51
|
Blood Circulating CD133+ Extracellular Vesicles Predict Clinical Outcomes in Patients with Metastatic Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14051357. [PMID: 35267665 PMCID: PMC8909146 DOI: 10.3390/cancers14051357] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/28/2022] [Accepted: 03/05/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary In this study, we explored the prognostic and predictive value of blood circulating EVs expressing selected surface proteins in patients with metastatic colorectal cancer (mCRC). A recently patented flow cytometry protocol was used for the identification and subtyping of blood circulating EVs in a cohort of patients with stage IV colorectal cancer (n = 54) and in a cohort of healthy controls (n = 48). We observed an increased blood concentration of tumor-induced blood circulating EVs in the mCRC cohort as compared to healthy controls. Additionally, we show an intriguing link between circulating CD133+ EVs and poor clinical outcomes in patients with mCRC. This study provides novel insights about the potential impact of EVs as a relevant source of candidate biomarkers in mCRC. Abstract Colorectal cancer (CRC) is one of the most incident and lethal malignancies worldwide. Recent treatment advances prolonged survival in patients with metastatic colorectal cancer (mCRC). However, there are still few biomarkers to guide clinical management and treatment selection in mCRC. In this study, we applied an optimized flow cytometry protocol for EV identification, enumeration, and subtyping in blood samples of 54 patients with mCRC and 48 age and sex-matched healthy controls (HCs). The overall survival (OS) and overall response rate (ORR) were evaluated in mCRC patients enrolled and treated with a first line fluoropyrimidine-based regimen. Our findings show that patients with mCRC presented considerably higher blood concentrations of total EVs, as well as CD133+ and EPCAM+ EVs compared to HCs. Overall survival analysis revealed that increased blood concentrations of total EVs and CD133+ EVs before treatment were significantly associated with shorter OS in mCRC patients (p = 0.001; and p = 0.0001, respectively). In addition, we observed a correlation between high blood levels of CD133+ EVs at baseline and reduced ORR to first-line systemic therapy (p = 0.045). These findings may open exciting perspectives into the application of novel blood-based EV biomarkers for improved risk stratification and optimized treatment strategies in mCRC.
Collapse
|
52
|
Yu D, Li Y, Wang M, Gu J, Xu W, Cai H, Fang X, Zhang X. Exosomes as a new frontier of cancer liquid biopsy. Mol Cancer 2022; 21:56. [PMID: 35180868 PMCID: PMC8855550 DOI: 10.1186/s12943-022-01509-9] [Citation(s) in RCA: 331] [Impact Index Per Article: 165.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/15/2022] [Indexed: 02/08/2023] Open
Abstract
Liquid biopsy, characterized by minimally invasive detection through biofluids such as blood, saliva, and urine, has emerged as a revolutionary strategy for cancer diagnosis and prognosis prediction. Exosomes are a subset of extracellular vesicles (EVs) that shuttle molecular cargoes from donor cells to recipient cells and play a crucial role in mediating intercellular communication. Increasing studies suggest that exosomes have a great promise to serve as novel biomarkers in liquid biopsy, since large quantities of exosomes are enriched in body fluids and are involved in numerous physiological and pathological processes. However, the further clinical application of exosomes has been greatly restrained by the lack of high-quality separation and component analysis methods. This review aims to provide a comprehensive overview on the conventional and novel technologies for exosome isolation, characterization and content detection. Additionally, the roles of exosomes serving as potential biomarkers in liquid biopsy for the diagnosis, treatment monitoring, and prognosis prediction of cancer are summarized. Finally, the prospects and challenges of applying exosome-based liquid biopsy to precision medicine are evaluated.
Collapse
Affiliation(s)
- Dan Yu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yixin Li
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Maoye Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Jianmei Gu
- Department of Clinical Laboratory Medicine, Nantong Tumor Hospital, Nantong, 226361, Jiangsu, China
| | - Wenrong Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Hui Cai
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Hospital of Jiangsu University, Lanzhou, 730000, Gansu, China
| | - Xinjian Fang
- Department of Oncology, Lianyungang Hospital Affiliated to Jiangsu University, Lianyungang, 222000, Jiangsu, China.
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Hospital of Jiangsu University, Lanzhou, 730000, Gansu, China.
