able to induce production and activation of platelets. Once activated, platelets serve as allies of cancer cells in tumor growth and metastasis. They can protect circulating tumor cells (CTCs) against the immune system and detachment-induced apoptosis while facilitating angiogenesis and tumor cell adhesion and invasion. Therefore, antiplatelet agents and platelet-based therapies should be developed for cancer treatment. Here, we discuss the mechanisms underlying the bidirectional cancer-platelet crosstalk and platelet-based therapeutic approaches.
Collapse
Affiliation(s)
- Liuting Yu
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yao Guo
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhiguang Chang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Dengyang Zhang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Shiqiang Zhang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hanzhong Pei
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Jun Pang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhizhuang Joe Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Yun Chen
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
15
|
Li W, Qu Y, Wen F, Yu R, He X, Jia H, Liu H, Yu H. Prognostic nutritional index and systemic immune-inflammation index are prognostic biomarkers for non-small-cell lung cancer brain metastases. Biomark Med 2021; 15:1071-1084. [PMID: 34397267 DOI: 10.2217/bmm-2020-0786] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Aim: This research aimed to elucidate the prognosis values of prognostic nutritional index (PNI) and systemic immune-inflammation index (SII) and clinical characteristics in NSCLC patients with brain metastases (BM) underwent radiotherapy. Materials & methods: Cut-off points of hematological indicators were determined by receiver operating characteristic curve. Overall survival was evaluated by Kaplan-Meier method and Cox proportional hazards model. Results: We retrospectively analyzed 214 patients from January 2009 to December 2018. The result demonstrated the independent prognostic values of PNI (hazard ratio: 0.600; p = 0.004) and SII (hazard ratio: 1.486; p = 0.019). The remaining clinicopathologic factors, including brain radiotherapy modality, smoking history, numbers of brain metastases, intracranial symptoms and Radiation Therapy Oncology Group - recursive partitioning analysis, were independently related to survival (p < 0.05). Conclusion: PNI and SII could be critical prognostic indicators for NSCLC patients with BM.
Collapse
Affiliation(s)
- Wang Li
- Dalian Medical University, Dalian, Liaoning, 116044, PR China.,Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, no. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, PR China
| | - Yanli Qu
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, no. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, PR China
| | - Fengyun Wen
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, no. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, PR China
| | - Ruoxi Yu
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, no. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, PR China
| | - Xiaoyi He
- Dalian Medical University, Dalian, Liaoning, 116044, PR China
| | - Hongying Jia
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, no. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, PR China
| | - Hangyu Liu
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, no. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, PR China
| | - Hong Yu
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, no. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, PR China
| |
Collapse
|
16
|
Santoro C, Capone V, Canonico ME, Gargiulo G, Esposito R, Sanna GD, Parodi G, Esposito G. Single, Dual, and Triple Antithrombotic Therapy in Cancer Patients with Coronary Artery Disease: Searching for Evidence and Personalized Approaches. Semin Thromb Hemost 2021; 47:950-961. [PMID: 34261150 DOI: 10.1055/s-0041-1726298] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Improvement in life expectancy of patients suffering from oncohematologic disorders has turned cancer from an acute into a chronic condition, making the management of comorbidities problematic, especially when it comes to both acute and chronic cardiovascular diseases. Treatment-related adverse events and drug-drug interactions often influence the therapeutic approach of patients with active malignancies and cardiovascular disease. Furthermore, tumor cells and platelets maintain a complex crosstalk that on one hand enhances tumor dissemination and on the other hand induces hemostasis abnormalities. Hence, clinicians should move carefully in the intricate land mines established by patients with active cancer under antithrombotic therapy. To date, there is no consensus on the antithrombotic treatment of patients with cardiovascular diseases and concomitant malignancies. The aim of this review is to collect the available scientific evidence, including the latest clinical trials and guidelines, in order to provide guidance on the management of antithrombotic treatment (both antiplatelet and anticoagulant therapy) in cancer patients with either pre-existent or new-onset coronary artery disease. Randomized-controlled trials on antithrombotic treatment in oncologic populations, which by far have thus far been excluded, have to be promoted to supply recommendations in the oncohematologic setting.