- Department of Oncology, Lianyungang Hospital Affiliated to Jiangsu University, Lianyungang, 222000, Jiangsu, China.
| |
Collapse
|
53
|
Campos-Silva C, Cáceres-Martell Y, Sánchez-Herrero E, Sandúa A, Beneitez-Martínez A, González Á, Provencio M, Romero A, Jara-Acevedo R, Yáñez-Mó M, Valés-Gómez M. A simple immunoassay for extracellular vesicle liquid biopsy in microliters of non-processed plasma. J Nanobiotechnology 2022; 20:72. [PMID: 35135541 PMCID: PMC8822649 DOI: 10.1186/s12951-022-01256-5] [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: 10/18/2021] [Accepted: 01/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Extracellular vesicles (EVs), released by most cell types, provide an excellent source of biomarkers in biological fluids. However, in order to perform validation studies and screenings of patient samples, it is still necessary to develop general techniques permitting rapid handling of small amounts of biological samples from large numbers of donors. RESULTS Here we describe a method that, using just a few microliters of patient's plasma, identifies tumour markers exposed on EVs. Studying physico-chemical properties of EVs in solution, we demonstrate that they behave as stable colloidal suspensions and therefore, in immunocapture assays, many of them are unable to interact with a stationary functionalised surface. Using flocculation methods, like those used to destabilize colloids, we demonstrate that cationic polymers increase EV ζ-potential, diameter, and sedimentation coefficient and thus, allow a more efficient capture on antibody-coated surfaces by both ELISA and bead-assisted flow cytometry. These findings led to optimization of a protocol in microtiter plates allowing effective immunocapture of EVs, directly in plasma without previous ultracentrifugation or other EV enrichment. The method, easily adaptable to any laboratory, has been validated using plasma from lung cancer patients in which the epithelial cell marker EpCAM has been detected on EVs. CONCLUSIONS This optimized high throughput, easy to automate, technology allows screening of large numbers of patients to phenotype tumour markers in circulating EVs, breaking barriers for the validation of proposed EV biomarkers and the discovery of new ones.
Collapse
Affiliation(s)
- Carmen Campos-Silva
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, CNB-CSIC, Madrid, Spain
| | - Yaiza Cáceres-Martell
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, CNB-CSIC, Madrid, Spain
| | - Estela Sánchez-Herrero
- Laboratorio de Biopsia Líquida, Instituto de Investigación Sanitaria Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain.,Atrys Health, Barcelona, Spain
| | - Amaia Sandúa
- Service of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Álvaro González
- Service of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
| | - Mariano Provencio
- Laboratorio de Biopsia Líquida, Instituto de Investigación Sanitaria Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain.,Medical Oncology Department, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Atocha Romero
- Laboratorio de Biopsia Líquida, Instituto de Investigación Sanitaria Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain.,Medical Oncology Department, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | | | - María Yáñez-Mó
- Department of Molecular Biology, UAM - Centro de Biología Molecular Severo Ochoa, Madrid, Spain.,Instituto de Investigación del Hospital Universitario La Princesa, Madrid, Spain
| | - Mar Valés-Gómez
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, CNB-CSIC, Madrid, Spain.
| |
Collapse
|
54
|
Liquid Biopsies: Flowing Biomarkers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:341-368. [DOI: 10.1007/978-3-031-04039-9_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
55
|
Zhao S, Mi Y, Zheng B, Wei P, Gu Y, Zhang Z, Xu Y, Cai S, Li X, Li D. Highly-metastatic colorectal cancer cell released miR-181a-5p-rich extracellular vesicles promote liver metastasis by activating hepatic stellate cells and remodelling the tumour microenvironment. J Extracell Vesicles 2022; 11:e12186. [PMID: 35041299 PMCID: PMC8765330 DOI: 10.1002/jev2.12186] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/21/2021] [Accepted: 12/29/2021] [Indexed: 01/11/2023] Open
Abstract
Liver metastasis of colorectal cancer (CRLM) is the most common cause of CRC-related mortality, and is typically caused by interactions between CRC cells and the tumour microenvironment (TME) in the liver. However, the molecular mechanisms underlying the crosstalk between tumour-derived extracellular vesicle (EV) miRNAs and the TME in CRLM have yet to be fully elucidated. The present study demonstrated that highly metastatic CRC cells released more miR-181a-5p-rich EVs than cells which exhibit a low metastatic potential, in-turn promoting CRLM. Additionally, we verified that FUS mediated packaging of miR-181a-5p into CRC EVs, which in-turn persistently activated hepatic stellate cells (HSCs) by targeting SOCS3 and activating the IL6/STAT3 signalling pathway. Activated HSCs could secrete the chemokine CCL20 and further activate a CCL20/CCR6/ERK1/2/Elk-1/miR-181a-5p positive feedback loop, resulting in reprogramming of the TME and the formation of pre-metastatic niches in CRLM. Clinically, high levels of serum EV containing miR-181a-5p was positively correlated with liver metastasis in CRC patients. Taken together, highly metastatic CRC cells-derived EVs rich in miR-181a-5p could activate HSCs and remodel the TME, thereby facilitating liver metastasis in CRC patients. These results provide novel insight into the mechanism underlying liver metastasis in CRC.
Collapse
Affiliation(s)
- Senlin Zhao
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yushuai Mi
- Department of Gastrointestinal SurgeryThe Second HospitalCheeloo College of MedicineShandong UniversityJinanChina
| | - Binbin Zheng
- Department of General SurgeryShanghai General HospitalSchool of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Ping Wei
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Yanzi Gu
- Department of BiobankFudan University Shanghai Cancer CenterShanghaiChina
| | - Zhengxiang Zhang
- Department of OncologyYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
| | - Ye Xu
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Sanjun Cai
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xinxiang Li
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Dawei Li
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
56
|
Morasso C, Ricciardi A, Sproviero D, Truffi M, Albasini S, Piccotti F, Sottotetti F, Mollica L, Cereda C, Sorrentino L, Corsi F. Fast quantification of extracellular vesicles levels in early breast cancer patients by Single Molecule Detection Array (SiMoA). Breast Cancer Res Treat 2021; 192:65-74. [PMID: 34935096 PMCID: PMC8841315 DOI: 10.1007/s10549-021-06474-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/02/2021] [Indexed: 10/25/2022]
Abstract
PURPOSE Preliminary reports suggest that extracellular vesicles (EVs) might be a promising biomarker for breast cancer (BC). However, the quantification of plasmatic levels of EVs is a complex task. To overcome these limitations, we developed a new, fast, and easy to use assay for the quantification of EVs directly in plasma based on the use of Single-Molecule Array (SiMoA). METHODS By using SiMoA to identify CD9+/CD63+ EVs, we analyzed plasma samples of 181 subjects (95 BC and 86 healthy controls, HC). A calibration curve, made of a serial dilution of lyophilized standards from human plasma, was used in each run to ensure the obtainment of quantitative results from the assay. In a subgroup of patients, EVs concentrations were estimated in plasma before and after 30 days from cancer surgery. Additional information on the size of EVs were also acquired using a Nanosight system to obtain a clearer understanding of the mechanism underlying the releases of EVs associated with the presence of cancer. RESULTS The measured levels of EVs resulted significantly higher in BC patients (median values 1179.1 ng/µl vs 613.0 ng/µl, p < 0.0001). ROC curve was used to define the optimal cut-off level of the test at 1034.5 ng/µl with an AUC of 0.75 [95% CI 0.68-0.82]. EVs plasmatic concentrations significantly decreased after cancer surgery compared to baseline values (p = 0.014). No correlation was found between EVs concentration and clinical features of BC. CONCLUSION SiMoA assay allows plasmatic EVs levels detection directly without any prior processing. EVs levels are significantly higher in BC patients and significantly decreases after cancer surgery.
Collapse
Affiliation(s)
- Carlo Morasso
- Laboratory of Nanomedicine, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Alessandra Ricciardi
- Laboratory of Nanomedicine, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Daisy Sproviero
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Marta Truffi
- Laboratory of Nanomedicine, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Sara Albasini
- Breast Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Francesca Piccotti
- Laboratory of Nanomedicine, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Federico Sottotetti
- Medical Oncology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Ludovica Mollica
- Medical Oncology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Cristina Cereda
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Luca Sorrentino
- Colorectal Surgery Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Fabio Corsi
- Breast Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy. .,Dipartimento di Scienze Biomediche e Cliniche "L. Sacco", Università di Milano, Via G.B. Grassi, 74, 20157, Milan, Italy.