Collapse
Affiliation(s)
- Ciro Santoro
- Department of Advanced Biomedical Sciences, Federico II University Hospital, Naples, Italy
| | - Valentina Capone
- Department of Advanced Biomedical Sciences, Federico II University Hospital, Naples, Italy
| | - Mario Enrico Canonico
- Department of Advanced Biomedical Sciences, Federico II University Hospital, Naples, Italy.,Clinical and Interventional Cardiology, Sassari University Hospital, Sassari, Italy
| | - Giuseppe Gargiulo
- Department of Advanced Biomedical Sciences, Federico II University Hospital, Naples, Italy
| | - Roberta Esposito
- Department of Advanced Biomedical Sciences, Federico II University Hospital, Naples, Italy
| | | | - Guido Parodi
- Clinical and Interventional Cardiology, Sassari University Hospital, Sassari, Italy
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, Federico II University Hospital, Naples, Italy
| |
Collapse
|
17
|
Tyrosine Kinase Inhibitor Sunitinib Delays Platelet-Induced Coagulation: Additive Effects of Aspirin. Thromb Haemost 2021; 122:92-104. [PMID: 34130349 DOI: 10.1055/s-0041-1730312] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Sunitinib is a multitarget tyrosine kinase inhibitor (TKI) used for cancer treatment. In platelets, sunitinib affects collagen-induced activation under noncoagulating conditions. We investigated (1) the effects of sunitinib on thrombus formation induced by other TK-dependent receptors, and (2) the effects under coagulating conditions. Cardiovascular disease is a comorbidity in cancer patients, resulting in possible aspirin treatment. Sunitinib and aspirin are associated with increased bleeding risk, and therefore we also investigated (3) the synergistic effects of these compounds on thrombus and fibrin formation. METHODS Blood or isolated platelets from healthy volunteers or cancer patients were incubated with sunitinib and/or aspirin or vehicle. Platelet activation was determined by TK phosphorylation, flow cytometry, changes in [Ca2+]i, aggregometry, and whole blood perfusion over multiple surfaces, including collagen with(out) tissue factor (TF) was performed. RESULTS Sunitinib reduced thrombus formation and phosphatidylserine (PS) exposure under flow on collagen type I and III. Also, sunitinib inhibited glycoprotein VI-induced TK phosphorylation and Ca2+ elevation. Upon TF-triggered coagulation, sunitinib decreased PS exposure and fibrin formation. In blood from cancer patients more pronounced effects of sunitinib were observed in lung and pancreatic as compared to neuroglioblastoma and other cancer types. Compared to sunitinib alone, sunitinib plus aspirin further reduced platelet aggregation, thrombus formation, and PS exposure on collagen under flow with(out) coagulation. CONCLUSION Sunitinib suppresses collagen-induced procoagulant activity and delays fibrin formation, which was aggravated by aspirin. Therefore, we urge for awareness of the combined antiplatelet effects of TKIs with aspirin, as this may result in increased risk of bleeding.
Collapse
|
18
|
Hong YM, Yoon KT, Cho M. Systemic immune-inflammation index predicts prognosis of sequential therapy with sorafenib and regorafenib in hepatocellular carcinoma. BMC Cancer 2021; 21:569. [PMID: 34006248 PMCID: PMC8130266 DOI: 10.1186/s12885-021-08124-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 03/29/2021] [Indexed: 12/26/2022] Open
Abstract
Background Regorafenib has shown promising results as a second-line therapy for patients with hepatocellular carcinoma (HCC) who progressed on sorafenib. Although there have been several data regarding the efficacy of sequential therapy with sorafenib and that of regorafenib in real-life, specific inflammation markers for predicting the prognosis have not been studied. This study aimed to investigate prognostic value of systemic inflammatory markers in patients with HCC who received sorafenib-regorafenib sequential therapy. Methods We retrospectively analyzed medical data of patients who received regorafenib for the treatment of HCC after sorafenib failure. Progression free survival (PFS) and overall survival (OS) were assessed using the Kaplan–Meier survival curves. Univariate and multivariate analyses were performed to analyze the factors associated with survival. Results A total of 58 patients who received at least one dose of regroafenib and fulfilled the eligibility criteria, good performance status (Eastern Cooperative Oncology Group [ECOG] 0–1) and preserved liver function (Child-Pugh-A), were included in the analysis. The median PFS was 3 months (95% confidence interval [CI] = 0.981–5.019) and the median OS was 8 months (95% CI = 5.761–10.239). Elevated systemic immune-inflammation index (SII ≥340) was independently associated with poor OS. In multivariate analysis, the SII (hazard ratio [HR] = 2.211, 95% CI = 1.089–4.489, P = 0.028) and alpha-fetoprotein (AFP) (HR = 2.750, 95% CI = 1.259–6.010, P = 0.011) were independent predictors of OS. Conclusion Elevated SII is associated with poor OS in patients with HCC who received sequential therapy with sorafenib and regorafenib. In addition, when selecting a treatment strategy, the SII can be used in combination with the AFP level as a promising prognostic tool for HCC.