| |
Collapse
|
57
|
Dai Y, Wang Y, Cao Y, Yu P, Zhang L, Liu Z, Ping Y, Wang D, Zhang G, Sang Y, Wang X, Tao Z. A Multivariate Diagnostic Model Based on Urinary EpCAM-CD9-Positive Extracellular Vesicles for Prostate Cancer Diagnosis. Front Oncol 2021; 11:777684. [PMID: 34900726 PMCID: PMC8652292 DOI: 10.3389/fonc.2021.777684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/29/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction Prostate cancer (PCa) is one of the most frequently diagnosed cancers and the leading cause of cancer death in males worldwide. Although prostate-specific antigen (PSA) screening has considerably improved the detection of PCa, it has also led to a dramatic increase in overdiagnosing indolent disease due to its low specificity. This study aimed to develop and validate a multivariate diagnostic model based on the urinary epithelial cell adhesion molecule (EpCAM)-CD9-positive extracellular vesicles (EVs) (uEVEpCAM-CD9) to improve the diagnosis of PCa. Methods We investigated the performance of uEVEpCAM-CD9 from urine samples of 193 participants (112 PCa patients, 55 benign prostatic hyperplasia patients, and 26 healthy donors) to diagnose PCa using our laboratory-developed chemiluminescent immunoassay. We applied machine learning to training sets and subsequently evaluated the multivariate diagnostic model based on uEVEpCAM-CD9 in validation sets. Results Results showed that uEVEpCAM-CD9 was able to distinguish PCa from controls, and a significant decrease of uEVEpCAM-CD9 was observed after prostatectomy. We further used a training set (N = 116) and constructed an exclusive multivariate diagnostic model based on uEVEpCAM-CD9, PSA, and other clinical parameters, which showed an enhanced diagnostic sensitivity and specificity and performed excellently to diagnose PCa [area under the curve (AUC) = 0.952, P < 0.0001]. When applied to a validation test (N = 77), the model achieved an AUC of 0.947 (P < 0.0001). Moreover, this diagnostic model also exhibited a superior diagnostic performance (AUC = 0.917, P < 0.0001) over PSA (AUC = 0.712, P = 0.0018) at the PSA gray zone. Conclusions The multivariate model based on uEVEpCAM-CD9 achieved a notable diagnostic performance to diagnose PCa. In the future, this model may potentially be used to better select patients for prostate transrectal ultrasound (TRUS) biopsy.
Collapse
Affiliation(s)
- Yibei Dai
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yiyun Wang
- Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Cao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Pan Yu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lingyu Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenping Liu
- Department of Laboratory Medicine, The First People's Hospital of Yuhang District, Hangzhou, China
| | - Ying Ping
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Danhua Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Gong Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yiwen Sang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xuchu Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
58
|
Li JW, Shi D, Wan XC, Hu J, Su YF, Zeng YP, Hu ZJ, Yu BH, Zhang QL, Wei P, Zhou XY. Universal extracellular vesicles and PD-L1+ extracellular vesicles detected by single molecule array technology as circulating biomarkers for diffuse large B cell lymphoma. Oncoimmunology 2021; 10:1995166. [PMID: 34745768 PMCID: PMC8565827 DOI: 10.1080/2162402x.2021.1995166] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Plasma extracellular vesicles (EVs) have been reported to be a promising source of diagnostic and prognostic biomarkers in various cancers. However, further research in this area is needed due to the limitations of circulating extracellular vesicles detection methods. Using the Single Molecule array (SiMoa) technology, we developed two extracellular vesicle detection assays, CD9-CD63 and PD-L1-CD63, to determine circulating universal EVs and PD-L1 positive EVs, respectively. A total of 164 diffuse large B-cell lymphoma (DLBCL) patients were retrospectively included in this study. Compared with healthy volunteers (n = 25), elevated CD9-CD63 and PD-L1-CD63 signals were detected in the plasma of DLBCL patients (n = 164). High CD9-CD63 signals was associated with molecular subtype, extranodal site and treatment response in DLBCL. A high PD-L1-CD63 signal was also associated with certain clinical features, including extranodal site and treatment response. CD9-CD63 and PD-L1-CD63 signals were found to be important prognostic factors for both progression-free and overall survival. Furthermore, PD-L1-positive EVs were found in all patients, though PD-L1 protein expression was positive in only 35.4% (17/48) of tumor biopsies. No correlation was found between circulating PD-L1+ EVs and soluble PD-L1 (sPD-L1) levels. Our results show that plasma universal EV and PD-L1-positive EV levels are significantly elevated in DLBCL and might serve as biomarkers for predicting survival outcomes in DLBCL patients.