Collapse
Affiliation(s)
- Young Mi Hong
- Department of Internal Medicine, Liver Center, Pusan National University School of Medicine, Pusan National University Yangsan Hospital, 20 Geumo-ro, Gyeongnam, 50612, Yangsan, South Korea
| | - K T Yoon
- Department of Internal Medicine, Liver Center, Pusan National University School of Medicine, Pusan National University Yangsan Hospital, 20 Geumo-ro, Gyeongnam, 50612, Yangsan, South Korea
| | - Mong Cho
- Department of Internal Medicine, Liver Center, Pusan National University School of Medicine, Pusan National University Yangsan Hospital, 20 Geumo-ro, Gyeongnam, 50612, Yangsan, South Korea.
| |
Collapse
|
19
|
Abstract
Until recently, the nucleic acid content of platelets was considered to be fully determined by their progenitor megakaryocyte. However, it is now well understood that additional mediators (eg, cancer cells) can intervene, thereby influencing the RNA repertoire of platelets. Platelets are highly dynamic cells that are able to communicate and influence their environment. For instance, platelets have been involved in various steps of cancer development and progression by supporting tumor growth, survival, and dissemination. Cancer cells can directly and/or indirectly influence platelet RNA content, resulting in tumor-mediated "education" of platelets. Alterations in the tumor-educated platelet RNA profile have been described as a novel source of potential biomarkers. Individual platelet RNA biomarkers as well as complex RNA signatures may be used for early detection of cancer and treatment monitoring. Here, we review the RNA transfer occurring between cancer cells and platelets. We explore the potential use of platelet RNA biomarkers as a liquid biopsy biosource and discuss methods to evaluate the transcriptomic content of platelets.
Collapse
|
20
|
Kao CC, Kung PH, Tai CJ, Tsai MC, Cheng YB, Wu CC. Juglone prevents human platelet aggregation through inhibiting Akt and protein disulfide isomerase. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 82:153449. [PMID: 33387969 DOI: 10.1016/j.phymed.2020.153449] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/24/2020] [Accepted: 12/22/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND/PURPOSE Juglone, a natural compound widely found in Juglandaceae plants, has been suggested as a potential drug candidate for treating cancer, inflammation, and diabetic vascular complications. In the present study, the antiplatelet effect and underlying mechanisms of juglone were investigated for the first time. STUDY DESIGN/METHODS Human platelet aggregation and activation were measured by turbidimetric aggregometry, flow cytometry, and Western blotting. In vitro antithrombotic activity of juglone was assessed using collagen-coated flow chambers under whole-blood flow conditions. The effect of juglone on protein disulfide isomerase (PDI) activity was determined by the dieosin glutathione disulfide assay. RESULTS Juglone (1 - 5 μM) inhibited platelet aggregation and glycoprotein (GP) IIb/IIIa activation caused by various agonists. In a whole blood flow chamber system, juglone reduced thrombus formation on collagen-coated surfaces under arterial shear rates. Juglone abolished intracellular Ca2+ elevation and protein kinase C activation caused by collagen, but had no significant effect on that induced by G protein-coupled receptor agonists. In contrast, Akt activation caused by various agonists were inhibited in juglone-treated platelets. Additionally, juglone showed inhibitory effects on both recombinant human PDI and platelet surface PDI at concentrations similar to those needed to prevent platelet aggregation. CONCLUSION Juglone exhibits potent in vitro antiplatelet and antithrombotic effects that are associated with inhibition of Akt activation and platelet surface PDI activity.
Collapse
Affiliation(s)
- Ching-Chieh Kao
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Hsiung Kung
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Jung Tai
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Family Medicine, Pingtung Hospital, Ministry of Health and Welfare, Pingtung, Taiwan
| | - Meng-Chun Tsai
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuan-Bin Cheng
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chin-Chung Wu
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
21
|
Gutiontov SI, Choe KS, Miller JL, Liauw SL. Improved outcomes after radiotherapy for prostate cancer: Anticoagulation, antiplatelet therapy, and platelet count as key factors in disease progression. Cancer Med 2020; 9:4667-4675. [PMID: 32400122 PMCID: PMC7333841 DOI: 10.1002/cam4.3087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Several studies have suggested that antiplatelet (AP) or anticoagulant (AC) therapy may improve outcome in men with prostate cancer. We evaluated the effects of AP/AC therapy and tested the hypothesis that platelet count may also be associated with outcomes. METHODS A total of 482 patients received primary radiotherapy (median dose 72 Gy) for nonmetastatic prostate cancer; 49% received androgen deprivation therapy. NCCN risk was low/intermediate/high risk in 39%/39%/22%. AP/AC therapy and platelet counts were analyzed with respect to freedom from biochemical failure (FFBF, nadir+2), distant metastasis (FFDM), and cause specific survival (CSS). RESULTS After a median follow-up of 103 months, 10-year FFBF, FFDM, and CSS were 77%, 92%, and 96%, respectively. The 10-year cumulative incidence of BF and DM (with death as a competing event) was 19% and 7.0%, respectively. The 32% of men on AP/AC therapy had a lower incidence of 10-year BF (P = .016) and a trend toward a lower incidence of DM (P = .084) and CSS (P = .091). In the entire cohort, lowest platelet quartile (platelet count <187) was associated with higher 10-year BF (31% vs 16%, P = .0042) but not DM (9.4% vs 5.2%, P = .22) nor CSS (P = .76) compared with those patients with platelet count ≥187. AP/AC therapy was associated with a larger absolute reduction in BF for men with lowest platelet quartile (10-year BF of 21% vs 38%, P = .092) vs platelet ≥187 (10-year BF of 10% vs 18%, P = .053). Lowest platelet quartile remained associated with higher BF and DM on multivariable analysis controlling for risk category, WBC, and Hg. CONCLUSION AP/AC was associated with improved FFBF. Low platelet count was associated with inferior FFBF and FFDM after prostate radiotherapy. This association was tempered when antiplatelet and anticoagulant therapy was administered.