Collapse
Affiliation(s)
- Ji-Wei Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Di Shi
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Xiao-Chun Wan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Jue Hu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Yi-Fan Su
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Yu-Peng Zeng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Zi-Juan Hu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Bao-Hua Yu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Qun-Ling Zhang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ping Wei
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Xiao-Yan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| |
Collapse
|
59
|
Ma F, Vayalil J, Lee G, Wang Y, Peng G. Emerging role of tumor-derived extracellular vesicles in T cell suppression and dysfunction in the tumor microenvironment. J Immunother Cancer 2021; 9:jitc-2021-003217. [PMID: 34642246 PMCID: PMC8513270 DOI: 10.1136/jitc-2021-003217] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2021] [Indexed: 02/07/2023] Open
Abstract
Immunotherapeutic drugs including immune checkpoint blockade antibodies have been approved to treat patients in many types of cancers. However, some patients have little or no reaction to the immunotherapy drugs. The mechanisms underlying resistance to tumor immunotherapy are complicated and involve multiple aspects, including tumor-intrinsic factors, formation of immunosuppressive microenvironment, and alteration of tumor and stromal cell metabolism in the tumor microenvironment. T cell is critical and participates in every aspect of antitumor response, and T cell dysfunction is a severe barrier for effective immunotherapy for cancer. Emerging evidence indicates that extracellular vesicles (EVs) secreted by tumor is one of the major factors that can induce T cell dysfunction. Tumor-derived EVs are widely distributed in serum, tissues, and the tumor microenvironment of patients with cancer, which serve as important communication vehicles for cancer cells. In addition, tumor-derived EVs can carry a variety of immune suppressive signals driving T cell dysfunction for tumor immunity. In this review, we explore the potential mechanisms employed by tumor-derived EVs to control T cell development and effector function within the tumor microenvironment. Especially, we focus on current understanding of how tumor-derived EVs molecularly and metabolically reprogram T cell fates and functions for tumor immunity. In addition, we discuss potential translations of targeting tumor-derived EVs to reconstitute suppressive tumor microenvironment or to develop antigen-based vaccines and drug delivery systems for cancer immunotherapy.
Collapse
Affiliation(s)
- Feiya Ma
- Biology, Saint Louis University, Saint Louis, Missouri, USA
| | - Jensen Vayalil
- Biology, Saint Louis University, Saint Louis, Missouri, USA
| | - Grace Lee
- Biology, Saint Louis University, Saint Louis, Missouri, USA
| | - Yuqi Wang
- Biology, Saint Louis University, Saint Louis, Missouri, USA
| | - Guangyong Peng
- Internal Medicine, Saint Louis University, Saint Louis, Missouri, USA
| |
Collapse
|
60
|
Yang L, Zheng S, Liu Q, Liu T, Zhang Q, Han X, Tuerxun A, Lu X. Plasma‑derived exosomal pyruvate kinase isoenzyme type M2 accelerates the proliferation and motility of oesophageal squamous cell carcinoma cells. Oncol Rep 2021; 46:216. [PMID: 34396437 PMCID: PMC8377463 DOI: 10.3892/or.2021.8167] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/17/2021] [Indexed: 01/15/2023] Open
Abstract
Exosomal pyruvate kinase isoenzyme type M2 (PKM2) has been found to play a key role in the progression of human hepatocarcinoma. However, exosomal PKM2 (especially plasma‑derived exosomal PKM2), in patients with oesophageal squamous cell carcinoma (ESCC) has not been well defined. In the present study, plasma‑derived exosomes were isolated from healthy controls and patients with ESCC, and identified by transmission electronic microscopy, western blotting, nano‑flow cytometry, nanoparticle tracking and phagocytosis analysis; exosomal PKM2 was detected by western blotting and ELISA. In addition, changes in cellular proliferation and motility in recipient cells (Eca109) were assessed using Cell Counting Kit‑8, colony formation, wound‑healing and Transwell assays. The PKM2 content was higher in exosomes from patients with ESCC than in those from healthy donors. Furthermore, exosomes from patients with ESCC enhanced the proliferation and motility of ESCC cells in vitro. Notably, PKM2 was found to be transferred by exosomes, and was able to act by activating STAT3. To verify the association between PKM2 and STAT3, immunohistochemistry was employed to analyse the protein levels of PKM2 and pSTAT3Tyr705. These data revealed that PKM2 and pSTAT3Tyr705 were upregulated and associated with overall survival in patients with ESCC. Therefore, the present study highlights that exosomes from patients with ESCC enhance the migration and invasiveness of ESCC cells by transferring PKM2.