Collapse
Affiliation(s)
| | - Kevin S. Choe
- Radiation Oncology AssociatesInova HospitalFairfaxVAUSA
| | | | - Stanley L. Liauw
- Department of Radiation and Cellular OncologyUniversity of ChicagoChicagoILUSA
| |
Collapse
|
22
|
Gu W, Yu F, Mao Q, Qiu F. Prognostic analysis of peripheral blood inflammatory markers in 99 nasopharyngeal carcinoma patients with recurrence and metastasis. PRECISION RADIATION ONCOLOGY 2020. [DOI: 10.1002/pro6.1084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Weiguo Gu
- First Affiliated Hospital of Nanchang University Nanchang China
- Nanchang University Nanchang China
| | - Feng Yu
- First Affiliated Hospital of Nanchang University Nanchang China
| | - Qiao Mao
- Shangrao Maternity and Child Health Hospital Shangrao China
| | - Feng Qiu
- First Affiliated Hospital of Nanchang University Nanchang China
| |
Collapse
|
23
|
Marturano A, Hendrickx ML, Falcinelli E, Sebastiano M, Guglielmini G, Hassanzadeh-Ghassabeh G, Muyldermans S, Declerck PJ, Gresele P. Development of anti-matrix metalloproteinase-2 (MMP-2) nanobodies as potential therapeutic and diagnostic tools. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 24:102103. [DOI: 10.1016/j.nano.2019.102103] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/13/2019] [Accepted: 09/27/2019] [Indexed: 12/14/2022]
|
24
|
Sun L, Li Q, Guo Y, Yang Q, Yin J, Ran Q, Liu L, Zhao Z, Wang Y, Li Y, Chen Y, Weng X, Cai W, Zhu X. Extract of Caulis Spatholobi, a novel platelet inhibitor,efficiently suppresses metastasis of colorectal cancer by targeting tumor cell-induced platelet aggregation. Biomed Pharmacother 2020; 123:109718. [PMID: 31918208 DOI: 10.1016/j.biopha.2019.109718] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/17/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022] Open
Abstract
Tumor cell-induced platelet aggregation (TCIPA) is the core mechanism potentiating high viability for circulatory tumor cells,which is the rate-limiting factor for metastasis.Additionally,as supported by the successful application of aspirin,the pro-malignant effects during tumor-platelets interaction can be largely neutralized by pharmacological deactivation of platelets.Caulis Spatholobi is widely used as an anti-coagulation herb in traditional Chinese medicine,indicating its potential against TCIPA.In our study,three fractions of Caulis Spatholobi extracts were firstly prepared.In colorectal cancer(CRC) model,the anti-metastatic potential was evaluated both in vitro and in vivo followed by the detection of their platlet regulatory effects.Results showed that all three extracts significantly suppressed the invasion and metastasis of CRC.Mechanistically,by blocking platelet-derived PDGF-B releasing,they reversed the enhanced epithelial mesenchymal transition during MC38-platelets interation.Further,ethyl acetate fraction shows the most promising efficacy for the future application in treatment.Overall,our study have for the first time proved CaulisSpatholobi extracts,especially the ethyl acetate fraction,as a potent TCIPA inhibitor during metastatic progression,which provided a novel candidate for pharmacologically blockage of metastasis in CRC.
Collapse
Affiliation(s)
- Lidong Sun
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qi Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuan Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qing Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jie Yin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qingsen Ran
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zheng Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yajie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yujie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiaogang Weng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Weiyan Cai
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiaoxin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
25
|
Pantazi D, Ntemou N, Brentas A, Alivertis D, Skobridis K, Tselepis AD. Molecular Requirements for the Expression of Antiplatelet Effects by Synthetic Structural Optimized Analogues of the Anticancer Drugs Imatinib and Nilotinib. Drug Des Devel Ther 2019; 13:4225-4238. [PMID: 31849454 PMCID: PMC6913343 DOI: 10.2147/dddt.s211907] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/12/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Platelets play important roles in cancer progression and metastasis, as well as in cancer-associated thrombosis (CAT). Tyrosine kinases are implicated in several intracellular signaling pathways involved in tumor biology, thus tyrosine kinase inhibitors (TKIs) represent an important class of anticancer drugs, based on the concept of targeted therapy. PURPOSE The objective of this study is the design and synthesis of analogues of the TKIs imatinib and nilotinib in order to develop tyrosine kinase inhibitors, by investigating their molecular requirements, which would express antiplatelet properties. METHODS Based on a recently described by us improved approach in the preparation of imatinib and/or nilotinib analogues, we designed and synthesized in five-step reaction sequences, 8 analogues of imatinib (I-IV), nilotinib (V, VI) and imatinib/nilotinib (VII, VIII). Their inhibitory effects on platelet aggregation and P-selectin membrane expression induced by arachidonic acid (AA), adenosine diphosphate (ADP) and thrombin receptor activating peptide-6 (TRAP-6), in vitro, were studied. Molecular docking studies and calculations were also performed. RESULTS The novel analogues V-VIII were well established with the aid of spectroscopic methods. Imatinib and nilotinib inhibited AA-induced platelet aggregation, exhibiting IC50 values of 13.30 μΜ and 3.91 μΜ, respectively. Analogues I and II exhibited an improved inhibitory activity compared with imatinib. Among the nilotinib analogues, V exhibited a 9-fold higher activity than nilotinib. All compounds were less efficient in inhibiting platelet aggregation towards ADP and TRAP-6. Similar results were obtained for the membrane expression of P-selectin. Molecular docking studies showed that the improved antiplatelet activity of nilotinib analogue V is primarily attributed to the number and the strength of hydrogen bonds. CONCLUSION Our results show that there is considerable potential to develop synthetic analogues of imatinib and nilotinib, as TKIs with antiplatelet properties and therefore being suitable to target cancer progression and metastasis, as well as CAT by inhibiting platelet activation.
Collapse
Affiliation(s)
- Despoina Pantazi
- Department of Chemistry, Atherothrombosis Research Centre, Laboratory of Biochemistry, University of Ioannina, Ioannina45110, Greece
| | - Nikoleta Ntemou
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, Ioannina45110, Greece
| | - Alexios Brentas
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, Ioannina45110, Greece
| | - Dimitrios Alivertis
- Department of Biological Applications and Technology, University of Ioannina, Ioannina45110, Greece
| | - Konstantinos Skobridis
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, Ioannina45110, Greece
| | - Alexandros D Tselepis
- Department of Chemistry, Atherothrombosis Research Centre, Laboratory of Biochemistry, University of Ioannina, Ioannina45110, Greece
| |
Collapse
|
26
|
Platelet Indices in Colorectal Cancer Patients with Synchronous Liver Metastases. Gastroenterol Res Pract 2019; 2019:6397513. [PMID: 31781195 PMCID: PMC6874931 DOI: 10.1155/2019/6397513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 09/21/2019] [Accepted: 10/15/2019] [Indexed: 01/02/2023] Open
Abstract
Aims Liver metastases occur in approximately 25% of colorectal cancer (CRC) patients and cause more than 90% of deaths in CRC. Platelets play a crucial role in cancer progression and metastases. We aimed to investigate the relationship between platelet indices and CRC with synchronous liver metastases. Methods We conducted a retrospective clinical study including 206 CRC patients without metastases and 200 CRC patients with synchronous liver metastases from January 1, 2015, to December 31, 2017. Data of the patients' clinicopathological characteristics were collected. Results Platelet distribution width (PDW) was decreased in CRC patients with liver metastases compared with CRC patients without liver metastases. In addition, the prevalence of liver metastases reduced as PDW quartiles increased. After adjusting for other risk factors, the odds ratios (95% confidence intervals) for CRC liver metastases according to PDW quartiles were 1.000, 0.289 (0.156-0.535), 0.482 (0.271-0.860), and 0.190 (0.101-0.358). Conclusions Compared with CRC patients without metastases, PDW is reduced in CRC patients with liver metastases. Moreover, PDW was independently associated with the presence of CRC liver metastases.
Collapse
|
27
|
Li MM, Yue CX, Fu S, Zhang X, Zhao CJ, Wang RT. Platelet Volume Is Reduced In Metastasing Breast Cancer: Blood Profiles Reveal Significant Shifts. Cancer Manag Res 2019; 11:9067-9072. [PMID: 31695497 PMCID: PMC6817343 DOI: 10.2147/cmar.s221976] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/10/2019] [Indexed: 12/17/2022] Open
Abstract
Background Platelets play a crucial role in breast cancer (BC) progression and metastases. Mean platelet volume (MPV) is an indicator of platelet activation. The aim of the present study was to assess whether there is a difference in MPV between patients with metastatic BC with liver metastases and those with BC without liver metastases. Methods Between January 2014 and December 2017, 211 metastatic BC patients with synchronous liver metastases and 215 BC patients without metastases were retrospectively analyzed. Patients’ clinicopathological characteristics data were collected. Results MPV levels were reduced in patients with liver metastases compared with those in patients without liver metastases. There were significant differences in MPV levels according to liver metastases status both in premenopausal and in postmenopausal non-TNBC or non-HER2+ patients. Moreover, in postmenopausal HER2+ or TNBC patients, MPV levels were lower in patients with liver metastases compared with those in patients without liver metastases. In the group with non-liver metastasis, platelet distribution width was significantly associated with tumor N stage. In addition, the prevalence of BC liver metastases decreased as MPV quartiles increased. After adjusting for other risk factors, the odds ratios for liver metastases according to MPV quartiles were 1.000, 0.267 (0.134–0.530), 0.072 (0.034–0.152), and 0.137 (0.066–0.281), respectively. Conclusion MPV is reduced in BC patients with liver metastases compared with that in BC patients without metastases. Moreover, MPV is independently associated with the presence of liver metastases.