Collapse
Affiliation(s)
- Lifei Yang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
- First Department of Lung Cancer Chemotherapy, Cancer Hospital Affiliated with Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 83000, P.R. China
| | - Shutao Zheng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Qing Liu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Tao Liu
- Department of Clinical Laboratory, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 83000, P.R. China
| | - Qiqi Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Xiujuan Han
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Aerziguli Tuerxun
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Xiaomei Lu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
- XinJiang Branch of Key Laboratory of Cancer Immunotherapy and Radiotherapy, Chinese Academy of Medical Sciences, Urumqi, Xinjiang Uygur Autonomous Region 83000, P.R. China
| |
Collapse
|
61
|
Li P, Wang J, Gao M, Wang J, Ma Y, Gu Y. Membrane Feature-Inspired Profiling of Extracellular Vesicles for Pancreatic Cancer Diagnosis. Anal Chem 2021; 93:9860-9868. [PMID: 34247486 DOI: 10.1021/acs.analchem.1c01712] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) have recently emerged as a promising tumor biomarker, and EV phenotyping offers many benefits for cancer diagnosis. However, the practicality of EV assays remains a challenge due to macromolecule disturbances, biomarker heterogeneities, and EV abundance limitations. Here, we demonstrate a membrane-based biosensor for precise and sensitive EV identification. The sensor synergistically integrates EV capture and detection by virtue of EV membrane features (membrane protein and lipid bilayer), comprising antibody-conjugated magnetic beads (AbMBs) and duplex-specific nuclease (DSN)-mediated amplification cycles. Bivalent cholesterol (biChol)-modified RNA-DNA duplexes are designed to insert into the EV membrane, transforming EV signals into RNA signals and initiating the signal amplification. The membrane-based signal production pattern eliminates protein interference. By employing four antibodies specific to PCa-related membrane proteins, the AbMB-biChol platform enables the successful differentiation and monitoring of PCa-related EVs and distinguishes PCa patients from healthy donors with improved efficacy, exhibiting superior efficiency over the analyses based on clinically used biomarker CA19-9 and PCa-related proteins. As such, the developed system has great potential for clinical PCa diagnosis.
Collapse
Affiliation(s)
- Ping Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Jie Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China
| | - Mengqiu Gao
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Jue Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Ma
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yueqing Gu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
62
|
Gualerzi A, Picciolini S, Carlomagno C, Rodà F, Bedoni M. Biophotonics for diagnostic detection of extracellular vesicles. Adv Drug Deliv Rev 2021; 174:229-249. [PMID: 33887403 DOI: 10.1016/j.addr.2021.04.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/26/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
Extracellular Vesicles (EVs) are versatile carriers for biomarkers involved in the pathogenesis of multiple human disorders. Despite the increasing scientific and commercial interest in EV application in diagnostics, traditional biomolecular techniques usually require consistent sample amount, rely on operator-dependent and time- consuming procedures and cannot cope with the nano-size range of EVs, limiting both sensitivity and reproducibility of results. The application of biophotonics, i.e. light-based methods, for the diagnostic detection of EVs has brought to the development of innovative platforms with excellent sensitivity. In this review, we propose an overview of the most promising and emerging technologies used in the field of EV-related biomarker discovery. When tested on clinical samples, the reported biophotonic approaches in most cases have managed to discriminate between nanovesicles and contaminants, achieved much higher resolution compared to traditional procedures, and reached moderate to excellent diagnostic accuracy, thus demonstrating great potentialities for their clinical translation.
Collapse
|
63
|
Liu MX, Zhang H, Zhang XW, Chen S, Yu YL, Wang JH. Nanozyme Sensor Array Plus Solvent-Mediated Signal Amplification Strategy for Ultrasensitive Ratiometric Fluorescence Detection of Exosomal Proteins and Cancer Identification. Anal Chem 2021; 93:9002-9010. [PMID: 34143614 DOI: 10.1021/acs.analchem.1c02010] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tumor exosomes with molecular marker-proteins inherited from their parent cells have emerged as a promising liquid biopsy biomarker for cancer diagnosis. However, facile, robust, and sensitive detection of exosomal proteins remains challenging. Therefore, a nanozyme sensor array is constructed by using aptamer-modified C3N4 nanosheets (Apt/C3N4 NSs) together with a solvent-mediated signal amplification strategy for ratiometric fluorescence detection of exosomal proteins. Three aptamers specific to exosomal proteins are selected to construct Apt/C3N4 NSs for high specific recognition of exosomal proteins. The adsorption of aptamers enhances the catalytic activity of C3N4 NSs as a nanozyme for oxidation of o-phenylenediamine (oPD) to 2,3-diaminophenazine (DAP). In the presence of target exosomes, the strong affinity between aptamer and exosome leads to the disintegration of Apt/C3N4 NSs, resulting in a decrease of catalytic activity, thereby reducing the production of DAP. The ratiometric fluorescence signal based on a photoinduced electron transfer (PET) effect between DAP and C3N4 NSs is dependent on the concentration of DAP generated, thus achieving highly facile and robust detection of exosomal proteins. Remarkably, the addition of organic solvent-1,4-dioxane can sensitize the luminescence of DAP without affecting the intrinsic fluorescence of C3N4 NSs, achieving the amplification of the aptamer-exosome recognition events. The detection limit for exosome is 2.5 × 103 particles/mL. In addition, the accurate identification of cancer can be achieved by machine learning algorithms to analyze the difference of exosomal proteins from different patients' blood. We hope that this facile, robust, sensitive, and versatile nanozyme sensor array would become a promising tool in the field of cancer diagnosis.