Collapse
Affiliation(s)
- Ming-Ming Li
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, People's Republic of China
| | - Chen-Xi Yue
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, People's Republic of China
| | - Shuang Fu
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, People's Republic of China
| | - Xin Zhang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, People's Republic of China
| | - Chang-Jiu Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, People's Republic of China
| | - Rui-Tao Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, People's Republic of China
| |
Collapse
|
28
|
Ward Y, Lake R, Faraji F, Sperger J, Martin P, Gilliard C, Ku KP, Rodems T, Niles D, Tillman H, Yin J, Hunter K, Sowalsky AG, Lang J, Kelly K. Platelets Promote Metastasis via Binding Tumor CD97 Leading to Bidirectional Signaling that Coordinates Transendothelial Migration. Cell Rep 2019; 23:808-822. [PMID: 29669286 DOI: 10.1016/j.celrep.2018.03.092] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 02/16/2018] [Accepted: 03/20/2018] [Indexed: 02/02/2023] Open
Abstract
Tumor cells initiate platelet activation leading to the secretion of bioactive molecules, which promote metastasis. Platelet receptors on tumors have not been well-characterized, resulting in a critical gap in knowledge concerning platelet-promoted metastasis. We identify a direct interaction between platelets and tumor CD97 that stimulates rapid bidirectional signaling. CD97, an adhesion G protein-coupled receptor (GPCR), is an overexpressed tumor antigen in several cancer types. Purified CD97 extracellular domain or tumor cell-associated CD97 stimulated platelet activation. CD97-initiated platelet activation led to granule secretion, including the release of ATP, a mediator of endothelial junction disruption. Lysophosphatidic acid (LPA) derived from platelets induced tumor invasiveness via proximal CD97-LPAR heterodimer signaling, coupling coincident tumor cell migration and vascular permeability to promote transendothelial migration. Consistent with this, CD97 was necessary for tumor cell-induced vascular permeability in vivo and metastasis formation in preclinical models. These findings support targeted blockade of tumor CD97 as an approach to ameliorate metastatic spread.
Collapse
Affiliation(s)
- Yvona Ward
- Laboratory of Genitourinary Cancer Pathogenesis, NCI, Bethesda, MD 20892, USA
| | - Ross Lake
- Laboratory of Genitourinary Cancer Pathogenesis, NCI, Bethesda, MD 20892, USA
| | - Farhoud Faraji
- Laboratory of Cancer Biology and Genetics, NCI, Bethesda, MD 20892, USA
| | - Jamie Sperger
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Philip Martin
- Center for Advanced Preclinical Research, NCI, Frederick, MD 21702, USA
| | - Cameron Gilliard
- Molecular Biology and Genetics Section, NIDDK, Bethesda, MD 20892, USA
| | - Kimberly P Ku
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Tamara Rodems
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David Niles
- Depatment of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Heather Tillman
- Laboratory of Genitourinary Cancer Pathogenesis, NCI, Bethesda, MD 20892, USA
| | - JuanJuan Yin
- Laboratory of Genitourinary Cancer Pathogenesis, NCI, Bethesda, MD 20892, USA
| | - Kent Hunter
- Laboratory of Cancer Biology and Genetics, NCI, Bethesda, MD 20892, USA
| | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, NCI, Bethesda, MD 20892, USA
| | - Joshua Lang
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kathleen Kelly
- Laboratory of Genitourinary Cancer Pathogenesis, NCI, Bethesda, MD 20892, USA.
| |
Collapse
|
29
|
Abstract
For over 100 years, a link has been recognized between thrombosis and cancer. However, whether this was a causal or correlational relationship was debated. It is now well established that cancer and thrombosis are mechanistically related in intricate ways and can directly fuel each other. Here, we present an historical perspective of platelets and how their physiological function in hemostasis can contribute to tumor development and metastasis. This emerging field has garnered great interest as aspirin therapy has been proposed as a prevention strategy for some malignancies. We highlight the advances that have been made, presenting platelets as a key component that supports many of the hallmarks of cancer that have been described and conclude with future directions and studies that are needed to clarify the role of platelets in cancer and solidify platelet modulating therapies within oncology.
Collapse
Affiliation(s)
- Aime T Franco
- Department of Physiology & Biophysics, University of Arkansas for Medical Sciences, Slot 505, 4301 W. Markham Street, Little Rock, AR, 72205, USA.
| | - Jerry Ware
- Department of Physiology & Biophysics, University of Arkansas for Medical Sciences, Slot 505, 4301 W. Markham Street, Little Rock, AR, 72205, USA
| |
Collapse
|
30
|
Mamdooh N, Kasabri V, Al‐Hiari Y, Almasri I, Al‐Alawi S, Bustanji Y. Evaluation of selected commercial pharmacotherapeutic drugs as potential pancreatic lipase inhibitors and antiproliferative compounds. Drug Dev Res 2018; 80:310-324. [DOI: 10.1002/ddr.21499] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 11/11/2022]
Affiliation(s)
- Noor Mamdooh
- School of PharmacyUniversity of Jordan Amman Jordan
| | | | | | - Ihab Almasri
- Pharmaceutical Chemistry DepartmentAl‐Azhar University Gaza Palestinian Territory
| | | | - Yasser Bustanji
- School of PharmacyUniversity of Jordan Amman Jordan
- Hamdi Mango Centre for Scientific ResearchUniversity of Jordan Amman Jordan
| |
Collapse
|
31
|
Anti-platelet treatments in cancer: Basic and clinical research. Thromb Res 2018; 164 Suppl 1:S106-S111. [PMID: 29703466 DOI: 10.1016/j.thromres.2017.12.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 12/27/2017] [Indexed: 02/07/2023]
Abstract
Over the past few decades the central role that platelets play in cancer development and progression, and especially in metastasis, has been elucidated. The molecular mechanisms responsible for initiating and mediating tumor cell-induced platelet aggregation and secretion have been largely unravelled. Considerable mechanistic insight into how platelets contribute to tumor angiogenesis, immunoevasion and cancer cell invasion have been clarified and, consequently, platelets have been identified as potential new drug targets for cancer therapy. This article gives an overview of the platelet-targeted pharmacologic approaches that have been attempted in the prevention of cancer development, progression and metastasis, including the application of antiplatelet drugs currently used for cardiovascular disease and of new and novel strategies.
Collapse
|
32
|
Di Virgilio F, Sarti AC, Falzoni S, De Marchi E, Adinolfi E. Extracellular ATP and P2 purinergic signalling in the tumour microenvironment. Nat Rev Cancer 2018; 18:601-618. [PMID: 30006588 DOI: 10.1038/s41568-018-0037-0] [Citation(s) in RCA: 493] [Impact Index Per Article: 70.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Modulation of the biochemical composition of the tumour microenvironment is a new frontier of cancer therapy. Several immunosuppressive mechanisms operate in the milieu of most tumours, a condition that makes antitumour immunity ineffective. One of the most potent immunosuppressive factors is adenosine, which is generated in the tumour microenvironment owing to degradation of extracellular ATP. Accruing evidence over the past few years shows that ATP is one of the major biochemical constituents of the tumour microenvironment, where it acts at P2 purinergic receptors expressed on both tumour and host cells. Stimulation of P2 receptors has different effects depending on the extracellular ATP concentration, the P2 receptor subtype engaged and the target cell type. Among P2 receptors, the P2X purinergic receptor 7 (P2X7R) subtype appears to be a main player in host-tumour cell interactions. Preclinical studies in several tumour models have shown that P2X7R targeting is potentially a very effective anticancer treatment, and many pharmaceutical companies have now developed potent and selective small molecule inhibitors of P2X7R. In this Review, we report on the multiple mechanisms by which extracellular ATP shapes the tumour microenvironment and how its stimulation of host and tumour cell P2 receptors contributes to determining tumour fate.
Collapse
Affiliation(s)
- Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.
| | - Alba Clara Sarti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Simonetta Falzoni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Elena De Marchi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Elena Adinolfi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
33
|
Abstract
Several lines of evidence are consistent with the hypothesis that activated platelets contribute to colorectal tumorigenesis and metastatization through direct cell-cell interactions and the release of different lipid and protein mediators, and microvesicles. This review examines the clinical pharmacology of low-dose aspirin as a basis for discussing the mechanisms underlying the contribution of platelets to neoplastic transformation and progression of cancer via the development of metastases.
Collapse
Affiliation(s)
- Paola Patrignani
- a Department of Neuroscience, Imaging and Clinical Sciences, Section of Cardiovascular and Pharmacological Sciences, and CeSI-MeT (Centro Scienze dell' Invecchiamento e Medicina Traslazionale) , "G. d'Annunzio" University , Chieti , Italy
| | - Carlo Patrono
- b Department of Pharmacology , Catholic University School of Medicine , Rome , Italy
| |
Collapse
|
34
|
Abstract
Thrombosis is a frequent issue in cancer patients. Tumor-induced platelet activation and coagulation does not only constitute a significant risk for thrombosis, but also contribute to tumor progression by promoting critical processes such as angiogenesis and metastasis. In addition to their role in hemostasis, platelets are increasingly recognized as regulators of inflammation. By modulating the immune system, platelets regulate several aspects of cancer-associated pathology. Platelets influence the inflammatory response in cancer by affecting the activation status of the endothelium and by recruiting leukocytes to primary and metastatic tumor sites, as well as to distant organs unaffected by tumor growth. Furthermore, platelets participate in the formation of neutrophil extracellular traps, which can promote metastasis, thrombosis, and contribute to organ failure. In this review, we discuss the role of platelets as coordinators of the immune system during malignant disease and the potential of targeting platelets to prevent cancer-associated pathology.