Collapse
Affiliation(s)
- Meng-Xian Liu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - He Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Xue-Wei Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Shuai Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Yong-Liang Yu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| |
Collapse
|
64
|
Wu F, Gu Y, Kang B, Heskia F, Pachot A, Bonneville M, Wei P, Liang J. PD-L1 detection on circulating tumor-derived extracellular vesicles (T-EVs) from patients with lung cancer. Transl Lung Cancer Res 2021; 10:2441-2451. [PMID: 34295653 PMCID: PMC8264343 DOI: 10.21037/tlcr-20-1277] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/06/2021] [Indexed: 12/26/2022]
Abstract
Background Recent breakthroughs in therapies with immune checkpoint inhibitors (ICIs) have revolutionized the treatment of lung cancer. However, only 15–25% of patients respond to the ICIs therapy, and methods to identify those responsive patients are currently a hot research topic. PD-L1 expression measured on tumor tissues using immunohistochemistry (IHC) was approved as one of the companion diagnostic methods, but it is invasive and cannot be used to monitor dynamic changes in PD-L1 expression during treatments. Methods In this study, we developed an Epcam-PD-L1 extracellular vesicle (EV) detection prototype using the Simoa platform. This assay detected PD-L1 expression levels on tumor-derived exosomes from the lung cancer cell lines A549 and SK-MES1. In addition, 35 plasma samples from patients with lung cancer were tested with this assay and the results were compared to the tissue PD-L1 expression levels represented by the tumor proportion score (TPS). Results PD-L1 TPS-positive patients (≥1% IHC TPS) had significantly higher Simoa Epcam-PD-L1 signals than TPS-negative patients (<1% IHC TPS, P=0.026). The Simoa Epcam-PD-L1 area under curve (AUC) reached 0.776, with a sensitivity of 92.86% and a specificity of 71.43%. When PD-L1 TPS-positive patients were defined as having an IHC TPS ≥10%, the greatest difference in Epcam-PD-L1 signals was observed between IHC TPS-positive and IHC TPS-negative groups (P=0.0024) and the Simoa Epcam-PD-L1 AUC reached 0.832. Finally, the Spearman’s correlation coefficient showed a significant correlation between the TPS and Simoa Epcam-PD-L1 signals (0.428, P=0.0104). Conclusions Based on our results, our Simoa Epcam-PD-L1 EV detection assay is a potential liquid biopsy method to predict the PD-L1 expression level in patients with lung cancer.