Collapse
Affiliation(s)
- A K Olsson
- a Department of Medical Biochemistry and Microbiology , Science for Life Laboratory, Uppsala University, Biomedical Center , Uppsala , Sweden
| | - J Cedervall
- a Department of Medical Biochemistry and Microbiology , Science for Life Laboratory, Uppsala University, Biomedical Center , Uppsala , Sweden
| |
Collapse
|
35
|
Deng J, Zhang P, Sun Y, Peng P, Huang Y. Prognostic and clinicopathological significance of platelet to lymphocyte ratio in esophageal cancer: a meta-analysis. J Thorac Dis 2018; 10:1522-1531. [PMID: 29707302 DOI: 10.21037/jtd.2018.02.58] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background The prognostic and clinicopathological significance of the platelet to lymphocyte ratio (PLR) has been studied in various cancers. However, studies examining the role of PLR in esophageal cancer have not yielded consistent results. The purpose of this meta-analysis was to study the prognostic and clinicopathological significance of PLR in esophageal cancer patients. Methods We performed a literature search in three major databases: PubMed, Web of Science and Embase (up until May 1, 2017). The clinicopathologic significance of PLR and its prognostic significance were analyzed. Results Our meta-analysis consisted of 13 studies with 4,621 patients. The pooled hazard ratios (HRs) showed that a high PLR was associated with poor survival of esophageal cancer [HR =1.283; 95% confidence interval (CI): 1.173-1.404; P<0.001]. Subgroup analysis revealed that elevated PLR was associated with poor survival in esophageal squamous cell carcinoma (HR =1.281; 95% CI: 1.098-1.493; P=0.002). The pooled odds ratio (OR) indicated that high PLR was also associated with the depth of tumor invasion (OR =1.543, 95% CI: 1.269-1.876, P<0.001), lymph node metastasis (OR =1.427, 95% CI: 1.195-1.705, P<0.001), tumor length (OR =1.81, 95% CI: 1.331-2.461, P<0.001), and Tumor stage (OR =1.459, 95% CI: 1.235-1.724, P<0.001). Conclusions Our results demonstrate that elevated PLR was significantly associated with poor prognosis of esophageal cancer. Furthermore, the high PLR might predict worse clinicopathological features of esophageal cancer patients.
Collapse
Affiliation(s)
- Juhong Deng
- Department of Endocrinology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Peng Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yue Sun
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ping Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
36
|
Rwibasira Rudinga G, Khan GJ, Kong Y. Protease-Activated Receptor 4 (PAR4): A Promising Target for Antiplatelet Therapy. Int J Mol Sci 2018; 19:E573. [PMID: 29443899 PMCID: PMC5855795 DOI: 10.3390/ijms19020573] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/09/2018] [Accepted: 02/09/2018] [Indexed: 12/29/2022] Open
Abstract
Cardiovascular diseases (CVDs) are currently among the leading causes of death worldwide. Platelet aggregation is a key cellular component of arterial thrombi and major cause of CVDs. Protease-activated receptors (PARs), including PAR1, PAR2, PAR3 and PAR4, fall within a subfamily of seven-transmembrane G-protein-coupled receptors (GPCR). Human platelets express PAR1 and PAR4, which contribute to the signaling transduction processes. In association with CVDs, PAR4 not only contributes to platelet activation but also is a modulator of cellular responses that serve as hallmarks of inflammation. Although several antiplatelet drugs are available on the market, they have many side effects that limit their use. Emerging evidence shows that PAR4 targeting is a safer strategy for preventing thrombosis and consequently may improve the overall cardiac safety profile. Our present review summarizes the PAR4 structural characteristics, activation mechanism, role in the pathophysiology of diseases and understanding the association of PAR4 targeting for improved cardiac protection. Conclusively, this review highlights the importance of PAR4 antagonists and its potential utility in different CVDs.
Collapse
Affiliation(s)
- Gamariel Rwibasira Rudinga
- School of Life Science & Technology, China Pharmaceutical University, 24 Tong Jia Street, Nanjing 210009, China.
| | - Ghulam Jilany Khan
- Jiangsu Center for Pharmacodynamics Research, Evaluation and Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| | - Yi Kong
- School of Life Science & Technology, China Pharmaceutical University, 24 Tong Jia Street, Nanjing 210009, China.
| |
Collapse
|