Collapse
Affiliation(s)
- Fei Wu
- Fudan University Shanghai Cancer Center-Institut Mérieux Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,bioMérieux (Shanghai) Company Limited, Shanghai, China
| | - Yanzi Gu
- Biobank, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Bin Kang
- Fudan University Shanghai Cancer Center-Institut Mérieux Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,bioMérieux (Shanghai) Company Limited, Shanghai, China
| | - Fabienne Heskia
- Global Medical Affairs, bioMérieux SA, Marcy l'Etoile, France
| | - Alexandre Pachot
- Open Innovation & Partnerships Department, bioMérieux SA, Marcy l'Etoile, France
| | | | - Ping Wei
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ji Liang
- Fudan University Shanghai Cancer Center-Institut Mérieux Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,bioMérieux (Shanghai) Company Limited, Shanghai, China
| |
Collapse
|
65
|
Thangavelu B, Boutté AM. Single Molecule Assay for Ultrasensitive Detection of Cathepsin B in Human Blood. ACS OMEGA 2021; 6:9609-9616. [PMID: 33869941 PMCID: PMC8047647 DOI: 10.1021/acsomega.1c00180] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/19/2021] [Indexed: 05/23/2023]
Abstract
Cathepsin B (catB) is a lysosomal cysteine protease expressed in several cells and organs, where it plays a role in protein degradation and turnover. Extracellular, secreted catB has utility as a biomarker for a host of pathological or physiological states, including a myriad of cancers or neurological diseases and injuries. Analytical or diagnostic assessment may be limited by biological sample volume availability. Pathologically relevant changes in catB levels may occur at low-moderate concentrations that require accurate measurement to differentiate from basal levels. Furthermore, biological samples like plasma and serum, often occlude accurate catB measurements because of background and high variance, vastly limiting the ability to detect catB as a peripheral biomarker. Techniques for ultrasensitive protein detection that require low volumes of sample are necessary. To overcome these challenges, a digital enzyme-linked immunosorbent assay (ELISA) was developed for differential detection of catB within less than 5 μL of serum and plasma using the single molecule array (SiMoA) platform, which offers 1000-times more sensitivity and vastly reduced variance compared to colorimetric tests. In buffer, curve-fitting estimated the limit of detection (LoD) to be ∼1.56 and ∼8.47 pg/mL using two-step or three-step assay configurations, respectively. After correcting for endogenous levels, the estimated LoD was ∼4.7 pg/mL in the serum or plasma with the two-step assay. The lower limit of quantitation was ∼2.3 pg/mL in the buffer and ∼9.4 pg/mL in the serum or plasma, indicting the ability to measure small changes above endogenous levels within blood samples.
Collapse
|
66
|
Lorico A, Lorico-Rappa M, Karbanová J, Corbeil D, Pizzorno G. CD9, a tetraspanin target for cancer therapy? Exp Biol Med (Maywood) 2021; 246:1121-1138. [PMID: 33601913 DOI: 10.1177/1535370220981855] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the present minireview, we intend to provide a brief history of the field of CD9 involvement in oncogenesis and in the metastatic process of cancer, considering its potential value as a tumor-associated antigenic target. Over the years, CD9 has been identified as a favorable prognostic marker or predictor of metastatic potential depending on the cancer type. To understand its implications in cancer beside its use as an antigenic biomarker, it is essential to know its physiological functions, including its molecular partners in a given cell system. Moreover, the discovery that CD9 is one of the most specific and broadly expressed markers of extracellular membrane vesicles, nanometer-sized entities that are released into extracellular space and various physiological body fluids and play a role in intercellular communication under physiological and pathological conditions, notably the establishment of cancer metastases, has added a new dimension to our knowledge of CD9 function in cancer. Here, we will discuss these issues as well as the possible cancer therapeutic implications of CD9, their limitations, and pitfalls.
Collapse
Affiliation(s)
- Aurelio Lorico
- Touro University College of Medicine, Henderson, NV 89014, USA.,Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | | | - Jana Karbanová
- Biotechnology Center and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany
| | - Denis Corbeil
- Biotechnology Center and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany
| | - Giuseppe Pizzorno
- University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Erlanger Health System, Chattanooga, TN 37403 , USA
| |
Collapse
|
67
|
He X, Zhong X, Hu Z, Zhao S, Wei P, Li D. An insight into small extracellular vesicles: Their roles in colorectal cancer progression and potential clinical applications. Clin Transl Med 2020; 10:e249. [PMID: 33377655 PMCID: PMC7733319 DOI: 10.1002/ctm2.249] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers and a leading cause of mortality worldwide. Small extracellular vesicles (sEVs) are nano-sized extracellular vesicles containing a variety of bioactive molecules, such as nucleic acids, proteins, lipids, and metabolites. Recent evidence from CRC has revealed that sEVs contribute to tumorigenesis, progression, and drug resistance, and serve as a tool for "liquid biopsy" and a drug delivery system for therapy. In this review, we summarize information about the roles of sEVs in the proliferation, invasion, migration, epithelial-mesenchymal transition, formation of the premetastatic niche, and drug resistance to elucidate the mechanisms governing sEVs in CRC and to identify novel targets for therapy and prognostic and diagnostic biomarkers.
Collapse
Affiliation(s)
- Xuefeng He
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xinyang Zhong
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zijuan Hu
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
- Institute of PathologyFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Senlin Zhao
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Ping Wei
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
- Institute of PathologyFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Dawei Li
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|